VASOPRESSIN FORMULATIONS FOR USE IN TREATMENT OF HYPOTENSION

Abstract
Provided herein are peptide formulations comprising polymers as stabilizing agents. The peptide formulations can be more stable for prolonged periods of time at temperatures higher than room temperature when formulated with the polymers. The polymers used in the present invention can decrease the degradation of the constituent peptides of the peptide formulations.
Description
BACKGROUND

Vasopressin is a potent endogenous hormone, responsible for maintaining plasma osmolality and volume in most mammals. Vasopressin can be used clinically in the treatment of sepsis and cardiac conditions, and in the elevation of patient's suffering from low blood pressure. Current formulations of vasopressin suffer from poor long-term stability.


INCORPORATION BY REFERENCE

Each patent, publication, and non-patent literature cited in the application is hereby incorporated by reference in its entirety as if each was incorporated by reference individually.


SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Feb. 16, 2018, is named 47956702314_SL.txt and is 5266 bytes in size.


SUMMARY OF THE INVENTION

In some embodiments, the invention provides a pharmaceutical composition comprising, in a unit dosage form: a) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin, or a pharmaceutically-acceptable salt thereof; and b) a polymeric pharmaceutically-acceptable excipient in an amount that is from about 1% to about 10% by mass of the unit dosage form or the pharmaceutically-acceptable salt thereof, wherein the unit dosage form exhibits from about 5% to about 10% less degradation of the vasopressin or the pharmaceutically-acceptable salt thereof after storage for about 1 week at about 60° C. than does a corresponding unit dosage form, wherein the corresponding unit dosage form consists essentially of: A) vasopressin, or a pharmaceutically-acceptable salt thereof; and B) a buffer having acidic pH.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 is a chromatogram of a diluent used in vasopressin assay.



FIG. 2 is a chromatogram of a sensitivity solution used in a vasopressin assay.



FIG. 3 is a chromatogram of an impurity marker solution used in a vasopressin assay.



FIG. 4 is a zoomed-in depiction of the chromatogram in FIG. 3.



FIG. 5 is a chromatogram of a vasopressin standard solution.



FIG. 6 is a chromatogram of a sample vasopressin preparation.



FIG. 7 is a UV spectrum of a vasopressin sample.



FIG. 8 is a UV spectrum of a vasopressin standard.



FIG. 9 plots vasopressin stability across a range of pH as determined experimentally.



FIG. 10 illustrates the effects of various stabilizers on vasopressin stability.



FIG. 11 plots vasopressin stability across a range of pH at 25° C.



FIG. 12 plots vasopressin impurities across a range of pH at 25° C.



FIG. 13 plots vasopressin stability across a range of pH at 40° C.



FIG. 14 plots vasopressin impurities across a range of pH at 40° C.



FIG. 15 illustrates vasopressin impurities across a range of pH at 25° C.



FIG. 16 illustrates vasopressin impurities across a range of pH at 40° C.



FIG. 17 illustrates the effect of pH on vasopressin at 25° C.



FIG. 18 illustrates the effect of pH on vasopressin at 40° C.



FIG. 19 depicts the % LC of vasopressin formulations stored for 15 months at 25° C.



FIG. 20 below shows a diagram of a cap to be used on a vial described herein.



FIG. 21 depicts a stopper to be used in a vial described herein.



FIG. 22 shows a vial for use in storing a 10 mL vasopressin formulation described herein.



FIG. 23 shows the vasopressin assay results (represented as % label claim) of studies performed at 25° C. with upright storage.



FIG. 24 shows the vasopressin assay results (represented as % label claim) of the studies performed at 25° C. with inverted storage.



FIG. 25 shows the amount of Gly9-AVP when a 10 mL vasopressin formulation was stored at 25° C. in an upright position.



FIG. 26 shows the amount of Gly9-AVP when a 10 mL vasopressin formulation was stored at 25° C. in an inverted position.



FIG. 27 shows the amount of Glu4-AVP when a 10 mL vasopressin formulation was stored at 25° C. in an upright position.



FIG. 28 shows the amount of Glu4-AVP when a 10 mL vasopressin formulation was stored at 25° C. in an inverted position.



FIG. 29 shows the total impurities when a 10 mL vasopressin formulation was stored at 25° C. in an upright position.



FIG. 30 shows the total impurities when a 10 mL vasopressin formulation was stored at 25° C. in an inverted position.





DETAILED DESCRIPTION
Vasopressin and Peptides of the Invention.

Vasopressin, a peptide hormone, acts to regulate water retention in the body and is a neurotransmitter that controls circadian rhythm, thermoregulation, and adrenocorticotrophic hormone (ACTH) release. Vasopressin is synthesized as a pro-hormone in neurosecretory cells of the hypothalamus, and is subsequently transported to the pituitary gland for storage. Vasopressin is released upon detection of hyperosmolality in the plasma, which can be due to dehydration of the body. Upon release, vasopressin increases the permeability of collecting ducts in the kidney to reduce renal excretion of water. The decrease in renal excretion of water leads to an increase in water retention of the body and an increase in blood volume. At higher concentrations, vasopressin raises blood pressure by inducing vasoconstriction.


Vasopressin acts through various receptors in the body including, for example, the V1, V2, V3, and oxytocin-type (OTR) receptors. The V1 receptors occur on vascular smooth muscle cells, and the major effect of vasopressin action on the V1 receptor is the induction of vasoconstriction via an increase of intracellular calcium. V2 receptors occur on the collecting ducts and the distal tubule of the kidney. V2 receptors play a role in detection of plasma volume and osmolality. V3 receptors occur in the pituitary gland and can cause ACTH release upon vasopressin binding. OTRs can be found on the myometrium and vascular smooth muscle. Engagement of OTRs via vasopressin leads to an increase of intracellular calcium and vasoconstriction.


Vasopressin is a nonapeptide, illustrated below (SEQ ID NO. 1):




embedded image


At neutral to acidic pH, the two basic groups of vasopressin, the N-terminal cysteine, and the arginine at position eight, are protonated, and can each carry an acetate counterion. The amide groups of the N-terminal glycine, the glutamine at position four, and the asparagine at position five, are susceptible to modification when stored as clinical formulations, such as unit dosage forms. The glycine, glutamine, and asparagine residues can undergo deamidation to yield the parent carboxylic acid and several degradation products as detailed in EXAMPLE 1 and TABLE 1 below.


Deamidation is a peptide modification during which an amide group is removed from an amino acid, and can be associated with protein degradation, apoptosis, and other regulatory functions within the cell. Deamidation of asparagine and glutamine residues can occur in vitro and in vivo, and can lead to perturbation of the structure and function of the affected proteins. The susceptibility to deamidation can depend on primary sequence of the protein, three-dimensional structure of the protein, and solution properties including, for example, pH, temperature, ionic strength, and buffer ions. Deamidation can be catalyzed by acidic conditions. Under physiological conditions, deamidation of asparagine occurs via the formation of a five-membered succinimide ring intermediate by a nucleophilic attack of the nitrogen atom in the following peptide bond on the carbonyl group of the asparagine side chain. Acetylation is a peptide modification whereby an acetyl group is introduced into an amino acid, such as on the N-terminus of the peptide.


Vasopressin can also form dimers in solution and in vivo. The vasopressin dimers can occur through the formation of disulfide bridges that bind a pair of vasopressin monomers together. The dimers can form between two parallel or anti-parallel chains of vasopressin.


Vasopressin and associated degradation products or peptides are listed in TABLE 1 below. All amino acids are L-stereoisomers unless otherwise denoted.











TABLE 1





Name
Sequence
SEQ ID NO.

















Vasopressin (AVP; arginine
CYFQNCPRG-NH2
1


vasopressin)


Gly9-vasopressin (Gly9-AVP)
CYFQNCPRG
2


Asp5-vasopressin
CYFQDCPRG-NH2
3


(Asp5-AVP)


Glu4-vasopressin (Glu4-AVP)
CYFENCPRG-NH2
4


Glu4Gly9-vasopressin
CYFENCPRG
5


(Glu4Gly9-AVP)


AcetylAsp5-vasopressin
Ac-CYFQDCPRG-NH2
6


(AcetylAsp5-AVP)


Acetyl-vasopressin
Ac-CYFQNCPRG-NH2
7


(Acetyl-AVP)


His2-vasopressin (His2-AVP)
CHFQNCPRG-NH2
8


Leu7-vasopressin (Leu7-AVP)
CYFQNCLRG-NH2
9


D-Asn-vasopressin
CYFQ(D-Asn)CPRG-NH2
10


(DAsn-AVP)


D-Cys1-vasopressin
(D-Cys)YFQNCPRG-NH2
11


D-Tyr-vasopressin
C(D-Tyr)FQNCPRG-NH2
12


D-Phe-vasopressin
CY(D-Phe)QNCPRG-NH2
13


D-Gln-vasopressin
CYF(D-Gln)NCPRG-NH2
14


D-Cys6-vasopressin
CYFQN(D-cys)PRG-NH2
15


D-Pro-vasopressin
CYFQNC(D-pro)RG-NH2
16


D-Arg-vasopressin
CYFQNCP(D-Arg)G-NH2
17









Therapeutic Uses.

A formulation of vasopressin can be used to regulate plasma osmolality and volume and conditions related to the same in a subject. Vasopressin can be used to modulate blood pressure in a subject, and can be indicated in a subject who is hypotensive despite treatment with fluid and catecholamines.


Vasopressin can be used in the treatment of, for example, vasodilatory shock, post-cardiotomy shock, sepsis, septic shock, cranial diabetes insipidus, polyuria, nocturia, polydypsia, bleeding disorders, Von Willebrand disease, haemophilia, platelet disorders, cardiac arrest, liver disease, liver failure, hypovolemia, hemorrhage, oesophageal variceal haemorrhage, hypertension, pulmonary hypertension, renal disease, polycystic kidney disease, blood loss, injury, hypotension, meniere disease, uterine myomas, brain injury, mood disorder. Formulations of vasopressin can be administered to a subject undergoing, for example, surgery or hysterectomy.


Plasma osmolality is a measure of the plasma's electrolyte-water balance and relates to blood volume and hydration of a subject. Normal plasma osmolality in a healthy human subject range from about 275 milliosmoles/kg to about 295 milliosmoles/kg. High plasma osmolality levels can be due to, for example, diabetes insipidus, hyperglycemia, uremia, hypernatremia, stroke, and dehydration. Low plasma osmolality can be due to, for example, vasopressin oversecretion, improper functioning of the adrenal gland, lung cancer, hyponatremia, hypothyroidism, and over-consumption of water or other fluids.


Septic shock can develop due to an extensive immune response following infection and can result in low blood pressure. Causes of sepsis can include, for example, gastrointestinal infections, pneumonia, bronchitis, lower respiratory tract infections, kidney infection, urinary tract infections, reproductive system infections, fungal infections, and viral infections. Risk factors for sepsis include, for example, age, prior illness, major surgery, long-term hospitalization, diabetes, intravenous drug use, cancer, use of steroidal medications, and long-term use of antibiotics. The symptoms of sepsis can include, for example, cool arms and legs, pale arms and legs, extreme body temperatures, chills, light-headedness, decreased urination, rapid breathing, edema, confusion, elevated heart rate, high blood sugar, metabolic acidosis, respiratory alkalosis, and low blood pressure.


Vasopressin can also be administered to regulate blood pressure in a subject. Blood pressure is the measure of force of blood pushing against blood vessel walls. Blood pressure is regulated by the nervous and endocrine systems and can be used as an indicator of a subject's health. Chronic high blood pressure is referred to as hypertension, and chronic low blood pressure is referred to as hypotension. Both hypertension and hypotension can be harmful if left untreated.


Blood pressure can vary from minute to minute and can follow the circadian rhythm with a predictable pattern over a 24-hour period. Blood pressure is recorded as a ratio of two numbers: systolic pressure (mm Hg), the numerator, is the pressure in the arteries when the heart contracts, and diastolic pressure (mm Hg), the denominator, is the pressure in the arteries between contractions of the heart. Blood pressure can be affected by, for example, age, weight, height, sex, exercise, emotional state, sleep, digestion, time of day, smoking, alcohol consumption, salt consumption, stress, genetics, use of oral contraceptives, and kidney disease.


Blood pressure for a healthy human adult between the ages of 18-65 can range from about 90/60 to about 120/80. Hypertension can be a blood pressure reading above about 120/80 and can be classified as hypertensive crisis when there is a spike in blood pressure and blood pressure readings reach about 180/110 or higher. Hypertensive crisis can be precipitated by, for example, stroke, myocardial infarction, heart failure, kidney failure, aortic rupture, drug-drug interactions, and eclampsia. Symptoms of hypertensive crisis can include, for example, shortness of breath, angina, back pain, numbness, weakness, dizziness, confusion, change in vision, nausea, and difficulty speaking.


Vasodilatory shock can be characterized by low arterial blood pressure due to decreased systemic vascular resistance. Vasodilatory shock can lead to dangerously low blood pressure levels and can be corrected via administration of catecholamines or vasopressin formulations. Vasodilatory shock can be caused by, for example, sepsis, nitrogen intoxication, carbon monoxide intoxication, hemorrhagic shock, hypovolemia, heart failure, cyanide poisoning, metformin intoxication, and mitochondrial disease.


Post-cardiotomy shock can occur as a complication of cardiac surgery and can be characterized by, for example, inability to wean from cardiopulmonary bypass, poor hemodynamics in the operating room, development of poor hemodynamics post-surgery, and hypotension.


Pharmaceutical Formulations.

Methods for the preparation of compositions comprising the compounds described herein can include formulating the compounds with one or more inert, pharmaceutically-acceptable excipients. Liquid compositions include, for example, solutions in which a compound is dissolved, emulsions comprising a compound, or a solution containing liposomes, micelles, or nanoparticles comprising a compound as disclosed herein. These compositions can also contain minor amounts of nontoxic, auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, and other pharmaceutically-acceptable additives.


Non-limiting examples of dosage forms suitable for use in the disclosure include liquid, elixir, nanosuspension, aqueous or oily suspensions, drops, syrups, and any combination thereof. Non-limiting examples of pharmaceutically-acceptable excipients suitable for use in the disclosure include granulating agents, binding agents, lubricating agents, disintegrating agents, anti-adherents, anti-static agents, surfactants, anti-oxidants, coloring agents, flavoring agents, plasticizers, preservatives, suspending agents, emulsifying agents, plant cellulosic material and spheronization agents, and any combination thereof.


Vasopressin can be formulated as an aqueous formulation or a lyophilized powder, which can be diluted or reconstituted just prior to use. Upon dilution or reconstitution, the vasopressin solution can be refrigerated for long-term stability for about one day. Room temperature incubation or prolonged refrigeration can lead to the generation of degradation products of vasopressin.


In some embodiments, a pharmaceutical composition of the invention can be formulated for long-term storage of vasopressin at room temperature in the presence of a suitable pharmaceutically-acceptable excipient. The pharmaceutically-acceptable excipient can increase the half-life of vasopressin when stored at any temperature, such as room temperature. The presence of the pharmaceutical excipient can decrease the rate of decomposition of vasopressin at any temperature, such as room temperature.


In some embodiments, a pharmaceutical composition has a shelf life of at least about 12 months, at least about 13 months, at least about 14 months, at least about 15 months, at least about 16 months, at least about 17 months, at least about 18 months, at least about 19 months, at least about 20 months, at least about 21 months, at least about 22 months, at least about 23 months, at least about 24 months, at least about 25 months, at least about 26 months, at least about 27 months, at least about 28 months, at least about 29 months, or at least about 30 months. The shelf life can be at any temperature, including, for example, room temperature and refrigeration (i.e., 2-8° C.). As used herein, “shelf life” means the period beginning from manufacture of a formulation beyond which the formulation cannot be expected beyond reasonable doubt to yield the therapeutic outcome approved by a government regulatory agency


In some embodiments, a vasopressin formulation of the invention comprises a pharmaceutically-acceptable excipient, and the vasopressin has a half-life that is at least about 1%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 100%, at least about 150%, at least about 200%, at least about 250%, at least about 300%, at least about 350%, at least about 400%, at least about 450%, at least about 500%, at least about 600%, at least about 700%, at least about 800%, at least about 900%, or at least about 1000% greater than the half-life of vasopressin in a corresponding formulation that lacks the pharmaceutically-acceptable excipient.


In some embodiments, a vasopressin formulation of the invention has a half-life at about 5° C. to about 8° C. that is no more than about 1%, no more than about 5%, no more than about 10%, no more than about 15%, no more than about 20%, no more than about 25%, no more than about 30%, no more than about 35%, no more than about 40%, no more than about 45%, no more than about 50%, no more than about 55%, no more than about 60%, no more than about 65%, no more than about 70%, no more than about 75%, no more than about 80%, no more than about 85%, no more than about 90%, no more than about 95%, no more than about 100%, no more than about 150%, no more than about 200%, no more than about 250%, no more than about 300%, no more than about 350%, no more than about 400%, no more than about 450%, no more than about 500%, no more than about 600%, no more than about 700%, no more than about 800%, no more than about 900%, or no more than about 1000% greater than the half-life of the formulation at another temperature, such as room temperature.


The half-life of the compounds of the invention in a formulation described herein at a specified temperature can be, for example, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 30 hours, about 36 hours, about 42 hours, about 48 hours, about 60 hours, about 3 days, about 4 days, about 5 days, about 6 days, or about one week.


A formulation described herein can be stable for or be stored for, for example, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 30 hours, about 36 hours, about 42 hours, about 48 hours, about 60 hours, about 3 days, about 4 days, about 5 days, about 6 days, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 13 months, about 14 months, about 15 months, about 16 months, about 17 months, about 18 months, about 19 months, about 20 months, about 21 months, about 22 months, about 23 months, about 2 years, about 25 months, about 26 months, about 27 months, about 28 months, about 29 months, about 30 months, about 31 months, about 32 months, about 33 months, about 34 months, about 35 months, or about 3 years prior to administration to a subject.


A unit dosage form described herein can be stable for or be stored for, for example, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 30 hours, about 36 hours, about 42 hours, about 48 hours, about 60 hours, about 3 days, about 4 days, about 5 days, about 6 days, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 13 months, about 14 months, about 15 months, about 16 months, about 17 months, about 18 months, about 19 months, about 20 months, about 21 months, about 22 months, about 23 months, about 2 years, about 25 months, about 26 months, about 27 months, about 28 months, about 29 months, about 30 months, about 31 months, about 32 months, about 33 months, about 34 months, about 35 months, or about 3 years prior to administration to a subject.


A diluted unit dosage form described herein can be stable for or be stored for, for example, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 30 hours, about 36 hours, about 42 hours, about 48 hours, about 60 hours, about 3 days, about 4 days, about 5 days, about 6 days, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 13 months, about 14 months, about 15 months, about 16 months, about 17 months, about 18 months, about 19 months, about 20 months, about 21 months, about 22 months, about 23 months, about 2 years, about 25 months, about 26 months, about 27 months, about 28 months, about 29 months, about 30 months, about 31 months, about 32 months, about 33 months, about 34 months, about 35 months, or about 3 years prior to administration to subject.


The stability of a formulation described herein can be measured after, for example, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 30 hours, about 36 hours, about 42 hours, about 48 hours, about 60 hours, about 3 days, about 4 days, about 5 days, about 6 days, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, about 8 weeks, about 9 weeks, about 10 weeks, about 11 weeks, about 3 months, about 4 months, about 5 months, about 6 months, about 7 months, about 8 months, about 9 months, about 10 months, about 11 months, about 1 year, about 13 months, about 14 months, about 15 months, about 16 months, about 17 months, about 18 months, about 19 months, about 20 months, about 21 months, about 22 months, about 23 months, about 2 years, about 25 months, about 26 months, about 27 months, about 28 months, about 29 months, about 30 months, about 31 months, about 32 months, about 33 months, about 34 months, about 35 months, or about 3 years.


A formulation or unit dosage form described herein can exhibit, for example, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9% about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, about 3%, about 3.1%, about 3.2%, about 3.3%, about 3.4%, about 3.5%, about 3.6%, about 3.7%, about 3.8%, about 3.9%, about 4%, about 4.1%, about 4.2%, about 4.3%, about 4.4%, about 4.5%, about 4.6%, about 4.7%, about 4.8%, about 4.9%, about 5%, about 5.5%, about 6%, about 6.5%, about 7%, about 7.5%, about 8%, about 8.5%, about 9%, about 9.5%, or about 10% degradation over a specified period of time. The degradation of a formulation or a unit dosage form disclosed herein can be assessed after about 24 hours, about 36 hours, about 72 hours, about 96 hours, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, about 8 months, about 10 months, about 1 year, about 13 months, about 14 months, about 15 months, about 16 months, about 17 months, about 18 months, about 19 months, about 20 months, about 21 months, about 22 months, about 23 months, about 2 years, about 25 months, about 26 months, about 27 months, about 28 months, about 29 months, about 30 months, about 31 months, about 32 months, about 33 months, about 34 months, about 35 months, or about 3 years of storage. The degradation of a formulation or a unit dosage form disclosed herein can be assessed at a temperature of, for example, about 0° C., about 1° C., about 2° C., about 3° C., about 4° C., about 5° C., about 6° C., about 7° C., about 8° C., about 9° C., about 10° C., about 11° C., about 12° C., about 13° C., about 14° C., about 15° C., about 16° C., about 17° C., about 18° C., about 19° C., about 20° C., about 21° C., about 22° C., about 23° C., about 24° C., about 25° C., about 26° C., about 27° C., about 28° C., about 29° C., about 30° C., about 31° C., about 32° C., about 33° C., about 34° C., about 35° C., about 36° C., about 37° C., about 38° C., about 39° C., about 40° C., about 41° C., about 42° C., about 43° C., about 44° C., about 45° C., about 46° C., about 47° C., about 48° C., about 49° C., about 50° C., or about 0° C. to about 5° C., about 1° C. to about 6° C., about 2° C. to about 7° C., about 2° C. to about 8° C., about 3° C. to about 8° C., about 4° C. to about 9° C., about 5° C. to about 10° C., about 6° C. to about 11° C., about 7° C. to about 12° C., about 8° C. to about 13° C., about 9° C. to about 14° C., about 10° C. to about 15° C., about 11° C. to about 16° C., about 12° C. to about 17° C., about 13° C. to about 18° C., about 14° C. to about 19° C., about 15° C. to about 20° C., about 16° C. to about 21° C., about 17° C. to about 22° C., about 18° C. to about 23° C., about 19° C. to about 24° C., about 20° C. to about 25° C., about 21° C. to about 26° C., about 22° C. to about 27° C., about 23° C. to about 28° C., about 24° C. to about 29° C., about 25° C. to about 30° C., about 26° C. to about 31° C., about 27° C. to about 32° C., about 28° C. to about 33° C., about 29° C. to about 34° C., about 30° C. to about 35° C., about 31° C. to about 36° C., about 32° C. to about 37° C., about 33° C. to about 38° C., about 34° C. to about 39° C., about 35° C. to about 40° C., about 36° C. to about 41° C., about 37° C. to about 42° C., about 38° C. to about 43° C., about 39° C. to about 44° C., about 40° C. to about 45° C., about 41° C. to about 46° C., about 42° C. to about 47° C., about 43° C. to about 48° C., about 44° C. to about 49° C., about 45° C. to about 50° C.


In some embodiments, a vasopressin formulation of the invention comprises an excipient and the vasopressin has a level of decomposition at a specified temperature that is about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, about 3%, about 3.1%, about 3.2%, about 3.3%, about 3.4%, about 3.5%, about 3.6%, about 3.7%, about 3.8%, about 3.9%, about 4%, about 4.1%, about 4.2%, about 4.3%, about 4.4%, about 4.5%, about 4.6%, about 4.7%, about 4.8%, about 4.9%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 250%, about 300%, about 350%, about 400%, about 450%, about 500%, about 600%, about 700%, about 800%, about 900%, or about 1000% less than the level of decomposition of a formulation of the invention in the absence of the excipient.


