throbber
Drug Deliv. and Transl. Res. (2013) 3:63–74
`DOI 10.1007/s13346-012-0092-0
`
`REVIEW ARTICLE
`
`A response to the opioid overdose epidemic:
`naloxone nasal spray
`
`Daniel P. Wermeling
`
`Published online: 1 August 2012
`# Controlled Release Society 2012
`
`Abstract Opioid overdose morbidity and mortality is rec-
`ognized to have epidemic proportions. Medical and public
`health agencies are adopting opioid harm reduction strate-
`gies to reduce the morbidity and mortality associated with
`overdose. One strategy developed by emergency medical
`services and public health agencies is to deliver the opioid
`antidote naloxone injection intranasally to reverse the
`effects of opioids. Paramedics have used this route to quick-
`ly administer naloxone in a needle-free system and avoiding
`needlestick injuries and contracting a blood-born pathogen
`disease such as hepatitis or human immunodeficiency virus.
`Public health officials advocate broader lay person access
`since civilians are likely witnesses or first responders to an
`opioid overdose in a time-acute setting. The barrier to great-
`er use of naloxone is that a suitable and optimized needle-
`free drug delivery system is unavailable. The scientific basis
`for design and study of an intranasal naloxone product is
`described. Lessons from nasal delivery of opioid analgesics
`are applied to the consideration of naloxone nasal spray.
`
`Keywords Intranasal . Naloxone . Opioid . Overdose .
`Antidote
`
`Introduction
`
`In 2008, poisoning surpassed motor vehicle accidents as the
`leading cause of “injury deaths” in the USA [1]. Nearly
`90 % of poisoning deaths are caused by drugs. During the
`past three decades, the number of drug poisoning deaths
`increased sixfold from about 6,100 in 1980 to 36,500 in
`
`D. P. Wermeling (*)
`University of Kentucky College of Pharmacy,
`789 South Limestone Street,
`Lexington, KY 40536-0596, USA
`e-mail: dwermel@uky.edu
`
`2008. Of the 36,500 drug poisoning deaths in 2008, 14,800
`involved prescription opioid analgesics. Approximately
`3,000 deaths also involved heroin overdose. In 2008, the
`overall death rate in the USA was 4.8 per 100,000 for
`nonmedical use of prescription opioids [2, 3].
`The opioid overdose crisis is a worldwide phenomenon
`crossing sovereign, cultural, and socio-economic bound-
`aries. The US Centers for Disease Control considers pre-
`scription drug overdose in epidemic proportions,
`in
`particular, the morbidity and mortality associated with use,
`abuse, and misuse of prescription opioids [4]. Hospitaliza-
`tions from prescription opioid poisoning increased by over
`50 % from 1999 to 2006, paralleling the increased prescrib-
`ing of these medications for the treatment of pain [5, 6].
`Although many deaths are associated with drug abuse, there
`is also a growing trend of therapeutic misadventures for pain
`patients prescribed powerful analgesics, including opioids.
`Chronic cancer and nonmalignant pain pharmacotherapy
`regimens frequently involve combinations of medications
`with additive or synergistic central nervous system depres-
`sion adverse effects.
`Injection drug use, principally heroin, is one of the most
`significant correlates to opiate use mortality. Eurasia, Aus-
`tralia, Canada, Italy, and Great Britain, among others, all
`describe significant injection drug use populations that ex-
`perience drug overdose with similar rates of mortality re-
`gardless of socioeconomic status [7–12].
`Government and nongovernment public health agencies,
`the pharmaceutical industry, and others are adopting preven-
`tion and intervention strategies in an attempt to reduce
`opioid overdose mortality. One “harm-reduction” strategy
`has been to provide education and training on opioid over-
`dose recognition and emergency treatment to addicts and
`their close daily contacts [13]. In addition, the addict and
`their loved ones are trained to rescue breathe, call emergen-
`cy medical services, and to administer the opioid antidote
`naloxone.
`
`Nalox1016
`Nalox-1 Pharmaceuticals, LLC
`Page 1 of 12
`
`

