Next Article in Journal
SARS-CoV-2 Detection for Diagnosis Purposes in the Setting of a Molecular Biology Research Lab
Previous Article in Journal
Transfection of hPSC-Cardiomyocytes Using Viafect™ Transfection Reagent
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Protocol

Comparison of Opioid-Free Anesthesia Versus Opioid-Containing Anesthesia for Elective Laparoscopic Surgery (COFA: LAP): A Protocol Measuring Recovery Outcomes

by
Anthony Eidan
1,2,
Angela Ratsch
3,4,*,
Elizabeth A. Burmeister
3,4 and
Geraldine Griffiths
3,4
1
Anesthetic Department, Bundaberg Hospital, Wide Bay Hospital and Health Service, Bundaberg 4670, Australia
2
School of Medicine, The University of Queensland, Brisbane 4072, Australia
3
School of Nursing, Midwifery and Social Work, The University of Queensland, Brisbane 4072, Australia
4
Nursing and Midwifery Services, Wide Bay Hospital and Health Service, Bundaberg 4670, Australia
*
Author to whom correspondence should be addressed.
Methods Protoc. 2020, 3(3), 58; https://doi.org/10.3390/mps3030058
Submission received: 1 July 2020 / Revised: 12 August 2020 / Accepted: 12 August 2020 / Published: 13 August 2020

Abstract

:
The administration of opioids is a central element in contemporary anesthetic techniques in Australia; however, opioids have a range of side effects. As an alternative, opioid-free anesthesia (OFA) is an emerging mode of anesthesia intended to avoid these side effects. This study is the first to publish the use of OFA in Australia and is conducted in a regional Queensland Health Service. The design will utilize a randomized clinical trial (RCT) to investigate the impact of OFA for patients having an elective laparoscopic cholecystectomy (n = 40) or tubal ligation (n = 40). Participant outcomes to be measured include: Quality of Recovery (QoR-15); Oral Morphine Equivalent Daily Dose (OMEDD) at 24-h post-operatively; time to first opioid (TTFO) dose; post-operative nausea and vomiting (PONV); Post Anesthetic Care Unit length of stay (PACU-LOS); and hospital length of stay (LOS). The findings may challenge the essentiality of opioids in the peri-operative period, which in turn would influence the future intra-operative management of surgical patients. Ultimately, a reduction in anesthesia-associated opioid use will support a more general decline in opioid use.

1. Introduction

Historical records indicate that humans have used opioids for at least 8000 thousand years as a mood enhancer, analgesic and hypnotic agent [1]. Evidence on Mesopotamian cuneiform tablets (c. 6000 BCE) refers to the medical properties of opium, and later, the Ebers papyrus (c. 1500 BCE) contains a record of the dried milky fluid from the poppy plant (opium) being used for headaches and as anesthesia [2]. The active ingredient in opium (morphine) was isolated in 1803, and its derivative, diacetaylmophine (heroin) in 1874. Opioids and their synthetic derivatives (Fentanyl) were first used in anesthesia in 1962 in Belgium and are now routinely administered via the oral, subcutaneous, intravenous, intramuscular, transdermal, epidural or via intrathecal routes specifically for their analgesic effects. Across the world, there has been a significant rise in the consumption of pharmaceutical opioids over the last twenty years. Australia now has one of the highest levels of use with almost 15 million opioid prescriptions dispensed in 2015 and with the use of high-potency opioids also increasing [3,4,5].
Traditionally in Australia, peri-operative analgesia has been provided by opioid analgesics, and current anesthetic practice is heavily dependent on opioid use during and after anesthesia for post-operative pain relief. However as White [6] indicates, ambulatory surgery involving high doses of opioids can be associated with increased post-operative complications including respiratory depression, paralytic ileus, nausea and vomiting, difficulty voiding, and pruritus. These can ironically increase length of stay, thereby reversing the objective of ambulatory surgery in providing rapid patient through-put. In addition, the intra-operative use of large bolus doses or continuous infusions of potent short-acting opioid analgesics (e.g., remifentanil) can increase post-operative pain due to their rapid elimination and the development of acute tolerance [7]. Accordingly, in the ambulatory peri-operative environment, anesthesiologists have explored the adjuvant use of non-opioid analgesics [8,9].
A non-opioid analgesic multimodal approach (opioid-free anesthesia (OFA)) is aimed at optimizing adjunctive options intra-operatively, utilizing anesthetic techniques targeting different neuroanatomical circuits and multiple neurophysiological mechanisms [10,11]. The pain (nociceptive) pathway commence with the A-delta and C-peripheral afferent nerve fibers. These fibers synapse on projection neurons in the dorsal horn of the spinal cord. The neurons then synapse in the brainstem including in the medulla at the nucleus of the tractus solitarius (NTS) [12,13]. The NTS initiates the autonomic response to nociceptive stimulus and then mediates, through the caudal ventral lateral medulla and the rostral ventral lateral medulla, (with projections to the adrenal medulla and the thoracolumbar sympathetic ganglia, the sympathetic output from the heart and peripheral blood vessels) [10,12,13]. In addition, the NTS synapses into the hypothalamus with the periventricular nucleus and supraoptic nucleus. The nucleus ambiguous synapses with the vagus nerve to the cardia sino-atrial node [12,13,14] to mediate the parasympathetic output. Blocking the autonomic nervous system response to nociceptive stimulus forms the major theoretical foundation of OFA [10,11,12,14].
Contemporaneous anesthetic practice calls for patient-centered approaches to assessing the recovery of patients after surgery. OFA has been shown to be an effective anesthetic technique for patients undergoing laparoscopic cholecystectomy [15] and, in addition, is associated with less peri-operative adverse events in cardiac surgery [16] and less nausea and vomiting in bariatric surgery [17] compared to opioid-containing anesthesia [18]. The measurement of Quality of Recovery (QoR) following anesthesia and surgery is broader than simply assessing pain management. It involves considerations of the patient’s post-operative status in comparison to their pre-operative status and includes return of self-care, household and work activities and mobility to the level pre-operatively. In measuring patient outcomes, there is a clear trend in favor of Patient Reported Outcome Measures (PROMs) i.e., the direct reporting of an individual’s health status by the individual [19]. In this clinical trial, the participant reported outcomes are central to the measurement of OFA therapeutic efficacy.
The research has been approved by the Royal Brisbane and Women’s Hospital Human Research Ethics Committee (HREC/2020/QRBW/62398), Queensland, Australia, and is registered with the Australian New Zealand Clinical Trials Registry (ANZCTR), trial number ACTRN12620000714987.