Pharmaceutical compositions of the invention can be used, stored, tested, analyzed or assayed at any suitable temperature. Non-limiting examples of temperatures include about 0° C., about 1° C., about 2° C., about 3° C., about 4° C., about 5° C., about 6° C., about 7° C., about 8° C., about 9° C., about 10° C., about 11° C., about 12° C., about 13° C., about 14° C., about 15° C., about 16° C., about 17° C., about 18° C., about 19° C., about 20° C., about 21° C., about 22° C., about 23° C., about 24° C., about 25° C., about 26° C., about 27° C., about 28° C., about 29° C., about 30° C., about 31° C., about 32° C., about 33° C., about 34° C., about 35° C., about 36° C., about 37° C., about 38° C., about 39° C., about 40° C., about 41° C., about 42° C., about 43° C., about 44° C., about 45° C., about 46° C., about 47° C., about 48° C., about 49° C., about 50° C., about 51° C., about 52° C., about 53° C., about 54° C., about 55° C., about 56° C., about 57° C., about 58° C., about 59° C., about 60° C., about 61° C., about 62° C., about 63° C., about 64° C., about 65° C., about 66° C., about 67° C., about 68° C., about 69° C., about 70° C., about 71° C., about 72° C., about 73° C., about 74° C., or about 75° C.


Pharmaceutical compositions of the invention can be used, stored, tested, analyzed or assayed at any suitable temperature. Non-limiting examples of temperatures include from about 0° C. to about 5° C., about 1° C. to about 6° C., about 2° C. to about 7° C., about 2° C. to about 8° C., about 3° C. to about 8° C., about 4° C. to about 9° C., about 5° C. to about 10° C., about 6° C. to about 11° C., about 7° C. to about 12° C., about 8° C. to about 13° C., about 9° C. to about 14° C., about 10° C. to about 15° C., about 11° C. to about 16° C., about 12° C. to about 17° C., about 13° C. to about 18° C., about 14° C. to about 19° C., about 15° C. to about 20° C., about 16° C. to about 21° C., about 17° C. to about 22° C., about 18° C. to about 23° C., about 19° C. to about 24° C., about 20° C. to about 25° C., about 21° C. to about 26° C., about 22° C. to about 27° C., about 23° C. to about 28° C., about 24° C. to about 29° C., about 25° C. to about 30° C., about 26° C. to about 31° C., about 27° C. to about 32° C., about 28° C. to about 33° C., about 29° C. to about 34° C., about 30° C. to about 35° C., about 31° C. to about 36° C., about 32° C. to about 37° C., about 33° C. to about 38° C., about 34° C. to about 39° C., about 35° C. to about 40° C., about 36° C. to about 41° C., about 37° C. to about 42° C., about 38° C. to about 43° C., about 39° C. to about 44° C., about 40° C. to about 45° C., about 41° C. to about 46° C., about 42° C. to about 47° C., about 43° C. to about 48° C., about 44° C. to about 49° C., about 45° C. to about 50° C., about 46° C. to about 51° C., about 47° C. to about 52° C., about 48° C. to about 53° C., about 49° C. to about 54° C., about 50° C. to about 55° C., about 51° C. to about 56° C., about 52° C. to about 57° C., about 53° C. to about 58° C., about 54° C. to about 59° C., about 55° C. to about 60° C., about 56° C. to about 61° C., about 57° C. to about 62° C., about 58° C. to about 63° C., about 59° C. to about 64° C., about 60° C. to about 65° C., about 61° C. to about 66° C., about 62° C. to about 67° C., about 63° C. to about 68° C., about 64° C. to about 69° C., about 65° C. to about 70° C., about 66° C. to about 71° C., about 67° C. to about 72° C., about 68° C. to about 73° C., about 69° C. to about 74° C., about 70° C. to about 74° C., about 71° C. to about 76° C., about 72° C. to about 77° C., about 73° C. to about 78° C., about 74° C. to about 79° C., or about 75° C. to about 80° C.


Pharmaceutical compositions of the invention can be used, stored, tested, analyzed or assayed at room temperature. The room temperature can be, for example, about 20.0° C., about 20.1° C., about 20.2° C., about 20.3° C., about 20.4° C., about 20.5° C., about 20.6° C., about 20.7° C., about 20.8° C., about 20.9° C., about 21.0° C., about 21.1° C., about 21.2° C., about 21.3° C., about 21.4° C., about 21.5° C., about 21.6° C., about 21.7° C., about 21.8° C., about 21.9° C., about 22.0° C., about 22.1° C., about 22.2° C., about 22.3° C., about 22.4° C., about 22.5° C., about 22.6° C., about 22.7° C., about 22.8° C., about 22.9° C., about 23.0° C., about 23.1° C., about 23.2° C., about 23.3° C., about 23.4° C., about 23.5° C., about 23.6° C., about 23.7° C., about 23.8° C., about 23.9° C., about 24.0° C., about 24.1° C., about 24.2° C., about 24.3° C., about 24.4° C., about 24.5° C., about 24.6° C., about 24.7° C., about 24.8° C., about 24.9° C., or about 25.0° C.


A pharmaceutical composition of the disclosed can be supplied, stored, or delivered in a vial or tube that is, for example, about 0.5 mL, about 1 mL, about 2 mL, about 3 mL, about 4 mL, about 5 mL, about 6 mL, about 7 mL, about 8 mL, about 9 mL, about 10 mL, about 11 mL, about 12 mL, about 13 mL, about 14 mL, about 15 mL, about 16 mL, about 17 mL, about 18 mL, about 19 mL, or about 20 mL in volume.


A pharmaceutical composition of the disclosure can be a combination of any pharmaceutical compounds described herein with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. The pharmaceutical composition facilitates administration of the compound to an organism. Pharmaceutical compositions can be administered in therapeutically-effective amounts, for example, intravenous, subcutaneous, intramuscular, transdermal, or parenteral administration.


Pharmaceutical preparations can be formulated for intravenous administration. The pharmaceutical compositions can be in a form suitable for parenteral injection as a sterile suspension, solution, or emulsion in oily or aqueous vehicles, and can contain formulation agents such as suspending, stabilizing, and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Suspensions of the active compounds can be prepared as oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. The suspension can also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient can be in powder form for constitution with a suitable vehicle, for example, sterile pyrogen-free water, before use.


Comparison Formulations.

A pharmaceutical composition described herein can be analyzed by comparison to a reference formulation. A reference formulation can be generated from any combination of compounds, peptides, excipients, diluents, carriers, and solvents disclosed herein. Any compound, peptide, excipient, diluent, carrier, or solvent used to generate the reference formulation can be present in any percentage, ratio, or amount, for example, those disclosed herein. The reference formulation can comprise, consist essentially of, or consist of any combination of any of the foregoing.


A non-limiting example of a comparison formulation comprises, consists essentially of, or consists of: an amount, such as about 20 Units or about 0.04 mg, of vasopressin or a pharmaceutically-acceptable salt thereof, an amount, such as about 5 mg, of chlorobutanol (for example, hydrous), an amount, such as about 0.22 mg, of acetic acid or a pharmaceutically-acceptable salt thereof or a quantity sufficient to bring pH to about 3.4 to about 3.6, and water as needed. Another non-limiting example of a comparison formulation comprises, consists essentially of, or consists of: vasopressin or a pharmaceutically-acceptable salt thereof, chlorobutanol, acetic acid, and a solvent such as water. Another non-limiting example of a comparison formulation comprises, consists essentially of, or consists of: vasopressin or a pharmaceutically-acceptable salt thereof, chlorobutanol, and a solvent such as water. Another non-limiting example of a comparison formulation comprises, consists essentially of, or consists of: vasopressin or a pharmaceutically-acceptable salt thereof, acetic acid, and a solvent such as water. Another non-limiting example of a comparison formulation comprises, consists essentially of, or consists of: vasopressin or a pharmaceutically-acceptable salt thereof and a solvent such as water. Another non-limiting example of a comparison formulation comprises, consists essentially of, or consists of: vasopressin or a pharmaceutically-acceptable salt thereof and a buffer having acidic pH, such as pH 3.5 or any buffer or pH described herein.


Methods.

Any formulation described herein can be diluted prior to administration to a subject. Diluents that can be used in a method of the invention include, for example, compound sodium lactate solution, 6% dextran, 10% dextran, 5% dextrose, 20% fructose, Ringer's solution, 5% saline, 1.39% sodium bicarbonate, 1.72% sodium lactate, or water. Upon dilution, any diluted formulation disclosed herein can be stored for, for example, about 24 hours, about 36 hours, about 72 hours, about 96 hours, about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 2 months, about 3 months, about 4 months, about 5 months, about 6 months, about 8 months, about 10 months, about 1 year, about 13 months, about 14 months, about 15 months, about 16 months, about 17 months, about 18 months, about 19 months, about 20 months, about 21 months, about 22 months, about 23 months, about 2 years, about 25 months, about 26 months, about 27 months, about 28 months, about 29 months, about 30 months, about 31 months, about 32 months, about 33 months, about 34 months, about 35 months, or about 3 years of storage. Upon dilution, any diluted formulation disclosed herein can be stored at, for example, about 0° C., about 1° C., about 2° C., about 3° C., about 4° C., about 5° C., about 6° C., about 7° C., about 8° C., about 9° C., about 10° C., about 11° C., about 12° C., about 13° C., about 14° C., about 15° C., about 16° C., about 17° C., about 18° C., about 19° C., about 20° C., about 21° C., about 22° C., about 23° C., about 24° C., about 25° C., about 26° C., about 27° C., about 28° C., about 29° C., about 30° C., about 31° C., about 32° C., about 33° C., about 34° C., about 35° C., about 36° C., about 37° C., about 38° C., about 39° C., about 40° C., about 41° C., about 42° C., about 43° C., about 44° C., about 45° C., about 46° C., about 47° C., about 48° C., about 49° C., about 50° C., or about 0° C. to about 5° C., about 1° C. to about 6° C., about 2° C. to about 7° C., about 2° C. to about 8° C., about 3° C. to about 8° C., about 4° C. to about 9° C., about 5° C. to about 10° C., about 6° C. to about 11° C., about 7° C. to about 12° C., about 8° C. to about 13° C., about 9° C. to about 14° C., about 10° C. to about 15° C., about 11° C. to about 16° C., about 12° C. to about 17° C., about 13° C. to about 18° C., about 14° C. to about 19° C., about 15° C. to about 20° C., about 16° C. to about 21° C., about 17° C. to about 22° C., about 18° C. to about 23° C., about 19° C. to about 24° C., about 20° C. to about 25° C., about 21° C. to about 26° C., about 22° C. to about 27° C., about 23° C. to about 28° C., about 24° C. to about 29° C., about 25° C. to about 30° C., about 26° C. to about 31° C., about 27° C. to about 32° C., about 28° C. to about 33° C., about 29° C. to about 34° C., about 30° C. to about 35° C., about 31° C. to about 36° C., about 32° C. to about 37° C., about 33° C. to about 38° C., about 34° C. to about 39° C., about 35° C. to about 40° C., about 36° C. to about 41° C., about 37° C. to about 42° C., about 38° C. to about 43° C., about 39° C. to about 44° C., about 40° C. to about 45° C., about 41° C. to about 46° C., about 42° C. to about 47° C., about 43° C. to about 48° C., about 44° C. to about 49° C., about 45° C. to about 50° C.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 24 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 48 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 96 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about one week.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about two weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about three weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about six weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about three months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about six months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about one year.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about two years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about three years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 24 hours; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 48 hours; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 96 hours; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 1 week; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 2 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about one week.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 6 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 months; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 6 months; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least one year; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least two years; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least three years; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 24 hours; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 24 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 48 hours; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 48 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 96 hours; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 96 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least one week; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about one week.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 2 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 2 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 months; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 6 months; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 6 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 1 year; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 1 year.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 2 years; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 2 years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 years; c) diluting the unit dosage form in a diluent to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 24 hours; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 48 hours; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 96 hours; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 1 week; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 2 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about one week.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 months; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 6 months; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 1 year; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 2 years; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 years; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 24 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 48 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 96 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 1 week.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 2 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 6 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 1 year.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 2 years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 24 hours; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 24 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 48 hours; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 48 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 96 hours; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 96 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least one week; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 1 week.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 2 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 2 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 4 weeks; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 months; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 6 months; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 6 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least one year; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about one year.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 2 years; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 2 years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising, in a unit dosage form: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) chlorobutanol; iii) acetic acid; and iv) water; b) storing the unit dosage form at 2-8° C., for example, 5° C., for at least 3 years; c) diluting the unit dosage form in 0.9% saline or 5% dextrose in water to provide a concentration from about 0.1 units/mL to about 1 unit/mL (about 0.21 μg/mL to about 2.1 μg/mL) of vasopressin or the pharmaceutically-acceptable salt thereof; and d) administering the diluted unit dosage form to the human by intravenous administration; wherein: the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; and the human is hypotensive, wherein the unit dosage form exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 24 hours; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 48 hours; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 96 hours; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 1 week; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 2 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 3 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 1 week.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 3 months; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 6 months; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 1 year; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 2 years; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 3 years; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 24 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 48 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 96 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 1 week.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about two weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about three weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about four weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 6 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 1 year.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 2 years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 24 hours; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 24 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 48 hours; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C. for about, for example, 5° C., 48 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 96 hours; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 96 hours.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 1 week; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 1 week.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 2 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 2 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 3 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 4 weeks; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 4 weeks.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 3 months; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 6 months; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 6 months.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 1 year; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 1 year.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 2 years; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 2 years.


The present invention provides a method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; ii) acetic acid; and iii) water; and b) storing the pharmaceutical composition at 2-8° C., for example, 5° C., for at least 3 years; and c) intravenously administering the pharmaceutical composition to the human, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.1 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute; wherein the human is hypotensive, wherein the pharmaceutical composition exhibits less than about 5% degradation after storage at 2-8° C., for example, 5° C., for about 3 years.


Dosage Amounts.


In practicing the methods of treatment or use provided herein, therapeutically-effective amounts of the compounds described herein are administered in pharmaceutical compositions to a subject having a disease or condition to be treated. A therapeutically-effective amount can vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compounds used, and other factors. Subjects can be, for example, humans, elderly adults, adults, adolescents, pre-adolescents, children, toddlers, infants, or neonates. A subject can be a patient.


Pharmaceutical compositions of the invention can be formulated in any suitable volume. The formulation volume can be, for example, about 0.1 mL, about 0.2 mL, about 0.3 mL, about 0.4 mL, about 0.5 mL, about 0.6 mL, about 0.7 mL, about 0.8 mL, about 0.9 mL, about 1 mL, about 1.1 mL, about 1.2 mL, about 1.3 mL, about 1.4 mL, about 1.5 mL, about 1.6 mL, about 1.7 mL, about 1.8 mL, about 1.9 mL, about 2 mL, about 2.1 mL, about 2.2 mL, about 2.3 mL, about 2.4 mL, about 2.5 mL, about 2.6 mL, about 2.7 mL, about 2.8 mL, about 2.9 mL, about 3 mL, about 3.1 mL, about 3.2 mL, about 3.3 mL, about 3.4 mL, about 3.5 mL, about 3.6 mL, about 3.7 mL, about 3.8 mL, about 3.9 mL, about 4 mL, about 4.1 mL, about 4.2 mL, about 4.3 mL, about 4.4 mL, about 4.5 mL, about 4.6 mL, about 4.7 mL, about 4.8 mL, about 4.9 mL, about 5 mL, about 5.1 mL, about 5.2 mL, about 5.3 mL, about 5.4 mL, about 5.5 mL, about 5.6 mL, about 5.7 mL, about 5.8 mL, about 5.9 mL, about 6 mL, about 6.1 mL, about 6.2 mL, about 6.3 mL, about 6.4 mL, about 6.5 mL, about 6.6 mL, about 6.7 mL, about 6.8 mL, about 6.9 mL, about 7 mL, about 7.1 mL, about 7.2 mL, about 7.3 mL, about 7.4 mL, about 7.5 mL, about 7.6 mL, about 7.7 mL, about 7.8 mL, about 7.9 mL, about 8 mL, about 8.1 mL, about 8.2 mL, about 8.3 mL, about 8.4 mL, about 8.5 mL, about 8.6 mL, about 8.7 mL, about 8.8 mL, about 8.9 mL, about 9 mL, about 9.1 mL, about 9.2 mL, about 9.3 mL, about 9.4 mL, about 9.5 mL, about 9.6 mL, about 9.7 mL, about 9.8 mL, about 9.9 mL, about 10 mL, about 11 mL, about 12 mL, about 13 mL, about 14 mL, about 15 mL, about 16 mL, about 17 mL, about 18 mL, about 19 mL, or about 20 mL.


A therapeutically-effective amount of a compound described herein can be present in a composition at a concentration of, for example, about 0.1 units/mL, about 0.2 units/mL, about 0.3 units/mL, about 0.4 units/mL, about 0.5 units/mL, about 0.6 units/mL, about 0.7 units/mL, about 0.8 units/mL, about 0.9 units/mL, about 1 unit/mL, about 2 units/mL, about 3 units/mL, about 4 units/mL, about 5 units/mL, about 6 units/mL, about 7 units/mL, about 8 units/mL, about 9 units/mL, about 10 units/mL, about 11 units/mL, about 12 units/mL, about 13 units/mL, about 14 units/mL, about 15 units/mL, about 16 units/mL, about 17 units/mL, about 18 units/mL, about 19 units/mL, about 20 units/mL, about 21 units/mL, about 22 units/mL, about 23 units/mL, about 24 units/mL about 25 units/mL, about 30 units/mL, about 35 units/mL, about 40 units/mL, about 45 units/mL, or about 50 units/mL.


A therapeutically-effective amount of a compound described herein can be present in a composition of the invention at a mass of about, for example, about 0.01 μg, about 0.05 μg, about 0.1 μg, about 0.15 μg, about 0.2 μg, about 0.25 μg, about 0.3 μg, about 0.35 μg, about 0.4 μg, about 0.5 μg, about 0.6 μg, about 0.7 μg, about 0.8 μg, about 0.9 μg, about 1 μg, about 2 μg, about 3 μg, about 4 μg, about 5 μg, about 10 μg, about 15 μg, about 20 μg, about 25 μg, about 30 μg, about 35 μg, about 40 μg, about 45 μg, about 50 μg, about 60 μg, about 70 μg, about 80 μg, about 90 μg, about 100 μg, about 125 μg, about 150 μg, about 175 μg, about 200 μg, about 250 μg, about 300 μg, about 350 μg, about 400 μg, about 450 μg, about 500 μg, about 600 μg, about 700 μg, about 800 μg, about 900 μg, about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, or about 10 mg.


A therapeutically-effective amount of a compound described herein can be present in a composition of the invention at a concentration of, for example, about 0.001 mg/mL, about 0.002 mg/mL, about 0.003 mg/mL, about 0.004 mg/mL, about 0.005 mg/mL, about 0.006 mg/mL, about 0.007 mg/mL, about 0.008 mg/mL, about 0.009 mg/mL, about 0.01 mg/mL, about 0.02 mg/mL, about 0.03 mg/mL, about 0.04 mg/mL, about 0.05 mg/mL, about 0.06 mg/mL, about 0.07 mg/mL, about 0.08 mg/mL, about 0.09 mg/mL, about 0.1 mg/mL, about 0.2 mg/mL, about 0.3 mg/mL, about 0.4 mg/mL, about 0.5 mg/mL, about 0.6 mg/mL, about 0.7 mg/mL, about 0.8 mg/mL, about 0.9 mg/mL, about 1 mg/mL, about 1.5 mg/mL, about 2 mg/mL, about 2.5 mg/mL, about 3 mg/mL, about 3.5 mg/mL, about 4 mg/mL, about 4.5 mg/mL, about 5 mg/mL, about 6 mg/mL, about 7 mg/mL, about 8 mg/mL, about 9 mg/mL, or about 10 mg/mL.


A therapeutically-effective amount of a compound described herein can be present in a composition of the invention at a unit of active agent/unit of active time. Non-limiting examples of therapeutically-effective amounts can be, for example, about 0.01 units/minute, about 0.02 units/minute, about 0.03 units/minute, about 0.04 units/minute, about 0.05 units/minute, about 0.06 units/minute, about 0.07 units/minute, about 0.08 units/minute, about 0.09 units/minute or about 0.1 units/minute.


Pharmaceutical compositions of the invention can be formulated at any suitable pH. The pH can be, for example, about 2, about 2.05, about 2.1, about 2.15, about 2.2, about 2.25, about 2.3, about 2.35, about 2.4, about 2.45, about 2.5, about 2.55, about 2.6, about 2.65, about 2.7, about 2.75, about 2.8, about 2.85, about 2.9, about 2.95, about 3, about 3.05, about 3.1, about 3.15, about 3.2, about 3.25, about 3.3, about 3.35, about 3.4, about 3.45, about 3.5, about 3.55, about 3.6, about 3.65, about 3.7, about 3.75, about 3.8, about 3.85, about 3.9, about 3.95, about 4, about 4.05, about 4.1, about 4.15, about 4.2, about 4.25, about 4.3, about 4.35, about 4.4, about 4.45, about 4.5, about 4.55, about 4.6, about 4.65, about 4.7, about 4.75, about 4.8, about 4.85, about 4.9, about 4.95, or about 5 pH units.


Pharmaceutical compositions of the invention can be formulated at any suitable pH. The pH can be, for example, from about 2 to about 2.2, about 2.05 to about 2.25, about 2.1 to about 2.3, about 2.15 to about 2.35, about 2.2 to about 2.4, about 2.25 to about 2.45, about 2.3 to about 2.5, about 2.35 to about 2.55, about 2.4 to about 2.6, about 2.45 to about 2.65, about 2.5 to about 2.7, about 2.55 to about 2.75, about 2.6 to about 2.8, about 2.65 to about 2.85, about 2.7 to about 2.9, about 2.75 to about 2.95, about 2.8 to about 3, about 2.85 to about 3.05, about 2.9 to about 3.1, about 2.95 to about 3.15, about 3 to about 3.2, about 3.05 to about 3.25, about 3.1 to about 3.3, about 3.15 to about 3.35, about 3.2 to about 3.4, about 3.25 to about 3.45, about 3.3 to about 3.5, about 3.35 to about 3.55, about 3.4 to about 3.6, about 3.45 to about 3.65, about 3.5 to about 3.7, about 3.55 to about 3.75, about 3.6 to about 3.8, about 3.65 to about 3.85, about 3.7 to about 3.9, about 3.7 to about 3.8, about 3.75 to about 3.95, about 3.75 to about 3.8, about 3.8 to about 3.85, about 3.75 to about 3.85, about 3.8 to about 4, about 3.85 to about 4.05, about 3.9 to about 4.1, about 3.95 to about 4.15, about 4 to about 4.2, about 4.05 to about 4.25, about 4.1 to about 4.3, about 4.15 to about 4.35, about 4.2 to about 4.4, about 4.25 to about 4.45, about 4.3 to about 4.5, about 4.35 to about 4.55, about 4.4 to about 4.6, about 4.45 to about 4.65, about 4.5 to about 4.7, about 4.55 to about 4.75, about 4.6 to about 4.8, about 4.65 to about 4.85, about 4.7 to about 4.9, about 4.75 to about 4.95, about 4.8 to about 5, about 4.85 to about 5.05, about 4.9 to about 5.1, about 4.95 to about 5.15, or about 5 to about 5.2 pH units.


In some embodiments, the addition of an excipient can change the viscosity of a pharmaceutical composition of the invention. In some embodiments the use of an excipient can increase or decrease the viscosity of a fluid by at least 0.001 Pascal-second (Pa·s), at least 0.001 Pa·s, at least 0.0009 Pa·s, at least 0.0008 Pa·s, at least 0.0007 Pa·s, at least 0.0006 Pa·s, at least 0.0005 Pa·s, at least 0.0004 Pa·s, at least 0.0003 Pa·s, at least 0.0002 Pa·s, at least 0.0001 Pa·s, at least 0.00005 Pa·s, or at least 0.00001 Pa·s.


