APOLLO-2: A Randomized, Placebo and Active-Controlled Phase III Study Investigating Oliceridine (TRV130), a G Protein-Biased Ligand at the μ-Opioid Receptor, for Management of Moderate to Severe Acute Pain Following Abdominoplasty

Neil K Singla, Franck Skobieranda, David G Soergel, Monica Salamea, David A Burt, Mark A Demitrack, Eugene R Viscusi, Neil K Singla, Franck Skobieranda, David G Soergel, Monica Salamea, David A Burt, Mark A Demitrack, Eugene R Viscusi

Abstract

Objectives: The clinical utility of conventional IV opioids is limited by the occurrence of opioid-related adverse events. Oliceridine is a novel G protein-biased μ-opioid receptor agonist designed to provide analgesia with an improved safety and tolerability profile. This phase III, double-blind, randomized trial (APOLLO-2 [NCT02820324]) evaluated the efficacy and safety of oliceridine for acute pain following abdominoplasty.

Methods: Patients received a loading dose of either placebo, oliceridine (1.5 mg), or morphine (4 mg), followed by demand doses via patient-controlled analgesia (0.1, 0.35, or 0.5 mg oliceridine; 1 mg morphine; or placebo) with a 6-minute lockout interval. The primary endpoint was the proportion of treatment responders over 24 hours for oliceridine regimens compared to placebo. Secondary outcomes included a predefined composite measure of respiratory safety burden (RSB, representing the cumulative duration of respiratory safety events) and the proportion of treatment responders vs. morphine.

Results: A total of 401 patients were treated with study medication. Effective analgesia was observed for all oliceridine regimens, with responder rates of 61.0%, 76.3%, and 70.0% for the 0.1-, 0.35-, and 0.5-mg regimens, respectively, compared with 45.7% for placebo (all P < 0.05) and 78.3% for morphine. Oliceridine 0.35- and 0.5-mg demand dose regimens were equi-analgesic to morphine using a noninferiority analysis. RSB showed a dose-dependent increase across oliceridine regimens (mean hours [standard deviation], 0.1 mg: 0.43 [1.56]; 0.35 mg: 1.48 [3.83]; 0.5 mg: 1.59 [4.26]; all comparisons not significant at P > 0.05 vs. placebo: 0.60 [2.82]). The RSB measure for morphine was 1.72 (3.86) (P < 0.05 vs. placebo). Gastrointestinal adverse events increased in a dose-dependent manner across oliceridine demand dose regimens (0.1 mg: 49.4%; 0.35 mg: 65.8%; 0.5 mg: 78.8%; vs. placebo: 47.0%; and morphine: 79.3%). In comparison to morphine, the proportion of patients experiencing nausea or vomiting was lower with the 2 equi-analgesic dose regimens of 0.35 and 0.5 mg oliceridine.

Conclusions: Oliceridine is a safe and effective IV analgesic for the relief of moderate to severe acute postoperative pain in patients undergoing abdominoplasty. Since the low-dose regimen of 0.1 mg oliceridine was superior to placebo but not as effective as the morphine regimen, safety comparisons to morphine are relevant only to the 2 equi-analgesic dose groups of 0.35 and 0.5 mg, which showed a favorable safety and tolerability profile regarding respiratory and gastrointestinal adverse effects compared to morphine. These findings support that oliceridine may provide a new treatment option for patients with moderate to severe acute pain where an IV opioid is warranted.

Keywords: abdominoplasty; analgesia; clinical trial; patient controlled; postoperative.

Conflict of interest statement

Mark Demitrack is a full‐time employees of Trevena Inc. and owns stock in Trevena Inc. David G. Soergel, Franck Skobieranda, Monica Salamea and David A. Burt were full‐time employees, and stockholders, of Trevena Inc. at the time the research was conducted and the manuscript was fully written. Neil Singla is the founder and CEO of Lotus Clinical Research LLC, an analgesic CRO and research site; in this capacity he has served as a consultant and provided clinical trial services for Trevena Inc. Eugene R. Viscusi has served as a consultant to Trevena Inc.

© 2019 The Authors. Pain Practice published by Wiley Periodicals, Inc. on behalf of World Institute of Pain.