`

`64
`
`Drug Deliv. and Transl. Res. (2013) 3:63–74
`
`Naloxone is the drug of choice to reverse respiratory and
`central nervous system depression caused by opioid over-
`dose [14]. Naloxone injection has been marketed in the
`USA for 41 years, initially under the trade name Narcan®.
`Naloxone hydrochloride (HCl), known chemically as 17-
`Allyl-4,5α,-epoxy-3, 14-dihydroxymorphinan-6-one hydro-
`chloride, is a potent mu-receptor antagonist. It has subse-
`quently become a multisource prescription generic drug
`manufactured by International Medication Systems, Limited
`and Hospira, Inc. [15, 16]. Ampoules of naloxone injection
`are also available in many countries. The injection is avail-
`able in two strengths, 0.4 and 1.0 mg/mL. Naloxone is a
`standard drug carried by emergency services personnel in
`ambulances and medication kits for reversal of suspected
`opioid overdose, whether accidental or intentional. Hospital
`emergency departments also use this medication routinely
`for this purpose. The initial adult dose of naloxone in known
`or suspected narcotic overdose is 0.4–2 mg, which may be
`repeated to a total dose of 10 mg. The current formulations
`of naloxone are approved for intravenous (IV), intramuscu-
`lar (IM), and subcutaneous (SC) administration, with IV
`being the recommended route [17, 18]. Naloxone is also
`indicated as a reversal agent when the effects of therapeutic
`use of opioids are no longer medically necessary, such as in
`reversal of opioid effects in general anesthesia [15, 16].
`Lastly, naloxone is coformulated with buprenorphine as an
`oral product providing an abuse-deterrent formulation for
`opioid maintenance in opioid dependent patients.
`In the last several years, the emergency medical systems
`(EMS) community in the USA and elsewhere has developed
`an interest in administering naloxone in a needleless system
`via the intranasal (IN) route. Some EMS programs have now
`moved toward intranasal administration of naloxone since
`many of the patients needing naloxone are injection drug
`users; 80 % of the injection drug user population in large
`metropolitan areas is hepatitis C positive or HIV positive.
`For example, the Denver and San Francisco EMS uses this
`drug administration technique as standard of care to prevent
`needlestick injuries to emergency medical technicians [17,
`19, 20].
`Some EMS programs have developed a system using
`existing technologies of an approved drug and an existing
`medical device to administer this opioid antidote, albeit in a
`non-Food and Drug Administration (FDA)-approved man-
`ner [19, 20]. This has been accomplished by using the
`injectable formulation (1 mg/mL) and administering 1 mL
`per nostril via a marketed nasal atomizer/nebulizer device.
`The system combines an FDA-approved naloxone injection
`product (with a Luer fitted tip, no needles) with a marketed
`[510(k) exempt] medical device called the Mucosal Atom-
`ization Device (MAD™ Nasal, Wolfe Tory Medical, Inc.).
`This initiative is consistent with the US Needlestick Safety
`and Prevention Act (Public Law 106–430) [21–25].
`
`The EMS programs recognize limitations of this system,
`one limitation being that it is not assembled and ready to
`use. Although this administration mode appears to be effec-
`tive in reversing narcosis, the formulation is not concentrat-
`ed for retention in the nasal cavity. The 1 mL delivery
`volume per naris is larger than that generally utilized for
`intranasal drug administration. Therefore, there is loss of
`drug from the nasal cavity, due either to drainage into the
`nasopharynx or externally from the nasal cavity. An im-
`provement would be to design a ready-to-use product spe-
`cifically optimized, concentrated, and formulated for nasal
`delivery.
`The drug abuse treatment and overdose prevention com-
`munities worldwide have also recognized the desire for a
`needle-free system for naloxone delivery [26]. Clients at
`needle-exchange centers provide kits containing naloxone,
`with either needles or nasal spray atomizers, and instruc-
`tions for use. Such programs are well-described in the USA
`and Great Britain, commonly operating in conjunction with
`needle-exchange programs [27–33]. In May 2012, the Brit-
`ish Advisory Council on the Misuse of Drugs published a
`report advocating for greater distribution of naloxone and
`training for administration [34]. The Council reiterates that
`naloxone is safe and effective, that there is evidence that
`take-home naloxone can be effective for reversing heroin
`overdoses. Cost-effectiveness of these programs is still be-
`ing assessed.
`An unmet medical need exists to provide greater access
`to the opioid antidote naloxone. A significant barrier to this
`goal is that naloxone is only available as an injection for IV,
`IM, or SC administration. A needleless system that integra-
`tes a concentrated formulation and a nasal delivery device
`would help satisfy this unmet need.
`
`Nasal physiology, drug, and formulation considerations
`for nasal delivery
`
`Nasal physiology
`
`Intranasal sprays of medication intended for systemic
`drug absorption are generally designed to target
`the
`turbinates on the medial wall of the nasal cavity. The
`turbinates serve as a baffle in which inspired air is
`humidified and filtered. This region of the nasal cavity
`is covered with a thin mucus layer, a monolayer ciliated
`epithelium, with an abundant underlying blood supply.
`These conditions are ideal
`to permit passive diffusion
`(transcellular) of medications with certain chemical
`characteristics across cell membranes and into the
`bloodstream. Some medications also transit to the blood
`stream by passing through the tight-cell
`junctions be-
`tween cells (paracellular) [35].
`
`Nalox1016
`Nalox-1 Pharmaceuticals, LLC
`Page 2 of 12
`
`

