434
Views
2
CrossRef citations to date
0
Altmetric
ORIGINAL RESEARCH

Sustained-Release Esketamine Based Nanoparticle-Hydrogel Delivery System for Neuropathic Pain Management

, , , , , , ORCID Icon & show all
Pages 1131-1143 | Received 08 Dec 2022, Accepted 21 Feb 2023, Published online: 07 Mar 2023

References

  • Campbell JN, Meyer RA. Mechanisms of neuropathic pain. Neuron. 2006;52(1):77–92. doi:10.1016/j.neuron.2006.09.021
  • Meacham K, Shepherd A, Mohapatra DP, Haroutounian S. Neuropathic pain: central vs. peripheral mechanisms. Curr Pain Headache Rep. 2017;21(6):28. doi:10.1007/s11916-017-0629-5
  • IsHak WW, Wen RY, Naghdechi L, et al. Pain and depression: a systematic review. Harv Rev Psychiatry. 2018;26(6):352–363. doi:10.1097/HRP.0000000000000198
  • Bannister K, Sachau J, Baron R, Dickenson AH. Neuropathic pain: mechanism-based therapeutics. Annu Rev Pharmacol Toxicol. 2020;60:257–274. doi:10.1146/annurev-pharmtox-010818-021524
  • Rasmussen-Barr E, Held U, Grooten WJ, et al. Non-steroidal anti-inflammatory drugs for sciatica. Cochrane Database Syst Rev. 2016;10(10):CD012382. doi:10.1002/14651858.CD012382
  • Garland EL. Treating chronic pain: the need for non-opioid options. Expert Rev Clin Pharmacol. 2014;7(5):545–550. doi:10.1586/17512433.2014.928587
  • Gangadhar M, Mishra RK, Sriram D, Yogeeswari P. Future directions in the treatment of neuropathic pain: a review on various therapeutic targets. CNS Neurol Disord Drug Targets. 2014;13(1):63–81. doi:10.2174/18715273113126660192
  • Attal N, Bouhassira D. Translational neuropathic pain research. Pain. 2019;160(Suppl 1):S23–S28. doi:10.1097/j.pain.0000000000001522
  • Yang Y, Maher DP, Cohen SP. Emerging concepts on the use of ketamine for chronic pain. Expert Rev Clin Pharmacol. 2020;13(2):135–146. doi:10.1080/17512433.2020.1717947
  • Robinson B, Gu Q, Kanungo J. Antidepressant actions of ketamine: potential role of l-type calcium channels. Chem Res Toxicol. 2021;34(5):1198–1207. doi:10.1021/acs.chemrestox.0c00411
  • Wagner LE, Gingrich KJ, Kulli JC, Yang J. Ketamine blockade of voltage-gated sodium channels: evidence for a shared receptor site with local anesthetics. Anesthesiology. 2001;95(6):1406–1413. doi:10.1097/00000542-200112000-00020
  • Lavender E, Hirasawa-Fujita M, Domino EF. Ketamine’s dose related multiple mechanisms of actions: dissociative anesthetic to rapid antidepressant. Behav Brain Res. 2020;390:112631. doi:10.1016/j.bbr.2020.112631
  • Porter SB, Schwenk ES. Ketamine and remote hyperalgesia. Minerva Anestesiol. 2018;84(4):432–433. doi:10.23736/S0375-9393.18.12521-1
  • Luginbühl M, Gerber A, Schnider TW, Petersen-Felix S, Arendt-Nielsen L, Curatolo M. Modulation of remifentanil-induced analgesia, hyperalgesia, and tolerance by small-dose ketamine in humans. Anesth Analg. 2003;96(3):726–732. doi:10.1213/01.ANE.0000048086.58161.18
  • Niesters M, Martini C, Dahan A. Ketamine for chronic pain: risks and benefits. Br J Clin Pharmacol. 2014;77(2):357–367. doi:10.1111/bcp.12094
  • Culp C, Kim HK, Abdi S. Ketamine use for cancer and chronic pain management. Front Pharmacol. 2021;11:599721. doi:10.3389/fphar.2020.599721
  • Kamp J, van Velzen M, Aarts L, Niesters M, Dahan A, Olofsen E. Stereoselective ketamine effect on cardiac output: a population pharmacokinetic/pharmacodynamic modelling study in healthy volunteers. Br J Anaesth. 2021;127(1):23–31. doi:10.1016/j.bja.2021.02.034