2. Experimental Design

2.1. Aim

This is an investigator-initiated, single site, prospective, randomized, parallel group, single-blind study, with concealed allocation of participants scheduled for elective laparoscopic tubal ligation or cholecystectomy surgery, randomized on a 1:1 basis to receive either a standard or opioid-free anesthesia protocol. The aim of this study is to compare clinical outcomes for participant’s receiving opioid-free anesthetic (OFA) with those receiving standard opioid-containing anesthesia.

2.2. Objective

The primary objective is to compare the quality of recovery from general anesthesia and surgery using the quality of recovery 15 item scale (QoR-15) [20]. The QoR-15 score (Table 1) is a shorter validated version of the QoR-40 and provides an efficient evaluation of post-operative recovery from the participant’s perspective with total QoR-15 score ranging from 0 (extremely poor recovery) to 150 (excellent recovery) [20]. Permission was obtained to use the scale in this research (P. Myles, personal communication, 11 February 2020). Minor amendments were made to two questions; the first to Question Seven changing the wording from “getting support”, to “able to get support”, and Question Eight changed from “able to return to work and usual home activities” to “able to do usual home activities”.
The secondary objectives and endpoints include comparing:
  • Post-operative complications of medication administration including:
    Respiratory depression;
    Nausea and vomiting;
    Delirium/hallucinations;
    Pruritus;
    Participant pain scores;
    Functional activity scores;
    Sedation level;
    Local anesthetic toxicity.
  • The use of analgesia:
    Time post-operatively to first analgesia;
    Type of analgesia;
    Oral Morphine Equivalent Daily Dose (OMEDD).
  • Time until post-operative mobilization.
  • Length-of-stay in PACU.
  • Hospital length-of-stay.
  • Peri-operative adverse events.

3. Procedure

3.1. Inclusion, Exclusion, Recruitment and Consent

All patients presenting for an elective cholecystectomy or tubal ligation (with or without oophorectomy) will be identified by the research team as per Table 2 and will be recruited and consented according to Table 3. Participation will be until Day 1 post-operatively. No long-term follow-up will be conducted to assess chronic complications.

3.2. Sample Size, Randomization and Blinding

3.2.1. Sample Size

Current literature reports that mean change QoR-15 scores (difference between baseline and Day 1 post-operative) is approximately 22 (on a scale of 0–150) with a standard deviation of four [20]. A difference in QoR-15 scores of eight has been reported as clinically significant [21]. To test (2-sided) the effect of a difference in mean change score of six with a standard deviation of four for OFA compared with non-OFA, with 80% power and alpha level of 0.01, the total sample size (30% compliance adjusted) required is 40 participants (20 required for both the OFA and the non-OFA groups). Allocations to treatment group will be stratified according to whether the procedure is a cholecystectomy or tubal ligation; therefore, the sample size will be doubled to enable combined and separate analyses of treatment groups between types of surgery. Accordingly, the total number of participants required will be 80, i.e., 40 in the OFA group (n = 20 OFA cholecystectomy and n = 20 OFA tubal ligation participants) and 40 in the non-OFA group (n = 20 non-OFA cholecystectomy and n = 20 non-OFA tubal ligation participants).