`

`Drug Deliv. and Transl. Res. (2013) 3:63–74
`
`65
`
`To reach the turbinates, the nasal spray device must be
`inserted fully into the nasal vestibule with the atomizer tip
`placed at the nasal valve, and then aimed laterally toward
`the turbinates. Activation of the device ejects the liquid as
`an atomized spray or plume. The bulk of the spray impacts
`the anterior and inferior portions of the nasal cavity as a
`function of straight-line impact of particles greater than
`10 μm in size [36–38]. The smallest particles, less than
`10 μm in size, may be carried by air currents more superi-
`orly in the nasal cavity and impact on the superior turbinate
`and possibly reach the olfactory region and nerve. There is
`substantial evidence in animals, and some evidence in man,
`that the olfactory nerve can absorb or actively transport
`medications to the central nervous system via the olfactory
`bulb (nose to brain theory). Differences in animal and hu-
`man nasal apparatus anatomy, and certain characteristics of
`the medication, seem to play roles as to whether medication
`is transported to the brain via this mechanism and if a
`pharmacologic effect is observed [39–41].
`Under ideal conditions, most medication is absorbed
`from the nasal cavity and into the bloodstream within 15–
`20 min, thus generally avoiding the gut first-pass metabo-
`lism [35, 36]. Medication remaining in the nasal cavity
`beyond this time is subject to elimination via various en-
`zyme systems present within the nasal mucus and by swal-
`lowing. A second absorption phase (oral) can be observed
`with nasally administered medications having incomplete
`nasal absorption that are not subject to high first pass gut
`metabolism.
`Nasal physiologic changes during pathologic conditions,
`such as polyposis and allergic and vasomotor rhinitis, could
`theoretically alter the biopharmaceutics of intranasal medi-
`cations intended for systemic drug administration [35, 36,
`42, 43]. Physical obstruction of the nasal passage(s) due to
`prior trauma and subsequent deflection of the passageways
`is another possibility. Concurrent use of medications with
`vasoconstriction or vasodilation properties may also affect
`drug absorption. Lastly, increases in mucus production and
`changes in mucociliary clearance rates could affect bioavail-
`ability [35–38].
`regulatory agencies have required
`Pharmaceutical
`studies of the effect of rhinitis on nasal drug delivery
`biopharmaceutics [43, 44]. It has been demonstrated that
`there is a lack of effect of nasal mucosal inflammation
`on the absorption of hydromorphone, butorphanol,
`buserelin, and triamcinolone acetonide—an exception
`was reported for desmopressin. Inconsistent results have
`been reported on the biopharmaceutical disposition of
`these medications when pretreatment with oral or topical
`decongestant was administered [37, 43, 44]. Small but
`statistically measureable changes in rate or extent of
`absorption have been reported when decongestants were
`co-administered.
`
`Drug and formulation considerations
`
`Many intranasal delivery products are designed to serve
`certain purposes or unmet medical needs. Clearly, a nasal
`spray can remove the needle from drug administration, as is
`the case with the conversion of the protein calcitonin from a
`daily injection to a nasal spray. Furthermore, beyond just
`removing the needle from delivery, intranasal products are
`designed for rapid action, such as those products designed to
`treat seizures (benzodiazepines) migraine headache (suma-
`triptan, butorphannol, zolmitriptan, dihydroergotamine) or
`pain in general (fentanyl, hydromorphone) [45–49].
`Successful intranasal products satisfied several design
`fundamentals necessary for intranasal delivery. The proper-
`ties of the drug generally follow these characteristics:
`& Mass of 20 mg or less per dose
`& Molecular weight <1,000 Da
`& Excellent water solubility
`&
`Ionization and pH control of aqueous solutions
`& Osmolality—isotonic to slightly hypertonic
`&
`Stability in processing and storage
`& Compatibility container closure and sprayer components
`& Compatible with excipients (buffers, antioxidants, cosol-
`vents, etc.)
`Physical–chemical properties of the candidates must also
`be considered. Water solubility is important for formulation
`considerations. Log P, derived from the octanol/water parti-
`tion coefficient, is a surrogate for lipophilicity and potential
`for compounds to diffuse across biologic membranes. Suc-
`cessful intranasal medications tend to be water soluble and
`have sufficient lipophilic character to readily cross biologic
`membranes [35, 36].
`The dose must have sufficient solubility to be adminis-
`tered in approximately 100–200 μL (one spray per naris) of
`solution. The nasal cavity can retain 100–150 μL without
`causing immediate runoff out the front of the nose or down
`the nasopharynx [35, 36]. Additional solubilization strate-
`gies may be necessary including the use of organic cosol-
`vents, excipients such as cyclodextrins or other agents to
`from water-soluble inclusion complexes, or preparation of
`emulsions. Permeation enhancers may also be necessary to
`enhance drug penetration through cell membranes [50].
`Design of the formulation must account for other factors
`as well. It is useful to design the formulation to be isotonic
`to slightly hypertonic to optimize absorption and tolerabili-
`ty. Viscosity enhancing agents such as methylcellulose and
`others can promote retention in the nasal cavity by slowing
`the ciliary movement of mucus [35, 36]. Surfactants or
`polymers can be employed to enhance transmembrane per-
`meation [50]. Lastly, the drug and formulation have to be
`stable in the device during processing, i.e., sterilization and
`storage, and thus may require stabilizers.
`
`Nalox1016
`Nalox-1 Pharmaceuticals, LLC
`Page 3 of 12
`
`

