Abstract
Nasal administration of treatments for neurologic conditions, including rescue therapies to treat seizure clusters among people with epilepsy, represents a meaningful advance in patient care. Nasal anatomy and physiology underpin the multiple advantages of nasal administration but also present challenges that must be addressed in any successful nasal formulation. Nasal cavity anatomy is complex, with a modest surface area for absorption that limits the dose volume of an intranasal formulation. The mucociliary clearance mechanism and natural barriers of the nasal epithelia must be overcome for adequate absorption. An extensive vasculature and the presence of olfactory nerves in the nasal cavity enable both systemic and direct-to-brain delivery of drugs targeting the central nervous system. Two intranasal benzodiazepine rescue therapies have been approved by the US Food and Drug Administration for seizure-cluster treatment, in addition to the traditional rectal formulation. Nasal sprays are easy to use and offer the potential for quick and consistent bioavailability. This review aims to increase the clinician’s understanding of nasal anatomy and physiology and of the formulation of intranasal rescue therapies and to facilitate patient education and incorporate intranasal rescue therapies for seizure clusters (also known as acute repetitive seizures) into their seizure action plans.
Keywords: Epilepsy, Benzodiazepines, Intranasal drug administration, Seizure cluster
Highlights
-
•
Intranasal neurologic drugs can advance ease of use and time to administration.
-
•
Nasal anatomy and physiology help to provide consistent bioavailability.
-
•
Intranasal formulations are designed specifically to optimize nasal absorption.
-
•
Two benzodiazepine nasal sprays are approved to treat seizure clusters in epilepsy.
-
•
Education on intranasal rescue therapies may increase patient comfort and care.
Introduction
Nasal administration represents an advance in treatment for a growing number of neurologic conditions, including triptans and ergotamine for migraine, esketamine for depression, naloxone for opioid overdose, and, of particular interest to clinicians treating patients with epilepsy, the benzodiazepines diazepam and midazolam as rescue therapy for seizure clusters (also known as acute repetitive seizures) [1], [2], [3], [4], [5], [6], [7], [8]. Epilepsy is a disabling condition with serious potential consequences. The US Centers for Disease Control and Prevention reports that 56% of adults taking antiseizure medications still experience seizures. Polytherapy is common in patients with drug-resistant epilepsy, while nondrug treatment options include surgery, neurostimulation, and diet [9]. Drug-resistant epilepsy may sometimes include treatment emergencies that may range on a continuum from multiple discrete events (seizure clusters) to continuous series without recovery between events and prolonged seizures (status epilepticus) [10]. Seizure clusters and their management are addressed in several recent reviews [11], [12], [13].
Rescue therapies for seizure clusters should be easy and safe to administer, with fast onset and reliable efficacy; administration should be socially acceptable to the patient and caregiver and can be administered during the seizure event [14]. Intravenous delivery of benzodiazepines is frequently used in the inpatient setting, particularly when venous access has already been established, however, it is not an option in outpatient settings. Rectal diazepam gel was approved by the U.S. Food and Drug Administration (FDA) in 1997 [15]. It is effective as an outpatient rescue therapy but has several drawbacks. It exhibits notable interpatient variability and may be difficult to administer during a seizure cluster, especially in adolescents and adults [16]. A 2002 survey of 91 adults with epilepsy reported that for seizure emergencies, they preferred early rectal benzodiazepine administration when compared with a visit to the emergency department, with some expressing embarrassment about the medication route [17]. More recently, other formulations of rescue therapy have been approved by the FDA, specifically intranasally delivered diazepam and midazolam; others are in development for prolonged seizures, including inhaled alprazolam [18]. Intranasal administration offers several advantages as a rescue therapy. Intranasal administration is noninvasive and easy to administer, with high patient acceptability [14]. It has potentially high and consistent bioavailability and rapid onset of action. It provides direct delivery into the circulation, circumventing first-pass metabolism in the liver, and it also offers the possibility of directly delivering the drug into the central nervous system (CNS), potentially facilitating more rapid seizure termination [14].
Other formulations, such as intravenous or oral formulations, are not designed for intranasal administration of CNS-targeted drugs and are thus likely suboptimal for addressing the challenges associated with nasal administration. These challenges are largely related to the anatomy and physiology of the nose. To be effective, nasal formulations must deliver adequate dosing in a limited dose volume (100–150 µL per nostril) [19]. Barriers to absorption include permeability across the nasal epithelia, which is poor for some molecules, and drug removal via mucociliary clearance and local cellular mechanisms [20]. There is the possibility that nasal irritation, congestion, or lesions may reduce absorption, and some patients and caregivers may have concerns about nasal administration. These concerns can typically be overcome with education, and patients who have used intranasal rescue therapy for seizure clusters generally have reported it to be easy to administer and preferable to rectal therapy [21], [22], [23].
This narrative review will address nasal anatomy and physiology within the context of intranasal drug delivery of rescue therapies for epilepsy. Elements of anatomy, neurovascular supply, physiology, and histology are discussed before turning to pathways of intranasal drug delivery to the CNS and challenges of intranasal drug formulations. Findings from studies of intranasal benzodiazepine rescue therapies highlight the importance of intranasal formulations for safety and effectiveness. The goal is to provide clinicians who prescribe rescue therapies with the context needed to understand and educate their patients on the benefits of intranasal formulations and the unique characteristics that make them key options in the care for patients with epilepsy who experience seizure clusters.
Anatomy and physiology
Nasal anatomy and development
The internal structures of the nose include a pair of large irregular cavities (Fig. 1) [24]. The roof is formed by the cribriform plate of the ethmoid at the base of the cranium, and each cavity extends down to the palate of the mouth. The septum, or inner wall, separates the 2 nasal cavities. The outer wall is convoluted into 3 (or 4) turbinates or conchae, each creating an associated canal, or meatus. The superior turbinate is located at the upper and posterior part of the cavity. It is the smallest, with the middle and inferior turbinates being increasingly larger in size. The total surface area of both nasal cavities in an adult is about 160 cm2, and the total volume is about 15 mL [24]. The roles of the nasal cavity include heating and humidifying air for respiration, clearing particles and dust, and olfaction [25].
Fig. 1.
Gross anatomy of the nasal cavity; frontal view (left) and sagittal view (right). Two irregularly shaped nasal cavities are bounded by the cribriform plate of the ethmoid at the top, the palate of the mouth on the bottom, the central septum, and outer walls with 3 turbinates and associated meatus. The nostrils open to the external environment and lead to the vestibule, lined with squamous mucosa. The central part of the cavity is divided into a large respiratory mucosa and a smaller olfactory mucosa above. The posterior of the cavity contains the nasopharynx-associated lymphoid tissue (NALT) and connects to the nasopharynx. The frontal sinuses, ethmoid sinuses, and maxillary sinuses drain into the middle and superior meatus through openings in the cavity wall. CSF, cerebrospinal fluid.
The nasal cavity contains 4 general regions: the vestibule, the respiratory region, the olfactory region, and the nasopharynx-associated lymphatic tissue (NALT) (Fig. 1). The vestibule rests behind the nostril openings and contains nasal hairs involved in filtering inhaled air. Its small surface area and squamous epithelial cell surface make it unsuited to extensive drug absorption [26]. Posterior to the vestibule is the nasal valve, a narrow, dynamic triangular slit leading to the respiratory and olfactory regions [27]. It is responsible for regulating the speed and direction of airflow during respiration. At the back of the cavity, the NALT, a part of the mucosal immune system, acts to prevent infection [28]. It is also important for intranasal vaccination via T-cell immunity, such as the seasonal flu vaccine [28], [29].
Together, the respiratory and olfactory regions form the large, central portion of the nasal cavity and are most relevant for intranasal drug delivery. The respiratory region includes the lateral walls of the cavity, including the turbinates, and is the largest (approximately 130 cm2) and most vascularized part of the nasal cavity, making it a prime site for systemic drug absorption after nasal administration [25], [26], [30]. The olfactory region comprises a small fraction (≤10 %) of the total surface area of the nasal cavity, 2–12.5 cm2 [24], [25], [31]. It is located in the uppermost part of the cavity, meaning it may be difficult to reach for nasally administered drugs [32]. The olfactory region contains olfactory sensory neurons (OSNs), and it represents the only site in the body where the CNS is in direct contact with the outer surface of the body, the nasal mucosal membrane in this case, and thus opens the possibility of direct-to-brain delivery of drugs [24], [32].