3.2.2. Randomization

Eligible participants will be randomly selected to receive OFA surgery or non-OFA surgery on a 1:1 basis. Treatment allocations will be randomized into blocks of six and stratified by type of surgery (tubal ligation or cholecystectomy). Following consent, participants will be allocated a study participation identification number (ID). An envelope with corresponding ID will be opened by the anesthetist just prior to surgery. This envelope will contain the random allocation of OFA or standard treatment and the respective treatment protocol for the allocation.

3.2.3. Blinding

Allocation to standard or OFA protocol will be completed prior to surgery as designated by the chief investigator. The participant and the outcome assessors (recovery and ward nurses) will be blinded to the allocation group. The chief investigator will not conduct any outcome assessments. The allocation/randomization arms will be as follows:
Group A: Investigational treatment—opioid-free anesthesia
Group B: Standard treatment—non-opioid-free anesthesia

3.3. Treatment Regimen

3.3.1. Intra-Operative Participant Medication

Induction and maintenance anesthesia will be given according to Table 4 and Table 5. All participants who do not have contraindication to receive intravenous lidocaine will receive Lidocaine as per the guidelines in Table 6.

3.3.2. Post-Operative Participant Assessment

The recovery room staff will be blinded to the participant’s anesthetic grouping. The intraoperative anesthetic charts (AARK) records will be printed without “anesthetic medications/agents administered intraoperatively”. The recovery nurses will receive the AARK print out with all other data, including procedure details, intraoperative monitoring and anesthetic notes. The recovery nurse will receive the Identification, Situation and Status, Background, Assessment and Actions, Recommendations and Responsibilities (ISBAR) handover from the anesthetic doctor/nurse. The recovery room staff will use the standard Post-anesthetic Pain Management Protocol (Supplementary Materials) for all participants. If there are any concerns during the recovery period, the recovery nurse will speak directly with the anesthetic registrar or consultant who delivered the intra-operative protocol for ongoing management for the participant.

3.4. Data Collection

3.4.1. Participant Documentation

Table 7 details the data (and their definitions) to be collected for the following source documents:
  • QoR-15 questionnaire (baseline and 24-h post-operatively).
  • Preoperative anesthetic clinic assessment form.
  • AARK.
  • Post-anesthetic care unit (PACU) documents.
  • National In-patient Medication Charts (NIMC).
  • Queensland Hospital Admitted Patient Data Collection.
  • Intravenous Patient Controlled Analgesia Order—Adult.
  • Analgesia monitoring forms, intravenous, PCA, epidural and regional analgesia.
  • Queensland Adult Deterioration Detection System (Q-ADDS).
  • Clinical notes.

3.4.2. Electronic Data Handling

Data will be collected and transcribed onto an electronic data collection sheet using Google Docs. A cloud-stored electronic database within Google Docs has been purpose built for this research by a research team member. Access is via two-factor authentication and password security. This methodology has been risk assessed by the Queensland Health Cyber Security team as meeting the needs for the storage of sensitive data.

3.5. Measures and Statistics

3.5.1. Measures

Outcomes are measured at baseline, then at the induction of anesthesia and at Day 1 post-operatively. The post-operative assessment will be conducted on the ward or by phone (if participant is already discharged). The primary outcome is the 15-item quality of recovery (QoR-15) score (Table 1). Each question is measured on a Likert scale ranging from 0–10; where 0 = none of the time [poor] to 10 = all of the time [excellent]. Results are considered clinically significant if there is a change of score of eight [21].

3.5.2. Statistical Methods

All participants randomized will be included in the analyses on an intention-to-treat basis. Ten percent of data collected and entered will be independently verified. If more than 0.5% disagreement between data collected/data entry exists, then a further 10% of data will be checked. This process will continue until no disagreement is found by the independent verifier.
Data distribution will be assessed. Descriptive statistics, including frequencies and percentages for categorical data, and means, standard deviations or medians, and ranges for continuous data, will be assessed to ensure randomization has succeeded, and to describe differences between groups.
Mean and total QoR-15 scores will be calculated per participant at baseline and on Day 1. QoR-15 change scores will be calculated to determine the difference between Day 1 and baseline scores for each participant. The effect of OFA versus non-OFA on recovery (QoR-15 score) will be evaluated using a mixed model with random effects (of individual participants) to estimate the effect of OFA on QoR-15 scores, accounting for baseline QoR-15 scores type of surgery and 24-h peri-operative opioid use. If there are no statistically significant random effects in the mixed-method model (tested using likelihood ratio test), univariate and multivariate linear regression models using the QoR-15 change score will be used for group comparisons and to estimate factors associated with outcomes. Interaction effects will be investigated and eliminated where possible through data transformation and the inclusion of interaction terms in the models. Stata 14 (StataCorp, College Station, TX, USA) and SPSS® version 20.0 (IBM® SPSS® Statistics 20) for Windows will be used for all analyses. All analyses will be reported at the 0.05 (95% confidence interval) statistical significance level, unless Bonferroni corrected levels are appropriate.