Figures

Figure 1
Figure 1
Patient disposition. All percentages are based on the number of patients randomized. Efficacy and safety analyses were conducted on all randomized patients who received ≥1 dose of study medication. Efficacy analyses were based on the randomized treatment assignment, while safety analyses were based on the actual treatment received. This explains the minor discrepancies seen between the number of patients analyzed for efficacy and safety in some treatment regimens. IPP, immediate postoperative period.
Figure 2
Figure 2
A, Cumulative use of rescue pain medication. B, First perceptible and first meaningful pain relief. In (A), the cumulative percentage of patients using rescue pain medication in each regimen is shown at predefined time points throughout the 24‐hour treatment period. Patients could receive open‐label rescue pain medication (oral etodolac 200 mg every 6 hours PRN) if they reported a score ≥ 4 on the pain NRS. In (B), the median time to first perceptible and first meaningful pain relief is presented, as reported by patients using the two‐stopwatch method. *P < 0.05 compared to morphine. CI, confidence interval; NRS, numeric rating scale; PRN, as needed.
Figure 3
Figure 3
A, Primary treatment response analysis of oliceridine compared with placebo (treatment responders at 24 hours). B, Treatment responders over the full treatment period. This primary endpoint analysis compared the percentage of treatment responders in each oliceridine regimen with the percentage of responders in the placebo regimen at 24 hours post loading dose. Responders were those who reached a ≥30% improvement in time‐weighted sum of pain intensity difference (SPID‐24) from baseline, whilst (1) not received rescue pain medication, (2) not discontinuing study medication early, and (3) without reaching dosing limits. *P < 0.05 vs. placebo (Hochberg adjusted). CI, confidence interval.
Figure 4
Figure 4
The key prespecified secondary endpoint of cumulative respiratory safety burden. During the randomized treatment period, patients were monitored on a protocol‐defined schedule by either a certified registered nurse anesthetist or an anesthesiologist, blinded to study medication assignment. The monitoring professional intervened when clinically indicated and determined the onset and resolution of each respiratory safety event. Respiratory safety burden was defined as the expected cumulative duration of respiratory safety events in a particular treatment group and was calculated as the mathematical product of the prevalence of respiratory safety events and the mean conditional duration (ie, mean duration in affected patients) of such events (see Table 3). *P < 0.05 compared to morphine (unadjusted). Mean respiratory safety burden from the model‐based estimate was 7, 5, 19, 25, and 32 minutes for the placebo, oliceridine 0.1, 0.35, and 0.5 mg, and morphine groups, respectively. SD, standard deviation.
Figure 5
Figure 5
Clinical interventions. A, The proportion of patients in each regimen who experienced any dosing interruption of study medication during the study is presented. Exploratory analyses showed that the odds ratio of an interruption compared to morphine was 0.09 (P < 0.0001) for placebo, 0.15 (P = 0.0002) for oliceridine 0.1 mg, 0.57 (P = 0.13) for oliceridine 0.35 mg, and 0.64 (P = 0.23) for oliceridine 0.5 mg regimens. B, The proportion of patients in each regimen who required supplemental oxygen therapy is presented. Exploratory analyses showed that the odds ratio of an interruption compared to morphine was 0.15 (P = 0.0002) for placebo, 0.18 (P = 0.0005) for oliceridine 0.1 mg, 0.54 (P = 0.11) for oliceridine 0.35 mg, and 0.65 (P = 0.25) for oliceridine 0.5 mg regimens. *P < 0.05 odds ratio vs. morphine.
Figure 6
Figure 6
(A) The proportion of responders over the first 60 minutes of treatment and (B) time to 1‐point improvement in NRS score. A, Treatment response during this period can largely be attributed to study drug (loading dose at Time 0 and demand doses starting at 10 minutes) since supplemental study medication doses were prohibited and rescue pain medication was discouraged during this time. *P < 0.05 compared to morphine. B, NRS pain scores were reported at baseline (up to 10 minutes before loading dose [Time 0]); at 5, 10, 15, 30, and 45 minutes; and at 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, and 24 hours post‐loading dose. *P < 0.05 vs. morphine. CI, confidence interval; NRS, numeric rating scale.
Figure 7
Figure 7
Rescue antiemetic use. This analysis examined post‐treatment rescue antiemetic use. Patients could receive rescue antiemetic medication if they were actively vomiting, or if they requested an antiemetic and reported moderate to severe nausea on a 4‐point scale (none, mild, moderate, severe). Prophylactic antiemetics were not permitted perioperatively or during the randomized treatment period. *P < 0.05 for odds ratio for rescue antiemetic use vs. morphine.

References

    1. Thomazeau J, Rouquette A, Martinez V, et al. Acute pain factors predictive of post‐operative pain and opioid requirement in multimodal analgesia following knee replacement. Eur J Pain. 2016;20:822–832.
    1. Fiala T. Tranversus abdominis plane block during abdominoplasty to improve postoperative patient comfort. Aesthet Surg J. 2015;35:72–80.
    1. Morales R Jr, Mentz H 3rd, Newall G, Patronella C, Masters O 3rd. Use of abdominal field block injections with liposomal bupivicaine to control postoperative pain after abdominoplasty. Aesthet Surg J. 2013;33:1148–1153.
    1. Feng LJ. Painless abdominoplasty: the efficacy of combined intercostal and pararectus blocks in reducing postoperative pain and recovery time. Plast Reconstr Surg. 2010;126:1723–1732.
    1. Khansa I, Koogler A, Richards J, Bryant R, Janis JE. Pain management in abdominal wall reconstruction. Plast Reconstr Surg Glob Open. 2017;5:e1400.
    1. Stein C. New concepts in opioid analgesia. Expert Opin Investig Drugs. 2018;27:765–775.
    1. Benyamin R, Trescot AM, Datta S, et al. Opioid complications and side effects. Pain Physician. 2008;11(2 suppl):S105–S120.
    1. Wheeler M, Oderda GM, Ashburn MA, Lipman AG. Adverse events associated with postoperative opioid analgesia: a systematic review. J Pain. 2002;3:159–180.
    1. Kessler ER, Shah M, Gruschkus SK, Raju A. Cost and quality implications of opioid‐based postsurgical pain control using administrative claims data from a large health system: opioid‐related adverse events and their impact on clinical and economic outcomes. Pharmacotherapy. 2013;33:383–391.
    1. Overdyk FJ, Dowling O, Marino J, et al. Association of opioids and sedatives with increased risk of in‐hospital cardiopulmonary arrest from an administrative database. PLoS One. 2016;11:e0150214.
    1. Habib AS, Chen Y‐T, Taguchi A, Hu XH, Gan TJ. Postoperative nausea and vomiting following inpatient surgeries in a teaching hospital: a retrospective database analysis. Curr Med Res Opin. 2006;22:1093–1099.
    1. Wardhan R, Chelly J. Recent advances in acute pain management: understanding the mechanisms of acute pain, the prescription of opioids, and the role of multimodal pain therapy. F1000Res. 2017;6:2065.
    1. Al‐Hasani R, Bruchas MR. Molecular mechanisms of opioid receptor‐dependent signaling and behavior. Anesthesiology. 2011;115:1363–1381.
    1. Waldhoer M, Bartlett SE, Whistler JL. Opioid receptors. Annu Rev Biochem. 2004;73:953–990.
    1. Groer CE, Schmid CL, Jaeger AM, Bohn LM. Agonist‐directed interactions with specific β‐arrestins determine μ‐opioid receptor trafficking, ubiquitination, and dephosphorylation. J Biol Chem. 2011;286:31731–31741.
    1. Bohn LM, Gainetdinov RR, Lin F‐T, Lefkowitz RJ, Caron MG. μ‐Opioid receptor desensitization by β‐arrestin‐2 determines morphine tolerance but not dependence. Nature. 2000;408:720–723.
    1. Raehal KM, Walker JK, Bohn LM. Morphine side effects in β‐arrestin 2 knockout mice. J Pharmacol Exp Ther. 2005;314:1195–1201.
    1. DeWire SM, Yamashita DS, Rominger DH, et al. A G protein‐biased ligand at the μ‐opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine. J Pharmacol Exp Ther. 2013;344:708–717.
    1. Soergel DG, Subach RA, Burnham N, et al. Biased agonism of the μ‐opioid receptor by TRV130 increases analgesia and reduces on‐target adverse effects versus morphine: a randomized, double‐blind, placebo‐controlled, crossover study in healthy volunteers. Pain. 2014;155:1829–1835.
    1. Bohn LM, Lefkowitz RJ, Gainetdinov RR, Peppel K, Caron MG, Lin F‐T. Enhanced morphine analgesia in mice lacking β‐arrestin 2. Science. 1999;286:2495–2498.
    1. Manglik A, Lin H, Aryal D, et al. Structure‐based discovery of opioid analgesics with reduced side effects. Nat Biotechnol. 2016;537:185–190.
    1. Viscusi ER, Webster L, Kuss M, et al. A randomized, phase 2 study investigating TRV130, a biased ligand of the μ‐opioid receptor, for the intravenous treatment of acute pain. Pain. 2016;157:264–272.
    1. Singla N, Minkowitz HS, Soergel DG, et al. A randomized, phase 2b study investigating oliceridine (TRV130), a novel μ receptor G protein pathway selective (μ‐GPS) modulator, for management of moderate to severe acute pain following abdominoplasty. J Pain Res. 2017;10:2413–2424.
    1. Apfel CC, Läärä E, Koivuranta M, Greim C‐A, Roewer N. A simplified risk score for predicting postoperative nausea and vomiting: conclusions from cross‐validations between two centers. Anesthesiology. 1999;91:693–700.
    1. Physician Desk Reference . Morphine Sulfate Drug Summary from Physician's Desk Reference Digital Format. (accessed January 14, 2019)
    1. U.S. Food and Drug Administration . Guidance for Industry. Analgesic Indications: Developing Drug and Biological Products. U.S. Department of Health and Human Services, Food and Drug Administration Center for Drug Evaluation and Research (CDER); 2014. (accessed January 14, 2019)
    1. Moline B, Roberts M, Houser J. Validity and interrater reliability of the Moline‐Roberts Pharmacologic Sedation Scale. Clin Nurse Spec. 2012;26:140–148.
    1. Sunshine A, Mulhern SA, Olson N, Elkind A, Almas M, Sikes C. Comparative sensitivity of stopwatch methodology and conventional pain assessment measures for detecting early response to triptans in migraine: results of a randomized, open‐label pilot study. Clin Ther. 2006;28:1107–1115.
    1. Modena V, Bianchi G, Roccatello D. Cost‐effectiveness of biologic treatment for rheumatoid arthritis in clinical practice: an achievable target? Autoimmun Rev. 2013;12:835–838.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol. 1995;57:289–300.
    1. Viscusi ER, Skobieranda F, Soergel DG, Cook E, Burt DA, Singla N. APOLLO‐1: a randomized, placebo‐ and active‐controlled Phase 3 study investigating oliceridine (TRV130), a novel μ receptor G protein pathway selective modulator, for management of moderate to severe acute pain following bunionectomy. J Pain Res. 2019;12:927–943.
    1. Gan TJ, Epstein R, Menzel B, et al. Physician practice patterns and treatment challenges in acute postoperative pain management. PAINWeek abstract book 2017. Postgrad Med. 2017;129(suppl 1):1–85 (abs. 96).
    1. Hsia H‐L, Takemoto S, van de Ven T, et al. Acute pain is associated with chronic opioid use after total knee arthroplasty. Reg Anesth Pain Med. 2018;43:705–711.
    1. Chen XT, Pitis P, Liu G, et al. Structure‐activity relationships and discovery of a G protein biased mu opioid receptor ligand, [(3‐methoxythiophen‐2‐yl)methyl]({2‐[(9R)‐9‐(pyridin‐2‐yl)‐6‐oxaspiro‐[4.5]decan‐ 9‐yl]ethyl})amine (TRV130), for the treatment of acute severe pain. J Med Chem. 2013;56:8019–8031.
    1. Jarzyna D, Jungquist CR, Pasero C, et al. American Society for Pain Management Nursing guidelines on monitoring for opioid‐induced sedation and respiratory depression. Pain Manag Nurs. 2011;12:118–145.e10.
    1. Patanwala AE, Keim SM, Erstad BL. Intravenous opioids for severe acute pain in the emergency department. Ann Pharmacother. 2010;44:1800–1809.
    1. Levy N, Sturgess J, Mills P. “Pain as the fifth vital sign” and dependence on the “numerical pain scale” is being abandoned in the US: why? Br J Anaesth. 2018;120:435–438.

Source: PubMed

3
Subscribe