`

`66
`
`Drug Deliv. and Transl. Res. (2013) 3:63–74
`
`The choice of delivery device for the medication is an-
`other critical consideration. Squeeze bottles are available but
`have no metering device appropriate to administer potent
`systemic medications. Multidose bottles provide a metered
`dose and are available for chronic drug administration. A
`standard syringe with Luer fitting to accept a nasal atomizer
`has been used to draw up and administer injection-based
`drug solutions into the nasal cavity for opiate overdoses,
`acute pain, and to deliver midazolam injection to the nasal
`cavity of a seizing patient. Unit–dose devices similar to
`those used for migraine treatment are also available and
`being used in development of benzodiazepine nasal spray
`products. The choice of device depends upon factors such as
`intended clinical use, setting, stability with the drug and
`formulation, among others [35, 36].
`Ideally, a well-designed formulation must not induce
`localized toxicity with acute or chronic use. For example,
`chronic vasoconstriction, irritation, or inflammation can in-
`duce tissue damage including ulceration, epistaxis, nasal–
`septal defects, and fistulae. Formulations should not cause
`damage to the cilia. Chronic, or daily use, of an irritating
`product could lead to more serious sequallae from nasal
`delivery [36].
`
`Properties of naloxone hydrochloride dihydrate
`
`Naloxone is supplied as naloxone HCl dihydrate. The em-
`pirical formula of naloxone HCl dihydrate is C19H22ClNO4,
`2H2O and its molecular weight is 399.9 g/mol. The struc-
`tural formula for naloxone is described in Fig. 1a [15, 16].
`Naloxone HCl has a physical form of white, or almost
`white, crystalline powder, and is hygroscopic. Its melting
`point is 200–205 °C. Naloxone HCl is freely soluble in
`water and 96 % alcohol, but practically insoluble in toluene.
`It is also slightly soluble in alcohol and practically insoluble
`in ether or chloroform. The dissociation constant pKa of
`naloxone is 7.9 and the log P is 1.92. These physical–
`chemical characteristics suggest a naloxone aqueous solu-
`tion is likely feasible [36].
`Given its high water solubility, naloxone HCl is an ex-
`cellent candidate to consider for intranasal delivery and
`satisfies many criteria necessary for this route. Naloxone is
`a high first-pass metabolism medication; oral bioavailability
`is reported to be ≤5 %. The parenteral dose is 2 mg or less; it
`is highly potent when injected [14–16].
`Nasal sprays of compounds chemically related to nalox-
`one have been described. Medications studied include the
`opioid antagonist naltrexone, and the opioid agonists hydro-
`morphone and butorphanol (Fig. 1b–d). Examination of the
`formulation methods and human biopharmaceutics of other
`chemically related compounds will be instructive for con-
`siderations of a naloxone nasal spray [36, 45, 48, 49].
`