4. Expected Results

The intent of this research is to consider the hypothesis that opioids are an essential component in the suite of anesthetic medications. In Western contemporary health care practices, the use of opioids during anesthetic is evidenced-based, or is it? The Australian and New Zealand College of Anesthetists is cognizant of the rising prescription and risk of opioids. Their position statement [22] around this matter strongly cautions against slow-release opioid prescription, particularly in the post-operative period. However, the millennia-long legacy of the “need” for opioids intra- and post-operatively appears unchallenged despite (or in spite of) the advanced science that exists around human anatomy and physiology, pharmacokinetics and pharmacodynamics. A growing body of evidence indicates that long-term opioid use begins with treatment of acute pain, including intra-operative and post-operative surgical pain [23,24,25]. Brat, et al. [26] show that among opioid-naïve patients, each refill and week of opioid use post-surgery is associated with a large increase in opioid misuse. This finding is confirmed by Macintyre, et al. [27] who suggest that post-discharge opioid use continues in some patients for some years after surgery.
The evidence provided by this research will contribute to the growing body of knowledge around the efficacy of newer opioid-free anesthetic techniques.

Supplementary Materials

The following is available online at https://www.mdpi.com/2409-9279/3/3/58/s1, Post-operative pain management of all patients.

Author Contributions

Conceptualization, A.E.; methodology, A.E., A.R., E.A.B. and G.G.; writing—original draft preparation, A.E., A.R. and G.G; writing—review and editing, A.E., A.R., E.A.B. and G.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors acknowledge the in-kind support provided by the Wide Bay Hospital and Health Service to the authors in the development of this protocol.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses or interpretation of data; in the writing of the manuscript or in the decision to publish the results.