Development of the nasal cavity during childhood
The nasal cavity changes dramatically during the first years of life. For example, the surface area of the turbinate region nearly doubles from age 3 (∼55 cm2) to age 5 (∼107 cm2), at which point it is approaching adult size (∼113 cm2) [33]. However, morphologic differences suggest that the area continues to develop after this age. Studies show heterogeneity in airway dimension and morphology in children, which impacts air flow dynamics and aerosol deposition [33]. Studies in adolescents show that the nasal passages reach adult proportions in the mid-teens, after undergoing a slight decrease in cross-sectional area sometime between the ages of 9 and 13 years [34]. The olfactory system is established before birth, with nasal chemoreception beginning during the last trimester of gestation. Vascular innervation in children follows the same general plan as adults, with nerves associated mainly with arteries but also veins in the nasal cavity [35], [36].
Children and adults with respiratory conditions may also have changes in the upper respiratory system that could affect nasal delivery of rescue therapies, such as decreased nasal patency with asthma or laryngomalacia and chronic rhinitis in children with Down syndrome [37], [38], [39]. However, changes may not always lead to decreased absorption. For example, the presence of polyps in the nasal cavity has been associated with longer residence time of compounds, and the inflammation seen with chronic sinusitis or allergic rhinitis may render the nasal mucosa more permeable to drugs [40], [41]. Clinical studies of several drugs, including zolmitriptan, hydromorphone, fentanyl, butorphanol, triamcinolone acetonide, testosterone, and buserelin, show that the presence of rhinitis does generally not seem to affect the absorption, bioavailability, clinical effect, and safety of drugs delivered intranasally [42].
Human vs animal models
Animal models, particularly the rat, but also the mouse, rabbit, dog, sheep, and monkey, are often used in studies of intranasal administration and CNS delivery of drugs, and care must be taken in extrapolating to humans because of anatomical and physiologic differences. For example, the nasal anatomy of dogs, which are dependent on their sense of smell, is complex, with branched turbinates, compared with the single scroll in humans. And the olfactory region in rats makes up almost 50% of the nasal cavity, a 5-fold higher percentage than in humans [26].
Neurovascular supply
Blood is supplied to the nasal cavity from one branch of the sphenopalatine artery, as well as the ethmoidal branches of the ophthalmic artery (Fig. 2) [24]. Arterial blood flow in the nasal cavity is anteriorly against the flow of air. The venous network matches approximately the arterial structure. Blood in the ethmoidal veins flows into the ophthalmic plexus then proceeds to the cavernous sinus (cavernous plexi) [24].
Fig. 2.
Neurovascular supply to the nasal cavity; arterial supply (upper), venous system (middle), and innervation (lower). The nasal cavity receives blood from multiple sources. One branch of the sphenopalatine artery enters behind the superior meatus. The ethmoidal branches of the ophthalmic artery, a second source, enter through openings in the cribriform plate and supply blood to the anterior part of the inner and outer wall of the nose and the ethmoidal and frontal sinuses.Veins in the nasal cavity begin in the venous plexus on the inferior turbinate and meatus and the back of the septum. Blood flows into the ophthalmic plexus, then proceeds to the cavernous sinus (cavernous plexi). The veins from the upper nasal cavity feed into veins in the skull’s interior through the foramen coecum and collect into the superior longitudinal sinus. These sinuses are venous channels that change blood volume in response to stimuli. The nasal cavity is innervated by the trigeminal nerve, the fifth cranial nerve, and longest one of the head and face (not shown). The first division of this nerve, the ophthalmic nerve, innervates the nasal cavity, frontal sinuses, integument of the nose, eyeball, and lachrymal gland. A branch of the ophthalmic nerve enters the nasal cavity through the anterior ethmoidal foramen and an opening on the anterior aspect of the cribriform plate. Two of 4 branches entering the pterygopalatine ganglion (Meckel’s ganglion) supply the nose. The posterior sections of the turbinates are served by the nasopalatine nerve, a branch of the maxillary nerve, and the anterior parts are served by anterior ethmoidal nerve, a branch of the ophthalmic nerve. These nerves course with adjacent arteries. Twenty olfactory nerves enter the nasal cavity through openings in the cribriform plate. Within the cavity, each nerve divides and spreads over the lateral walls of the superior turbinate and upper third of the septum. They are connected to the olfactory bulb and convey signals from the olfactory neurons in the olfactory mucosa to the olfactory bulb.
The lower part of the septum contains a dense vascular bed with a rich blood supply, particularly at the transition from squamous to respiratory epithelium. The vascular bed extends over the middle and inferior turbinate and is composed of fenestrated vessels and capillaries that are thought to be a source of fluid for humidification of air during respiration. This fenestration may also play a role in facilitating drug absorption [24].
With respect to innervation of the nasal cavity, sensory nerves involved in olfaction are spread over the upper septum and outer wall and are connected through small openings in the cribriform plate to the olfactory bulb, an oval mass sitting on the ethmoid. These olfactory nerves comprise axons from OSN bodies in the olfactory epithelia [24]. Signals are transmitted from OSNs to mitral and tufted cells within the olfactory bulb. The latter cells act as efferent neurons and transmit signals to the olfactory cortex located on the ventral surface of the forebrain [31]. The olfactory pathway bypasses the thalamus, unlike other sensory neurons [24], [31].
The trigeminal nerve, the largest cranial nerve, is mainly a sensory nerve that has 2 branches that innervate the respiratory and olfactory epithelia near the anterior part of the septum and the anterior part of the outer wall of the nostril. It enters the brainstem at the pons level. Posterior sections of the nasal cavity are served by branches of the palatine and maxillary nerves [24], [43].
Cilia, mucus, and mucosal clearance
The composition of mucus and its constant removal from the nasal cavity represents challenges to drug delivery that can be addressed through formulation strategies to extend contact time with the nasal mucosa, thereby potentially improving absorption. The mucus overlaying the epithelium in the nasal cavity insulates the epithelium, provides humidification to inhaled air, maintains hydration, and protects from irritants, toxins, and pathogens through a removal system that uses motile cilia on epithelial cells to constantly sweep mucus and trapped particles out of the cavity [44]. Motile cilia are found on over 80% of cells in the respiratory mucosa [28]. Mucus is composed primarily of water and mucin proteins, which have both hydrophobic and hydrophilic regions and thus can capture particles of varying hydrophobicity by forming low-affinity bonds [28]. The mucus layer covering the nasal epithelium is biphasic, with an outer gel layer of higher viscosity and adhesive properties [25]. The inner periciliary, or serous fluid, layer allows for the beating of the cilia and the unidirectional movement of the gel layer posteriorly toward the nasopharynx for removal [44]. In adults, cilia beat at a rate of approximately 1000strokes per minute [40]. It is estimated that the respiratory mucus layer is renewed every 10 to 20 min [44]. Rates have been reported to be higher in children and adolescents [45], [46]. Clearance time can be affected by disease. For example, nasal congestion can lengthen clearance time [40].
Nasal histology and drug delivery
Histology of nasal structures
The respiratory region is lined with a ciliated pseudostratified epithelium (Fig. 3) composed of 4 cell types: ciliated and nonciliated columnar cells, goblet cells, and basal cells [25]. The epithelium is covered by the mucus layer (described above). The lamina propria lies beneath the epithelium and contains a dense network of capillaries and blood vessels that drain directly into the systemic circulation [24]. Serous glands in the lamina propria and goblet cells in the epithelium keep the tissue moist and produce mucus. The olfactory epithelium is also a ciliated pseudostratified epithelium that covers the superior turbinate and the uppermost part of the septum in each cavity [25]. The olfactory epithelium is marked by the presence of OSNs and Bowman’s glands, involved in mucus production. The basement membrane beneath the epithelium contains blood and lymph vessels, olfactory axon bundles, and other nerve fibers, including the trigeminal nerve. OSNs are bipolar cells with dendrites on their apical surface that extend into the mucous membrane [24]. On the basal surface, the neurons form unmyelinated axon bundles that pass through the foramina in the cribriform plate to join the olfactory bulb [25]. The axon bundles are surrounded by olfactory ensheathing cells and olfactory nerve fibroblasts [25].
Fig. 3.
Histology and drug transport across the epithelium of the nasal cavity; respiratory epithelium (left), olfactory epithelium (right). Drug transport across the epithelium can take different routes, labeled (1) to (4). Drug molecules may pass through the epithelium using paracellular (1) or transcellular (2) mechanisms (via tight junctions between cells) to enter the lamina propria underlying the epithelium and then be transported through blood or lymphatic vessels to the systemic circulation and on to the brain. Olfactory sensory neurons in the olfactory epithelium offer the potential for direct-to-brain delivery through intracellular (3) or perineural (4) routes. Perineural routes along the channels surrounding the olfactory neurons connect the fluid in the lamina propria of the olfactory epithelium and the cerebrospinal fluid (CSF) in the subarachnoid space. Intracellular transport is also possible through trigeminal nerve endings located in the respiratory epithelium.