`Translation of intranasal opioid formulations
`to naloxone nasal spray
`
`Naturally occurring and semi-synthetic opioids and antago-
`nists share the core structure of thebaine [51]. The addition
`of functional groups to the core structure imparts different
`physical–chemical and pharmacologic properties. However,
`the molecular weights and pKa values are roughly similar
`and the acid salts tend to be freely soluble in water. Certain
`cogeners have relatively higher log P as compared to others
`and so transmembrane delivery may be more accommoda-
`tive for these molecules [36]. Functional group changes also
`impart pharmacological properties of being a full mu-
`receptor agonist, partial agonist, or antagonist. Similarly,
`these changes may affect the degree of first-pass metabolism
`upon oral administration. Many of these cogeners are also
`quite potent—doses may range from 0.5 to 10 mg.
`A recent manuscript has provided a comprehensive re-
`view on intranasal delivery of opioids [36]. Tables on phys-
`ical–chemical properties and biopharmaceutics of various
`agents are provided. Data for naltrexone, hydromorphone,
`and butorphanol are provided (Table 1). These molecules
`may be of particular interest since the intravenous and
`intranasal dosing appears to be similar and the molecules
`share chemical characteristics to naloxone. Table 2 provides
`a comparison of the biopharmaceutics of nasally adminis-
`tered naltrexone, hydromorphone and butorphanol. Interest-
`ingly, the clinical doses of these drugs (dose normalized for
`naltrexone) are comparable to that of naloxone injection.
`Concentration–time profiles for naltrexone, hydromor-
`phone, and butorphanol are provided in Fig. 2 (dose nor-
`malized). These data may suggest the likely outcome of an
`intranasally delivered concentrated solution of naloxone
`HCl. A 2 mg nasal solution dose will likely have a Cmax
`of 3–5 ng/mL and a tmax of approximately 20 min, similar to
`naltrexone and hydromorphone [48]. The greater absorption
`of butorphanol is likely related to its higher Log P and
`ability to diffuse across biologic membranes.
`
`Biopharmaceutics of intranasal naloxone
`
`The nasal administration of 3H-naloxone to anesthetized
`male rats (n03/group) at a single dose of 30 μg (0.1 mg/
`kg, based on their average weight of 270 g/rat) in 0.1 mL
`was compared to a similar dose in 0.1 mL given by the
`intravenous and intraduodenal routes [52]. Nasally admin-
`istered naloxone was rapidly and completely absorbed
`(Fig. 3). The plasma elimination half-life of radioactivity
`was found to be 40–45 min. The nasal bioavailability for
`naloxone calculated from the ratio of the AUC0- INF (na-
`sal/intravenous 01,517.5/1,498.7 ng·h/mL) was 101 %.
`The intraduodenal bioavailability for naloxone was only
`1.5 % (intraduodenal/intravenous022.0/1,498.7 ng·h/mL).
`
`Nalox1016
`Nalox-1 Pharmaceuticals, LLC
`Page 4 of 12
`
`

