A Novel Faster-Acting, Dry Powder-Based, Naloxone Intranasal Formulation for Opioid Overdose

Tair Lapidot, Mohammed Bouhajib, Janice Faulknor, Shabaz Khan, Galia Temtsin Krayz, Carolina Abrutzky, Dalia Megiddo, Tair Lapidot, Mohammed Bouhajib, Janice Faulknor, Shabaz Khan, Galia Temtsin Krayz, Carolina Abrutzky, Dalia Megiddo

Abstract

Objective: To examine the pharmacokinetics and safety of FMXIN001, a new intranasal powder-based naloxone formulation, in comparison to Narcan® nasal liquid spray.

Methods: FMXIN001, was developed by blending drug microspheres with larger lactose monohydrate particles, that serve as diluent and carrier, as well as a disaggregating agent. Scanning electron microscopy and X-ray were used to characterize the formulation and in vitro deposition was investigated using a nasal cast. We compared the pharmacokinetics and safety of FMXIN001 versus Narcan® in two clinical trials: a pilot study with 14 healthy adults and a pivotal trial in 42 healthy adults (NCT04713709). The studies were open-label, single-dose, randomized, two-period, two-treatment, two-sequence crossover studies to assess the pharmacokinetics and safety of FMXIN001 versus Narcan® nasal spray.

Results: FMXIN001 comprises naloxone microspheres (5-30 μM) and lactose particles (40-240 μM). Upon in vitro testing, naloxone deposits mainly to the middle turbinates region and the upper part of the nasal cavity of a nasal cast. In human subjects, FMXIN001 produced significantly higher exposure at the initial time points of 4, 10, and 30 min, post-administration, compared to Narcan®. Both treatments were safe and well tolerated. FMXIN001, powder-based spray, results in similar overall exposure to Narcan®, but with more rapid absorption in the first 30 min.

Conclusions: FMXIN001 is expected to have a shorter onset of action for a more effective therapeutic intervention to manage opioid overdose. Rapid administration of naloxone in cases of opioid overdose is imperative, given the alarming increase in mortality rates.

Keywords: bioavailability; dry powder; lactose; naloxone microspheres; nasal spray.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
SEM image of FMXIN001 new dry powder-based formulation
Fig. 2
Fig. 2
Particle size distribution FMXIN001 new dry powder-based formulation
Fig. 3
Fig. 3
Mean of percentage deposition of naloxone microspheres dry powder formulation in each nasal cast, by region. Nasal cast used courtesy of Aptar Pharma, DTF medical and University of Tours
Fig. 4
Fig. 4
Mean Plasma Unconjugated Naloxone Concentration-Time Profile in a Linear Scale (A: n = 42 / B: n = 42)
Fig. 5
Fig. 5
Mean Plasma Unconjugated Naloxone Concentration-Time Profile in a Linear Scale (A: n = 42 / B: n = 42) – First 1-Hour

References

    1. Tiozzo Fasiolo L, Manniello MD, Tratta E, Buttini F, Rossi A, Sonvico F, et al. Opportunity and challenges of nasal powders: drug formulation and delivery. Eur J Pharm Sci. 2018;113:2–17. doi: 10.1016/j.ejps.2017.09.027.
    1. Tanaka A. Optimization of nasal drug absorption from powder formulations: the feasibility of controlling drug absorption by the use of pharmaceutical excipients. Yakugaku Zasshi. 2018;138(12):1467–1472. doi: 10.1248/yakushi.18-00157.
    1. Chen Q, Larochelle MR, Weaver DT, Lietz AP, Mueller PP, Mercaldo S, et al. Prevention of prescription opioid misuse and projected overdose deaths in the United States. JAMA Netw Open. 2019;2(2):e187621. doi: 10.1001/jamanetworkopen.2018.7621.
    1. Jones MR, Viswanath O, Peck J, Kaye AD, Gill JS, Simopoulos TT. A brief history of the opioid epidemic and strategies for pain medicine. Pain Ther. 2018;7(1):13–21. doi: 10.1007/s40122-018-0097-6.
    1. Volkow ND, Frieden TR, Hyde PS, Cha SS. Medication-assisted therapies — tackling the opioid-overdose epidemic. N Engl J Med. 2014;370(22):2063–2066. doi: 10.1056/NEJMp1402780.
    1. WHO (2018). Information sheet on opioid overdose. Retrieved from
    1. Dunne RB. Prescribing naloxone for opioid overdose intervention. Pain Management. 2018;8(3):197–208. doi: 10.2217/pmt-2017-0065.
    1. Rzasa Lynn R, Galinkin J. Naloxone dosage for opioid reversal: current evidence and clinical implications. Therapeutic Advances in Drug Safety. 2018;9(1):63–88. doi: 10.1177/2042098617744161.
    1. Djupesland PG, Skretting A. Nasal deposition and clearance in man: comparison of a bidirectional powder device and a traditional liquid spray pump. Journal of Aerosol Medicine and Pulmonary Drug Delivery. 2012;25(5):280–289. doi: 10.1089/jamp.2011.0924.
    1. Djupesland PG. Nasal drug delivery devices: characteristics and performance in a clinical perspective—a review. Drug Deliv and Transl Res. 2013;3(1):42–62. doi: 10.1007/s13346-012-0108-9.
    1. Hoekman J, Ray S, Aurora SK, Shrewsbury SB. The upper nasal space—a novel delivery route ideal for central nervous system drugs. US Neurology. 2020;16(1):25. doi: 10.17925/USN.2020.16.1.25.
    1. FDA Advisory Committee on the Most Appropriate Dose or Doses of Naloxone to Reverse the Effects of Life-threatening Opioid Overdose in the Community Settings (2016) Page 149. Retrieved from
    1. CDC Report (2020). Increase in Fatal Drug Overdoses Across the United States Driven by Synthetic Opioids Before and During the COVID-19 Pandemic. Retrieved from
    1. Jain MS, Lohare GB, Bari MM, Chavan RB, Barhate SD, Shah CB. Spray drying in pharmaceutical industry: a review. Research Journal of Pharmaceutical Dosage Forms and Technology. 2012;4:74–79.
    1. Fransén, N., Studies on a novel powder formulation for nasal drug delivery; digital comprehensive summaries of Uppsala dissertations from the Faculty of Pharmacy 81, Acta Universitatis Upsaliensis, Uppsala, p 22, 2008.
    1. Djupesland PG, Messina JC, Mahmoud RA. Role of nasal casts for in vitro evaluation of nasal drug delivery and quantitative evaluation of various nasal casts. Ther Deliv. 2020;11(8):485–495. doi: 10.4155/tde-2020-0054.
    1. Le Guellec S, Le Pennec D, Gatier S, Leclerc L, Cabrera M, Pourchez J, et al. Validation of anatomical models to study aerosol deposition in human nasal cavities. Pharm Res. 2014;31(1):228–237. doi: 10.1007/s11095-013-1157-6.
    1. Liu G, Zong G, Doty RL, Sun Q. Prevalence and risk factors of taste and smell impairment in a nationwide representative sample of the US population: a cross-sectional study. BMJ Open. 2016;6(11):e013246. doi: 10.1136/bmjopen-2016-013246.
    1. Rawal S, Hoffman HJ, Honda M, Huedo-Medina TB, Duffy VB. The Taste and Smell Protocol in the 2011–2014 US National Health and Nutrition Examination Survey (NHANES): Test–Retest Reliability and Validity Testing. Chem Percept. 2015;8(3):138–148. doi: 10.1007/s12078-015-9194-7.
    1. Simons FER, Prenner BM, Finn A. Efficacy and safety of desloratadine in the treatment of perennial allergic rhinitis. J Allergy Clin Immunol. 2003;111(3):617–622. doi: 10.1067/mai.2003.168.
    1. FDA Guidance: allergic rhinitis: developing drug products for treatment guidance for industry (2018). Retrieved from .
    1. Ryan SA, Dunne RB. Pharmacokinetic properties of intranasal and injectable formulations of naloxone for community use: a systematic review. Pain Management. 2018;8(3):231–245. doi: 10.2217/pmt-2017-0060.
    1. Scherließ R. Nasal formulations for drug administration and characterization of nasal preparations in drug delivery. Ther Deliv. 2020;11(3):183–191. doi: 10.4155/tde-2019-0086.
    1. Trenkel M, Scherließ R. Nasal powder formulations: in-vitro characterisation of the impact of powders on nasal residence time and sensory effects. Pharmaceutics. 2021;13(3):385. doi: 10.3390/pharmaceutics13030385.
    1. Lin Y-W, Wong J, Qu L, Chan H-K, Zhou Q. Powder production and particle engineering for dry powder inhaler formulations. CPD. 2015;21(27):3902–3916. doi: 10.2174/1381612821666150820111134.
    1. Rahimpour Y, Hamishehkar H. Lactose Engineering for Better Performance in Dry Powder Inhalers. Advanced Pharmaceutical Bulletin; eISSN 2251–7308 [Internet]. 2012 [cited 2021 Nov 17]; Available from:
    1. Hsu DJ, Chuang MH. In-vivo measurements of micrometer-sized particle deposition in the nasal cavities of Taiwanese adults. Aerosol Sci Technol. 2012;46(6):631–638. doi: 10.1080/02786826.2011.652749.
    1. Cady R. A novel intranasal breath-powered delivery system for sumatriptan: a review of technology and clinical application of the investigational product AVP-825 in the treatment of migraine. Expert Opin Drug Deliv. 2015;12(9):1565–1577. doi: 10.1517/17425247.2015.1060959.
    1. FDA Draft Guidance: Bioequivalence Studies with Pharmacokinetic Endpoints for Drugs Submitted Under an ANDA (2013). Retrieved from
    1. NARCAN® prescribing information (2015). Retrieved from
    1. Vasa DM, Buckner IS, Cavanaugh JE, Wildfong PLD. Improved flux of levodopa via direct deposition of solid microparticles on nasal tissue. AAPS PharmSciTech. 2017;18(3):904–912. doi: 10.1208/s12249-016-0581-4.

Source: PubMed

3
Se inscrever