  • Pickering G, Morel V, Micallef J. Kétamine et douleur chronique: une revue narrative de son efficacité et sécurité [Ketamine and chronic pain: a narrative review of its efficacy and its adverse events]. Therapie. 2018;73(6):529–539. doi:10.1016/j.therap.2018.06.001
  • Cvrcek P. Side effects of ketamine in the long-term treatment of neuropathic pain. Pain Med. 2008;9(2):253–257. doi:10.1111/j.1526-4637.2007.00314.x
  • Zhou YL, Yang QQ, Yan YY, Zhu C, Zhang L, Tang JB. Localized delivery of miRNAs targets cyclooxygenases and reduces flexor tendon adhesions. Acta Biomater. 2018;70:237–248. doi:10.1016/j.actbio.2018.01.047
  • Dixon WJ. Efficient analysis of experimental observations. Annu Rev Pharmacol Toxicol. 1980;20:441–462. doi:10.1146/annurev.pa.20.040180.002301
  • Wang J, Huang J, Yang S, et al. Pharmacokinetics and safety of esketamine in Chinese patients undergoing painless gastroscopy in comparison with ketamine: a randomized, open-label clinical study. Drug Des Devel Ther. 2019;13:4135–4144. doi:10.2147/DDDT.S224553
  • Mei XP, Zhang H, Wang W, et al. Inhibition of spinal astrocytic c-Jun N-terminal kinase (JNK) activation correlates with the analgesic effects of ketamine in neuropathic pain. J Neuroinflammation. 2011;8(1):6. doi:10.1186/1742-2094-8-6
  • Mei X, Wang W, Wang W, et al. Inhibiting astrocytic activation: a novel analgesic mechanism of ketamine at the spinal level? J Neurochem. 2009;109(6):1691–1700. doi:10.1111/j.1471-4159.2009.06087.x
  • Vranken JH, Troost D, de Haan P, et al. Severe toxic damage to the rabbit spinal cord after intrathecal administration of preservative-free S(+)-ketamine. Anesthesiology. 2006;105(4):813–818. doi:10.1097/00000542-200610000-00028
  • Wang T, Hurwitz O, Shimada SG, et al. Anti-nociceptive effects of bupivacaine-encapsulated PLGA nanoparticles applied to the compressed dorsal root ganglion in mice. Neurosci Lett. 2018;668:154–158. doi:10.1016/j.neulet.2018.01.031
  • Hayek SM, Shah A. Nerve blocks for chronic pain. Neurosurg Clin N Am. 2014;25(4):809–817. doi:10.1016/j.nec.2014.07.006
  • Esposito MF, Malayil R, Hanes M, Deer T. Unique characteristics of the dorsal root ganglion as a target for neuromodulation. Pain Med. 2019;20(Suppl 1):S23–S30. doi:10.1093/pm/pnz012
  • Berta T, Qadri Y, Tan PH, Ji RR. Targeting dorsal root ganglia and primary sensory neurons for the treatment of chronic pain. Expert Opin Ther Targets. 2017;21(7):695–703. doi:10.1080/14728222.2017.1328057
  • Colombo E, Farina C. Astrocytes: key regulators of neuroinflammation. Trends Immunol. 2016;37(9):608–620. doi:10.1016/j.it.2016.06.006
  • Chen YL, Feng XL, Cheung CW, Liu JA. Mode of action of astrocytes in pain: from the spinal cord to the brain. Prog Neurobiol. 2022;219:102365. doi:10.1016/j.pneurobio.2022.102365
  • Sinner B, Graf BM. Ketamine. Handb Exp Pharmacol. 2008;(182):313–333. doi:10.1007/978-3-540-74806-9_15
  • Michaelis EK. Two different families of NMDA receptors in mammalian brain: physiological function and role in neuronal development and degeneration. Adv Exp Med Biol. 1993;341:119–128. doi:10.1007/978-1-4615-2484-7_11
  • Deng M, Chen SR, Pan HL. Presynaptic NMDA receptors control nociceptive transmission at the spinal cord level in neuropathic pain. Cell Mol Life Sci. 2019;76(10):1889–1899. doi:10.1007/s00018-019-03047-y
  • McDonald JW, Silverstein FS, Johnston MV. Neurotoxicity of N-methyl-D-aspartate is markedly enhanced in developing rat central nervous system. Brain Res. 1988;459(1):200–203. doi:10.1016/0006-8993(88)90306-x