`

`Drug Deliv. and Transl. Res. (2013) 3:63–74
`
`67
`
`Fig. 1 Chemical structure of
`naloxone (a), naltrexone (b),
`hydromorphone (c) and
`butorphanol (d) (Pub Chem,
`pubchem.ncbi.nlm.nih.gov)
`
`These results established the nasal route for the adminis-
`tration of naloxone in rats was equivalent to the parenteral
`route.
`The pharmacokinetic properties of intranasal naloxone
`in humans are not well described. A literature review
`found there are no papers describing the human phar-
`macokinetics of intranasal naloxone using what might
`be considered a highly concentrated nasal solution for-
`mulation. One paper describes pharmacokinetics of nasal
`administration of commercial injectable naloxone in man
`[53]. The intranasal formulation employed was an injec-
`tion in which 0.8 mg was administered in a volume of
`2 mL (1 mL/naris) even though it is commonly under-
`stood the nasal cavity can retain only 100–200 μL per
`naris. The study compared intravenous and intramuscu-
`lar administration to intranasal administration. The
`reported intranasal bioavailability of 4 % is dependent
`upon this non-optimized delivery volume, as it can be
`
`assumed that much of the medication ran away from the
`site of absorption. Therefore,
`the report may be mis-
`leading regarding predicting nasal naloxone absorption
`in humans using a solution concentrated and designed
`to accommodate the absorptive surface of the naris.
`A recent publication provides a more relevant exam-
`ination of the possible pharmacokinetic profile of a
`formulated naloxone nasal spray. The study provides
`information regarding the nasal absorption of naloxone
`in humans from a powder obtained from crushed Sub-
`oxone® (buprenorphine and naloxone sublingual tablets)
`[54]. After administration of 2 mg (naloxone) nasal
`powder, the absolute bioavailability was 30 %, with a
`tmax of 20 min and a Cmax of 1.6 ng/mL. A powder will
`behave somewhat differently than a solution adminis-
`tered intranasally because dissolution must occur during
`the time that
`the naloxone powder is present
`in the
`nasal cavity and before the ciliated epithelia sweep the
`
`Table 1 Chemical properties of naloxone, naltrexone, hydromor-
`phone, and butorhpanol [36]
`
`Table 2 Biopharmaceutics of intranasal naltrexone, hydromorphone
`and butorphanol [48, 49]
`
`Drug name
`
`Molecular weight
`
`pKa
`
`Log P
`
`Drug and dose
`
`Cmax
`(ng/mL)
`
`Tmax
`(minutes)
`
`Bioavailability (%)
`
`Naloxone
`Naltrexone
`Hydromorphone
`Butorphanol tartrate
`
`327
`341
`285
`327
`
`7.9
`8.1
`8.5
`8.6
`
`2
`1.9
`1.8
`3.77
`
`14.9
`Naltrexone HCl, 10 mg
`Hydromorphone HCl, 2 mg 3.5
`Butorphanol tartrate, 2 mg
`5.5
`
`22
`20
`10
`
`600 (to oral)
`50–60
`60–70
`
`Nalox1016
`Nalox-1 Pharmaceuticals, LLC
`Page 5 of 12
`
`