References

  1. Brooks, J.K. Chemical burn to the gingiva after misuse of an over-the-counter oral whitening mouthwash. Gen. Dent. 2017, 65, 34–36. [Google Scholar]
  2. Whysner, J. The Alchemy of Disease: How Chemicals and Toxins Cause Cancer and Other Illnesses; Columbia University Press: New York, NY, USA, 2020. [Google Scholar]
  3. Berterame, S.; Erthal, J.; Thomas, J.; Fellner, S.; Vosse, B.; Clare, P.; Hao, W.; Johnson, D.T.; Mohar, A.; Pavadia, J.; et al. Use of and barriers to access to opioid analgesics: A worldwide, regional, and national study. Lancet 2016, 387, 1644–1656. [Google Scholar] [CrossRef]
  4. Karanges, E.A.; Blanch, B.; Buckley, N.A.; Pearson, S.A. Twenty-five years of prescription opioid use in Australia: A whole-of-population analysis using pharmaceutical claims. Br. J. Clin. Pharmacol. 2016, 82, 255–267. [Google Scholar] [CrossRef] [Green Version]
  5. Campbell, G.; Lintzeris, N.; Gisev, N.; Larance, B.; Pearson, S.; Degenhardt, L. Regulatory and other responses to the pharmaceutical opioid problem. Med. J. Aust. 2019, 210, 6–8. [Google Scholar] [CrossRef]
  6. White, P.F. What are the advantages of non-opioid analgesic techniques in the management of acute and chronic pain? Expert Opin. Pharmacother. 2017, 18, 329–333. [Google Scholar] [CrossRef]
  7. Guignard, B.; Bossard, A.E.; Coste, C.; Sessler, D.I.; Lebrault, C.; Alfonsi, P.; Fletcher, D.; Chauvin, M. Acute opioid tolerance: Intraoperative remifentanil increases postoperative pain and morphine requirement. Anesthesiology 2000, 93, 409–417. [Google Scholar] [CrossRef]
  8. White, P.F. The role of non-opioid analgesic techniques in the management of pain after ambulatory surgery. Anesth. Analg. 2002, 94, 577–585. [Google Scholar] [CrossRef]
  9. Warren-Stomberg, M.; Brattwall, M.; Jakobsson, J.G. Non-opioid analgesics for pain management following ambulatory surgery: A review. Minerva Anestesiol. 2013, 79, 1077–1087. [Google Scholar]
  10. Gropper, M.A.; Miller, R.D.; Eriksson, L.I.; Fleisher, L.A.; Wiener-Kronish, J.P.; Cohen, N.H.; Leslie, K. (Eds.) Miller’s Anesthesia, 2-Volume Set E-Book, 9th ed.; Elsevier Health Sciences: New York, NY, USA, 2019. [Google Scholar]
  11. McMahon, S.B.; Koltzenburg, M.; Tracey, I.; Turk, D. (Eds.) Wall and Melzack’s Textbook of Pain, 6th ed.; Elsevier Health Sciences: New York, NY, USA, 2013. [Google Scholar]
  12. Matta, B.F.; Menon, D.K.; Turner, J.M. (Eds.) Textbook of Neuroanaesthesia and Critical Care; Cambridge University Press: Greenwich, UK, 2000. [Google Scholar]
  13. Egan, T.D. Are opioids indispensable for general anaesthesia? Br. J. Anaesth. 2019, 122, e127–e135. [Google Scholar] [CrossRef]
  14. Stoelting, R.; Flood, P.; Rathmell, J.; Shafer, S. (Eds.) Stoelting’s Handbook of Pharmacology & Physiology in Anaesthetic Practice, 3rd ed.; Wolters Kluwer: Philadelphia, PA, USA, 2015. [Google Scholar]
  15. Bakan, M.; Umutoglu, T.; Topuz, U.; Uysal, H.; Bayram, M.; Kadioglu, H.; Salihoglu, Z. Opioid-free total intravenous anesthesia with propofol, dexmedetomidine and lidocaine infusions for laparoscopic cholecystectomy: A prospective, randomized, double-blinded study. Braz. J. Anesthesiol. 2015, 65, 191–199. [Google Scholar] [CrossRef] [Green Version]
  16. Guinot, P.G.; Spitz, A.; Berthoud, V.; Ellouze, O.; Missaoui, A.; Constandache, T.; Grosjean, S.; Radhouani, M.; Anciaux, J.B.; Parthiot, J.P.; et al. Effect of opioid-free anaesthesia on post-operative period in cardiac surgery: A retrospective matched case-control study. BMC Anesthesiol. 2019, 19, 136. [Google Scholar] [CrossRef] [Green Version]
  17. Ziemann-Gimmel, P.; Goldfarb, A.A.; Koppman, J.; Marema, R.T. Opioid-free total intravenous anaesthesia reduces postoperative nausea and vomiting in bariatric surgery beyond triple prophylaxis. Br. J. Anaesth. 2014, 112, 906–911. [Google Scholar] [CrossRef] [Green Version]
  18. Frauenknecht, J.; Kirkham, K.R.; Jacot-Guillarmod, A.; Albrecht, E. Analgesic impact of intra-operative opioids vs. opioid-free anaesthesia: A systematic review and meta-analysis. Anaesthesia 2019, 74, 651–662. [Google Scholar] [CrossRef]
  19. Ursulet, E.; Bonnet, C.; Lopez, S.; Lavand’homme, P. Quality of recovery after non opioid versus opioid anesthesia for spinal surgery: A prospective observational study. J. Clin. Anesth. 2020, 61, 109689. [Google Scholar] [CrossRef]
  20. Stark, P.A.; Myles, P.S.; Burke, J.A. Development and psychometric evaluation of a postoperative quality of recovery score: The QoR-15. Anesthesiology 2013, 118, 1332–1340. [Google Scholar] [CrossRef] [Green Version]
  21. Myles, P.S.; Myles, D.B.; Galagher, W.; Chew, C.; MacDonald, N.; Dennis, A. Minimal Clinically Important Difference for Three Quality of Recovery Scales. Anesthesiology 2016, 125, 39–45. [Google Scholar] [CrossRef] [Green Version]
  22. Australian and New Zealand College of Anaesthetists: Faculty of Pain Medicine. Position Statement on the Use of Slow-Release Opioid Preparations in the Treatment of Acute Pain. Available online: http://www.anzca.edu.au/resources/endorsed-guidelines/position-statement-on-the-use-of-slow-release-opio (accessed on 26 June 2020).
