Fuxi Song 1, Chunmiao Ye 2, Feng Qi 3, Ping Zhang 2, Xuexiang Wang 4, Yanfeng Lü 5, Alejandro Fernandez-Escobar 6, Chao Zheng 2* and Liang Li 2*

Similar documents
DOI /yydb medetomidine a review of clinical applications J. Curr Opin Anaesthesiol

PDF of Trial CTRI Website URL -

Int. J. Pharm. Sci. Rev. Res., 36(1), January February 2016; Article No. 38, Pages: *Corresponding author s

Comparison of dexmedetomidine and propofol for conscious sedation in inguinal hernia repair: A prospective, randomized, controlled trial

Associate Professor, Department of Anaesthesiology, Government Thoothukudi Medical College, Thoothukudi, Tamil Nadu, India, 2

Hemodynamic effects of dexmedetomidine-- fentanyl vs. nalbuphine--propofol in plastic surgery

Role of Dexmedetomidine as an Anesthetic Adjuvant in Laparoscopic Surgery

Corresponding author: V. Dua, Department of Anaesthesia, BJ Wadia Hospital for Children, Parel, Mumbai, India.

A randomized control study of dexmedetomidine versus fentanyl as an anesthetic adjuvant in supratentorial craniotomies

Dexmedetomidine. Dr.G.K.Kumar,M.D.,D.A., Assistant Professor, Madras medical college,chennai. History

Study the Effect of Dexmedetomidine on Emergence Agitation after Nasal Surgeries

Original Article Effects of low dose midazolam on bradycardia and sedation during dexmedetomidine infusion

Comparison of dexmedetomidine and propofol in mechanically ventilated patients with sepsis: A pilot study

Original Article INTRODUCTION. Abstract

ASMIC 2016 DEXMEDETOMIDINE IN THE INTENSIVE CARE UNIT DR KHOO TIEN MENG

A Clinical Study of Dexmedetomidine under Combined Spinal Epidural Anaesthesia at a Tertiary Care Hospital

Study between clonidine and dexmedetomidine in attenuation of pressor response during endotracheal intubation

Comparison of Intensive Care Unit Sedation Using Dexmedetomidine, Propofol, and Midazolam

Rajaclimax Kirubahar, Bose Sundari, Vijay Kanna*, Kanakasabai Murugadoss

Comparison of Clonidine and Dexmedetomidine on Cardiovascular Stability in Laparoscopic Cholecystectomy

Susan Becker DNP, RN, CNS, CCRN, CCNS Marymount University, Arlington, VA

Haemodynamic and anaesthetic advantages of dexmedetomidine

Dexmedetomidine and its Injectable Anesthetic-Pain Management Combinations

Comparison of anesthesia with a morphine lidocaine ketamine infusion or a morphine lidocaine epidural on time to extubation in dogs

Dr. PratekKoolwal, Dr.BribalBaj, DrKashif M Madani, Dr.MohitSomani, Dr. Vijay Mathur.

Propofol vs Dexmedetomidine

A SYSTEMATIC REVIEW ON THE USE OF DEXMEDETOMIDINE AS A SOLE AGENT FOR INTRAVENOUS MODERATE SEDATION

Preliminary UK experience of dexmedetomidine, a novel agent for postoperative sedation in the intensive care unit

Effects of different doses of dexmedetomidine on inflammatory factors and T lymphocyte subsets in elderly patients undergoing laparoscopic surgery

T u l a n e U n i v e r s i t y I A C U C Guidelines for Rodent & Rabbit Anesthesia, Analgesia and Tranquilization & Euthanasia Methods

Original Article Dose-dependent effects of dexmedetomidine during one-lung ventilation in patients undergoing lobectomy

Comparison of several dosing schedules of intravenous dexmedetomidine in elderly patients under spinal anesthesia

Review Article The Effects of Intravenous Dexmedetomidine Injections on IOP in General Anesthesia Intubation: A Meta-Analysis

A COMPARATIVE STUDY OF MIDAZOLAM, PROPOFOL AND DEXMEDETOMIDINE INFUSIONS FOR SEDATION IN ME- CHANICALLY VENTILATED PATIENTS IN ICU

The comparison of the effects of intravenous ketamine or dexmedetomidine infusion on spinal block with bupivacaine

PAIN Effect of intra-articular dexmedetomidine on postoperative analgesia after arthroscopic knee surgery

Chronic subdural hematoma (CSDH) is one of the most

Efficacy of dexmedetomidine in reducing postoperative morphine consumption in patients undergoing total abdominal hysterectomy

Dıfferent Doses Of Dexmedetomidine On Controllıng Haemodynamıc Responses To Tracheal Intubatıon

Ashraf Darwish, Rehab Sami, Mona Raafat, Rashad Aref and Mohamed Hisham

Appendix: Outcomes when Using Adjunct Dexmedetomidine with Propofol Sedation in

Dexmedetomidine intravenous sedation using a patient-controlled sedation infusion pump: a case report

POST-OPERATIVE ANALGESIA AND FORMULARIES

A bispectral index guided study on the effect of dexmedetomidine on sevoflurane requirements during elective laparoscopic surgeries

International Journal of Health Sciences and Research ISSN:

S Kumar, B B Kushwaha, R Prakash, S Jafa, A Malik, R Wahal, J Aggarwal, R Kapoor

Intraoperative Sedation During Epidural Anesthesia: Dexmedetomidine Vs Midazolam

Effects of Dexmedetomidine on Serum Interleukin-6, Hemodynamic Stability, and Postoperative Pain Relief in Elderly Patients under Spinal Anesthesia

Premedication with alpha-2 agonists procedures for monitoring anaesthetic

Use of Dexmedetomidine for Sedation of Children Hospitalized in the Intensive Care Unit

Dexmedetomidine vs. Propofol for Short-Term Sedation of Postoperative Mechanically Ventilated Patients

SCIENTIFIC COOPERATIONS MEDICAL WORKSHOPS July, 2015, Istanbul - TURKEY

Pain Management in Racing Greyhounds

Parthasarathy et al. Sri Lankan Journal of Anaesthesiology: 25(2):76-81(2017)

Additive analgesic effect of dexmedetomidine and dezocine administered intrathecally in a mouse pain model