`

`68
`
`Drug Deliv. and Transl. Res. (2013) 3:63–74
`
`Fig. 2 Concentration–time profiles for naltrexone (10 mg intranasal and 50 mg oral; a), hydromorphone (2 mg IV and 1 and 2 mg intranasal; b), and
`butorphanol (1 and 2 mg intranasal; c) [48, 49]
`
`product posterior to be swallowed. A solution of nalox-
`one may have a slightly higher bioavailability and Cmax.
`Figure 4 presents a concentration–time profile for 0.5
`and 2.0 mg of intranasal naloxone powder from a Sub-
`oxone® tablet.
`The nasal delivery of naloxone powder, and subse-
`quent exposure, can be used as a surrogate marker as to
`the potential for naloxone nasal spray to reverse narco-
`sis. The essential question remains as to whether nalox-
`one nasal spray will produce an exposure that
`is
`clinically relevant. The Dowling paper suggests that
`naloxone nasal spray will produce a systemic exposure
`that is clinically relevant if compared to the profiles of
`intravenous and intramuscular administration. Figure 5
`presents the concentration–time profile for 0.8 mg nal-
`oxone, a clinically relevant dose within the naloxone
`prescribing label, for intravenous and intramuscular ad-
`ministration. The intramuscular administration is partic-
`ularly relevant given the absorption phase. A Cmax of
`about 1.5 ng/mL and a tmax 12 min were derived from
`this route of administration [53]. These data are not
`greatly different from the profile demonstrated from
`intranasal naloxone powder administration.
`
`Clinical experience with intranasal naloxone in reversing
`narcosis
`
`Emergency medicine practitioners and drug abuse treatment
`and prevention clinicians have considerable experience with
`administering naloxone injection for treatment of suspected
`opioid overdose. It appears the practice of nasal naloxone
`administration has outpaced the biopharmaceutic and clini-
`cal pharmacologic aspects typical of understanding the
`properties of a medication and delivery system. Regardless,
`the system appears to work and is further described.
`Nasal administration of naloxone was first reported by
`Loimer in 1992 [55]. Naloxone was studied using a 1 mg
`intranasal dose to identify potentially physically dependent
`opioid users. Withdrawal distress, pupillary response, pulse
`rate and blood pressure were recorded. Withdrawal symp-
`toms highly correlated with subject history and the presence
`of opioid metabolites in urine. The authors conclude that
`nasal delivery of naloxone is as effective as intravenous
`injection to identify physically dependent opioid users and
`could be useful in emergency medicine treatment.
`A second study by Loimer was conducted in 17 opioid-
`dependent patients to compare the efficacy of 1 mg of
`
`Nalox1016
`Nalox-1 Pharmaceuticals, LLC
`Page 6 of 12
`
`

`

`Drug Deliv. and Transl. Res. (2013) 3:63–74
`
`69
`
`Fig. 3 Mean plasma naloxone levels in rats after a single nasal (open
`circle), intravenous (filled circle), or intraduodenal (empty square) dose
`of 30 μg of 3H-naloxone [52]
`
`intranasal naloxone to intramuscular and intravenous nalox-
`one administration [56]. Withdrawal symptoms and vital
`sign changes were again used as endpoints at 1, 5, 15, 45,
`90, and 180 min after administration. The data demonstrated
`that intranasal administration had a more rapid onset and
`intensity of withdrawal as compared to intramuscular ad-
`ministration, but was not as rapid or as intense as intrave-
`nous administration. All endpoints returned to baseline by
`180 min in all groups.
`Naloxone is approved for use in the USA by IV, IM, or
`SC routes [14–16]. It is suggested that the onset of action of
`
`Fig. 4 Mean±SEM for plasma concentrations of naloxone in volun-
`teers, analyzed for up to 8 h after 0.5 or 2 mg dose of naloxone from
`Suboxone® tablets [54]
`
`Fig. 5 Concentration–time profile for 0.8 mg naloxone give IV and IM
`[53]
`
`the IV will be faster, so is preferred in emergency situations.
`However, obtaining IV access in the prehospital setting,
`especially among injection drug abusers can be time con-
`suming and difficult. Wanger conducted a study to deter-
`mine the actual onset of effect (defined as a respiratory rate
`of ≥10 breaths/minute) calculated from the time of arrival at
`the patient’s side using two approved injection routes [57].
`That is, this approach takes into account the time to set up
`and deliver the drug by the intended route.
`The study utilized naloxone injection, as used in nonhos-
`pital settings for presumed heroin overdoses. Patients were
`enrolled in series, rather than randomized. The first series
`treated patients with naloxone 0.4 mg IV followed by an
`additional 0.4 mg IV in the case of unsatisfactory response
`by 5 min (satisfactory response was ≥10 breaths/min). The
`second phase of the protocol treated patients with 0.8 mg
`naloxone SC. If a satisfactory response was not observed by
`5 min, the IV protocol was followed.
`While the time to effect after IV administration was faster,
`it was offset by the time needed to obtain IV access and
`administer the drug, as compared to SC administration, such
`that there was no difference in time to response. The SC
`
`Nalox1016
`Nalox-1 Pharmaceuticals, LLC
`Page 7 of 12
`
`