  • Zeilhofer HU, Swandulla D, Geisslinger G, Brune K. Differential effects of ketamine enantiomers on NMDA receptor currents in cultured neurons. Eur J Pharmacol. 1992;213(1):155–158. doi:10.1016/0014-2999(92)90248-3
  • Haeseler G, Tetzlaff D, Bufler J, et al. Blockade of voltage-operated neuronal and skeletal muscle sodium channels by S (+)- and R(-)-ketamine. Anesth Analg. 2003;96(4):1019–1026. doi:10.1213/01.ANE.0000052513.91900.D5
  • Hayashi Y, Kawaji K, Sun L, et al. Microglial Ca (2+)-activated K (+) channels are possible molecular targets for the analgesic effects of S-ketamine on neuropathic pain. J Neurosci. 2011;31(48):17370–17382. doi:10.1523/JNEUROSCI.4152-11.2011
  • Ardalan M, Elfving B, Rafati AH, et al. Rapid effects of S-ketamine on the morphology of hippocampal astrocytes and BDNF serum levels in a sex-dependent manner. Eur Neuropsychopharmacol. 2020;32:94–103. doi:10.1016/j.euroneuro.2020.01.001
  • Lalani J, Patil S, Kolate A, Lalani R, Misra A. Protein-functionalized PLGA nanoparticles of lamotrigine for neuropathic pain management. AAPS PharmSciTech. 2015;16(2):413–427. doi:10.1208/s12249-014-0235-3
  • Kim SI, Shin J, Tran Q, et al. Application of PLGA nanoparticles to enhance the action of duloxetine on microglia in neuropathic pain. Biomater Sci. 2021;9(18):6295–6307. doi:10.1039/d1bm00486g
  • Babaie S, Taghvimi A, Hong JH, Hamishehkar H, An S, Kim KH. Recent advances in pain management based on nanoparticle technologies. J Nanobiotechnology. 2022;20(1):290. doi:10.1186/s12951-022-01473-y
  • Vasvani S, Kulkarni P, Rawtani D. Hyaluronic acid: a review on its biology, aspects of drug delivery, route of administrations and a special emphasis on its approved marketed products and recent clinical studies. Int J Biol Macromol. 2020;151:1012–1029. doi:10.1016/j.ijbiomac.2019.11.066
  • Zhou Y, Peng Z, Seven ES, Leblanc RM. Crossing the blood-brain barrier with nanoparticles. J Control Release. 2018;270:290–303. doi:10.1016/j.jconrel.2017.12.015
  • Feng J, Lepetre-Mouelhi S, Gautier A, et al. A new painkiller nanomedicine to bypass the blood-brain barrier and the use of morphine. Sci Adv. 2019;5(2):eaau5148. doi:10.1126/sciadv.aau5148
  • da Silva A, Lepetre-Mouelhi S, Couvreur P. Micro- and nanocarriers for pain alleviation. Adv Drug Deliv Rev. 2022;187:114359. doi:10.1016/j.addr.2022.114359
  • Hashimoto K. Detrimental side effects of repeated ketamine infusions in the brain. Am J Psychiatry. 2016;173(10):1044–1045. doi:10.1176/appi.ajp.2016.16040411
  • Vranken JH, Troost D, Wegener JT, Kruis MR, van der Vegt MH. Neuropathological findings after continuous intrathecal administration of S(+)-ketamine for the management of neuropathic cancer pain. Pain. 2005;117(1–2):231–235. doi:10.1016/j.pain.2005.06.014
  • Lin CS, Lin YC, Lao HC, Chen CC. Interventional treatments for postherpetic neuralgia: a systematic review. Pain Physician. 2019;22(3):209–228. doi:10.36076/ppj/2019.22.209
  • Collins KL, Russell HG, Schumacher PJ, et al. A review of current theories and treatments for phantom limb pain. J Clin Invest. 2018;128(6):2168–2176. doi:10.1172/JCI94003
  • Bobo D, Robinson KJ, Islam J, Thurecht KJ, Corrie SR. Nanoparticle-based medicines: a review of FDA-approved materials and clinical trials to date. Pharm Res. 2016;33(10):2373–2387. doi:10.1007/s11095-016-1958-5
  • Zhao N, Francis NL, Calvelli HR, Moghe PV. Microglia-targeting nanotherapeutics for neurodegenerative diseases. APL Bioeng. 2020;4(3):30902. doi:10.1063/5.0013178