  23. Shah, A.; Hayes, C.J.; Martin, B.C. Characteristics of Initial Prescription Episodes and Likelihood of Long-Term Opioid Use—United States, 2006–2015. MMWR. Morb. Mortal. Wkly. Rep. 2017, 66, 265–269. [Google Scholar] [CrossRef] [Green Version]
  24. Allen, M.L.; Kim, C.C.; Braat, S.; Jones, K.; Winter, N.; Hucker, T.R.; Chia, A.; Lang, C.; Brooks, S.L.; Williams, D.L. Post-discharge opioid use and handling in surgical patients: A multicentre prospective cohort study. Anaesth. Intensive Care 2020, 48, 36–42. [Google Scholar] [CrossRef]
  25. Roughead, E.E.; Lim, R.; Ramsay, E.; Moffat, A.K.; Pratt, N.L. Persistence with opioids post discharge from hospitalisation for surgery in Australian adults: A retrospective cohort study. BMJ Open 2019, 9, e023990. [Google Scholar] [CrossRef]
  26. Brat, G.A.; Agniel, D.; Beam, A.; Yorkgitis, B.; Bicket, M.; Homer, M.; Fox, K.P.; Knecht, D.B.; McMahill-Walraven, C.N.; Palmer, N.; et al. Postsurgical prescriptions for opioid naive patients and association with overdose and misuse: Retrospective cohort study. BMJ 2018, 360, j5790. [Google Scholar] [CrossRef] [Green Version]
  27. Macintyre, P.E.; Huxtable, C.A.; Flint, S.L.P.; Dobbin, M.D.H. Costs and Consequences: A Review of Discharge Opioid Prescribing for ongoing Management of Acute Pain. Anaesth. Intensive Care 2014, 42, 558–574. [Google Scholar] [CrossRef] [Green Version]
Table 1. QoR–15 patient survey.
Table 1. QoR–15 patient survey.
How Have you Been Feeling in the Last 24 h?
(0 to 10, where 0 = none of the time [poor] and 10 = all of the time [excellent]
1.Able to breathe easilyNone of
the time
012345678910All of
the time
2.Able to enjoy foodNone of
the time
012345678910All of
the time
3.Feeling restedNone of
the time
012345678910All of
the time
4.Have had a good sleepNone of
the time
012345678910All of
the time
5.Able to look after personal
toilet and hygiene unaided
None of
the time
012345678910All of
the time
6.Able to communicate with
family or friends
None of
the time
012345678910All of
the time
7.Able to get support from
hospital doctors and nurses
None of
the time
012345678910All of
the time
8.Able to do usual
home activities
None of
the time
012345678910All of
the time
9.Feeling comfortable and
in control
None of
the time
012345678910All of
the time
10.Having a feeling of general
well-being
None of
the time
012345678910All of
the time
How Have you Had Any of the Following in the Last 24 h?
(0 to 10, where 0 = none of the time [excellent] and 10 = all of the time [poor]
1.Moderate painNone of
the time
012345678910All of
the time
2.Severe painNone of
the time
012345678910All of
the time
3.Nausea or vomitingNone of
the time
012345678910All of
the time
4.Feeling worried or anxiousNone of
the time
012345678910All of
the time
5.Feeling sad
or depressed
None of
the time
012345678910All of
the time
Table 2. Inclusion and exclusion criteria.
Table 2. Inclusion and exclusion criteria.
InclusionExclusion
Aged 18–65 yearsPregnant women
Booked for elective laparoscopic cholecystectomy or tubal ligation (with or without oophorectomyBody Mass Index > 35
Independent capacity to consent to participate in the trialAllergy to opioids
American Society of Anesthesiologists (ASA) physical health score of I-IIAllergy to adjuvant drugs
Persistent opioid use. Patients with OMEDD greater than 0 during previous week pre-op and taken for seven successive days or more including recreational drug use.
Non-elective surgery
Non-English speaking
Table 3. Recruitment and consent.
Table 3. Recruitment and consent.
Step 1The research nurse investigator will liaise with the theater booking team to identify potentially eligible patients scheduled on theater lists.
Step 2At the patient’s pre-anesthetic appointment, the research nurse investigator will undertake an initial screening of those patient’s clinical records for inclusion and exclusion criteria.
Step 3Potentially eligible patients will then be approached by the research nurse and the project outlined with them. Patients who initially indicate interest will be provided with an information sheet; the researcher nurse will talk about the project broadly and encourage the patient to ask questions. The research nurse will contact the principal investigator if response clarification is required.
Step 4Those patients who have been provided with an information sheet will then see the anesthetist who will explain in detail the project, the mechanism by which OFA works, and the process of randomization.
Step 5Patients will be assured that the treating doctors will follow all usual procedures to ensure the patient does not experience pain. That is, if it is evident that the patient is in pain, standard pain management protocols will be enacted during surgery and while the patient is in recovery.
Step 6If the patient is comfortable with participation, the formal consent procedures will be undertaken. The random allocation as to whether the participant receives OFA or standard anesthesia will be explained to participants. It will also be explained that participants can choose to withdraw consent at any time and that the right of any participant to refuse to participate in the trial at any time without giving reasons will be respected and will not prejudice their further treatment.
Table 4. Induction of anesthesia.
Table 4. Induction of anesthesia.
Standard Anesthetic Induction Opioid-Free Anesthesia Induction
Anxiolytic agent: 1 min prior to induction
Midazolam 1–3 mg IV
Anxiolytic agent: 1 min prior to induction
Midazolam 1–3 mg IV
Fentanyl 1–2 mcg/kg IVClonidine 1–3 mcg/kg IV in 3 divided doses (at induction, mid-surgery and end)
Lidocaine as per Table 5Lidocaine as per Table 5
Propofol 1–2.5 mg/kg IV bolus or as per programmed Target Control Infusion (TCI)Propofol 1–2.5 mg/kg IV bolus or as per programmed Target Control Infusion (TCI)
Magnesium sulphate 40 mg/kg (ideal body weight IBW)
Dexamethasone 8 mg IV and
Parecoxib 40 mg IV
Dexamethasone 8 mg IV and
Parecoxib 40 mg IV
Neuromuscular blocker (NMB): Choice of any
NMB is at the anesthetist’s discretion with
NMB monitoring
Neuromuscular blocker (NMB): Choice of any
NMB is at the anesthetist’s discretion with
NMB monitoring
Beta-blocker: Esmolol 10–50 mg IV bolus 20 s prior to intubation
On standby IV bolus medications:
  • Ephedrine 3 mg/mL
  • Phenylephrine 100 mcg/mL or
  • Metaraminol 0.5 mg/mL
On standby IV bolus medications:
  • Ephedrine 3 mg/mL
  • Phenylephrine 100 mcg/mL or
  • Metaraminol 0.5 mg/mL
Table 5. Maintenance of anesthesia: standard and opioid-free.
Table 5. Maintenance of anesthesia: standard and opioid-free.
Maintenance of Standard AnesthesiaMaintenance of Opioid-Free Anesthesia
Lidocaine 1.5–2.5 mg/kg/h IVI as per Table 5Lidocaine 1.5–2.5 mg/kg/h IVI as per Table 5
Opioids analgesics:
  • Fentanyl 1–3 mcg/kg IV or
  • Morphine 0.1–0.2 mg/kg IV
Magnesium sulphate: 20 mg/kg/h IVI (IBW)
Sevoflurane/desflurane 0.6–0.8 MAC (Minimal alveolar concentration) with BIS (Bispectral Index Score) target around 40 or
Propofol TCI infusion (TCI 4-8 mcg/mL)
Sevoflurane/desflurane 0.6–0.8 MAC (Minimal alveolar concentration) with BIS (Bispectral Index Score) target around 40 or
Propofol TCI infusion (TCI 4–8mcg/mL)
Ketamine 0.25–0.5 mg/kg IV (bolus before end of surgery)
Paracetamol IV 1000 mg IV Paracetamol IV 1000 mg
Clonidine 1–3 mcg/kg IV in 3 divided doses (at induction, mid surgery and end)
Ondansetron 4 mg IVOndansetron 4 mg IV
Table 6. Intravenous lidocaine administration guidelines.
Table 6. Intravenous lidocaine administration guidelines.
CriteriaPoints Scored
Hypoalbuminaemia1 point
Heart block (first degree)1 point
Participant on Vaughan–Williams Type 1 medications; sodium channel blocker1 point
Deranged liver function or hemihepatectomy1 point
CKD stages 3–41 point
Age ≥ 65 years2 points
Risk categorySum of points scored
Low risk<2 points
Intermediate risk2–3 points
High risk>3 points
Low risk
Administer a loading dose of 1.5 mg/kg by intravenous injection over 2–5 min. Maximum loading dose of 150 mg. Followed by a maintenance intravenous infusion at 2.5 mg/kg/h until local anesthetic infiltration by surgeon after wound closure.
Intermediate risk
Administer a loading dose of 1.25 mg/kg by intravenous injection over 2–5 min. Maximum loading dose of 150 mg. Followed by a maintenance intravenous infusion at a rate of 2 mg/kg/h until local anesthetic infiltration by surgeon after wound closure.
High risk
Administer a loading dose of 1 mg/kg by intravenous injection over 2–5 min. Maximum loading dose of 100 mg. Followed by a maintenance intravenous infusion at a rate of 1.5 mg/kg/h until local anesthetic infiltration by surgeon after wound closure.
Table 7. Definitions, variables and measures.
Table 7. Definitions, variables and measures.
MeasureDefinitionAssessment
Pre-opIntra-opPost-op
Administration of all peri-operative analgesia (theater/PACU and ward/unit)
  • Time to analgesia
  • Type of analgesia
  • Route of analgesia
  • Dose of analgesia
Time, type and dose of all analgesia peri-operatively xx
Adverse events (AE) and serious adverse event (SAE)Adverse events are considered “serious” if they threaten life or function. Due to the significant information they provide, serious adverse events require expedited reporting.
Serious adverse events (SAEs) are defined as any AE which:
  • Results in death
  • Is life-threatening
  • Requires in-patient hospitalization or prolongation of existing hospitalization
  • Results in persistent or significant disability/incapacity
The term “life-threatening” in the definition of “serious” refers to an event in which the participant was immediately at risk of death at the time of event. It does not refer to an event, which hypothetically might have caused death if it were more severe. However, important clinical events may be considered a serious adverse experience if they require clinical intervention to prevent one of the listed definitions, e.g., an “allergic bronchospasm”, which required intensive treatment in an emergency room or at home.
SAEs will be reported, whether or not it is considered related to trial treatment.
xx
American Society of Anesthesiologists (ASA) scoreThe ASA score is a subjective assessment of a participant’s overall health that is based on five classes (I to V) and will be completed at eligibility screening. Only participants scoring I or II are eligible for the study.
I
Participant is a completely healthy and fit participant
II
Participant has mild systemic disease
III
Participant has severe systemic disease that is not incapacitating
IV
Participant has incapacitating disease that is a constant threat to life
V
A moribund participant who is not expected to live 24 h with or without surgery
x
Analgesia monitoring formHospital analgesia monitoring form for intravenous, PCA, epidural and regional analgesia—adult xxx
Clinical assessmentStandard clinical observations, temperature, pulse, blood pressure, physical examination and cognitive assessment, height and weight—BMIxxx
Complications–anesthetic and surgicalYes/no plus text description of complication xx
Date and time discharged from hospitalParticipant notes, discharge summary x
Date of surgeryParticipant clinical notes x
Day 1 observationsAssessment conducted at 24 h (± two-hours) post-discharge from theater (i.e., admission to PACU time) on Day 1 post-operatively x
Delirium/hallucinationsAny evidence/report of = Yes x
Documentation confirming participant eligibilityTrial eligibility and consent form in participant notesxx
Duration of surgeryAARK times. xx
Functional activity scoresScored A = activity unlimited, B = activity mild to moderately limited by pain, C = activity severely limited by pain. x x
Local anesthetic toxicityAny evidence of symptoms = Yes xx
Medical history/comorbiditiesParticipant clinical notes- text descriptionx
Medication administeredIntra-operative, PACU and post-operative list of medications, dose, route xx
Oral morphine equivalent daily dose (OMEDD) Oral morphine equivalent daily dose (OMEDD) allows for comparison of other opioids from all routes, oral, sublingual, transdermal, parental or rectal preparations to be summed and converted to an equivalent total oral morphine daily dose. Calculated with the Faculty of Pain Medicine (FPM), Australia and New Zealand College of Anesthetists (ANZCA) Opioid Calculator
Participant self-report of prescription or recreational use of opioids pre-surgery. OMEDD scores will be calculated at baseline (for eligibility criteria) and at Day 1 to include surgery, PACU and ward/home
xxx
Overall length of stay (LOS)LOS from theater start time to discharge from hospital (hours: minutes)xxx
PACUPost-anesthetic care unit
PACU LOSPACU LOS from automated anesthetic record keeping (AARK) admission time to PACU discharge time x
PainSeverity of pain is measured using the numerical rating scale, a points scale from zero to ten. Pain is documented as stated by the participant. 0 = no pain, 1 to 3 = mild pain, 4 to 6 = moderate pain and 7 to 10 = severe pain. x x
Pain score on Day 10 = no pain, 1 to 3 = mild pain, 4 to 6 = moderate pain and 7 to 10 = severe pain. x
Pain score–highest pain score recorded while in PACU0 = no pain, 1 to 3 = mild pain, 4 to 6 = moderate pain and 7 to 10 = severe pain. x
Participant demographics and clinical characteristicsCollected at baseline including:
Date of birth (age)
Gender
Smoking status
Medical history/comorbiditiesCurrent weight and height (Body Surface Area/Body Mass Index)
x
Quality of recovery form (QoR-15)Evaluation of post-operative recovery from the participant’s perspective with total QoR-15 score ranging from 0 (extremely poor recovery) to 150 (excellent recovery). Assessment at baseline and at 24-h post-operatively. x x
Sedation levelsSedation scored as 0 = awake, 1 = mild (easy to rouse), 2 = moderate (rousable but unable to keep eyes open more than 10 s) and 3 = severe (difficult or unable to rouse) x
Time until post-operative mobilizationTime from admission to PACU until the time first mobilized x
Time admitted to PACUArrival time in PACU x
Type of surgeryTubal ligation/cholecystectomyxx
Urinary complicationsUrinary system complications x
Vital signsAll vital signs from baseline until hospital discharge including:
Respiratory rate
Blood pressure
Oxygen saturation
Pulse rate
Temperature
xxx

Share and Cite

MDPI and ACS Style

Eidan, A.; Ratsch, A.; Burmeister, E.A.; Griffiths, G. Comparison of Opioid-Free Anesthesia Versus Opioid-Containing Anesthesia for Elective Laparoscopic Surgery (COFA: LAP): A Protocol Measuring Recovery Outcomes. Methods Protoc. 2020, 3, 58. https://doi.org/10.3390/mps3030058

AMA Style

Eidan A, Ratsch A, Burmeister EA, Griffiths G. Comparison of Opioid-Free Anesthesia Versus Opioid-Containing Anesthesia for Elective Laparoscopic Surgery (COFA: LAP): A Protocol Measuring Recovery Outcomes. Methods and Protocols. 2020; 3(3):58. https://doi.org/10.3390/mps3030058

Chicago/Turabian Style

Eidan, Anthony, Angela Ratsch, Elizabeth A. Burmeister, and Geraldine Griffiths. 2020. "Comparison of Opioid-Free Anesthesia Versus Opioid-Containing Anesthesia for Elective Laparoscopic Surgery (COFA: LAP): A Protocol Measuring Recovery Outcomes" Methods and Protocols 3, no. 3: 58. https://doi.org/10.3390/mps3030058

Article Metrics

Back to TopTop