![]() Intranasal epinephrine formulations and methods for the treatment of disease
专利摘要:
Drug products adapted for nasal delivery comprising formulations with epinephrine and devices comprising such formulations are provided. Methods of treating anaphylaxis with epinephrine products are also provided. 公开号:ES2791775A2 申请号:ES202090032 申请日:2019-02-06 公开日:2020-11-05 发明作者:Richard Lowenthal;Edward T Maggio;Robert G Bell;Pratik Shah 申请人:Ars Pharmaceuticals Inc;Aegis Therapeutics LLC; IPC主号:
专利说明:
[0002] INTRANASAL EPINEPHRINE FORMULATIONS AND METHODS FOR TREATMENT [0004] CROSS REFERENCE [0005] This application claims the benefit of US Patent Application No. 15 / 890,131 filed February 6, 2018; and U.S. Provisional Patent Application No. 62 / 784,057 filed December 21, 2018; each of which is incorporated herein by reference in its entirety. [0007] FIELD OF THE INVENTION [0008] This document describes intranasal (IN) Epinephrine formulations and methods for using such formulations in the treatment of conditions or diseases. [0010] FIELD OF THE INVENTION [0011] Anaphylaxis is a medical emergency that may require resuscitation measures, such as airway management, supplemental oxygen, large volumes of intravenous fluids, and close monitoring. Epinephrine administration is the treatment of choice. There is a need for needle-free and non-invasive methods for the dosing of Epinephrine. Provided herein are methods, formulations, and devices for the treatment of anaphylaxis and other conditions. [0013] SUMMARY OF THE INVENTION [0014] Methods, pharmaceutical formulations of Epinephrine and methods of using the same in the treatment of conditions such as type 1 hypersensitivity reactions (systemic allergic reaction), asthma and cardiac arrest are described herein. [0015] Anaphylaxis is a severe and life-threatening type 1 hypersensitivity reaction (systemic allergic reaction) that affects many body systems, with a rapid onset that usually averages between approximately 5 and 30 minutes after intravenous exposure to an antigen and approximately 2 hours. after oral exposure Anaphylaxis is the result of the release of inflammatory mediators and cytokines from mast cells and basophils, usually due to an immunological reaction, but sometimes due to non-immunological mechanisms. The most common areas of the affected body include: skin (80-90%), respiratory (70%), gastrointestinal (30-45%), heart and vasculature (10-45%) and central nervous system (10-15%) with two or more people involved in a single episode. [0016] Anaphylaxis is a medical emergency that may require resuscitation measures, such as such as airway management, supplemental oxygen, large volumes of intravenous fluids, and close monitoring. Epinephrine administration is the treatment of choice with antihistamines and steroids (eg, dexamethasone) that are often used as supplements. Due to concerns of biphasic anaphylaxis, a hospital observation period of between 2 and 24 hours is often required for people once they have returned to normal. [0017] Epinephrine (adrenaline, (fi) -4- (1-Hydroxy-2- (methylamino) ethyl) benzene-1,2-diol) is the primary treatment for anaphylaxis without absolute contraindication for its use. Currently, Epinephrine is administered as a solution administered by injection, preferably into the mid anterolateral thigh as soon as anaphylaxis is suspected. The injection can be repeated every 5 to 15 minutes if there is not enough response. A second dose is needed in 16-35% of episodes, but more than two doses are rarely required. The intramuscular route is preferred over subcutaneous administration because the latter may have a delayed absorption of epinephrine. However, although only minor adverse effects of Epinephrine (tremors, anxiety, headaches, and palpitations) are reported, there have been numerous reports of highly variable exposures of injection products depending on the injection location (intramuscular or subcutaneous ) and other factors such as body mass index (BMI). [0018] There is a significant need in the medical community to develop products that help improve the clinical management of anaphylaxis in an out-of-hospital setting. While epinephrine is effective when administered by intramuscular injection, there is published evidence that pharmacokinetics are highly variable depending on the injection site, whether intramuscular or subcutaneous. There have also been significant product quality issues with approved autoinjectors using complex technologies, leading to many recalls of these products by the FDA in the United States. Epinephrine auto-injectors, such as EpiPen®, are also difficult to transport and require training and time to administer properly in a life-threatening situation. [0019] The need for alternative, needle-free, and non-invasive methods for dosing Epinephrine is well documented, as many patients are fearful of injection and, as a result, are reluctant to use an autoinjector of any kind. Also, auto-injectors are large and heavy, so many patients who need them do not have an Epinephrine injector in their presence at all times. There is also a well-documented reluctance to self-administer a dose in public settings. [0020] Therefore, there is a need for improved or alternative methods of dosing Epinephrine in an emergency situation, as well as improved or alternative formulations and devices. Desirable enhancements include: individually and in combinations, convenience (intranasal versus intramuscular), faster administration, more reliable dosing, more consistent, needle-free, more discreet dose in public and administrable by an untrained or non-professional individual. [0021] Accordingly, provided herein are methods, formulations and devices for the treatment of anaphylaxis and other conditions comprising administering an intranasal formulation of Epinephrine using a small compact unit dose spray device. [0022] In one aspect, a nasal aerosol pharmaceutical formulation comprising between about 0.40 mg and about 2.4 mg of Epinephrine, or a salt thereof, is described herein. In another aspect, described herein is a nasal pharmaceutical aerosol formulation comprising between about 0.40 mg and about 2.4 mg of Epinephrine, or a salt thereof, in a single dose of the nasal pharmaceutical aerosol formulation. In another aspect, disclosed herein is a nasal aerosol pharmaceutical formulation comprising from about 0.40 mg to about 2.4 mg of Epinephrine, or a salt thereof, in a single dose nasal pharmaceutical aerosol formulation. In some embodiments, the nasal aerosol pharmaceutical formulation comprises between about 0.40 mg and about 2.0 mg of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol pharmaceutical formulation comprises between about 0.40 mg and about 1.8 mg of Epinephrine, or a salt thereof. In some embodiments, a single dose of the nasal aerosol pharmaceutical formulation comprises between about 0.5 mg and about 2.0 mg of Epinephrine, or a salt thereof. In some embodiments, a single dose of the nasal pharmaceutical aerosol formulation comprises between about 0.5 mg and about 1.5 mg of Epinephrine, or a salt thereof. In some embodiments, a single dose of the nasal pharmaceutical aerosol formulation comprises between about 0.5 mg and about 0.7 mg of Epinephrine, or a salt thereof. In some embodiments, a single dose of the nasal pharmaceutical aerosol formulation comprises about 1.0 mg of Epinephrine, or a salt thereof. In some embodiments, a single dose of the nasal pharmaceutical aerosol formulation comprises between about 1.3 mg and about 1.5 mg of Epinephrine, or a salt thereof. In some embodiments, intranasal administration of a single dose of the nasal pharmaceutical aerosol formulation to a subject provides a concentration of Epinephrine in plasma that is effective for the treatment of an acute hypersensitivity reaction. In some embodiments, the nasal aerosol pharmaceutical formulation is an aqueous solution, aqueous suspension, aqueous emulsion, non-aqueous solution, non-aqueous suspensions, non-aqueous emulsion, or dry powder. [0023] In one aspect, a nasal aerosol formulation is described herein which comprises between about 0.40 mg and about 2.4 mg per dose of Epinephrine, or a salt thereof, dispensed from the device. In some embodiments, a nasal aerosol formulation comprising between about 0.5 mg and about 2.0 mg of Epinephrine, or a salt thereof, per dose dispensed from the device is described herein; between about 0.5 mg and about 1.5 mg of Epinephrine, or a salt thereof, per dose dispensed from the device; between about 0.5 mg and about 0.7 mg of Epinephrine, or a salt thereof, per dose dispensed from the device; of approximately 1.0 mg of Epinephrine, or a salt thereof, per dose dispensed from the device; or between about 1.3 mg and about 1.5 mg of Epinephrine, or a salt thereof, per dose dispensed from the device. In some embodiments, a single dose of the nasal aerosol formulation when administered intranasally provides plasma concentrations of Epinephrine that are effective for the treatment of an acute hypersensitivity reaction. In some embodiments, the Epinephrine or its salt is present in the pharmaceutical formulation in an amount effective for treating an acute hypersensitivity reaction. In some embodiments, the nasal aerosol formulation is an aqueous solution, aqueous suspension, aqueous emulsion, non-aqueous solution, non-aqueous suspension, or non-aqueous emulsion. [0024] In some embodiments, the nasal aerosol formulation comprises between about 1 mg / mL and about 40 mg / mL of Epinephrine, or a salt thereof, per dose. In some embodiments, the nasal aerosol formulation comprises between about 5 mg / mL and about 40 mg / mL of Epinephrine, or a salt thereof, per dose. In some embodiments, the nasal aerosol formulation comprises between about 1 mg / mL and about 20 mg / mL of Epinephrine, or a salt thereof, per dose. In some embodiments, the nasal aerosol formulation comprises between about 3 mg / mL and about 20 mg / mL of Epinephrine, or a salt thereof, per dose. In some embodiments, the nasal aerosol formulation comprises between about 3 mg / mL and about 15 mg / mL of Epinephrine, or a salt thereof, per dose. In some embodiments, the nasal aerosol formulation comprises about 3 mg / mL, about 4 mg / mL, about 5 mg / mL, about 6 mg / mL, about 7 mg / mL, about 8 mg / mL, about 9 mg / mL. mL, about 10 mg / mL, about 11 mg / mL, about 12 mg / mL, about 13 mg / mL, about 14 mg / mL, about 15 mg / mL, about 16 mg / mL, about 17 mg / mL, approximately 18 mg / mL, approximately 19 mg / mL, or approximately 20 mg / mL of Epinephrine, or a salt thereof, per dose. In some embodiments, a dose of the nasal spray formulation comprises about 100 pL of the nasal spray Epinephrine formulation described herein. [0025] In some embodiments, a nasal spray formulation described herein comprises about 1 mg / mL to about 40 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a nasal aerosol formulation described herein comprises about 1 mg / mL to about 20 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a nasal spray formulation described herein comprises about 1 mg / mL to about 18 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a nasal spray formulation described herein comprises about 1 mg / mL, about 2 mg / mL, about 3 mg / mL, about 4 mg / mL, about 5 mg / mL, about 6 mg / mL. , about 7 mg / mL, about 8 mg / mL, about 9 mg / mL, about 10 mg / mL, about 11 mg / mL, about 12 mg / mL, about 13 mg / mL, about 14 mg / mL, about 15 mg / mL, approximately 16 mg / mL, approximately 17 mg / mL, approximately 18 mg / mL, approximately 19 mg / mL, or approximately 20 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a nasal spray formulation described herein comprises about 3 mg / mL, about 5 mg / mL, about 6 mg / mL, about 6.5 mg / mL, about 7 mg / mL, about 7.5 mg / mL. , about 8 mg / mL, about 8.5 mg / mL, about 9 mg / mL, about 9.5 mg / mL, about 10 mg / mL, about 10.5 mg / mL, about 11 mg / mL, about 11.5 mg / mL, about 12 mg / mL, approximately 12.5 mg / mL, approximately 13 mg / mL, approximately 13.5 mg / mL, approximately 14 mg / mL, approximately 14.5 mg / mL, or approximately 15 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a nasal aerosol formulation described herein comprises about 10 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a nasal spray formulation described herein comprises about 6 mg / mL to about 8 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a nasal aerosol formulation described herein comprises about 13 mg / mL to about 15 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a dose of the nasal spray formulation comprises about 100 pL of the nasal spray Epinephrine formulation described herein. [0026] In some embodiments, a dose of about 100 pL of the nasal aerosol formulation described herein comprises from 1 mg / mL to about 40 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a dose of about 100 pL of the nasal aerosol formulation described herein comprises 1 mg / mL to 20 mg / mL of Epinephrine, or a salt thereof. In some embodiments, a dose of approximately 100 pL of the nasal spray formulation described herein comprises 3 mg / mL, 3.5 mg / mL, 4 mg / mL, 4.5 mg / mL, 5 mg / mL, 6 mg / mL, 6.5 mg./ mL, 7 mg / mL, 7.5 mg / mL, 8 mg / mL, 8.5 mg / mL, 9 mg / mL, 9.5 mg / mL, 10 mg / mL, 10.5 mg / mL, 11 mg / mL, 11.5 mg / mL, 12 mg / mL, 12.5 mg / mL, 13 mg / mL, 13.5 mg / mL, 14 mg / mL, 14.5 mg / mL, or 15 mg / mL of Epinephrine, or a salt thereof. [0027] In some embodiments, the nasal aerosol formulation comprises one or more absorption enhancers. [0028] In some embodiments, the nasal spray formulation provides pharmacokinetics similar to intramuscular (IM) injection when IM injection is dosed in the lateral thigh, or absorption similar to subcutaneous (SC) or in the middle. [0029] In some embodiments, the nasal aerosol formulation provides absorption similar to intramuscular (IM) injection. [0030] In some embodiments, the nasal aerosol formulation provides similar absorption to subcutaneous (SC) and the pharmacokinetic profile of SC has a C max of at least 100 pg / mL and AUC 0-240 min of 150 h * pg / mL. [0031] In some embodiments, intranasal administration of a single dose of the nasal pharmaceutical aerosol formulation to a subject provides absorption similar to intramuscular (IM) injection. [0032] In some embodiments, the nasal spray formulation when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC 0-t are at least 80% of AUC 0-20min and AUC 0-t of what a 0.3 mg intramuscular injection yields; a mean C max that is at least 80% of C max and no more than 150% of C max of what a 0.3 mg intramuscular injection yields; a mean W of less than 45 minutes; o Absorption similar to IM injection under optimal dosing conditions in the thigh. In some embodiments, the nasal spray formulation when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC 0-t are at least 80% of the AUC 0-20min and AUC 0-t of what a 0.3 mg intramuscular injection yields; a mean C max that is at least 80% of C max and no more than 150% of C max of what a 0.3 mg intramuscular injection yields; a mean t max of less than 45 minutes; and absorption as IM injection under optimal dosing conditions in the thigh. [0033] In some embodiments, the nasal aerosol formulation when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC 04 are at least 80% of AUC 0-20min and AUC 04 of what an intramuscular injection of 0.15 mg yields; a mean C max that is at least 80% of the C max and no more than 150% of the C max of what an intramuscular injection of 0.15 mg yields; a mean t max of less than 45 minutes; or absorption as IM injection under optimal conditions of dosage on the thigh. In some embodiments, the nasal aerosol formulation when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC 0-t are at least 80% of AUC 0-20min and AUC 0-t of what an intramuscular injection of 0.15 mg yields; a mean C max that is at least 80% of the C max and no more than 150% of the C max of what an intramuscular injection of 0.15 mg yields; a mean W of less than 45 minutes; and absorption as IM injection under optimal dosing conditions in the thigh. [0034] In some embodiments, the nasal aerosol formulation when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC 0-t are at least 80% of the AUC 0-20min and AUC 0-t of what a 0.5 mg intramuscular injection yields; a mean C max that is at least 80% of C max and no more than 150% of C max of what a 0.5 mg intramuscular injection yields; a mean W of less than 45 minutes; or absorption as IM injection under optimal dosing conditions in the thigh. In some embodiments, the nasal aerosol formulation when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC 0-t are at least 80% of AUC 0-20min and AUC 0-t of what a 0.5 mg intramuscular injection yields; a mean C max that is at least 80% of C max and no more than 150% of C max of what a 0.5 mg intramuscular injection yields; a mean t max of less than 45 minutes; and absorption as IM injection under optimal dosing conditions in the thigh. [0035] In some embodiments, the nasal aerosol formulation comprises between about 0.5 and about 1.1 molar equivalents of acid for each mole of Epinephrine. In some embodiments, the acid is adipic acid, ammonium chloride, citric acid, acetic acid, hydrochloric acid, lactic acid, phosphoric acid, propionic acid, sulfuric acid, or tartaric acid. In some embodiments, the acid is hydrochloric acid. In some embodiments, no base is added to the nasal spray formulation during its preparation. In some embodiments, the nasal aerosol formulation has a pH between about 2.0 and about 6.0. In some embodiments, the nasal aerosol formulation has a pH of about 4.0. [0036] In some embodiments, the nasal aerosol formulation comprises between about 5 mg / mL and about 40 mg / mL per dose of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.9 mg and about 2.40 mg per dose dispensed from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.5 mg and about 2.0 mg per dose dispensed from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.9 mg and about 1.5 mg per dose. dispensed from the device Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.75 mg and about 1.5 mg per dose dispensed from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.45 mg and about 1.15 mg per dose dispensed from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 1.0 mg and about 2.0 mg per dose dispensed from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.5 mg and about 2.0 mg of Epinephrine, or a salt thereof, per dose dispensed from the device. In some embodiments, the nasal aerosol formulation comprises between about 0.5 mg and about 1.5 mg of Epinephrine, or a salt thereof, per dose dispensed from the device. In some embodiments, the nasal aerosol formulation comprises between about 0.5 mg and about 0.7 mg of Epinephrine, or a salt thereof, per dose dispensed from the device. In some embodiments, the nasal aerosol formulation comprises about 1.0 mg of Epinephrine, or a salt thereof, per dose dispensed from the device. In some embodiments, the nasal aerosol formulation comprises between about 1.3 mg and about 1.5 mg of Epinephrine, or a salt thereof, per dose dispensed from the device. [0037] In some embodiments, the nasal aerosol pharmaceutical formulation comprises one or more absorption enhancing agents; and optionally one or more agents selected from isotonicity agents; stabilizing agents; preservatives flavor masking agents; viscosity modifiers; buffer antioxidants and pH adjusting agents; wherein the pH of the nasal pharmaceutical aerosol formulation is between about 2.0 and about 6.0. [0038] In some embodiments, the nasal pharmaceutical aerosol formulation has a pH between about 3.0 and about 5.0. In some embodiments, the nasal aerosol pharmaceutical formulation has a pH of about 4.0. In some embodiments, the nasal aerosol pharmaceutical formulation comprises pH adjusting agents. In some embodiments, the pH adjusting agent is an acid, a base, a buffer, or a combination thereof. In some embodiments, the acid is adipic acid, ammonium chloride, citric acid, acetic acid, hydrochloric acid, lactic acid, phosphoric acid, propionic acid, sulfuric acid, or tartaric acid; the base is sodium hydroxide, sodium citrate, sodium bicarbonate, sodium carbonate; and the buffer is a phosphate buffer, acetate buffer, or citrate buffer. In some embodiments, the nasal aerosol pharmaceutical formulation comprises between about 0.5 and about 1.1 molar equivalents of acid for each mole of Epinephrine. In some embodiments, the acid is hydrochloric acid. [0039] In some embodiments, the nasal aerosol formulation comprises one or more absorption enhancers selected from dodecyl maltoside, benzalkonium chloride, oleic acid or a salt thereof, polysorbate 20, polysorbate 80, and sodium lauryl sulfate. [0040] In some embodiments, the formulation comprises one or more absorption enhancers selected from alcohol, aprotinin, benzalkonium chloride, benzyl alcohol, capric acid, ceramides, cetylpyridinium chloride, chitosan, cyclodextrins, deoxycholic acid, decanoyl, dimethyl sulfoxide, glyceryl monooleate. , glycofurol, glycofurol, glycosylated sphingosines, glycyrrhetinic acids, 2-Hydroxypropyl-p-cyclodextrin, laureth-9, lauric acid, lauroyl carnitine, lysophosphatidylcholine, menthol, poloxamer 407 or F68, poly-L-lauryl ether 9-polyoxyl ether , isopropyl myristate, isopropyl palmitate, lanolin, light mineral oil, linoleic acid, menthol, myristic acid, myristyl alcohol, oleic acid or a salt thereof, oleyl alcohol, palmitic acid, polysorbate 20, polysorbate 80, propylene glycol, alkyl ethers of polyoxyethylene, polyoxylglycerides, pyrrolidone, quillaia saponin, salicylic acid, sodium salt, p-sitosterol pD-glucoside, s lauryl sulfate Hate, sucrose cocoate, taurocholic acid, taurodeoxycholic acid, taurodihydrofussidic acid, thymol, tricaprylin, triolein, and alkylsaccharides. [0041] In some embodiments, the formulation comprises one or more absorption enhancers selected from dodecyl maltoside, benzalkonium chloride, oleic acid or a salt thereof, polysorbate 20, polysorbate 80, and sodium lauryl sulfate. [0042] In some embodiments, the one or more absorption enhancers are: from about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside; or from about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride; or from about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; or a combination of about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) of oleic acid, or salts thereof ; or a combination of about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof . [0043] In some embodiments, the one or more absorption enhancers are: from about 0.005% (w / v) to about 0.08% (w / v) benzalkonium chloride; or from about 0.01% (w / v) to about 0.06% (w / v) of benzalkonium chloride; or from about 0.01% (w / v) to about 0.04% (w / v) of benzalkonium chloride; wherein benzalkonium chloride is the only absorption enhancing agent in the formulation or is present in the formulation with one or more additional absorption enhancing agents. [0044] In some embodiments, the formulation comprises a preservative. In some embodiments, the preservative is benzalkonium chloride. [0045] In some embodiments, the nasal aerosol pharmaceutical formulation comprises an isotonicity agent. In some embodiments, the isotonicity agent is dextrose, glycerin, mannitol, potassium chloride, or sodium chloride. In some embodiments, the isotonicity agent is sodium chloride. [0046] In some embodiments, the nasal aerosol formulation additionally comprises a stabilizing agent. In some embodiments, the stabilizing agent is ethylenediaminetetra acetic acid (EDTA) or a salt thereof. In some embodiments, the EDTA is disodium EDTA. In some embodiments, the nasal aerosol formulation comprises from about 0.001% (w / v) to about 1% (w / v) of disodium EDTA. [0047] In some embodiments, the nasal aerosol formulation additionally comprises a preservative. In some embodiments, the preservative is benzalkonium chloride. [0048] In some embodiments, the nasal aerosol formulation comprises one or more absorption enhancers selected from alkylglycosides, benzalkonium chloride, oleic acid, or salts thereof, polysorbate 20, polysorbate 80, sodium lauryl sulfate, cyclodextrins, a medium chain of and long fatty acids, or their salts, saturated and unsaturated fatty acids, or their salts, alcohol, glycerin, propylene glycol, PEG 300/400 and benzyl alcohol. [0049] In some embodiments, the nasal aerosol formulation further comprises an antioxidant. In some embodiments, the nasal aerosol formulation further comprises an antioxidant selected from alpha tocopherol, arachidonic acid, ascorbic acid, ascorbyl palmitate, benzethonium chloride, benzethonium bromide, benzalkonium chloride, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT). ), capric acid, caproic acid, carbon dioxide, cetylpyridium chloride, chelating agents, chitosan derivatives, citric acid monohydrate, dodecyl dimethyl aminopropionate, enanthic acid, erythorbic acid, ethyl oleate, fumaric acid, glycerol oleate, Glyceryl monostearate, lauric acid, limonene, linolenic acid, lysine, malic acid, menthol, methionine, monothioglycerol, myristic acid, oleic acid, palmitic acid, pelargonic acid, peppermint oil, phosphoric acid, polysorbates, potassium metabisulfite, propionic acid , propyl gallate, sodium ascorbate, sodium bisulfite, sodium caprate, sodium deoxycholate, sodium deoxyglycolate, for sodium maldehyde sulfoxylate, sodium glycocholate, sodium hydroxybenzoyal aminocaprylate, sodium lauryl sulfate, sodium metabisulfite, sodium sulfite, sodium taurocholate, sodium thiosulfate, stearic acid, sulfur dioxide, and a combination thereof. [0050] In some embodiments, the nasal aerosol formulation further comprises synergists with the antioxidants selected from citric acid monohydrate, tartaric acid, thymol, tocopherol (alpha tocopherol), tocopherasol, vitamin E and vitamin E polyethylene glycol succinate and a combination of the themselves. [0051] In some embodiments, the nasal aerosol formulation further comprises enhancers Permeation agents selected from alcohol, arachidonic acid, benzethonium chloride, benzethonium bromide, benzalkonium chloride, capric acid, caproic acid, carvone, cetylpyridium chloride, chitosans, citric acid, 6-cyclohexyl-1-hexyl-pD-maltopyranoside, n-decyl-pD-maltopyranoside, dimethyl sulfoxide, dodecyl dimethyl aminopropionate, 1-en-dodecyl-pD-maltopyranoside, dodecylpolyethylglycol ether, disodium edetate dihydrate, enanthic acid, glyceryl monooleate, glyceryl isopropyl monostearate, glyceryl isopropylphurolate, glyceryl isopropyl ester isopropyl, pelargonic acid, lanolin, lauric acid, light mineral oil, limonene, oleic acid, lysine, menthol, myristic acid, myristyl alcohol, oleic acid, oleyl alcohol, palmitic acid, peppermint oil, polyoxyethylene alkyl ethers, polyoxylglycerides, polysorbates, pyrrolidone, sodium caprate, sodium deoxycholate, sodium deoxycholate, sodium glycocholate, sodium hydroxybenzoyal aminocaprylate, d-lauryl sulfate and sodium, sodium taurocholate, stearic acid, thymol, tricaprylin, triolein, undecylenic acid, and a combination thereof. [0052] In some embodiments, the nasal aerosol formulation comprises: about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside; from about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; from about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; a combination of about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; a combination of about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof : or a combination of about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) of oleic acid, or salts of the same and 0.001% to 1% sodium metabisulfite; or a combination of about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof and about 0.001% to 10% polysorbate 80 and 0.001% to 1% sodium metabisulfite; or a combination of about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof and approximately 0.001% to 10% polysorbate 80 and 0.001% to 1% sodium metabisulfite and 0.001% to 1% citric acid. [0053] In some embodiments, the nasal aerosol formulation comprises: from about 0.005% (w / v) to about 0.08% (w / v) of benzalkonium chloride; from about 0.01% (w / v) to about 0.06% (w / v) of benzalkonium chloride; or about 0.01% (w / v) to approximately 0.04% (w / v) of benzalkonium chloride; wherein benzalkonium chloride is the only absorption enhancing agent in the nasal aerosol formulation or is present in the formulation with one or more additional absorption enhancing agents. [0054] In some embodiments, the nasal aerosol formulation comprises: about 0.001% to 1% of any of the antioxidants described herein, or a combination of any of the antioxidants described herein. [0055] In some embodiments, the nasal aerosol formulation comprises a buffering agent. Buffering agents include, but are not limited to, adipic acid, boric acid, calcium carbonate, calcium hydroxide, calcium lactate, calcium phosphate, tribasic, citric acid monohydrate, dibasic sodium phosphate, diethanolamine, glycine, maleic acid, acid Malic, Methionine, Monobasic Sodium Phosphate, Monoethanolamine, Monosodium Glutamate, Phosphoric Acid, Potassium Citrate, Sodium Acetate, Sodium Bicarbonate, Sodium Borate, Sodium Carbonate, Sodium Citrate Dihydrate, Sodium Hydroxide, Sodium Lactate and triethanolamine. In one aspect, provided herein is a method of treating an adrenergic receptor-mediated condition comprising intranasal administration of any of the formulations as described herein. In some embodiments, the condition is chosen from a type 1 hypersensitivity reaction (systemic allergic reaction), an acute asthmatic attack, cardiac arrest, and Stokes-Adams syndrome. In some embodiments, the condition is a type 1 hypersensitivity reaction (systemic allergic reaction). In some embodiments, the type 1 hypersensitivity reaction is selected from allergic asthma, allergic conjunctivitis, allergic rhinitis, anaphylaxis, angioedema, urticaria, eosinophilia, drug allergy, and food allergy. In some embodiments, the drug allergy is an antibiotic allergy. [0056] In one aspect, described herein is a nasal spray formulation comprising Epinephrine or a salt thereof, which when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0- [0057] 20min and AUC 0-t are at least 80% of AUC 0-20min and AUC 0-t of what an intramuscular injection of 0.3 mg yields; a mean C max that is at least 80% of C max and no more than 150% of C max of what a 0.3 mg intramuscular injection yields; a mean W of less than 45 minutes; or absorption as IM injection under optimal dosing conditions in the thigh. In another aspect, a nasal spray formulation comprising Epinephrine or a salt thereof is described herein, which when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC 0-t are at least 80% of the AUC 0-20min and AUC 0-t of what an intramuscular injection of 0.3 mg yields; a mean C max that is at least 80% of C max and no more than 150% of C max than an intramuscular injection of 0.3 mg yields; a mean t max of less than 45 minutes; and absorption as IM injection under optimal dosing conditions in the thigh. [0058] In another aspect, a nasal spray formulation comprising Epinephrine or a salt thereof is described herein, which when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC ot are at least 80% of the AUC or -20min and AUC ot of what an intramuscular injection of 0.15 mg yields; a mean C max that is at least 80% of C max and no more than 150% of C max of what an intramuscular injection of 0.15 mg yields; a mean t max of less than 45 minutes; or absorption as IM injection under optimal dosing conditions in the thigh. In another aspect, a nasal spray formulation comprising Epinephrine or a salt thereof is described herein, which when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC 0-t are at least 80% of the AUC 0-20min and AUC 0-t of what an intramuscular injection of 0.15 mg yields; a mean C max that is at least 80% of C max and no more than 150% of C max of what an intramuscular injection of 0.15 mg yields; a mean t max of less than 45 minutes; and absorption as IM injection under optimal dosing conditions in the thigh. [0059] In yet another aspect, described herein is a nasal spray formulation comprising Epinephrine or a salt thereof, which when administered to a subject produces one or more of the following pharmacokinetic characteristics: both a mean AUC 0-20min and AUC 0-t are at least 80% of the AUC 0-20min and AUC 0-t of what an intramuscular injection of 0.5 mg yields; a mean C max that is at least 80% of C max and no more than 150% of C max of what an intramuscular injection of 0.5 mg yields; a mean t max of less than 45 minutes; or absorption as IM injection under optimal dosing conditions in the thigh. [0060] In some embodiments, the nasal spray formulation is a pharmaceutical formulation. In some embodiments, the Epinephrine or salts thereof are present in the nasal aerosol formulation in an amount effective for the treatment of an acute hypersensitivity reaction. [0061] In some embodiments, intranasal administration of a single dose of the nasal pharmaceutical aerosol formulation to a subject provides a concentration of Epinephrine in plasma that is effective for the treatment of an acute hypersensitivity reaction. In some embodiments, a single dose of the nasal aerosol pharmaceutical formulation comprises between about 0.5 mg and about 2.0 mg of Epinephrine, or a salt thereof. In some embodiments, a single dose of the nasal pharmaceutical aerosol formulation comprises between about 0.5 mg and about 1.5 mg of Epinephrine, or a salt thereof. In some embodiments, a single dose of the nasal pharmaceutical aerosol formulation comprises between about 0.5 mg and about 0.7 mg of Epinephrine, or a salt thereof. In some embodiments, a single dose of the nasal pharmaceutical aerosol formulation comprises about 1.0 mg of Epinephrine, or a salt thereof. In some embodiments, a single dose of the nasal pharmaceutical aerosol formulation comprises between about 1.3 mg and about 1.5 mg of Epinephrine, or a salt thereof. In some embodiments, the formulation is an aqueous solution, aqueous suspension, aqueous emulsion, non-aqueous solution, non-aqueous suspension, non-aqueous emulsion, pressure metered dose inhalers, or dry powder. [0062] In some embodiments, the nasal aerosol formulation is an aqueous solution, aqueous suspensions, aqueous emulsion, non-aqueous solution, non-aqueous suspension, or non-aqueous emulsion. [0063] In some embodiments, the nasal spray formulation has similar pharmacokinetics to intramuscular (IM) injection when IM injection is dosed in the lateral thigh, or absorption similar to subcutaneous (SC) or in the middle. [0064] In some embodiments, the nasal aerosol formulation has similar absorption to subcutaneous (SC) and the SC pharmacokinetic profile has a Cmax of at least 100 pg / mL and AUC0-240 min of 150 h * pg / mL. [0065] In some embodiments, the nasal aerosol formulation has intramuscular (IM) absorption similar to injection. [0066] In some embodiments, the nasal aerosol formulation comprises an absorption enhancer. [0067] In some embodiments, the nasal aerosol pharmaceutical formulation comprises one or more absorption enhancing agents; and optionally one or more agents selected from isotonicity agents; stabilizing agents; preservatives flavor masking agents; viscosity modifiers; buffer antioxidants and pH adjusting agents; wherein the pH of the nasal pharmaceutical aerosol formulation is between about 2.0 and about 6.0. In some embodiments, the nasal aerosol pharmaceutical formulation has a pH between about 3.0 and about 5.0. In some embodiments, the nasal aerosol pharmaceutical formulation has a pH of about 4.0. [0068] In some embodiments, the nasal aerosol pharmaceutical formulation comprises pH adjusting agents. In some embodiments, the pH adjusting agent is an acid, a base, a buffer, or a combination thereof. In some embodiments, the acid is adipic acid, ammonium chloride, citric acid, acetic acid, hydrochloric acid, lactic acid, phosphoric acid, propionic acid, sulfuric acid, or tartaric acid; the base is sodium hydroxide, sodium citrate, sodium bicarbonate, sodium carbonate; and the buffer is a phosphate buffer, acetate buffer, or citrate buffer. [0069] In some embodiments, the nasal aerosol formulation comprises between about 0.5 and approximately 1.1 molar equivalents of acid to each mole of Epinephrine. In some embodiments, the acid is adipic acid, ammonium chloride, citric acid, acetic acid, hydrochloric acid, lactic acid, phosphoric acid, propionic acid, sulfuric acid, or tartaric acid. In some embodiments, the acid is hydrochloric acid. In some embodiments, no base is added to the formulation during its preparation. In some embodiments, the nasal aerosol formulation has a pH between about 2.0 and about 6.0. In some embodiments, the nasal aerosol formulation has a pH of about 4.0. [0070] In some embodiments, the nasal aerosol formulation comprises between about 5 mg / mL and about 40 mg / mL per dose of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.40 mg and about 2.40 mg per dose dispensed from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.9 mg and about 2.40 mg per dispensed dose from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.5 mg and about 2.0 mg per dispensed dose from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.9 mg and about 1.5 mg per dispensed dose from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.75 mg and about 1.5 mg per dispensed dose from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 0.45 mg and about 1.15 mg per dose dispensed from the device of Epinephrine, or a salt thereof. In some embodiments, the nasal aerosol formulation comprises between about 1.0 mg and about 2.0 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0071] In some embodiments, the nasal spray formulation comprises one or more absorption enhancers selected from alcohol, aprotinin, benzalkonium chloride, benzyl alcohol, capric acid, ceramides, cetylpyridinium chloride, chitosan, cyclodextrins, deoxycholic acid, decanoyl, dimethyl sulfoxide. , glyceryl monooleate, glycofurol, glycofurol, glycosylated sphingosines, glycyrrhetinic acids, 2-Hydroxypropyl-p-cyclodextrin, laureth-9, lauric acid, lauroyl carnitine, lysophosphatidylcholine, menthol, poloxamer 407 or polyox-arginine, polyol-arginine- 9-lauryl ether, isopropyl myristate, isopropyl palmitate, lanolin, light mineral oil, oleic acid, menthol, myristic acid, myristyl alcohol, oleic acid, oleyl alcohol, palmitic acid, polysorbate 20, polysorbate 80, propylene glycol, polyoxyethylene alkyl ethers , polyoxylglycerides, pyrrolidone, quillaia saponin, salicylic acid, sodium salt, p-sitosterol pD-glucoside, s lauryl sulfate Hate, Sucrose Cocoate, Taurocholic Acid, Taurodeoxycholic Acid, taurodihydrofusidic acid, thymol, tricapryline, triolein, and alkylsaccharides. [0072] In some embodiments, the nasal spray formulation comprises one or more absorption enhancers selected from dodecyl maltoside, benzalkonium chloride, oleic acid, or salts thereof, polysorbate 20, polysorbate 80, and sodium lauryl sulfate. [0073] In some embodiments, the nasal aerosol formulation comprises: from about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside; from about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; from about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; a combination of about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; a combination of about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof and about 0.001 to 1% of an antioxidant (eg, sodium metabisulfite). In some embodiments, the nasal aerosol formulation comprises: from about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside; from about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; from about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; a combination of about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; a combination of about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof . In some embodiments, the nasal aerosol formulation comprises: from about 0.005% (w / v) to about 0.08% (w / v) of benzalkonium chloride; from about 0.01% (w / v) to about 0.06% (w / v) of benzalkonium chloride; or from about 0.01% (w / v) to about 0.04% (w / v) of benzalkonium chloride; wherein benzalkonium chloride is the sole absorption enhancing agent in the nasal spray formulation or is present in the formulation with one or more additional absorption enhancing agents. [0074] In some embodiments, the nasal spray formulation additionally comprises a stabilizing agent. In some embodiments, the stabilizing agent is ethylenediaminetetra acetic acid (EDTA) or a salt thereof. In some embodiments, the EDTA is disodium EDTA. In some embodiments, EDTA is present in an amount that is about 0.001% to about 1%. [0075] In some embodiments, the nasal spray formulation additionally comprises a preservative. In some embodiments, the preservative is benzalkonium chloride. [0076] In one aspect, a method of treating an adrenergic receptor-mediated condition comprising intranasal administration of any of the formulations described herein is described herein. In some embodiments, the condition is chosen from a type 1 hypersensitivity reaction (systemic allergic reaction), an acute asthmatic attack, cardiac arrest, and Stokes-Adams syndrome. In some embodiments, the condition is a type 1 hypersensitivity reaction (systemic allergic reaction). In some embodiments, the type 1 hypersensitivity reaction is selected from allergic asthma, allergic conjunctivitis, allergic rhinitis, anaphylaxis, angioedema, urticaria, eosinophilia, drug allergy, and food allergy. In some embodiments, the drug allergy is an antibiotic allergy. [0077] In another aspect, described herein is a method of treating anaphylaxis comprising intranasal administration of an intranasal formulation of Epinephrine in an amount less than about 2.0 mg. In some embodiments, the nasal pharmaceutical formulation comprises between about 0.5 mg and about 1.5 mg of Epinephrine, or a salt thereof. In some embodiments, the nasal pharmaceutical formulation comprises between about 0.5 mg and about 0.7 mg of Epinephrine, or a salt thereof. In some embodiments, the nasal pharmaceutical formulation comprises about 1.0 mg of Epinephrine, or a salt thereof. In some embodiments, the nasal pharmaceutical formulation comprises between about 1.3 mg and about 1.5 mg of Epinephrine, or a salt thereof. [0078] In some embodiments of the treatment methods, the intranasal formulation comprises: one or more absorption enhancing agents; an isotonicity agent; a stabilizing agent; a preservative an optional antioxidant; and optional pH adjusting agents. In some embodiments of the treatment methods, the one or more absorption enhancing agents are selected from: dodecyl maltoside; benzalkonium chloride; oleic acid, or salts thereof; sodium lauryl sulfate; a combination of dodecyl maltoside and benzalkonium chloride; a combination of dodecyl maltose and oleic acid, or salts thereof; and a combination of benzalkonium chloride and oleic acid, or salts thereof. In some embodiments of the treatment methods, the one or more absorption enhancing agents are selected from: about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside; from about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; from about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; a combination of about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride; a combination of about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof . In some embodiments of the treatment methods, the formulation comprises: from about 0.005% (w / v) to about 0.08% (w / v) of benzalkonium chloride; from about 0.01% (w / v) to about 0.06% (w / v) of benzalkonium chloride; or from about 0.01% (w / v) to about 0.04% (w / v) of benzalkonium chloride; wherein benzalkonium chloride is the only absorption enhancing agent in the formulation or is present in the formulation with one or more absorption enhancing agents. In some embodiments of the treatment methods, the isotonicity agent is sodium chloride. In some embodiments of the treatment methods, the stabilizing agent is EDTA. In some embodiments of the treatment methods, the stabilizing agent is EDTA in an amount from about 0.001% (w / v) to about 1% (w / v). In some embodiments of the treatment methods, the preservative is benzalkonium chloride. In some embodiments of the treatment methods, the preservative is benzalkonium chloride in an amount from about 0.001% (w / v) to about 1% (w / v). [0079] Articles of manufacture, including packaging material, a nasal spray formulation described herein within the packaging material, a label indicating that the nasal spray formulation is used for the treatment of any of the Conditions described in this document (eg, anaphylaxis) are provided. [0080] Other objects, features, and advantages of the compositions and methods described herein will be apparent from the following detailed description. However, it should be understood that the detailed description and specific examples, while indicating specific embodiments, are given by way of illustration only, since various changes and modifications within the spiritual scope of instant disclosure will be apparent to those skilled in the art of this detailed description. [0082] BRIEF DESCRIPTION OF THE FIGURES [0083] Figure 1 shows the area under the plasma concentration versus time curve in time 0-120 minutes (AUC0-120 min) of absorption of Epinephrine at different doses and routes of administration as described in Srisawat et al., "A preliminary study of intranasal Epinephrine administration as a potential route for anaphylaxis treatment, " Asian Pac J Allergy Immunol, 2016 Mar; 34 (1): 38-43, described below. [0084] Figure 2 shows the graphs of plasma Epinephrine concentration versus time after administration of IN saline, 5 mg IN Epinephrine, and 0.3 mg IM Epinephrine, as described in Srisawat et al. [0085] Figure 3 shows the mean plasma Epinephrine concentrations above the baseline curves (pg / mL) versus time (min) of the first clinical study described in Example 2A comparing 0.3 mg of IM and IN Epinephrine; the curve with the square represents IM, and the curve with circles, IN. [0086] Figure 4 shows the mean plasma Epinephrine concentrations above baseline curves (pg / mL) versus time (min) from the second clinical study described in Example 2B comparing 0.5, 1.0. and 2.0 mg of Epinephrine IN. The circled curve represents 0.5 mg; the curve with triangles, 1.0 mg, and the curve with squares, 2.0 mg. [0087] Figure 5 shows the first 30 minutes of mean plasma Epinephrine concentrations above baseline curves (pg / mL) versus time (min) of the second clinical study described in Example 2B comparing 0.5, 1.0. and 2.0 mg of Epinephrine IN. The circled curve represents 0.5 mg; the curve with triangles, 1.0 mg, and the curve with squares, 2.0 mg. [0088] Figure 6 repeats the data from Figure 4 and overlays it with the mean plasma concentration versus time curve for IM Epinephrine from the first clinical study described in Example 2A. [0089] Figure 7 repeats the data from Figure 5 and overlaps with the first 30 minutes of the mean plasma concentration versus time curve for IM Epinephrine from the first clinical study described in Example 2A. [0090] Figure 8 shows mean plasma Epinephrine concentrations above baseline curves (pg / mL) versus time (min) of the comparative bioavailability portion of the second clinical study described in Example 2B, comparing 1.0 mg of IN Epinephrine ( curve w / circles) to 0.3 mg Epinephrine IM (Epinephrine autosampler, curve w / triangles). [0091] Figure 9 shows the first 30 minutes of mean plasma Epinephrine concentrations above baseline curves (pg / mL) versus time (min) for the comparative bioavailability portion of the second clinical study described in Example 2B, comparing 1.0 mg of Epinephrine IN (curve w / circles) to 0.3 mg of Epinephrine IM (EpiPen®, curve w / triangles). [0093] DETAILED DESCRIPTION [0094] Described herein are methods and formulations useful for the treatment of anaphylaxis and other conditions, which comprise administering an intranasal formulation of Epinephrine. Devices adapted for the nasal delivery of a pharmaceutical formulation to a patient are also provided, including single, multiple dose, and multi-dose delivery comprising a therapeutically effective amount of Epinephrine and its salts. pharmaceutically acceptable. [0095] Intranasal epinephrine has a long history of use in low doses as a decongestant and as a vasoconstrictor, often formulated in combination with an anesthetic, in nasal and nasal surgery. Historically, Epinephrine has been difficult to formulate as an intranasal solution for systemic delivery. See, for example, Srisawat C et al., "A preliminary study of intranasal Epinephrine administration as a potential route for anaphylaxis treatment," Asian Pac J Allergy Immunol, 2016 Mar; 34 (1): 38-43. Srisawat showed that significant systemic absorption of Epinephrine via the IN route was observed only at 5 mg (see Figure 1) and the pharmacokinetic parameters of Epinephrine IN even at 5 mg were also not significantly different from those of the Epinephrine IM group ( see Table 1. below). Table 1. Intramuscular and Intranasal Administration of Epinephrine (from Srisawat (2016). [0100] Figure 1 is reproduced from Srisawat C et al. and shows that Srisawat et al. A blood level of Epinephrine was not observed at the 2.5 mg intranasal dose level and a cotinuation. [0101] Furthermore, Figure 2 shows that even at a 5 mg dose, Srisawat was unable to make an intranasal formulation that could achieve a higher plasma concentration than intramuscular epinephrine administered by autoinjector at any time before approximately 60 minutes, therefore, absorption during early critical points was delayed when rapid absorption is needed to stop the systemic allergic reaction (anaphylaxis). This is potentially detrimental in severe conditions, such as anaphylaxis, where immediate treatment, and therefore injection-like pharmacokinetics, is desirable. The PK profile of IM injection in the thigh is considered the optimal dosage method by the literature, since the greater vascularization of the leg muscle allows a faster absorption and distribution of Epinephrine, providing a rapid increase in plasma levels. to stop the anaphylaxis reaction much earlier. than other routes of administration. Figure 2 also shows that the Srisawat 5 mg formulation, in contrast to intramuscular epinephrine administered by autoinjector, was almost completely cleared from plasma in approximately two hours. Finally, Epinephrine is known to be associated with dose-related cardiac side effects, including myocardial infarction, at doses as low as 0.3 to 0.5 mg intramuscularly; accordingly, dose as high as 5 mg would likely be risky in the general population if nasal conditions exist that allow excessive absorption. Therefore, lower dose preparations that would avoid such risks are preferred as a safer nasal preparation. [0102] Described herein are intranasal formulations of Epinephrine, and nasal spray devices comprising the formulations, that solve the problems of past attempts. Various aspects can contribute to the success of the formulations, devices, and methods of use described herein. [0103] For example, in certain embodiments, formulating Epinephrine in an aqueous solution with the appropriate addition of acid molar equivalents to each mole of said Epinephrine helps to solubilize and stabilize Epinephrine. This allows the formulation to avoid the use of buffering agents commonly used in aqueous pharmaceutical compositions for injection, including phosphate, acetate, and citrate buffers, which are sometimes avoided in the nasal formulations described herein. Other salts of Epinephrine, such as Epinephrine acetate, Epinephrine hydrochloride, Epinephrine tartrate, Epinephrine bitartrate, Epinephrine hydrogen tartrate, and Epinephrine borate can also be used to formulate aqueous solutions of Epinephrine. [0104] Certain embodiments of the formulations, devices, and methods of use described herein offer advantages over Epinephrine formulated in other ways. Epinephrine is considered a narrow therapeutic index drug. As a sympathomimetic catecholamine, epinephrine has a narrow therapeutic index and serious adverse reactions including cardiovascular and cerebrovascular reactions may be associated with its use. However, the use of Epinephrine for this indication saves lives and the benefits of its use outweigh the potential safety risks. The intranasal formulation and administration is suitable for the safe and painless administration of drugs such as Epinephrine because of the constant uniformity of the content, the amount of administration and the absorption, thus minimizing serious adverse reactions, including cardiovascular and cerebrovascular reactions that can be associated with its use through injection mechanisms. Injection weights have low variability and consistently deliver the labeled dose. [0105] In one aspect, a pharmaceutical composition comprising: a) Epinephrine; and b) an alkylglycoside; wherein the pharmaceutical composition is formulated for administration into the circulatory system of a subject via the intranasal, inhalation, or pulmonary route of administration. In some embodiments, a pharmaceutical composition comprising: a) Epinephrine; and b) an alkylglycoside; wherein the pharmaceutical composition is a liquid formulated for intranasal administration. [0106] In some embodiments, the alkylglycoside has an alkyl chain that includes between 8 and 20 carbons. In some embodiments, the alkylglucoside is selected from the group consisting of undecyl maltoside, dodecyl maltoside, tridecyl maltoside, tetradecyl maltoside, sucrose mono dodecanoate, sucrose mono tridecanoate, and sucrose mono tetradecanoate. In some embodiments, the alkylglycoside is dodecyl-beta-D-maltoside. In some embodiments, the alkylglycoside concentration is between about 0.001% and 10.0% (w / v). In some embodiments, the alkylglycoside concentration is between about 0.05% and 0.5% (w / v). [0107] In some embodiments, the composition further comprises a membrane penetration enhancing agent. In some embodiments, the membrane penetration enhancing agent is a surfactant, a bile salt, a phospholipid, an alcohol, an enamine, a long chain amphipathic molecule, a small hydrophobic molecule, sodium or a derivative of salicylic acid, a glycerol ester of acetoacetic acid, a cyclodextrin, medium or long chain fatty acids, a chelating agent, an amino acid or salts thereof, an enzyme or a combination thereof. In some embodiments, the membrane penetration enhancing agent is selected from the group consisting of citric acid, sodium citrate, propylene glycol, glycerin, ascorbic acid, sodium metabisulfite, ethylenediaminetetra acetic acid (EDTA) disodium, benzalkonium chloride, hydroxyquinolone, sodium hydroxide, and combinations thereof. In some embodiments, the membrane penetration enhancing agent is selected from the group consisting of citric acid, sodium citrate, propylene glycol, glycerin, ascorbic acid, sodium metabisulfite, ethylenediaminetetra acetic acid (EDTA) disodium, benzalkonium chloride, sodium hydroxide, and combinations thereof. In some embodiments, the membrane penetration enhancing agent is benzalkonium chloride, EDTA, or a combination thereof. [0108] In some embodiments, the composition provides a C max for Epinephrine in a subject that is approximately 2-fold or greater compared to administration without alkylglycoside. [0109] In some embodiments, the composition provides a T max for Epinephrine in a subject that is approximately 2-fold or less compared to administration without alkylglycoside. [0110] In some embodiments, the composition provides a T max for Epinephrine of about 0.3 hours or less in a subject. [0111] In some embodiments, the composition has a pH of about 2.0 to 6.0. In some embodiments, the composition has a pH of about 2.0 to 5.0. [0112] In another aspect, a method for increasing the bioavailability of Epinephrine in a subject is described herein which comprises administering to a subject a pharmaceutical composition comprising Epinephrine and an alkylglycoside, thereby increasing the bioavailability of Epinephrine in the subject; wherein the pharmaceutical composition is formulated for administration into the circulatory system of a subject via the intranasal, inhalation, or pulmonary route of administration. In some embodiments, a method of increasing the bioavailability of Epinephrine in a subject is described herein which comprises administering to a subject a pharmaceutical composition comprising Epinephrine and an alkylglycoside, thereby increasing the bioavailability of Epinephrine in the subject; wherein the pharmaceutical composition is a liquid formulated for intranasal administration. In some embodiments, increasing the bioavailability of Epinephrine allows lower dose amounts of epeinpehrine to be administered intramuscularly and to be effective in treating anaphylaxis. In some embodiments, exposure to larger doses of Epinephrine can cause an Epinephrine overdose. There is a growing interest and need to develop alternative non-invasive Epinephrine dosage forms that provide plasma concentrations of Epinephrine equivalent to those obtained by Epinephrine autoinjectors, available in a range of doses, have a long shelf life, and are anxiety free by needles, possibility of administration error, unintentional injection and injuries. The nasal dosage forms of Epinephrine described in this document offer the potential to be non-invasive and easy-to-use alternatives for the emergency first aid treatment of anaphylaxis in community settings. [0113] In some embodiments, the alkylglycoside has an alkyl chain that includes between 8 and 20 carbons. In some embodiments, the alkylglucoside is selected from the group consisting of undecyl maltoside, dodecyl maltoside, tridecyl maltoside, tetradecyl maltoside, sucrose mono dodecanoate, sucrose mono tridecanoate, and sucrose mono tetradecanoate. In some embodiments, the alkylglycoside is dodecyl-beta-D-maltoside. In some embodiments, the alkylglycoside concentration is between about 0.001% and 10.0% (w / v). In some embodiments, the alkylglycoside concentration is between about 0.05% and 0.5% (w / v). [0114] In some embodiments, the composition further comprises a membrane penetration enhancing agent. In some embodiments, the membrane penetration enhancing agent is a surfactant, a bile salt, a phospholipid, an alcohol, an enamine, a medium and / or long chain amphipathic molecule, a hydrophobic small molecule, sodium, or a derivative. of salicylic acid, a glycerol acetoacetic acid ester, a cyclodextrin, medium or long chain fatty acids, a chelating agent, an amino acid or salts thereof, an enzyme or a combination thereof. In some embodiments, the membrane penetration enhancing agent is selected from the group consisting of citric acid, sodium citrate, propylene glycol, glycerin, ascorbic acid, sodium metabisulfite, ethylenediaminetetra acetic acid (EDTA) disodium, benzalkonium chloride, sodium hydroxide, and combinations thereof. In some embodiments, the penetration enhancing agent of the membrane is benzalkonium chloride, EDTA, or a combination thereof. [0115] In some embodiments, the composition provides a C max for Epinephrine in the subject that is approximately 2-fold or greater compared to administration without alkylglycoside. [0116] In some embodiments, the composition provides a T max for Epinephrine in the subject that is approximately 2-fold or less compared to administration without alkylglycoside. [0117] In some embodiments, the composition provides a Tmax for Epinephrine of about 0.3 hours or less in the subject. [0118] In some embodiments, the composition has a pH of about 2.0 to 6.0. In some embodiments, the composition has a pH of about 2.0 to 5.0. [0119] In some embodiments, the compositions described herein are liquid compositions suitable for intranasal administration. [0120] In one aspect, the invention provides a method of increasing the absorption of Epinephrine into the circulatory system of a subject by administering, nasal, inhalation, or pulmonary delivery, of a composition comprising: (a) Epinephrine; (b) an increasing amount of absorption of a suitable non-toxic nonionic alkylglycoside having a hydrophobic alkyl group linked by a bond to a hydrophilic saccharide; and (c) a mucosal release enhancing agent. [0121] The term "mucosal delivery enhancing agent" includes agents that enhance release or solubility (eg, of a formulation delivery vehicle), rate of diffusion, penetration capacity and time, absorption, residence time, stability, effective half-life, peak or sustained concentration levels, clearance, and other desired mucosal release characteristics (for example, measured at the site of delivery, or at a selected target site of activity, such as the bloodstream or system central nervous system) of a compound (s) (e.g., biologically active compound). Improved mucosal release can occur by any of a variety of mechanisms, including, for example, increasing diffusion, transport, persistence or stability of the compound, increasing membrane fluidity, modulating availability, or the action of calcium and other ions that regulate intracellular or paracellular permeation, solubilizing components of the mucous membrane (for example, lipids), changing the levels of sulfhydryl of non-protein protein and in the mucosal tissues, increasing the flow of water through the mucosal surface, modulating the physiology of the epithelial junction, reducing the viscosity of the mucosa that covers the mucosal epithelium, reducing the rates of mucociliary clearance, and other mechanisms. [0122] Examples of mucosal delivery enhancing agents include the following agents and any combination thereof: [0123] (a) an aggregation inhibitor agent; [0124] (b) a charge modifying agent; [0125] (c) a pH control agent; [0126] (d) a degrading enzyme inhibitory agent; [0127] (e) a mucolytic or mucosa-removing agent; [0128] (f) a ciostatic agent; [0129] (g) a membrane penetration enhancing agent selected from: [0130] (i) a surfactant; [0131] (ii) a bile salt; [0132] (iii) a phospholipid additive, mixed micelle, liposome, or vehicle; [0133] (iv) an alcohol; [0134] (v) an enamine; [0135] (vi) a non-donor compound; [0136] (vii) a long chain amphipathic molecule; [0137] (viii) a small hydrophobic penetration enhancer; [0138] (ix) sodium or a derivative of salicylic acid; [0139] (x) a glycerol ester of acetoacetic acid; [0140] (xi) a cyclodextrin or beta-cyclodextrin derivative; [0141] (xii) a medium chain fatty acid; [0142] (xiii) a chelating agent; [0143] (xiv) an amino acid or salts thereof; [0144] (xv) an N-acetylamino acid or salts thereof; [0145] (xvi) an enzyme degrading a selected membrane component; [0146] (xvii) an inhibitor of fatty acid synthesis; [0147] (xviii) an inhibitor of cholesterol synthesis; Y [0148] (ix) any combination of the membrane penetration enhancing agents mentioned in (i) - (x); [0149] (h) a modulating agent of the physiology of the epithelial junction; [0150] (i) a vasodilator agent; [0151] (j) a selective transport enhancing agent; Y [0152] (k) a stabilizing delivery vehicle, carrier, mucoadhesive, support, or complex-forming species with which the compound combines, associates, contains, encapsulates, or binds effectively resulting in stabilization of the compound for improved delivery of the compound. nasal mucosa, wherein formulation of the compound with the intranasal administration enhancing agents provides increased bioavailability of the compound in the blood plasma of a subject. [0153] Additional mucosal delivery enhancing agents include, for example example, citric acid, sodium citrate, propylene glycol, glycerin, ascorbic acid (for example, L-ascorbic acid), sodium metabisulfite, ethylenediaminetetra acetic acid (EDTA) disodium, benzalkonium chloride, sodium hydroxide, and mixtures thereof . For example, EDTA or its salts (eg, sodium or potassium) are used in amounts ranging from about 0.01% to 2% by weight of the alkylsaccharide preservative-containing composition. [0154] In yet another aspect, disclosed herein is a pharmaceutical composition having a suitable non-toxic nonionic alkylglycoside having a hydrophobic alkyl group bonded to a hydrophilic saccharide in combination with a selected mucosal delivery enhancing agent. from: [0155] (a) an aggregation inhibitor agent; [0156] (b) a charge modifying agent; [0157] (c) a pH control agent; [0158] (d) a degrading enzyme inhibitory agent; [0159] (e) a mucolytic or mucosa-removing agent; [0160] (f) a ciostatic agent; [0161] (g) a membrane penetration enhancing agent selected from: [0162] (i) a surfactant; [0163] (ii) a bile salt; [0164] (iii) a phospholipid additive, mixed micelle, liposome, or vehicle; [0165] (iv) an alcohol; [0166] (v) an enamine; [0167] (vi) a non-donor compound; [0168] (vii) a long chain amphipathic molecule; [0169] (viii) a small hydrophobic penetration enhancer; [0170] (ix) sodium or a derivative of salicylic acid; [0171] (x) a glycerol ester of acetoacetic acid; [0172] (xi) a cyclodextrin or beta-cyclodextrin derivative; [0173] (xii) a medium chain fatty acid; [0174] (xiii) a chelating agent; [0175] (xiv) an amino acid or salts thereof; [0176] (xv) an N-acetylamino acid or salts thereof; [0177] (xvi) an enzyme degrading a selected membrane component; [0178] (xvii) an inhibitor of fatty acid synthesis; [0179] (xviii) an inhibitor of cholesterol synthesis; Y [0180] (ix) any combination of the membrane penetration enhancing agents mentioned in (i) - (x); [0181] (h) a modulating agent of the physiology of the epithelial junction; [0182] (i) a vasodilator agent; [0183] (j) a selective transport enhancing agent; Y [0184] (k) a stabilizing delivery vehicle, carrier, mucoadhesive, support, or complex-forming species with which the compound combines, associates, contains, encapsulates, or binds effectively resulting in stabilization of the compound for improved delivery of the compound. nasal mucosa, wherein formulation of the compound with the intranasal administration enhancing agents provides increased bioavailability of the compound in the blood plasma of a subject. [0185] In another embodiment, a method of administering an alkylglycoside composition is described herein by administering a therapeutically effective amount of at least one alkylglycoside having an alkyl chain length of from about 12 to about 14 carbon atoms, at least one saccharide. with an antibacterial activity and Epinephrine. [0186] In one aspect, provided herein is an antibacterial alkylsaccharide composition, including n-dodecyl-4-O-a-D-glucopyranosyl-p-D-glucopyranoside or n-tetradecyl-4-O-a-D-glucopyranosyl-p-D-glucopyranoside. [0187] Accordingly, this document provides Embodiment 1. a nasal spray formulation comprising between about 0.40 mg and about 2.40 mg per dispensed dose from the Epinephrine device, or a salt thereof. Alternative Embodiment 1. A nasal spray formulation comprising between about 0.40 mg and about 2.0 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0188] Embodiment 2. The nasal aerosol formulation as described in Embodiment 1. wherein the formulation is a pharmaceutical formulation. [0189] Embodiment 3. The nasal aerosol formulation as described in Embodiment 2. wherein Epinephrine or salts thereof is present in the pharmaceutical formulation in an amount effective for treating an acute hypersensitivity reaction. [0190] Embodiment 4. The nasal aerosol formulation as described in any of Embodiments 1-3, wherein the formulation is aqueous. [0191] Embodiment 5. The nasal aerosol formulation as described in any of Embodiments 1-4, wherein the formulation comprises an absorption enhancer. In an alternative Embodiment 5. The nasal aerosol formulation as described in any of Embodiments 1-4, wherein the formulation comprises one or more absorption enhancers. [0192] Embodiment 6. The nasal aerosol formulation as described in any of Embodiments 1-5, wherein the formulation has intramuscular (IM) absorption similar to injection or subcutaneous (SQ), or in between. [0193] Embodiment 7. Nasal aerosol formulation as described in Embodiment 6, wherein it has intramuscular (IM) absorption similar to injection. [0194] Embodiment 8. The nasal aerosol formulation as described in Embodiment 6, wherein the formulation has subcutaneous (SC) -like absorption. [0195] Embodiment 9. Nasal aerosol formulation as described in Embodiment 8 where the SC pharmacokinetic profile has a Cmax of at least 100 pg / mL and AUC0-240min of 150 h * pg / mL. [0196] Embodiment 10. The nasal aerosol formulation as described in any of Embodiments 1-9, wherein the formulation, when administered to a subject, produces one or more of the following pharmacokinetic characteristics: [0197] • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.3 mg yields; [0198] • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.3 mg yields; [0199] • a mean W of less than 45 minutes; Y [0200] • an absorption similar to IM injection under optimal dosing conditions in the thigh. [0201] Embodiment 11. The nasal aerosol formulation as described in any of Embodiments 1-9, wherein the formulation, when administered to a subject, produces one or more of the following pharmacokinetic characteristics: [0202] • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.15 mg yield; [0203] • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.15 mg yields; [0204] • a mean W of less than 45 minutes; Y [0205] • an absorption similar to IM injection under optimal dosing conditions in the thigh. [0206] Embodiment 12. The nasal aerosol formulation as described in any of Embodiments 1-9, wherein the formulation, when administered to a subject, produces one or more of the following pharmacokinetic characteristics: [0207] • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.5 mg yield; [0208] • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.5 mg yields; [0209] • a mean tmax of less than 45 minutes; Y [0210] • an absorption similar to IM injection under optimal dosing conditions in the thigh. [0211] Embodiment 13. The nasal aerosol formulation as described in Embodiment 1-12, wherein the formulation, when administered to a subject, produces a t max of less than 40 minutes, a t max of less than 35 minutes, a t max between 30 and 45 minutes, a t max between 30 and 40 minutes, or a t max between 30 and 35 minutes. An alternative Embodiment 13. Nasal aerosol formulation as described in Embodiment 1-12, wherein the formulation, when administered to a subject, produces a t max of less than 40 minutes, a t max of less than 35 minutes , a t max of between 15 and 45 minutes, a t max of between 20 and 45 minutes, a t max of between 25 and 45 minutes, a t max of between 30 and 45 minutes, a t max of between 30 and 40 minutes , a t max of between 30 and 35 minutes, a t max of between 15 and 20 minutes, a t max of between 15 and 25 minutes, or a t max of between 15 and 30 minutes. [0212] Embodiment 14. The nasal aerosol formulation as described in any of Embodiments 1-13, wherein the formulation comprises less than one molar equivalency of acid for each mole of Epinephrine. [0213] Embodiment 15. The nasal aerosol formulation as described in any of Embodiments 1-13, wherein the formulation comprises between about 0.5 and about 1.1 molar equivalents of acid in each mole of Epinephrine. [0214] Embodiment 16. The nasal aerosol formulation is as described in any of Embodiments 14 and 15, wherein the acid is hydrochloric acid. An alternative Embodiment 16, wherein the nasal aerosol formulation is as described in any of Embodiments 14 and 15, wherein the acid is acetic acid, adipic acid, ammonium chloride, boric acid, citric acid, hydrochloric acid, acid lactic acid, phosphoric acid, propionic acid, sulfuric acid, or tartaric acid. [0215] Embodiment 17. The nasal aerosol formulation as described in any of Embodiments 1-16, wherein the formulation has a pH between about 3.0 and about 6.0. An Alternative Embodiment 17. The nasal aerosol formulation as described in any of Embodiments 1-16, wherein the formulation has a pH between about 2.0 and about 6.0. [0216] Embodiment 18. The nasal aerosol formulation as described in Embodiment 17, wherein the formulation has a pH between about 3.5 and about 5.0. [0217] Embodiment 19. The nasal aerosol formulation as described in Embodiment 17, wherein the formulation has a pH between about 4.0 and about 4.5. [0218] Embodiment 20, The nasal aerosol formulation as described in Embodiment 17, wherein the formulation has a pH of about 4.5. [0219] Embodiment 21. The nasal aerosol formulation as described in Embodiment 17, wherein the formulation has a pH of about 4.0. [0220] Embodiment 22. Nasal aerosol formulation as described in any of the Embodiments 1-21, wherein the formulation comprises between about 5 mg / mL and about 40 mg / mL of Epinephrine, or a salt thereof. An alternative Embodiment 22. The nasal aerosol formulation as described in any of Embodiments 1 21, wherein the formulation comprises between about 3 mg / mL and about 40 mg / mL of Epinephrine, or a salt thereof. [0221] Embodiment 23. The nasal aerosol formulation as described in any of Embodiments 1-22, wherein the formulation comprises between about 0.9 mg and about 2.4 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0222] Embodiment 24. The nasal aerosol formulation as described in any of Embodiments 1-22, wherein the formulation comprises between about 0.5 mg and about 2.0 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0223] Embodiment 25. The nasal aerosol formulation as described in any of Embodiments 1-22, wherein, the formulation comprises between about 0.75 mg and about 1.5 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0224] Embodiment 26. The nasal aerosol formulation as described in any of Embodiments 1-22, wherein the formulation comprises between about 0.9 mg and about 1.15 mg per dispensed dose from the Epinephrine device, or a salt thereof. [0225] Embodiment 27. The nasal aerosol formulation as described in any of Embodiments 1-22, wherein, the formulation comprises approximately 1.0 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0226] Embodiment 28. The nasal aerosol formulation as described in any of Embodiments 1-22, wherein the formulation comprises between about 0.45 mg and about 1.15 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0227] Embodiment 29. The nasal aerosol formulation as described in any of Embodiments 1-22, wherein the formulation comprises between about 1.0 mg and about 2.0 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0228] Embodiment 30. The nasal aerosol formulation as described in any of Embodiments 1-29, wherein the formulation further comprises a stabilizing agent. [0229] Embodiment 31. The nasal aerosol formulation as described in Embodiment 30. wherein the stabilizing agent is ethylenediaminetetra acetic acid (EDTA) or a salt thereof. [0230] Embodiment 32. The nasal aerosol formulation as described in Embodiment 31, wherein the EDTA is disodium EDTA. [0231] Embodiment 33. The nasal aerosol formulation as described in Embodiment 31 or 32, wherein EDTA is present in an amount that is 5% to 15% of the amount of Epinephrine, both measured in mmol. An alternative Embodiment 33. The nasal aerosol formulation as described in Embodiment 31 or 32, wherein EDTA is present in an amount that is 0.001% (w / v) to 1% (w / v). [0232] Embodiment 34. The nasal aerosol formulation as described in Embodiment 31 or 32, wherein the mmol of EDTA is approximately 10% of the mmol of Epinephrine. [0233] Embodiment 35. The nasal aerosol formulation as described in any of Embodiments 1-34, wherein the formulation further comprises a preservative. [0234] Embodiment 36. The nasal aerosol formulation as described in Embodiment 35, wherein the preservative is benzalkonium chloride. [0235] Embodiment 37. The nasal aerosol formulation as described in any of Embodiments 1-34, wherein the formulation further comprises an absorption enhancer. [0236] Embodiment 38. The nasal aerosol formulation as described in Embodiment 37, wherein the absorption enhancer is an alkylsaccharide. [0237] Embodiment 39. The nasal aerosol formulation as described in Embodiment 38, wherein the absorption enhancer is dodecyl maltoside. [0238] Embodiment 40. The nasal aerosol formulation as described in Embodiment 39, wherein, the formulation comprises about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside. [0239] Embodiment 41. The nasal aerosol formulation as described in Embodiment 40. wherein, the formulation comprises about 0.1% (w / v) to about 0.5% (w / v) of dodecyl maltose. [0240] Embodiment 42. The nasal aerosol formulation as described in Embodiment 41, wherein the formulation comprises approximately 0.25% (w / v) dodecyl maltose. An alternative Embodiment 42. The nasal aerosol formulation as described in Embodiment 41, wherein, the formulation comprises about 0.25% (w / v) dodecyl maltose and about 0.001 (w / v) to about 1% (p / v) of benzalkonium chloride. An alternative Embodiment 42. The nasal aerosol formulation is as described in Embodiment 41, wherein, the formulation comprises about 0.25% (w / v) dodecyl maltoside and about 0.001 (w / v) to about 1% (p / v) of oleic acid, or salts thereof. An alternative Embodiment 42. The nasal aerosol formulation as described in Embodiment 41, wherein, the formulation comprises about 0.25% (w / v) of dodecyl maltoside, about 0.001 to about 1% (w / v) of benzalkonium chloride and about 0.001 to about 1% (w / v) of oleic acid, or salts thereof. [0241] Embodiment 43a. In certain embodiments, the formulation comprises between about 0.75 mg and about 1.5 mg per dispensed dose from the device of Epinephrine, or a salt thereof, and when administered as a nasal spray, a subject produces one or more of the following pharmacokinetic characteristics : [0242] • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.3 mg yields; [0243] • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.3 mg yields; [0244] • a mean tmax of less than 45 minutes; Y [0245] • an absorption similar to IM injection under optimal dosing conditions in the thigh. [0246] Embodiment 43b. In certain embodiments, the formulation comprises between about 0.5 mg and about 1.15 mg per dispensed dose from the device of Epinephrine, or a salt thereof, and when administered as a nasal spray, a subject produces one or more of the following pharmacokinetic characteristics : [0247] • both a mean AUC 0-20 min and AUC 0 - t is the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.15 mg yield; [0248] • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.15 mg yields; [0249] • a mean tmax of less than 45 minutes [0250] • an absorption similar to IM injection under optimal dosing conditions in the thigh. [0251] Embodiment 43c. In certain embodiments, the formulation comprises between about 1.0 mg and about 2.0 mg per dispensed dose from the device of Epinephrine, or a salt thereof, and when administered as a nasal spray, a subject produces one or more of the following pharmacokinetic characteristics : [0252] • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.5 mg yield; [0253] • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.5 mg yields; [0254] • a mean tmax of less than 45 minutes; Y [0255] • an absorption similar to IM injection under optimal dosing conditions in the thigh. [0256] Also provided are embodiments in which any of Embodiments 43a, 43b, and 43c comprise one or more of the limitations described above in Embodiments. 2-22 and 30-42. [0257] Also provided herein is Embodiment 44, wherein a nasal spray formulation comprising Epinephrine, or a salt thereof, which, when administered to a subject, produces one or more of the following pharmacokinetic characteristics: [0258] • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 min and AUCo-t than intramuscular injection of 0.3 mg yields; [0259] • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.3 mg yields; [0260] • a mean W of less than 45 minutes; Y [0261] • an absorption similar to IM injection under optimal dosing conditions in the thigh. [0262] Also provided herein is Embodiment 45, a nasal spray formulation comprising Epinephrine, or a salt thereof, which, when administered to a subject, produces one or more of the following pharmacokinetic characteristics: [0263] • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.15 mg yield; [0264] • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.15 mg yields; [0265] • a mean W of less than 45 minutes; Y [0266] • an absorption similar to IM injection under optimal dosing conditions in the thigh, [0267] Also provided herein is Embodiment 46, wherein a nasal spray formulation comprising Epinephrine, or a salt thereof, which, when administered to a subject, produces one or more of the following pharmacokinetic characteristics: [0268] • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.5 mg yield; [0269] • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.5 mg yields; [0270] • a mean W of less than 45 minutes; Y [0271] • an absorption similar to IM injection under optimal dosing conditions in the thigh. [0272] Embodiment 47. The nasal aerosol formulation as described in any of Embodiments 45-46, comprising between about 0.4 mg and about 2.40 mg per dispensed dose from the Epinephrine device, or a salt thereof. [0273] Embodiment 48. The nasal aerosol formulation as described in Embodiment 47, wherein the formulation is a pharmaceutical formulation. [0274] Embodiment 49. The nasal aerosol formulation as described in Embodiment 48, at wherein Epinephrine or salts thereof is present in the pharmaceutical formulation in an effective amount for the treatment of an acute hypersensitivity reaction. [0275] Embodiment 50. The nasal aerosol formulation as described in any of Embodiments 44-49, wherein the formulation is aqueous. [0276] Embodiment 51. The nasal aerosol formulation as described in any of Embodiments 44-50. wherein the formulation has intramuscular (IM) absorption similar to injection or subcutaneous (SQ). [0277] Embodiment 52. The nasal aerosol formulation as described in Embodiment 51, wherein the formulation has intramuscular (IM) absorption similar to injection. [0278] Embodiment 53. The nasal aerosol formulation as described in Embodiment 51, wherein the formulation has subcutaneous-type (SQ) absorption. [0279] Embodiment 54. The nasal aerosol formulation as described in Embodiment 1-51 where the SC pharmacokinetic profile has a Cmax of at least 100 pg / mL and AUC0-240min of 150 h * pg / mL. [0280] Embodiment 55. The nasal aerosol formulation as described in any of Embodiments 44-54, wherein the formulation comprises between about 5 mg / mL and about 40 mg / mL of Epinephrine, or a salt thereof. [0281] Embodiment 56. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein the formulation comprises between about 0.9 mg and about 2.4 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0282] Embodiment 57. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein the formulation comprises between about 0.5 mg and about 2.0 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0283] Embodiment 58. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein, the formulation comprises between about 0.75 mg and about 1.5 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0284] Embodiment 59. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein the formulation comprises between about 0.9 mg and about 1.15 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0285] Embodiment 60. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein, the formulation comprises about 1.0 mg per dispensed dose of Epinephrine device, or a salt thereof. [0286] Embodiment 61. The nasal aerosol formulation as described in any of the Embodiments 44-55, wherein the formulation comprises between about 0.45 mg and about 1.15 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0287] Embodiment 62. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein the formulation comprises between about 0.5 mg and about 2.0 mg per dispensed dose from the Epinephrine device, or a salt thereof. [0288] Embodiment 63. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein, the formulation comprises approximately 0.5 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0289] Embodiment 64. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein, the formulation comprises approximately 0.75 mg per dispensed dose of Epinephrine device, or a salt thereof. [0290] Embodiment 65. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein, the formulation comprises about 1.0 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0291] Embodiment 66. The nasal aerosol formulation as described in any of Embodiments 44-55, wherein, the formulation comprises approximately 1.5 mg per dispensed dose from the device of Epinephrine, or a salt thereof. [0292] Embodiment 67. The nasal aerosol formulation as described in any of Embodiments 44-66, wherein the formulation comprises less than one molar equivalency of acid for each mole of Epinephrine. [0293] Embodiment 68. The nasal aerosol formulation as described in any of Embodiments 44-66, wherein the formulation comprises between about 0.5 and about 1.1 molar equivalents of acid in each mole of Epinephrine. [0294] Embodiment 69. The nasal spray formulation is as described in any of Embodiments 66 and 67, wherein the acid is a strong acid. Strong acids include hydrochloric acid, phosphoric acid, and sulfuric acid. [0295] Embodiment 70. The nasal aerosol formulation as described in Embodiment 69, wherein the acid is hydrochloric acid. [0296] Embodiment 71. The nasal aerosol formulation as described in any of Embodiments 44-70. where no base is added to the formulation during its preparation. Embodiment 72. The nasal aerosol formulation as described in any of Embodiments 44-71, wherein the formulation has a pH between about 3.0 and about 6.0. An alternative Embodiment 72. The nasal aerosol formulation as described in any of Embodiments 44-71, wherein the formulation has a pH between about 2.0 and about 6.0. [0297] Embodiment 73. The nasal aerosol formulation as described in Embodiment 72, wherein the formulation has a pH between about 3.5 and about 5.0. [0298] Embodiment 74. The nasal aerosol formulation as described in Embodiment 72, wherein the formulation has a pH between about 4.0 and about 4.5. [0299] Embodiment 75. The nasal aerosol formulation as described in Embodiment 72, wherein the formulation has a pH of about 4.5. [0300] Embodiment 76. The nasal aerosol formulation as described in Embodiment 72, wherein the formulation has a pH of about 4.0. [0301] Embodiment 77. The nasal aerosol formulation as described in any of Embodiments 44-76, wherein, the formulation further comprises a stabilizing agent. [0302] Embodiment 78. The nasal aerosol formulation as described in Embodiment 77, wherein the stabilizing agent is ethylenediaminetetra acetic acid (EDTA) or a salt thereof. [0303] Embodiment 79. The nasal aerosol formulation as described in Embodiment 78, wherein the EDTA is disodium EDTA. [0304] Embodiment 80. The nasal aerosol formulation as described in Embodiment 78, wherein EDTA is present in an amount that is 5% to 15% of the amount of Epinephrine, both measured in mmol. An alternative Embodiment 80. The nasal aerosol formulation as described in Embodiment 78, wherein EDTA is present in an amount that is from about 0.001% (w / v) to about 1% (w / v). [0305] Embodiment 81. The nasal aerosol formulation as described in Embodiment 79, wherein the mmol of EDTA is approximately 10% of the mmol of Epinephrine. [0306] Embodiment 82. The nasal aerosol formulation as described in any of Embodiments 44-81, wherein the formulation additionally comprises a preservative. Embodiment 83. The nasal aerosol formulation as described in Embodiment 82, wherein the preservative is benzalkonium chloride. [0307] Embodiment 84. The nasal aerosol formulation as described in any of Embodiments 44-83, wherein the formulation further comprises an absorption enhancer. In an alternative Embodiment 84, the nasal aerosol formulation as described in any of Embodiments 44-83, wherein the formulation additionally comprises one or more absorption enhancers. [0308] Embodiment 85. The nasal aerosol formulation as described in Embodiment 84, wherein the absorption enhancer is an alkylsaccharide. In an alternative Embodiment 85, the nasal aerosol formulation as described in Embodiment 84, wherein the absorption enhancer is an alkylsaccharide and / or benzalkonium chloride. [0309] Embodiment 86. The nasal aerosol formulation as described in Embodiment 85, wherein the absorption enhancer is dodecyl maltoside. In an alternative Embodiment 86, the nasal aerosol formulation as described in Embodiment 85, wherein the absorption enhancer is dodecyl maltoside, benzalkonium chloride, or a combination of dodecyl maltoside and benzalkonium chloride. [0310] Embodiment 87. The nasal aerosol formulation as described in Embodiment 86, wherein, the formulation comprises about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside. [0311] Embodiment 88. The nasal aerosol formulation as described in Embodiment 87, wherein, the formulation comprises about 0.1% (w / v) to about 0.5% (w / v) of dodecyl maltoside. [0312] Embodiment 89. The nasal aerosol formulation as described in Embodiment 88, wherein, the formulation comprises about 0.25% (w / v) dodecyl maltose. Also provided in Embodiment 90. is a method of treating an adrenergic receptor-mediated condition comprising intranasal administration of the formulation as mentioned in any of Embodiments 1-89 above. [0313] Embodiment 91. The method as described in Embodiment 90. wherein the condition is selected from a type 1 hypersensitivity reaction (systemic allergic reaction), an acute asthmatic attack, cardiac arrest, and Stokes-Adams syndrome. [0314] Embodiment 92. The method as described in Embodiment 91, wherein the condition is a type 1 hypersensitivity reaction (systemic allergic reaction). [0315] Embodiment 93. The method as described in Embodiment 92, wherein the type 1 hypersensitivity reaction (systemic allergic reaction) is selected from allergic asthma, allergic conjunctivitis, allergic rhinitis, anaphylaxis, angioedema, urticaria, eosinophilia, drug allergy, and Food Allergy. [0316] Embodiment 94. The method as described in Embodiment 93, wherein the drug allergy is an antibiotic allergy. [0317] Embodiment 95. Also provided herein is the method of treating a systemic allergic reaction and anaphylaxis which comprises intranasal administration of an unbuffered intranasal formulation of Epinephrine in an amount less than about 2.0 mg. Also provided are embodiments wherein Embodiment 95 comprises one or more of the limitations described above in Embodiments 2-22, 25-28, 30-55, 58-61, and 63-89. [0318] Embodiment 96. A pharmaceutical composition comprising: a) Epinephrine; and b) an alkylglycoside; wherein the composition is formulated for administration into the circulatory system of a subject via the intranasal, inhalation, or pulmonary route of administration. An Alternative Embodiment 96. A pharmaceutical composition comprising: a) Epinephrine; and b) an alkylglycoside; wherein the composition is a liquid formulated for intranasal administration. [0319] Embodiment 97. The pharmaceutical composition of Embodiment 96, wherein the alkylglycoside has an alkyl chain that includes between 8 to 20 carbons. [0320] Embodiment 98. The pharmaceutical composition of Embodiment 97, wherein the alkylglycoside is selected from the group consisting of undecyl maltoside, dodecyl maltoside, tridecyl maltoside, tetradecyl maltoside, sucrose monododecanoate, sucrose mono-tridecanoate, sucrose mono-tetradecanoate. [0321] Embodiment 99. The pharmaceutical composition of Embodiment 98, wherein the alkylglycoside is dodecyl-beta-D-maltoside. [0322] Embodiment 100. The pharmaceutical composition of Embodiment 96, wherein the alkylglycoside concentration is between about 0.001% and 10.0% (w / v). [0323] Embodiment 101. The pharmaceutical composition of Embodiment 100. wherein the alkylglycoside concentration is between about 0.05% and 0.5% (w / v). [0324] Embodiment 102. The pharmaceutical composition of Embodiment 96, wherein the composition further comprises a membrane penetration enhancing agent. The pharmaceutical composition of Embodiment 96, wherein the composition further comprises a membrane penetration enhancing agent, pH modifier, buffering agents, isotonicity agent, antioxidant, chelator, preservative, or a combination thereof. [0325] Embodiment 103. The pharmaceutical composition of Embodiment 102, wherein the membrane penetration enhancing agent is a surfactant, a bile salt, a phospholipid, an alcohol, an enamine, a medium and / or long chain amphipathic molecule, a small hydrophobic molecule, a sodium or salicylic acid derivative or a glycerol ester of acetoacetic acid, a cyclodextrin, a medium chain or long chain fatty acid, a chelating agent, an amino acid or salts thereof, an enzyme or a combination of them. [0326] Embodiment 104. The pharmaceutical composition of Embodiment 102, wherein the membrane penetration enhancing agent is selected from the group consisting of citric acid, sodium citrate, propylene glycol, glycerin, ascorbic acid, sodium metabisulfite, ethylenediaminetetra acetic acid ( EDTA) disodium, benzalkonium chloride, sodium hydroxide, and combinations thereof. [0327] Embodiment 105. The pharmaceutical composition of Embodiment 102, wherein the membrane penetration enhancing agent is benzalkonium chloride, EDTA, or a combination thereof. [0328] Embodiment 106. The pharmaceutical composition of Embodiment 96, wherein the composition provides a Cmax for Epinephrine in a subject that is approximately 2-fold or greater compared to an administration without alkylglycoside. [0329] Embodiment 107. The pharmaceutical composition of Embodiment 96, wherein the composition provides a W for Epinephrine in a subject that is about 2 times or less compared to an administration without alkyl glucoside. [0330] Embodiment 108. The pharmaceutical composition of Embodiment 96, wherein the composition provides a W for Epinephrine of about 0.3 hours or less in a subject. [0331] Embodiment 109. The pharmaceutical composition of Embodiment 96, wherein the composition has a pH of about 2.0 to 5.0. [0332] Embodiment 110. A method of increasing the bioavailability of Epinephrine in a subject comprising administering to a subject a composition comprising Epinephrine and an alkylglycoside, thereby increasing the bioavailability of Epinephrine in the subject, wherein the composition is administered into the circulatory system of the subject by intranasal, inhalation, or pulmonary route of administration. An Alternative Embodiment 110. A method of increasing the bioavailability of Epinephrine in a subject comprising administering to a subject a composition comprising Epinephrine and an alkylglycoside, thus increasing the bioavailability of Epinephrine in the subject, wherein the composition is an administered liquid composition intranasally. [0333] Embodiment 111. The method of Embodiment 110. wherein the alkylglycoside has an alkyl chain that includes between 8 to 20 carbons. [0334] Embodiment 112. The method of Embodiment 111, wherein the alkylglycoside is selected from the group consisting of undecyl maltoside, dodecyl maltoside, tridecyl maltoside, tetradecyl maltoside, sucrose mono-dodecanoate, sucrose mono-tridecanoate, sucrose monotetradecanoate. [0335] Embodiment 113. The method of Embodiment 112, wherein the alkylglycoside is dodecylbeta-D-maltoside. [0336] Embodiment 114. The method of Embodiment 110. wherein the alkylglycoside concentration is between about 0.001% and 10.0% (w / v). [0337] Embodiment 115. The method of Embodiment 114, wherein the alkylglycoside concentration is between about 0.05% and 0.5% (w / v). [0338] Embodiment 116. The method of Embodiment 110. wherein the composition further comprises an agent that enhances membrane penetration. [0339] Embodiment 117. The method of Embodiment 116, wherein the membrane penetration enhancing agent is a surfactant, a bile salt, a phospholipid, an alcohol, an enamine, a long-chain amphipathic molecule, a hydrophobic small molecule, a sodium or salicylic acid derivative, a glycerol ester of acetoacetic acid, a cyclodextrin, a medium chain fatty acid, a chelating agent, an amino acid or salts thereof, an enzyme or a combination thereof. [0340] Embodiment 118. The method of Embodiment 117, wherein the membrane penetration enhancing agent is selected from the group consisting of citric acid, sodium citrate, propylene glycol, glycerin, ascorbic acid, sodium metabisulfite, ethylenediaminetetra acid disodium acetic acid (EDTA), benzalkonium chloride, sodium hydroxide, and combinations thereof. [0341] Embodiment 119. The method of Embodiment 116, wherein the membrane penetration enhancing agent is benzalkonium chloride, EDTA, or a combination thereof. [0342] Embodiment 120, The method of Embodiment 110. wherein the composition provides a Cmax for Epinephrine in the subject that is approximately 2-fold or greater compared to administration without alkyl glucoside. [0343] Embodiment 121. The method of Embodiment 110. wherein the composition provides a tmax for Epinephrine in the subject that is approximately 2-fold or less compared to an administration without alkyl glucoside. [0344] Embodiment 122. The method of Embodiment 110. wherein the composition provides a tmax for Epinephrine of about 0.3 hours or less in the subject. [0345] Embodiment 123. The method of Embodiment 110. wherein the composition has a pH of about 2.0 to 6.0. An alternative Embodiment 123. The method of Embodiment 110. wherein the composition has a pH of about 2.0 to 5.0. [0346] Definitions [0347] As used herein, the following terms have the indicated meanings. [0348] When ranges of values are mentioned, Using the notation "from ni ... an 2 " or "between ni ... and n 2 " is used, when ni and n 2 are the numbers, then, unless otherwise specified, this notation is intended to include the numbers themselves and the range of numbers between them. This range can be integral or continuous between and, including final values. By way of example, the range "2 to 6 carbons" is intended to include two, three, four, five, and six carbons, since carbons come in whole units. Compare, as an example, the range "1 to 3 ^ M (micromolar)," which is intended to include 1 ^ M, 3 ^ M, and all between any number of significant figures (for example, 1.255 ^ M, 2.1 ^ M, 2.9999 ^ M, etc.). [0349] The term "approximately", as used herein, is intended to qualify the numerical values that it modifies, denoting said value as a variable within a range. When no range is recited, such as a margin of error or standard deviation, a given mean value in a table or Data Table, the term "approximately" should be understood as a mean of the largest range that would encompass the recited value and the range that would be included when rounding up or down in that figure as well, considering significant figures, and the range that the recited value would cover plus or minus 20%. [0350] "Weight by volume" or "w / v" refers to the mass in grams of a dissolved solute divided by the volume in milliliters of the entire solution. Typically, weight by volume is expressed as a percentage. [0351] The term "absorption enhancer", as used herein, refers to a functional excipient included in formulations to enhance the absorption of an active agent such as a pharmacologically active drug. This term generally refers to an agent whose function is to increase absorption by improving penetration of the nasal mucous membrane, rather than by increasing solubility. As such, such agents are sometimes referred to as permeation enhancers or penetration enhancers. In particular, the absorption enhancers described here can enhance paracellular transport (i.e., passage through intercellular spaces and tight junctions), transcellular transport (i.e., passive diffusion or active transport across cell membranes. ), or transcytosis (i.e., vesicular cell uptake). Ozsoy et al., Molecules 14: 3754-79, 2009. [0352] Examples of absorption enhancers include alcohol, aprotinin, benzalkonium chloride, benzyl alcohol, capric acid, ceramides, cetylpyridinium chloride, chitosan, cyclodextrins, deoxycholic acid, decanoyl, dimethyl sulfoxide, glyceryl monooleate, glycofurol, glycolofuroflucif, Glycerophosciflycol, glycerophosphatidylcholine, 2-Hydroxypropyl-pcyclodextrin, laureth-9, lauric acid, lauroyl carnitine, sodium lauryl sulfate, lysophosphatidylcholine, menthol, poloxamer 407 or F68, poly-L-arginine, polyoxyethylene-myrne 9-isopropyl ether , isopropyl palmitate, lanolin, light mineral oil, oleic lin acid, menthol, myristic acid, myristyl alcohol, oleic acid, or sales thereof, oleyl alcohol, palmitic acid, polysorbate 80, propylene glycol, polyoxyethylene alkyl ethers, polyoxylglycerides, pyrrolidone, quillaia saponin, salicylic acid, sodium salt, p-sitosterol pD-glucoside, sucrose cocoate, taurocholic acid, tauroid acid soxycholic, taurodihydrofussidic acid, thymol, tricapryline, triolein, and alkylsaccharides, and combinations thereof, including, but not limited to, dodecyl maltoside, dodecyl-pD-maltoside, tetradecyl maltoside, tetradecyl-pD-maltoside, and sucrose dodecanoate. Alkylsaccharides (for example, nonionic alkylsaccharide surfactants such as alkylglycosides and sucrose esters of fatty acids consisting of an aliphatic hydrocarbon chain coupled to a sugar moiety by a glucosidic ester bond, respectively), cyclodextrins (cyclic oligosaccharides composed of six or more monosaccharide units with a central cavity, which form inclusion complexes with hydrophobic molecules and have been used mainly to increase drug solubility and dissolution and improve absorption of low molecular weight drug), Chitosans (cationic polysaccharides produced from deacetylation of chitin), bile salts and their derivatives (such as sodium glycocholate, sodium taurocholate, and sodium taurodihydrofusidate) tend to be among the best tolerated absorption enhancers. See, for example, Aungst BJ, AAPS Journal 14 (1): 10-8, 2011; and Maggio, ET, Excipients and Food Chem. 5 (2): 100-12, 2014. Due to their chemical properties, certain absorption enhancers can function as preservatives and / or surfactants cationic in certain circumstances, depending on the concentration in the formulation and other factors. [0353] In this document compositions comprising Epinephrine and at least one absorption enhancer and / or preservative and / or surfactant are described wherein the at least one absorption enhancer and / or preservative and / or surfactant comprises at least one alkylglycoside and / or at least one alkyl ester saccharide. [0354] As used herein, the term "alkylsaccharide" (also referred to herein as "alkylglycoside") refers to a type of absorption enhancer. As used herein, an alkylsaccharide refers to any sugar linked by a bond to any hydrophobic alkyl, as is known in the art. Alkylsaccharides include, but are not limited to: alkylsaccharides, such as octyl-, nonyl-, decyl-, undecyl-, dodecyl-, tridecyl-, tetradecyl-, pentadecyl-, hexadecyl-, heptadecyl-, and octadecyl- □ - or Alkali DD-maltoside, -glucoside, or -sucrosidothiomaltosides, such as heptyl, octyl, dodecyl-, tridecyl-, and tetradecyl-DD-thiomaltoside; alkali thioglycosides, such as heptyl or octyl 1-thio □ - or dD-glucopyranoside; alkaline thiosucroses; alkaline maltotriosides; long-chain aliphatic carbonic acid amides of sucrose D-amino-alkyl ethers; palatinose and isomaltamine derivatives amide linked to an alkaline chain; isomaltamine derivatives linked by urea to an alkaline chain; long chain aliphatic carbonic acid ureides of sucrose D-amino-alkyl ethers; and sucrose amides of long chain aliphatic carbonic acid D-amino-alkyl ethers. The hydrophobic alkyl can be chosen in any desired size, depending on the desired hydrophobicity and the hydrophilicity of the saccharide moiety. For example, a preferred range of alkyl chains is from about 9 to about 24 carbon atoms. An even more preferred range is from about 9 to about 16 or about 14 carbon atoms. Similarly, some preferred saccharides include maltose, sucrose, and glucose linked by glucosidic bonds and an alkyl chain of 9, 10. 12, 13, 14, 16, 18, 20, 22, or 24 carbon atoms, for example, nonyl -, decyl-, dodecyl- and tetradecyl sucroside, glucoside, and maltoside, etc. The alkyl chain of an alkylsaccharide is often linked to the saccharide through a glycosidic bond, and accordingly, alkylsaccharides are often referred to interchangeably as alkylglucosides. [0355] Any "suitable" alkylglucoside means one that meets the characteristics contemplated herein, that is, that the alkylglucoside is non-toxic and nonionic, and that increases the absorption of a compound (eg, Epinephrine) when administered with the compound via the nasal delivery route. [0356] As used herein, a "saccharide" includes monosaccharides, oligosaccharides or polysaccharides in the form of a ring or straight chain, or a combination thereof to form a saccharide chain. Oligosaccharides are saccharides that have two or more monosaccharide residues. The saccharide can be chosen, for example, from any species of saccharide currently commercially available or can be synthesized. Some examples of the many possible saccharides that are used include glucose, maltose, maltotriose, maltotetraose, sucrose, and trehalose. Preferable saccharides include maltose, sucrose, and glucose. [0357] In some Embodiments, compositions including at least one alkylglucoside and / or saccharide alkyl ester and Epinephrine, methods for administering and using the compositions via the nasal route of administration, and methods for ameliorating a disease state in a subject are described herein. By administering such compositions. [0358] In some Embodiments, a method of administering a composition having at least one alkylglucoside and / or alkyl saccharide ester mixed, mixed, mixed with Epinephrine and administered or administered to a subject is described herein, wherein the alkyl has of about 10 to 24, 10 to 20, 10 to 16, or 10 to 14 carbon atoms, wherein the at least one alkylglucoside and / or alkyl ester saccharide increases the stability and bioavailability of the therapeutic agent. [0359] In some embodiments, contemplated alkylsaccharides have a hydrophobic alkyl group attached to a hydrophilic saccharide. The bond between the hydrophobic alkyl group and the hydrophilic saccharide can include, among other possibilities, a glycosidic, thioglucosidic (Horton) amide, amide (Carbohydrates as Organic Raw Materials, FW Lichtenthaler ed., VCH Publishers, New York, 1991), ureide (Austrian Pat. 386,414 (1988); Chem. Abstr. [0360] 110: 137536p (1989); see Gruber, H. and Greber, G., "Reactive Sucrose Derivatives" in Carbohydrates as Organic Raw Materials, pp. 95-116) or ester bond (Sugar Esters: Preparation and Application, J. C. Colbert ed., (Noyes Data Corp., New Jersey), (1974)). In addition, preferred glycosides may include maltose, sucrose, glucose linked by glucosidic bond, and an alkyl chain of about 9-16 carbon atoms, for example, nonyl-, decyl-, dodecyl- and tetradecyl sucroside, glucoside, and malthoes. These compositions are amphipathic and non-toxic, because they degrade an alcohol and an oligosaccharide. [0361] The above examples are illustrative of the types of glycosides contemplated, but the list is not exhaustive. Derivatives of the above compounds that meet the criteria described herein are also contemplated when choosing an alkylsaccharide. [0362] In some embodiments, contemplated membrane penetration enhancing agents serve as antibacterial agents. An agent is an "antibacterial" agent or substance if the agent or its equivalent kills bacteria, or suppresses bacterial growth or reproduction. [0363] The term "active ingredient" or "pharmaceutically active compound" is defined in the context of a "formulation" and is intended to mean a component of a pharmaceutical formulation that provides the primary pharmacological effect, as opposed to an "inactive ingredient" which would generally be recognized as providing no pharmaceutical benefit. [0364] The term "actuation", as used herein, refers to an operation of the device such that the pharmaceutical formulation is administered therefrom. The term "antimicrobial preservative", as used herein, refers to a pharmaceutically acceptable excipient with antimicrobial properties to which a pharmaceutical formulation is added to maintain microbiological stability. Antimicrobial preservatives include, but are not limited to, antibacterial agents, antifungal agents, antioxidants, and preservatives. [0365] The term "AUC", as used herein, refers to the area under the plasma drug concentration-time curve. The term "AUC 0 -t," as used herein, refers to the area under the plasma drug concentration-time curve from t = 0 to the last measured or measurable concentration. The term "AUC 0 -", "or equivalently," AUC 0 -inf, "as used herein, refers to the area under the plasma drug concentration-time curve extrapolated by infinity (~). [0366] As used herein, the term "benzalkonium chloride" ("BZK") refers to a member of the class of quaternary ammonium compounds that have the following structure: [0370] where n is an integer. Benzalkonium chloride is a mixture of alkylbenzyl dimethylammonium chlorides, where a mixture of more than one n is used. In certain Embodiments, n is 8, 10. 12, 14, 16, or 18. In other Embodiments, n is 10. 12, or 14. In some Embodiments, a mixture of n is 10. 12 and / or 14 predominates. In some embodiments, a mixture of n 10. 12, 14 and / or 16 predominates. In some embodiments, benzalkonium chloride functions as a preservative (even in small amounts), an antiseptic, a disinfectant, a wetting and solubilizing agent, and / or a cationic surfactant. In some cases, benzalkonium chloride refers to a type of absorption enhancer. [0371] The term "bioavailability (F)", as used herein, refers to a fraction of a drug dose that is absorbed from its site of administration and reaches, in an unaltered form, the systemic circulation. The term "absolute bioavailability" is used when the fraction of the drug absorbed is related to its bioavailability IV. It can be calculated using the following formula: [0375] The term "relative bioavailability (Frel)" is used to compare two different extravascular routes of drug administration and can be calculated using the following formula: [0378] The term "clearance (CL)", as used herein, refers to the rate at which a drug is eliminated divided by its plasma concentration, giving a plasma volume of which the drug is completely eliminated per unit of weather. CL is equal to the constant elimination rate (A) multiplied by the volume of distribution (Vd), where "Vd" is the volume of fluid that would be needed to contain the amount of drug present in the body at the same concentration as in plasma. The term "apparent clearance (CL / F)," as used herein, refers to an clearance that does not take into account the bioavailability of the drug. It is the ratio of the dose over the AUC. [0379] The term "Cmax," as used herein, refers to the maximum observed plasma concentration. [0380] The term "coefficient of variation (CV)", as used herein, refers to the ratio of the sample standard deviation to the sample mean. It is often expressed as a percentage. [0381] The term "confidence interval", as used herein, refers to a range of values that will include the true average value of a parameter a specific percentage of the time. [0382] The term "device", as used herein, refers to an apparatus capable of delivering a drug to a patient in need of it. [0383] The term "delivery time", as used herein, refers to the amount of time that elapses between a determination made by a healthcare professional, or an untrained individual that an individual needs a nasal delivery of Epinephrine and complete delivery. [0384] The term "disease", as used in this document, is intended to be generally synonymous, and is used interchangeably with the terms "disorder", "syndrome", and "condition" (as in a medical condition), in that sense they reflect a abnormal condition of the human or animal body of one of its parts that impairs normal functioning, is typically manifested by distinctive signs and symptoms, and causes the human or animal to have a reduced duration or quality of life. [0385] As used herein, the "dose dispensed from the device" is typically measured in the nasal spray configuration by the difference in weight of a device before and after activation and release of a dose of the formulation contained in the device. same. The volume of the liquid formulation and the weight in milligrams of the active residue contained therein can be determined by standard calculations. [0386] The term "elimination rate constant (A)," as used herein, refers to the fractional rate of elimination of drugs from the body. This rate is constant in first-order kinetics and is independent of the concentration of the drug in the body. A is the slope of the plasma concentration-time line (on a logarithmic scale y). The term “Az,” as used herein, refers to the terminal phase elimination rate constant, where the "terminal phase" of the drug plasma concentration time curve is a straight line when plotted on a graph semi-logarithmic. The terminal phase is often called the "elimination phase" because the main mechanism for decreasing drug concentration during the terminal phase is the elimination of the drug from the body. The hallmark of the terminal elimination phase is that the relative proportion of drug in plasma and peripheral volumes of distribution remains constant. During this "terminal phase", the drug returns from the fast and slow volumes of distribution in plasma, and is permanently eliminated from plasma by renal metabolism or excretion. [0387] The term "equal", as used herein, means essentially the same as (ie, insignificantly different from) in amount, amount, value, grade, or size. The term "equal" may, in certain embodiments, include "bioequivalence", but the terms are not coincident. [0388] The term "bioequivalence", as used herein, describes the relationship between a reference and a presumed equivalence or alternative medicine, per 21 C.F.R. § 320.1, means that there is no significant difference in the rate and degree to which the active ingredient or active fraction in pharmaceutical equivalents or pharmaceutical alternatives are available at the site of action of the drug when administered at the same molar dose in Similar conditions under appropriate conditions designed study The rate and degree of absorption can be determined from, or reported by, Cmax and AUC, respectively. In certain embodiments, statistical criteria can be used, for example, between 80% and 125% of a reference value, or 90% CI. [0389] The term "molar equivalence", as used herein, refers to an amount of Epinephrine that is equimolar to a specific amount of acid. [0390] The term "excipient", as used herein, refers to a natural or synthetic substance formulated together with the active ingredient in a drug. An excipient is included in a formulation for a variety of reasons such as, but not limited to, long-term stabilization, solid bulking formulations, or to confer a therapeutic enhancement to the active ingredient in the final dosage form, such as facilitating the absorption of the drug, reducing viscosity, or improving solubility. [0391] The term "filled", as used herein, refers to an association between a device and a pharmaceutical formulation, for example, when a pharmaceutical formulation described herein comprising a therapeutically effective amount of Epinephrine is present within of a reservoir that is part of a device described here. The term "formulation", with or without the modifier "pharmaceutical", as used herein, refers to a composition comprising at least one ingredient. physiologically active (eg, a drug); including, but not limited to, salts, solvates and hydrates of Epinephrine and related compounds disclosed herein, whereby the formulation is usable for a specific and effective result in a mammal (eg, without limitation, a human). [0392] The term "pharmaceutical formulation", as used herein, alone or in combination, refers to a formulation that is suitable for use for the treatment (or in certain embodiments, prevention) of a disease in a subject. [0393] The term "hydrate", as used herein, refers to an Epinephrine described herein or a salt thereof that further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces. [0394] The term "in need of treatment" and the term "in need" when referring to treatment are used interchangeably and refers to a judgment made by a caregiver (eg, doctor, nurse, nurse practitioner, that a patient will benefit from the treatment. [0395] As used herein, an "intramuscular (IM) injection" of Epinephrine is typically administered via an IM epinephrine delivered by autoinjector into the thigh, eg, into the lateral vestibular muscle (referred to herein as "conditions optimal dosing rate on the thigh "). As such, when comparing the pharmacokinetic parameters produced by IM epinephrine injection with those obtained by IN epinephrine administration, the comparison should be assumed as if the IM injection was in the thigh, which is the optimal dosing method for Epinephrine. In one Embodiment, IM Epinephrine injection is accomplished with the EpiPen® Autoinjector (0.3mg / 0.3ml Epinephrine Injection, USP, Preloaded Autoinjector; Milan Specialty L.P.). [0396] As used herein, a "subcutaneous injection (SQ)" of Epinephrine is typically administered by injection into the subcutaneous layer of the deltoid region in the upper arm. Simons et al. Epinephrine absorption in adults: Intramuscular versus subcutaneous injection. J Allergy. Clin. Immunol. 2001; 108: 871-3. [0397] As used herein, two embodiments are "mutually exclusive" when one is defined as something different from the other. For example, an Embodiment in which the concentration of Epinephrine is specified as 5 mg / ml is mutually exclusive with an Embodiment in which the amount of Epinephrine is specified as 10 mg / ml. However, an Embodiment in which the amount of Epinephrine is specified as 5 mg / ml is not mutually exclusive with an Embodiment in which less than 10% of the pharmaceutical formulation exits the nasal cavity through the drainage into the nasopharynx externally. . [0398] The term "pharmaceutically acceptable", as used herein, refers to a component of a formulation, often referred to as a carrier or excipient, that is compatible with the other ingredients of the formulation and not too harmful to the recipient of the same. [0399] The term "pre-priming", as used herein, refers to a device, such as a nasal spray that can deliver a formulation to a patient in need of it with the first actuation of the aerosol pump, that is, without the need to prime the pump prior to dosing, such as operating the pump one or more times until sprinkling occurs. [0400] The term "prone", as used herein, refers to a patient who is lying face down. [0401] As used herein, the term "protective packaging" refers to a wrapper. The term "solvate," as used herein, refers to an Epinephrine described herein or a salt thereof, which further includes a stoichiometric or non-stoichiometric amount of a solvent bound by non-covalent intermolecular forces. Preferred solvents are volatile, non-toxic, and / or acceptable for administration to humans in trace amounts. [0402] The term "storage stable", as used herein, refers to a formulation in which at least about 90%, 115% of the active ingredient remains within acceptable regulatory specifications after storage of the formulation at specified temperature and humidity for a period of time, for example, for at least 12 months at 25 ° C and 60% relative humidity and approximately six months at approximately 40 ° C and approximately 75% relative humidity. [0403] The term "subject", as used herein, is intended to be synonymous with "patient", and refers to any mammal (preferably human) affected by a condition that is likely to be a benefit of treatment with a therapeutically effective amount of Epinephrine, for example, a subject experiencing a type 1 hypersensitivity reaction (systemic allergic reaction) such as anaphylaxis. [0404] The term "supine", as used herein, refers to a patient who is lying on his back. [0405] The term "nostril", as used herein, is synonymous with "naris." The term "therapeutically effective amount" or "therapeutically effective dose", as used herein, refers to the amount or dose of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system or individual that is being sought by a researcher, healthcare provider, or individual.A therapeutically effective amount may, but not necessarily, eliminate one, more, or all symptoms of a disease, disorder, or condition to be treated.A therapeutically effective amount may also prevent the progression of the disease or the appearance of additional symptoms. [0406] The term "W or" half-life, "as used herein, refers to the amount of time required for half a drug or other analyte of interest (eg, an adrenergic receptor agonist) that can be remove from the body or the time required for the concentration of a drug to drop by half. [0407] The term "tonicity agent", as used herein, refers to a compound that modifies the osmolality of a formulation, for example, makes it isotonic. Tonicity agents include dextrose, lactose, sodium chloride, calcium chloride, magnesium chloride, sorbitol, sucrose, mannitol, trehalose, raffinose, polyethylene glycol, hydroxyethyl starch, glycine, and the like. In some embodiments, the formulations contemplated herein include one or more tonicity agents selected from dextrose, glycerin, mannitol, potassium chloride, and sodium chloride. In some embodiments, the formulations contemplated herein include sodium chloride as a tonicity agent. [0408] The term "tomography", as used herein, refers to a sectional imaging process. Images can be viewed individually, as a series of two-dimensional slices, or together as a computer-generated three-dimensional representation. [0409] The term "Tmax," as used herein, refers to the time, from administration, for a drug or other analyte to reach the maximum plasma drug concentration (C max). [0410] Epinephrine [0411] The term "Epinephrine" as used herein refers to the compound (R) -4- (1-Hydroxy-2- (methylamino) ethyl) benzene-1,2-diol, also known as adrenaline, shows a cotinuation and that has the following structure, elemental composition, molecular weight and CAS Registry Number: [0415] CAS Registry Number: 51-43-4 [0416] The term includes any metabolite, salt, ester, hydrate, anhydride, solvate, isomer, isotope, enantiomer, free acid form, free base form, crystalline form, cocrystalline form, complexes, amorphous form, prodrug (including pro-drug ester ), racemate, polymorph, chelate, isomer, tautomer, or optically active form thereof, or a mixture of two or more of the foregoing. [0417] Pharmacological products adapted for nasal administration of Epinephrine, including formulations and devices. Epinephrine works by binding a variety of adrenergic receptors. Epinephrine is a non-selective agonist of all adrenergic receptors, including the major subtypes a1, a2, p1, p2, and p3. Its actions vary according to the type of tissue and the tissue expression of adrenergic receptors. For example, high levels of Epinephrine cause relaxation of the smooth muscle in the airways, but it causes contraction of the smooth muscle that lines most arterioles. [0418] Formulations are provided, devices adapted for the nasal administration of a formulation to a patient, kits comprising the above methods, and of using the same in treatment, each of which comprises a therapeutically effective amount of Epinephrine. [0419] Epinephrine may be present in the formulations administered herein at concentrations between 1 mg / mL and 40 mg / mL, for example, at concentrations of about 5 mg / mL, about 10 mg / mL, or about 20 mg / mL. Epinephrine can be present in formulations administered herein at doses between 0.1 mg and 4 mg, for example, at doses of about 0.5 mg, about 1.0 mg, or about 2.0 mg. These doses can be scaled according to the molecular weight of a counterion if a salt is used to prepare the formulation. [0420] Epinephrine can optionally exist as a pharmaceutically acceptable salt including pharmaceutically acceptable acid addition salts prepared from non-toxic pharmaceutically acceptable acids including inorganic and organic acids. Representative acids include, but are not limited to, acetic, benzenesulfonic, benzoic, camphor sulfonic, citric, ethenesulfonic, dichloroacetic, formic, fumaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic. , mucic, nitric, oxalic, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, oxalic, p-toluenesulfonic, and the like, as the pharmaceutically acceptable salts listed by Berge et al., Journal of Pharmaceutical Sciences, 66: 1-19 ( 1977). Acid addition salts can be obtained as direct synthesis products of compounds. Alternatively, the free base can be dissolved in a suitable solvent containing the appropriate acid and isolated salt by evaporating the solvent or otherwise separating the salt and solvent. Due to the perceived insolubility of the Epinephrine base, finished dosage forms of Epinephrine used in healthcare (solutions, sprays, etc.) are typically salts, eg, hydrochloride, bitartrate, or borate salts. In certain embodiments, the formulations contemplated herein include a salt form of Epinephrine which is Epinephrine acetate, Epinephrine hydrochloride, Epinephrine tartrate, Epinephrine bitartrate, Epinephrine hydrogen tartrate, or Epinephrine borate. Accordingly, provided herein are pharmaceutical formulations for intranasal administration comprising Epinephrine. In certain embodiments, the formulation it is an aqueous solution. In certain embodiments, the formulation comprises, per dose, between about 25 and about 250 pL of the aqueous solution. In certain embodiments, the formulation comprises, per dose, between about 50 and about 250 pL of the aqueous solution. In certain embodiments, the formulation comprises, per dose, between about 50 and about 200 pL of the aqueous solution. In certain embodiments, the formulation comprises, per dose, no more than about 140 pL. In certain embodiments, the formulation comprises, per dose, no more than about 100 pL. In certain embodiments, the formulation comprises, per dose, about 100 pL. The formulation may comprise, per dose, about 25 pL, about 50 pL, about 75 pL, about 100 pL, about 125 pL, about 150 pL, about 175 pL, about 200 pL, or about 250 pL of the aqueous solution. [0421] Pharmaceutical formulations for intranasal administration comprising Epinephrine described herein avoid potential metabolic conversion in the gastrointestinal tract and first-pass hepatic metabolism, and reach the systemic circulation in a pharmacologically active form. Epinephrine is extensively metabolized after oral administration by catechol-O-methyltransferase in the gastrointestinal tract and by monoamine oxidase in the gastrointestinal tract and liver. Avoiding first-pass authorization ensures that more of the epinephrine being administered will be available in an anaphylaxis to treat. By avoiding first-pass liver clearance, the bioavailability of Epinephrine increases. [0422] Formulations [0423] [00302] Pharmaceutical formulations comprising Epinephrine are also provided. Certain embodiments of the present disclosure include a method of producing a formulation comprising mixing Epinephrine and a pharmaceutically acceptable carrier. The pharmaceutical formulations are applied directly to the nasal cavity using the devices described in this document. In the case of a spray, this can be achieved, for example, by a metering atomizing spray pump, for example a single, two-dose, multi-purpose spray device, with or without propellant. [0424] Liquid preparations include solutions, suspensions, and emulsions, eg, water, or water-ethanol, or water-propylene glycol solutions. Typically the formulation is an aqueous liquid solution. Additional ingredients in liquid preparations may include preservatives, stabilizing agents, tonicity agents, absorption enhancers, pH adjusting agents, antioxidants, buffers, sweeteners / flavoring agents / task masking agents, and optionally other ingredients. Ingredients in liquid preparations can serve different functions. The function (s) of a particular ingredient will depend on a number of factors including, but not limited to, the presence or absence of other ingredients, concentration (s) and other factors. [0425] Preservatives include: benzalkonium chloride, methyl paraben, sodium benzoate, benzoic acid, phenyl alcohol, and the like, and mixtures thereof. Due to their chemical properties, certain preservatives can function as surfactants and / or absorption enhancers in certain circumstances, depending on the concentration in the formulation and other factors. [0426] Other preservatives include: alcohol, benzalkonium chloride, benzethonium chloride, benzoic acid, benzyl alcohol, boric acid, bronopol, butylated hydroxyanisole (BHA), butylene glycol, butyl paraben, calcium acetate, calcium chloride, calcium lactate, carbon dioxide , bentonite, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, citric acid monohydrate, cresol, dimethyl ether, ethylparaben, glycerin, hexetidine, imidurea, magnesium trisilicate, isopropyl alcohol, methylpalactic acid, monothoserol, parabenzene, methylpalactic acid (methyl, ethyl and propyl), penthetic acid, phenol, phenylethyl alcohol, phenylmercuric acetate, phenylmercuric borate, phenylmercuric nitrate, potassium benzoate, potassium metabisulfite, potassium sorbate, propionic acid, propyl gallate, propylene glycol, propyl paraben, propyl paraben, sodium , sodium acetate, sodium benzoate, sodium borate, sodium lactate, sodium metabisulfite, sodium propionate, sulfite sodium, sorbic acid, sulfobutyletherbcyclodextrin, sulfur dioxide, edetic acid, thimerosal, and xylitol. [0427] In some embodiments, preservatives include, but are not limited to, antibacterial agents, antifungal agents, and antioxidants. [0428] Antibacterial agents include, but are not limited to, chlorocresol, diazolidinyl urea, dimethyl sulfoxide, glacial acetic acid, imidurea, iodine / edetic acid, phenylmercuric acetate, phenylmercuric borate, phenylmercuric hydroxide, potassium sorbate, sodium hydroxide, and sorbic acid. thymol, antiseptics and disinfectants. [0429] Antifungal agents include, but are not limited to, benzoic acid, butylene glycol, butyl paraben, chlorocresol, coconut oil, dimethyl sulfoxide, ethyl paraben, glacial acetic acid, imidurea, methyl paraben, phenylmercuric acetate, phenylmercuric borate, phenylmercuric potassium hydroxide, phenylmercuric hydroxide. , potassium sorylmercurbate, phenylmercuro hydroxide, propylparaben, sodium propionate, sodium thiosulfate, thymol, and vanillin. [0430] Surfactants include, but are not limited to: Polysorbate 80NF, polyoxyethylene 20 sorbitan monolaurate, polyoxyethylene (4) sorbitan monolaurate, polyoxyethylene 20 sorbitan monopalmitate, polyoxyethylene 20 sorbitan monostearate, polyoxyethane, polyethylene sorbitan, polyoxyethane, monoethylene, sorbethylene, sorbethylene, sorbethylene, sorbitan monoethylene, sorbet, polyoxethylene 20, polyoxethylene sorbet 20, polyoxyethylene sorbet 20 (5) sorbitan monooleate, polyoxyethylene 20 sorbitan trioleate, polyoxyethylene 20 sorbitan monoisotearate, sorbitan monooleate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monopalmitate, sorbitan monopalmitate , sorbitan trilaurate, sorbitan trioleate, sorbitan mixtures, similar to the trialeates of sorbitan, similar mixtures thereof. Due to their chemical properties, certain surfactants can function as preservatives and / or absorption enhancers in certain circumstances, depending on the concentration in the formulation and other factors. Surfactants include, but are not limited to: cationic, anionic, nonionic, and zwitterionic surfactants. [0431] Surfactants also include: anionic surfactants (e.g., carboxylate sulfonates, petroleum sulfonates, alkylbenzene sulfonates, naphthalene sulfonates, olefin sulfonates, alkyl sulfates, sulfates, sulfated natural oils and fats, sulfated esters, sulphated alkanolamides, ethoxylated alkyl phenoxylate sulfate alkylbentane sulfate), non-ionic surfactants (e.g., ethoxylated aliphatic alcohol, polyoxyethylene surfactants, carboxylic esters, polyethylene glycol esters, anhydrosorbitol esters and their ethoxylated derivatives, glycol esters of fatty acids, carboxylic amides, carboxylic amides, condensed carboxylic acids Polyoxyethylene fatty acid amides, cationic surfactants (cationic surfactants) salts, amide linked amines, polyoxyethylene alkyl and alicyclic amines, 4.n, n, n ', n' tetrakis substituted ethylenediamines, 2-alkyl 1-hydroxethyl 2- imidazolines), amphoteric surfactants (amphoteric surfactants contain both an acidic and basic hydrophilic moiety on its surface, for example, n-coco 3-aminopropionic acid / sodium salt, n-tallow 3-iminodipropionate, disodium salt, n-carboxymethyl n dimethyl n-9 octadecenyl ammonium hydroxide, n- cocoamidethyl n Hydroxyethylglycine, sodium salt, etc.). [0432] Antioxidants include, but are not limited to, tocopherol, arachidonic acid, ascorbic acid, ascorbyl palmitate, benzethonium chloride, benzethonium bromide, benzalkonium chloride, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), capric acid, acid caproic, carbon dioxide, cetylpyridium chloride, chelating agents, chitosan derivatives, citric acid monohydrate, dodecyl dimethyl aminopropionate, enanthic acid, erythorbic acid, ethyl oleate, fumaric acid, glycerol oleate, glyceryl monostearate, lauric acid, limonene, linolenic acid, lysine, lysine, acid lysine, menthol, methionine, monothioglycerol, myristic acid, oleic acid, sales of the same, palmitic acid, pelargonic acid, peppermint oil, phosphoric acid, polysorbates, potassium metabisulfite, propionic acid , propyl gallate, sodium ascorbate, sodium bisulfite, sodium caprate, sodium deoxycholate, sodium deoxyglycolate, sodium formaldehyde sulfoxylate, g sodium lucocholate, sodium hydroxybenzoyal aminocaprylate, sodium lauryl sulfate, sodium metabisulfite, sodium sulfite, sodium taurocholate, sodium thiosulfate, stearic acid, sulfur dioxide, and a combination thereof. [0433] Buffers include, but are not limited to, phosphate buffers, acetate buffers, citrate buffers. [0434] In some embodiments, the nasal spray formulation comprises a buffering agent. Buffering agents include, but are not limited to, adipic acid, boric acid, carbonate of calcium, calcium hydroxide, calcium lactate, calcium phosphate, tribasic, citric acid monohydrate, dibasic sodium phosphate, diethanolamine, glycine, maleic acid, malic acid, methionine, monobasic sodium phosphate, monoethanolamine, monosodium glutamate, phosphoric acid , potassium citrate, sodium acetate, sodium bicarbonate, sodium borate, sodium carbonate, sodium citrate dihydrate, sodium hydroxide, sodium lactate, and triethanolamine. Isotonicity agents include sodium chloride, calcium chloride, magnesium chloride, sorbitol, sucrose, dextrose, lactose, mannitol, trehalose, raffinose, polyethylene glycol, hydroxyethyl starch, glycerin, glycine, and the like, and mixtures thereof. In certain embodiments, the isotonicity agent is selected from dextrose, glycerin, mannitol, potassium chloride, and sodium chloride. In certain embodiments, the isotonicity agent is sodium chloride. In certain embodiments, the formulations described herein contain sodium chloride in an amount sufficient for the final composition to have a nasally acceptable osmolality, preferably 240-350 mOsm / kg. In certain embodiments, the formulations contain 0.3-1.9% sodium chloride. [0435] Sweeteners / flavoring agents / task masking agents include, but are not limited to, sucrose, dextrose, lactose, sucralose, acesulfame-K, aspartame, saccharin, sodium saccharin, citric acid, aspartic acid, eucalyptol, mannitol, glycerin, xylitol, menthol, glycyrrhizic acid, cinnamon oils, wintergreen oil, peppermint oils, clover oil, bay oil, anise oil, eucalyptus, vanilla, citrus oil such as lemon oil, orange oil, grape oil and grapefruit, fruit essences including apple, peach, pear, strawberry, raspberry, cherry, plum, pineapple, apricot, etc. and combinations thereof. In some embodiments, the formulations contain from about 0.0001 percent to about 1 percent of a sweetening agent / flavoring / task masking agent, and may be present in lower or higher amounts as a factor of one or more potency of effect. on the taste, solubility of the flavoring, effects of the flavoring on the solubility or other physicochemical or pharmacokinetic properties of other components of the formulation, or other factors. [0436] In certain embodiments, the pharmaceutical formulation further comprises an isotonicity agent. The intranasal formulation may comprise between about 0.2% (w / v) and about 1.2% (w / v) of isotonicity agent, such as about 0.2% (w / v), about 0.3% (w / v), about 0.4 % (p / v), approximately 0.5% (p / v), approximately 0.6% (p / v), approximately 0.7% (p / v), approximately 0.8% (p / v), approximately 0.9% (p / v ), about 1.0% (w / v), about 1.1% (w / v), or about 1.2% (w / v). The intranasal formulation may comprise greater than about 0.1% (w / v) isotonicity agent. The intranasal formulation may comprise less than about 1.2% (w / v) isotonicity agent. In other embodiments, the intranasal formulation may comprise between about 0.2% (w / v) and about 1.9% (w / v) of isotonicity agent, such as about 0.2% (w / v), about 0.3% (w / v), about 0.4% (w / v), about 0.5% (w / v ), about 0.6% (w / v), about 0.7% (w / v), about 0.8% (w / v), about 0.9% (w / v), about 1.0% (w / v), about 1.1% (w / v), about 1.2% (w / v), about 1.3% (w / v), about 1.4% (w / v), about 1.5% (w / v), about 1.6% (w / v) , about 1.7% (w / v), about 1.8% (w / v), or about 1.9% (w / v). The intranasal formulation may comprise less than about 1.9% (w / v) isotonicity agent. [0437] In certain embodiments, the formulation further comprises an absorption enhancer. In certain embodiments, the pharmaceutical formulation comprises between about 0.005% (w / v) to about 2.5% (w / v) of the absorption enhancer. In certain embodiments, the pharmaceutical formulation comprises between about 0.05% (w / v) to about 2.5% (w / v) of the absorption enhancer. In certain embodiments, the pharmaceutical formulation comprises between about 0.1% (w / v) to about 0.5% (w / v) of the absorption enhancer. In certain embodiments, the pharmaceutical formulation comprises about 0.25% (w / v) of the absorption enhancer. In certain embodiments, the pharmaceutical formulation comprises about 0.18% (w / v) of the absorption enhancer. [0438] In certain embodiments, the absorption enhancer is selected from benzalkonium chloride, cyclodextrins, chitosan, deoxycholic acid, an alkylsaccharide (for example, a nonionic alkylsaccharide surfactant such as an alkylglucoside and a fatty acid sucrose ester consisting of a aliphatic hydrocarbon chain coupled to a sugar residue by a glycosidic or ester bond, respectively), derivatives of fusidic acid, glycocholic acid, laureth-9, phosphatidylcholines, taurocholic acid, taurodihydrofusidic acid, microspheres and liposomes, and bile salts. In certain embodiments, the absorption enhancer is benzalkonium chloride. The formulation can comprise from about 0.01% (w / v) to about 1% (w / v) of benzalkonium chloride. In certain embodiments, the pharmaceutical formulation comprises about 0.005% (w / v) to about 0.015% (w / v) of benzalkonium chloride. In certain embodiments, the pharmaceutical formulation comprises about 0.01% (w / v), about 0.02% (w / v), about 0.03% (w / v), or about 0.04% (w / v) of benzalkonium chloride. In certain embodiments, the pharmaceutical formulation comprises about 0.01% (w / v) benzalkonium chloride. In certain embodiments, the pharmaceutical formulation comprises about 0.02% (w / v) benzalkonium chloride. In certain embodiments, the pharmaceutical formulation comprises about 0.04% benzalkonium chloride. In certain embodiments, the pharmaceutical formulation comprises benzalkonium chloride in an amount between about 0.001% (w / v) and about 1% (w / v). In others Certain embodiments, the pharmaceutical formulation comprises benzalkonium chloride in an amount between about 0.001% (w / v) and about 0.5% (w / v). In certain other embodiments, the pharmaceutical formulation comprises benzalkonium chloride in an amount between about 0.001% (w / v) and about 0.2% (w / v). In some embodiments, the pharmaceutical formulation comprises 0.001% (w / v), 0.003% (w / v), 0.005% [0439] (p / v), 0.007% (p / v), 0.009% (p / v), 0.01% (p / v), 0.02% (p / v), 0.03% (p / v), 0.04% (p / v), 0.05% [0440] (p / v), 0.06% (p / v), 0.07% (p / v), 0.08% (p / v), 0.09% (p / v), 0.1% (p / v), 0.11% (p / v), 0.12% (p / v), 0.13% (p / v), 0.14% (p / v), 0.15% (p / v), 0.16% (p / v), 0.17% (p / v), 0.18% (p / v), 0.19% (p / v), 0.2% [0441] (p / v), 0.31% (p / v), 0. 22% (p / v), 0.23% (p / v), 0.24% (p / v), 0.25% (p / v), 0. 26% (p / v), 0.27% [0442] (p / v), 0.28% (p / v), 0.29% (p / v), 0.3% (p / v), 0.31% (p / v), 0.32% (p / v), 0.33% (p / v), 0.34% (p / v), 0.35% (p / v), 0.36% (p / v), 0.37% (p / v), 0.38% (p / v), 0.39% (p / v ), 0.4% (p / v), 0.41% (p / v), 0.42% [0443] v [0445] [0447] 0.95% (p / v), 0.96% (p / v), 0.97% (p / v), 0.98% (p / v), 0.99% (p / v), or 1% (p / v) of chloride of benzalkonium. [0448] In certain embodiments, the absorption enhancer is an alkylsaccharide. In certain embodiments, the alkylsaccharide is selected from dodecyl maltoside, tetradecyl maltoside (TDM), and sucrose dodecanoate. [0449] In certain embodiments, the alkylsaccharide is dodecyl maltoside (the alkylglucoside 1-O-ndodecyl-pD-maltopyranoside, alternatively referred to as lauryl-pD-maltopyranoside, dodecyl maltopyranoside, and DDM; C 24 H 46 Q 11 , often referred to by the name Intravail® commercial). Alkylsaccharides are used in commercial food and personal care products and have been designated Generally Recognized As Safe Substances (GRAS) for food applications. They are non-irritating transmucosal absorption enhancers that are odorless, tasteless, non-toxic, non-mutagenic and non-sensitizing in the Draize test up to a concentration of 25%. Alkylsaccharides increase absorption by increasing paracellular permeability, as indicated by a decrease in transepithelial electrical resistance; They can also increase transcytosis. The effect is short-lived. Other alkylsaccharides include tetradecyl maltoside (TDM) and sucrose dodecanoate. [0450] In certain embodiments, an intranasal formulation comprises between about [0451] 0.05% (w / v) and approximately 2.5% (w / v) of Intravail®. In certain embodiments, an intranasal formulation comprises between about 0.1% (w / v) and about [0452] 0.5% (w / v) of Intravail®. In certain embodiments, an intranasal formulation comprises between about 0.15% (w / v) and about 0.35% (w / v) of Intravail®. In certain embodiments, an intranasal formulation comprises between about 0.15% (w / v) and about 0.2% (w / v) of Intravail®. In certain embodiments, an intranasal formulation comprises about 0.18% (w / v) of Intravail®. In certain embodiments, an intranasal formulation comprises about 0.2% (w / v) to about 0.3% (w / v) of Intravail®. In certain embodiments, an intranasal formulation comprises approximately 0.25% (w / v) Intravail®. [0453] In certain embodiments, the absorption enhancer is Intravail® (dodecyl maltoside). [0454] In certain embodiments, the absorption enhancer in the intranasal formulation is a combination of dodecyl maltoside and benzalkonium chloride. While the use of dodecyl maltoside or benzalkonium chloride as an absorption enhancer in the intranasal formulations described in this document provides bioavailability of intransal Epinephrine, the combination of dodecyl maltose and benzalkonium chloride as absorption enhancers in the intranasal formulations described in this document it provides a pharmacokinetics that closely resembles the pharmacokinetics obtained by intramuscular injection of epinephrine. [0455] In certain embodiments, each dose dispensed from the pharmaceutical formulation device comprises between about 0.4 mg and about 2.40 mg per dose of Epinephrine, or a salt thereof, and between 0.1 and 0.50 mg Intravail® (dodecyl maltoside). In certain embodiments, each dispensed dose from the formulation device comprises between about 0.5 mg and about 2.0 mg per dose of Epinephrine, or a salt thereof, and about between 0.1 and 0.50 mg Intravail® (dodecyl maltose). [0456] In certain embodiments, each dispensed dose from the formulation device comprises between about 0.75 mg and about 1.5 mg per dose of Epinephrine, or a salt thereof, and between 0.1 and 0.50 mg Intravail® (dodecyl maltose). [0457] In certain embodiments, each dispensed dose of the formulation device comprises between about 0.9 mg and about 1.15 mg per dose of Epinephrine, or a salt thereof, and about 0.25 mg Intravail® (dodecyl maltose). [0458] In certain embodiments, each dispensed dose from the formulation device comprises about 1.0 mg per dose of Epinephrine, or a salt thereof, and about 0.25 mg Intravail® (dodecyl maltoside). [0459] In certain embodiments, the pharmaceutical formulation further comprises a chelating or antioxidant agent (stabilizing agent) to improve stability. In certain embodiments, the chelating agent / stabilizer is EDTA. [0460] Examples of additional stabilizing agents include: acacia, agar, albumin, alginic acid, aluminum stearate, ammonium alginate, ascorbic acid, ascorbyl palmitate, Bentonite, Butylated Hydroxytoluene (BHT), Calcium Alginate, Calcium Stearate, Calcium Carboxymethyl Cellulose, Sodium Carboxymethyl Cellulose, Carrageenan, Cellulose, Microcrystalline, Sodium Carboxymethyl Cellulose, Keratonia, Colloidal Silicon Dioxide, Ethylene Cyclodextrins, Diethanolamine, Ethyletheol ethanolamine , glycerin monostearate, guar gum, hectorite, hydroxpropyl betadex, hydroxypropyl cellulose, hypromellose, inulin, invert sugar, lauric acid, lecithin, aluminum magnesium silicate, mineral oil and lanolin alcohols, monoethanolamine, pectin, pentatic acid, Phospholipids, Polacrylin Potassium, Poloxamer, Polyvinyl Alcohol, Potassium Alginate, Potassium Chloride, Povidone, Propyl Gallate, Propylene Glycol, Propylene Glycol Alginate, Raffinose, Sodium Acetate, Sodium Alginate, Sodium Borate, Sodium Stearyl Fumarate , sorbitol, stearyl alcohol, sulfobutylether b-cyclodextrin, tagatose, trehalose, triethanol amine, white wax, xanthan gum, xylitol, yellow wax, and zinc acetate. [0461] Examples of additional chelating agents include: citric acid monohydrate, edetate disodium, edetate calcium disodium, edetate acid, fumaric acid, malic acid, maltol, pentetic acid, edetate sodium, and edetate trisodium. [0462] In certain embodiments, the pharmaceutical formulation comprises benzalkonium chloride. In certain embodiments, the pharmaceutical formulation comprises about 0.005% (w / v) to about 1% (w / v) of benzalkonium chloride. [0463] In its capacity as a surfactant, benzalkonium chloride can affect the surface tension of the drops from a delivered nasal spray column, producing spherical or substantially spherical particles that have a narrow drop size distribution (DSD), as well as the viscosity of a liquid formulation (DSD). [0464] In certain embodiments, the absorption enhancer is an alkylsaccharide, for example, a nonionic alkylsaccharide surfactant such as an alkylglucoside and a fatty acid sucrose ester consisting of an aliphatic hydrocarbon chain coupled to a sugar moiety by a bond. glycosidic ester, respectively. In certain embodiments, the absorption enhancer is an alkylmaltoside ( eg, a tetradecyl maltoside (TDM), a dodecyl maltoside (DDM), etc.). In certain embodiments, the absorption enhancer is sucrose dodecanoate. Alkylsaccharides are used in commercial food and personal care products and have been designated Generally Recognized As Safe (GRAS) substances for food applications. They are non-irritating transmucosal absorption enhancers that are odorless, tasteless, non-toxic, non-mutagenic and non-sensitizing in the Draize test up to a concentration of 25%. Without being bound by theory, it is believed that alkylsaccharides increase absorption by increasing paracellular permeability, as indicated by a decrease in transepithelial electrical resistance; They can also increase transcytosis. The effect can be short-lived. In its capacity as an absorption enhancer, alkylmaltosides ( for example, a tetradecyl maltoside (TDM), a dodecyl maltoside (DDM), etc.) can affect the surface tension of drops from a supplied nasal spray column, producing spherical or substantially spherical particles that have a narrow drop size distribution (DSD), as well as the viscosity of a formulation liquid. [0465] In certain embodiments, the absorption enhancer is 1-On-dodecyl-pD-maltopyranoside alkylsaccharide (alternatively known as lauryl-pD-maltopyranoside, dodecyl maltopyranoside, dodecyl maltoside, and DDM; C 24 H 46 Q 11 ; often referred to under the Intravail® trademark). In certain embodiments, an intranasal formulation comprises from about 0.01% (w / v) to about 2.5% (w / v) of DDM. In certain embodiments, an intranasal formulation comprises from about 0.1% (w / v) to about 0.5% (w / v) of DDM. In certain embodiments, an intranasal formulation comprises from about 0.15% (w / v) to about 0.35% (w / v) of DDM. In certain embodiments, an intranasal formulation comprises from about 0.15% (w / v) to about 0.2% (w / v) of DDM. In certain embodiments, an intranasal formulation comprises about 0.18% (w / v) DDM. In certain embodiments, an intranasal formulation comprises from about 0.2% (w / v) to about 0.3% (w / v) of DDM. In certain embodiments, an intranasal formulation comprises about 0.25% (w / v) DDM. [0466] In sugar chemistry, an anomer is a pair of cyclic stereoisomers (designated a or p) of a sugar or glucoside, differing only in the configuration of the hemiacetal (or hemiketal) carbon, also called anomeric carbon or reducing carbon. If the structure is analogous to the Hydroxyl group on the anomeric carbon in the axial position of glucose, then the sugar is an alpha anomer. However, if Hydroxil is equatorial, the sugar is a beta anomer. For example, a-D-glucopyranose and p-D-glucopyranose, the two cyclic forms of glucose, are anomers. Also, alkylglucosides occur as anomers. For example, dodecyl p-D-maltoside and dodecyl a-D-maltoside are two cyclic forms of dodecyl maltoside. The two different anomers are two different chemical structures, so they have different physical and chemical properties. In one aspect of the invention, the alkylglycoside of the present invention is a p anomer. In an exemplary aspect, the alkylglucoside is a p-anomer of an alkylmaltoside, such as tetradecyl-p-D-maltoside (TDM). [0467] In some embodiments, the alkylglucoside used is a substantially pure alkylglucoside. As used herein, a "substantially pure" alkylglucoside refers to an anomeric form of the alkylglucoside (the a or p anomeric forms) with less than about 2% of the other anomeric form, preferably less than about 1.5% of the another anomeric form, and more preferably less than about 1% of the other anomeric form. In one aspect, a substantially pure alkylgycoside contains more than 98% of the a or p anomer. In another respect, a Substantially pure alkylgycoside contains more than 99% of the a or p anomer. In another aspect, a substantially pure alkylgycoside contains greater than 99.5% of either a or p anomers. In another aspect, a substantially pure alkylgycoside contains greater than 99.9% of either a or p anomers. [0468] [00340] In certain embodiments, described herein, it is an aqueous formulation suitable for intranasal administration comprising: Epinephrine; Water; and one or more ingredients selected from absorption enhancers, chelating agents, antioxidants, stabilizing agents, surfactants, isotonicity agents, and pH adjusting agents. [0469] [00341] In certain embodiments, described herein, it is an aqueous formulation suitable for intranasal administration comprising: Epinephrine; Water; and one or more ingredients selected from alkylglucosides, chitosan, alkylcyclodextrins, benzalkonium chloride, sodium chloride, and EDTA. [0470] [00342] In certain embodiments, described herein, it is an aqueous formulation suitable for intranasal administration comprising: Epinephrine; Water; and one or more ingredients selected from Dodecyl Maltoside (DDM), Tetradecyl Maltoside (TDM), Benzalkonium Chloride, Sodium Chloride, Hydrochloric Acid, and EDTA. In certain other embodiments, described herein is an aqueous formulation suitable for intranasal administration comprising: Epinephrine; Water; and one or more ingredients selected from dodecyl maltoside (DDM), benzalkonium chloride, sodium chloride, and EDTA. [0471] [00343] In certain embodiments, described herein, it is an aqueous formulation suitable for intranasal administration comprising: Epinephrine; Water; dodecyl maltoside (DDM); and one or more ingredients selected from benzalkonium chloride, sodium chloride, pH adjusting agent, and EDTA. [0472] [00344] In certain embodiments, described herein, it is an aqueous formulation suitable for intranasal administration comprising: Epinephrine; Water; benzalkonium chloride; and one or more ingredients selected from dodecyl maltose (DDM), sodium chloride, pH adjusting agents, and EDTA. [0473] In certain embodiments, an aqueous formulation suitable for intranasal administration is described herein comprising: Epinephrine; Water; Dodecyl Maltoside (DDM) or Benzalkonium Chloride or a combination of Dodecyl Maltoside (DDM) and Benzalkonium Chloride; and one or more additional ingredients selected from sodium chloride, pH adjusting agents, and EDTA. [0474] PH adjusting agents include acids described herein (eg, hydrochloric acid, citric acid), buffers (eg, phosphate, acetate, and citrate buffers), bases, and (eg, sodium hydroxide, citrate sodium, sodium bicarbonate, sodium carbonate). [0475] In certain embodiments, an aqueous formulation suitable for intranasal administration is disclosed herein comprising: about 0.5% (w / v) to about 2.5% (w / v) Epinephrine; one or more aqueous ingredients selected from: about 0.05% (w / v) to about 2.5% (w / v) dodecyl maltoside (DDM); from about 0.005% (w / v) to about 1% (w / v) of benzalkonium chloride; from about 0.2% (w / v) to about 1.2% (w / v) of sodium chloride, optional hydrochloric acid or sodium hydroxide in an amount sufficient to adjust the pH to a final pH of about 4.0 to about 5.0; and about 0.05% (w / v) to about 2.0% (w / v) EDTA. [0476] In certain embodiments, an aqueous formulation suitable for intranasal administration is described herein comprising: i) from about 0.5% (w / v) to about 2.5% (w / v) Epinephrine; ii) water; iii) about 0.05% (w / v) to about 2.5% (w / v) of dodecyl maltoside (DDM) or about 0.005% (w / v) to about 1% (w / v) of benzalkonium chloride, or a combination of about 0.05% (w / v) to about 2.5% (w / v) of dodecyl maltoside (DDM) and about 0.005% (w / v) to about 1% (w / v) of benzalkonium chloride; and iv) one or more ingredients selected from a) from about 0.2% (w / v) to about 1.2% (w / v) of sodium chloride; b) optional hydrochloric acid or sodium hydroxide in an amount sufficient to adjust the pH to the final pH of about 4.0 to about 5.0; and c) from about 0.05% (w / v) to about 2.0% (w / v) EDTA. [0477] In certain embodiments, an aqueous formulation suitable for intranasal administration is described herein comprising: i) from about 0.9% (w / v) to about 2.0% (w / v) of Epinephrine; ii) water; iii) from about 0.05% (w / v) to about 2.5% (w / v) of dodecyl maltoside (DDM), or from about 0.005% (w / v) to about 1% (w / v) of benzalkonium chloride , or a combination of about 0.05% (w / v) to about 2.5% (w / v) of dodecyl maltoside (DDM) and from about 0.005% (w / v) to about 1% (w / v) of chloride benzalkonium; and iv) one or more ingredients selected a) from about 0.2% (w / v) to about 1.2% (w / v) sodium chloride; b) optional hydrochloric acid or sodium hydroxide hydrochloric acid in an amount sufficient to adjust the pH to a final pH of from about 4.0 to about 5.0; and c) from about 0.05% (w / v) to about 2.0% (w / v) EDTA. [0478] In certain embodiments, an aqueous formulation suitable for intranasal administration is described herein comprising: from about 0.5% (w / v) to about 2.5% (w / v) Epinephrine; Water; from about 0.05% (w / v) to about 2.5% (w / v) dodecyl maltoside (DDM); from about 0.005% (w / v) to about 1% (w / v) of benzalkonium chloride; from about 0.2% (w / v) to about 1.2% (w / v) of sodium chloride, hydrochloric acid in an amount sufficient to adjust the pH to a final pH of about 4.0 to about 5.0; and about 0.05% (w / v) to about 2.0% (w / v) EDTA. [0479] In certain embodiments, an aqueous formulation suitable for intranasal administration is described herein comprising: Epinephrine; Water; one or more absorption enhancing agents; an isotonicity agent; a stabilizing agent; a preservative and optional pH adjusting agents for adjusting the pH to a pH of 3 to 6. In certain embodiments, an aqueous formulation suitable for intranasal administration is described herein comprising: Epinephrine; Water; one or more absorption enhancing agents (eg, dodecyl maltoside; benzalkonium chloride; or a combination of dodecyl maltoside and benzalkonium chloride); an isotonicity agent (eg, sodium chloride); a stabilizing agent (eg, EDTA or disodium EDTA); a preservative (eg, benzalkonium chloride); and optional pH adjusting agents to adjust the pH to a pH of 3 to 6. In certain embodiments, an aqueous formulation suitable for intranasal administration is described herein comprising: Epinephrine; Water; one or more absorption enhancing agents (eg, dodecyl maltoside; benzalkonium chloride; or a combination of dodecyl maltoside and benzalkonium chloride); an isotonicity agent (eg, sodium chloride); a stabilizing agent (eg, EDTA or disodium EDTA); a preservative (eg, benzalkonium chloride); an antioxidant; a buffering agent; and optional pH adjusting agents to adjust the pH to a pH of 3 to 6. [0480] In certain embodiments, an aqueous formulation suitable for intranasal administration is described herein comprising: Epinephrine; Water; dodecyl maltoside or benzalkonium chloride or a combination of dodecyl maltoside and benzalkonium chloride; sodium chloride; EDTA or EDTA disodium; and optional pH adjusting agents to adjust the pH to a pH of 3 to 6. [0481] In certain embodiments, an aqueous formulation suitable for intranasal administration is disclosed herein comprising: about 0.5% (w / v) to about 2.5% (w / v) Epinephrine; Water; from about 0.05% (w / v) to about 2.5% (w / v) of dodecyl maltoside or from about 0.005% (w / v) to about 1% (w / v) of benzalkonium chloride or a combination of about 0.05 % (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.005% (w / v) to about 1% (w / v) of benzalkonium chloride; from about 0.2% (w / v) to about 1.2% (w / v) of sodium chloride; from about 0.05% (w / v) to about 2.0% (w / v) EDTA or disodium EDTA; and optional pH adjusting agents to adjust the pH to a pH of 3 to 6. [0482] In some embodiments, a 100 pL sample of the aqueous formulation suitable for Intranasal administration comprises less than about 2.5mg of Epinephrine. In some embodiments, a 100 pL sample of the aqueous formulation suitable for intranasal administration comprises from about 0.5mg to about 2.5mg of Epinephrine. In some embodiments, a 100 pL sample of the aqueous formulation suitable for intranasal administration comprises about 0.5 mg, about 0.6 mg, about 0.7 mg, about 0.8 mg, about 0.9 mg, about 1.0 mg, about 1.1 mg, about 1.2 mg, about 1.3 mg, about 1.4 mg, about 1.5 mg, about 1.6 mg, about 1.7 mg, about 1.8 mg, about 1.9 mg, about 2.0 mg, about 2.1 mg, about 2.2 mg, about 2.3 mg, about 2.4 mg, or about 2.5 mg of Epinephrine. [0483] Nasal Drug Delivery Devices & Kits [0484] Nasal drug delivery devices are also provided comprising a formulation described herein. In certain embodiments, the device is pre-prepared. In certain Embodiments, the device may be printed prior to use. In certain embodiments, the device can be operated with one hand. [0485] Nasal delivery is considered an attractive, safe, and easy to administer route for needle-free systemic drug delivery, especially when a rapid absorption effect is desired. Additionally, nasal delivery can help address issues related to poor bioavailability, slow absorption, drug breakdown, adverse events (AEs) in the gastrointestinal tract, and prevents first-pass metabolism in the liver. [0486] Liquid nasal formulations are primarily aqueous solutions, but suspensions, emulsions, liposomes, and microspheres can also be administered. Other liquid formulations can comprise liposomes, microspheres, aqueous and mixed organic formulations, non-aqueous formulations, dry powder, and retentive formulations (gels). In traditional spray pump systems, antimicrobial preservatives are generally required to maintain microbiological stability in liquid formulations. Dominated spray pumps have dominated the nasal drug delivery market since their introduction. The pumps typically deliver 100 pL (25-250 pL) per spray, and offer high reproducibility of emitted dose and column geometry in in vitro tests. [0487] Examples of standard metered spray pumps include those offered by Aptar Pharma, Inc., as the "classic technology platform" multi-dose nasal spray devices, and by BD Medical-Pharmaceutical Systems, as the Accuspray ™ system. Such devices comprise a reservoir containing multiple doses of the nasal spray formulation (for example, 50, 100, 150, 200, 60, or 120 doses), a closure (for example, screw, crimp, or complement), and an actuator that delivers 45 to 1000 pL (for example, 50. [0488] 100, 140, 150, or 200 pL) of fluid per actuation comprises a single dose. The actuator can be configured to count a dose, deliver gel formulations, deliver in an inverted configuration, etc. [0489] In traditional multipurpose spray pump systems, antimicrobial preservatives are typically required to maintain microbiological stability in liquid formulations. However, preservative-free systems are also available, for example Aptar's Advanced Preservative-Free (APF) system, which is vented, contains a filter membrane for air flow that prevents contamination, has a fluid pathway free of metals for oxidizing formulations, and can be used in any orientation. Additional nasal spray devices from Aptar and others are optimized with clog-free dispensing tips (useful for high-viscosity, high-volatility formulations), actuators that do not need priming after long periods of disuse, etc. boosted propellant. However, additional nasal spray devices include dry powder inhalers. [0490] The particle size and geometry of the column can vary within certain limits and depend on the properties of the pump, the formulation, the actuator orifice and the applied force. The droplet size distribution of a nasal spray is a critical parameter, as it significantly influences the in vivo deposition of the drug in the nasal cavity. The droplet size is influenced by the drive parameters of the device and the formulation. The Mediana of the average droplet size should be between about 30 and about 100 pm. If the droplets are too large (> about 120 pm), the stool occurs mainly in the anterior parts of the nose, and if the droplets are too small (<about 10 pm), they can possibly be inhaled and reach the lungs and through orally. cavity, which should be avoided for safety reasons. In their capacity as a surfactant, benzalkonium chloride and alkylmaltosides (for example, a tetradecyl maltoside (TDM), a dodecyl maltoside (DDM), etc.) can affect the surface tension of the droplets from a supplied nasal spray cloud, producing Spherical or substantially spherical particles that have a narrow drop size distribution (DSD) as well as the viscosity of a liquid formulation. [0491] The column geometry, droplet size and DSD of the delivered column after spraying can be measured under specified experimental and instrumental conditions by appropriate analytical and / or calibrated procedures known in the art. These include photography, laser diffraction, and impaction systems (cascade impaction, NGI). Column geometry, droplet size, and DSD can affect pharmacokinetic results such as Cmax, Tmax, and dose proportionally. [0492] The droplet size distribution can be controlled in terms of ranges for D10. [0493] D50. D90. span [(D90-D10) / D50], and percentage of drops less than 10 mm. In certain embodiments, the formulation will have a narrow DSD. In certain embodiments, the formulation will have a D (v, 50) of 30-70 gm and a D (v, 90) <100 gm. [0494] In certain Embodiments, the percentage of drops less than 10 gm will be less than 10%. In certain Embodiments, the percentage of drops less than 10 gm will be less than 5%. In certain Embodiments, the percentage of drops less than 10 gm will be less than 2%. In certain Embodiments, the percentage of drops less than 10 gm will be less than 1%. [0495] In certain Embodiments, the formulation when dispensed by actuation of the device will produce a uniform circular column with an ovality ratio close to 1. The ovality ratio is calculated as the ratio of the maximum diameter (Dmax) and the minimum diameter (Dmin) of a spray pattern taken orthogonal to the direction of the spray flow ( eg, from "above"). In certain Embodiments, the ovality ratio is less than ± 2.0. In certain Embodiments, the ovality ratio is less than ± 1.5. In certain Embodiments, the ovality ratio is less than ± 1.3. In certain Embodiments, the ovality ratio is less than ± 1.2. In certain Embodiments, the ovality ratio is less than ± 1.1. [0496] The details and mechanical principles of particle generation have been described for different types of nasal spray devices. See, Vidgren and Kublik, Adv. Drug Deliv. Rev. [0497] 29: 157-77, 1998. Traditional spray pumps replace the emitted liquid with air, so preservatives are required to prevent contamination. However, driven by studies suggesting possible negative effects of preservatives, pump manufacturers have developed different spray systems that avoid the need for preservatives. These systems use a collapsible bag, movable piston, or compressed gas to compensate for the volume of liquid emitted (on the World Wide Web at aptar.com and on the World Wide Web at rexam.com). Solutions with a collapsible bag and a movable piston that compensates for the volume of liquid emitted offer the additional advantage that they can be emitted backwards, without the risk of drawing air into the dip tube and compromising the subsequent spray. This may be useful for some products where patients are bedridden and where a head-down application is recommended. Another method used to avoid preservatives is that the air that replaces the emitted liquid is filtered through an aseptic air filter. In addition, some systems have a ball valve at the tip to prevent contamination of the liquid within the applicator tip. More recently, the pumps have been designed with side drive. The pump was designed with a shorter tip to avoid contact with sensitive mucosal surfaces. New designs reduce the need to prime and re-prime, pumps incorporating pressure point features improve dose reproducibility, and dose counters and locking mechanisms for better dose control and safety are available (on the World Wide Web at rexam.com and on the World Wide Web at aptar.com). [0498] Traditional, single, single, bi-dose and multipurpose metered dose spray pumps require priming and some degree of overfilling to maintain dose compliance for the number of doses marked. They are suitable for drugs that are administered daily for a prolonged period, but due to the priming procedure and limited dosage control, unless a specialized device is selected, they are less suitable for drugs with a narrow therapeutic time period in which use the device, especially if they are not used frequently. For expensive drugs and drugs intended for single administration or sporadic use and where tight control of dosage and formulation is important, single dose (UDS) or two dose sprinkler (BDS) devices are preferred (on the World Wide Web in aptar .com). LMA offers a simple variant of a Single Dose Spray Device (MAD ™) (LMA, Salt Lake City, UT, USA; on the World Wide Web at lmana.com). A spray tip nozzle is equipped with a standard syringe. The liquid medicine to be administered is first put into the syringe and then the spray tip is attached to the syringe. This device has been used in academic studies and delivers, for example, a topical steroid in patients with chronic rhinosinusitis and in a vaccine study. A pre-filled device based on the same principle for one or two doses (Accuspray ™, Becton Dickinson Technologies, Research Triangle Park, NC, USA; on the World Wide Web at bdpharma.com) is used to deliver the FluMist ™ influenza vaccine (on the World Wide Web at flumist.com), approved for both adults and children in the US market. A Swiss company marketed a similar two-dose device for the delivery of another flu vaccine a decade ago. [0499] Single and pre-primed dosing devices are also available, consisting of a reservoir, piston, vortex chamber (See, for example, UDS UnitDose ™ and BDS BiDose ™ devices from Aptar, formerly Pfeiffer). Spray is formed when liquid is expelled through the swirl chamber. These devices are held between the second and third fingers with the thumb on the actuator. A pressure point mechanism incorporated in some devices ensures the reproducibility of the actuation force and the characteristics of the emitted pen. Currently, nasal migraine medications such as Imitrex® (on the World Wide Web at gsk.com) and Zomig® (on the World Wide Web at az.com; Pfeiffer / Aptar single-dose device), the vaccine marketed against influenza Flu-Mist (on the World Wide Web at flumist.com; Becton Dickinson single-dose spray device), and the intranasal formulation of naloxone for the rescue of opioid overdose, Narcan Nasal® (on the World Wide Web at narcan.com; Adapt Pharma) are delivered with this type of device. [0500] In certain embodiments, the 90% confidence interval for the dose administered by drive is ± approximately 2%. In certain embodiments, the 95% confidence interval for the dose delivered per actuation is ± about 2.5%. [0501] Historically, the intranasal administration of drugs in large volume, such as from syringes fitted with mucosal spray devices (MAD), has encountered difficulties due to the tendency of some of the formulations to drip through the nose into the nasopharynx. Consequently, In certain Embodiments, when nasally administering said pharmaceutical formulation to said patient, less than 20% of said pharmaceutical formulation exits the nasal cavity through drainage into the nasopharynx externally. In certain embodiments, after nasal delivery of said pharmaceutical formulation to said patient, less than 10% of said pharmaceutical formulation exits the nasal cavity through drainage into the nasopharynx externally. In certain embodiments, after nasal delivery of said pharmaceutical formulation to said patient, less than 5% of said pharmaceutical formulation exits the nasal cavity through drainage into the nasopharynx or externally. [0502] Current designs of container closure systems for inhalation aerosol pharmaceuticals include premeasured and device-metered presentations utilizing mechanical or power assistance and / or patient inspiration energy for spray column production. Premeasured presentations contain pre-metered doses or a fraction of doses in some types of units (eg, single or multiple ampoules or other cavities) that are subsequently inserted into the device by the patient during manufacture or before use. Typical device metered units have a reservoir containing a formulation sufficient for multiple doses that the device itself delivers as metered aerosols when activated by the patient. [0503] [00371] With aseptic techniques, the use of preservatives may not be necessary in previously primed devices, but an overfill is required which results in a residue fraction similar to multi-dose metered dose aerosols. emits 100 pL, a 125 pL volume is filled into the device (Pfeiffer / Aptar single dose device) used for intranasal migraine medications Imitrex ™ (sumatriptan) and Zomig ™ (zolmitriptan) and about half of that for a bi-dose design. Sterile pharmaceuticals can be produced by aseptic processing or terminal sterilization. Terminal sterilization generally involves filling and sealing product containers under high-quality ambient conditions. Products are filled and sealed in this type of environment to minimize the microbial particle content of the in-process product and help ensure that the subsequent sterilization process is successful. In most cases, the product, container, and closure have a low bioburden, but are not sterile. The product in its final container undergoes a sterilization process such as heat, irradiation, or chemical (gas). In an aseptic process, the drug, the The container and closure are first subjected to separate sterilization methods, as appropriate, and then put together. Since there is no process to sterilize the product in its final container, it is critical that the containers are filled and sealed in an efficient quality environment. Aseptic processing involves more variables than terminal sterilization. Before aseptic assembly into a final product, individual parts of the final product can generally undergo various sterilization processes. For example, glass containers are subjected to dry heat; rubber closures are subjected to humid heat; Liquid dosage forms are filtered. Each of these manufacturing processes requires validation and control. [0504] The devices mentioned herein can employ any of the pharmaceutical formulations, and are useful in the methods described herein. [0505] Accordingly, provided herein are devices adapted for the nasal delivery of a pharmaceutical formulation to a patient, comprising a reservoir with a therapeutically effective amount of Epinephrine. [0506] In certain embodiments, Epinephrine is the only pharmaceutically active compound in the pharmaceutical formulation. [0507] In certain embodiments, the volume of the pharmaceutical formulation in the reservoir is no greater than about 140 pL. [0508] In certain embodiments, the volume of the pharmaceutical formulation in the reservoir is greater than about 125 pL and less than 140 pL. [0509] In certain embodiments, approximately 100 pL of the pharmaceutical formulation in the reservoir is delivered to the patient in one actuation. [0510] In some embodiments, about 100 pL of the pharmaceutical formulation in the reservoir is delivered to the patient in one actuation and comprises less than about 2.5 mg of Epinephrine. In some embodiments, about 100 pL of the pharmaceutical formulation in the reservoir is delivered to the patient in one actuation and comprises about 0.5 mg to about 2.5 mg of Epinephrine. In some embodiments, approximately 100 pL of the pharmaceutical formulation in the reservoir is delivered to the patient in one actuation and comprises approximately 0.5mg, approximately 0.6mg, approximately 0.7mg, approximately 0.8mg, approximately 0.9mg, approximately 1.0mg, approximately 1.1mg , about 1.2 mg, about 1.3 mg, about 1.4 mg, about 1.5 mg, about 1.6 mg, about 1.7 mg, about 1.8 mg, about 1.9 mg, about 2.0 mg, about 2.1 mg, about 2.2 mg, about 2.3 mg, about 2.4 mg, or about 2.5 mg of Epinephrine. [0511] In certain embodiments, the pharmaceutical formulation further comprises one or more Excipients selected from water, EDTA and sodium chloride. In certain embodiments, the pharmaceutical formulation further comprises benzalkonium chloride. [0512] In some embodiments, approximately 100 pl of the aqueous pharmaceutical formulation in the reservoir is delivered to the patient in a single action and comprises Epinephrine, dodeclimaltoside or benzalkonium chloride or a combination of dodeclimaltoside and benzalkonium chloride, EDTA, and NaCl. [0513] In certain embodiments, the pharmaceutical formulation is substantially free of antimicrobial preservatives. [0514] In certain embodiments, the pharmaceutical formulation further comprises a compound that acts as a preservative, absorption enhancer, and / or a cationic surfactant; an isotonicity agent; a stabilizing agent; and an amount of acid or base sufficient to achieve a pH of from about 3.5 to about 6.0. The use of absorption enhancers, such as alkylsaccharides, cyclodextrins, and chitosans can increase the rate at which epinephrine is absorbed. In general, absorption enhancers provide better pharmacokinetic results, such as increasing Cmax, decreasing Tmax, and dose proportionally compared to intramuscular formulations and intranasal formulations that do not contain an absorption enhancer. Without being bound by theory, such absorption enhancers typically work by affecting two primary mechanisms for nasal absorption: paracellular transport through the opening of tight junctions between cells, and transcellular transport or transcytosis through cells. cells via vesicle transporters. [0515] Some excipients that improve absorption can alter the transcellular and / or paracellular pathways, others can extend the residence time in the nasal cavity or prevent metabolic changes. Without an absorption enhancer, the molecular weight limit for nasal absorption is approximately 1 kDa, whereas administration of drugs in conjunction with absorption enhancers may allow the absorption of 1 to 30 kDa molecules. However, intranasal administration of most absorption enhancers can cause damage to the nasal mucosa. See Maggio, J. Excipients and Food Chem. [0516] 5 (2): 100-12, 2014. Examples of absorption enhancers include aprotinin, benzalkonium chloride, benzyl alcohol, capric acid, ceramides, cetylpyridinium chloride, chitosan, cyclodextrins, deoxycholic acid, decanoyl carnitine, EDTA, acid Glycocholic, Glycodeoxycholic Acid, Glycofurol, Glycosylated Sphingosines, Glycyrrhetinic Acids, 2-Hydroxypropyl-pcyclodextrin, Laureth-9, Lauric Acid, Lauroyl Carnitine, Lauryl Sulfate, Lysophosphatidylcholine, Menthol, Poloxamer-Arginyl-Arginyl-Poloxamer 407, Poloxamer-L-Arginyl-Arginyl -polyoxyethyl-ether-polyoxyethyl-ether-polyoxyether polysorbate 80, propylene glycol, quillaia saponin, salicylic acid, p-sitosterol-pD-glucoside, sucrose cocoate, taurocholic acid, taurodeoxycholic acid, taurodihydrofusdecyl acid, mallido-tetrahydrofusidic acid, mallido-talidodecyltosis, and malidocyl saccharides Y sucrose dodecanoate. [0517] Epinephrine can optionally exist as pharmaceutically acceptable salts including pharmaceutically acceptable acid addition salts prepared from non-toxic pharmaceutically acceptable acids including inorganic and organic acids. Representative acids include, but are not limited to, acetic, benzenesulfonic, benzoic, camphor sulfonic, citric, ethenesulfonic, dichloroacetic, formic, fumaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic , mucic, nitric, oxalic, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, oxalic, p-toluenes similar. Acid addition salts can be obtained as direct synthesis products of compounds. Alternatively, the free base can be dissolved in a suitable solvent containing the appropriate acid and isolated salt by evaporating the solvent or otherwise separating the salt and solvent. The salt can form solvates with standard low molecular weight solvents using methods known to those skilled in the art. [0518] In certain embodiments, the device is filled with the pharmaceutical formulation using a sterile filling. [0519] In certain embodiments, the pharmaceutical formulation is chemically stable on storage for about twelve months at about 25 ° C and about 60% relative humidity and about six months at about 40 ° C and about 75% relative humidity. [0520] In some embodiments, intranasal epinephrine is administered as an aqueous solution, aqueous suspension, aqueous emulsion, nonaqueous solution, nonaqueous suspensions, nonaqueous emulsion, a solution with halogenated hydrocarbon propellant (s), or as a dry powder. In some embodiments, the aqueous formulations are sprayed into the nostril. In some embodiments, the aqueous formulations are aerosolized by liquid nebulizers employing hydraulic or ultrasonic atomization. Propellant-based systems can use suitable pressurized metered dose inhalers (pMDI). Dry powders can use dry powder inhaler (DPI) devices, which are able to disperse the drug substance effectively. [0521] Typically used propellants include chlorofluorocarbons, hydrochlorofluorocarbons, hydrofluorocarbons, hydrocarbons, and compressed gases. [0522] In some embodiments, intranasal epinephrine is delivered as a nasal spray produced by a nasal pressurized metered dose inhaler (pMDI). In some embodiments, the pMDI is a hydrofluoroalkane (HFA) -based pMDI for nasal use. Like spray pumps, nasal pMDI produces a localized deposition in the non-ciliated anterior epithelium of the nasal vestibule and anterior parts of the narrow nasal valve, but due to rapid evaporation of the aerosol delivered with a pMDI, a Noticeable "dripping" may be less of a problem. [0523] In some embodiments, the Epinephrine is delivered with a nebulizer. Nebulizers use compressed gases (air, oxygen, nitrogen) or ultrasonic or mechanical energy to dissolve medical solutions and suspensions into small aerosol droplets that can be inhaled directly into the nose. Smaller particles and the slow velocity of the nebulized aerosol increase penetration to target sites in the middle and superior meatus and sinuses. [0524] In some embodiments, the Epinephrine is delivered with a pulsating aerosol generated through a perforated vibrating membrane. In some embodiments, the pulsating membrane nebulizer is VibrENT (PARI Pharma GmbH). In some embodiments, Epinephrine is delivered with a pulsating spray in combination with breathing techniques. In some embodiments, Epinephrine is delivered with Bi-Directional ™ delivery technology (eg, Bi-Directional ™ Exhalation Delivery Systems (EDS); OptiNose). [0525] In some embodiments, Epinephrine is delivered with a spray bottle. In some embodiments, the atomizer is a portable, battery-operated atomizer intended for nasal administration of medications. In some embodiments, the atomizer atomizes liquids producing a vortex flow in the droplets as they exit the device. Such devices include the ViaNaseTM atomizer (by Kurve Technology Inc., Lynnwood, WA, USA). In some embodiments, the atomizer is a highly pressurized nitrogen gas powered nasal atomizer. [0526] In some embodiments, intranasal epinephrine is administered with a nasal powder device. In some embodiments, the nasal powder device is a nasal powder inhaler, a nasal powder sprayer, or a nasal powder insufflator. Dust sprinklers generally have a compressible compartment that provides a pressure that, when released, creates a cloud of dust particles quite similar to that of a liquid aerosol. Breath-activated inhalers require the user to use their own breath and inhale the powder into the nostril from an ampoule or capsule. Nasal insufflation devices consist of a mouthpiece and a mouthpiece that are fluidly connected. Labor occurs when the subject exhales into the mouthpiece and closes the veil, and the airflow carries the dust particles to the nose through the mouthpiece of the device. [0527] In some embodiments, the nasal powder inhaler is a blister-based powder inhaler. Typically, the blister is pierced prior to use of the nose piece of the device placed in a nostril. The subject closes the other nostril with the finger and inhales the powder into the nose. Representative devices include BiDose ™ / Prohaler ™, and Twin-lizer ™. [0528] Representative nasal powder sprayers include, but are not limited to, UnidoseDP ™, Fit-lizer ™, Monopowder ™, SoluVent ™) [0529] In some embodiments, the nasal powder sprayer is a single dose powder device. based on capsules. In one of these embodiments, the capsule-based single-dose powder device consists of a chamber that cuts the top and bottom of the capsule when inserted. A plastic chamber is compressed by hand, compressed air passes through a one-way valve and capsule during actuation, and dust is emitted. In some embodiments, the nasal powder sprayer consists of an air-filled compartment that is compressed until a pin ruptures a membrane and releases a pressure that emits a column of powder. [0530] In some embodiments, the nasal powder sprayer consists of a plunger that when depressed creates a positive pressure that ruptures a membrane and expels the powder. [0531] In some Embodiments, the nasal powder insufflator requires the subject a blow into one end of the tube while the other end is inserted into the vestibule of the nostril. [0532] In some embodiments, intranasal Epinephrine is administered with a breath-powered Bi-Directional ™ delivery device. A Bi-Directional ™ Nasal Breathing Device uses exhaled breath to deliver medicine to the nose. Bi-Directional ™ breath-driven devices consist of a mouthpiece and sealing mouthpiece with an optimized frusto-conical shape and a comfortable surface that mechanically expands the first part of the nasal valve. The user slides a sealing nosepiece into a nostril until a seal is formed with the flexible soft tissue of the nostril opening, at which point, it mechanically expands the narrow, slit-shaped portion of the triangular nasal valve. The user then exhales through an attached mouthpiece. As you exhale into the mouthpiece against the resistance of the device, the soft palate (or veil) is automatically raised by positive oropharyngeal pressure, isolating the nasal cavity from the rest of the respiratory system. Due to the sealing nose piece, the dynamic pressure that is transferred from the mouth through the device to the nose further expands the slit-shaped nasal passages. This "breath-driven" mechanism allows the release of liquid or dust particles into a stream of air that enters one of the nostrils, passes completely around the nasal septum, and exits through the opposite nose. Triggering drug release in devices using this approach uses manual or automatic flow and / or pressure triggered mechanisms. [0533] Single Dose Devices [0534] In certain embodiments, the device is a single dose device, wherein the pharmaceutical formulation is present in a reservoir, and wherein the therapeutically effective amount of Epinephrine is administered essentially by actuation of the device. [0535] Also provided herein is a single-use pre-primed device adapted for nasal delivery of a pharmaceutical formulation to a patient by actuation. of the device in a nostril of the patient, having a single reservoir comprising approximately 100 pL of a pharmaceutical formulation as described herein. In certain embodiments, the device is operable with one hand. [0536] In certain Embodiments, the delivery time is less than about 30 seconds. In certain Embodiments, the delivery time is less than about 25 seconds. In certain embodiments, the delivery time is less than about 20 seconds. In certain embodiments, the delivery time is less than about 15 seconds. In certain embodiments, the 90% confidence interval for the dose delivered by actuation is ± about 2%. In certain embodiments, the 95% confidence interval for the dose delivered per actuation is ± about 2.5%. [0537] In certain embodiments, after nasal administration of the formulation to the patient, less than about 20%, less than about 15%, less than about 10%, or less than about 5%, of the formulation exits the nasal cavity through drainage into the nasopharynx or externally, as provided above. [0538] In certain embodiments, said formulation is chemically stable on storage for about twelve months at about 25 ° C and about 60% relative humidity and / or about six months at about 40 ° C and about 75% relative humidity. [0539] Os D D devices Osis [0540] In certain embodiments, said device is a two-dose device, wherein a first volume of said formulation is present in a first reservoir and a second volume of said formulation is present in a second reservoir, and in which said amount is administered. therapeutically effective essentially by a first actuation of said device in a first nostril of said patient and a second actuation of said device in a second nostril of said patient. [0541] In certain embodiments, said first volume and said second volume combined equals no more than about 380 pL. [0542] In certain embodiments, approximately 100 pL of said first volume of said formulation is delivered by said first actuation. [0543] In certain embodiments, approximately 100 pL of said second volume of said formulation is delivered by said second actuation. [0544] In certain embodiments, said dosing device is operable with one hand. [0545] In certain Embodiments, the delivery time is less than about 30 seconds. In certain Embodiments, the delivery time is less than about 25 seconds. In certain embodiments, the delivery time is less than about 20 seconds. In certain embodiments, the delivery time is less than about 15 seconds. In certain embodiments, the 90% confidence interval for the dose administered by actuation is ± approximately 2%. In certain embodiments, the 95% confidence interval for the delivered dose per actuation is ± about 2.5%. [0546] In certain embodiments, after nasal administration of the formulation to the patient, less than about 20%, less than about 15%, less than about 10%, or less than about 5%, the formulation exits the nasal cavity through the drain. towards the nasopharynx or externally. [0547] Embodiments are also provided wherein any of the above Embodiments can be combined with any of these Embodiments, as long as the combination is not mutually exclusive. [0548] indications [0549] Also provided are formulations and devices for use in treating adrenergic receptor mediated conditions, one or more symptoms thereof, and methods of treating such conditions which comprise administering the formulations and using the devices described herein. [0550] In certain embodiments, the condition is (1) acute hypersensitivity treatment, such as a type 1 hypersensitivity reaction (eg, such as an anaphylactoid reaction (systemic allergic reaction) to foods, drugs, animal sera, insect stings and stings, and other allergens, see a cotination), (2) treatment of acute asthmatic attacks and relief of uncontrolled bronchospasm by inhalation or subcutaneous administration of other solutions of the medicine, (3) treatment and prophylaxis of cardiac arrest and / or attacks of transient atrioventricular (AV) heart block with syncope crises (Stokes-Adams syndrome), (4) an increase in mean arterial pressure in adult patients with hypotension associated with septic shock, (5) for induction and maintenance of mydriasis during surgery intraocular. [0551] In certain embodiments, the type 1 hypersensitivity reaction (systemic allergic reaction) is selected from allergic asthma, allergic conjunctivitis, allergic rhinitis (hay fever), anaphylaxis, angioedema, urticaria (urticaria), eosinophilia, allergy to antibiotics (eg, a penicillin or cephalosporin), and a food allergy (for example, a peanut or shellfish). [0552] In certain embodiments, the type 1 hypersensitivity reaction (systemic allergic reaction) is anaphylaxis. [0553] Symptoms of anaphylaxis include hives, generalized itching, nasal congestion, wheezing, shortness of breath, cough, cyanosis, lightheadedness, dizziness, confusion, difficulty speaking, rapid pulse, palpitations, nausea and vomiting, abdominal pain or cramps, redness of the skin or inflammation, nasal flaring and intercostal retractions. [0554] In certain embodiments, the symptom of type 1 hypersensitivity reaction (systemic allergic reaction) is chosen from generalized urticaria (urticatria), itching (pruritus), redness, swelling (angioedema) of the affected tissues, a burning sensation the skin (common in people with angi iodem a), h swelling of the tongue or throat, symptoms of breathing such as shortness of breath, drowsiness, or shortness of breath from stridor, spasm or of the coronary artery, myocardial infarction, dysrhythmia, or cardiac arrest (those with underlying coronary diseases have a higher risk of cardiac effects), tachycardia, bradycardia, Bezold-Jarisch reflex. [0555] In certain realizations, the type 1 hypersensitivity reaction (systemic allergic reaction) is caused by stinging insects (for example, order H and m eneoptera, including bees, wasps, hornets, wasps yellow and fire ants), stinging insects (for example, triatoms, mosquitoes), allergen immunotherapy, limes, medications, substances for diagnostic tests (for e jem plo, radiological contrast media) and other allergens, as well as idiopathic anaphylaxis or exercise-induced anaphylaxis. [0556] [00425] In certain embodiments, cardiac arrest is cardiac arrest outside of the hospital. [0557] [00426] It is also possible to carry out the realizations where the previous Realizations can be combined with one or more of these Realizations, provided that the combination is not mutually exclusive. [0558] EXAMPLES [0560] The following examples are provided for illustrative purposes only and do not limit the scope of the Claims provided in this document. [0561] Example 1. Formulations Of Epinephrine For Clinical Use [0562] A representative procedure for the preparation of formulations for clinical use is described. The Formulation Excipient Solution (FES) can be prepared in advance (up to 7 days) and stored at room temperature. Epinephrine Stock Solution (ESS) should be made fresh within 72 hours after dosing, protected from light and excessive oxidation, and stored at 2-8 ° C for up to 2 hours before use. A mixture of equal volumes of sterile filtered FES and ESS will result in a solution of Epinephrine, dodecylmaltoside (DDM), EDTA, benzalkonium chloride (BZK) in saline for use in clinical monitoring protocols. [0563] A 200 ml batch of formulation excipient solution (FES) is prepared by weighing 0.80 g (0.75-0.85 g) of EDTA into a 200 ml volumetric flask and dissolving it in ~ 150 ml of sterile saline; with a weight of 1.00 g (0.95-1.05 g) of Intravail® DDM, quantitatively transferring an EDTA solution, and mixing until dissolved (the solution must be clear and colorless); if necessary, using a light warming aid solution (40-60 ° C), then cooling to room temperature once dissolved; adding the desired amount of a BZK solution (or adding BZK as a solid) and adding an Intravail® / EDTA mixture; adding the appropriate amount of 1N HCl to achieve a pH of 4 (eg, approximately 20 ml), and diluting QS one volume with sterile saline, and stirring until the mixture is uniform. The pH of the FES solution can be measured and recorded. [0564] Epinephrine Stock Solution (ESS) 10 mg / ml must be freshly prepared, protected from light (e.g. with aluminum foil, use of brown lights, etc.), and used within 72 hours to dosage. Formulate a 100 ml batch of 10 mg / ml final product: make sure the 100 ml volumetric flask is wrapped in aluminum foil before adding the FES Solution; Add 50 ml of FES solution to each of the two 100 ml flasks wrapped in aluminum foil (50 ml per flask); weigh and add 1.0 g (0.95-1.05 g) of Epinephrine (E4250 Sigma Aldrich) to each of the two 100 mL flasks; mix each until smooth; measure the pH of each flask and record. [0565] Final dosage formulations (FDFs) are prepared by filling appropriate sprayers capable of delivering 100 gl per spray with appropriate amounts of ESS (e.g., about 5.0 ml of ESS for Aptar multi-dose spray devices or about 125 gL of ESS for spray devices. single dose spray). [0566] Representative formulations of Epinephrine for clinical use are presented in Table 2. [0567] Table 3, and Table 4. [0568] Table 2. Representative Formulations Of Epinephrine For Clinical Use [0570] Table 3. Representative Formulations Of Epinephrine For Clinical Use [0572] Table 4. Representative Formulations Of Epinephrine For Clinical Use [0573] [0575] [0578] Example 2: Clinical Protocols [0579] The following clinical protocols were carried out, or can be carried out, in healthy human volunteers to evaluate the safety, optimal dosage, and pharmacokinetics of intranasal epinephrine. [0580] Example 2A: First Clinical Study [0581] Objective. The main objective of this study was to evaluate the comparative bioavailability of Epinephrine after intranasal administration and intramuscular administration as intramuscular epinephrine administered by autoinjector in healthy fasting volunteers. A secondary objective was to evaluate the safety and tolerability of intranasal (IN) epinephrine in healthy volunteers. [0582] Study Design. A Phase 1, single-dose, two-treatment, randomized, open-label crossover study was conducted, consisting of a screening period, base period, and an open-label treatment period. In the evaluation period, subjects underwent evaluation within 21 days prior to entry into the open-label treatment phase of the study. In the base period, within 24 hours after dosing, initial evaluations were taken; In some cases, the assessment and base visits may be combined if all assessments are done within 24 hours of administration. [0583] In the Open-Label Treatment Period, twelve (12) eligible subjects were randomized after an overnight fast and received single doses of 0.3 mg intranasal Epinephrine and intramuscular Epinephrine administered by autoinjector. Blood samples were collected for 360 minutes after dosing. Treatments were separated by a minimum 24 hour washout period. Safety assessments were performed on each study day and subjects released after discharge assessments on day 1. Subjects were followed for 6 hours after administration of the last dose of study drug. [0584] Plasma samples from all subjects who completed two study periods were analyzed. Blood samples were collected for the measurement of plasma concentrations of Epinephrine, norEpinephrine and diHydroxyphenylglycol (DHPG) (metabolite) before (0. predose) and to 2. 4, 6, 8, 10. 12.5, 15, 20, 25, 45 , 60, 90, 120, 150, 180, 240 and 360 minutes after dosing. Actual blood collection times may vary as follows: 1) ± 1 minutes for samples from 2 to 20 minutes, 2) ± 2 minutes for samples samples from 25 to 90 minutes, and 3) ± 5 minutes for samples from 120 to 360 minutes. Actual sampling times were recorded. [0585] Study of Drugs and Administration. Each formulation of intranasal Epinephrine with a dose of 100 pL IN contained, in addition to 0.3 mg of Epinephrine, 0.25% (w / v) of dodecylmaltoside (0.25 mg), 0.04% (w / v) of benzalkonium chloride (BZK) (0.04 mg), ethylenediaminetetra acetic acid (EDTA) in 10 mM acetate buffer pH 4.0. Intramuscular Epinephrine administered by autoinjector delivered 0.3 mg of Epinephrine by intramuscular injection. [0586] Subjects were fasted prior to an IN or IM epinephrine administration. Each 100 pL aerosol was delivered into the left nostril via a commercially available multiple dose nasal spray device sold by Aptar Pharma. Device priming (5 times activation) was performed in a priming bell or box within 30 minutes prior to subject dosing. [0587] Participants. The study included healthy adult male volunteers (up to 12) between the ages of 18 and 55, inclusive, who gave their written informed consent. Other inclusion criteria include: body weight greater than 50 kg; mass index between 18 and 28 kg / (height in m) 2 (BMI), inclusive; no medical history of hypertension and cardiovascular disease; blood pressure and heart rate within normal range at detection and baseline; no clinically significant abnormal findings in medical history, physical examination, electrocardiogram (QTcF <450 ms), or clinical laboratory results during screening; and agree to remain homebound until the end of the study and to be willing to comply with all required study procedures. [0588] Exclusion criteria included: clinically significant gastrointestinal, renal, hepatic, neurological, hematological, endocrine, oncological, respiratory, immunological, psychiatric history, cardiovascular disease, severe seasonal or non-seasonal allergies, nasal polyps, no nasal perforations, or any abnormality nasal passage that may interfere with the administration of the nasal spray, or any other condition that, in the opinion of the Principal Investigator, endangers the safety of the subject or affects the validity of the study results; smoked within 6 months prior to a screening test; significant traumatic injury, major surgery, or open biopsy within 30 days of a study examination; history of allergic or adverse reactions to Epinephrine or any comparable or similar product; an abnormal diet (such as one that severely restricts specific staple food groups [for example, ketogenic diet], limits calories [for example, fast], and / or requires the use of daily supplements as a substitute for foods normally eaten with meals), during the four (4) weeks prior to the study; donation of blood or plasma within 30 days of the first dose of study drug; participation in a clinical trial within 30 days prior to the first dose of study drug (acceptable non-interventional trial); inadequate or difficult venous access that could compromise the quality or timing of PK samples; positive blood test for HIV, hepatitis B surface antigen (HbSAg), or hepatitis C, or positive urine test for alcohol (a saliva base test may be used), drug abuse, or cotinine. [0589] Additionally, during the study, subjects were not allowed to: take over-the-counter products, including vitamins and supplements, for the seven (7) days prior to the study; to use any prescription drug within 14 days prior to the first dose of study drug during the study unless approved by the principal investigator and medical monitor; a use of oral and / or nasal decongestants within 14 days prior to the first dose of study drug during the study; smoking or using tobacco products for six (6) months prior to the first dose of study drug and for the duration of the study; or participate in strenuous exercise during the study confinement period. [0590] Security. Adverse events were collected and reviewed to assess the safety and tolerability of intranasal epinephrine (IN). Other safety measures included measuring vital signs. Objective evaluations of nasal irritation after each study drug administration were evaluated using a 6-point score. (0 ® 5). Scoring was performed by a trained observer based on an assessment of the nasal mucosa prior to dosing (base) and at 30 (± 5 min) minutes, and 1 (± 10 min), 2 (± 15 min), 4 ( ± 30 min), and 6 (± 30 min) hours after the dose. Irritation was evaluated by evaluating the degree of mucosal inflammation and bleeding. Subjects were asked to report any incidents of bleeding or swelling between the actual assessment time points. [0591] An unrestricted visual analog scale (VAS) consisting of a 10 cm (100 mm) horizontal straight line was used to assess acute pain after each administration of the intranasal intranasal (IN) epinephrine drug product. The extremes of the scale were defined as extreme limits of pain sensation: 0 = no pain, 10 = extreme pain. Subjects were asked to mark a point on the scale that best described their intensity of pain and discomfort just before a dose (base) and at 15 (± 2 min) and 30 (± 5 min) minutes, and 1 ( ± 10 min) hour after the dose. The location of the mark at each time point was measured and noted as the reported score. [0592] Pharmacokinetic Analysis. The pharmacokinetic parameters for Epinephrine, norEpinephrine and DHPG will be calculated using non-compartmental analysis: maximum plasma concentrations (Cmax), time at Cmax (W), area under the curve were calculated once the final time with a concentration equal to or greater than the lower limit of quantification [AUC ( 0 -t)] and an infinite [AUC (inf)], constant elimination rate (Az) and half-life (ty2), and, only for Epinephrine, removal (CL / F) and volume of distribution (Vz / F) uncorrected for bioavailability (F). [0593] Non-GCP pharmacokinetic analysis was performed using WinNonlin version 7.0. The lower limit of bioanalytical quantification was 20 pg / ml. The plasma concentration designated BLQ received a value of 20 pg / ml. Pharmacokinetic parameters. Cmax, AUC ( 0 -t), and AUC (inf) for Epinephrine, norEpinephrine, and DHPG were compared between treatments using an analysis of variance (ANOVA) model with treatment, period, sequence, and subject within sequence as the variables. classification using the natural logarithms of the data. The corrected base Cmax was calculated from the corrected pre-dose Cmax concentration. The AUCo-t corrected base was calculated from the predose concentration AUCo-t - PreDose corrected x tlast. Confidence intervals (90%) were constructed for the geometric mean relationships, intranasal to intramuscular epinephrine for the three parameters using the logarithmically transformed data and the two-sided t-test procedure. Point estimates and confidence limits will be displayed on the original scale. Comparability between intranasal (IN) Epinephrine and intramuscular Epinephrine was evaluated from the geometric relationships of the means and the 90% confidence intervals for the three parameters. [0594] Results. The mean plasma concentration of Epinephrine from IN administration remained significantly cotinational than that of Epinephrine from intramuscular epinephrine administered by autoinjector throughout the study, as shown in Figure 3. Intranasal administration of Epinephrine using the above intranasal Epinephrine formulation resulted in significantly lower exposure (Cmax and AUC 0 -O of parent compound Epinephrine compared to intramuscular epinephrine administered by autoinjector, as a cotinuation shown in Table 5. No related adverse events were reported. pH was between 3 and 4. [0595] Table 5. Intramuscular and Intranasal Administration of Epinephrine [0600] Example 2B: Second Clinical Study [0601] Objective. The primary objectives of this study were to determine the optimal dose of an Intranasal Epinephrine (IN-Epi) formulation to be used in a study of, and in that study an evaluation, the comparative bioavailability of Epinephrine after intranasal administration and intramuscular administration by injection (EpiPen®) injection in healthy fasting volunteers. A secondary objective was to evaluate the safety and tolerability of the intranasal epinephrine formulation in healthy volunteers. [0602] Study design. A Phase 1 dose escalation followed by 12 open-label, randomized, single-dose, two-treatment, two-period, crossover studies was performed as follows. [0603] A dose escalation was performed in three subjects to determine the optimal dose of Epinephrine. In the Evaluation Period, subjects underwent an evaluation within 28 days prior to study entry. Three (3) subjects were subsequently enrolled and received IN-Epi doses of 0.5 mg, 1.0 mg, and 2.0 mg of Epinephrine by IN administration (formulated at pH 5.5 to 6.0) after an overnight fast. Blood samples were collected for 360 minutes after dosing. Treatments were separated by a minimum 24 hour washout period. [0604] The comparative bioavailability of the intranasal formulation of an intramuscular injection was then evaluated in twelve subjects in an open-label, randomized, single-dose, two-treatment, two-period, crossover study consisting of a screening period, base period, and a open treatment period. In the Evaluation Period, subjects underwent an evaluation within 28 days prior to study entry. In the open-label treatment period, twelve (12) eligible subjects were randomly assigned to a 1.0 mg dose of IN-Epi or a 0.3 mg dose of Epinephrine by IM (EpiPen®) administration after an overnight fast and receive doses. unique. Blood samples are collected for 360 minutes after dosing. Treatments are separated by a minimum 24-hour washout period. [0605] Safety assessments were conducted on each day of the study and subjects could be released after discharge assessment. Subjects were followed for 6 hours after administration of the last dose of study drug. [0606] The study was conducted in part as described above. For all parts of the study, the following procedures were performed as follows. [0607] Study of Drugs and Administration. Each formulation of Epinephrine with a dose of 100 pL IN contained, in addition to 0.5 mg, 1.0 mg, or 2.0 mg (5 mg / mL, 10 mg / mL, or 20 mg / mL) of IN-Epi, 0.25% (p / v) dodecylmaltoside (0.25 mg), 0.04% (w / v) of benzalkonium chloride (BZK) (0.04mg), and ethylenediaminetetra acetic acid (EDTA) in saline 0.9% (w / v), at pH 4.5 ( 3.5 to 5.0). The commercially available EpiPen® delivers 0.3 mg of Epinephrine by intramuscular injection. Subjects were fasted prior to an IN or IM epinephrine administration. Each 100 pL aerosol was delivered into the left nostril via a commercially available multiple dose nasal spray device sold by Aptar Pharma. The Device priming (activation 5 times) was performed in a priming bell or box within 30 minutes prior to subject dosing. [0608] Participants. A total of fifteen (15) men were enrolled in the study. Plasma samples from all subjects who completed the study were analyzed. Blood samples were collected for the measurement of plasma concentrations of Epinephrine before (0. predose) and at 2. 4, 6, 8, 10. 12.5, 15, 20, 25, 45, 60. 90, 120, 150. 180, 240 and 360 minutes after dosing. Actual blood collection times may vary as follows: 1) ± 1 minutes for 2 to 20 minute samples, 2) ± 2 minutes for 25 to 90 minute samples, and 3) ± 5 minutes for samples from 120 to 360 minutes. [0609] Inclusion criteria. Participants: they were male, between 18 and 30 years old, inclusive; gave your informed consent in writing; had a body weight of more than 50 kg and a mass index between 18 and 28 kg / m2, inclusive; had no family / medical history of hypertension and cardiovascular disease in the past 10 years; you have blood pressure within the normal range (ie, <140/90 mmHg) on screening; had no clinically significant abnormal findings on medical history, physical examination, electrocardiogram (QTcF <450 ms), or clinical laboratory results during screening; and agreed to remain homebound during appropriate study times and willing to comply with all required study procedures. [0610] Exclusion criteria. Exclusion criteria included a history of clinically significant gastrointestinal, kidney, liver, neurologic, hematologic, endocrine, oncologic, pulmonary, immunologic, psychiatric, or cardiovascular disease, severe or non-seasonal seasonal allergies, nasal polyps, or any nasal passage abnormality that could interfere with nasal spray administration, or any other condition that, in the opinion of the Principal Investigator, would endanger the safety of the subject or impact the validity of the study results; had smoked within the 6 months prior to a screening test; significant traumatic injury, major surgery, or open biopsy within 30 days of a study examination; history of allergic or adverse reactions to Epinephrine or any comparable or similar product; had been on an abnormal diet (such as one that severely restricts specific staple food groups [for example, ketogenic diet], limits calories [for example, fast], and / or required the use of daily supplements as a substitute for foods typically eaten at meals), during the four (4) weeks prior to the study; donated blood or plasma within 30 days of the first dose of study drug; participation in a clinical trial within 30 days prior to the first dose of study drug; inadequate or difficult venous access that may compromise the quality or timing of PK samples; positive blood test for HIV, hepatitis B surface antigen (HbSAg), or hepatitis C, or a positive urine test for alcohol, drugs of abuse, or cotinine. [0611] Additionally, during the study, subjects were not allowed to: take over-the-counter products, including vitamins and supplements, for the seven (7) days prior to the study; to use any prescription drug within 14 days prior to the first dose of study drug during the study unless approved by the principal investigator and medical monitor; a use of oral and / or nasal decongestants within 14 days prior to the first dose of study drug during the study; smoking or using tobacco products for six (6) months prior to the first dose of study drug and for the duration of the study; or participate in strenuous exercise during the study confinement period. [0612] Security. Adverse events were collected and will be reviewed to assess the safety and tolerability of IN-Epi. Other safety measures will include measurements of vital signs. Objective evaluations of nasal irritation were assessed after each study drug administration using a 6 point score (0 ® 5). Scoring was performed by a medically trained observer based on an assessment of the nasal mucosa prior to dosing (base) and at 30 (± 5 min) minutes, and 1 (± 10 min), 2 (± 15 min), 4 (± 30 min), and 6 (± 30 min) hours after the dose. Irritation was evaluated by evaluating the degree of mucosal inflammation and bleeding. Subjects were also asked to report any incidents of bleeding or swelling between the actual test time points. An unrestricted visual analog scale (VAS) consisting of a 10 cm (100 mm) horizontal straight line was used to assess acute pain after each IN-Epi drug administration. The extremes of the scale were defined as extreme limits of pain sensation: 0 = no pain, 10 = extreme pain. Subjects were asked to mark a point on the scale that best describes their intensity of pain and discomfort just before a dose (base) and at 15 (± 2 min) and 30 (± 5 min) minutes, and 1 ( ± 10 min) hour after the dose. The location of the mark at each time point was measured and noted as the reported score. [0613] Pharmacokinetic analysis. The pharmacokinetic parameters for epinephrine were calculated by non-compartmental analysis: maximum plasma concentration (Cmax), time to Cmax (tmax), area under the curve once the final time with a concentration equal to or greater than the lower limit of quantification [AUC ( 0 -t)] and an [AUC (inf)] infinite, constant elimination rate (Az) and half-life (ty2), and, only for Epinephrine, clearance (CL / F) and volume of distribution (Vz / F ) uncorrected for bioavailability (F). [0614] Pharmacokinetic parameters Cmax, AUC ( 0 -t), and AUC (inf) for Epinephrine were compared between treatments using an analysis of variance (ANOVA) model with treatment, period, sequence, and subject within sequence as the classification variables using the natural logarithms of the data. Confidence intervals (90%) were constructed for the Geometric Mean relationships, IN-Epi-to-EpiPen®, of the three parameters using the logarithmically transformed data and the two-sided t-test procedure. Point estimates and confidence limits were displayed on the original scale. The comparability between IN and IM Epinephrine was evaluated from the Geometric Mean relationships and the 90% confidence intervals for the three parameters. [0615] Results. Results for the dose escalation portion of the study are listed in Table 6 and Figures 4, 5, 6, and 7. Intranasal formulations of Epinephrine formulated as described above at doses of 0.5, 1.0. and 2.0 mg in saline at pH 4.0 (3.5-5.0 acceptable) were administered to three subjects. Table 6 below provides the pharmacokinetic parameters of the media for the three doses. Table 6. Pharmacokinetic Parameters of the Media for Three Dose Intranasal Epinephrine [0617] [0620] As can be seen, Tmax was lower, and Cmax and AUC higher, for all doses of intranasal Epinephrine than in the previous study. These trends were more marked with increasing doses. In particular, the 1.0 and 2.0 mg formulations exhibited a tmax lower than intramuscular epinephrine administered by autoinjector, as used in the previous study, and a higher Cmax. The AUC for all intranasal formulations was higher than for intramuscular epinephrine administered by autoinjector for all doses. Figures 4 and 5 show the time versus concentration curves of the media for intranasal formulations of Epinephrine of 0.5, 1.0 and 2.0 mg. Figures 6 and 7 duplicate the data in Figures 4 and 5, but overlap it on the Epinephrine autoinjector data from Study 2A and illustrate the pharmacokinetic differences between, for example, intranasal doses of 1.0 and 2.0 mg of Epinephrine and the intramuscular epinephrine autoinjector. These figures also provide a relevant contrast to Figure 3, where intranasal Epinephrine was formed in acetate buffer at pH 3-4. [0621] Results from the dose escalation portion of the study show, in contrast to previous studies, that Epinephrine can be formulated to achieve bioavailability. significant. At certain doses, the pharmacokinetics of intranasal epinephrine thus formulated appears superior to intramuscular epinephrine administered by autoinjector, achieving a rapid absorption rate similar to IM injection in the first 20 minutes. [0622] The results for the part of the study comparing the bioavailability of IN in an IM injection are listed in Tables 7-9c and Figures 8 and 9. Intranasal epinephrine formulated as described above at a dose of 1.0 mg in saline solution at about pH 4.0 it was administered to twelve subjects; Twelve others were administered intramuscular epinephrine administered by autoinjector (0.3 mg) in the thigh. Table 7 below shows the Mean PK parameters for IN and IM Epinephrine formulated as described above. [0624] Table 7. Mean PK Parameters for IN and IM Epinephrine [0629] Figures 8 and 9 also demonstrate that the plasma time versus concentration curve for 1.0 mg IN Epinephrine is very similar to that for 0.3 mg IM Epinephrine (EpiPen®) administered to the thigh. [0630] Table 8 below shows partial data for Cmax and AUC comparing the IM and IN pathways. The intranasal ratio is given as a reference percentage for AUCs. The data below demonstrates that 1.0 mg intranasal Epinephrine can be formulated to be very similar to or better than an intramuscular injection of 0.3 mg Epinephrine. [0631] Table 8. Comparison Of Key Pharmacokinetic Parameters Between Intranasal And Intramuscular Administration --- Relationship Defined As Intranasal / Intramuscular With A Confidence Interval Of 90%. [0636] Tables 9a - 9c below show the comparisons of 9a) the Median of W, 9b) the distribution values of w, and 9c) the percentage of W subjects meeting the indicated condition between intranasal and intramuscular Epinephrine. [0637] Table 9a. Distribution of Resulting Tmax Values After Intranasal and Intramuscular Administration [0642] Table 9b. Tmax Values Listed in Ascending Order After Intranasal and Intramuscular Administration [0647] Table 9c: Percentage of Subjects With tmax Meeting the Indicated Condition After Intranasal and Intramuscular Administration [0652] IN-Epi appeared to be safe and well tolerated, and demonstrated that PK equivalences and in some respects (eg, Cmax) parameters are better than the Epinephrine autoinjector. [0653] Furthermore, throughout the study, no significant PK differences were observed between 1.0 mg of Epinephrine and 0.3 mg of IM Epinephrine. [0655] Other Realizations [0656] [00475] Embodiments can also be made wherein any previous Embodiment can be combined with one or more of these Embodiments, as long as the combination is not mutually exclusive. Also provided herein are uses in treating indications of one or more symptoms thereof as disclosed herein, and uses in the manufacture of medicaments for treating indications of one or more symptoms thereof as disclosed herein. , equivalences in scope to any previously disclosed Embodiment, or any combination thereof that is not mutually exclusive. The methods and uses may employ any of the devices described herein, or any combination thereof that is not mutually exclusive, or any of the pharmaceutical formulations disclosed herein, or any combination thereof that is not mutually exclusive. [0657] [00476] Although the present invention has been described with reference to specific details of certain embodiments thereof in the above Examples, modifications and variations will be understood to be encompassed within the spirit and scope of the invention.
权利要求:
Claims (89) [1] 1. A nasal spray pharmaceutical formulation comprising between about 0.40 mg and about 2.4 mg of Epinephrine, or a salt thereof, in a single dose of the nasal spray pharmaceutical formulation. [2] 2. The formulation of claim 1. wherein intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides a concentration of Epinephrine in plasma that is effective for the treatment of an acute hypersensitivity reaction. [3] The formulation of claim 1 or claim 2, wherein the nasal spray pharmaceutical formulation is an aqueous solution, aqueous suspension, aqueous emulsion, non-aqueous solution, non-aqueous suspensions, non-aqueous emulsion, or dry powder. [4] The formulation of any of claims 1-3, wherein intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides injection-like intramuscular (IM) pharmacokinetics when the IM injection is dosed. in the lateral thigh, or subcutaneous absorption (SC) or similar between. [5] The formulation of any one of claims 1-3, wherein intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides subcutaneous absorption (SC) and the SC pharmacokinetic profile has a Cmax of at the least 100 pg / mL and AUC 0-240 min 150 hr * pg / mL. [6] 6. The formulation of any one of claims 1-3, wherein intranasal administration of a single dose of the pharmaceutical nasal spray formulation to a subject provides intramuscular (IM) absorption similar to injection. [7] The formulation of any one of claims 1-3, wherein the intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides one or more of the following pharmacokinetic characteristics: • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.3 mg yields; • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.3 mg yields; • a mean W of less than 45 minutes; Y • an absorption similar to IM injection under optimal dosing conditions in the thigh. A single dose intranasal nasal spray pharmaceutical formulation to a subject provides one or more of the following pharmacokinetic characteristics: • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.15 mg yield; • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.15 mg yields; • a mean W of less than 45 minutes; Y [8] • An absorption similar to IM injection under optimal dosing conditions in the thigh. [9] The formulation of any one of claims 1-3, wherein intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides one or more of the following pharmacokinetic characteristics: • both a mean AUC 0-20 min and AUC 0 -t are the least 80% of the AUC 0-20 and AUC 0 min -t than intramuscular injection of 0.5 mg yield; • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax of what an intramuscular injection of 0.5 mg yields; • a mean W of less than 45 minutes; Y • An absorption similar to IM injection under optimal dosing conditions in the thigh. [10] The formulation of any one of claims 1-9, wherein a single dose of the nasal spray pharmaceutical formulation comprises between about 0.5 mg and about 2.0 mg of Epinephrine, or a salt thereof. [11] The formulation of any one of claims 1-9, wherein a single dose of the nasal spray pharmaceutical formulation comprises about 0.5 mg and about 1.5 mg of Epinephrine, or a salt thereof. [12] The formulation of any one of claims 1-9, wherein a single dose of the pharmaceutical nasal spray formulation comprises about 0.5 mg and about 0.7 mg of Epinephrine, or a salt thereof. [13] The formulation of any one of claims 1-9, wherein a single dose of the nasal spray pharmaceutical formulation comprises about 1.0 mg of Epinephrine, or a salt thereof. [14] The formulation of any of claims 1-9, wherein a single dose of the nasal spray pharmaceutical formulation comprises about 1.3 mg and about 1.5 mg of Epinephrine, or a salt thereof. [15] 15. The formulation of any of claims 1-14, wherein the nasal spray pharmaceutical formulation comprises one or more isotonicities; stabilizing agents; preservatives taste masking agents; viscosity modifiers; buffer antioxidants and pH adjusting agents; wherein the pH of the nasal spray pharmaceutical formulation is between about 2.0 and about 6.0 [16] 16. The formulation of any of claims 1-15, wherein the nasal spray pharmaceutical formulation has a pH between about 3.0 and about 5.0. [17] 17. The formulation of any of claims 1-15, wherein the nasal spray pharmaceutical formulation has a pH of about 4.0. [18] 18. The formulation of any one of claims 15-17, wherein the nasal spray pharmaceutical formulation comprises pH adjusting agents. [19] 19. The formulation of claim 18, wherein the pH adjusting agent is an acid, a base, a buffer, or a combination thereof. [20] 20. The formulation of claim 19, wherein: the acid is adipic acid, ammonium chloride, citric acid, acetic acid, hydrochloric acid, lactic acid, phosphoric acid, propionic acid, sulfuric acid or tartaric acid; the base is sodium hydroxide, sodium citrate, sodium bicarbonate, sodium carbonate; Y the buffer is a phosphate buffer, acetate buffer, or citrate buffer. [21] 21. The formulation of claim 15, wherein the nasal spray pharmaceutical formulation comprises between about 0.5 and about 1.1 molar equivalents of acid in each mole of Epinephrine. [22] 22. The formulation of claim 21, wherein the acid is hydrochloric acid. [23] 23. The formulation of any of claims 15-22, wherein the formulation comprises one or more absorption enhancers selected from alcohol, aprotinin, benzalkonium chloride, benzyl alcohol, capric acid, ceramides, cetylpyridinium chloride, chitosan, cyclodextrins, deoxycholic acid, decanoyl, dimethylsulfoxide, glycerol monooleate, glycofurol, glycofurol, glycosylated sphingosines, glycyrrhetinic acids, 2-Hydroxypropyl-p-cyclodextrin, laureth-9, lauric acid, lauroyl carnitine, lysophosphatidylcholine68, polyphosphatidylcholine68, polyphosphatidylcholine68 L-arginine, polyoxyethylene-9-lauryl ether, isopropyl myristate, isopropyl palmitate, lanolin, light mineral oil, linoleic acid, menthol, myristic acid, myristyl alcohol, oleic acid or a salt thereof, oleyl alcohol, palmitic acid, polysorbate 20, polysorbate 80, propylene glycol, polyoxyethylene alkyl ethers, polyoxylglycerides, pyrrolidone, quillaia saponin, salicylic acid, sodium salt, ps itosterol pD-glucoside, sodium lauryl sulfate, sucrose cocoate, taurocholic acid, taurodeoxycholic acid, taurodihydrofusidic acid, thymol, [24] 24. The formulation of any of claims 15-22, wherein the formulation comprises one or more absorption enhancers selected from dodecyl malttoside, benzalkonium chloride, oleic acid, or sales thereof, polysorbate 20, polysorbate 80 and lauryl sulfate of sodium. [25] 25. The formulation of any of claims 15-22, wherein the one or more absorption enhancers are: about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside; or about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride; or about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; or a combination of about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof. [26] 26. The formulation of any of claims 15-22, wherein the one or more absorption enhancers are: about 0.005% (w / v) to about 0.08% (w / v) of benzalkonium chloride; or about 0.01% (w / v) to about 0.06% (w / v) of benzalkonium chloride; or about 0.01% (w / v) to about 0.04% (w / v) of benzalkonium chloride; wherein benzalkonium chloride is the only absorption enhancing agent in the formulation or is present in the formulation with one or more additional absorption enhancing agents. [27] 27. The formulation of any of claims 15-26, wherein the nasal spray pharmaceutical formulation comprises a stabilizing agent. [28] 28. The formulation of claim 27, wherein the stabilizing agent is ethylenediaminetetra acetic acid (EDTA) or a salt thereof. [29] 29. The formulation of claim 28, wherein the EDTA is disodium EDTA. [30] 30. The formulation of claim 29, wherein the formulation comprises from about 0.001% (w / v) to about 1% (w / v) of disodium EDTA. [31] includes a conservator. [32] 32. The formulation of claim 31, wherein the preservative is benzalkonium chloride. [33] 33. The formulation of any of claims 15-32, wherein the nasal spray pharmaceutical formulation comprises an isotonicity agent. [34] 34. The formulation of claim 33, wherein the isotonicity agent is dextrose, glycerin, mannitol, potassium chloride, or sodium chloride [35] 35. The formulation of claim 33, wherein the isotonicity agent is sodium chloride. [36] 36. A method of treating an adrenergic receptor-mediated condition comprising intranasal administration of the formulation of any of claims 1-35. [37] 37. The method of claim 36, wherein the condition is selected from a Type-1 hypersensitivity reaction (systemic allergic reaction), an acute asthmatic attack, cardiac arrest, and Stokes-Adams syndrome. [38] 38. The method of claim 37, wherein the condition is a type 1 hypersensitivity reaction (systemic allergic reaction). [39] 39. The method of claim 38, wherein the Type 1 hypersensitivity reaction is selected from allergic asthma, allergic conjunctivitis, allergic rhinitis, anaphylaxis, angioedema, urticaria, eosinophilia, drug allergy, and food allergy. [40] 40. The method of claim 39, wherein the drug allergy is an allergy to antibiotics. [41] 41. A nasal spray pharmaceutical formulation comprising Epinephrine or a salt thereof, wherein intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides one or more of the following pharmacokinetic characteristics: • both mean AUC 0-20 min and mean AUC 0 -t that are the least 80% of the mean AUC 0-20 min and mean AUC 0 -t than intramuscular injection provides; • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax that an intramuscular injection provides; • a mean W of less than 45 minutes; Y • an intramuscular (IM) type absorption under optimal dosing conditions in the thigh. [42] 42. A nasal spray pharmaceutical formulation comprising Epinephrine or a salt thereof, wherein intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides one or more of the following • both mean AUC 0 - 20 min and mean AUC 0 -t which are at least 80% of the mean AUC 0 - 20 min and mean AUC 0 -t than what an intramuscular injection of 0.15mg provides; • a mean Cmax that is at least 80% of the Cmax and no more than 150% of the Cmax than what an intramuscular injection of 0.15mg provides; • a mean W of less than 45 minutes; Y • an intramuscular (IM) type absorption under optimal dosing conditions in the thigh. [43] 43. A nasal spray pharmaceutical formulation comprising Epinephrine or a salt thereof, wherein intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides one or more of the following pharmacokinetic characteristics: • both mean AUC 0-20 min and mean AUC 0 -t that are the least 80% of the mean AUC 0-20 min and mean AUC 0 -t a 0.5 mg intramuscular injection provides; • a mean Cmax that is at least 80% of the Cmax and not more than 150% of the Cmax that a 0.5 mg intramuscular injection provides; • a mean W of less than 45 minutes; Y • an intramuscular (IM) type absorption under optimal dosing conditions in the thigh. [44] 44. The formulation of any one of claims 41-43, wherein intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides injection-type intramuscular (IM) absorption. [45] 45. The formulation of any of claims 41-44, wherein intranasal administration of a single dose of the nasal spray pharmaceutical formulation to a subject provides a plasma concentration of Epinephrine that is effective for treating a hypersensitivity reaction. sharp. [46] 46. The formulation of any of claims 41-45, wherein a single dose of the nasal spray pharmaceutical formulation comprises between about 0.5 mg and about 2.0 mg of Epinephrine, or a salt thereof. [47] 47. The formulation of any of claims 41-45, wherein a single dose of the nasal spray pharmaceutical formulation comprises between about 0.5 mg and about 1.5 mg of Epinephrine, or a salt thereof. [48] 48. The formulation of any of claims 42, 44, and 45, wherein a single dose of the nasal spray pharmaceutical formulation comprises between about 0.5 mg and about 0.7 mg of Epinephrine, or a salt thereof. [49] A single nasal spray pharmaceutical formulation comprises about 1.0 mg of Epinephrine, or a salt thereof. [50] 50. The formulation of any of claims 43-45, wherein a single dose of the nasal spray pharmaceutical formulation comprises between about 1.3 mg and about 1.5 mg of Epinephrine, or a salt thereof. [51] 51. The formulation of any of claims 41-50. wherein the formulation is an aqueous solution, aqueous suspension, aqueous emulsion, non-aqueous solution, non-aqueous suspension, non-aqueous emulsion, pressure metered dose inhalers, or dry powder. [52] 52. 52. The formulation of any of claims 41-51, wherein the pharmaceutical nasal spray formulation comprises one or more absorption enhancing agents; and optionally one or more agents selected from isotonicity agents; stabilizing agents; preservatives taste masking agents; viscosity modifiers; buffer antioxidants and pH adjusting agents; wherein the pH of the nasal spray pharmaceutical formulation is between about 2.0 and about 6.0. [53] 53. 53. The formulation of any of claims 41-52, wherein the pharmaceutical nasal spray formulation has a pH between about 3.0 and about 5.0. [54] 54. 54. The formulation of any of claims 41-52, wherein the nasal spray pharmaceutical formulation has a pH of about 4.0. [55] 55. 55. The formulation of any one of claims 52-54, wherein the nasal spray pharmaceutical formulation comprises pH adjusting agents. [56] 56. The formulation of claim 55, wherein the pH adjusting agent is an acid, a base, a buffer, or a combination thereof. [57] 57. The formulation of claim 56, wherein: the acid is adipic acid, ammonium chloride, citric acid, acetic acid, hydrochloric acid, lactic acid, phosphoric acid, propionic acid, sulfuric acid or tartaric acid; the base is sodium hydroxide, sodium citrate, sodium bicarbonate, sodium carbonate; Y the buffer is a phosphate buffer, acetate buffer, or citrate buffer. [58] 58. The formulation of any of claims 52-57, wherein the formulation comprises one or more absorption enhancers selected from alcohol, aprotinin, benzalkonium chloride, benzyl alcohol, capric acid, ceramides, cetylpyridinium chloride, chitosan, cyclodextrins, deoxycholic acid, decanoyl, dimethyl sulfoxide, glyceryl monooleate, glycofurol, glycosylated sphingosines, glycyrrhetinic acid, 2 menthol, poloxamer 407 or F68, poly-L-arginine, polyoxyethylene-9-lauryl ether, isopropyl myristate, isopropyl palmitate, lanolin, light mineral oil, linoleic acid, menthol, myristic acid, myristyl alcohol, oleic acid or a salt thereof, oleyl alcohol, palmitic acid, polysorbate 20, polysorbate 80, propylene glycol, polyoxyethylene alkyl ethers, polyoxylglycerides, pyrrolidone, quillaia saponin, salicylic acid, sodium salt, p-sitosterol pD-glucoside, sodium lauryl cocoate, sucrose , taurocholic acid, taurodeoxycholic acid, taurodihydrofusidic acid, thymol, tricaprylin, triolein, and alkylsaccharides. [59] The formulation of any of claims 52-57, wherein the formulation comprises one or more absorption enhancers selected from dodecyl maltoside, benzalkonium chloride, oleic acid, or sales thereof, polysorbate 20, polysorbate 80, and lauryl sulfate. sodium. [60] The formulation of any of claims 52-57, wherein the one or more absorption enhancers are: about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside; or about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride; or about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; or a combination of about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof. [61] The formulation of any one of claims 52-57, wherein the one or more absorption enhancers are: about 0.005% (w / v) to about 0.08% (w / v) of benzalkonium chloride; or about 0.01% (w / v) to about 0.06% (w / v) of benzalkonium chloride; or about 0.01% (w / v) to about 0.04% (w / v) of benzalkonium chloride; wherein benzalkonium chloride is the only absorption enhancing agent in the formulation or is present in the formulation with one or more absorption enhancing agents [62] 62. The formulation of any of claims 52-61, wherein the nasal spray pharmaceutical formulation comprises a stabilizing agent. [63] 63. The formulation of claim 62, wherein the stabilizing agent is ethylenediaminetetra acetic acid (EDTA) or a salt thereof. [64] 64. The formulation of claim 63, wherein the EDTA is disodium EDTA. [65] 65. The formulation of claim 64, wherein the formulation comprises from about 0.001% (w / v) to about 1% (w / v) of disodium EDTA. [66] 66. The formulation of any of claims 52-65, wherein the formulation comprises a preservative. [67] 67. The formulation of claim 66, wherein the preservative is benzalkonium chloride. [68] 68. The formulation of any of claims 52-67, wherein the nasal spray pharmaceutical formulation comprises an isotonicity agent. [69] 69. The formulation of claim 68, wherein the isotonicity agent is dextrose, glycerin, mannitol, potassium chloride, or sodium chloride [70] 70. The formulation of claim 68, wherein the isotonicity agent is sodium chloride. [71] 71. A method of treating an adrenergic receptor-mediated condition comprising intranasal administration of the formulation of any of claims 41-70. [72] 72. The method of claim 71, wherein the condition is selected from a Type-1 hypersensitivity reaction (systemic allergic reaction), an acute asthmatic attack, cardiac arrest, and Stokes-Adams syndrome. [73] 73. The method of claim 72, wherein the condition is a type 1 hypersensitivity reaction (systemic allergic reaction). [74] 74. The method of claim 73, wherein the Type 1 hypersensitivity reaction is selected from allergic asthma, allergic conjunctivitis, allergic rhinitis, anaphylaxis, angioedema, urticaria, eosinophilia, drug allergy, and food allergy. [75] 75. The method of claim 74, wherein the drug allergy is an antibiotic allergy. [76] 76. A method of treating anaphylaxis which comprises intranasal administration of a nasal pharmaceutical formulation of Epinephrine, or a salt thereof, in an amount less than about 2.0 mg. [77] 77. The method of claim 76, wherein the nasal pharmaceutical formulation comprises between about 0.5 mg and about 1.5 mg of epinephrine, or a salt thereof. [78] 78. The method of claim 76, wherein the nasal pharmaceutical formulation a salt of it. [79] 79. The method of claim 76, wherein the nasal pharmaceutical formulation comprises about 1.0 mg of Epinephrine, or a salt thereof. [80] 80. The method of claim 76, wherein the nasal pharmaceutical formulation comprises between about 1.3 mg and about 1.5 mg of Epinephrine, or a salt thereof. [81] 81. The method of any one of claims 76-80. wherein the intranasal formulation comprises: one or more absorption enhancing agents; an isotonicity agent; a stabilizing agent; an optional antioxidant; an optional buffering agent; a conservative; Y optional pH adjusting agents. [82] 82. The method of claim 81, wherein the absorption enhancing agents are selected from: dodecyl maltoside; benzalkonium chloride; oleic acid, or salts thereof; a combination of dodecyl maltoside and benzalkonium chloride; a combination of dodecyl maltose and oleic acid, or salts thereof; and a combination of benzalkonium chloride and oleic acid, or salts thereof. [83] 83. The method of claim 81, wherein the absorption enhancing agents are selected from: about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside; about 0.001 (w / v) to about 1% (w / v) benzalkonium chloride; about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; a combination of about 0.005% (w / v) to about 2.5% (w / v) of dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride; a combination of about 0.005% (w / v) to about 2.5% (w / v) dodecyl maltoside and about 0.001 (w / v) to about 1% (w / v) oleic acid, or salts thereof; or a combination of about 0.001 (w / v) to about 1% (w / v) of benzalkonium chloride and about 0.001 (w / v) to about 1% (w / v) of [84] 84. The method of any of claims 81-83, wherein the formulation comprises: about 0.005% (w / v) to about 0.08% (w / v) of benzalkonium chloride; about 0.01% (w / v) to about 0.06% (w / v) of benzalkonium chloride; or about 0.01% (w / v) to about 0.04% (w / v) of benzalkonium chloride; wherein benzalkonium chloride is the only absorption enhancing agent in the formulation or is present in the formulation with one or more absorption enhancing agents. [85] 85. The method of any of claims 81-84, wherein: the isotonicity agent is sodium chloride. [86] 86. The method of any of claims 81-85, wherein: the stabilizing agent is EDTA. [87] 87. The method of any of claims 81-85, wherein: the stabilizing agent is EDTA in an amount from about 0.001% (w / v) to about 1% (w / v). [88] 88. The method of any of claims 81-87, wherein: the preservative is benzalkonium chloride. [89] 89. The method of any of claims 81-87, wherein: the preservative is benzalkonium chloride in an amount from about 0.001% (w / v) to about 1% (w / v).
类似技术:
公开号 | 公开日 | 专利标题 US11191838B2|2021-12-07|Intranasal epinephrine formulations and methods for the treatment of disease ES2626134T3|2017-07-24|Bepotastin compositions US10806709B2|2020-10-20|Intranasal formulation of epinephrine for the treatment of anaphylaxis RU2421209C2|2011-06-20|Pharmaceutical compositions containing cyclosporine ES2792682T3|2020-11-11|Methods for the treatment of lung diseases with mast cell stabilizers JP5863641B2|2016-02-16|Pharmaceutical formulation for stabilizing concentrated mast cells ES2617255T3|2017-06-16|Compositions comprising azelastine and methods of use thereof ES2891729T3|2022-01-31|Fixed dose stable pharmaceutical composition comprising mometasone and olopatadine for nasal administration ES2281021T3|2007-09-16|LIQUID FORMULATION CONTAINING TOBRAMYCIN. ES2791775A2|2020-11-05|Intranasal epinephrine formulations and methods for the treatment of disease KR102375232B1|2022-03-16|Intranasal epinephrine preparations and methods of treatment of diseases CA3124202A1|2020-06-25|Compositions, devices, and methods for the treatment of overdose and reward-based disorders
同族专利:
公开号 | 公开日 MA50106A|2021-04-21| CA3088989A1|2019-08-15| SG11202007527XA|2020-09-29| KR20200121818A|2020-10-26| ES2791775R1|2021-07-02| EP3678649A4|2020-10-14| BR112020015646A2|2020-12-08| GB202016960D0|2020-12-09| IL276485D0|2020-09-30| EP3678649A1|2020-07-15| AU2021254650A1|2021-11-18| DE112019000683T5|2020-10-15| AU2019217643B2|2021-10-28| GB2583051B|2020-12-02| GB2583051A|2020-10-14| AU2019217643A1|2020-08-13| GB2587934B|2022-01-19| GB2587934A|2021-04-14| GB202012086D0|2020-09-16| GB202117505D0|2022-01-19| CN110505873A|2019-11-26|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 US5369095A|1990-02-14|1994-11-29|Alcon Laboratories, Inc.|Compositions and method comprising substituted glycosides as mucus membrane permeation enhancers| WO2015095389A1|2013-12-18|2015-06-25|Aegis Therapeutics, Llc|Compositions for drug administration| US9895444B2|2004-08-25|2018-02-20|Aegis Therapeutics, Llc|Compositions for drug administration| WO2011139838A2|2010-04-28|2011-11-10|Zelos Therapeutics, Inc.|Intranasal formulations| US9789071B2|2012-06-27|2017-10-17|G2B Pharma, Inc.|Intranasal formulation of epinephrine for the treatment of anaphylaxis| WO2014127015A1|2013-02-12|2014-08-21|Ys Pharm Tech|Epinephrine formulations for medicinal products| US10039710B2|2015-09-18|2018-08-07|Insys Development Company, Inc.|Epinephrine spray formulations|
法律状态:
2020-11-05| BA2A| Patent application published|Ref document number: 2791775 Country of ref document: ES Kind code of ref document: A2 Effective date: 20201105 | 2021-07-02| EC2A| Search report published|Ref document number: 2791775 Country of ref document: ES Kind code of ref document: R1 Effective date: 20210625 |
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US15/890,131|US10576156B2|2004-08-25|2018-02-06|Compositions for drug administration| US201862685049P| true| 2018-06-14|2018-06-14| US201862734550P| true| 2018-09-21|2018-09-21| US201862784057P| true| 2018-12-21|2018-12-21| PCT/US2019/016918|WO2019157099A1|2018-02-06|2019-02-06|Intranasal epinephrine formulations and methods for the treatment of disease| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|