Transmucosal drug transport
Following drug deposition, absorption is critical for successful drug delivery. In general, drugs can follow either extracellular or intracellular routes to gain access to the circulatory system or the nervous system (Fig. 3).
In the respiratory region, the main challenge to drug absorption is to cross the epithelium. Extracellular transport across the columnar cells of the epithelium can occur via paracellular diffusion [47]. Tight junctions (zonula occludens) are hydrophilic and generally impermeable to many drugs. In its natural state, the epithelium allows passage of molecules up to 1000 Da, with decreasing absorption with increasing molecular weight [41], [48]. Molecules passing through the epithelium enter the lamina propria where they can then enter the vasculature and systemic circulation [30]. Intracellular transport via transcytosis is another possible mechanism to reach the lamina propria [47]. Several intranasal drug formulation strategies aim to increase permeability of the epithelium by disrupting the cellular barrier or loosening tight junctions, as discussed below.
In the olfactory region, intracellular transport in olfactory neurons begins with internalization of the compound into the neuron and then transportation within the neuron toward the CNS, where it is released [49]. Olfactory neurons have been shown to use both endocytosis and pinocytosis to transport a range of molecules using receptor-mediated and nonreceptor-mediated mechanisms. Extracellular pathways in the olfactory epithelium use paracellular mechanisms to allow molecules to cross the epithelium into the lamina propria, similar to mechanisms in the respiratory mucosa [49].
Pathways to the CNS
After absorption through the nasal epithelium, multiple pathways are possible for delivery of neurotherapeutic drugs to the brain (Fig. 4). Drugs absorbed into the bloodstream and systemic circulation require transport across the blood–brain barrier. Olfactory and trigeminal nerve pathways offer the potential for direct nose-to-brain drug delivery. It is likely that individual drugs use the pathways to varying degrees depending on different factors, such as the molecular nature of the drug, the composition of the formulation, and the delivery system.
Fig. 4.
Pathways to the central nervous system (CNS) following intranasal drug delivery. A drug absorbed through the respiratory mucosa enters the general circulation and then passes through the blood–brain barrier (BBB) to reach its site of action, the CNS. A drug in general circulation will also be distributed to organs and tissues and be eliminated. Absorption in the respiratory mucosa may also lead to intracellular transport along trigeminal nerves pathways and direct transport to the CNS, though this route is thought to be slow (17–56 h). Absorption in the olfactory mucosa can lead to direct-to-brain delivery through olfactory sensory neurons via intracellular or extracellular pathways, with evidence suggesting that delivery along extracellular pathways can take minutes. Finally, excess drug delivered to the nasal cavity may be swallowed and absorbed by the gastrointestinal (GI) tract, similar to an orally administered drug.
Vascular pathway
The rich vascularization of the respiratory region, and the olfactory region to a lesser extent, allows drugs to be absorbed into the systemic circulation [30]. The vascular endothelia in these regions are both continuous and fenestrated, allowing both large and small molecules to enter the general circulation. Generally, lipophilic and low-molecular-weight drugs can pass through the blood–brain barrier, and thus CNS drug formulations must overcome the challenge of hydrophobic tight junctions and a lipophilic blood–brain barrier. There is also evidence from animal models that drug transfer, including diazepam, from venous to carotid blood supply (and thereby on to brain) can occur via local countercurrent mechanisms [50], [51]. Finally, there is the possibility of entry into the brain through the lymphatic system as the lymphatic system has connections with perineural spaces and the cerebrospinal fluid (CSF) [25], [52]. Nasal administration may avoid first-pass metabolism. Because passage through the systemic circulation raises the possibility of systemic toxicities, intranasal formulations may show better tolerability compared to other routes of delivery, such as oral or intravenous [31].
Research in rats and rabbits suggest that diazepam delivered intranasally reaches the brain by passing through the blood–brain barrier after entering the systemic circulation, as suggested by similar diazepam concentration–time profiles in plasma, CSF, and brain tissue [53]. The study authors also concluded that direct-to-brain delivery of diazepam did not occur in either animal model, based on a lack of higher drug targeting efficiency in brain tissues.
Olfactory and trigeminal pathways
The olfactory pathway involves OSNs as a direct route to the brain and CNS. Drugs may enter the brain directly via intraneuronal transport or the CSF via paraneuronal transport, before then being transferred into neuronal cells. Studies have reported intracellular translocation times along the olfactory nerves of 1.5–6 h or even longer, up to 24 h in some early mouse studies [28], [49], [54]. Thus, it is not likely to be a primary mechanism for delivery to the brain and CNS [28]. For paraneuronal transport, compounds must first pass through the olfactory mucosa via transcellular or paracellular mechanisms. From there, drugs may move via diffusion extracellularly through the perineural channels that exist between the olfactory neuron–ensheathing cells and the surrounding olfactory neural fibroblasts. These channels run the length of the olfactory neuron from olfactory epithelium to olfactory bulb and create a continuous connection between the fluid in the lamina propria of the olfactory epithelium and the CSF in the subarachnoid space [49]. Transport along this path is faster than intraneuronal transport, with reported times ranging from 5 min to 2.3 h [47], [55]. Evidence for intracellular or extracellular transport along the olfactory pathway has been obtained for compounds such as nerve growth factor, dihydroergotamine, morphine, carbamazepine, and albumin [55], [56], [57], [58], [59]. Transcellular pathways may be more relevant for lipophilic compounds, while hydrophilic compounds may follow passive diffusion along fluid-filled channels, with an inverse relationship to molecular weight of the compound [25]. Finally, arteries adjacent to the olfactory nerve bundles have been proposed to create a “perivascular pump” based on the high-pressure waves created by movement of blood through vessels after systole. This pump can move fluids within the perivascular space around arteries, moving solutes toward the brain in this region [49].
The trigeminal nerve, which innervates the respiratory epithelium, is less studied as a pathway of drug delivery to the brain [25]. Although trigeminal nerve endings are not directly exposed in the nasal cavity, it is assumed that drugs pass through the epithelium and then are transported extracellularly or intracellularly to the brain. Studies have implicated trigeminal nerves in the transport of insulin-like growth factor 1, lidocaine, and interferon-β-1b to the brain after intranasal administration [60], [61], [62]. Reported transport times are longer along the trigeminal nerve, 17–56 h, than the olfactory nerve [28].
Drug formulation considerations
Optimizing deposition in the nasal cavity
Because the volume of the nasal cavity is limited, the optimal administration volume is 100 µL (maximum 150 µL) per nostril in an adult [14], [19]. Thus, high concentrations of drug may be needed to ensure adequate dosing in a limited volume. Nasal formulations of hydrophobic molecules may require specific approaches to ensure adequate absorption (discussed below). The targeting of drug deposition and subsequent absorption within the nasal cavity is affected by the geometry of the spray plume as influenced by drug formulation, device mechanics, and patient/caregiver technique [63]. Droplet size also affects deposition, with droplets > 10 µm retained in the nasal cavity, while pulmonary deposition occurs with smaller droplets [64]. Narrower plume angles have been associated with better deposition in the respiratory and olfactory regions of the cavity, while wider angles result in more deposition on the nasal valve. Droplet velocity, formulation viscosity, and device type have less impact on deposition [63]. Minimizing off-target deposition is important so as to avoid loss of dose through leaking out of the nostrils, by swallowing into the gastrointestinal (GI) tract, or by inhaling or aspirating into the lungs. Excessive ingestion and absorption into the GI tract may impact bioavailability [14], while deposition in the lungs may lead to a range of adverse effects, from mild coughing to toxicity or infection [31].
Congestion in the nasal cavity is a potential consideration; however, rhinitis, including allergic, infectious, and vasomotor rhinitis, does not seem to have a clinical impact on nasal drug absorption, as seen in multiple studies [65], [66], [67], [68]. Moreover, in a study of diazepam nasal spray to treat seizure clusters, rates of second dose administration within 24 h of the first were similar between patients with and without history or concomitant treatment of seasonal allergies or rhinitis [42]. Finally, there is the potential for local toxicity to the active compound or excipients, such as solvents, absorption enhancers, or mucoadhesives, which are discussed next.
Enhancing absorption
Cellular processes that can impact drug absorption include cell membrane permeability, drug efflux, and drug metabolism [20]. The physiologic process of mucociliary clearance can also affect absorption. Absorption can be enhanced through multiple mechanisms, including enhancing solubility of the drug, increasing permeability of the mucosa, and increasing residence time on the nasal mucosa. Enhanced absorption can be obtained by modifying the drug, adjusting the pH of the solution, adding excipients, or creating new drug delivery systems.