`

`70
`
`Drug Deliv. and Transl. Res. (2013) 3:63–74
`
`response data may be quite relevant to assessing possible
`doses and response profile of naloxone nasal spray. A sec-
`ond dose was needed in 35 % of the patients treated with IV
`naloxone and a second dose (given IV) was needed in 15 %
`of the patients treated with SC naloxone. Subjectively, the
`ambulance attendants administering the naloxone indicated
`that emergence was more gradual, resulting in less violence
`and aggression at the scene after SC administration as com-
`pared to IV administration. A similar effect may be seen
`after intranasal route of administration (see Table 3).
`Barton was the first to demonstrate intranasal delivery of
`naloxone by paramedics. Thirty patients in Denver, CO,
`USA encountered by paramedics received 2 mg of naloxone
`(injection formulation, 1 mg/mL) administered intranasally
`with the disposable nasal spray atomizer called the Mucosal
`Atomization Device [58]. One milliliter was administered
`into each naris upon initial patient contact. Paramedics then
`assessed/provided airway management and IV line place-
`ment unless the patient initially responded to the naloxone
`and no additional treatment was needed. Nasal abnormali-
`ties were also noted. Eighty-three percent of the patients
`with an opioid overdose responded to intranasal naloxone,
`with an average response time of 3.4 min. One patient
`responded to IV naloxone and not to IN naloxone alone.
`Sixty-four percent of the naloxone responders did not re-
`quire an IV placement. This was the first paper demonstrat-
`ing use of this delivery route in clinical practice.
`Barton published a final report of the aforementioned
`study. In this analysis of 95 patients, response rates to
`treatment remained similar [59]. That is, 83 % of naloxone
`responders responded to intranasal naloxone. Seven (16 %)
`of the intranasal responders required additional doses of IV
`naloxone. None of the “naloxone responders” were reported
`to have severe withdrawal reactions from either IV or IN
`naloxone. Nasal abnormalities (epistaxis, mucus, trauma, or
`septal abnormalities) were noted in five patients (of nine)
`who did not respond to IN naloxone. None of the IN
`naloxone responders had any nasal abnormality noted by
`paramedics.
`The mean time from drug administration to clinical re-
`sponse was slightly longer with intranasal (4.2 min) as
`compared to intravenous administration (3.7 min); the
`medians were not different (3.0 min for each route). How-
`ever, because of the time needed to obtain IV access in these
`
`patients, the time from first patient contact to time of clinical
`response is not different between the routes. The median
`time from arrival to the patient’s side to clinical response
`was 8 min for IN and 10 min for IV (means reported as 9.9
`and 12.8 min, respectively). Thus, the nasal route can be a
`quick first response while additional intervention and mon-
`itoring are conducted by rescue personnel.
`A randomized trial comparing 2 mg intranasal to 2 mg
`intramuscular naloxone was reported by Kelly in 2005 [22].
`One hundred fifty-five patients (71 IM and 84 IN) requiring
`prehospital treatment for suspected overdose received 2 mg/
`5 mL naloxone by one of the routes of administration. Sixty-
`three percent of the IN patients had 10 or more spontaneous
`respirations within 8 min of treatment as compared to 82 %
`of the intramuscular naloxone treated patients. Additional
`rescue naloxone was needed in 13 % of IM patients and
`26 % of IN patients (p0NS). The time to achieve a Glasgow
`Coma Scale (GCS) greater than 11 was not significantly
`different between groups. There were no major adverse
`events in either group, although the intramuscular group
`more commonly experienced withdrawal symptoms, espe-
`cially agitation and/or irritation. The authors conclude that
`IN naloxone wa

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket