278
Views
2
CrossRef citations to date
0
Altmetric
Review

New and developing anesthesia drugs

&
Pages 195-204 | Received 03 Nov 2016, Accepted 06 Jan 2017, Published online: 20 Jan 2017

References

  • Sneyd JR, Rigby-Jones AE. New drugs and technologies, intravenous anaesthesia is on the move (again). Br J Anaesth. 2010;105:246–254.
  • Sneyd JR. Remimazolam: new beginnings or just a me-too? Anesth Analg. 2012;115:217–219.
  • Goudra BG, Singh PM. Remimazolam: the future of its sedative potential. Saudi J Anaesth. 2014;8:388–391.
  • Wiltshire HR, Kilpatrick GJ, Tilbrook GS, et al. A placebo- and midazolam-controlled phase I single ascending-dose study evaluating the safety, pharmacokinetics, and pharmacodynamics of remimazolam (CNS 7056): part II. Population pharmacokinetic and pharmacodynamic modeling and simulation. Anesth Analg. 2012;115:284–296.
  • Antonik LJ, Goldwater DR, Kilpatrick GJ, et al. A placebo- and midazolam-controlled phase I single ascending-dose study evaluating the safety, pharmacokinetics, and pharmacodynamics of remimazolam (CNS 7056): part I. Safety, efficacy, and basic pharmacokinetics. Anesth Analg. 2012;115:274–283.
  • Saari TI, Uusi-Oukari M, Ahonen J, et al. Enhancement of GABAergic activity: neuropharmacological effects of benzodiazepines and therapeutic use in anesthesiology. Pharmacol Rev. 2011;63:243–267.
  • Worthington MT, Antonik LJ, Goldwater DR, et al. A phase Ib, dose-finding study of multiple doses of remimazolam (CNS 7056) in volunteers undergoing colonoscopy. Anesth Analg. 2013;117:1093–1100.
  • Rogers WK, McDowell TS. Remimazolam, a short-acting GABA(A) receptor agonist for intravenous sedation and/or anesthesia in day-case surgical and non-surgical procedures. IDrugs. 2010;13:929–937.
  • Sato S, Doi M, Morita K, et al. Remimazolam a new ultra-short acting anesthetic shows similar efficacy and superior hemodynamic stability vs. propofol in general surgery patients with TIVA: results of a randomised, non-inferiority, phase IIb/III trial. Abstract Presentation, ASA Annual Meeting; 2015 Oct 28. p. A5018.
  • Chahar P, Cummings KC. Liposomal bupivacaine: a review of a new bupivacaine formulation. J Pain Res. 2012;5:257–264.
  • Hu D, Onel E, Singla N, et al. Pharmacokinetic profile of liposome bupivacaine injection following a single administration at the surgical site. Clin Drug Investig. 2013;33:109–115.
  • Davidson EM, Barenholz Y, Cohen R, et al. High-dose bupivacaine remotely loaded into multivesicular liposomes demonstrates slow drug release without systemic toxic plasma concentrations after subcutaneous administration in humans. Anesth Analg. 2010;110:1018–1023.
  • Langford RM, Chappell GM, Karrasch JA, et al. A single administration of depobupivacaine intraoperatively results in prolonged detectable plasma bupivacaine and analgesia in patients undergoing inguinal hernia repair. Poster presented at: 62nd Annual Postgraduate Assembly of the New York State Society of Anesthesiologists; 2008 Dec 12–16; New York, NY.
  • Dullenkopf A, Borgeat A. Local anesthetics. Differences and similarities in the “-cains”. Anaesthesist. 2003;52:329–340.
  • Covino BG. Pharmacology of local anaesthetic agents. Br J Anaesth. 1986;58:701–716.
  • Longhorn RK, Cross M. Bupivacaine toxicity. J Cardiothorac Vasc Anesth. 2000;14:759–760.
  • Bourne E, Wright C, Royse C. A review of local anesthetic cardiotoxicity and treatment with lipid emulsion. Local Reg Anesth. 2010;3:11–19.
  • Viscusi ER, Sinatra R, Onel E, et al. The safety of liposome bupivacaine, a novel local analgesic formulation. Clin J Pain. 2014;30:102–110.
  • Gorfine SR, Onel E, Patou G, et al. Bupivacaine extended-release liposome injection for prolonged postsurgical analgesia in patients undergoing hemorrhoidectomy: a multicenter, randomized, double-blind, placebo-controlled trial. Dis Colon Rectum. 2011;54:1552–1559.
  • Golf M, Daniels SE, Onel E. A phase 3, randomized, placebo-controlled trial of DepoFoam® bupivacaine (extended-release bupivacaine local analgesic) in bunionectomy. Adv Ther. 2011;28:776–788.
  • Haas E, Onel E, Miller H, et al. A double-blind, randomized, active-controlled study for post-hemorrhoidectomy pain management with liposome bupivacaine, a novel local analgesic formulation. Am Surg. 2012;78:574–581.
  • Smoot JD, Bergese SD, Onel E, et al. The efficacy and safety of DepoFoam bupivacaine in patients undergoing bilateral, cosmetic, submuscular augmentation mammoplasty: a randomized, double-blind, active-control study. Aesthet Surg J. 2012;32:69–76.
  • Cohen SM. Extended pain relief trial utilizing infiltration of Exparel(®), a long-acting multivesicular liposome formulation of bupivacaine: a Phase IV health economic trial in adult patients undergoing open colectomy. J Pain Res. 2012;5:567–572.
  • Viscusi ER, Candiotti KA, Onel E, et al. The pharmacokinetics and pharmacodynamics of liposome bupivacaine administered via a single epidural injection to healthy volunteers. Reg Anesth Pain Med. 2012;37:616–622.
  • Ilfeld BM, Viscusi ER, Hadzic A, et al. Safety and side effect profile of liposome bupivacaine (Exparel) in peripheral nerve blocks. Reg Anesth Pain Med. 2015;40:572–582.
  • Hamilton TW, Athanassoglou V, Trivella M, et al. Liposomal bupivacaine peripheral nerve block for the management of postoperative pain. Cochrane Database Syst Rev. 2016 Aug 25;(8). Art. No.: CD011476.
  • Tong YC, Kaye AD, Urman RD. Liposomal bupivacaine and clinical outcomes. Best Pract Res Clin Anaesthesiol. 2014;28:15–27.
  • Dasta J, Ramamoorthy S, Patou G, et al. Bupivacaine liposome injectable suspension compared with bupivacaine HCl for the reduction of opioid burden in the postsurgical setting. Curr Med Res Opin. 2012;28:1609–1615.
  • Craig RG, Hunter JM. Neuromuscular blocking drugs and their antagonists in patients with organ disease. Anaesthesia. 2009;64(Suppl 1):55–65.
  • Naguib M. Sugammadex: another milestone in clinical neuromuscular pharmacology. Anesth Analg. 2007;104:575–581.
  • Bom A, Bradley M, Cameron K, et al. A novel concept of reversing neuromuscular block: chemical encapsulation of rocuronium bromide by a cyclodextrin-based synthetic host. Angewandte Chemie. 2002;41:266–270.
  • Full prescribing information for Bridon (Sugammadex). Patheon Manufacturing Services LLC, Greenville, NC27834, USA for Merck Sharp & Dohme Corp, Merck & Co, Inc; 2015. [Revised 2016 Sep].
  • Gijsenbergh F, Ramael S, Houwing N, et al. First human exposure of Org 25969, a novel agent to reverse the action of rocuronium bromide. Anesthesiology. 2005;103:695–703.
  • Keating GM. sugammadex: a review of neuromuscular blockade reversal. Drugs. 2016;76:1041–1052.
  • Hemmerling TM, Zaouter C, Geldner G, et al. Sugammadex: a short review and clinical recommendations for the cardiac anesthesiologist. Ann Card Anaesth. 2010;13:206–216.
  • Staals LM, Snoeck MMJ, Driessen JJ, et al. Reduced clearance of rocuronium and sugammadex in patients with severe to end-stage renal failure: a pharmacokinetic study. Br J Anaesth. 2010;104:31–39.
  • Dahl V, Pendeville PE, Hollmann MW, et al. Safety and efficacy of sugammadex for the reversal of rocuronium-induced neuromuscular blockade in cardiac patients undergoing noncardiac surgery. Eur J Anaesthesiol. 2009;26:874–884.
  • Cammu G, Coart D, De Graeve K, et al. Reversal of rocuroniuminduced neuromuscular block with sugammadex in heart failure patients: a prospective observational study. Acta Anaesthesiol Belg. 2012;63:69–73.
  • de Kam PJ, van Kuijk J, Prohn M, et al. Effects of sugammadex doses up to 32 mg/kg alone or in combination with rocuronium or vecuronium on QTc prolongation: a thorough QTc study. Clin Drug Investig. 2010;30:599–611.
  • Zwiers A, van den Heuvel M, Smeets J, et al. Assessment of the potential for displacement interactions with sugammadex: a pharmacokinetic-pharmacodynamic modelling approach. Clin Drug Investig. 2011;31:101–111.
  • Abrishami A, Ho J, Wong J, et al. Cochrane corner: sugammadex, a selective reversal medication for preventing postoperative residual neuromuscular blockade. Anesth Analg. 2010;110:1239.