The addition of enhancers can increase the permeability of the epithelium up to 30-fold vs the natural state, allowing molecules up to 30 kDa to pass [69]. For many compounds, absorption is increased with lower pH, while the opposite is true for other compounds [70], [71], [72], [73]. Excipients added to a formulation, such as permeation enhancers and mucoadhesive agents, may aid absorption (Table 1). Some absorption enhancers, however, can be associated with nasal irritation or cytotoxicity [74].
Table 1.
| Description | Potential Disadvantages | Examples | |
|---|---|---|---|
| Permeation enhancers | Enhanced movement across the nasal mucosa, often by opening tight junctions or increasing the solubility of the drug | Possible local toxicity and irritation of nasal mucosa with some agents (eg, cosolvents isopropyl alcohol, PEG400, propylene glycol) | Cosolvents (alcohol, PEG400, propylene glycol) Ionic and some nonionic surfactants (PEG400, PEG3500, polysorbate 20, polysorbate 80, polyoxyl 400 stearate) Selected fatty acids (capric acid, DHA, EPA, oleic acid, oleyl alcohol, palmitic acid, palmitoleic acid, stearic acid) Chitosan Cyclodextrins Modified vitamin E (TPGS) pH modifiers (sodium hydroxide, sulfuric acid, hydrochloric acid) Polymers |
| Mucoadhesive agents | Slowed removal of drug by mucociliary clearance by increasing residence time via increased viscosity or slowed removal action | Reduced barrier function of mucosa | Chitosan Hyaluronan Gellan gum |
| Agents used in research studies | |||
| Enzyme inhibitors | Inhibition of nasal cavity enzymes, prolonging drug stability and absorption | Reduced barrier function of nasal cavity | Fluvoxamine, α- aminoboronic acid derivatives |
| Lectin-mediated transport | Addition of agglutinin to target the nasal mucosa and increase brain uptake | Not specific to the nasal mucosa | Wheat germ agglutinin |
| P-glycoprotein inhibitors | Inhibit efflux of compounds from the brain, increasing concentrations in brain | Potential for reduced efflux of harmful compounds | Cyclosporin A, rifampin, pantoprazole, elacridar |
| Prodrugs | Chemical modification of drug to increase drug stability and absorption; drug activated by enzymatic modification after absorption (eg, esterases) | Enzymatic activation required | l-dopa butyl ester |
| Vasoconstrictors | Vasoconstriction of nasal blood vessels, leading to enhanced mucosal residence and decreased uptake in circulation | Increased blood pressure | Phenylephrine |
DHA, docosahexaenoic acid; EPA, eicosapentaenoic acid; PEG, polyethylene glycol; TPGS, D-ɑ-tocopheryl polyethylene glycol succinate.
Benzodiazepines as a class have low solubility in aqueous solutions and enhancing techniques have been used in the development of the FDA-approved intranasal rescue therapies [20]. The approved intranasal formulation of midazolam contains the cosolvents polyethylene glycol (PEG) 400, PEG-6 methyl ether, propylene glycol, and ethanol [7], [20], [75], [76]. Diazepam nasal spray contains a vitamin E–based formula to enhance the solubility of diazepam and dodecyl-beta-d-maltopyranoside, a nonionic alkylglycoside surfactant that is thought to enhance absorption by paracellular and transcellular routes (Fig. 5) [74]. It also contains benzyl and anhydrous alcohol [8].
Fig. 5.
Diagrammatic representation of mechanism of action of permeation enhancers used with diazepam nasal spray. Dodecyl-beta-d-maltopyranoside (DDM) is a nonionic alkylglycoside surfactant that alters electrical resistance values, mucosal viscosity, and membrane fluidity and can enhance absorption by temporarily loosening cell–cell junctions and increasing mucosal permeation. The related compound tetradecyl maltoside has been associated with vesicle-based transcellular transport. A vitamin E–based formula increases the solubility of diazepam.
Importantly, intranasal formulations have been specifically designed to optimize the bioavailability, effectiveness, and safety of intranasal delivery, while intravenous or oral formulations do not account for the specific parameters of intranasal delivery. In contrast, intravenous midazolam delivered using a spray tip or mucosal atomization device (MAD) to treat seizure clusters requires a large dose volume, which can result in significant loss of drug from the nasal cavity and variable dosing and absorption [77]. In addition, the low pH of the formulation may lead to irritation of nasopharyngeal tissues [78].
Using effective delivery devices
Integral to an intranasal formulation of a drug is the delivery device. Traditional nasal droppers require difficult maneuvers by the patient for correct administration and optimal deposition [79]. A MAD attached to a syringe has been used for intranasal delivery of several CNS-targeted drugs, including naloxone, ketamine, fentanyl, and the intravenous formulation of midazolam [78], [80], [81], [82]. However, this method has disadvantages: it can be inconvenient and challenging as it requires multiple steps to assemble. The device may be assembled in a pharmacy, with a limited shelf-life after preparation, or be assembled during the stress of the seizure event, requiring additional time and training for the person administering the therapy. Finally, it may be difficult to administer a consistent, exact dose [76]. Both currently FDA-approved intranasal benzodiazepine rescue therapies are formulated as fixed-dose delivery systems [7], [8]. These systems are designed for simple, intuitive use, which can be particularly important for rescue therapies delivered by nonmedical personnel in a stressful situation. They also deliver a premeasured drug volume, contributing to more consistent dosing. Although studies have indicated that spray administration angle can affect deposition, inhalation patterns during administration do not seem to have a significant influence [63]. Neither intranasal spray provides any recommendations on head position [7], [83], and the device itself can be used in any position [84]. Patients and caregivers report that training on the use of the device is easy and could be part of a larger program to develop a seizure action plan for patients with epilepsy [85].
Studies of intranasal rescue therapies compared with other routes of administration
The pharmacokinetic profiles of approved and off-label seizure-cluster rescue therapies are listed in Table 2. The time to maximum plasma concentration varies between the 2 available intranasal therapies, but therapeutic levels are achieved quickly for both benzodiazepines (∼70–200 ng/mL for diazepam [86], [87] and > 30–100 ng/mL for midazolam [88]). Changes in electroencephalogram (EEG) spike-wave activity occur very soon after administration of diazepam to treat status epilepticus: within 5 min after intravenous administration and after 10 min with rectal administration [89]. Peak serum diazepam concentrations were achieved at 50 min for intravenous administration and 58 min for rectal administration [89]. Similar results to intravenous administration have been obtained with an intranasal formulation of midazolam, with cessation of status epilepticus on EEG occurring within a median time of about 5 min after intranasal administration in a hospital setting [90]. Using an increase in β-band power as an early surrogate marker, researchers found evidence of midazolam exerting its effect on the brain at as soon as 4:07 min, on average, after administration [90].
Table 2.
Pharmacokinetic and pharmacodynamic parameters of common rescue therapies to treat seizure clusters [7], [8], [75], [96], [101], [102].
| Single Dose (maximum) | Route | Onset* (min) | tmax (range) | Bioavailability† (%) | t1/2 (hr) | |
|---|---|---|---|---|---|---|
| Diazepam (Valtoco®) | 5–20 mg (20 mg) | IN | 2–10 | 1.5 hr (0.8–4.0) ‡ | 97 | ∼49 |
| Diazepam (Valium®) | 5–10 mg (30 mg)§ | IV | 1–3 | 100 | ||
| Diazepam (Diastat®) | 0.2–0.5 mg/kg (20 mg) | PR | 2–10 | 1.2 hr** |
90 | ∼46 |
| Lorazepam (Ativan® injection) | 4 mg††¶ | IV | 1.6 | 100 | 9–19 | |
| Lorazepam (Ativan® sublingual tablets)‡‡ | 0.05 mg/kg (4 mg)§§ | SL | 15–17 | 1 hr | >90 | 12–15 |
| Midazolam (Nayzilam®) | 5 mg (5 mg) | IN | 3–10 | 17.3 min (7.8–28.2) | ∼44 | 2–6 |
| Midazolam + atomizer | 0.2 mg/kg (15 mg) | IV solution given IN | 6–14 | 44–83 | ||
| Midazolam (Versed®) | 0.3–0.35 mg/kg*** | IV | 1.5–2.5 | 100 | 2–6 |
IN, intranasal; IV, intravenous; PR, rectal; SL, sublingual; t1/2, elimination half-life; tmax, time to peak plasma concentration.
Relative to intravenous formulation.
10-mg dose.
Maximum dose according to US prescribing information to treat status epilepticus and severe recurrent convulsive seizures in adults.
Onset of action is less well studied than other pharmacokinetic parameters listed and should be considered an approximation.
15-mg dose.
For the treatment of status epilepticus in adults.
Not available in the US.
To treat excessive anxiety prior to surgical procedures in adults.
Initial dose to induce general anesthesia in an average adult under the age of 55 years.
In clinical trials, intranasal administration of diazepam rescue therapy led to seizure termination within 10 min for the majority of patients and was comparable to the results with rectal diazepam [91], [92]. In a safety study of adults and children with epilepsy using diazepam nasal spray, median overall duration from seizure onset to cessation was 6 min, with nearly two-thirds (63 %) of seizures ending within 10 min [91]. In adolescents and adults with epilepsy (age > 14 years) taking intranasal midazolam to treat seizure clusters, just over half (54 %) of seizures ended within 10 min with no recurrence within the next 6 h [93]. Moreover, studies suggest that intranasal administration leads to shorter total times to cessation compared with intravenous delivery, in part because of the time required to administer the treatments [94].
The duration of action of rescue therapy has been evaluated through assessment of the need for retreatment of seizure clusters within a certain period following initial dosing. For rectal diazepam gel, in an open-label clinical trial, 77 % of administrations prevented further seizures for the 12 h following the first dose [95], [96]. With intranasal midazolam, 38.5 % of episodes in an outpatient cohort required a second dose between 10 min to 6 h after the initial dose [7], [78], while a separate analysis of those data found that, among patients treated with a single dose within 10 min, 56.2 % of seizure-cluster episodes were treated with another dose or were followed by another seizure within 24 h [97]. Finally, in a long-term safety study of diazepam nasal spray, 12.6 % of treated seizure clusters were treated with a second dose within the full 24 h after the initial dose [92]. However, as there are no head-to-head studies for comparison of the above presented results, any comparison should be done with the understanding of the differences in their patient populations, including severity of epilepsy and historic duration of seizure clusters; availability of second doses; and study designs.
As a class, benzodiazepines carry a boxed warning on risks of concomitant use with opioids due to the possibility of profound respiratory depressive effects and the potential for abuse, misuse, and addiction. In clinical trials, the common adverse events associated with the intermittent use of benzodiazepine nasal sprays included somnolence, nasal discomfort, and headache. No signs of respiratory depression were observed during the long-term safety study of diazepam nasal spray [92]. During the test-dose phase of a phase 3 trial of intranasal midazolam (n = 292), 5 patients experienced an adverse event of acute central respiratory depression leading to or contributing to discontinuation [93]. Safety data for approved intranasal rescue therapies have not identified an impact on olfaction [78], [92]. Intranasal corticosteroids, which have been used to treat allergic rhinitis and other conditions of the upper respiratory tract for decades, have shown a good safety profile with no evidence of atrophy or deleterious pathologic changes in the nasal mucosa after 6 months to 5 years of use [98].
Conclusion
The anatomy and physiology of the nasal cavity present unique opportunities and challenges to implementing intranasal treatments for neurologic conditions. Intranasal formulations are designed to meet the specific parameters of the nasal anatomy and physiology in ways that oral, intravenous, or other formulations may not. Intranasal delivery offers an alternative route to rectal administration that is preferred by many patients for rapid delivery of rescue therapy to treat seizure clusters in people with epilepsy in outpatient settings, such as at home, school, or work. Rescue therapies are also an important part of a seizure action plan among appropriate patients. By overcoming specific challenges, the successful formulation of intranasal rescue therapies offers the opportunity of improved patient acceptance, better management, and potentially better patient outcomes.
Declaration of Competing Interest
The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Dr Chung is a consultant for Neurelis, Inc.; SK Life Science; Sunovion; and UCB and is a speaker for Eisai, Jazz Pharma, SK Life Science, Sunovion, and UCB. Dr Peters has served as a speaker and consultant for Greenwich Biosciences; Neurelis, Inc.; and Novartis. Dr Detyniecki is a consultant for Aquestive; Greenwich Biosciences; Neurelis, Inc.; and UCB. Dr Tatum receives a stipend from Elsevier as Editor-in-Chief of Epilepsy & Behavior Reports and is on the editorial board of the Journal of Clinical Neurophysiology. He has received grant funding from Esai, Martin Family Foundation, Mayo Clinic, and McElvey Fund and industry research support from Cerevel, Engage, Liva Nova, and Xenon. He serves as a consultant for Bioserenity; Medtronics; Neurelis, Inc.; and Zimmer-Biomet. He receives royalties from Demos, Springer, and Wiley publishers. He has patents/patents pending on intraoperative monitoring devices (#62527896; #62770362). Dr Rabinowicz is an employee of and has received stock options from Neurelis, Inc. Dr Carrazana is an employee of and has received stock and stock options from Neurelis, Inc.
Acknowledgments
Acknowledgments
Editorial support was provided by David McLay, PhD, of The Curry Rockefeller Group, LLC (Tarrytown, NY), and was funded by Neurelis, Inc. (San Diego, CA).
Funding
This work was funded by Neurelis, Inc. (San Diego, CA).
References
- 1.Bausch Health Companies Inc. Migranal nasal spray (dihydrorgotamine mesylate). Full Prescribing Information. Bausch Health Companies Inc.; 2022.
- 2.GlaxoSmithKline. Imitrex nasal spray (sumatriptan). Full Prescribing Information. GlaxoSmithKline; 2017.
- 3.Pharmaceuticals A. Full Prescribing Information; Amneal Pharmaceuticals: 2019. Zomig (zolmitriptan spray, metered) [Google Scholar]
- 4.US Food and Drug Administration . US Food and Drug Administration; Silver Spring, MD: 2019. Nayzilam (midazolam) NDA approval. [Google Scholar]
- 5.Pharmaceuticals J. Full Prescribing Information; Janssen Pharmaceutical Companies: 2020. Spravato nasal spray (esketamine) [Google Scholar]
- 6.Adapt Pharma, Inc. Narcan® Nasal Spray (naloxone hydrochloride). Full Prescribing Information. Plymouth Meeting, PA: Adapt Pharma, Inc.; 2020.
- 7.UCB, Inc., UCB, Inc.; Smyrna, GA: 2021. Nayzilam® (midazolam nasal spray). Full Prescribing Information. [Google Scholar]
- 8.Neurelis, Inc. VALTOCO (diazepam nasal spray). Full Prescribing Information. San Diego, CA: Neurelis, Inc.; 2022.
- 9.Laxer K.D., Trinka E., Hirsch L.J., Cendes F., Langfitt J., Delanty N., et al. The consequences of refractory epilepsy and its treatment. Epilepsy Behav. 2014;37:59–70. doi: 10.1016/j.yebeh.2014.05.031. [DOI] [PubMed] [Google Scholar]
- 10.Higdon L.M., Sperling M.R. A review of a diazepam nasal spray for the treatment of acute seizure clusters and prolonged seizures. Expert Rev Neurother. 2021;21:1207–1212. doi: 10.1080/14737175.2021.1965880. [DOI] [PubMed] [Google Scholar]
- 11.Chung S., Szaflarski J.P., Choi E.J., Wilson J.C., Kharawala S., Kaur G., et al. A systematic review of seizure clusters: prevalence, risk factors, burden of disease and treatment patterns. Epilepsy Res. 2021;177 doi: 10.1016/j.eplepsyres.2021.106748. [DOI] [PubMed] [Google Scholar]
- 12.Gidal B., Klein P., Hirsch L.J. Seizure clusters, rescue treatments, seizure action plans: unmet needs and emerging formulations. Epilepsy Behav. 2020;112 doi: 10.1016/j.yebeh.2020.107391. [DOI] [PubMed] [Google Scholar]
- 13.Jafarpour S., Hirsch L.J., Gainza-Lein M., Kellinghaus C., Detyniecki K. Seizure cluster: definition, prevalence, consequences, and management. Seizure. 2019;68:9–15. doi: 10.1016/j.seizure.2018.05.013. [DOI] [PubMed] [Google Scholar]
- 14.Cloyd J., Haut S., Carrazana E., Rabinowicz A.L. Overcoming the challenges of developing an intranasal diazepam rescue therapy for the treatment of seizure clusters. Epilepsia. 2021;62:846–856. doi: 10.1111/epi.16847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.US Food and Drug Administration. FDA Approved Drugs: NDA: 020648 - Diastat (diazepam rectal gel) Drug Approval, http://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&varApplNo=020648 1997 [accessed Jan 21, 2022].
- 16.Garnett W.R., Barr W.H., Edinboro L.E., Karnes H.T., Mesa M., Wannarka G.L. Diazepam autoinjector intramuscular delivery system versus diazepam rectal gel: a pharmacokinetic comparison. Epilepsy Res. 2011;93:11–16. doi: 10.1016/j.eplepsyres.2010.10.001. [DOI] [PubMed] [Google Scholar]
- 17.Tatum W.O. Adult patient perceptions of emergency rectal medications for refractory seizures. Epilepsy Behav. 2002;3:535–538. doi: 10.1016/s1525-5050(02)00559-0. [DOI] [PubMed] [Google Scholar]
- 18.French J.A., Wechsler R., Gelfand M.A., Pollard J.R., Vazquez B., Friedman D., et al. Inhaled alprazolam rapidly suppresses epileptic activity in photosensitive participants. Epilepsia. 2019;60:1602–1609. doi: 10.1111/epi.16279. [DOI] [PubMed] [Google Scholar]
- 19.Pires A., Fortuna A., Alves G., Falcao A. Intranasal drug delivery: how, why and what for? J Pharm Pharm Sci. 2009;12:288–311. doi: 10.18433/j3nc79. [DOI] [PubMed] [Google Scholar]
- 20.Kapoor M., Cloyd J.C., Siegel R.A. A review of intranasal formulations for the treatment of seizure emergencies. J Control Release. 2016;237:147–159. doi: 10.1016/j.jconrel.2016.07.001. [DOI] [PubMed] [Google Scholar]
- 21.Wheless JW, Sperling, M.R., Liow, K., et al. Safety of Valtoco (NRL-1; diazepam nasal spray) in patients with epilepsy: interim results from a phase 3, open-label, 12-month repeat dose study. Presented at: American Epilepsy Society . Baltimore; MD: 2019. 2019. [Google Scholar]
- 22.Penovich P., Wheless J.W., Hogan R.E., Guerra C., Cook D.F., Carrazana E., et al. Examining the patient and caregiver experience with diazepam nasal spray for seizure clusters: results from an exit survey of a phase 3, open-label, repeat-dose safety study. Epilepsy Behav. 2021;121 doi: 10.1016/j.yebeh.2021.108013. [DOI] [PubMed] [Google Scholar]
- 23.Nunley S., Glynn P., Rust S., Vidaurre J., Albert D.V.F., Patel A.D. A hospital-based study on caregiver preferences on acute seizure rescue medications in pediatric patients with epilepsy: intranasal midazolam versus rectal diazepam. Epilepsy Behav. 2019;92:53–56. doi: 10.1016/j.yebeh.2018.12.007. [DOI] [PubMed] [Google Scholar]
- 24.Gizurarson S. Anatomical and histological factors affecting intranasal drug and vaccine delivery. Curr Drug Deliv. 2012;9:566–582. doi: 10.2174/156720112803529828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bourganis V., Kammona O., Alexopoulos A., Kiparissides C. Recent advances in carrier mediated nose-to-brain delivery of pharmaceutics. Eur J Pharm Biopharm. 2018;128:337–362. doi: 10.1016/j.ejpb.2018.05.009. [DOI] [PubMed] [Google Scholar]
- 26.Erdo F., Bors L.A., Farkas D., Bajza A., Gizurarson S. Evaluation of intranasal delivery route of drug administration for brain targeting. Brain Res Bull. 2018;143:155–170. doi: 10.1016/j.brainresbull.2018.10.009. [DOI] [PubMed] [Google Scholar]
- 27.Djupesland P.G. Nasal drug delivery devices: characteristics and performance in a clinical perspective-a review. Drug Deliv Transl Res. 2013;3:42–62. doi: 10.1007/s13346-012-0108-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ganger S., Schindowski K. Tailoring formulations for intranasal nose-to-brain delivery: a review on architecture, physico-chemical characteristics and mucociliary clearance of the nasal olfactory mucosa. Pharmaceutics. 2018;10 doi: 10.3390/pharmaceutics10030116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Pizzolla A., Wang Z., Groom J.R., Kedzierska K., Brooks A.G., Reading P.C., et al. Nasal-associated lymphoid tissues (NALTs) support the recall but not priming of influenza virus-specific cytotoxic T cells. Proc Natl Acad Sci U S A. 2017;114:5225–5230. doi: 10.1073/pnas.1620194114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Dhuria S.V., Hanson L.R., Frey W.H., 2nd Intranasal delivery to the central nervous system: mechanisms and experimental considerations. J Pharm Sci. 2010;99:1654–1673. doi: 10.1002/jps.21924. [DOI] [PubMed] [Google Scholar]
- 31.Keller L.A., Merkel O., Popp A. Intranasal drug delivery: opportunities and toxicologic challenges during drug development. Drug Deliv Transl Res. 2022;12:735–757. doi: 10.1007/s13346-020-00891-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Martins P.P., Smyth H.D.C., Cui Z. Strategies to facilitate or block nose-to-brain drug delivery. Int J Pharm. 2019;570 doi: 10.1016/j.ijpharm.2019.118635. [DOI] [PubMed] [Google Scholar]
- 33.Xi J., Si X., Zhou Y., Kim J., Berlinski A. Growth of nasal and laryngeal airways in children: implications in breathing and inhaled aerosol dynamics. Respir Care. 2014;59:263–273. doi: 10.4187/respcare.02568. [DOI] [PubMed] [Google Scholar]
- 34.Crouse U., Laine-Alava M.T., Warren D.W., Wood C.L. A longitudinal study of nasal airway size from age 9 to age 13. Angle Orthod. 1999;69:413–418. doi: 10.1043/0003-3219(1999)069<0413:ALSONA>2.3.CO;2. [DOI] [PubMed] [Google Scholar]
- 35.Schaal B., Saxton T.K., Loos H., Soussignan R., Durand K. Olfaction scaffolds the developing human from neonate to adolescent and beyond. Philos Trans R Soc Lond B Biol Sci. 2020;375:20190261. doi: 10.1098/rstb.2019.0261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Figueroa J.M., Mansilla E., Suburo A.M. Innervation of nasal turbinate blood vessels in rhinitic and nonrhinitic children. Am J Respir Crit Care Med. 1998;157:1959–1966. doi: 10.1164/ajrccm.157.6.9709019. [DOI] [PubMed] [Google Scholar]
- 37.Chin C.J., Khami M.M., Husein M. A general review of the otolaryngologic manifestations of Down syndrome. Int J Pediatr Otorhinolaryngol. 2014;78:899–904. doi: 10.1016/j.ijporl.2014.03.012. [DOI] [PubMed] [Google Scholar]
- 38.Straszek S.P., Moeller A., Hall G.L., Zhang G., Stick S.M., Franklin P.J. Reference values for acoustic rhinometry in children from 4 to 13 years old. Am J Rhinol. 2008;22:285–291. doi: 10.2500/ajr.2008.22.3147. [DOI] [PubMed] [Google Scholar]
- 39.Thorstensen W.M., Sue-Chu M., Bugten V., Steinsvag S.K. Nasal flow, volumes, and minimal cross sectional areas in asthmatics. Respir Med. 2013;107:1515–1520. doi: 10.1016/j.rmed.2013.07.021. [DOI] [PubMed] [Google Scholar]
- 40.Illum L. Nasal clearance in health and disease. J Aerosol Med. 2006;19:92–99. doi: 10.1089/jam.2006.19.92. [DOI] [PubMed] [Google Scholar]
- 41.Donovan M.D., Huang Y. Large molecule and particulate uptake in the nasal cavity: the effect of size on nasal absorption. Adv Drug Deliv Rev. 1998;29:147–155. doi: 10.1016/s0169-409x(97)00066-5. [DOI] [PubMed] [Google Scholar]
- 42.Vazquez B., Wheless J., Desai J., Rabinowicz A.L., Carrazana E. Lack of observed impact of history or concomitant treatment of seasonal allergies or rhinitis on repeated doses of diazepam nasal spray administered per seizure episode in a day, safety, and tolerability: Interim results from a phase 3, open-label, 12-month repeat-dose safety study. Epilepsy Behav. 2021;118 doi: 10.1016/j.yebeh.2021.107898. [DOI] [PubMed] [Google Scholar]
- 43.Mistry A., Stolnik S., Illum L. Nanoparticles for direct nose-to-brain delivery of drugs. Int J Pharm. 2009;379:146–157. doi: 10.1016/j.ijpharm.2009.06.019. [DOI] [PubMed] [Google Scholar]
- 44.Edizer D.T., Yigit O., Rudenko M. In: All Around the Nose. Cingi C., Bayar Muluk N., editors. Springer International Publishing; Cham: 2020. Mucociliary Clearance and Its Importance; pp. 65–70. [Google Scholar]
- 45.Chilvers M.A., Rutman A., O'Callaghan C. Functional analysis of cilia and ciliated epithelial ultrastructure in healthy children and young adults. Thorax. 2003;58:333–338. doi: 10.1136/thorax.58.4.333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Roth Y., Aharonson E.F., Teichtahl H., Baum G.L., Priel Z., Modan M. Human in vitro nasal and tracheal ciliary beat frequencies: comparison of sampling sites, combined effect of medication, and demographic relationships. Ann Otol Rhinol Laryngol. 1991;100:378–384. doi: 10.1177/000348949110000506. [DOI] [PubMed] [Google Scholar]
- 47.Lochhead J.J., Thorne R.G. Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev. 2012;64:614–628. doi: 10.1016/j.addr.2011.11.002. [DOI] [PubMed] [Google Scholar]
- 48.McMartin C., Hutchinson L.E., Hyde R., Peters G.E. Analysis of structural requirements for the absorption of drugs and macromolecules from the nasal cavity. J Pharm Sci. 1987;76:535–540. doi: 10.1002/jps.2600760709. [DOI] [PubMed] [Google Scholar]
- 49.Crowe T.P., Greenlee M.H.W., Kanthasamy A.G., Hsu W.H. Mechanism of intranasal drug delivery directly to the brain. Life Sci. 2018;195:44–52. doi: 10.1016/j.lfs.2017.12.025. [DOI] [PubMed] [Google Scholar]
- 50.Muszak J., Krzymowski T., Gilun P., Stefanczyk-Krzymowska S. Countercurrent transfer of dopamine from venous blood in the cavernous sinus to the arterial blood supplying the brain - the perfused rabbit head as an experimental model. J Physiol Pharmacol. 2014;65:641–648. [PubMed] [Google Scholar]
- 51.Einer-Jensen N., Larsen L. Local transfer of diazepam, but not of cocaine, from the nasal cavities to the brain arterial blood in rats. Pharmacol Toxicol. 2000;87:276–278. doi: 10.1034/j.1600-0773.2000.pto870606.x. [DOI] [PubMed] [Google Scholar]
- 52.Johnston M., Zakharov A., Papaiconomou C., Salmasi G., Armstrong D. Evidence of connections between cerebrospinal fluid and nasal lymphatic vessels in humans, non-human primates and other mammalian species. Cerebrospinal Fluid Res. 2004;1:2. doi: 10.1186/1743-8454-1-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kaur P., Kim K. Pharmacokinetics and brain uptake of diazepam after intravenous and intranasal administration in rats and rabbits. Int J Pharm. 2008;364:27–35. doi: 10.1016/j.ijpharm.2008.07.030. [DOI] [PubMed] [Google Scholar]
- 54.Kristensson K., Olsson Y. Uptake of exogenous proteins in mouse olfactory cells. Acta Neuropathol. 1971;19:145–154. doi: 10.1007/BF00688493. [DOI] [PubMed] [Google Scholar]
- 55.Falcone J.A., Salameh T.S., Yi X., Cordy B.J., Mortell W.G., Kabanov A.V., et al. Intranasal administration as a route for drug delivery to the brain: evidence for a unique pathway for albumin. J Pharmacol Exp Ther. 2014;351:54–60. doi: 10.1124/jpet.114.216705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Barakat N.S., Omar S.A., Ahmed A.A. Carbamazepine uptake into rat brain following intra-olfactory transport. J Pharm Pharmacol. 2006;58:63–72. doi: 10.1211/jpp.58.1.0008. [DOI] [PubMed] [Google Scholar]
- 57.Frey W.H., Liu J., Chen X., Thorne R.G., Fawcett J.R., Ala T.A., et al. Delivery of125I-NGF to the brain via the olfactory route. Drug Deliv. 2008;4:87–92. doi: 10.3109/10717549709051878. [DOI] [Google Scholar]
- 58.Westin U., Piras E., Jansson B., Bergstrom U., Dahlin M., Brittebo E., et al. Transfer of morphine along the olfactory pathway to the central nervous system after nasal administration to rodents. Eur J Pharm Sci. 2005;24:565–573. doi: 10.1016/j.ejps.2005.01.009. [DOI] [PubMed] [Google Scholar]
- 59.Wang Y., Aun R., Tse F.L. Brain uptake of dihydroergotamine after intravenous and nasal administration in the rat. Biopharm Drug Dispos. 1998;19:571–575. doi: 10.1002/(sici)1099-081x(199812)19:9<571::aid-bdd142>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
- 60.T.M. Ross P.M. Martinez J.C. Renner R.G. Thorne L.R. Hanson W.H. Frey Intranasal administration of interferon beta bypasses the blood–brain barrier to target the central nervous system and cervical lymph nodes: a non-invasive treatment strategy for multiple sclerosis J Neuroimmunol 2004;151:66-77. doi: 10.1016/j.jneuroim.2004.02.011. [DOI] [PubMed]
- 61.Thorne R.G., Pronk G.J., Padmanabhan V., Frey W.H. Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration. Neuroscience. 2004;127:481–496. doi: 10.1016/j.neuroscience.2004.05.029. [DOI] [PubMed] [Google Scholar]
- 62.Johnson N.J., Hanson L.R., Frey W.H. Trigeminal pathways deliver a low molecular weight drug from the nose to the brain and orofacial structures. Mol Pharm. 2010;7:884–893. doi: 10.1021/mp100029t. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Foo M.Y., Cheng Y.S., Su W.C., Donovan M.D. The influence of spray properties on intranasal deposition. J Aerosol Med. 2007;20:495–508. doi: 10.1089/jam.2007.0638. [DOI] [PubMed] [Google Scholar]
- 64.Trows S., Wuchner K., Spycher R., Steckel H. Analytical challenges and regulatory requirements for nasal drug products in Europe and the U.S. Pharmaceutics. 2014;6:195–219. doi: 10.3390/pharmaceutics6020195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Perelman M., Fisher A.N., Smith A., Knight A. Impact of allergic rhinitis and its treatment on the pharmacokinetics of nasally administered fentanyl. Int J Clin Pharmacol Ther. 2013;51:349–356. doi: 10.5414/CP201825. [DOI] [PubMed] [Google Scholar]
- 66.Kaila T., Suonpaa J., Grenman R., Iisalo E. Vasomotor rhinitis and the systemic absorption of ipratropium bromide. Rhinology. 1990;28:83–89. [PubMed] [Google Scholar]
- 67.Argenti D., Colligon I., Heald D., Ziemniak J. Nasal mucosal inflammation has no effect on the absorption of intranasal triamcinolone acetonide. J Clin Pharmacol. 1994;34:854–858. doi: 10.1002/j.1552-4604.1994.tb02051.x. [DOI] [PubMed] [Google Scholar]
- 68.Lunell E., Molander L., Andersson M. Relative bioavailability of nicotine from a nasal spray in infectious rhinitis and after use of a topical decongestant. Eur J Clin Pharmacol. 1995;48:71–75. doi: 10.1007/BF00202176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Maggio E.T., Pillion D.J. High efficiency intranasal drug delivery using Intravail® alkylsaccharide absorption enhancers. Drug Deliv Transl Res. 2013;3:16–25. doi: 10.1007/s13346-012-0069-z. [DOI] [PubMed] [Google Scholar]
- 70.Tengamnuay P., Sahamethapat A., Sailasuta A., Mitra A.K. Chitosans as nasal absorption enhancers of peptides: comparison between free amine chitosans and soluble salts. Int J Pharm. 2000;197:53–67. doi: 10.1016/s0378-5173(99)00451-2. [DOI] [PubMed] [Google Scholar]
- 71.Shimoda N., Maitani Y., Machida Y., Nagai T. Effects of dose, pH and osmolarity on intranasal absorption of recombinant human erythropoietin in rats. Biol Pharm Bull. 1995;18:734–739. doi: 10.1248/bpb.18.734. [DOI] [PubMed] [Google Scholar]
- 72.Ahmed S., Sileno A.P., deMeireles J.C., Dua R., Pimplaskar H.K., Xia W.J., et al. Effects of pH and dose on nasal absorption of scopolamine hydrobromide in human subjects. Pharm Res. 2000;17:974–977. doi: 10.1023/a:1007551927177. [DOI] [PubMed] [Google Scholar]
- 73.Ohwaki T., Ando H., Kakimoto F., Uesugi K., Watanabe S., Miyake Y., et al. Effects of dose, pH, and osmolarity on nasal absorption of secretin in rats. II: histological aspects of the nasal mucosa in relation to the absorption variation due to the effects of pH and osmolarity. J Pharm Sci. 1987;76:695–698. doi: 10.1002/jps.2600760905. [DOI] [PubMed] [Google Scholar]
- 74.Rabinowicz A.L., Carrazana E., Maggio E.T. Improvement of intranasal drug delivery with Intravail® alkylsaccharide excipient as a mucosal absorption enhancer aiding in the treatment of conditions of the central nervous system. Drugs R D. 2021;21:361–369. doi: 10.1007/s40268-021-00360-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Bouw M.R., Chung S.S., Gidal B., King A., Tomasovic J., Wheless J.W., et al. Clinical pharmacokinetic and pharmacodynamic profile of midazolam nasal spray. Epilepsy Res. 2021;171 doi: 10.1016/j.eplepsyres.2021.106567. [DOI] [PubMed] [Google Scholar]
- 76.Marx D., Williams G., Birkhoff M. In: Drug Discovery and Development — From Molecules to Medicine. Vallisuta O., Olimat S., editors. InTech Open; London, UK: 2015. Intranasal drug administration — an attractive delivery route for some drugs; pp. 299–320. [Google Scholar]
- 77.Bancke L.L., Dworak H.A., Rodvold K.A., Halvorsen M.B., Gidal B.E. Pharmacokinetics, pharmacodynamics, and safety of USL261, a midazolam formulation optimized for intranasal delivery, in a randomized study with healthy volunteers. Epilepsia. 2015;56:1723–1731. doi: 10.1111/epi.13131. [DOI] [PubMed] [Google Scholar]
- 78.Wheless J.W., Meng T.C., Van Ess P.J., Detyniecki K., Sequeira D.J., Pullman W.E. Safety and efficacy of midazolam nasal spray in the outpatient treatment of patients with seizure clusters: an open-label extension trial. Epilepsia. 2019;60:1809–1819. doi: 10.1111/epi.16300. [DOI] [PubMed] [Google Scholar]
- 79.Vidgren M.T., Kublik H. Nasal delivery systems and their effect on deposition and absorption. Adv Drug Deliv Rev. 1998;29:157–177. doi: 10.1016/s0169-409x(97)00067-7. [DOI] [PubMed] [Google Scholar]
- 80.Tippey K.G., Yovanoff M., McGrath L.S., Sneeringer P. Comparative human factors evaluation of two nasal naloxone administration devices: NARCAN® nasal spray and naloxone prefilled syringe with nasal atomizer. Pain Ther. 2019;8:89–98. doi: 10.1007/s40122-019-0118-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Buonsenso D., Barone G., Valentini P., Pierri F., Riccardi R., Chiaretti A. Utility of intranasal ketamine and midazolam to perform gastric aspirates in children: a double-blind, placebo controlled, randomized study. BMC Pediatr. 2014;14:67. doi: 10.1186/1471-2431-14-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.O'Donnell D.P., Schafer L.C., Stevens A.C., Weinstein E., Miramonti C.M., Kozak M.A. Effect of introducing the mucosal atomization device for fentanyl use in out-of-hospital pediatric trauma patients. Prehosp Disaster Med. 2013;28:520–522. doi: 10.1017/S1049023X13003579. [DOI] [PubMed] [Google Scholar]
- 83.Neurelis, Inc. VALTOCO. Instructions for use for 5 mg and 10 mg doses 2022.
- 84.Pharma A. Aptar Brochure: Nasal spray device with uni-dose and bi-dose systems. In. 2016 [Google Scholar]
- 85.Penovich P., Glauser T., Becker D., Patel A.D., Sirven J., Long L., et al. Recommendations for development of acute seizure action plans (ASAPs) from an expert panel. Epilepsy Behav. 2021;123 doi: 10.1016/j.yebeh.2021.108264. [DOI] [PubMed] [Google Scholar]
- 86.Boddu S.H.S., Kumari S. A Short review on the intranasal delivery of diazepam for treating acute repetitive seizures. Pharmaceutics. 2020:12. doi: 10.3390/pharmaceutics12121167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Dhir A., Rogawski M.A. Determination of minimal steady-state plasma level of diazepam causing seizure threshold elevation in rats. Epilepsia. 2018;59:935–944. doi: 10.1111/epi.14069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kanto J.H. Midazolam: the first water-soluble benzodiazepine. Pharmacology, pharmacokinetics and efficacy in insomnia and anesthesia. Pharmacotherapy. 1985;5:138–155. doi: 10.1002/j.1875-9114.1985.tb03411.x. [DOI] [PubMed] [Google Scholar]
- 89.Milligan N., Dhillon S., Richens A., Oxley J. Rectal diazepam in the treatment of absence status: a pharmacodynamic study. J Neurol Neurosurg Psychiatry. 1981;44:914–917. doi: 10.1136/jnnp.44.10.914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kay L., Merkel N., von Blomberg A., Willems L.M., Bauer S., Reif P.S., et al. Intranasal midazolam as first-line inhospital treatment for status epilepticus: a pharmaco-EEG cohort study. Ann Clin Transl Neurol. 2019;6:2413–2425. doi: 10.1002/acn3.50932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wheless J., Peters J., Misra S.N., Becker D., Rabinowicz A.L., Sirven J., et al. Comment on “Intranasal midazolam versus intravenous/rectal benzodiazepines for acute seizure control in children: a systematic review and meta-analysis”. Epilepsy Behav. 2022;128 doi: 10.1016/j.yebeh.2021.108550. [DOI] [PubMed] [Google Scholar]
- 92.Wheless J.W., Miller I., Hogan R.E., Dlugos D., Biton V., Cascino G.D., et al. Final results from a phase 3, long-term, open-label, repeat-dose safety study of diazepam nasal spray for seizure clusters in patients with epilepsy. Epilepsia. 2021;62:2485–2495. doi: 10.1111/epi.17041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Detyniecki K., Van Ess P.J., Sequeira D.J., Wheless J.W., Meng T.C., Pullman W.E. Safety and efficacy of midazolam nasal spray in the outpatient treatment of patients with seizure clusters-a randomized, double-blind, placebo-controlled trial. Epilepsia. 2019;60:1797–1808. doi: 10.1111/epi.15159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chhabra R., Gupta R., Gupta L.K. Intranasal midazolam versus intravenous/rectal benzodiazepines for acute seizure control in children: a systematic review and meta-analysis. Epilepsy Behav. 2021;125 doi: 10.1016/j.yebeh.2021.108390. [DOI] [PubMed] [Google Scholar]
- 95.Mitchell W.G., Conry J.A., Crumrine P.K., Kriel R.L., Cereghino J.J., Groves L., et al. An open-label study of repeated use of diazepam rectal gel (Diastat) for episodes of acute breakthrough seizures and clusters: safety, efficacy, and tolerance. North American Diastat Group Epilepsia. 1999;40:1610–1617. doi: 10.1111/j.1528-1157.1999.tb02047.x. [DOI] [PubMed] [Google Scholar]
- 96.Bausch Health US, LLC. Diastat® C-IV (diazepam rectal gel). Full Prescribing Information. Bridgewater, NJ: Bausch Health US, LLC; 2021.
- 97.J.W. Wheless M. Brunnert F. Floricel S. Dimova B. Fannon Early intervention with midazolam nasal spray and efficacy in patients with seizure clusters: post-hoc analysis of an open-label extension trial Presented at: American Epilepsy Society Annual Meeting December 3–7, 2021, 2021; Chicago.
- 98.Sastre J., Mosges R. Local and systemic safety of intranasal corticosteroids. J Investig Allergol Clin Immunol. 2012;22:1–12. [PubMed] [Google Scholar]
- 99.Yang C., Wu T., Qi Y., Zhang Z. Recent advances in the application of vitamin E TPGS for drug delivery. Theranostics. 2018;8:464–485. doi: 10.7150/thno.22711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ghadiri M., Young P.M., Traini D. Strategies to enhance drug absorption via nasal and pulmonary routes. Pharmaceutics. 2019:11. doi: 10.3390/pharmaceutics11030113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Agarwal S.K., Kriel R.L., Brundage R.C., Ivaturi V.D., Cloyd J.C. A pilot study assessing the bioavailability and pharmacokinetics of diazepam after intranasal and intravenous administration in healthy volunteers. Epilepsy Res. 2013;105:362–367. doi: 10.1016/j.eplepsyres.2013.02.018. [DOI] [PubMed] [Google Scholar]
- 102.Cloyd J.C., Lalonde R.L., Beniak T.E., Novack G.D. A single-blind, crossover comparison of the pharmacokinetics and cognitive effects of a new diazepam rectal gel with intravenous diazepam. Epilepsia. 1998;39:520–526. doi: 10.1111/j.1528-1157.1998.tb01415.x. [DOI] [PubMed] [Google Scholar]