Study of Dexmedetomidine as intramuscular premedication in outpatient cataract surgery: A placebo controlled study

Syddansk Universitet. Published in: Cochrane Database of Systematic Reviews. DOI: / CD pub2. Publication date: 2016

Dexmedetomidine and stress response Madhusudan et al

Comparison of dexmedetomidine v/s propofol used as adjuvant with combined spinal epidural anaesthesia for joint replacement surgeries

Associate Professor, Department of Anaesthesiology, Rangaraya Medical College, Kakinada, East Godavari, Andhra Pradesh, India, 2

Quality of MRI pediatric sedation: Comparison between intramuscular and intravenous dexmedetomidine

Alfaxan. (alfaxalone 10 mg/ml) Intravenous injectable anesthetic for use in cats and dogs. TECHNICAL NOTES DESCRIPTION INDICATIONS

Evaluation of efficacy of sedative and analgesic effects of single IV dose of dexmedetomidine in post-operative patients

RETRACTED. Dexmedetomidine infusion is associated with enhanced renal function after thoracic surgery

A New Advancement in Anesthesia. Your clear choice for induction.

The effects of intravenous dexmedetomidine on spinal anesthesia: comparision of different dose of dexmedetomidine

Therapeutics and clinical risk management (2011) Vol.7:291~299. Dexmedetomidine hydrochloride as a long-term sedative.

A Comparative Evaluation of Intranasal Dexmedetomidine and Intranasal Midazolam for Premedication in Pediatric Surgery

Evaluation of clinical analgesic efficacy of dexmedetomidine. combined with sufentanil on uterine contraction pain after

Clinical applicability of dexmedetomidine for sedation, premedication and analgesia in cats 1 / 2007

Dexmedetomidine for prevention of delirium in elderly patients after non-cardiac surgery: a randomised, double-blind, placebo-controlled trial

Day 90 Labelling, PL LABELLING AND PACKAGE LEAFLET

Disclosures. Dexmedetomidine: The Good, The Bad and The Delirious. The Delirious. Objectives. Characteristics of Delirium. Definition of Delirium

Original Article Dexmedetomidine reduces shivering during epidural anesthesia

Highly variable pharmacokinetics of dexmedetomidine during intensive care: a case report

SUMMARY OF PRODUCT CHARACTERISTICS

Maduranthagam. 2 Associate Professor, Department Of Anesthesia, Karpagavinayaga Medical College And Hospital, Maduranthagam.

SUMMARY OF PRODUCT CHARACTERISTICS

What dose of methadone should I use?

A comparison of dexmedetomidine and midazolam for sedation in third molar surgery*

Post-graduate Trainee, Department of Anaesthesiology and Critical Care, Gauhati Medical College and Hospital, Guwahati, Assam, India, 2

Evaluation of dexmedetomine in anesthesia care for elderly patients with obstructive sleep apnea

Comparative Study of Dexmedetomidine and Propofol for Intraoperative Sedation During Surgery Under Regional Anaesthesia

Procedure # IBT IACUC Approval: December 11, 2017

Dexmedetomidine as an additive to local anesthetics compared with intravenous dexmedetomidine in peribulbar block for cataract surgery

SUMMARY OF PRODUCT CHARACTERISTICS

Perioperative Care of Swine

Study of efficacy and safety of intravenous Dexmedetomidine infusion as an adjuvant to Bupivacaine spinal anaesthesia in Abdominal hysterectomy

Yuan Han 1,2, Liu Han 1, Mengmeng Dong 1, Qingchun Sun 1, Ke Ding 1, Zhenfeng Zhang 1, Junli Cao 1,2* and Yueying Zhang 1*

Prophylactic antibiotic timing and dosage. Dr. Sanjeev Singh AIMS, Kochi

Summary of Product Characteristics

Dexmedetomidine for Emergence Agitation after Sevoflurane Anesthesia in Preschool Children Undergoing Day Case Surgery: Comparative Dose-Ranging Study

Comparison of two doses of intranasal dexmedetomidine as premedication in children

Introduc/on. Introduc/on 2/3/16. Garret Pach/nger, VMD, DACVECC COO, VETgirl. Jus/ne A. Lee, DVM, DACVECC, DABT CEO, VETgirl

Cheung, CW; Ying, CLA; Chiu, WK; Wong, GTC; Ng, KFJ; Irwin, MG

N.C. A and T List of Approved Analgesics 1 of 5

DREXEL UNIVERSITY COLLEGE OF MEDICINE ANIMAL CARE AND USE COMMITTEE POLICY FOR PREOPERATIVE AND POSTOPERATIVE CARE FOR NON-RODENT MAMMALS

Effective dose of dexmedetomidine to induce adequate sedation in elderly patients under spinal anesthesia

TELAZOL (tiletamine and zolazepam for injection) IV Induction Claim FAQs 1, 2

A Comparison of Dexmedetomidine and Midazolam for Sedation in Gynecologic Surgery Under Epidural Anesthesia

Transcription:

Song et al. BMC Anesthesiology (2018) 18:145 https://doi.org/10.1186/s12871-018-0608-3 RESEARCH ARTICLE Open Access Effect of perioperative infusion of Dexmedetomidine combined with Sufentanil on quality of postoperative analgesia in patients undergoing laparoscopic nephrectomy: a CONSORTprospective, randomized, controlled trial Fuxi Song 1, Chunmiao Ye 2, Feng Qi 3, Ping Zhang 2, Xuexiang Wang 4, Yanfeng Lü 5, Alejandro Fernandez-Escobar 6, Chao Zheng 2* and Liang Li 2* Abstract Background: Postoperative pain is one of the most common symptoms after surgery, which brings physical discomfort to patients. In addition, it may cause a series of complications, and even affect the long-term quality of life. The purpose of this prospective, randomized, double-blinded, controlled trial is to investigate the efficacy and safety of dexmedetomidine combined with sufentanil to attenuate postoperative pain in patients after laparoscopic nephrectomy. Methods: Ninety patients undergoing laparoscopic nephrectomy were randomized into three groups: the control (sufentanil 0.02 μg/kg/h, Group C), sufentanil plus low dose of dexmedetomidine (0.02 μg/kg/h each, Group D1), and sufentanil plus high dose of dexmedetomidine (0.04 μg/kg/h, Group D2). The patient-controlled analgesia was programmed to deliver a bolus dose of 0.5 ml, followed by an infusion of 2 ml/h and a lockout time of 10 min. The primary goal was to calculate the cumulative amount of self-administered sufentanil; the secondary goals were to estimate pain intensity using the numerical rating scale (NRS), level of sedation, the first bowel movement, concerning adverse effects as well as duration of postoperative hospital stay. Results: The total consumption of sufentanil in group D1 and D2 were significantly lower than in group C during the first 8haftersurgery(P < 0.05), whereas there were no statistically significant differences (P > 0.05) between group D1 and D2. Compared with group C, the NRS scores at rest during first 8 h after surgery were significantly lower in group D1 (P < 0.05). The NRS scores, neither at rest nor with movement, show statistically significant differences between group D1 and D2 at each time point following surgery (P > 0.05). The time to first flatus was shorter in group D1 compared with the control group (P < 0.05). In addition, compared with group C, group D1 and D2 had a shorter time for first defecation (P <0.05). Conclusions: Dexmedetomidine combined with sufentanil showed better postoperative analgesia without adverse effects, as well as facilitated bowel movements for patients undergoing laparoscopic nephrectomy. (Continued on next page) * Correspondence: chaozheng@sdu.edu.cn; liliang@sdu.edu.cn 2 Department of Breast Surgery, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan 250033, China Full list of author information is available at the end of the article The Author(s). 2018 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Song et al. BMC Anesthesiology (2018) 18:145 Page 2 of 10 (Continued from previous page) Trial registration: We registered this study in a Chinese Clinical Trial Registry (ChiCTR) centre on Dec 23 2015 and received the registration number: ChiCTR-IPR-15007628. Keywords: Dexmedetomidine, Sufenanil, Postoperative analgesia, Flatus, Defecation Background Postoperative pain is a common postoperative complication that can aggravate the body s stress response. It can disturb the endocrine and immune function, as well as it is considered to be a risk factor for postoperative chronic pain. Meanwhile, severe postoperative pain will delay the recovery of patients after surgery, and even influence the quality of long-term survival [1 3]. Therefore, effective postoperative analgesia has a positive impacts on the recovery of patients following surgery. Ideal analgesia is defined as a measure that is easy to implement, effective, and has limited number of adverse effects. Postoperative patient-controlled analgesia (PCA) was introduced in the early 1980s. It is a delivery system that can be self-controlled by patients. Compared with traditional intramuscular injection of analgesia, PCA can be impactful as it alleviates postoperative pain, reduces drug consumption as well as it has less frequent postoperative complications [4]. Sufentanil is a selective μ - opioid receptor agonist that is characterized by its a fast onset, short duration of action and a strong analgesic effect (12-fold greater intrinsic potency than fentanyl) [5], has now become a common medication used in PCA. However, similar to other opioid drugs, sufentanil has significant side effects such as respiratory depression, nausea, vomiting and constipation [6]. In addition, it can potentially cause severe gastrointestinal adverse effects that may lead to dehydration, electrolyte imbalance, and a delay in enteral nutrition [7]. These will ultimately prolong hospital stay and increase the cost of hospitalization. Therefore, the ideal PCA should consist of a multimodal approach using a variety of drugs with different mechanisms of action that act synergistically, thereby reducing the opioid use and the incidence of side effects. Dexmedetomidine is a highly selective α 2-adrenergic receptor agonist with sedative and analgesic properties that has been associated with a relatively low incidence of adverse effects [8]. It has been widely used in intraoperative sedation, as well as in intravenous PCA as an adjuvant drug. However, there is still a paucity of evidence regarding the effect of dexmedetomidine in combination with sufentanil on postoperative outcomes for patients undergoing laparoscopic nephrectomy. This prospective, randomized, double-blinded, controlled trial was designed to investigate whether a continuous application of dexmedetomidine and sufentanil for laparoscopic nephrectomy during perioperative period can decrease postoperative sufentanil consumption and pain intensity, as well as to validate the effectiveness and safety profile of this combined therapy. Methods Participants This prospective, randomized, double-blinded, controlled trial was approved by the Institutional Medical Ethics Committee of Qilu Hospital of Shandong University on December 23, 2015 as it was in accordance with the current guidelines of the institution. This study was also registered at chictr.org (ChiCTR-IPR-15007628). Informed consent was obtained from all of the participants. We recruited patients who underwent laparoscopic nephrectomy under general anesthesia at Qilu Hospital from December 2015 to June 2016. This study was supported by the National Natural Science Foundation of China NSFC Grant No. 81702603, and the Key Project of the Natural Science Foundation of Shandong Province Grant No. ZR2014HZ004. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Patients were selected to participate on the study based on the following inclusion criteria: age between 30 and 70 years; American Society of Anesthesiologists (ASA) physical status classification of I or II; intubated with an L-double-lumen endobronchial tube; received 48 h of intravenous PCA after surgery that was used with sufficient competence; agreed to cooperate and signed the informed consent. Patients were excluded if they met the following criteria: body mass index (BMI) greater than 32 kg/m 2 ; basal heart rate (HR) less than 55 beats/minute; history of ischemic heart disease within 6 months prior to this surgical procedure; second-degree or third-degree atrioventricular block; liver and kidney disfunction; history of chronic pain and long-term use of analgesic drugs (> 3 months); hypersensitivity to any of the test drugs; change in surgical approach was required (from laparoscopic to open surgery); failed to complete the data collection. Randomization and masking After obtaining the informed consent, patients were allocated into three equal groups by an independent anesthetist who was blinded to the study, using a computer-generated randomization table. On the day of

Song et al. BMC Anesthesiology (2018) 18:145 Page 3 of 10 the surgery, this anesthetist prepared the intraoperative and postoperative intravenous PCA agents. During the postoperative period, the following outcomes were assessed: pain intensity using a numerical rating scale (NRS), both at rest and with movement; cumulative amount of self-administered sufentanil; level of sedation; concerning adverse effects such as nausea, vomiting, severe abdominal pain, abdominal distention, shivering, delirium, and serious respiratory depression. Anesthesia All patients received atropine 0.5 mg by intramuscular injection in the ward, 30 min before entering the operating room (OR). After arriving to the OR, patients were randomly allocated into three groups by the anesthetist and a venous access was established on patients right upper limb by a nurse. All patients were continuously monitored using a 5-lead electrocardiogram, noninvasive blood pressure cuff, pulse oximetry saturation (SpO 2 ), and end-tidal carbon dioxide (EtCO 2 ) by an automated system (Philips IntelliVue MP50; Philips Company, Beijing, China). If these indicators appeared to be within the test standard, then a continuous infusion of the test drug would be immediately administered to the patient for a period of 10 min (groups D1 and D2 with 0.5 μg/kg DEX, group C with 0.9% NS). Propofol, sufentanil and rocuronium were given for anesthetic induction. After laryngeal mask airway (LMA) intubation, an arterial cannula was placed in the left radial artery. Anesthesia was maintained with sevoflurane (end-tidal concentration 1 2%), dexmedetomidine (0.9% NS in group C, 0.2 μg/kg/h in group D1 and 0.4 μg/kg/h in group D2), and rocuronium. The latter was given to provide a satisfactory level of muscle relaxation. Dexmedetomidine was stopped approximately 1 h prior to the end of surgery. If a fluctuation of more than 20% of the baseline level was evidenced in the mean arterial pressure (MAP), vasoactive drugs (serotonin 0.1 0.2 mg or nitroglycerin 50 100 μg) were used to maintain patients hemodynamically stable. In addition, if HR decreased to less than 50 beats/minute, atropine 0.5 mg was used. On the other hand, if HR increased to more than 100 beats/ minute, esmolol 0.5 mg/kg was given to keep the patient stable. The EtCO 2 was maintained at 28 to 38 mmhg. Thirty minutes before the end of surgery, palonosetron 0.25 mg was given to prevent post-operative nausea and vomiting (PONV). Neostigmine 0.04 mg/kg and atropine 0.02 mg/kg were administrated to revert neuromuscular blockade at the end of surgery. After endotracheal extubation, all patients were sent to the ward. Postoperative analgesia management Before the surgery, all patients were instructed about the NRS, which ranged from 0 (no pain) to 10 (worst possible pain), as well as the how to use the intravenous PCA pump. The PCA was programmed to deliver a bolus dose of 0.5 ml followed by an infusion of 2 ml/h and a lockout time of 10 min. At the end of surgery, the PCA pump was started (Group C with sufentanil 0.02 μg/kg/h; group D1 with both sufentanil and DEX 0.02 μg/kg/h; group D2 with sufentanil 0.02 μg/kg/h and DEX 0.04 μg/kg/h). The goal of using the PCA was to maintain the NRS score less than 3 (at rest) in the first 48 h after surgery. If the NRS was greater than 3 (at rest), patients were given an additional bolus of sufentanil. If patients still reported pain or the NRS score was greater than 6 (at rest), supplemental rescue boluses of intravenous flurbiprofen axetil injection of 50 mg were administered. If the rescue analgesia was ineffective 30 min after administration, intravenous injection of tramadol (100 mg) was given. Outcome measures The primary outcome measure was the cumulative amount of self-administered sufentanil. Furthermore, the secondary outcome measure were: pain intensity using NRS, both at rest and with movement; level of sedation (5-point scale: 0, patient is fully awake; 1, patient is drowsy/closed eyes; 2, patient is asleep/easily aroused with light tactile stimulation or a simple verbal command; 3, patient is asleep/arousable only by strong physical stimulation; and 4, patient is unarousable) [9], concerning adverse effects, the first bowel movement, and duration of postoperative hospital stay. HR, MAP, and SpO 2 were recorded at the following time points: 5 min after entering the OR (T0), 5 min after induction of anesthesia (T1), 5 min after establishment of pneumoperitoneum (T2), 1 h after establishment of pneumoperitoneum (T3), 5 min after stopping the sevoflurane (T4) and 5 min after extubation (T5). EtCO 2 was recorded from T1 to T4. The cumulative amount of self-administered sufentanil and pain intensity were recorded at 1, 2, 4, 8, 16, 24 and 48 h after surgery; level of sedation was recorded at the time of tracheal extubation, 1, 2 and 4 h after surgery; The total number of rescue analgesia and the postoperative adverse effects (nausea and vomiting, severe abdominal pain, abdominal distention, drowsiness, shivering, delirium, and serious respiratory depression) were also recorded at the end of the study. Statistical analysis Statistical analysis was performed with SPSS for Windows Version 21.0 (SPSS Inc. Chicago, IL). We used the Kolmogorov - Smirnov test to assess distribution of the variables. Homogeneity of variance was compared among the three groups by Levene tests. Normally distributed data were expressed as mean standard deviation, skewed data distribution was expressed using median, and categorical data were expressed as frequency (n) and percentage (%). Parameters like age,

Song et al. BMC Anesthesiology (2018) 18:145 Page 4 of 10 weight, operation time, anesthesia time, bowel movement recovery time, dosage of sufentanil and MAP, HR, EtCO 2, SpO 2 at different time points among the groups were compared using 2-way analysis of variance (ANOVA). Multiple comparisons were performed using the LSD post hoc test. NRS and level of sedation were compared among the three groups with Mann-Whitney test, and categorical variables were analyzed using x 2 test. All data with Probability (P) values < 0.05 were considered statistically significant. Results A total of 112 patients undergoing laparoscopic nephrectomy were screened between December 2015 and June 2016, of which 17 were excluded because they either did not meet the inclusion criteria or refused to participate in the trial. The remaining 95 patients were randomly allocated into three groups. Two patients were eliminated due to conversions to open nephrectomy (one from group D1 and one from group D2), and one patient cancelled the surgery (from group D2). Another two patients were excluded after surgery due to incomplete clinical data (one from group C, one from group D1). (Fig. 1). Demographic data and surgery/anesthesia related information Baseline characteristics and demographics of patients were compared among the three groups (Table 1). No statistically significant differences were found in sex, age, height, body weight, BMI and ASA grade (P > 0.05; Table 1). Hypertension was the most frequent type of basic disease as it affected 36.67%, 36.67%, 43.33% of group C, D1 and D2, respectively; there were no statistically significant differences in type of underlying disease and surgeries. The intraoperative data and recovery time among the three groups appeared to have no statistically significant differences (P > 0.05; Table 1). Changes in MAP, HR, EtCO 2 and SpO 2 among the three groups showed no statistical significance (P > 0.05; Fig. 2). Postoperative outcome The total consumption of sufentanil was lower in group D1 and D2 compared with group C at 1, 2, 4 and 8 h after surgery (P < 0.05; Table 2), whereas the total dosage of sufentanil had no statistically significant differences between group D1 and D2 at any time point after surgery (P > 0.05; Table 2). In addition, patients that needed additional self-administered sufentanil within 48 h after surgery in group D1 (10 cases) and group D2 (11 cases) were significantly lower than group C (20 cases) (P < 0.05 Table 2). The NRS score at rest during the first 8 h after surgery was significantly lower in group D1 compared with group C (P = 0.012). In addition, the NRS scores with movement at 1, 2, 4, 8, 24 and 48 h after surgery were significantly lower in both group D1 and D2 (P < 0.05; Fig. 3). The NRS scores both at rest and with Fig. 1 Patient enrolment flow diagram. This illustrates the flow of all patients screened, excluded, and randomized

Song et al. BMC Anesthesiology (2018) 18:145 Page 5 of 10 Table 1 Clinical Characteristics of Patients in Group C, D1 and D2 Group O Group D1 Group D2 P value Sex, F/M,n 10(33.33%)/20(66.67%) 5(16.67%)/25(83.33%) 8(26.67%)/22(73.33%) 0.330 Age, years 51.50 ± 12.06 50.27 ± 10.06 50.94 ± 9.91 0.890 BMI, kg/m 2 26.24 ± 2.84 26.09 ± 1.46 25.83 ± 2.23 0.785 basic disease(hypertension /DM/CHD), n(%) 7(23.33%)/3(10.00%)/1 (3.33%) 9(30.00%)/2(6.67%)/ 0(0%) 11(36.67%)/2(6.67%)/0(0%) 0.840 Type of surgery (radical/ partial), n(%) 14(46.67%)/16 (53.33%) 16(53.33%)/14(46.67%) 14(46.67%)/16(53.33%) 0.850 ASA I/II, n(%) 4(13.33%)/26(86.67%) 2(6.67%)/28(93.33%) 1(3.33%)/29(96.67%) 0.338 Duration of anaesthesia, min 152.50 ± 63.63 149.67 ± 53.38 143.67 ± 57.63 0.836 Duration of surgery, min 134.00 ± 58.63 136.33 ± 53.22 128.83 ± 57.11 0.870 Duration of analgesia, min 20.67 ± 12.98 15.50 ± 5.78 17.90 ± 6.32 0.089 2%propofol, ml 14.43 ± 1.85 14.73 ± 1.25 14.13 ± 1.36 0.311 Rocuronium, mg/h 35.73 ± 6.55 36.26 ± 6.37 37.52 ± 6.55 0.552 Sufentanil,μg 34.50 ± 10.45 30.67 ± 1.72 31.50 ± 3.97 0.063 Number of using vasoactive agent, n (%) 17 (56.67%) 14 (46.67%) 13 (43.33%) 0.219 Postoperative stay in hospital, d 9.40 ± 1.10 8.90 ± 0.85 9.20 ± 0.83 0.243 Variables presented as mean SD or number of patients n (%). None showed any statistical significance (P > 0.05) ASA American Society of Anesthesiologists, BMI body mass index, CHD coronary heart disease, DM diabetes mellitus, SD standard deviation Fig. 2 Changes in MAP, HR, EtCO 2 and SpO 2 among the three groups. Continuous variables presented as mean standard deviation. None showed any statistical significance (P > 0.05). (T0, 5 min after entering the OR; T1, 5 min after induction of anesthesia; T2, 5 min after establishment of pneumoperitoneum; T3, 1 h after establishment of pneumoperitoneum; T4, 5 min after stop sevoflurane; T5, 5 min after extubation), HR = heart rate, MAP = mean arterial pressure, SpO 2 = pulse oxygen saturation, EtCO 2 = end-tidal carbon dioxide partial pressure, OR = operating room

Song et al. BMC Anesthesiology (2018) 18:145 Page 6 of 10 Table 2 Total Dosage of Sufentanil in Group C, D1 and D2 Time point Group C Group D1 Group D2 P values(p c and D1,P c and D2, P D1 and D2 ) Concentration of sufentanil, μg/ml 0.77 ± 0.11 0.76 ± 0.06 0.72 ± 0.06 0.163(0.552/ 0.063/ 0.200) Dosage of sufentanil, ml 1 h 2.28 ± 0.41 2.00 ± 0.00 2.05 ± 0.15 0.003(0.001**/0.007**/0.531) 2 h 4.27 ± 0.41 4.05 ± 0.15 4.12 ± 0.29 0.013(0.011*/0.011*/1.000) 4 h 8.40 ± 0.50 8.10 ± 0.30 8.13 ± 0.22 0.020(0.012*/0.020*/0.829) 8 h 16.48 ± 0.57 16.16 ± 0.46 16.17 ± 0.34 0.058(0.032*/0.047*/0.866) 24 h 48.55 ± 0.69 48.25 ± 0.77 48.17 ± 0.34 0.143(0.135/ 0.063/ 0.706) 48 h 96.55 ± 0.69 96.30 ± 0.92 96.17 ± 0.35 0.227(0.258/ 0.092/ 0.570) Needed additional self- administered sufentanil, n(%) 48 h 20(66.67%) 10(33.33%) 11(36.67%) 0.051(0.030*/0.039*/0.930) Needed rescue analgesia, n(%) 48 h 0 (0%) 0 (0%) 0 (0%) 1.000 Variables presented as mean SD or number of patients n (%). The total consumption of sufentanil were significantly lower in group D1 and D2 than group C at 1, 2, 4 and 8 h after surgery. Patients that needed additional self-administered sufentanil within 48 h after surgery in group D1 and group D2 were significantly lower than in group C. (* meant P < 0.05 compared with Group C, **meant P < 0.01 compared with Group C) No patients required rescue analgesia in all the three groups 48 h after surgery Fig. 3 Pain score (NRS) during 48 h after surgery in group C, D1, and D2. Variables presented as mean standard deviation. The NRS score at rest during the first 8 h after surgery was significantly lower in group D1 compared with group C (P = 0.012). The NRS scores with movement at 1, 2, 4, 8, 24 and 48 h after surgery were significantly lower compared group D1 with group C (P =<0.001,<0.001,0.010,0.032,0.024,0.006,respectively).Atthesame time, compared with group C, group D2 also show significantly lower NRS scores with movement at 1, 2, 4, 8, 24 and 48 h (P = < 0.001, 0.002, 0.005, 0.027, 0.001, 0.028, respectively). (*meant P < 0.05 compared group D1 with group C; # meant P < 0.05 compared group D2 with group C). NRS = numerical rating scale

Song et al. BMC Anesthesiology (2018) 18:145 Page 7 of 10 movement showed no statistically significant differences between group D1 and D2 at each time point after surgery (P > 0.05; Fig. 3). No patients required rescue analgesia in any of the three groups, 48 h after surgery (Table 2). Level of sedation was monitored within the first 4 h post-surgery. The findings were summarized in Table 3. Between the three groups, there were no statistically significant differences among the patients at any time point after surgery (P >0.05; Table 3). Patients in group D2 had more drowsiness than patients in group D1 and C (P < 0.05; Table 4). Other adverse effects had no statistically significant differences among the three groups (P > 0.05; Table 4). The first flatus and defecation time after surgery of patients in group D1 and D2 were significantly shorter than group C (P < 0.05, Fig. 4). Moreover, the difference between D1 and D2 in terms of postoperative bowel movement had no statistical significance (P > 0.05, Fig. 4). Discussion This prospective, randomized, double-blinded, controlled trial showed that using dexmedetomidine combined with sufentanil in patients could decrease the total dosage of sufentanil and improve postoperative analgesia during the first 8 and 48 h after surgery, respectively. In addition, the first flatus and defecation time after surgery in patients of group D1 and D2 were evidently shorter than group C. Patients that were administered a high dose of dexmedetomidine (0.04 μg/kg/h) experienced more drowsiness compared to the participants in the other two groups. The beneficial effects of dexmedetomidine for postoperative analgesia have been reported in many reviews [10 12]. In some clinical studies, the combination of dexmedetomidine and opioids for postoperative intravenous PCA could significantly reduce the consumption of opioids in more than 24 h after surgery [3, 13, 14]. In contrast to those reports, in our study, using dexmedetomidine demonstrated to lessen the required dosage of sufentanil, but the degree of reduction was not clear compared with the total uses of sufentanil. The pain scores (NRS) at rest had no statistically significant differences except during the first 8 h after surgery (NRS at 8 h after surgery in group D1 was evidently lower than in group C), while the pain scores (NRS) with movement in group D1 and D2 were clearly lower than in group C at any time point within 48 h after surgery. The reasons to explain these differences may be complex. Firstly, opioid and α 2-adrenergic receptor agonists are both potent analgesic drugs that can work synergistically when co-administered. Some studies have also shown that the combination of α 2-adrenergic receptor agonists and opioid agonists in sub-analgesic dose can also achieve an effective analgesic effect [15, 16]. This synergistic interaction takes effect in the spinal cord due to the presence of the receptors that mediate these interactions and cause downstream signaling events that ultimately enable the synergy to occur [17]. Secondly, compared with traditional open nephrectomy, laparoscopic nephrectomy has benefits of a smaller incision and less postoperative pain [18, 19]. In our study, the administration of sufentanil 0.2 μg/kg/h was sufficient to satisfy the needs of postoperative analgesia in patients undergoing laparoscopic nephrectomy; only a few patients required additional self-administered doses of sufentanil. The level of pain in patients at rest demonstrated no statistically significant different among the three groups. Therefore, infusion of dexmedetomidine could only slightly decrease the amount of sufentanil. In the meantime, the pain scores (NRS) in the three groups showed no statistically significant differences. Further research is necessary to explain the reasons for the differences observed with regards to the effect of dexmedetomidine on pain between rest and movement. This pilot study showed that dexmedetomidine could increase the incidence of drowsiness that is easily arousable and does not cause severe respiratory depression; this is consistent with its sedative effect [8, 20]. According to Song et al. [21], dexmedetomidine added to an opioid-based analgesic regimen could prevent PONV in highly susceptible patients. However, in our study, there appeared to be no differences in the occurrence of PONV between the three groups. This was mainly prevented by an adequate bowel preparation and antiemetic drug application during the perioperative period. Additionally, laparoscopic nephrectomy had less stimulation of gastrointestinal tract compared to an open nephrectomy. All of these contributed to a low incidence of PONV among the three groups. Talke Table 3 Level of sedation in Group C, D1 and D2 Time point Group O Group D1 Group D2 P values(p c and D1,P c and D2, P D1 and D2 ) level of sedation (0/1/2/3), n extubation 2 (1 3) 2 (1 2) 2(1 2) 0.542/0.737/ 0.755 1h 1(0 2) 1 (0 1) 1 (0 2) 0.060/ 0.630/ 0.007 2h 0(0 2) 0 (0 1) 1 (0 2) 0.450/ 0.771/ 0.287 4h 0(0 1) 0 (0 1) 0 (0 1) 0.080/ 0.218/ 0.553 Variables presented as median (interquartile range) or number of patients, n

Song et al. BMC Anesthesiology (2018) 18:145 Page 8 of 10 Table 4 Adverse reactions after Surgery in Group C, D1 and D2 Group O Group D1 Group D2 Nausea and Vomiting 7 (43.33%) 5(30%) 5 (26.67%) 0.495 Severe abdominal pain and distention 4 (13.33%) 5 (16.67%) 4 (20%) 0.787 Drowsiness 1 (0%) 2 (6.67%) 7 (23.33%) 0.031 * Delirium 0 (0%) 0 (0%) 0 (0%) 1.000 Shivering 1 (3.33%) 1 (6.67%) 0 (0%) 0.537 serious respiratory depression 0 (0%) 0 (0%) 0 (0%) 1.000 Variables presented as number of patients, n (%) *meant P < 0.05 P values et al. [22] reported that bradycardia and hypotension are the most common side effects of dexmedetomidine. However, in our study, no patients were found to have bradycardia or hypotension. It may have been primarily due to the criteria of patient enrollment that was adopted in our study (patients with a history of ischemic heart disease 6 months prior to this event or history of second-degree or third-degree atrioventricular blocks were excluded). In addition, age, PCA settings, monitoring protocol and type of surgery could have also influenced the results. In this prospective study, we also found that the combination of dexmedetomidine with opioids improved the recovery of bowel movement after surgery which is in accordance with the previous report by Jin et al. [23]. Sympathetic activation has been demonstrated that it is related to the occurrence and development of postoperative ileus [24 26]. Dexmedetomidine can inhibit the excessive activation of the sympathetic nervous system [27], therefore causing a decreased inhibitory effect in the gastrointestinal tract. In addition, the inhibitory effect of sympathetic nervous system can decrease acetylcholine release, which is crucial for the activation of the enteric nervous system. Opioids such as sufentanil can induced bowel dysfunction [28], while the use of dexmedetomidine reduces the dosage of sufentanil used after surgery, thereby reducing the inhibition caused by it. Although some studies have proved that dexmedetomidine can decrease the incidence of postoperative delirium [29, 30], there is no sufficient evidence in our trial to support this argument. However, delirium usually occurs in elderly patients or patients with prolonged-operative time, while in our study, all surgical procedures had a short operative time, therefore decreasing the likelihood of finding delirium in our patients. In addition, the incidence of other adverse effects can also be attributed to a similar reason. Some limitations exist in our study. First of all, instead of monitoring the depth of anesthesia by BIS value, we maintained the depth of anesthesia by adjusting the sevoflurane end-tidal concentration, but it could not accurately reflect the depth of anesthesia. Secondly, dexmedetomidine was administered at a rate of 0.5 μg/kg for 10 minutes before the induction of anesthesia and then at a rate of 0.2 to 0.4 μg/kg/h during the operation. We did not measure the serum concentration of dexmedetomidine at any time point, so we could not determine the effects of the plasma concentrations of dexmedetomidine on intraoperative hemodynamics. Finally, laparoscopic nephrectomy has two different surgical approaches, intra-abdominal and retroperitoneal. Therefore, different surgical techniques could have influenced the postoperative Fig. 4 Bowel movement recovery after surgery among the three groups. Variables presented as mean standard deviation.* meant P < 0.05 compared group D1 with group C; # meant P < 0.05 compared group D2 with group C

Song et al. BMC Anesthesiology (2018) 18:145 Page 9 of 10 analgesia and bowel movement recovery, but we did not take this into account. To further investigate this effect on postoperative outcomes, more studies are required. Conclusions In conclusion, we found that combination of sufentanil and dexmedetomidine (0.02 0.04 μg/kg/h) was associated with better analgesic effect, and faster recovery of bowel movement without additional severe adverse effects in patients after laparoscopic nephrectomy compared with sufentanil alone. To decrease the incidence of drowsiness, we suggested using low dose of dexmedetomidine (0.02 μg/kg/h) instead of high dose of dexmedetomidine (0.04 μg/kg/h). More studies are still required to determine the optimal dose of dexmedetomidine for improving postoperative analgesia in patients undergoing other types of surgeries. Abbreviations ASA: American Society of Anesthesiologists; BMI: Body mass index; CHD: Coronary heart disease; DM: Diabetes mellitus; EtCO 2 : End-tidal carbon dioxide partial pressure; HR: Heart rate; LMA: Laryngeal mask airway; MAP: Mean arterial pressure; NRS: Numerical rating scale; NS: Normal saline; OR: Operating room; PCA: Patient-controlled analgesia; PONV: Postoperative nausea and vomiting; SpO 2 : Pulse oxygen saturation Funding This work was supported by the National Natural Science Foundation of China NSFC Grant No. 81702603, and the Key Project of the Natural Science Foundation of Shandong Province Grant No. ZR2014HZ004, Medical and Health Technology Development Program of Shandong Province Grant No. 2015WS0309. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Availability of data and materials The datasets generated and analysed during the current study are available from the corresponding author on reasonable request. Authors contributions FS helped collect, analyze, and interpret the data, and write the manuscript. CY helped analyze the data. FQ helped design the study, analyze the data, and conduct the study. PZ helped collect the data. XW helped collect the data and write the manuscript. AFE helped revise the manuscript. YL helped collect the data. CZ helped design the study, interpret the data, and revise the manuscript. LL helped design the study, conduct the study, and revise the manuscript. All authors read and approved the final manuscript. Ethics approval and consent to participate This study was approved by the Institutional Medical Ethics Committee of Qilu Hospital of Shandong University on December 23, 2015 as it was in accordance with the current guidelines of the institution. This study was also registered at chictr.org (ChiCTR-IPR-15007628). Written informed consent was obtained from all of the participants. Consent for publication Not applicable. Competing interests The authors declare that they have no competing interests. Publisher s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Author details 1 Department of Anaesthesiology, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan 250033, China. 2 Department of Breast Surgery, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan 250033, China. 3 Department of Anaesthesiology, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan 250012, China. 4 The Institute for Translational Medicine, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan 250033, China. 5 Department of Anoproctology, the Second Hospital of Shandong University, No. 247 Beiyuan Street, Jinan 250033, China. 6 Translational Research Program, University of Toronto, 27 King s College Circle, Toronto, ON M5S 1A1, Canada. Received: 26 June 2018 Accepted: 5 October 2018 References 1. Perkins FM, Kehlet H. Chronic pain as an outcome of surgery. A review of predictive factors. Anesthesiology. 2000;93(4):1123 33. 2. Callesen T, Bech K, Kehlet H. Prospective study of chronic pain after groin hernia repair. Br J Surg. 1999;86(12):1528 31. 3. Nie Y, Liu Y, Luo Q, Huang S. Effect of dexmedetomidine combined with sufentanil for post-caesarean section intravenous analgesia: a randomised, placebo-controlled study. Eur J Anaesthesiol. 2014;31(4):197 203. 4. Momeni M, Crucitti M, De Kock M. Patient-controlled analgesia in the management of postoperative pain. Drugs. 2006;66(18):2321 37. 5. Scott JC, Cooke JE, Stanski DR. Electroencephalographic quantitation of opioid effect: comparative pharmacodynamics of fentanyl and sufentanil. Anesthesiology. 1991;74(1):34 42. 6. Gadsden J, Hart S, Santos AC. Post-cesarean delivery analgesia. Anesth Analg. 2005;101(5 Suppl):S62 9. 7. Watcha MF, White PF. Postoperative nausea and vomiting. Its etiology, treatment, and prevention. Anesthesiology. 1992;77(1):162 84. 8. Ebert TJ, Hall JE, Barney JA, Uhrich TD, Colinco MD. The effects of increasing plasma concentrations of dexmedetomidine in humans. Anesthesiology. 2000;93(2):382 94. 9. Lin TF, Yeh YC, Lin FS, Wang YP, Lin CJ, Sun WZ, Fan SZ. Effect of combining dexmedetomidine and morphine for intravenous patientcontrolled analgesia. Br J Anaesth. 2009;102(1):117 22. 10. Chan AK, Cheung CW, Chong YK. Alpha-2 agonists in acute pain management. Expert Opin Pharmacother. 2010;11(17):2849 68. 11. Abdel Hamid MHE. Intravenous Dexmedetomidine infusion compared with that of fentanyl in patients undergoing arthroscopic shoulder surgery under general anesthesia. Anesth Essays Res. 2017;11(4):1070 4. 12. Sharma R, Gupta R, Choudhary R, Singh Bajwa SJ. Postoperative analgesia with intravenous paracetamol and Dexmedetomidine in laparoscopic cholecystectomy surgeries: a prospective randomized comparative study. Int J Appl Basic Med Res. 2017;7(4):218 22. 13. Ramsay MA, Newman KB, Leeper B, Hamman BL, Hebeler RF Jr, Henry AC, Kourlis H Jr, Wood RE, Stecher JA, Hein HA. Dexmedetomidine infusion for analgesia up to 48 hours after lung surgery performed by lateral thoracotomy. Proc (Baylor Univ Med Cent). 2014;27(1):3 10. 14. Peng K, Liu HY, Wu SR, Cheng H, Ji FH. Effects of combining Dexmedetomidine and opioids for postoperative intravenous patientcontrolled analgesia: a systematic review and meta-analysis. Clin J Pain. 2015;31(12):1097 104. 15. Drasner K, Fields HL. Synergy between the antinociceptive effects of intrathecal clonidine and systemic morphine in the rat. Pain. 1988;32(3): 309 12. 16. Plummer JL, Cmielewski PL, Gourlay GK, Owen H, Cousins MJ. Antinociceptive and motor effects of intrathecal morphine combined with intrathecal clonidine, noradrenaline, carbachol or midazolam in rats. Pain. 1992;49(1):145 52. 17. Chabot-Dore AJ, Schuster DJ, Stone LS, Wilcox GL. Analgesic synergy between opioid and alpha2 -adrenoceptors. Br J Pharmacol. 2015;172(2): 388 402. 18. Fonouni H, Mehrabi A, Golriz M, Zeier M, Muller-Stich BP, Schemmer P, Werner J. Comparison of the laparoscopic versus open live donor nephrectomy: an overview of surgical complications and outcome. Langenbeck s Arch Surg. 2014;399(5):543 51.

Song et al. BMC Anesthesiology (2018) 18:145 Page 10 of 10 19. Zhao PT, Richstone L, Kavoussi LR. Laparoscopic partial nephrectomy. Int J Surg. 2016;36(Pt C):548 53. 20. Szumita PM, Baroletti SA, Anger KE, Wechsler ME. Sedation and analgesia in the intensive care unit: evaluating the role of dexmedetomidine. Am J Health Syst Pharm. 2007;64(1):37 44. 21. Song Y, Shim JK, Song JW, Kim EK, Kwak YL. Dexmedetomidine added to an opioid-based analgesic regimen for the prevention of postoperative nausea and vomiting in highly susceptible patients: a randomised controlled trial. Eur J Anaesthesiol. 2016;33(2):75 83. 22. Talke P, Li J, Jain U, Leung J, Drasner K, Hollenberg M, Mangano DT. Effects of perioperative dexmedetomidine infusion in patients undergoing vascular surgery. The study of perioperative ischemia research group. Anesthesiology. 1995;82(3):620 33. 23. Cho JS, Kim HI, Lee KY, An JY, Bai SJ, Cho JY, Yoo YC. Effect of intraoperative Dexmedetomidine infusion on postoperative bowel movements in patients undergoing laparoscopic gastrectomy: a prospective, randomized, placebocontrolled study. Medicine. 2015;94(24):e959. 24. Dubois A, Weise VK, Kopin IJ. Postoperative ileus in the rat: physiopathology, etiology and treatment. Ann Surg. 1973;178(6):781 6. 25. Nilsson F, Jung B. Gastric evacuation and small bowel propulsion after laparotomy. A study with a double isotope technique in rat. Acta Chir Scand. 1973;139(8):724 30. 26. Luckey A, Livingston E, Tache Y. Mechanisms and treatment of postoperative ileus. Arch Surg. 2003;138(2):206 14. 27. Carollo DS, Nossaman BD, Ramadhyani U. Dexmedetomidine: a review of clinical applications. Curr Opin Anaesthesiol. 2008;21(4):457 61. 28. Brock C, Olesen SS, Olesen AE, Frokjaer JB, Andresen T, Drewes AM. Opioidinduced bowel dysfunction: pathophysiology and management. Drugs. 2012;72(14):1847 65. 29. Su X, Meng ZT, Wu XH, Cui F, Li HL, Wang DX, Zhu X, Zhu SN, Maze M, Ma D. Dexmedetomidine for prevention of delirium in elderly patients after non-cardiac surgery: a randomised, double-blind, placebo-controlled trial. Lancet (London, England). 2016;388(10054):1893 902. 30. Riker RR, Shehabi Y, Bokesch PM, Ceraso D, Wisemandle W, Koura F, Whitten P, Margolis BD, Byrne DW, Ely EW, et al. Dexmedetomidine vs midazolam for sedation of critically ill patients: a randomized trial. JAMA. 2009;301(5):489 99.