  • de Kam PJ, Grobara P, Prohn M, et al. Effects of sugammadex on activated partial thromboplastin time and prothrombin time in healthy subjects. Int J Clin Pharmacol Ther. 2014;52:227–236.
  • Dirkmann D, Britten MW, Pauling H, et al. Anticoagulant effect of sugammadex: just an in vitro artifact. Anesthesiology. 2016;124:1277–1285.
  • de Kam P-J, El Galta R, Kruithof AC, et al. No clinically relevant interaction between sugammadex and aspirin on platelet aggregation and coagulation parameters. Int J Clin Pharmacol Ther. 2013;51:976–985.
  • de Kam P-J, Kruithof AC, van Lierop M-J, et al. Lack of a clinically relevant effect of sugammadex on anti-Xa activity or activated partial thromboplastin time following pretreatment with either unfractionated or low-molecular-weight heparin in healthy subjects. Int J Clin Pharmacol Ther. 2014;52:631–641.
  • Rahe-Meyer N, Fennema H, Sam Schulman S, et al. Effect of reversal of neuromuscular blockade with sugammadex versus usual care on bleeding risk in a randomized study of surgical patients. Anesthesiology. 2014;121:969–977.
  • Raft J, Betala Belinga JF, Jurkolow G, et al. Clinical evaluation of post-surgical bleeding after a sugammadex injection. Ann Fr Anesth Reanim. 2011;30:714–717.
  • Tsur A, Kalansky A. Hypersensitivity associated with sugammadex administration: a systematic review. Anaesthesia. 2014;69:1251–1257.
  • Ho G, Clarke RC, Sadleir PH, et al. The first case report of anaphylaxis caused by the inclusion complex of rocuronium and sugammadex. A A Case Rep. 2016;7:190–192.
  • Del Valle EM. Cyclodextrins and their uses: a review. Process Biochem. 2004;39:1033–1046.
  • Mirakhur RK. Sugammadex in clinical practice. Anaesthesia. 2009;64(Suppl 1):45–54.
  • White PF, Tufanogullari B, Sacan O, et al. The effect of residual neuromuscular blockade on the speed of reversal with sugammadex. Anesth Analg. 2009;108:846–851.
  • Lee C, Jahr JS, Candiotti K, et al. Reversal of profound rocuronium NMV with sugammadex is faster than recovery from succinylcholine. Anesthesiology. 2007;107:A988.
  • Won YJ, Lim BG, Lee DK, et al. Sugammadex for reversal of rocuronium-induced neuromuscular blockade in pediatric patients: a systematic review and meta-analysis. Medicine (Baltimore). 2016;95:e4678.
  • Abad-Gurumeta A, Ripolles-Melchor J, Casans-Frances R, et al. A systematic review of sugammadex vs neostigmine for reversal of neuromuscular blockade. Anaesthesia. 2015;70:1441–1452.
  • Sacan O, White PF, Tufanogullari B, et al. Sugammadex reversal of rocuronium-induced neuromuscular blockade: a comparison with neostigmine-glycopyrrolate and edrophonium-atropine. Anesth Analg. 2007;104:569–574.
  • Brueckmann B, Sasaki N, Grobara P, et al. Effects of sugammadex on incidence of postoperative residual neuromuscular blockade: a randomized, controlled study. Br J Anaesth. 2015;115:743–751.
  • Haerter F, Simons JCP, Foerster U, et al. Comparative effectiveness of calabadion and sugammadex to reverse non-depolarizing neuromuscular-blocking agents. Anesthesiology. 2015;123:1337–1349.
  • Hoffmann U, Grosse-Sundrup M, Eikermann-Haerter K, et al. Calabadion: a new agent to reverse the effects of benzylisoquinoline and steroidal neuromuscular-blocking agents. Anesthesiology. 2013;119:317–325.
  • Diaz-Gil D, Haerter F, Falcinelli S, et al. A novel strategy to reverse general anesthesia by scavenging with the acyclic Cucurbit[n]uril-type molecular container calabadion 2. Anesthesiology. 2016;125:333–345.
  • Heerdt PM, Sunaga H, Owen JS, et al. Dose-response and cardiopulmonary side effects of the novel neuromuscular-blocking drug CW002 in man. Anesthesiology. 2016;125:1136–1143.
  • Sunaga H, Savarese JJ, McGilvra JD, et al. Preclinical pharmacology of CW002: a nondepolarizing neuromuscular blocking drug of intermediate duration, degraded and antagonized by l-cysteine-additional studies of safety and efficacy in the anesthetized rhesus monkey and cat. Anesthesiology. 2016;125:732–743.
  • National institute of drug abuse. Nationwide Trends. [Revised 2015 Jun]. Available from: https://www.drugabuse.gov/publications/drugfacts/nationwide-trends
  • Kunins HV. Abuse-deterrent opioid formulations: part of a public health strategy to reverse the opioid epidemic. JAMA Intern Med. 2015;175:987–988.
  • Abramowicz M (ed). From the Medical Letter on Drugs and Therapeutics: Abuse-deterrent opioid formulations. JAMA. 2015; 314:1744–1745.
  • Bartholomaeus JH, Arkenau-Marić E, Galia E. Opioid extended-release tablets with improved tamper-resistant properties. Expert Opin Drug Deliv. 2012;9:879–889.
  • Gudin J, Levy-Cooperman N, Kopecky EA, et al. Comparing the effect of tampering on the oral pharmacokinetic profiles of two extended-release oxycodone formulations with abuse-deterrent properties. Pain Med. 2015;16:2142–2151.
  • Fleming AB, Carlson DR, Varanasi RK, et al. Evaluation of an extended-release, abuse-deterrent, microsphere-in-capsule analgesic for the management of patients with chronic pain with dysphagia (CPD). Pain Pract. 2016;16:334–344.
  • Katz N, Kopecky EA, OʼConnor M, et al. A phase 3, multicenter, randomized, double-blind, placebo-controlled, safety, tolerability, and efficacy study of Xtampza ER in patients with moderate-to-severe chronic low back pain. Pain. 2015;156:2458–2467.
  • Pflomm JM (ed). Extended-release hydrocodone (Hysingla ER) for pain. Med Lett Drugs Ther. 2015;57:71–72.
  • Dhillon S. Hydrocodone Bitartrate ER (Hysingla® ER): a review in chronic pain. Clin Drug Investig. 2016;36:969–980.
  • Khan MF, Gharibo C. Abuse deterrent opioids. Tech Reg Anesth Pain Manag. 2010;14:99–103.
  • Johnson FK, Stark JG, Bieberdorf FA, et al. Relative oral bioavailability of morphine and naltrexone derived from crushed morphine sulfate and naltrexone hydrochloride extended-release capsules versus intact product and versus naltrexone solution: a single-dose, randomized-sequence, open-label, three-way crossover trial in healthy volunteers. Clin Ther. 2010;32:1149–1164.
  • Johnson F, Setnik B. Morphine sulfate and naltrexone hydrochloride extended-release capsules: naltrexone release, pharmacodynamics, and tolerability. Pain Physician. 2011;14:391–406.
  • Ruan X, Chen T, Gudin J, et al. Acute opioid withdrawal precipitated by ingestion of crushed embeda (morphine extended release with sequestered naltrexone): case report and the focused review of the literature. J Opioid Manag. 2010;6:300–303.
  • Katz N, Sun S, Fow L, et al. Efficacy and safety evaluation of an extended-release morphine sulfate formulation with sequestered naltrexone for the treatment of osteoarthritis. J Pain. 2008;9:41.
  • Severtson SG, Bartelson BB, Davis JM, et al. Reduced abuse, therapeutic errors, and diversion following reformulation of extended-release oxycodone in 2010. J Pain. 2013;14:1122–1130.
  • Butler SF, Cassidy TA, Chilcoat H, et al. Abuse rates and routes of administration of reformulated extended-release oxycodone: initial findings from a sentinel surveillance sample of individuals assessed for substance abuse treatment. J Pain. 2013;14:351–358.
  • Degenhardt L, Bruno R, Ali R, et al. The introduction of a potentially abuse deterrent oxycodone formulation: early findings from the Australian national opioid medications abuse deterrence (NOMAD) study. Drug Alcohol Depend. 2015;151:56–67.
  • Coplan PM, Chilcoat HD, Butler SF, et al. The effect of an abuse-deterrent opioid formulation (OxyContin) on opioid abuse-related outcomes in the postmarketing setting. Clin Pharmacol Ther. 2016;100:275–286.
  • Absalom A, Pledger D, Kong A. Adrenocortical function in critically ill patients 24 h after a single dose of etomidate. Anaesthesia. 1999;54:861–867.
  • Bruder EA, Ball IM, Ridi S, et al. Single induction dose of etomidate versus other induction agents for endotracheal intubation in critically ill patients. Cochrane Database Syst Rev. 2015 Jan 8;1:CD010225.
  • Pejo E, Zhou XJ, Shaukat Husain S, et al. Sedative-hypnotic binding to 11β-hydroxylase. Anesthesiology. 2016;125:943–951.
  • de Jong FH, Mallios C, Jansen C, et al. Etomidate suppresses adrenocortical function by inhibition of 11 beta-hydroxylation. J Clin Endocrinol Metab. 1984;59:1143–1147.
  • Pejo E, Cotten JF, Kelly EW, et al. In vivo and in vitro pharmacological studies of methoxycarbonyl-carboetomidate. Anesth Analg. 2012;115:297–304.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.