In some embodiments, the addition of an excipient to a pharmaceutical composition of the invention can increase or decrease the viscosity of the composition by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99%. In some embodiments, the addition of an excipient to a pharmaceutical composition of the invention can increase or decrease the viscosity of the composition by no greater than 5%, no greater than 10%, no greater than 15%, no greater than 20%, no greater than 25%, no greater than 30%, no greater than 35%, no greater than 40%, no greater than 45%, no greater than 50%, no greater than 55%, no greater than 60%, no greater than 65%, no greater than 70%, no greater than 75%, no greater than 80%, no greater than 85%, no greater than 90%, no greater than 95%, or no greater than 99%.


Any compound herein can be purified. A compound can be at least 1% pure, at least 2% pure, at least 3% pure, at least 4% pure, at least 5% pure, at least 6% pure, at least 7% pure, at least 8% pure, at least 9% pure, at least 10% pure, at least 11% pure, at least 12% pure, at least 13% pure, at least 14% pure, at least 15% pure, at least 16% pure, at least 17% pure, at least 18% pure, at least 19% pure, at least 20% pure, at least 21% pure, at least 22% pure, at least 23% pure, at least 24% pure, at least 25% pure, at least 26% pure, at least 27% pure, at least 28% pure, at least 29% pure, at least 30% pure, at least 31% pure, at least 32% pure, at least 33% pure, at least 34% pure, at least 35% pure, at least 36% pure, at least 37% pure, at least 38% pure, at least 39% pure, at least 40% pure, at least 41% pure, at least 42% pure, at least 43% pure, at least 44% pure, at least 45% pure, at least 46% pure, at least 47% pure, at least 48% pure, at least 49% pure, at least 50% pure, at least 51% pure, at least 52% pure, at least 53% pure, at least 54% pure, at least 55% pure, at least 56% pure, at least 57% pure, at least 58% pure, at least 59% pure, at least 60% pure, at least 61% pure, at least 62% pure, at least 63% pure, at least 64% pure, at least 65% pure, at least 66% pure, at least 67% pure, at least 68% pure, at least 69% pure, at least 70% pure, at least 71% pure, at least 72% pure, at least 73% pure, at least 74% pure, at least 75% pure, at least 76% pure, at least 77% pure, at least 78% pure, at least 79% pure, at least 80% pure, at least 81% pure, at least 82% pure, at least 83% pure, at least 84% pure, at least 85% pure, at least 86% pure, at least 87% pure, at least 88% pure, at least 89% pure, at least 90% pure, at least 91% pure, at least 92% pure, at least 93% pure, at least 94% pure, at least 95% pure, at least 96% pure, at least 97% pure, at least 98% pure, at least 99% pure, at least 99.1% pure, at least 99.2% pure, at least 99.3% pure, at least 99.4% pure, at least 99.5% pure, at least 99.6% pure, at least 99.7% pure, at least 99.8% pure, or at least 99.9% pure.


Compositions of the invention can be packaged as a kit. In some embodiments, a kit includes written instructions on the administration or use of the composition. The written material can be, for example, a label. The written material can suggest conditions methods of administration. The instructions provide the subject and the supervising physician with the best guidance for achieving the optimal clinical outcome from the administration of the therapy. In some embodiments, the label can be approved by a regulatory agency, for example the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA), or other regulatory agencies.


A composition described herein can be provided as a pharmaceutical formulation that can be stored in, for example, a vial or a container. The vial or container can contain, at the opening, a dispensing region from which a portion of the pharmaceutical formulation can be withdrawn for administration to a subject. In some embodiments, the dispensing region is punctured by, for example, a hypodermic needle to access the pharmaceutical formulation. The dispensing region can be punctured, for example, one time, two times, three times, four times, five times, six times, seven time, eight time, nine times, ten times, 11 times, 12 times, 13 times, 14 times, 15 times, or as many times as needed for treatment of the subject. The dispensing region can be made of, for example, aluminum, rubber, plastic, wax, or any combination thereof.


The dispensing region of the contain can be punctured within one hour, two hours, three hours, four hours, five hours, six hours, seven hours, eight hours, nine hours, ten hours, 11 hours, 12 hours, one day (24 hours), two days (48 hours), three days, four days, five days, six days, one week, two weeks, three weeks, one month (30 days), two months, three months, four months, five months, six months, seven months, eight months, nine months, ten months, eleven months, or one year from the first puncture of the dispensing region of the container.


Pharmaceutically-Acceptable Excipients.

Non-limiting examples of pharmaceutically-acceptable excipients can be found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa. 1975; Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999), each of which is incorporated by reference in its entirety.


In some embodiments, the pharmaceutical composition provided herein comprises a sugar as an excipient. Non-limiting examples of sugars include trehalose, sucrose, glucose, lactose, galactose, glyceraldehyde, fructose, dextrose, maltose, xylose, mannose, maltodextrin, starch, cellulose, lactulose, cellobiose, mannobiose, and combinations thereof.


In some embodiments, the pharmaceutical composition provided herein comprises a buffer as an excipient. Non-limiting examples of buffers include potassium phosphate, sodium phosphate, saline sodium citrate buffer (SSC), acetate, saline, physiological saline, phosphate buffer saline (PBS), 4-2-hydroxyethyl-1-piperazineethanesulfonic acid buffer (HEPES), 3-(N-morpholino)propanesulfonic acid buffer (MOPS), and piperazine-N,N′-bis(2-ethanesulfonic acid) buffer (PIPES), or combinations thereof.


In some embodiments, a pharmaceutical composition of the invention comprises a source of divalent metal ions as an excipient. A metal can be in elemental form, a metal atom, or a metal ion. Non-limiting examples of metals include transition metals, main group metals, and metals of Group 1, Group 2, Group 3, Group 4, Group 5, Group 6, Group 7, Group 8, Group 9, Group 10, Group 11, Group 12, Group 13, Group 14, and Group 15 of the Periodic Table. Non-limiting examples of metals include lithium, sodium, potassium, cesium, magnesium, calcium, strontium, scandium, titanium, vanadium, chromium, manganese, iron, cobalt, nickel, copper, zinc, yttrium, zirconium, niobium, molybdenum, palladium, silver, cadmium, tungsten, rhenium, osmium, iridium, platinum, gold, mercury, cerium, and samarium.


In some embodiments, the pharmaceutical composition provided herein comprises an alcohol as an excipient. Non-limiting examples of alcohols include ethanol, propylene glycol, glycerol, polyethylene glycol, chlorobutanol, isopropanol, xylitol, sorbitol, maltitol, erythritol, threitol, arabitol, ribitol, mannitol, galactilol, fucitol, lactitol, and combinations thereof.


Pharmaceutical preparations can be formulated with polyethylene glycol (PEG). PEGs with molecular weights ranging from about 300 g/mol to about 10,000,000 g/mol can be used. Non-limiting examples of PEGs include PEG 200, PEG 300, PEG 400, PEG 540, PEG 550, PEG 600, PEG 1000, PEG 1450, PEG 1500, PEG 2000, PEG 3000, PEG 3350, PEG 4000, PEG 4600, PEG 6000, PEG 8000, PEG 10,000, and PEG 20,000.


Further excipients that can be used in a composition of the invention include, for example, benzalkonium chloride, benzethonium chloride, benzyl alcohol, butylated hydroxyanisole, butylated hydroxytoluene, chlorobutanol, dehydroacetic acid, ethylenediamine, ethyl vanillin, glycerin, hypophosphorous acid, phenol, phenylethyl alcohol, phenylmercuric nitrate, potassium benzoate, potassium metabisulfite, potassium sorbate, sodium bisulfite, sodium metabisulfite, sorbic acid, thimerasol, acetic acid, aluminum monostearate, boric acid, calcium hydroxide, calcium stearate, calcium sulfate, calcium tetrachloride, cellulose acetate phthalate, microcrystalline celluose, chloroform, citric acid, edetic acid, and ethylcellulose.


In some embodiments, the pharmaceutical composition provided herein comprises an aprotic solvent as an excipient. Non-limiting examples of aprotic solvents include perfluorohexane, α,α,α-trifluorotoluene, pentane, hexane, cyclohexane, methylcyclohexane, decalin, dioxane, carbon tetrachloride, freon-11, benzene, toluene, carbon disulfide, diisopropyl ether, diethyl ether, t-butyl methyl ether, ethyl acetate, 1,2-dimethoxyethane, 2-methoxyethyl ether, tetrahydrofuran, methylene chloride, pyridine, 2-butanone, acetone, N-methylpyrrolidinone, nitromethane, dimethylformamide, acetonitrile, sulfolane, dimethyl sulfoxide, and propylene carbonate.


The amount of the excipient in a pharmaceutical composition of the invention can be about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 60%, about 70%, about 80%, about 90%, about 100%, about 200%, about 300%, about 400%, about 500%, about 600%, about 700%, about 800%, about 900%, or about 1000% by mass of the vasopressin in the pharmaceutical composition.


The amount of the excipient in a pharmaceutical composition of the invention can be about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 3%, about 3.5%, about 4%, about 4.5%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55% about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 99%, or about 100% by mass or by volume of the unit dosage form.


The ratio of vasopressin to an excipient in a pharmaceutical composition of the invention can be about 100:about 1, about 95:about 1, about 90:about 1, about 85:about 1, about 80:about 1, about 75:about 1, about 70:about 1, about 65:about 1, about 60:about 1, about 55:about 1, about 50:about 1, about 45:about 1, about 40:about 1, about 35:about 1 about 30:about 1, about 25:about 1, about 20:about 1, about 15:about 1, about 10:about 1, about 9:about 1, about 8:about 1, about 7:about 1, about 6:about 1, about 5:about 1, about 4:about 1, about 3:about 1, about 2:about 1, about 1:about 1, about 1:about 2, about 1:about 3, about 1:about 4, about 1:about 5, about 1:about 6, about 1:about 7, about 1:about 8, about 1:about 9, or about 1:about 10.


Pharmaceutically-Acceptable Salts.

The invention provides the use of pharmaceutically-acceptable salts of any therapeutic compound described herein. Pharmaceutically-acceptable salts include, for example, acid-addition salts and base-addition salts. The acid that is added to the compound to form an acid-addition salt can be an organic acid or an inorganic acid. A base that is added to the compound to form a base-addition salt can be an organic base or an inorganic base. In some embodiments, a pharmaceutically-acceptable salt is a metal salt. In some embodiments, a pharmaceutically-acceptable salt is an ammonium salt.


Metal salts can arise from the addition of an inorganic base to a compound of the invention. The inorganic base consists of a metal cation paired with a basic counterion, such as, for example, hydroxide, carbonate, bicarbonate, or phosphate. The metal can be an alkali metal, alkaline earth metal, transition metal, or main group metal. In some embodiments, the metal is lithium, sodium, potassium, cesium, cerium, magnesium, manganese, iron, calcium, strontium, cobalt, titanium, aluminum, copper, cadmium, or zinc.


In some embodiments, a metal salt is a lithium salt, a sodium salt, a potassium salt, a cesium salt, a cerium salt, a magnesium salt, a manganese salt, an iron salt, a calcium salt, a strontium salt, a cobalt salt, a titanium salt, an aluminum salt, a copper salt, a cadmium salt, or a zinc salt.


Ammonium salts can arise from the addition of ammonia or an organic amine to a compound of the invention. In some embodiments, the organic amine is triethyl amine, diisopropyl amine, ethanol amine, diethanol amine, triethanol amine, morpholine, N-methylmorpholine, piperidine, N-methylpiperidine, N-ethylpiperidine, dibenzylamine, piperazine, pyridine, pyrrazole, pipyrrazole, imidazole, pyrazine, or pipyrazine.


In some embodiments, an ammonium salt is a triethyl amine salt, a diisopropyl amine salt, an ethanol amine salt, a diethanol amine salt, a triethanol amine salt, a morpholine salt, an N-methylmorpholine salt, a piperidine salt, an N-methylpiperidine salt, an N-ethylpiperidine salt, a dibenzylamine salt, a piperazine salt, a pyridine salt, a pyrrazole salt, a pipyrrazole salt, an imidazole salt, a pyrazine salt, or a pipyrazine salt.


Acid addition salts can arise from the addition of an acid to a compound of the invention. In some embodiments, the acid is organic. In some embodiments, the acid is inorganic. In some embodiments, the acid is hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, nitrous acid, sulfuric acid, sulfurous acid, a phosphoric acid, isonicotinic acid, lactic acid, salicylic acid, tartaric acid, ascorbic acid, gentisinic acid, gluconic acid, glucaronic acid, saccharic acid, formic acid, benzoic acid, glutamic acid, pantothenic acid, acetic acid, propionic acid, butyric acid, fumaric acid, succinic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, oxalic acid, or maleic acid.


In some embodiments, the salt is a hydrochloride salt, a hydrobromide salt, a hydroiodide salt, a nitrate salt, a nitrite salt, a sulfate salt, a sulfite salt, a phosphate salt, isonicotinate salt, a lactate salt, a salicylate salt, a tartrate salt, an ascorbate salt, a gentisinate salt, a gluconate salt, a glucaronate salt, a saccarate salt, a formate salt, a benzoate salt, a glutamate salt, a pantothenate salt, an acetate salt, a propionate salt, a butyrate salt, a fumarate salt, a succinate salt, a methanesulfonate (mesylate) salt, an ethanesulfonate salt, a benzenesulfonate salt, a p-toluenesulfonate salt, a citrate salt, an oxalate salt, or a maleate salt.


Peptide Sequence.

As used herein, the abbreviations for the L-enantiomeric and D-enantiomeric amino acids are as follows: alanine (A, Ala); arginine (R, Arg); asparagine (N, Asn); aspartic acid (D, Asp); cysteine (C, Cys); glutamic acid (E, Glu); glutamine (Q, Gln); glycine (G, Gly); histidine (H, His); isoleucine (I, Ile); leucine (L, Leu); lysine (K, Lys); methionine (M, Met); phenylalanine (F, Phe); proline (P, Pro); serine (S, Ser); threonine (T, Thr); tryptophan (W, Trp); tyrosine (Y, Tyr); valine (V, Val). In some embodiments, the amino acid is a L-enantiomer. In some embodiments, the amino acid is a D-enantiomer.


A peptide of the disclosure can have about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 81%, about 82%, about 83%, about 84%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100% amino acid sequence homology to SEQ ID NO. 1.


In some embodiments, a pharmaceutical composition of the invention comprises one or a plurality of peptides having about 80% to about 90% sequence homology to SEQ ID NO. 1, about 88% to about 90% sequence homology to SEQ ID NO. 1 or 88% to 90% sequence homology to SEQ ID NO. 1. In some embodiments, a pharmaceutical composition of the invention comprises vasopressin and one or more of a second, third, fourth, fifth, sixth, seventh, eighth, ninth, and tenth peptide.


The ratio of vasopressin to another peptide in a pharmaceutical composition of the invention can be, for example, about 1000:about 1, about 990:about 1, about 980:about 1, about 970:about 1, about 960:about 1, about 950:about 1, about 800:about 1, about 700: about 1, about 600:1, about 500:about 1, about 400:about 1, about 300:about 1, about 200: about 1, about 100:about 1, about 95:about 1, about 90:about 1, about 85:about 1, about 80: about 1, about 75:about 1, about 70:about 1, about 65:about 1, about 60:about 1, about 55: about 1, about 50:about 1, about 45:about 1, about 40:about 1, about 35:about 1, about 30:about 1, about 25:about 1, about 20:about 1, about 19:about 1, about 18:about 1, about 17:about 1, about 16:about 1, about 15:about 1, about 14:about 1, about 13:about 1, about 12:about 1, about 11:about 1, or about 10:about 1.


The amount of another peptide or impurity in a composition of the invention can be, for example, about 0.01%, about 0.02%, about 0.03%, about 0.04%, about 0.05%, about 0.06%, about 0.07%, about 0.08%, about 0.09%, about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.1%, about 1.2%, about 1.3%, about 1.4%, about 1.5%, about 1.6%, about 1.7%, about 1.8%, about 1.9%, about 2%, about 2.1%, about 2.2%, about 2.3%, about 2.4%, about 2.5%, about 2.6%, about 2.7%, about 2.8%, about 2.9%, about 3%, about 3.1%, about 3.2%, about 3.3%, about 3.4%, about 3.5%, about 3.6%, about 3.7%, about 3.8%, about 3.9%, about 4%, about 4.1%, about 4.2%, about 4.3%, about 4.4%, about 4.5%, about 4.6%, about 4.7%, about 4.8%, about 4.9%, about 5%, about 5.5%, about 6%, about 6.5%, about 7%, about 7.5%, about 8%, about 8.5%, about 9%, about 9.5%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% by mass of vasopressin.


Another peptide or impurity present in a composition described herein can be, for example, SEQ ID NO.: 2, SEQ ID NO.: 3, SEQ ID NO.: 4, SEQ ID NO.: 5, SEQ ID NO.: 6, SEQ ID NO.: 7, SEQ ID NO.: 8, SEQ ID NO.: 9, SEQ ID NO.: 10, SEQ ID NO.: 11, SEQ ID NO.: 12, SEQ ID NO.: 13, SEQ ID NO.: 14, SEQ ID NO.: 15, SEQ ID NO.: 16, SEQ ID NO.: 17, a dimer of SEQ ID NO.: 1, an unidentified impurity, or any combination thereof.


Non-limiting examples of methods that can be used to identify peptides of the invention include high-performance liquid chromatography (HPLC), mass spectrometry (MS), Matrix Assisted Laser Desorption Ionization Time-of-Flight (MALDI-TOF), electrospray ionization Time-of-flight (ESI-TOF), gas chromatography-mass spectrometry (GC-MS), liquid chromatography-mass spectrometry (LC-MS), and two-dimensional gel electrophoresis.


HPLC can be used to identify peptides using high pressure to separate components of a mixture through a packed column of solid adsorbent material, denoted the stationary phase. The sample components can interact differently with the column based upon the pressure applied to the column, material used in stationary phase, size of particles used in the stationary phase, the composition of the solvent used in the column, and the temperature of the column. The interaction between the sample components and the stationary phase can affect the time required for a component of the sample to move through the column. The time required for component to travel through the column from injection point to elution is known as the retention time.


Upon elution from the column, the eluted component can be detected using a UV detector attached to the column. The wavelength of light at which the component is detected, in combination with the component's retention time, can be used to identify the component. Further, the peak displayed by the detector can be used to determine the quantity of the component present in the initial sample. Wavelengths of light that can be used to detect sample components include, for example, about 200 nM, about 225 nm, about 250 nm, about 275 nm, about 300 nm, about 325 nm, about 350 nm, about 375 nm, and about 400 nm.


Mass spectrometry (MS) can also be used to identify peptides of the invention. To prepare samples for MS analysis, the samples, containing the proteins of interest, are digested by proteolytic enzymes into smaller peptides. The enzymes used for cleavage can be, for example, trypsin, chymotrypsin, glutamyl endopeptidase, Lys-C, and pepsin. The samples can be injected into a mass spectrometer. Upon injection, all or most of the peptides can be ionized and detected as ions on a spectrum according to the mass to charge ratio created upon ionization. The mass to charge ratio can then be used to determine the amino acid residues present in the sample.


The present disclosure provides several embodiments of pharmaceutical formulations that provide advantages in stability, administration, efficacy, and modulation of formulation viscosity. Any embodiments disclosed herein can be used in conjunction or individually. For example, any pharmaceutically-acceptable excipient, method, technique, solvent, compound, or peptide disclosed herein can be used together with any other pharmaceutically-acceptable excipient, method, technique, solvent, compound, or peptide disclosed herein to achieve any therapeutic result. Compounds, excipients, and other formulation components can be present at any amount, ratio, or percentage disclosed herein in any such formulation, and any such combination can be used therapeutically for any purpose described herein and to provide any viscosity described herein.


EXAMPLES
Example 1: Impurities of Vasopressin as Detected by HPLC

To analyze degradation products of vasopressin that can be present in an illustrative formulation of vasopressin, gradient HPLC was performed to separate vasopressin from related peptides and formulation components. TABLE 2 below depicts the results of the experiment detailing the chemical formula, relative retention time (RRT), molar mass, and structure of vasopressin and detected impurities.


Vasopressin was detected in the eluent using UV absorbance. The concentration of vasopressin in the sample was determined by the external standard method, where the peak area of vasopressin in sample injections was compared to the peak area of vasopressin reference standards in a solution of known concentration. The concentrations of related peptide impurities in the sample were also determined using the external standard method, using the vasopressin reference standard peak area and a unit relative response factor. An impurities marker solution was used to determine the relative retention times of identified related peptides at the time of analysis.


Experimental conditions are summarized in TABLE 2 below.










TABLE 2







Column
YMC-Pack ODS-AM, 3 μm, 120 Å pore,



4.6 × 100 mm


Column Temperature
25° C.


Flow Rate
1.0 mL/min


Detector
215 nm



Note: For Identification a Diode Array



Detector (DAD) was used with the range



of 200-400 nm.


Injection Volume
100 μL


Run time
55 minutes


Autosampler Vials
Polypropylene vials















Time (min)
% A
% B
Flow





Pump (gradient)
0
90
10
1.0



40
50
50
1.0



45
50
50
1.0



46
90
10
1.0



55
90
10
1.0









The diluent used for the present experiment was 0.25% v/v Acetic Acid, which was prepared by transferring 2.5 mL of glacial acetic acid into a 1-L volumetric flask containing 500 mL of water. The solution was diluted to the desired volume with water.


Phosphate buffer at pH 3.0 was used for mobile phase A. The buffer was prepared by weighing approximately 15.6 g of sodium phosphate monobasic monohydrate into a beaker. 1000 mL of water was added, and mixed well. The pH was adjusted to 3.0 with phosphoric acid. The buffer was filtered through a 0.45 μm membrane filter under vacuum, and the volume was adjusted as necessary.


An acetonitrile:water (50:50) solution was used for mobile phase B. To prepare mobile phase B, 500 mL of acetonitrile was mixed with 500 mL of water.


The working standard solution contained approximately 20 units/mL of vasopressin. The standard solution was prepared by quantitatively transferring the entire contents of 1 vial of USP Vasopressin RS with diluent to a 50-mL volumetric flask.


The intermediate standard solution was prepared by pipetting 0.5 mL of the working standard solution into a 50-mL volumetric flask.


The sensitivity solution was prepared by pipetting 5.0 mL of the intermediate standard solution into a 50-mL volumetric flask. The solution was diluted to the volume with Diluent and mixed well.


A second working standard solution was prepared as directed under the standard preparation.


A portion of the vasopressin control sample was transferred to an HPLC vial and injected. The control was stable for 120 hours when stored in autosampler vials at ambient laboratory conditions.


To prepare the impurities marker solution, a 0.05% v/v acetic acid solution was prepared by pipetting 200.0 mL of a 0.25% v/v acetic acid solution into a 1-L volumetric flask. The solution was diluted to the desired volume with water and mixed well.


To prepare the vasopressin impurity stock solutions, the a solution of each impurity was prepared in a 25 mL volumetric flask and diluted with 0.05% v/v acetic acid to a concentration suitable for HPLC injection.


To prepare the MAA/H-IBA (Methacrylic Acid/α-Hydroxy-isobutyric acid) stock solution, a stock solution containing approximately 0.3 mg/mL H-IBA and 0.01 mg/mL in 0.05% v/v acetic acid was made in a 50 mL volumetric flask.


To prepare the chlorobutanol diluent, about one gram of hydrous chlorobutanol was added to 500 mL of water. Subsequently, 0.25 mL of acetic acid was added and the solution was stirred to dissolve the chlorobutanol.


To prepare the impurity marker solution, vasopressin powder was mixed with the impurity stock solutions prepared above.


The solutions were diluted to volume with the chlorobutanol diluent. The solutions were aliquoted into individual crimp top vials and stored at 2-8° C. At time of use, the solutions were removed from refrigeration (2-8° C.) and allowed to reach room temperature.


The vasopressin impurity marker solution was stable for at least 120 hours when stored in auto-sampler vials at ambient laboratory conditions. The solution was suitable for use as long as the chromatographic peaks could be identified based on comparison to the reference chromatogram.


To begin the analysis, the HPLC system was allowed to equilibrate for at least 30 minutes using mobile phase B, followed by time 0 min gradient conditions until a stable baseline was achieved.


The diluent was injected at the beginning of the run, and had no peaks that interfered with Vasopressin at around 18 minutes as shown in FIG. 1.


A single injection of the sensitivity solution was performed, wherein the signal-to-noise ratio of the Vasopressin was greater than or equal to ten as shown in FIG. 2.


A single injection of the impurities marker solution was then made. The labeled impurities in the reference chromatogram were identified in the chromatogram of the marker solution based on their elution order and approximate retention times shown in FIG. 3 and FIG. 4. FIG. 4 is a zoomed in chromatograph of FIG. 3 showing the peaks that eluted between 15 and 30 minutes. The nomenclature, structure, and approximate retention times for individual identified impurities are detailed in TABLE 3.


A single injection of the working standard solution was made to ensure that the tailing factor of the vasopressin peak was less than or equal to about 2.0 as shown in FIG. 5.


A total of five replicate injections of the working standard solution were made to ensure that the relative standard deviation (% RSD) of the five replicate vasopressin peak areas was not more than 2.0%.


Two replicate injections of the check standard preparation were to confirm that the check standard conformity was 99.0%-101.0%. One injection of the control sample was made to confirm that the assay of the control sample met the control limits established for the sample.


Then, one injection of the working standard solution was made.


Following the steps above done to confirm system suitability, a single injection of each sample preparation was made. The chromatograms were analyzed to determine the vasopressin and impurity peak areas. The chromatogram is depicted in FIG. 6.


The working standard solution was injected after 1 to 4 sample injections, and the bracketing standard peak areas were averaged for use in the calculations to determine peak areas of vasopressin and associated impurities.


The relative standard deviation (% RSD) of vasopressin peak areas for the six injections of working standard solution was calculated by including the initial five injections from the system suitability steps above and each of the subsequent interspersed working standard solution injections. The calculations were done to ensure that each of the % RSD were not more than 2.0%.


The retention time of the major peak in the chromatogram of the sample preparation corresponded to that of the vasopressin peak in the working standard solution injection that preceded the sample preparation injection.


The UV spectrum (200-400 nm) of the main peak in the chromatogram of the sample preparation compared to the UV spectrum of vasopressin in the working standard preparation. FIG. 7 depicts a UV spectrum of a vasopressin sample and FIG. 8 depicts a UV spectrum of vasopressin standard.


To calculate the vasopressin units/mL, the following formula was used:







Vasopressin





units


/


mL

=



R
U


R
S


×
Conc





STD





where:


RU=Vasopressin peak area response of Sample preparation.


RS=average vasopressin peak area response of bracketing standards.


Conc STD=concentration of the vasopressin standard in units/mL


To identify the impurities, the % Impurity and identity for identified impurities (TABLE 3) that are were greater than or equal to 0.10% were reported. Impurities were truncated to 3 decimal places and then rounded to 2 decimal places, unless otherwise specified.


The impurities were calculated using the formula below:







%





impurity

=



R
I


R
S


×


Conc





STD


20





U


/


mL


×
100

%





where:


RI=Peak area response for the impurity


20 U/mL=Label content of vasopressin


TABLE 3 below details the chemical formula, relative retention time (RRT in minutes), molar mass, and structure of vasopressin and detected impurities.












TABLE 3







Appr.
Molar


Name
Formula
RRT
Mass (g)







Vasopressin
C46H65N15O12S2
1.00
1084.23


(Arginine Vasopressin, AVP)







CYFQNCPRG-NH2 SEQ ID NO.: 1


(disulfide bridge between cys residues)










Gly9-vasopressin
C46H64N14O13S2
1.07
1085.22


(Gly9-AVP)







CYFQNCPRG SEQ ID NO.: 2 (disulfide bridge between cys residues)










Asp5-vasopressin
C46H64N14O13S2
1.09
1085.22


(Asp5-AVP)







CYFQDCPRG-NH2 SEQ ID NO.: 3


(disulfide bridge between cys residues)










Glu4-vasopressin
C46H64N14O13S2
1.12
1085.22


(Glu4-AVP)







CYFENCPRG-NH2 SEQ ID NO.: 4


(disulfide bridge between cys residues)










Acetyl-vasopressin
C48H67N15O13S2
1.45
1126.27


(Acetyl-AVP)







Ac-CYFQNCPRG-NH2 SEQ ID NO.: 7


(disulfide bridge between cys residues)










D-Asn-vasopressin
C46H65N15O12S2
0.97
1084.23


(DAsn-AVP)







CYFQ(D-Asn)CPRG-NH2 SEQ ID NO.: 10 (disulfide bridge between cys


residues)










Dimeric-vasopressin
C92H130N30O24S4
1.22
2168.46


(Dimer-AVP) (monomers


cross linked by disulfide


bridges)









Example 2: Investigation of pH

To determine a possible pH for a vasopressin formulation with good shelf life, vasopressin formulations were prepared in 10 mM citrate buffer diluted in isotonic saline across a range of pH. Stability was assessed via HPLC as in EXAMPLE 1 after incubation of the formulations at 60° C. for one week. FIG. 9 illustrates the results of the experiment. The greatest level of stability was observed at pH 3.5. At pH 3.5, the percent label claim (% LC) of vasopressin was highest, and the proportion of total impurities was lowest.


Example 3: Effect of Peptide Stabilizers on Vasopressin Formulation

To observe the effect of stabilizers on the degradation of vasopressin, a series of peptide stabilizers were added to a vasopressin formulation as detailed in TABLE 4. Stability of vasopressin was assessed via HPLC after incubation of the formulations at 60° C. for one week.














TABLE 4






PEG

Poloxamer

n-Methylpyrrolidone


Ethanol
400
Glycerol
188
HPbCDa
(NMP)




















1%
1%
1%
1%
1%
1%


10%
10%
10%
10%
10%
10%






aHydroxypropyl beta-Cyclodextrin








FIG. 10 illustrates the stability of vasopressin in terms of % label claim at varying concentrations of stabilizer. The results indicate that the tested stabilizers provided a greater stabilizing effect at 1% concentration than at 10%. Also, in several cases the stabilization effect was about 5% to about 10% greater than that observed in the experiments of EXAMPLE 2.


Example 4: Effect of Buffer and Divalent Metals on Vasopressin Formulation

To determine whether different combinations of buffers and use of divalent metals affect vasopressin stability, vasopressin formulations with varying concentrations of citrate and acetate buffers and variable concentrations of calcium, magnesium, and zinc ions were prepared. Solutions of 0 mM, 10 mM, 20 mM, and 80 mM calcium, magnesium, and zinc were prepared and each was combined with 1 mM or 10 mM of citrate or acetate buffers to test vasopressin stability.


The tested combinations provided vasopressin stability comparable to that of a vasopressin formulation lacking buffers and divalent metals. However, that the addition of divalent metal ions was able to counteract the degradation of vasopressin caused by the use of a citrate buffer.


Example 5: Illustrative Formulations for Assessment of Vasopressin Stability

An aqueous formulation of vasopressin is prepared using 10% trehalose, 1% sucrose, or 5% NaCl and incubated at 60° C. for one week, at which point stability of vasopressin is assessed using HPLC.


A formulation containing 50 units of vasopressin is lyophilized. The lyophilate is reconstituted with water and either 100 mg of sucrose or 100 mg of lactose, and the stability of vasopressin is tested via HPLC after incubation at 60° C. for one week.


Co-solvents are added to a vasopressin solution to assess vasopressin stability. 95% solvent/5% 20 mM acetate buffer solutions are prepared using propylene glycol, DMSO, PEG300, NMP, glycerol, and glycerol:NMP (1:1), and used to create formulations of vasopressin. The stability of vasopressin is tested after incubation at 60° C. for one week.


Amino acid and phosphate buffers are tested with vasopressin to assess vasopressin stability. Buffers of 10 mM glycine, aspartate, phosphate are prepared at pH 3.5 and 3.8 and used to create formulations of vasopressin. The stability of vasopressin is tested after incubation at 60° C. for one week.


A vasopressin formulation in 10% polyvinylpyrrolidone is prepared to assess vasopressin stability. The stability of vasopressin will be tested after incubation at 60° C. for one week.


A vasopressin formulation that contains 0.9% saline, 10 mM acetate buffer, 0.2 unit/mL API/mL in 100 mL of total volume is prepared. The pH of the solution is varied from pH 3.5-3.8 to test the stability of vasopressin.


A vasopressin formulation in about 50% to about 80% DMSO (for example, about 80%), about 20% to about 50% ethyl acetate (for example, about 20%), and about 5% to about 30% polyvinylpyrrolidone (PVP) (for example, about 10% by mass of the formulation) is prepared to assess vasopressin stability. PVP K12 and PVP K17 are each independently tested in the formulation. The stability of vasopressin is tested after incubation at 60° C. for one week.


A vasopressin formulation in about 70% to about 95% ethyl acetate, and about 5% to about 30% PVP is prepared to assess vasopressin stability. PVP K12 and PVP K17 are each independently tested in the formulation. The stability of vasopressin is tested after incubation at 60° C. for one week.


A vasopressin formulation in 90% DMSO and 10% PVP is prepared to test vasopressin stability. PVP K12 and PVP K17 are each independently tested in the formulation. The stability of vasopressin is tested after incubation at 60° C. for one week.


Example 6: Illustrative Vasopressin Formulation for Clinical Use

A formulation for vasopressin that can be used in the clinic is detailed in TABLE 5 below:











TABLE 5





Ingredient
Function
Amount (per mL)







Vasopressin, USP
Active Ingredient
20 Units (~0.04 mg)


Chlorobutanol, Hydrous NF
Preservative
5.0 mg


Acetic Acid, NF
pH Adjustment
To pH 3.4-3.6




(~0.22 mg)


Water for injection, USP/EP
Diluent
QS









Example 7: Illustrative Regimen for Therapeutic Use of a Vasopressin Formulation

Vasopressin is indicated to increase blood pressure in adults with vasodilatory shock (for example, adults who are post-cardiotomy or septic) who remain hypotensive despite fluids and catecholamines.


Preparation and Use of Vasopressin.

Vasopressin is supplied in a carton of 25 multi-dose vials each containing 1 mL vasopressin at 20 units/mL.


Vasopressin is stored between 15° C. and 25° C. (59° F. and 77° F.), and is not frozen. Alternatively, a unit dosage form of vasopressin can be stored between 2° C. and 8° C. for about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, or 8 weeks.


Vials of vasopressin are to be discarded 48 hours after first puncture.


Vasopressin is prepared according to TABLE 6 below:











TABLE 6









Mix










Fluid Restriction?
Final Concentration
Vasopressin
Diluent





No
0.1 units/mL
2.5 mL (50 units)
500 mL


Yes
  1 unit/mL
5 mL (100 units)
100 mL









Vasopressin is diluted in normal saline (0.9% sodium chloride) or 5% dextrose in water (D5W) prior to use to either 0.1 units/mL or 1 unit/mL for intravenous administration. Unused diluted solution is discarded after 18 hours at room temperature or after 24 hours under refrigeration.


Diluted vasopressin should be inspected for particulate matter and discoloration prior to use whenever solution and container permit.


The goal of treatment with vasopressin is optimization of perfusion to critical organs, but aggressive treatment can compromise perfusion of organs, like the gastrointestinal tract, for which function is difficult to monitor. Titration of vasopressin to the lowest dose compatible with a clinically-acceptable response is recommended.


For post-cardiotomy shock, a dose of 0.03 units/minute is used as a starting point. For septic shock, a dose of 0.01 units/minute is recommended. If the target blood pressure response is not achieved, titrate up by 0.005 units/minute at 10- to 15-minute intervals. The maximum dose for post-cardiotomy shock is 0.1 units/minute and for septic shock 0.07 units/minute. After target blood pressure has been maintained for 8 hours without the use of catecholamines, taper vasopressin by 0.005 units/minute every hour as tolerated to maintain target blood pressure.


Vasopressin is provided at 20 units per mL of diluent, which is packaged as 1 mL of vasopressin per vial, and is diluted prior to administration.


Contraindications, Adverse Reactions, and Drug-Drug Interactions.

Vasopressin is contraindicated in patients with known allergy or hypersensitivity to 8-L-arginine vasopressin or chlorobutanol. Additionally, use of vasopressin in patients with impaired cardiac response can worsen cardiac output.


Adverse reactions have been observed with the use of vasopressin, which adverse reactions include bleeding/lymphatic system disorders, specifically, hemorrhagic shock, decreased platelets, intractable bleeding; cardiac disorders, specifically, right heart failure, atrial fibrillation, bradycardia, myocardial ischemia; gastrointestinal disorders, specifically, mesenteric ischemia; hepatobiliary disorders, specifically, increased bilirubin levels; renal/urinary disorders, specifically, acute renal insufficiency; vascular disorders, specifically, distal limb ischemia; metabolic disorders, specifically, hyponatremia; and skin disorders, specifically, and ischemic lesions.


These reactions are reported voluntarily from a population of uncertain size. Thus, reliable estimation of frequency or establishment of a causal relationship to drug exposure is unlikely.


Vasopressin has been observed to interact with other drugs. For example, use of vasopressin with catecholamines is expected to result in an additive effect on mean arterial blood pressure and other hemodynamic parameters. Use of vasopressin with indomethacin can prolong the effect of vasopressin on cardiac index and systemic vascular resistance. Indomethacin more than doubles the time to offset for vasopressin's effect on peripheral vascular resistance and cardiac output in healthy subjects.


Further, use of vasopressin with ganglionic blocking agents can increase the effect of vasopressin on mean arterial blood pressure. The ganglionic blocking agent tetra-ethylammonium increases the pressor effect of vasopressin by 20% in healthy subjects.


Use of vasopressin with furosemide increases the effect of vasopressin on osmolar clearance and urine flow. Furosemide increases osmolar clearance 4-fold and urine flow 9-fold when co-administered with exogenous vasopressin in healthy subjects.


Use of vasopressin with drugs suspected of causing SIADH (Syndrome of inappropriate antidiuretic hormone secretion), for example, SSRIs, tricyclic antidepressants, haloperidol, chlorpropamide, enalapril, methyldopa, pentamidine, vincristine, cyclophosphamide, ifosfamide, and felbamate can increase the pressor effect in addition to the antidiuretic effect of vasopressin. Additionally, use of vasopressin with drugs suspected of causing diabetes insipidus for example, demeclocycline, lithium, foscarnet, and clozapine can decrease the pressor effect in addition to the antidiuretic effect of vasopressin.


Halothane, morphine, fentanyl, alfentanyl and sufentanyl do not impact exposure to endogenous vasopressin.


Use of Vasopressin in Specific Populations.

Vasopressin is a Category C Drug for Pregnancy.


Due to a spillover into the blood of placental vasopressinase, the clearance of exogenous and endogenous vasopressin increases gradually over the course of a pregnancy. During the first trimester of pregnancy the clearance is only slightly increased. However, by the third trimester the clearance of vasopressin is increased about 4-fold and at term up to 5-fold. Due to the increased clearance of vasopressin in the second and third trimester, the dose of vasopressin can be up-titrated to doses exceeding 0.1 units/minute in post-cardiotomy shock and 0.07 units/minute in septic shock. Vasopressin can produce tonic uterine contractions that could threaten the continuation of pregnancy. After delivery, the clearance of vasopressin returns to preconception levels.


Overdosage.

Overdosage with vasopressin can be expected to manifest as a consequence of vasoconstriction of various vascular beds, for example, the peripheral, mesenteric, and coronary vascular beds, and as hyponatremia. In addition, overdosage of vasopressin can lead less commonly to ventricular tachyarrhythmias, including Torsade de Pointes, rhabdomyolysis, and non-specific gastrointestinal symptoms. Direct effects of vasopressin overdose can resolve within minutes of withdrawal of treatment.


Pharmacology of Vasopressin.

Vasopressin is a polypeptide hormone that causes contraction of vascular and other smooth muscles and antidiuresis, which can be formulated as a sterile, aqueous solution of synthetic arginine vasopressin for intravenous administration. The 1 mL solution contains vasopressin 20 units/mL, chlorobutanol, NF 0.5% as a preservative, and water for injection, USP adjusted with acetic acid to pH 3.4-3.6.


The chemical name of vasopressin is Cyclo (1-6) L-Cysteinyl-L-Tyrosyl-L-Phenylalanyl-L-Glutaminyl-L-Asparaginyl-L-Cysteinyl-L-Prolyl-L-Arginyl-L-Glycinamide. Vasopressin is a white to off-white amorphous powder, freely soluble in water. The structural formula of vasopressin is:




embedded image


Molecular Formula: C46H65N15O12S2; Molecular Weight: 1084.23


One mg of vasopressin is equivalent to 530 units. Alternatively, one mg of vasopressin is equivalent to 470 units.


The vasoconstrictive effects of vasopressin are mediated by vascular V1 receptors. Vascular V1 receptors are directly coupled to phospholipase C, resulting in release of calcium, leading to vasoconstriction. In addition, vasopressin stimulates antidiuresis via stimulation of V2 receptors which are coupled to adenyl cyclase.


At therapeutic doses, exogenous vasopressin elicits a vasoconstrictive effect in most vascular beds including the splanchnic, renal, and cutaneous circulation. In addition, vasopressin at pressor doses triggers contractions of smooth muscles in the gastrointestinal tract mediated by muscular V1-receptors and release of prolactin and ACTH via V3 receptors. At lower concentrations typical for the antidiuretic hormone, vasopressin inhibits water diuresis via renal V2 receptors. In patients with vasodilatory shock, vasopressin in therapeutic doses increases systemic vascular resistance and mean arterial blood pressure and reduces the dose requirements for norepinephrine.


Vasopressin tends to decrease heart rate and cardiac output. The pressor effect is proportional to the infusion rate of exogenous vasopressin. Onset of the pressor effect of vasopressin is rapid, and the peak effect occurs within 15 minutes. After stopping the infusion, the pressor effect fades within 20 minutes. There is no evidence for tachyphylaxis or tolerance to the pressor effect of vasopressin in patients.


At infusion rates used in vasodilatory shock (0.01-0.1 units/minute), the clearance of vasopressin is 9 to 25 mL/min/kg in patients with vasodilatory shock. The apparent half-life of vasopressin at these levels is ≤10 minutes. Vasopressin is predominantly metabolized and only about 6% of the dose is excreted unchanged in urine. Animal experiments suggest that the metabolism of vasopressin is primarily by liver and kidney. Serine protease, carboxipeptidase and disulfide oxido-reductase cleave vasopressin at sites relevant for the pharmacological activity of the hormone. Thus, the generated metabolites are not expected to retain important pharmacological activity.


Carcinogenesis, Mutagenesis, Impairment of Fertility.

Vasopressin was found to be negative in the in vitro bacterial mutagenicity (Ames) test and the in vitro Chinese hamster ovary (CHO) cell chromosome aberration test. In mice, vasopressin can have an effect on function and fertilizing ability of spermatozoa.


Clinical Studies.

Increases in systolic and mean blood pressure following administration of vasopressin were observed in seven studies in septic shock and eight studies in post-cardiotomy vasodilatory shock.


Example 8: Effect of Temperature on Vasopressin Formulations

To test the effect of temperature on the stability of vasopressin formulation, solutions containing 20 units/mL vasopressin and chlorobutanol, adjusted to pH 3.5 with acetic acid, were prepared. One mL of each vasopressin formulations was then filled into 3 cc vials. Each Vasopressin Formulation was stored either inverted or upright for at least three months, up to 24 months, at: (i) 5° C.; (ii) 25° C. and 60% relative humidity; or (iii) 40° C. and 75% humidity, and the amount of vasopressin (U/mL) and % total impurities were measured periodically. TABLES 7-12 below display the results of the experiments at 5° C. The results of the experiments at 25° C. are included in TABLES 13-18. All of the experiments were performed in triplicate. The results of the experiments at 40° C. are included in TABLES 19-24. For each temperature tested, three lots of the vasopressin formulation were stored for 24 months (5° C. and 25° C.) and 3 months (40° C.), and measurements were taken at regular intervals during the testing periods. “NMT” as used in the tables denotes “not more than.”


The vasopressin and impurity amounts observed in the experiments conducted at 5° C. are shown in TABLES 7-12 below (AVP=Vasopressin).









TABLE 7







Samples stored inverted at 5° C.









Time in months
















Test
Initial
1
2
3
6
9
12
18
24





AVP
19.4
19.4
19.4
19.3
19.5
19.4
19.5
19.4
19.3


As-


say



2.3%
2.0%
2.1%
2.3%
2.2%
2.3%
2.6%
2.9%
2.9%
















TABLE 8







Samples stored inverted at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24





AVP
16.0-21.0
19.7
19.7
19.7
19.7
19.9
19.7
19.8
19.7
19.5


As-
U/mL


say



Total
2.7%
2.2%
2.3%
2.4%
2.1%
2.3%
2.7%
2.9%
2.9%



Impurities:



NMT 17.0%
















TABLE 9







Samples stored inverted at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24





AVP
16.0-21.0
19.7
19.7
19.6
19.7
19.8
19.7
19.9
19.8
19.5


As-
U/mL


say



Total
2.2%
1.9%
2.0%
2.2%
2.0%
2.1%
2.4%
2.6%
2.8%



Impurities:



NMT 17.0%
















TABLE 10







Samples stored upright at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24





AVP
16.0-21.0
19.4
19.5
19.4
19.4
19.5
19.5
19.5
19.4
19.3


As-
U/mL


say



Total
2.3%
2.1%
2.1%
2.3%
2.1%
2.3%
2.5%
2.9%
2.9%



Impurities:



NMT 17.0%
















TABLE 11







Samples stored upright at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24





AVP
16.0-21.0
19.7
19.7
19.6
19.7
19.8
19.7
19.8
19.7
19.5


As-
U/mL


say



Total
2.7%
2.1%
2.2%
2.2%
2.2%
2.3%
2.6%
2.9%
2.8%



Impurities:



NMT 17.0%
















TABLE 12







Samples stored upright at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24





AVP
16.0-21.0
19.7
19.7
19.6
19.7
19.8
19.7
19.9
19.8
19.5


As-
U/mL


say



Total
2.2%
1.8%
2.0%
2.2%
2.2%
2.1%
2.4%
2.8%
2.7%



Impurities:



NMT 17.0%









The vasopressin and impurity amounts observed in the experiments conducted at 25° C. and 60% relative humidity are shown in TABLES 13-18 below.









TABLE 13







Samples stored inverted at 25° C. and 60% Relative Humidity










Acceptance
Time in months















Test
Criteria
Initial
3
6
9
12
18
24





AVP
16.0-21.0
19.8
19.4
19.1
18.8
18.3
17.5
17.3


As-
U/mL


say



Total
1.1%
2.4%
3.7%
4.7%
5.9%
9.0%
13.6%



Impurities:



NMT 17.0%
















TABLE 14







Samples stored inverted at 25° C. and 60% Relative Humidity










Acceptance
Time in months















Test
Criteria
Initial
3
6
9
12
18
24





AVP
16.0-21.0
20.1
19.7
19.3
19
18.6
17.6
17.6


As-
U/mL


say



Total
1.3%
2.5%
3.4%
4.6%
5.6%
9.0%
13.4%



Impurities:



NMT 17.0%
















TABLE 15







Samples stored inverted at 25° C. and 60% Relative Humidity










Acceptance
Time in months















Test
Criteria
Initial
3
6
9
12
18
24





AVP
16.0-21.0
19.9
19.6
19.2
19
18.7
18
17.4


As-
U/mL


say



Total
1.5%
2.6%
3.3%
4.6%
5.9%
9.0%
12.9%



Impurities:



NMT 17.0%
















TABLE 16







Samples stored upright at 25° C. and 60% Relative Humidity










Acceptance
Time in months















Test
Criteria
Initial
3
6
9
12
18
24





AVP
16.0-21.0
19.8
19.4
19.1
18.8
18.3
17.5
17.4


As-
U/mL


say



Total
1.1%
2.4%
3.2%
4.8%
5.6%
9.2%
13.1%



Impurities:



NMT 17.0%
















TABLE 17







Samples stored upright at 25° C. and 60% Relative Humidity










Acceptance
Time in months















Test
Criteria
Initial
3
6
9
12
18
24





AVP
16.0-21.0
20.1
19.7
19.4
18.9
18.6
17.8
17.7


As-
U/mL


say



Total
1.3%
2.5%
3.3%
4.5%
5.7%
9.1%
13.3%



Impurities:



NMT 17.0%
















TABLE 18







Samples stored upright at 25° C. and 60% Relative Humidity










Acceptance
Time in months















Test
Criteria
Initial
3
6
9
12
18
24





AVP
16.0-21.0
19.9
19.6
19.2
19
18.5
18.1
17.4


As-
U/mL


say



Total
1.5%
2.5%
3.7%
4.7%
5.9%
9.1%
13.3%



Impurities:



NMT 17.0%









The vasopressin and impurity amounts observed in the experiments conducted at 40° C. and 75% relative humidity are shown in TABLES 19-24 below.









TABLE 19







Samples stored inverted at 40° C.










Acceptance
Time in months












Test
Criteria
Initial
1
2
3





Vasopressin
18.0-21.0 U/mL
19.8
19.1
18.6
17.3


Assay



Total Impurities:
1.1%
3.7%
7.3%
10.6%



NMT 17.0%
















TABLE 20







Samples stored Upright at 40° C.










Acceptance
Time in months












Test
Criteria
Initial
1
2
3





Vasopressin
18.0-21.0 U/mL
19.8
18.9
18.5
17.2


Assay



Total Impurities:
1.1%
3.6%
7.2%
10.3%



NMT 17.0%
















TABLE 21







Samples stored inverted at 40° C.










Acceptance
Time in months












Test
Criteria
Initial
1
2
3





Vasopressin
18.0-21.0 U/mL
20.1
19.3
18.7
17.6


Assay



Total Impurities:
1.3%
3.6%
7.3%
10.3%



NMT 17.0%
















TABLE 22







Samples stored Upright at 40° C.










Acceptance
Time in months












Test
Criteria
Initial
1
2
3





Vasopressin
18.0-21.0 U/mL
20.1
18.9
18.7
17.4


Assay



Total Impurities:
1.3%
3.5%
7.1%
10.2%



NMT 17.0%
















TABLE 23







Samples stored inverted at 40° C.










Acceptance
Time in months












Test
Criteria
Initial
1
2
3





Vasopressin
18.0-21.0 U/mL
19.9
19.2
18.3
17.4


Assay



Total Impurities:
1.5%
3.7%
6.3%
10.3%



NMT 17.0%
















TABLE 24







Samples stored Upright at 40° C.










Acceptance
Time in months












Test
Criteria
Initial
1
2
3





Vasopressin
18.0-21.0 U/mL
19.9
19.2
18.3
17.5


Assay



Total Impurities:
1.5%
3.8%
6.3%
10.5%



NMT 17.0%









The results of the above experiments suggested that storage in either an upright or inverted position did not markedly affect the stability of vasopressin. The samples held at 5° C. exhibited little fluctuation in vasopressin amounts over 24 months, and the amount of total impurities did not increase above 3% during the testing period (TABLES 7-12). The samples held at 25° C. and 60% relative humidity exhibited a decrease in vasopressin amount of about 10-12% after 24 months (TABLES 13-18). The amount of impurities observed in the samples stored at 25° C. and 60% relative humidity after 24 months exceeded 13% in some samples, whereas the amount of impurities observed in the samples stored at 5° C. did not exceed 3% after 24 months. After about three months, the samples held at 40° C. exhibited a decrease in the amount of vasopressin of about 10-12%. The amount of impurities observed at 40° C. exceeded 10% after three months, whereas the amount of impurities observed in the samples stored at 5° C. was less than 3% after three months (TABLES 19-24).


Experiments were also conducted on the same samples above over the course of the experiments to measure the amount of individual impurities in the samples, pH of the samples, chlorobutanol content, particulate matter, antimicrobial effectiveness, and bacterial endotoxin levels (TABLES 25-42). (NR=no reading; ND=not determined; UI=unidentified impurity).


The anti-microbial effectiveness of the solution was established to determine the amount of antimicrobial agents in the formulation that protect against bacterial contamination. The bullets in the tables below indicate that the sample was not tested for anti-microbial effectiveness at that specific time point.


The bacterial endotoxin levels were also measured for some of the formulations. The bullets in the tables below indicate that the sample was not tested for bacterial endotoxin levels at that specific time point.









TABLE 25







Samples stored inverted at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24





Vasopressin
16.0-21.0
 19.4
 19.4
 19.4
 19.3
 19.5
 19.4
 19.5
 19.4
 19.3


Assay
U/mL


Related
SEQ ID
0.5%
0.5%
0.6%
0.6%
0.6%
0.6%
0.7%
0.8%
0.9%


Substances
NO.: 2



NMT 6.0%



SEQ ID
0.6%
0.6%
0.6%
0.7%
0.7%
0.7%
0.8%
0.9%
1.0%



NO.: 4:



NMT 6.0%



SEQ ID
0.3%
0.3%
0.3%
0.4%
0.3%
0.3%
0.4%
0.4%
0.3%



NO.: 10:



NMT 1.0%



Asp5-AVP:
0.1%
0.1%
0.1%
0.1%
0.1%
0.1%
0.2%
0.2%
0.2%



NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



Acetyl-AVP:
0.3%
0.2%
0.3%
0.3%
0.2%
0.2%
0.3%
0.3%
0.3%



NMT 1.0%



UI-0.84:
NR
NR
0.1%
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.03:
0.2%
0.2%
0.2%
0.3%
0.2%
0.2%
0.3%
0.3%
0.2%



NMT 1.0%



UI-1.67:
NR
NR
NR
NR
NR
NR
NR
NR
0.2%



NMT 1.0%



UI-1.85:
0.2%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-2.05:
0.1%
NR
0.1%
NR
NR
NR
NR
NR
NR



NMT 1.0%




Total
2.3%
2.0%
2.1%
2.3%
2.2%
2.3%
2.6%
2.9%
2.9%



Impurities:



NMT 17.0%


pH
2.5-4.5
  3.5
  3.5
  3.5
  3.5
  3.5
  3.5
  3.5
  3.8
  3.5


Chlorobutanol
0.25-0.60%
0.48% 
0.49% 
0.48% 
0.48% 
0.47% 
0.48% 
0.48% 
0.49% 
0.49% 



w/v


Particulate
NMT 6000
0
1
1
1
2
16 
2
4
1


Matter (USP)
(≥10 μm)



NMT 600
0
0
0
0
0
0
0
0
0



(≥25 μm)


Anti-
Meets Test











Microbial


Effectiveness


Bacterial
NMT 29











Endotoxin
EU/mL
















TABLE 26







Samples stored inverted at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24





Vasopressin
16.0-21.0
 19.7
 19.7
 19.7
 19.7
 19.9
 19.7
 19.8
 19.7
 19.5


Assay
U/mL


Related
SEQ ID
0.6%
0.5%
0.5%
0.6%
0.5%
0.6%
0.7%
0.8%
0.8%


Substances
NO.: 2:



NMT 6.0%



SEQ ID
0.6%
0.6%
0.6%
0.6%
0.6%
0.7%
0.7%
0.8%
0.9%



NO.: 4:



NMT 6.0%



SEQ ID
0.3%
0.3%
0.3%
0.4%
0.3%
0.4%
0.4%
0.4%
0.3%



NO.: 10:



NMT 1.0%



Asp5-AVP:
0.1%
0.1%
0.1%
0.1%
0.1%
0.1%
0.2%
0.2%
0.2%



NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



Acetyl-AVP:
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%



NMT 1.0%



UI-0.75-0.78:
0.2%
0.2%
0.2%
0.2%
NR
0.1%
0.2%
0.2%
0.2%



NMT 1.0%



UI-0.83-0.84:
0.1%
0.1%
0.1%
NR
0.1%
NR
NR
NR
NR



NMT 1.0%



UI-1.02-1.03:
0.2%
0.2%
0.2%
0.3%
0.2%
0.2%
0.3%
0.3%
0.3%



NMT 1.0%



UI-1.67:
NR
NR
NR
NR
NR
NR
NR
NR
0.2%



NMT 1.0%



UI-1.85:
0.2%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-2.05:
0.2%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%




Total
2.7%
2.2%
2.3%
2.4%
2.1%
2.3%
2.7%
2.9%
2.9%



Impurities:



NMT 17.0%


pH
2.5-4.5
  3.6
  3.6
  3.6
  3.6
  3.6
  3.6
  3.6
  3.6
  3.6


Chlorobutanol
0.25-0.60%
0.48% 
0.48% 
0.48% 
0.47% 
0.48% 
0.48% 
0.49% 
0.48% 
0.49% 



w/v


Particulate
NMT 6000
1
1
1
1
1
15 
2
3
2


Matter (USP)
(≥10 μm)



NMT 600
0
0
0
0
0
0
0
0
0



(≥25 μm)


Anti-
Meets Test











Microbial


Effectiveness


Bacterial
NMT 29











Endotoxin
EU/mL
















TABLE 27







Samples stored inverted at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24





Vasopressin
16.0-21.0
 19.7
 19.7
 19.6
 19.7
 19.8
 19.7
 19.9
 19.8
 19.5


Assay
U/mL


Related
SEQ ID
0.5%
0.5%
0.5%
0.5%
0.5%
0.6%
0.6%
0.8%
0.8%


Substances
NO.: 2:



NMT 6.0%



SEQ ID
0.5%
0.5%
0.5%
0.6%
0.6%
0.7%
0.7%
0.8%
0.9%



NO.: 4:



NMT 6.0%



SEQ ID
0.3%
0.3%
0.3%
0.4%
0.3%
0.3%
0.4%
0.4%
0.3%



NO.: 10:



NMT 1.0%



Asp5-AVP:
0.1%
0.1%
0.1%
0.1%
0.1%
0.1%
0.1%
0.2%
0.2%



NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



Acetyl-AVP:
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%



NMT 1.0%



UI-0.75-0.78:
NR
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-0.83-0.84:
NR
NR
0.1%
NR
NR
NR
NR
NR
0.1%



NMT 1.0%



UI-1.02-1.03:
0.2%
0.2%
0.2%
0.3%
0.2%
0.2%
0.3%
0.3%
0.2%



NMT 1.0%



UI-1.67:
NR
NR
NR
NR
NR
NR
NR
NR
0.2%



NMT 1.0%



UI-1.76:
NR
NR
NR
0.1%
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.85:
0.2%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-2.05:
0.1%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%




Total
2.2%
1.9%
2.0%
2.2%
2.0%
2.1%
2.4%
2.6%
2.8%



Impurities:



NMT 17.0%


pH
2.5-4.5
  3.6
  3.5
  3.6
  3.5
  3.5
  3.5
  3.6
  3.5
  3.5


Chlorobutanol
0.25-0.60%
0.47% 
0.48% 
0.47% 
0.47% 
0.47% 
0.47% 
0.48% 
0.48% 
0.48% 



w/v


Particulate
NMT 6000
1
2
1
2
1
4
2
1
3


Matter (USP)
(≥10 μm)



NMT 600
0
0
0
0
0
0
0
0
0



(≥25 μm)


Anti-
Meets Test











Microbial


Effectiveness


Bacterial
NMT 29











Endotoxin
EU/mL
















TABLE 28







Samples stored upright at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24




















Vasopressin
16.0-21.0 U/mL
19.4
19.5
19.4
19.4
19.5
19.5
19.5
19.4
19.3


Assay


Related
SEQ ID
0.5%
0.6%
0.6%
0.6%
0.6%
0.6%
0.7%
0.8%
0.9%


Substances
NO.: 2:



NMT 6.0%



SEQ ID
0.6%
0.6%
0.6%
0.7%
0.7%
0.7%
0.7%
0.9%
1.0%



NO.: 4:



NMT 6.0%



SEQ ID
0.3%
0.3%
0.3%
0.4%
0.3%
0.3%
0.4%
0.4%
0.3%



NO.: 10:



NMT 1.0%



Asp5-AVP:
0.1%
0.1%
0.1%
0.1%
0.1%
0.1%
0.2%
0.2%
0.2%



NMT 1.5%



AVP-
NR
NR
NR
NR
NR
NR
NR
NR
NR



Dimer:



NMT 1.0%



Acetyl-
0.3%
0.3%
0.3%
0.2%
0.2%
0.3%
0.3%
0.3%
0.3%



AVP: NMT



1.0%



UI-0.84:
NR
NR
0.1%
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.03:
0.2%
0.2%
0.2%
0.3%
0.2%
0.2%
0.3%
0.3%
0.2%



NMT 1.0%



UI-1.67:
NR
NR
NR
NR
NR
NR
NR
NR
0.2%



NMT 1.0%



UI-1.85:
0.2%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-2.05:
0.1%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%




Total
2.3%
2.1%
2.1%
2.3%
2.1%
2.3%
2.5%
2.9%
2.9%



Impurities:



NMT 17.0%


pH
2.5-4.5
3.5
3.5
3.5
3.5
3.5
3.5
3.5
3.8
3.5


Chlorobutanol
0.25-0.60%
0.48% 
0.48% 
0.48% 
0.48% 
0.48% 
0.48% 
0.48% 
0.49% 
0.49% 



w/v


Particulate
NMT 6000
0
2
2
2
1
2
2
4
1


Matter (USP)
(≥10 μm)



NMT 600 (≥25 μm)
0
0
0
0
0
0
0
0
0


Anti-
Meets Test











Microbial


Effectiveness


Bacterial
NMT 29 EU/mL











Endotoxin
















TABLE 29







Samples stored upright at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24




















Vasopressin
16.0-21.0 U/mL
19.7
19.7
19.6
19.7
19.8
19.7
19.8
19.7
19.5


Assay


Related
SEQ ID
0.6%
0.5%
0.5%
0.5%
0.6%
0.6%
0.6%
0.8%
0.7%


Substances
NO.: 2:



NMT 6.0%



SEQ ID
0.6%
0.6%
0.6%
0.6%
0.6%
0.7%
0.7%
0.8%
0.8%



NO.: 4:



NMT 6.0%



SEQ ID
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%
0.4%
0.4%
0.3%



NO.: 10:



NMT 1.0%



Asp5-AVP:
0.1%
0.1%
0.1%
0.1%
0.1%
0.1%
0.2%
0.2%
0.2%



NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



Acetyl-
0.3%
0.3%
0.3%
0.3%
0.2%
0.3%
0.3%
0.3%
0.3%



AVP: NMT



1.0%



UI-0.75-0.78:
0.2%
0.2%
NR
0.2%
0.2%
0.2%
0.2%
0.2%
0.2%



NMT



1.0%



UI-0.83-0.84:
0.1%
NR
0.1%
NR
NR
NR
NR
NR
NR



NMT



1.0%



UI-1.02-1.03:
0.2%
0.2%
0.2%
0.2%
0.2%
0.2%
0.3%
0.3%
0.2%



NMT



1.0%



UI-1.67:
NR
NR
NR
0.2%
NR
NR
NR
NR
0.2%



NMT 1.0%



UI-1.85:
0.2%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-2.05:
0.2%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%




Total
2.7%
2.1%
2.2%
2.2%
2.2%
2.3%
2.6%
2.9%
2.8%



Impurities:



NMT 17.0%


pH
2.5-4.5
3.6
3.6
3.6
3.6
3.6
3.6
3.6
3.6
3.6


Chlorobutanol
0.25-0.60%
0.48% 
0.48% 
0.48% 
0.48% 
0.48% 
0.48% 
0.49% 
0.49% 
0.49% 



w/v


Particulate
NMT 6000
1
1
1
2
2
6
4
4
1


Matter (USP)
(≥10 μm)



NMT 600 (≥25 μm)
0
0
0
0
0
0
0
0
0


Anti-
Meets Test











Microbial


Effectiveness


Bacterial
NMT 29 EU/mL











Endotoxin
















TABLE 30







Samples stored upright at 5° C.










Acceptance
Time in months

















Test
Criteria
Initial
1
2
3
6
9
12
18
24




















Vasopressin
16.0-21.0 U/mL
19.7
19.7
19.6
19.7
19.8
19.7
19.9
19.8
19.5


Assay


Related
SEQ ID
0.5%
0.5%
0.5%
0.5%
0.5%
0.6%
0.6%
0.8%
0.8%


Substances
NO.: 2:



NMT 6.0%



SEQ ID
0.5%
0.5%
0.5%
0.6%
0.6%
0.7%
0.7%
0.8%
0.9%



NO.: 4:



NMT 6.0%



SEQ ID
0.3%
0.3%
0.3%
0.4%
0.3%
0.3%
0.4%
0.4%
0.3%



NO.: 10:



NMT 1.0%



Asp5-AVP:
0.1%
NR
0.1%
0.1%
0.1%
0.1%
0.2%
0.2%
0.2%



NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



Acetyl-
0.3%
0.3%
0.3%
0.3%
0.2%
0.3%
0.3%
0.3%
0.3%



AVP: NMT



1.0%



UI-0.75-0.78:
NR
NR
NR
NR
0.2%
NR
NR
NR
NR



NMT



1.0%



UI-0.83-0.84:
NR
NR
0.1%
NR
NR
NR
NR
0.1%
NR



NMT



1.0%



UI-1.02-1.03:
0.2%
0.2%
0.2%
0.3%
0.2%
0.2%
0.3%
0.3%
0.2%



NMT



1.0%



UI-1.67:
NR
NR
NR
NR
NR
NR
NR
NR
0.2%



NMT 1.0%



UI-1.76:
NR
NR
NR
0.1%
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.85:
0.2%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-2.05:
0.1%
NR
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%




Total
2.2%
1.8%
2.0%
2.2%
2.2%
2.1%
2.4%
2.8%
2.7%



Impurities:



NMT 17.0%


pH
2.5-4.5
3.6
3.5
3.6
3.5
3.5
3.5
3.6
3.5
3.5


Chlorobutanol
0.25-0.60%
0.47% 
0.48% 
0.47% 
0.47% 
0.48% 
0.47% 
0.48% 
0.48% 
0.48% 



w/v


Particulate
NMT 6000
1
1
1
1
1
3
2
1
3


Matter (USP)
(≥10 μm)



NMT 600 (≥25 μm)
0
0
0
0
0
0
0
0
0


Anti-
Meets Test











Microbial


Effectiveness


Bacterial
NMT 29 EU/mL











Endotoxin
















TABLE 31







Samples stored inverted at 25° C. and 60% Relative Humidity










Acceptance
Time in months
















Test
Criteria
Initial
3
6
9
12
18
24
30



















Vasopressin
16.0-21.0 U/mL
19.8
19.4
19.1
18.8
18.3
17.5
17.3



Assay


Related
SEQ ID
0.1%
0.5%
1.1%
1.6%
2.0%
3.3%
4.6%



Substances
NO.: 2:



NMT 6.0%



SEQ ID
0.1%
0.6%
1.2%
1.8%
2.2%
3.7%
5.2%




NO.: 4:



NMT 6.0%



SEQ ID
0.3%
0.4%
0.5%
0.5%
0.4%
0.2%
0.3%




NO.: 10:



NMT 1.0%



Asp5-AVP:
NR
0.1%
0.3%
0.4%
0.5%
0.7%
1.0%




NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR




NMT 1.0%



Acetyl-
0.3%
0.3%
0.3%
0.2%
0.2%
0.2%
0.3%




AVP: NMT



1.0%



UI-0.83:
NR
NR
<0.10
NR
NR
NR
0.1%




NMT 1.0%



UI-0.99:
NR
NR
NR
NR
0.1%
NR
NR




NMT 1.0%



UI-1.03:
0.2%
0.2%
0.3%
0.3%
0.3%
0.2%
0.2%




NMT 1.0%



UI-1.14:
NR
NR
NR
NR
NR
NR
0.1%




NMT 1.0%



UI-1.18:
NR
NR
NR
NR
NR
0.1%
0.3%




NMT 1.0%



UI-1.20:
NR
NR
NR
NR
NR
NR
0.1%




NMT 1.0%



UI-1.22:
NR
NR
NR
NR
NR
NR
0.1%




NMT 1.0%



UI-1.56-1.57:
NR
NR
<0.10
0.1%
0.1%
0.2%
0.2%




NMT



1.0%



UI-1.60:
NR
NR
NR
0.1%
0.1%
0.2%
NR




NMT 1.0%



UI-1.74:
NR
NR
NR
NR
NR
0.2%
NR




NMT 1.0%



UI-1.85-1.88:
NR
0.2%
NR
NR
NR
0.1%
0.1%




NMT



1.0%



UI-2.09-2.10:
NR
0.2%
NR
NR
NR
NR
0.4%




NMT



1.0%



UI-2.15-2.16:
NR
NR
0.1%
NR
NR
NR
0.5%




NMT



1.0%




Total
1.1%
2.4%
3.7%
4.7%
5.9%
9.0%
13.6% 




Impurities:



NMT 17.0%


pH
2.5-4.5
3.5
3.5
3.5
3.5
3.4
3.3
3.2



Chlorobutanol
0.25-0.60%
0.49% 
0.48% 
0.48% 
0.47% 
0.47% 
0.48% 
0.47




w/v


Particulate
NMT 6000
1
1
1
1
8
4
1



Matter (USP)
(≥10 μm)



NMT 600 (≥25 μm)
0
0
0
0
0
0
0



AntiMicrobial
Meets Test
Pass
·
·
·
Pass

Pass



Effectiveness


Bacterial
NMT 29 EU/mL
<1
·
·
·
<1

<1



Endotoxin
















TABLE 32







Samples stored inverted at 25° C. and 60% Relative Humidity










Acceptance
Time in months
















Test
Criteria
Initial
3
6
9
12
18
24
30



















Vasopressin
16.0-21.0 U/mL
20.1
19.7
19.3
19
18.6
17.6
17.6



Assay


Related
SEQ ID NO.:
0.1%
0.5%
0.9%
1.5%
1.9%
3.1%
4.4%



Substances
2: NMT 6.0%



SEQ ID NO.:
0.1%
0.5%
1.1%
1.6%
2.2%
3.4%
4.9%




4: NMT 6.0%



SEQ ID NO.:
0.3%
0.4%
0.3%
0.4%
0.3%
0.4%
0.3%




10:



NMT 1.0%



Asp5-AVP:
NR
0.1%
0.2%
0.3%
0.4%
0.7%
0.9%




NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR




NMT 1.0%



Acetyl-AVP:
0.3%
0.3%
0.3%
0.2%
0.2%
0.2%
0.3%




NMT 1.0%



UI-0.75-0.76:
NR
0.2%
0.2%
0.2%
0.2%
0.2%
0.2%




NMT 1.0%



UI-0.83:
0.2%
NR
0.1%
NR
NR
0.1%
0.1%




NMT 1.0%



UI-0.99:
NR
NR
NR
NR
0.1%
NR
NR




NMT 1.0%



UI-1.02-1.03:
0.2%
0.2%
0.2%
0.3%
0.2%
0.3%
0.2%




NMT 1.0%



UI-1.14:
NR
NR
NR
NR
NR
NR
0.1%




NMT 1.0%



UI-1.18:
NR
NR
NR
NR
NR
0.1%
0.3%




NMT 1.0%



UI-1.20:
NR
NR
NR
NR
NR
NR
0.1%




NMT 1.0%



UI-1.22:
NR
NR
NR
NR
NR
NR
0.1%




NMT 1.0%



UI-1.56-1.57:
NR
NR
0.1%
0.1%
0.2%
0.2%
0.2%




NMT 1.0%



UI-1.60:
NR
NR
0.1%
0.1%
0.2%
0.2%
NR




NMT 1.0%



UI-1.74:
NR
NR
NR
NR
NR
0.2%
NR




NMT 1.0%



UI-1.85-1.88:
NR
0.2%
NR
NR
NR
0.1%
0.1%




NMT 1.0%



UI-2.09-2.10:
NR
0.2%
NR
NR
NR
NR
0.4%




NMT 1.0%



UI-2.15-2.16:
NR
NR
NR
NR
NR
NR
0.6%




NMT 1.0%




Total
1.3%
2.5%
3.4%
4.6%
5.6%
9.0%
13.4% 




Impurities:



NMT 17.0%


pH
2.5-4.5
3.6
3.6
3.5
3.5
3.2
3.3
3.4



Chlorobutanol
0.25-0.60%
0.48% 
0.49% 
0.48% 
0.47% 
0.47% 
0.47% 
0.47




w/v


Particulate
NMT 6000
2
1
1
3
4
1
2



Matter
(≥10 μm)


(USP)
NMT 600 (≥25 μm)
0
0
0
0
0
0
0



AntiMicrobial
Meets Test
Pass



Pass

Pass



Effectiveness


Bacterial
NMT 29 EU/mL
<1



<1

<1



Endotoxin
















TABLE 33







Samples stored inverted at 25° C. and 60% Relative Humidity










Acceptance
Time in months
















Test
Criteria
Initial
3
6
9
12
18
24
30



















Vasopressin
16.0-21.0 U/mL
19.9
19.6
19.2
19
18.7
18
17.4



Assay


Related
SEQ ID NO.:
0.2%
0.5%
1.0%
1.5%
2.0%
3.2%
4.5%



Substances
2: NMT 6.0%



SEQ ID NO.:
0.1%
0.6%
1.1%
1.8%
2.2%
3.7%
5.0%




4: NMT 6.0%



SEQ ID NO.:
0.4%
0.4%
0.3%
0.4%
0.4%
0.3%
0.5%




10:



NMT 1.0%



Asp5-AVP:
NR
0.1%
0.2%
0.4%
0.5%
0.7%
1.0%




NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
0.1%




NMT 1.0%



Acetyl-AVP:
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%




NMT 1.0%



UI-0.12:
NR
0.1%
NR
NR
NR
NR
NR



NMT 1.0%



UI-0.75-0.76:
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-0.83-0.84:
NR
0.1%
0.1%

0.1%
0.1%
0.1%



NMT 1.0%



UI-0.93:
NR
NR
NR
NR
NR
NR
0.1%



NMT 1.0%



UI-0.99:
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.02-1.03:
0.3%
0.2%
0.2%
0.3%
0.3%
0.3%
0.3%




NMT 1.0%



UI-1.15:
NR
NR
NR
NR
NR
NR
0.1%



NMT 1.0%



UI-1.18:
NR
NR
NR
NR
NR
0.1%
0.3%



NMT 1.0%



UI-1.20:
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.22:
NR
NR
NR
NR
NR
NR
0.1%



NMT 1.0%



UI-1.26:
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.35:
0.3%
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.56-1.57:
NR
NR
0.1%
NR
0.1%
0.2%
0.3%



NMT 1.0%



UI-1.60:
NR
NR
0.1%
NR
0.1%
NR
NR




NMT 1.0%



UI-1.74:
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.84-1.89:
NR
0.1%
NR
NR
NR
NR
0.2%



NMT 1.0%



UI-1.96:
0.2%
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-2.09-2.10:
NR
20.0% 
NR
NR
NR
<0.10
0.1%



NMT 1.0%



UI-2.15-2.16:
NR
NR
0.1%
NR
NR
0.1%
NR



NMT 1.0%




Total
1.5%
2.6%
3.3%
4.6%
5.9%
9.0%
12.9% 




Impurities:



NMT 17.0%


pH
2.5-4.5
3.6
3.5
3.5
3.5
3.4
3.4
3.3



Chlorobutanol
0.25-0.60%
0.48% 
0.47% 
0.47% 
0.46% 
0.46% 
0.46% 
0.45% 




w/v


Particulate
NMT 6000
1
2
3
3
3
1
2



Matter
(≥10 μm)


(USP)
NMT 600 (≥25 μm)
0
0
0
0
0
0
0



Anti-
Meets Test
Pass



Pass

Pass



Microbial


Effectiveness


Bacterial
NMT 29 EU/mL
<1



<1

<1



Endotoxin
















TABLE 34







Samples stored upright at 25° C. and 60% Relative Humidity










Acceptance
Time in months
















Test
Criteria
Initial
3
6
9
12
18
24
30



















Vasopressin
16.0-21.0 U/mL
19.8
19.4
19.1
18.8
18.3
17.5
17.4



Assay


Related
SEQ ID NO.:
0.1%
0.5%
1.1%
1.6%
2.0%
3.2%
4.5%



Substances
2: NMT 6.0%



SEQ ID NO.:
0.1%
0.6%
1.2%
1.8%
2.3%
3.6%
5.0%




4: NMT 6.0%



SEQ ID NO.:
0.3%
0.4%
0.3%
0.4%
0.3%
0.2%
0.3%




10:



NMT 1.0%



Asp5-AVP:
NR
0.1%
0.2%
0.4%
0.4%
0.7%
0.9%




NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR




NMT 1.0%



Acetyl-AVP:
0.3%
0.3%
0.3%
0.2%
0.2%
0.2%
0.3%




NMT 1.0%



UI-0.83:
NR
NR
<0.10
NR
NR
0.1%
0.1%




NMT 1.0%



UI-0.99:
NR
NR
NR
NR
NR
NR
NR




NMT 1.0%



UI-1.03:
0.2%
0.2%
0.2%
0.3%
0.2%
0.2%
0.2%




NMT 1.0%



UI-1.14:
NR
NR
NR
NR
NR
NR
0.1%




NMT 1.0%



UI-1.18:
NR
NR
NR
NR
NR
0.1%
0.3%




NMT 1.0%



UI-1.20:
NR
NR
NR
NR
NR
NR
0.1%




NMT 1.0%



UI-1.22:
NR
NR
NR
NR
NR
NR
NR




NMT 1.0%



UI-1.56-1.57:
NR
NR
NR
0.1%
0.1%
0.2%
0.2%




NMT 1.0%



UI-1.60:
NR
NR
NR
NR
NR
0.1%
NR




NMT 1.0%



UI-1.74:
NR
NR
NR
NR
NR
0.2%
NR




NMT 1.0%



UI-1.85-1.88:
NR
0.2%
NR
NR
NR
0.1%
0.1%




NMT 1.0%



UI-2.09-2.10:
NR
0.2%
NR
NR
NR
NR
0.3%




NMT 1.0%



UI-2.15-2.16:
NR
NR
NR
NR
NR
NR
0.5%




NMT 1.0%




Total
1.1%
2.4%
3.2%
4.8%
5.6%
9.2%
13.1% 




Impurities:



NMT 17.0%


pH
2.5-4.5
3.5
3.5
3.5
3.5
3.4
3.3
3.3



Chlorobutanol
0.25-0.60%
0.49% 
0.48% 
0.48% 
0.48% 
0.47% 
0.48% 
0.47




w/v


Particulate
NMT 6000
1
2
2
2
2
4
2



Matter
(≥10 μm)


(USP)
NMT 600 (≥25 μm)
0
0
0
0
0
0
0



AntiMicrobial
Meets Test
Pass



Pass

Pass



Effectiveness


Bacterial
NMT 29 EU/mL
<1



<1

<1



Endotoxin
















TABLE 35







Samples stored upright at 25° C. and 60% Relative Humidity










Acceptance
Time in months
















Test
Criteria
Initial
3
6
9
12
18
24
30



















Vasopressin
16.0-21.0 U/mL
20.1
19.7
19.4
18.9
18.6
17.8
17.7



Assay


Related
SEQ ID
0.1%
0.5%
0.9%
1.4%
1.9%
3.1%
4.3%



Substances
NO.: 2:



NMT 6.0%



SEQ ID
0.1%
0.5%
1.1%
1.6%
2.2%
3.4%
4.9%




NO.: 4:



NMT 6.0%



D-Asn-
0.3%
0.4%
0.3%
0.3%
0.3%
0.4%
0.3%




AVP:



NMT 1.0%



Asp5-AVP:
NR
0.1%
0.2%
0.3%
0.4%
0.7%
0.9%




NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR




NMT 1.0%



Acetyl-
0.30% 
0.30% 
0.30% 
0.20% 
0.20% 
0.20% 
0.3%




AVP: NMT



1.0%



UI-0.75-0.76:
NR
0.2%
0.2%
0.2%
0.2%
0.2%
0.2%



NMT



1.0%



UI-0.83:
0.2%
NR
<0.10
NR
NR
0.1%
0.1%



NMT 1.0%



UI-0.99:
NR
NR
NR
NR
0.1%
NR
NR



NMT 1.0%



UI-1.02-1.03:
0.2%
0.2%
0.2%
0.2%
0.2%
0.3%
0.2%




NMT



1.0%



UI-1.14:
NR
NR
NR
NR
NR
NR
0.1%



NMT 1.0%



UI-1.18:
NR
NR
NR
NR
NR
0.1%
0.3%



NMT 1.0%



UI-1.20:
NR
NR
NR
NR
NR
NR
0.1%



NMT 1.0%



UI-1.22:
NR
NR
NR
NR
NR
NR
0.4%



NMT 1.0%



UI-1.56-1.57:
NR
NR
0.1%
0.1%
0.2%
0.2%
0.3%



NMT



1.0%



UI-1.60:
NR
NR
0.1%
0.1%
0.2%
0.2%
NR




NMT 1.0%



UI-1.74:
NR
NR
NR
NR
NR
0.2%
NR



NMT 1.0%



UI-1.85-1.88:
NR
0.2%
NR
NR
NR
0.1%
0.1



NMT



1.0%



UI-2.09-2.10:
NR
0.2%
NR
NR
NR
0.1%
0.3



NMT



1.0%



UI-2.15-2.16:
NR

NR
NR
NR
NR
0.5



NMT



1.0%




Total
1.3%
2.5%
3.3%
4.5%
5.7%
9.1%
13.3% 




Impurities:



NMT 17.0%


pH
2.5-4.5
3.6
3.6
3.5
3.5
3.4
3.3
3.3



Chlorobutanol
0.25-0.60%
0.48% 
0.49% 
0.48% 
0.47% 
0.47% 
0.48% 
0.46




w/v


Particulate
NMT 6000
2
1
1
2
5
1
4



Matter (USP)
(≥10 μm)



NMT 600 (≥25 μm)
0
0
0
0
0
0
0



Anti-
Meets Test
Pass



Pass

Pass



Microbial


Effectiveness


Bacterial
NMT 29 EU/mL
<1



<1

<1



Endotoxin
















TABLE 36







Samples stored upright at 25° C. and 60% Relative Humidity










Acceptance
Time in months
















Test
Criteria
Initial
3
6
9
12
18
24
30





Vasopressin
16.0-21.0
 19.9
 19.6
 19.2
19 
 18.5
 18.1
 17.4



Assay
U/mL


Related
SEQ ID
0.2%
0.5%
1.0%
1.5%
2.1%
3.3%
4.7%



Substances
NO.: 2:



NMT 6.0%



SEQ ID
0.1%
0.6%
1.1%
1.7%
2.3%
3.7%
5.3%




NO.: 4:



NMT 6.0%



D-Asn-AVP:
0.4%
0.4%
0.3%
0.4%
0.4%
0.3%
0.5%




NMT 1.0%



Asp5-AVP:
NR
0.1%
0.2%
0.3%
0.5%
0.7%
1.0%




NMT 1.5%



AVP-Dimer:
NR
NR
NR
NR
NR
NR
NR




NMT 1.0%



Acetyl-AVP:
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%
0.3%




NMT 1.0%



UI-0.12:
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-0.75-0.76:
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-0.83-0.84:
NR
0.1%
0.1%
0.1%
NR
0.1%
0.1%



NMT 1.0%



UI-0.93:
NR
NR
NR
NR
NR
NR
0.1%



NMT 1.0%



UI-0.99:
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.02-1.03:
0.3%
0.2%
0.2%
0.3%
0.3%
0.3%
0.3%




NMT 1.0%



UI-1.15:
NR
NR
NR
NR
NR
NR
0.2%



NMT 1.0%



UI-1.18:
NR
NR
NR
NR
NR
0.1%
0.3%



NMT 1.0%



UI-1.20:
NR
NR
NR
NR
NR
NR
0.1%



NMT 1.0%



UI-1.22:
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.26:
NR
NR
0.4%
NR
NR
NR
NR



NMT 1.0%



UI-1.35:
0.1%
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.56-1.57:
NR
NR
0.1%
0.1%
NR
0.2%
0.3%



NMT 1.0%



UI-1.60:
NR
NR
NR
NR
NR
NR
NR




NMT 1.0%



UI-1.74:
NR
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-1.84-1.89:
NR
0.1%
NR
NR
NR
NR
0.2%



NMT 1.0%



UI-1.96:
0.2%
NR
NR
NR
NR
NR
NR



NMT 1.0%



UI-2.09-2.10:
NR
NR
NR
NR
NR
<0.10
NR



NMT 1.0%



UI-2.15-2.16:
NR
NR
0.1%
NR
NR
0.2%
NR



NMT 1.0%




Total
1.5%
2.5%
3.7%
4.7%
5.9%
9.1%
13.3% 




Impurities:



NMT 17.0%


pH
2.5-4.5
  3.6
  3.5
  3.5
  3.5
  3.4
  3.4
  3.3



Chlorobutanol
0.25-0.60%
0.48% 
0.48% 
0.47% 
0.47% 
0.46% 
  0.45
  0.46




w/v


Particulate
NMT 6000
1
0
1
3
7
0
3



Matter (USP)
(≥10 μm)



NMT 600
0
0
0
0
0
0
0




(≥25 μm)


Anti-Microbial
Meets Test
Pass



Pass

Pass



Effectiveness


Bacterial
NMT 29
<1 



<1 

<1 



Endotoxin
EU/mL
















TABLE 37







Samples stored inverted at 40° C.










Acceptance
Time in months












Test
Criteria
Initial
1
2
3















Vasopressin
18.0-21.0 U/mL
19.8
19.1
18.6
17.3


Assay


Related
SEQ ID NO.: 2:
0.1%
1.0%
2.4%
3.8%


Substances
NMT 6.0%



SEQ ID NO.: 4:
0.1%
1.1%
2.7%
4.3%



NMT 6.0%



D-Asn-AVP:
0.3%
0.4%
0.3%
0.3%



NMT 1.0%



Asp5-AVP: NMT
ND
0.2%
0.5%
0.8%



1.5%



AVP-Dimer:
ND
ND
ND
ND



NMT 1.0%



Acetyl-AVP:
0.3%
0.2%
0.2%
0.2%



NMT 1.0%



UI-0.13: NMT
ND
0.1%
ND
ND



1.0%



UI-0.75-0.78:
ND
ND
ND
ND



NMT 1.0%



UI-0.83-0.84:
ND
ND
ND
ND



NMT 1.0%



UI-1.02-1.03:
0.2%
0.3%
0.2%
0.3%



NMT 1.0%



UI-1.18: NMT
ND
ND
ND
0.2%



1.0%



UI-1.56-1.57:
ND
0.2%
0.4%
0.4%



NMT 1.0%



UI-1.67: NMT
ND
ND
ND
ND



1.0%



UI-1.76: NMT
ND
ND
ND
ND



1.0%



UI-1.83-1.85:
ND
ND
0.2%
0.2%



NMT 1.0%



UI-1.87-1.88:
ND
ND
0.2%
0.2%



NMT 1.0%



UI-1.93: NMT
ND
0.1%
ND
ND



1.0%



UI-2.05-2.08:
ND
ND
0.2%
ND



NMT 1.0%




Total Impurities:
1.1%
3.7%
7.3%
10.6% 



NMT 17.0%


pH
2.5-4.5
3.5
3.3
3.2
3.1


Chlorobutanol
0.25-0.60% w/v
0.49% 
0.48% 
0.50% 
0.47% 


Particulate
NMT 6000
1
1
1
1


Matter (USP)
(≥10 μm)



NMT 600
0
0
0
0



(≥25 μm)
















TABLE 38







Samples stored inverted at 40° C.











Acceptance

Time in months












Test
Criteria
Initial
1
2
3















Vasopressin
18.0-21.0 U/mL
20.1
19.3
18.7
17.6


Assay







Related
SEQ ID NO.: 2:
 0.1%
 0.9%
 2.2%
 3.6%


Substances
NMT 6.0%







SEQ ID NO.: 4:
 0.1%
 1.0%
 2.5%
 3.9%



NMT 6.0%







D-Asn-AVP:
 0.3%
 0.4%
 0.3%
 0.3%



NMT 1.0%







Asp5-AVP: NMT
ND
 0.2%
 0.5%
 0.8%



1.5%







AVP-Dimer:
ND
ND
ND
ND



NMT 1.0%







Acetyl-AVP:
 0.3%
 0.2%
 0.3%
 0.2%



NMT 1.0%







UI-0.13: NMT
ND
0.1%
ND
ND



1.0%







UI-0.75-0.78:
ND
ND
 0.2%
 0.2%



NMT 1.0%







UI-0.80-0.84:
 0.2%
 0.2%
ND
ND



NMT 1.0%







UI-1.02-1.03:
 0.2%
 0.3%
 0.2%
 0.3%



NMT 1.0%







UI-1.18: NMT
ND
ND
 0.3%
 0.2%



1.0%







UI-1.56-1.57:
ND
 0.2%
ND
 0.4%



NMT 1.0%







UI-1.67: NMT
ND
ND
ND
ND



1.0%







UI-1.76: NMT
ND
ND
ND
ND



1.0%







UI-1.81-1.85:
ND
ND
 0.2%
 0.2%



NMT 1.0%







UI-1.87-1.88:
ND
ND
 0.2%
 0.2%



NMT 1.0%







UI-1.93: NMT
ND
 0.1%
ND
ND



1.0%







UI-2.03-2.08:
ND
ND
 0.2%
 0.1%



NMT 1.0%







UI-2.14: NMT
ND
ND
 0.2%
ND



1.0%







Total Impurities:
 1.3%
 3.6%
 7.3%
10.3%



NMT 17.0%






pH
2.5-4.5
3.6
3.3
3.2
3.1


Chlorobutanol
0.25-0.60% w/v
0.48%
0.48%
0.50%
0.47%


Particulate
NMT 6000 (≥10
2
2
1
1


Matter (USP)
μm)







NMT 600 (≥25
0
0
0
0



μm)
















TABLE 39







Samples stored inverted at 40° C.











Acceptance

Time in months












Test
Criteria
Initial
1
2
3















Vasopressin
18.0-21.0 U/mL
19.9
19.2
18.3
17.4


Assay







Related
SEQ ID NO.: 2:
 0.2%
 0.9%
 2.2%
 3.8%


Substances
NMT 6.0%







SEQ ID NO.: 4:
 0.1%
 1.0%
 2.4%
 4.0%



NMT 6.0%







D-Asn-AVP:
 0.4%
 0.3%
 0.3%
 0.3%



NMT 1.0%







Asp5-AVP: NMT
ND
 0.2%
 0.5%
 0.8%



1.5%







AVP-Dimer:
ND
ND
ND
ND



NMT 1.0%







Acetyl-AVP:
 0.3%
 0.3%
 0.3%
 0.2%



NMT 1.0%







UI-0.13: NMT
ND
ND
ND
ND



1.0%







UI-0.75-0.78:
ND
ND
ND
ND



NMT 1.0%







UI-0.80-0.84:
ND
ND
ND
ND



NMT 1.0%







UI-1.02-1.03:
 0.3%
 0.2%
 0.2%
 0.3%



NMT 1.0%







UI-1.18: NMT
ND
ND
ND
 0.2%



1.0%







UI-1.35: NMT
 0.1%
ND
ND
ND



1.0%







UI-1.52-1.58:
ND
 0.2%
 0.3%
 0.4%



NMT 1.0%







UI-1.67: NMT
ND
ND
ND
ND



1.0%







UI-1.76: NMT
ND
ND
ND
ND



1.0%







UI-1.81-1.85:
ND
ND
ND
ND



NMT 1.0%







UI-1.86-1.88:
ND
 0.1%
 0.2%
ND



NMT 1.0%







UI-1.91-1.96:
 0.2%
 0.2%
ND
ND



NMT 1.0%







UI-2.02-2.08:
ND
ND
ND
 0.2%



NMT 1.0%







UI-2.11-2.14:
ND
 0.2%
ND
ND



NMT 1.0%







Total Impurities:
 1.5%
 3.7%
 6.3%
10.3%



NMT 17.0%






pH
2.5-4.5
3.6
3.4
3.2
3.1


Chlorobutanol
0.25-0.60% w/v
0.48%
0.47%
0.46%
0.46%


Particulate
NMT 6000 (≥10
2
2
1
1


Matter (USP)
μm)







NMT 600 (≥25
0
0
0
0



μm)
















TABLE 40







Samples stored Upright at 40° C.











Acceptance

Time in months












Test
Criteria
Initial
1
2
3















Vasopressin
18.0-21.0 U/mL
19.8
18.9
18.5
17.2


Assay







Related
SEQ ID NO.: 2:
 0.1%
 1.0%
 2.4%
 3.8%


Substances
NMT 6.0%







SEQ ID NO.: 4:
 0.1%
 1.1%
 2.7%
 4.3%



NMT 6.0%







D-Asn-AVP:
 0.3%
 0.3%
 0.3%
 0.3%



NMT 1.0%







Asp5-AVP: NMT
ND
 0.2%
 0.5%
 0.8%



1.5%







AVP-Dimer:
ND
ND
ND
ND



NMT 1.0%







UI-0.13: NMT
ND
 0.1%
ND
ND



1.0%







UI-0.75-0.78:
ND
ND
ND
ND



NMT 1.0%







UI-0.83-0.84:
ND
ND
ND
ND



NMT 1.0%







UI-1.02-1.03:
 0.2%
 0.2%
 0.2%
 0.2%



NMT 1.0%







UI-1.18: NMT
ND
ND
ND
 0.2%



1.0%







UI-1.56-1.57:
ND
 0.2%
 0.3%
 0.3%



NMT 1.0%







UI-1.67: NMT
ND
ND
ND
ND



1.0%







UI-1.76: NMT
ND
ND
ND
ND



1.0%







UI-1.83-1.85:
ND
ND
 0.2%
ND



NMT 1.0%







UI-1.87-1.88:
ND
ND
 0.2%
 0.2%



NMT 1.0%







UI-1.93: NMT
ND
 0.1%
ND
ND



1.0%







UI-2.05-2.08:
ND
ND
 0.2%
ND



NMT 1.0%







Total Impurities:
 1.1%
 3.6%
 7.2%
10.3%



NMT 17.0%







Total Impurities:
 1.1%
 3.6%
 7.2%
10.3%



NMT 17.0%






pH
2.5-4.5
3.5
3.3
3.2
3.1


Chlorobutanol
0.25-0.60% w/v
0.49%
0.48%
0.50%
0.48%


Particulate
NMT 6000 (≥10
1
1
1
1


Matter (USP)
μm)







NMT 600 (≥25
0
0
0
0



μm)




















TABLE 41







Samples stored Upright at 40° C.











Acceptance

Time in months












Test
Criteria
Initial
1
2
3















Vasopressin
18.0-21.0 U/mL
20.1
18.9
18.7
17.4


Assay







Related
SEQ ID NO.: 2:
 0.1%
 0.9%
 2.3%
 3.7%


Substances
NMT 6.0%







SEQ ID NO.: 4:
 0.1%
 1.0%
 2.5%
 3.9%



NMT 6.0%







D-Asn-AVP:
 0.3%
 0.4%
 0.3%
 0.3%



NMT 1.0%







Asp5-AVP: NMT
ND
 0.2%
 0.5%
 0.8%



1.5%







AVP-Dimer:
ND
ND
ND
ND



NMT 1.0%







Acetyl-AVP:
 0.3%
 0.2%
 0.3%
 0.2%



NMT 1.0%







UI-0.13: NMT
ND
ND
ND
ND



1.0%







UI-0.75-0.78:
ND
ND
 0.2%
 0.2%



NMT 1.0%







UI-0.80-0.84:
 0.2%
 0.2%
ND
ND



NMT 1.0%







UI-1.02-1.03:
 2.0%
 0.3%
 0.2%
 0.3%



NMT 1.0%







UI-1.18: NMT
ND
ND
ND
 0.2%



1.0%







UI-1.56-1.57:
ND
 0.2%
 0.4%
 0.4%



NMT 1.0%







UI-1.67: NMT
ND
ND
ND
ND



1.0%







UI-1.76: NMT
ND
ND
ND
ND



1.0%







UI-1.83-1.85:
ND
ND
 0.2%
 0.1%



NMT 1.0%







UI-1.87-1.88:
ND
ND
 0.2%
 0.2%



NMT 1.0%







UI-1.93: NMT
ND
 0.1%
ND
ND



1.0%







UI-2.05-2.08:
ND
ND
 0.2%
ND



NMT 1.0%







UI-2.14: NMT
ND
ND
ND
ND



1.0%







Total Impurities:
 1.3%
 3.5%
 7.1%
10.2%



NMT 17.0%






pH
2.5-4.5
3.6
3.3
3.2
3.1


Chlorobutanol
0.25-0.60% w/v
0.48%
0.48%
0.49%
0.47%


Particulate
NMT 6000 (≥10
2
1
1
1


Matter (USP)
μm)







NMT 600 (≥25
0
0
0
0



μm)




















TABLE 42







Samples stored Upright at 40° C.











Acceptance

Time in months












Test
Criteria
Initial
1
2
3















Vasopressin
18.0-21.0 U/mL
19.9
19.2
18.3
17.5


Assay







Related
SEQ ID NO.: 2:
 0.2%
 1.0%
 2.2%
 3.9%


Substances
NMT 6.0%







SEQ ID NO.: 4:
 0.1%
 1.1%
 2.4%
 4.2%



NMT 6.0%







D-Asn-AVP:
 0.4%
 0.3%
 0.3%
 0.3%



NMT 1.0%







Asp5-AVP: NMT
ND
 0.2%
50.0%
 0.8%



1.5%







AVP-Dimer:
ND
ND
ND
ND



NMT 1.0%







Acetyl-AVP:
 0.3%
 0.3%
 0.3%
 0.2%



NMT 1.0%







UI-0.13: NMT
ND
ND
ND
ND



1.0%







UI-0.75-0.78:
ND
ND
ND
ND



NMT 1.0%







UI-0.80-0.84:
ND
ND
ND
ND



NMT 1.0%







UI-1.02-1.03:
 0.3%
 0.2%
 0.2%
 0.3%



NMT 1.0%







UI-1.18: NMT
ND
ND
ND
 0.2%



1.0%







UI-1.35: NMT
 0.1%
ND
ND
ND



1.0%







UI-1.52-1.58:
ND
 0.2%
 0.3%
 0.4%



NMT 1.0%







UI-1.67: NMT
ND
ND
ND
ND



1.0%







UI-1.76: NMT
ND
ND
ND
ND



1.0%







UI-1.83-1.85:
ND
ND
ND
ND



NMT 1.0%







UI-1.86-1.88:
ND
 0.1%
 0.2%
ND



NMT 1.0%







UI-1.91-1.96:
 0.2%
 0.2%
ND
ND



NMT 1.0%







UI-2.02-2.08:
ND
ND
ND
 0.1%



NMT 1.0%







UI-2.11-2.14:
ND
 0.2%
ND
ND



NMT 1.0%







Total Impurities:
 1.5%
 3.8%
 6.3%
10.5%



NMT 17.0%






pH
2.5-4.5
3.6
3.4
3.2
3.1


Chlorobutanol
0.25-0.60% w/v
0.48%
0.47%
0.47%
0.45%


Particulate
NMT 6000 (≥10
1
2
1
1


Matter (USP)
μm)







NMT 600 (≥25
0
0
0
0



μm)













Example 9: Effect of pH 3.5-4.5 on Vasopressin Formulations

To test of effect of pH on vasopressin formulations, solutions containing 20 units/mL vasopressin, adjusted to pH 3.5, 3.6, 3.7, 3.8, 3.9, 4, 4.1, 4.2, 4.3, 4.4, or 4.5 with 10 mM acetate buffer, were prepared. One mL of each of the vasopressin formulations was then filled into 10 cc vials.


The vasopressin formulations were stored for four weeks at: (i) 25° C.; or (ii) 40° C., and the assay (% label claim; vasopressin remaining) and % total impurities after four weeks were measured using the methods described in EXAMPLE 1. FIGS. 11 and 12 below display the results of the experiments at 25° C. The results of the experiments at 40° C. are included in FIGS. 13 and 14.


The results of the experiments suggested that the stability of a vasopressin formulation was affected by pH. At 25° C., the remaining vasopressin after four weeks was highest between pH 3.6 and pH 3.8 (FIG. 11). Within the range of pH 3.6 to pH 3.8, the level of impurities was lowest at pH 3.8 (FIG. 12). At 25° C., pH 3.7 provided the highest stability for vasopressin (FIG. 11).


At 40° C., the remaining vasopressin after four weeks was highest between pH 3.6 and pH 3.8 (FIG. 13). Within the range of pH 3.6 to pH 3.8, the level of impurities was lowest at pH 3.8 (FIG. 14). At 40° C., pH 3.6 provided the highest stability for vasopressin (FIG. 13),


Example 10: Effect of pH 2.5-4.5 of Vasopressin Formulations

To test of effect of pH on vasopressin formulations, solutions containing 20 units/mL vasopressin, adjusted to pH 2.5, 2.6, 2.7, 2.8, 2.9, 3, 3.1, 3.2, 3.3, or 3.4 with 10 mM acetate buffer were also prepared. One mL of each of the vasopressin formulations was then filled into 10 cc vials.


The amount of vasopressin, impurities, and associated integration values were determined using the methods describes in EXAMPLE 1. The results from the stability tests on the vasopressin formulations from pH 2.5 to 3.4 were plotted against the results from the stability tests on vasopressin formulations from pH 3.5 to 4.5 as disclosed in EXAMPLE 9, and are displayed in FIGS. 15-18.


The assay (% label claim; vasopressin remaining) and % total impurities in the vasopressin pH 2.5 to 3.4 formulations after four weeks are reported in TABLE 43.














TABLE 43






Target


Vasopressin
% Total


Batch
pH
Week
Condition
(% LC)
Impurities




















1A
2.5
0
25° C.
100.57
2.48


1B
2.6
0
25° C.
101.25
2.24


1C
2.7
0
25° C.
101.29
2.26


1D
2.8
0
25° C.
101.53
2.00


1E
2.9
0
25° C.
102.33
1.95


1F
3
0
25° C.
102.32
1.89


1G
3.1
0
25° C.
102.59
2.06


1H
3.2
0
25° C.
102.60
1.85


1I
3.3
0
25° C.
102.73
1.81


1J
3.4
0
25° C.
101.93
1.75


1A
2.5
0
40° C.
100.57
2.48


1B
2.6
0
40° C.
101.25
2.24


1C
2.7
0
40° C.
101.29
2.26


1D
2.8
0
40° C.
101.53
2.00


1E
2.9
0
40° C.
102.33
1.95


1F
3
0
40° C.
102.32
1.89


1G
3.1
0
40° C.
102.59
2.06


1H
3.2
0
40° C.
102.60
1.85


1I
3.3
0
40° C.
102.73
1.81


1J
3.4
0
40° C.
101.93
1.75


1A
2.5
4
25° C.
95.70
6.66


1B
2.6
4
25° C.
98.58
5.29


1C
2.7
4
25° C.
98.94
4.26


1D
2.8
4
25° C.
99.14
3.51


1E
2.9
4
25° C.
100.08
3.41


1F
3
4
25° C.
100.29
2.92


1G
3.1
4
25° C.
100.78
2.55


1H
3.2
4
25° C.
100.74
2.16


1I
3.3
4
25° C.
100.46
2.14


1J
3.4
4
25° C.
100.25
2.03


1A
2.5
4
40° C.
81.89
19.41


1B
2.6
4
40° C.
90.10
15.60


1C
2.7
4
40° C.
92.19
13.46


1D
2.8
4
40° C.
94.89
10.98


1E
2.9
4
40° C.
96.03
9.78


1F
3
4
40° C.
97.26
8.09


1G
3.1
4
40° C.
99.61
6.39


1H
3.2
4
40° C.
98.58
5.25


1I
3.3
4
40° C.
97.81
4.41


1J
3.4
4
40° C.
97.35
3.85









The % total impurities for the pH 2.5 to 3.4 formulations and the pH 3.5 to 4.5 formulations observed in the experiments conducted at 25° C. and 40° C. are shown in FIGS. 15 (25° C.) and 16 (40° C.).


The vasopressin assay amount for the vasopressin pH 2.5 to 3.4 formulations and the vasopressin pH 3.5 to 4.5 formulations observed in the experiments conducted at 25° C. and 40° C. are shown in FIGS. 17 (25° C.) and 18 (40° C.). The vasopressin assay is presented as a % assay decrease of vasopressin over the four-week study period, rather than absolute assay, because the amount of starting vasopressin varied between the vasopressin pH 2.5 to 3.4 formulations and the vasopressin pH 3.5 to 4.5 formulations.


The results of the above experiments suggested that the stability of a vasopressin formulation was affected by pH. At 25° C., the percent decrease in vasopressin after four weeks was lowest between pH 3.7 and pH 3.8 (FIG. 17). Within the range of pH 3.7 to pH 3.8, the level of impurities was lowest at pH 3.8 (FIG. 15). At 40° C., the percent decrease in vasopressin after four weeks was lowest between pH 3.6 and pH 3.8 (FIG. 18). Within the range of pH 3.6 to pH 3.8, the level of impurities was lowest at pH 3.8 (FIG. 16).


Example 11: Intra-Assay and Inter-Analysis Precision of Vasopressin pH Experiments

The methods used to determine the % assay decrease and amount of impurities in the vasopressin solutions over time in EXAMPLE 10 had both intra-assay and inter-analyst precision.


Intra-assay precision was demonstrated by performing single injections of aliquots of a vasopressin formulation (n=6; Chemist 1) from a common lot of drug product and determining the assay and repeatability (% RSD; relative standard deviation). Inter-analyst precision was demonstrated by two different chemists testing the same lot of drug product; however, the chemists used different instruments, reagents, standard preparations, columns, and worked in different laboratories. The procedure included a common pooling of 20 vials of vasopressin, which were assayed by the two chemists using different HPLC systems and different HPLC columns. The vasopressin assay results (units/mL) and repeatability (% RSD for n=6) were recorded and are reported in the TABLE 44 below.









TABLE 44







Precision of Vasopressin Results.










Chemist 1
Chemist 2


Sample
(units/mL)
(units/mL)





1
19.74   
19.65   


2
19.76   
19.66   


3
19.77   
19.66   


4
19.75   
19.72   


5
19.97   
19.73   


6
19.65   
19.73   


Mean
19.8    
19.7    


% RSD (≤2.0%)
0.5%
0.2%














%





Difference

=

0.5

%






(


accecptance





criteria


:




3.0

%


)









%





Difference

=



(


Chemist






1
Mean


-

Chemist






2
Mean



)


(


Chemist






1
Mean


+

Chemist






2
Mean



)


×
200
















The intra-assay repeatability met the acceptance criteria (% RSD≤2.0%) with values of 0.5% and 0.2%. The inter-analyst repeatability also met the acceptance criteria (% difference ≤3.0%) with a difference of 0.5%.


Example 12: Effect of Citrate Versus Acetate Buffer on Vasopressin Formulations

To test the effect of citrate and acetate buffer on vasopressin formulations, a total of twelve solutions of 20 Units/mL vasopressin were prepared in 1 mM citrate buffer, 10 mM citrate buffer, 1 mM acetate buffer, and 10 mM acetate buffer. All of the solutions were prepared in triplicate. Each solution was adjusted to pH 3.5 with hydrochloric acid.


The vasopressin formulations were stored at 60° C. for 7 days, and the assay (% label claim; vasopressin remaining) and % total impurities after 7 days were analyzed by HPLC using the procedure and experimental conditions described in EXAMPLE 1.


The assay (% label claim; vasopressin remaining) and % total impurities for each of the Vasopressin Buffered Formulations are reported in the TABLES 45 and 46 below.









TABLE 45







Assay (% label claim; vasopressin remaining) in the


vasopressin formulations after storage at 60° C. for 7 days.










Sample












Buffer
1
2
3
Average





 1 mM citrate buffer
89.5%
89.7%
90.6%
89.9%


10 mM citrate buffer
84.1%
84.4%
84.5%
84.3%


 1 mM acetate buffer
90.5%
91.1%
91.9%
91.2%


10 mM acetate buffer
90.9%
90.9%
92.4%
91.4%
















TABLE 46







% Total Impurities in the vasopressin formulations


after storage at 60° C. for 7 days.












Sample














Buffer
1
2
3
Average







 1 mM citrate buffer
3.4%
3.5%
2.5%
3.1%



10 mM citrate buffer
9.5%
9.0%
9.4%
9.3%



 1 mM acetate buffer
3.3%
2.8%
3.2%
3.1%



10 mM acetate buffer
2.9%
2.6%
3.1%
2.9%










The data indicated that the vasopressin assay in the vasopressin formulations with citrate buffer was lower than in the vasopressin formulations with acetate buffer. For example, at 10 mM of either citrate or acetate buffer, the average vasopressin assay was 91.4% in acetate buffer, but was 84.3% in citrate buffer. The data also indicated that % total impurities in the vasopressin formulations with citrate buffer were higher than in the vasopressin formulations with acetate buffer. For example, at 10 mM of either citrate or acetate buffer, the average % total impurities was 2.9% in acetate buffer, but was 9.3% in citrate buffer.


Further, as the citrate buffer concentration increased, the vasopressin assay further decreased (from an average of 89.9% to 84.3%), and the % total impurities increased (from an average of 3.1% to 9.3%). This effect was not observed in the vasopressin formulations with acetate buffer, where the average and % total impurities stayed fairly constant.


Example 13: Multi-Dose Vasopressin Formulation

A multi-dose formulation (10 mL) for vasopressin that can be used in the clinic is detailed in TABLE 47 below:









TABLE 47







Drug Product Description









Vasopressin,

20 Units


USP
Active Ingredient
(~0.04 mg)





Dosage Form
Injection



Route of
Intravenous



Administration




Description
Clear colorless to practically




colorless solution supplied




in a 10 mL clear glass vial with flip-off cap









The composition of a 10 mL formulation of vasopressin is provided below.









TABLE 48







Drug Product Composition











Ingredient
Grade
Function
Batch Quantity
Unit Formula
















Vasopressin, USP
USP
Active
3,000,000
Units
20
Units


Sodium Acetate Trihydrate
USP
Buffer
214.2
g
1.36
mg












Sodium Hydroxide
NF
pH Adjustor
40
g
QS to pH 3.8


Hydrochloric Acid
NF/EP
pH Adjustor
237.9
g
QS to pH 3.8













Chlorobutanol
NF
Preservative
0.8274
kg
5
mg











Water for Injection
USP
Solvent
QS
QS to 1 mL


Nitrogen
NF
Processing Aid











The 10 mL vasopressin formulation was compared to the guidelines for inactive ingredients provided by the Food and Drug Administration (FDA). The results are shown in TABLE 49 below.












TABLE 49






Vasopressin 10 mL
Concentration (%
Inactive Ingredients


Ingredient
Formulation (mg/mL)
w/v)
Guideline Acceptable Level







Sodium Acetate
1.36
0.136%
IV (infusion); Injection


Trihydrate


0.16%


Sodium Hydroxide
QS to pH 3.8
QS to pH 3.8
N/A


Hydrochloric
Acid
QS to pH 3.8
QS to pH 3.8 N/A


Chlorobutanol
5 mg
 0.5%
IV (Infusion); Injection 1%


Water for Injection
QS to 1 mL
QS to target
N/A




volume









Example 14: Alternative Vasopressin Formulation for Clinical Use

A 1 mL dosage of vasopressin was prepared. A description of the formulation is shown in TABLE 50 below.









TABLE 50







Drug Product Description











20




Units/mL


Vasopressin,

(~0.04


USP
Active Ingredient
mg)





Dosage Form
Injection



Route of
Intravenous



Administration




Description
Clear colorless to practically




colorless solution supplied in




a 3 mL vial with flip-off cap









The drug composition of the formulation is provided in TABLE 51.









TABLE 51







Drug Product Composition









Ingredient
Function
Quantity (mg/mL)





Vasopressin, USP
Active
20 Units


Sodium Acetate Trihydrate,
Buffer
1.36


USP




Sodium Hydroxide NF/EP
pH
QS for pH



Adjustor
adjustment to pH 3.8


Hydrochloric Acid, NF/EP
pH
QS for pH



Adjustor
adjustment to pH 3.8


Water for Injection
Solvent
QS to 1 mL









The 1 mL vasopressin formulation was compared to the guidelines for inactive ingredients provided by the Food and Drug Administration (FDA). The results are shown in TABLE 52 below.












TABLE 52






Vasopressin





1 mL
Concentration
Inactive Ingredients



Formulation
(%
Guideline


Ingredient
(mg/mL)
w/v)
Acceptable Level







Sodium Acetate
1.36
0.136%
0.16%  


Trihydrate


Sodium Hydroxide
QS to pH 3.8
QS to pH 3.8
 8%


Hydrochloric Acid
QS to pH 3.8
QS to pH 3.8
10%


Water for Injection
QS to 1 mL
QS to target
N/A




volume









Example 15: 15-Month Stability Data for Vasopressin Formulations

The drug product detailed in TABLE 51 was tested for stability over a 15-month period. Three different lots (X, Y, and Z) of the vasopressin drug formulation were stored at 25° C. for 15 months in an upright or inverted position. At 0, 1, 2, 3, 6, 9, 12, 13, 14, and 15 months, the amount of vasopressin (AVP), % label claim (LC), amount of various impurities, and pH was measured. The vasopressin and impurity amounts were determined using the HPLC method described above in EXAMPLE 1. The results of the stability experiment are shown in TABLES 53-54 below.









TABLE 53







Inverted Storage of Vasopressin Formulations at 25° C.




































UI-
UI
UI-
UI-
UI-
UI-
UI-
UI-






AVP






Ace-
0.81-
0.97
1.02-
1.72-
1.81-
1.90-
2.05-
2.09-
Total




(U/
%
Gly9
Glu4
D-Asn
Asp5
Dimer
tyl
0.86
0.99
1.03
1.76
1.89
1.96
2.07
2.10
Impuri-


Lot
Month
mL)
LC
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
ties
pH





























X
0
19.6
97.9





0.3
0.4

0.3





1.0
3.8


Y
0
19.7
98.6





0.3
0.4

0.3





1.1
3.8


Z
0
19.9
99.3
0.1




0.5


0.2





0.8
3.8


X
1
19.6
98.1
0.2
0.2
0.1


0.4
0.4

0.3





1.6
3.8


Y
1
19.6
97.9
0.2
0.2
0.1


0.4
0.4

0.3





1.6
3.9


Z
1
19.8
99
0.2
0.2



0.6
0.1

0.2





1.4
3.8


X
2
19.6
98.1
0.3
0.3
0.1


0.3
0.4

0.3





1.7
3.7


Y
2
19.5
97.5
0.2
0.3
0.1


0.3
0.4

0.3





1.6
3.8


Z
2
19.8
99
0.3
0.4



0.5


0.2





1.3
3.8


X
3
19.6
97.8
0.4
0.5
0.1
0.1

0.3
0.4

0.4





2.2


Y
3
19.5
97.4
0.4
0.4
0.1


0.3
0.4

0.4





2.0
3.8


Z
3
19.7
98.6
0.4
0.4



0.5


0.3





1.6


X
6
19.3
96.5
0.7
0.8
0.1
0.2

0.3
0.4

0.4





2.9
3.8


Y
6
19.2
95.9
0.6
0.7
0.1
0.1
0.1
0.3
0.4

0.4





2.5
3.9


Z
6
19.6
98
0.6
0.7

0.1

0.5


0.2





2.3
3.9


X
9
19
95
1.0
1.0

0.2

0.3
0.4

0.4

0.1



3.6


Y
9
18.9
94.5
0.8
1.0

0.2

0.3
0.4

0.4

0.1



3.1
3.9


Z
9
19.2
96
1.0
1.1

0.2

0.5


0.3





3.1
3.8


X
12
18.7
93.5
1.4
1.5
0.1
0.3

0.3
0.4

0.5

0.2



4.8
3.8


Y
12
18.6
93
1.1
1.2
0.2
0.2

0.3
0.4

0.5

0.3
0.2


4.4
3.8


Z
12
18.9
94.5
1.2
1.3

0.3

0.5


0.3

0.3
0.1


4.0
3.8


X
13
18.6
93
1.5
1.6
0.2
0.3

0.4
0.4
0.1
0.4

0.2
0.1


5.2
3.8


Y
13
18.5
92.5
1.2
1.3
0.2
0.3

0.3
0.4
0.1
0.5
0.1
0.4
0.2
0.2

5.2
3.9


Z
13
19
95
1.3
1.5
0.1
0.3

0.5
0.1

0.3
0.1
0.3
0.2
0.2

4.9
3.8


X
14
18.6
93
1.5
1.7
0.1
0.3

0.3
0.5
0.1
0.4

0.4
0.1
0.1

5.5
3.8


Y
14
18.5
92.5
1.2
1.4
0.1
0.3

0.3
0.5
0.1
0.4

0.5
0.2
0.2

5.3
3.9


Z
14
18.9
94.5
1.3
1.6

0.3

0.5
0.2

0.3

0.4
0.2
0.2

5.0
3.8


X
15
18.5
92.5
1.6
1.8
0.1
0.4

0.3
0.4
0.1
0.4

0.3
0.2
0.2

5.9
3.8


Y
15
18.4
92
1.3
1.5
0.1
0.3

0.3
0.4
0.1
0.4

0.5
0.3
0.1

5.3
3.9


Z
15
18.8
94
1.5
1.6

0.3

0.5


0.3

0.4
0.2
0.1

4.9
3.9
















TABLE 54







Upright Storage of Vasopressin Formulations at 25° C.




































UI-
UI
UI-
UI-
UI-
UI-
UI-
UI-






AVP






Ace-
0.81-
0.97
1.02-
1.72-
1.81-
1.90-
2.05-
2.09-
Total




(U/
%
Gly9
Glu4
D-Asn
Asp5
Dimer
tyl
0.86
0.99
1.03
1.76
1.89
1.96
2.07
2.10
Impuri-


Lot
Month
mL)
LC
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
(%)
ties
pH





























X
0
19.6
97.9





0.3
0.4

0.3





1.0
3.8


Y
0
19.7
98.6





0.3
0.4

0.3





1.1
3.8


Z
0
19.9
99.3
0.1




0.5


0.2





0.8
3.8


X
1
19.6
98
0.2
0.2
0.1


0.3
0.4

0.3





1.6
3.8


Y
1
19.5
97.7
0.2
0.2



0.3
0.4

0.3





1.4
3.9


Z
1
19.7
98.3
0.2
0.2



0.6


0.2





1.2
3.8


X
2
19.6
98.2
0.3
0.3



0.3
0.4

0.3





1.6
3.7


Y
2
19.5
97.4
0.2
0.3
0.1


0.4
0.4

0.3





1.6
3.8


Z
2
19.8
99
0.3
0.3



0.5


0.2





1.3
3.8


X
3
19.5
97.6
0.4
0.4
0.1


0.3
0.4

0.4





2.1
3.7


Y
3
19.5
97.5
0.4
0.4
0.1



0.4

0.4





1.9
3.8


Z
3
19.7
98.7
0.4
0.4

0.1

0.5


0.3





1.7


X
6
19.3
96.5
0.7
0.8
0.1
0.2

0.3
0.4

0.4





2.9
3.8


Y
6
19.2
96
0.5
0.7
0.1
0.1

0.3
0.4

0.4





2.5
3.9


Z
6
19.5
97.5
0.7
0.7

0.2

0.5


0.3





2.3
3.9


X
9
18.9
94.5
1.0
1.1

0.2

0.3
0.4



0.2
0.1


3.7
3.8


Y
9
18.9
94.5
0.8
0.9

0.2


0.4

0.4

0.2



3.1
3.9


Z
9
19.2
96
0.9
1.0

0.2

0.5


0.3





2.9
3.8


X
12
18.6
93
1.4
1.5
0.1
0.3

0.3
0.4

0.5

0.2
0.1


4.8
3.7


Y
12
18.7
93.5
1.1
1.2
0.1
0.3

0.3
0.4

0.5


0.2

0.2
4.6
3.9


Z
12
18.9
94.5
1.3
1.4

0.3

0.5


0.4

0.3
0.2


4.2
3.8


X
13
18.4
92
1.5
1.6
0.2
0.3

0.3
0.4
0.1
0.4

0.3
0.1
0.1

5.4
3.8


Y
13
18.6
93
1.1
1.3
0.2
0.3

0.3
0.4
0.1
0.4

0.3
0.2


4.6
3.9


Z
13
18.8
94
1.3
1.5

0.3

0.5
0.1

0.3

0.4
0.2
0.1

4.7
3.8


X
14
18.6
93
1.5
1.7
0.1
0.4

0.3
0.4
0.1
0.4

0.3
0.1


5.4
3.8


Y
14
18.5
92.5
1.2
1.4
0.1
0.3

0.3
0.5

0.4

0.5
0.3
0.3

5.4
3.9


Z
14
18.8
94
1.3
1.5

0.3

0.5
0.1

0.3

0.5
0.2
0.2

5.0
3.8


X
15
18.4
92
1.6
1.8
0.1
0.4

0.3
0.4
0.1
0.4

0.3
0.2


5.7
3.8


Y
15
18.4
92
1.3
1.5
0.2
0.3

0.3
0.4
0.1
0.4

0.5
0.3
0.3

5.4
3.9


Z
15
18.6
93
1.5
1.6

0.3

0.5


0.2

0.4
0.2
0.3

5.1
3.9









The results from TABLES 53-54 indicate that stability of the vasopressin formulations was not significantly affected by either inverted or upright storage. The impurities detected included Gly9 (SEQ ID NO.: 2), Glu4 (SEQ ID NO.: 4), D-Asn (SEQ ID NO.: 10), Asp5 (SEQ ID NO.: 3), Acetyl-AVP (SEQ ID NO.: 7), vasopressin dimer, and several unidentified impurities (UI). The unidentified impurities are labeled with a range of relative retention times at which the impurities eluted from the column.


The results indicate that the pH remained fairly constant over the 15-month period, fluctuating between 3.8 and 3.9 throughout the 15 months. The total impurities did not increase over 5.9%, and the % LC of vasopressin did not decrease below 92%.



FIG. 19 shows a graph depicting the % LC over the 15-month study period for the results provided in TABLES 53-54. The starting amounts of vasopressin were 97.9% LC for lot X, 98.6% LC for lot Y, and 99.3% LC for lot Z. The results indicate that the % LC of vasopressin decreased over the 15-month study period, but did not decrease below 92% LC.


The formula for the trend line of lot X was:





% LC=98.6−0.4262(month)


The formula for the trend line of lot Y was:





% LC=98.47−0.4326(month)


The formula for the trend line of lot Z was:





% LC=99.54−0.3906(month)


Example 16: 10 mL Vasopressin Formulation

The drug product description for a 10 mL vasopressin formulation is shown in TABLE 55 below:









TABLE 55





Drug Product Description
















Vasopressin,
Active Ingredient


USP


Strength
20 U/mL


Dosage Form
Injection


Route of
Intravenous


Administration


Description
Clear colorless to practically colorless solution supplied



in a 10 mL vial with orange flip-off cap









The composition of the 10 mL vasopressin formulation is shown in TABLE 56 below:









TABLE 56







Drug Product Composition











Ingredient
Grade
Function
Batch Quantity
Unit Formula
















Vasopressin, USP
USP
Active
3,000,000
Units
20
Units


Sodium Acetate Trihydrate
USP
Buffer
214.2
g
1.36
mg












Sodium Hydroxide
NF
pH Adjustor
40
g
QS to pH 3.8


Hydrochloric Acid
NF/EP
pH Adjustor
237.9
g
QS to pH 3.8













Chlorobutanol
NF
Preservative
0.8274
kg
5
mg












Water for Injection
USP
Solvent
QS
QS to 1
mL











Nitrogen
NF
Processing Aid











The container closure system used in the 10 mL vasopressin formulation is shown below in TABLE 57.









TABLE 57







Container/Closure Description










Part
Type







Vial
10 mL, 13 mm Type 1, Flint (clear) tubing vial



Stopper
13 mm, Westar RS, 4432/50 Flurotec



Cap
Flip-Off Cap, 13 mm Orange Peel










Labeled vials containing the 10 mL vasopressin formulation are packaged into secondary packaging consisting of preprinted paperboard cartons. The vials can be packaged in a single pack (one vial in one carton) or a 5-pack (5 vials in one carton).



FIG. 20 below shows a diagram of a cap to be used on a vial described herein. The left part of FIG. 20 shows an aluminum seal to be used in a cap of the vial described herein. The height of seal is from about 6.17 mm to about 6.38 mm. The middle of FIG. 20 shows the plastic button, which has a diameter of about 14.99 mm. The right portion of FIG. 20 depicts the assembled cap with the aluminum seal placed on top of the plastic button. The assembled cap has a height from about 7.62 mm to about 8.38 mm.



FIG. 21 depicts a stopper to be used in a vial described herein. The left part of FIG. 21 shows a top view, the center of FIG. 21 shows a cross-sectional side view, and the right part of FIG. 21 shows a bottom view of the stopper. FIG. 21 indicates that the diameter of the stopper can be about 12.75±0.3 mm. The width of the top part of the stopper can be about 2.11±0.3 mm. The height of the entire stopper can be about 6.3±0.51 mm.


A vial for use in storing the 10 mL vasopressin formulation is shown in FIG. 22. The dimensions for the vial are shown in TABLE 58 below:












TABLE 58







Dimension as denoted in FIG. 22
Length (mm)









d1
23.75 ± 0.25 



d2
13.15 ± 0.2 



d4
7.05 ± 0.2 



d59
1 (minimum)



h1
 50 ± 0.5



h3
8.5 ± 0.5



h21
3.9 ± 0.2



c51
35 (approximately)



s1
 1.2 ± 0.05



s2
0.7 (max)



t
0.76 (max)










Example 17: Container Closure Integrity Test for the 10 mL Vasopressin Formulation

Microbial ingress testing can be performed to demonstrate container closure system seal integrity for a stopper used in a vial described herein. Microbial ingress testing can evaluate the adequacy of the closure to maintain a sterile barrier. A sample of the container closure system was tested by immersing media filled containers into a media bath chamber inoculated with a high concentration of Brevundimonas diminuta, which containers were then subjected to pressure and vacuum. The containers were then removed from the pressure and vacuumed, incubated, and examined for bacterial growth.


Seventy-four washed and depyrogenated 10 mL tubing vials were hand-filled with approximately 5 mL of Trypticase Soy Broth and then hand stoppered. The vials were then capped, and incubated at 20-25° C. for seven days and then at 30-35° C. for eight days. At the end of the incubation period, none of the vials had any microbial growth.


From the original 74 vials, 20 vials were sent for microbial ingress testing, along with three positive controls and two negative controls. The test vials and positive controls were submerged in a bacterial suspension. The two negative controls were not exposed to the bacterial suspension, vacuum, or pressure, but were incubated for seven days at 30-35° C. The test vial samples were then incubated at 30-35° C. for seven days, and the positive controls were incubated at 30-35° C. for five days. At the end of the incubation intervals, all the samples were examined for evidence of microbial ingress by assessing the turbidity of the solutions. There was no microbial growth in 20 out of 20 of the test samples, nor was there growth in the negative controls. There was bacterial growth in 3 out of 3 of the positive controls.


Example 18: Formulation Stability for 10 mL Vasopressin Formulation

Three batches of the 10 mL vasopressin formulation were tested for stability at 2-8° C. and 25° C. for three months. The formulation used for the tests is shown in TABLE 56 above. The study layout is shown in TABLE 59 below. Lots A-C were the 10 mL vasopressin formulation, while Lots D-F were the 1 mL vasopressin formulation.












TABLE 59









Condition













Study
Lot
° C.
% RH
Orientation
Study Duration















1
A
5
Not Defined
Inverted
24 months


2
A
5
Not Defined
Upright
24 months


3
A
25
60
Inverted
 6 months


4
A
25
60
Upright
 6 months


1
B
5
Not Defined
Inverted
24 months


2
B
5
Not Defined
Upright
24 months


3
B
25
60
Inverted
 6 months


4
B
25
60
Upright
 6 months


1
C
5
Not Defined
Inverted
24 months


2
C
5
Not Defined
Upright
24 months


3
C
25
60
Inverted
 6 months


4
C
25
60
Upright
 6 months


1
D
5
Not Defined
Inverted
24 months


2
D
5
Not Defined
Upright
24 months


3
D
25
60
Inverted
24 months


4
D
25
60
Upright
24 months


1
E
5
Not Defined
Inverted
24 months


2
E
5
Not Defined
Upright
24 months


3
E
25
60
Inverted
24 months


4
E
25
60
Upright
24 months


1
F
5
Not Defined
Inverted
24 months


2
F
5
Not Defined
Upright
24 months


3
F
25
60
Inverted
24 months


4
F
25
60
Upright
24 months









The stability trends were calculated from a linear regression of the datasets. The estimated shelf-life (ESL) provided in the tables below refers to the shortest time in months at which the extrapolated 95% confidence interval for the linear model of the attribute in question exceeded the specification limit. All models assumed a concentration-independent linear decline or increase (zero-order models). The results were pooled to obtain the best estimates for average trends. Three linear degradation models were fit to the data and the expiration date of the batches was estimated following FDA-issued guidelines. The three models were the following:


Model 1: Different slopes and different intercepts for the batches.


Model 2: Common slope and different intercepts for the batches.


Model 3: Common slope and common intercept for the batches.


The recommended model was determined by the following procedure: Model 1 was fit with the time effect first in the model, followed by the batch effect, then the interaction. Using Type I (Sequential) sums-of-squares, the regressions were tested for equal slopes (Source C in the output). If the p-value was less than 0.25, the slopes were assumed to be different across batches. Then, the procedure was stopped and Model 1 was used to estimate the expiration date. If the p-value was greater than or equal to 0.25, the slopes were assumed to be common across batches. Then, the procedure continued to step 2. If the conclusion from step 1 was common slopes, then the regressions were tested for equal intercepts using Type I (Sequential) sums-of-squares from Model 1 (Source B in the output). If the p-value was less than 0.25, the intercepts were assumed to be different across batches, and Model 2 was used to estimate the expiration date. If the p-value was greater than or equal to 0.25, the intercepts were assumed to be common across batches, and Model 3 was used to estimate the expiration date. When Model 1 (different slopes and different intercepts) was used for estimating the expiration date, the MSE (mean squared error) was not pooled across batches. Prediction intervals were computed for each batch using individual mean squared errors, and the interval that crossed the specification limit first was used to estimate the expiration date.


The results indicated that the pH of the formulations ranged from 3.7 to 3.8, and no significant changes were observed through three months under long-term and accelerated storage conditions. The vasopressin assay values ranged from 100% to 100.3% at the time of study release. The results at all stability intervals through three months were within the proposed limits as shown in the data tables below. The stability study results for the 10 mL vasopressin formulation were compared to the 1 mL vasopressin formulation and are shown TABLE 60 below.


At 25° C., the shortest estimated shelf-life based on trend extrapolation at 25° C. was approximately 17.2 months, which was comparable to the 15.1 month shelf-life value obtained for the 1 mL vasopressin formulation as shown in TABLE 60.


At 5° C., the vasopressin assay ranged from 99.8% to 100.1% after three months.















TABLE 60









Intercept ±
Slope ±






ESL
Std. Error
Std. Error


Formulation
R2
Lot
(months)
(%)
(%/month)
Model Type





















10 mL
0.7
A
17.2
 100% ± 0.12%
−0.46% ± 0.06%
Common




B



slopes/common




C



intercepts


 1 mL
0.97
D
15.1
98.6% ± 0.27%
−0.53% ± 0.02%
Common




E

99.1% ± 0.27%

slopes/different




F

99.6% ± 0.27%

intercepts





Note:


For this regression analysis, upright and inverted studies were treated as independent replicates of a single batch.







FIG. 23 shows the vasopressin assay results (represented as % label claim) of the studies performed at 25° C. with upright storage. The linear equation for each of the lots was as follows:





% LC=100−0.42(month)  Lot A:





% LC=99.86−0.44(month)  Lot B:





% LC=100−0.4(month)  Lot C:





% LC=98.58−0.5359(month)  Lot D:





% LC=98.58−0.5359(month)  Lot E:





% LC=99.37−0.515(month)  Lot F:



FIG. 24 shows the vasopressin assay results (represented as % label claim) of the studies performed at 25° C. with inverted storage. The linear equation for each of the lots was as follows:





% LC=99.8−0.4(month)  Lot A:





% LC=99.97−0.63(month)  Lot B:





% LC=100−0.46(month)  Lot C:





% LC=98.68−0.5519(month)  Lot D:





% LC=100−0.5634(month)  Lot E:





% LC=99.49−0.52(month)  Lot F:


The data supported a 24-month refrigerated (2-8° C.) shelf life with a 12-month period out of refrigeration.


Vasopressin impurities, as detailed in TABLE 1, were tested for in the 1 mL and 10 mL vasopressin formulations. First, Gly9-AVP was measured in the formulations after storage for three months. Gly9-AVP is a deamidation product and major degradant of vasopressin. The concentration of Gly9-AVP was around <0.10% to 0.1% at time of release. The results at all stability intervals through three months were within the proposed limits (NMT 6%) as shown in TABLE 61 below.















TABLE 61









Intercept ±
Slope ±






ESL
Std. Error
Std. Error


Formulation
R2
Lot
(months)
(%)
(%/month)
Model Type





















10 mL
0.9
A
49.9
 0.15% ± 0.02%
0.10% ± 0.0%
Common




B

 0.1% ± 0.02%

slopes/different




C

 0.1% ± 0.02%

intercepts


 1 mL
0.99
D
32.2
−0.03% ± 0.06%
0.19% ± 0.0%
Different




E

−0.05% ± 0.06%
0.18% ± 0.0%
slopes/different




F

−0.04% ± 0.06%
0.19% ± 0.0%
intercepts





Note:


For this regression analysis, upright and inverted studies are treated as independent replicates of a single batch.






The stability study results for the 10 mL vasopressin formulations were compared to the 1 mL vasopressin formulation and plotted as shown in FIGS. 25 and 26. FIG. 25 shows the amount of Gly9-AVP when the formulations were stored at 25° C. in an upright position. The linear equation for each of the lots was as follows:






Y=0.0015+0.001(month)  Lot A:






Y=0.001+0.001(month)  Lot B:






Y=0.001+0.001(month)  Lot C:






Y=−0.000204+0.001825(month)  Lot D:






Y=−0.000204+0.001825(month)  Lot E:






Y=−0.000545+0.0019(month)  Lot F:



FIG. 26 shows the amount of Gly9-AVP when the formulations were stored at 25° C. in an inverted position. The linear equation for each of the lots was as follows:






Y=0.0015+0.001(month)  Lot A:






Y=0.001+0.001(month)  Lot B:






Y=0.001+0.001(month)  Lot C:






Y=−0.000431+0.001875(month)  Lot D:






Y=−0.000743+0.001794(month)  Lot E:






Y=−0.000234+0.001814(month)  Lot F:


The regression analysis and trend estimates for stability at 25° C. are shown above in TABLE 61. As indicated by the trend analysis and FIGS. 25 and 26, the trends in assay were well-represented by linear zero-order regression models. At the 25° C. conditions, the results did not pool intercepts but did pool slopes for the new formulation. The slope for this model was about 0.10%/month. The shortest estimated shelf-life based on trend extrapolation at 25° C. was about 49.9 months. This was a 17.7 month improvement compared to the 32.2 month value obtained from the 1 mL vasopressin results at 25° C. At 5° C., the vasopressin assay results were all 0.1% to 0.2% after 3 months.


Next, Glu4-AVP was measured in the formulations after storage for three months. Glu4-AVP is a deamidation product and major degradant of vasopressin. The concentration of Gly9-AVP was around <0.10% to 0.1% at time of release. The results at all stability intervals through three months were within the proposed limits (NMT 6%) as shown in TABLE 62 below.















TABLE 62









Intercept ±
Slope ±






ESL
Std. Error
Std. Error


Formulation
R2
Lot
(months)
(%)
(%/month)
Model Type





















10 mL
0.94
A
34
0.12% ± 0.03%
0.15% ± 0.01%
Different




B

0.11% ± 0.03%
0.14% ± 0.01%
slopes/different




C

0.13% ± 0.03%
0.11% ± 0.01%
intercepts


 1 mL
0.99
D
28.7
−0.05% ± 0.0% 
0.21% ± 0.0% 
Pooled




E



slopes/Pooled




F



intercepts









The stability study results for the 10 mL vasopressin formulations were compared to the 1 mL vasopressin formulation and plotted as shown in FIGS. 27 and 28. FIG. 27 shows the amount of Glu4-AVP when the formulations were stored at 25° C. in an upright position. The linear equation for each of the lots was as follows:






Y=0.0013+0.0013(month)  Lot A:






Y=0.0013+0.0013(month)  Lot B:






Y=0.0012+0.0012(month)  Lot C:






Y=−8.383×10−5+0.002036(month)  Lot D:






Y=−8.383×10−5+0.002036(month)  Lot E:






Y=−0.001102+0.002163(month)  Lot F:



FIG. 28 shows the amount of Glu4-AVP when the formulations were stored at 25° C. in an inverted position. The linear equation for each of the lots was as follows:






Y=0.0011+0.0016(month)  Lot A:






Y=0.0009+0.0014(month)  Lot B:






Y=0.0014+0.0009(month)  Lot C:






Y=−0.000599+0.002114(month)  Lot D:






Y=−0.000826+0.001997(month)  Lot E:






Y=−0.000473+0.00206(month)  Lot F:


The regression analysis and trend estimates for stability at 25° C. are shown above in TABLE 62. As indicated by the trend analysis and FIGS. 27 and 28, the trends in assay were well-represented by linear zero-order regression models. At the 25° C. conditions, the results did not pool intercepts or slopes for the 10 mL vasopressin formulation. The slopes for this model are from about 0.11% to 0.15% per month. The shortest estimated shelf-life based on trend extrapolation at 25° C. was about 34 months. This was a 5.3 month improvement compared to the 28.7 month value obtained from the 1 mL vasopressin results at 25° C. At 5° C., the vasopressin assay results were all 0.1% to 0.2% after 3 months.


The test results for D-Asn-AVP met the acceptance criteria (NMT 1%) at all conditions and intervals through three months on long-term, and accelerated, stability. All the results for D-Asn-AVP ranged from none detected to 0.1%. No significant changes were observed through three months under long-term and accelerated storage conditions.


The test results for Asp5-AVP met the acceptance criteria (NMT 1.5%) at all conditions and intervals through three months on long-term, and accelerated, stability. All the results for Asp5-AVP ranged from none detected to 0.1%. No significant changes were observed through three months under long-term and accelerated storage conditions.


The test results for AVP-Dimer met the acceptance criteria (NMT 1%) at all conditions and intervals through three months on long-term, and accelerated, stability. All the results for AVP-Dimer were none detected. No significant changes were observed through three months under long-term and accelerated storage conditions.


The test results for Acetyl-AVP met the acceptance criteria (NMT 1%) at all conditions and intervals through three months on long-term, and accelerated, stability. All the results for Acetyl-AVP ranged from 0.3% to 0.6%. No significant changes were observed through three months under long-term and accelerated storage conditions.


The test results for individual unidentified impurities met the acceptance criteria (NMT 1%) at all conditions and intervals through three months on long-term, and accelerated, stability. All unidentified impurities ranged from none detected to 0.4%. No significant changes were observed through three months under long-term and accelerated storage conditions.


The acceptance criteria for total impurities was NMT 17%. The total impurities included the sum of all identified and unidentified impurities. The values for total impurities were consistently low (1% to 1.2%) at the time of release. The results at all stability intervals through three months were within the proposed limits as shown in TABLE 63 below.















TABLE 63









Intercept ±
Slope ±






ESL
Std. Error
Std. Error


Formulation
R2
Lot
(months)
(%)
(%/month)
Model Type





















10 mL
0.88
A
52.3
1.07% ± 0.06%
0.26% ± 0.02%
Pooled




B

1.29% ± 0.06%

slopes/different




C

1.26% ± 0.06%

intercepts


 1 mL
0.98
D
32.3
0.67% ± 0.15%
0.49% ± 0.01%
Pooled




E



slopes/Pooled




F



intercepts









The stability study results for the 10 mL vasopressin formulations were compared to the 1 mL vasopressin formulation and plotted as shown in FIGS. 29 and 30. FIG. 29 shows the total impurities when the formulations were stored at 25° C. in an upright position. The linear equation for each of the lots was as follows:






Y=0.0108+0.0023(month)  Lot A:






Y=0.0126+0.0026(month)  Lot B:






Y=0.0123+0.0028(month)  Lot C:






Y=0.005617+0.004926(month)  Lot D:






Y=0.005617+0.004926(month)  Lot E:






Y=−0.008521+0.004824(month)  Lot F:



FIG. 30 shows the amount of total impurities when the formulations were stored at 25° C. in an inverted position. The linear equation for each of the lots was as follows:






Y=0.011+0.0025(month)  Lot A:






Y=0.0131+0.0026(month)  Lot B:






Y=0.0125+0.0025(month)  Lot C:






Y=0.006132+0.005015(month)  Lot D:






Y=0.006383+0.004907(month)  Lot E:






Y=0.008467+0.004705(month)  Lot F:


The regression analysis and trend estimates for stability at 25° C. are shown above in TABLE 63. As indicated by the trend analysis and FIGS. 29 and 30, the trends in assay were well-represented by linear zero-order regression models. At the 25° C. conditions, the results did not pool intercepts and did pool slopes for the 10 mL vasopressin formulation. The slopes for this pooled model is about 0.26% per month. The shortest estimated shelf-life based on trend extrapolation at 25° C. was about 52.3 months. This was a 20 month improvement compared to the 32.3 month value obtained from the 1 mL vasopressin results at 25° C. At 5° C., the vasopressin assay results were all 1% to 1.5% after 3 months.


The test results met the specification requirements for particulate matter under all study conditions. The highest reported values were about 13 counts (greater than 10 microns) and 1 count (greater than 25 microns).


The test results passed the bacterial endotoxin test, for which the acceptance criteria was NMT 29 EU/mL.


The test results passed the sterility test.


Overall, the data supported a 24-month refrigerated (2-8° C.) shelf life with a 12-month period out of refrigeration room temperature storage. A summary of the estimated shelf life for the measured vasopressin attributed is shown in TABLE 64 below.











TABLE 64







Estimated out of



Estimated out of Refrigerator
Refrigerator Storage


Trending
Storage Period (months) 10 mL
Period (months) 1 mL


COA
formulation
formulation

















Vasopressin
17.2
15.1


Glu4-AVP
34
28.7


Gly9-AVP
49.9
32.2


Total
52.3
32.3


Impurities









TABLES 65-76 provide the raw data for the different measured attributes of the 10 mL vasopressin formulations at 5° C. and 25° C./60% relative humidity.









TABLE 65







Lot A, Inverted, 5° C.










Test
Acceptance Criteria
Initial Amount
3 months





Assay
90%-110% LC
100.3% 
100.1% 











Related Substances
Gly9-AVP
NMT 6%
0.1%
0.2%



Glu4-AVP
NMT 6%
0.1%
0.2%



D-Asn-AVP
NMT 1%
ND
ND



Asp5-AVP
NMT 1.5%
ND
ND



AVP-Dimer
NMT 1%
ND
ND



Acetyl-AVP
NMT 1%
0.6%
0.6%



Unidentified
NMT 1%
RRT 0.84 = <0.1%
RRT 0.1 = <0.1%





RRT 1.03 = 0.2%
RRT 0.11 = <0.1%






RRT 0.37 = <0.1%






RRT 0.83 = <0.1%






RRT 1.03 = 0.2%



Total Impurities
NMT 17%
  1%
1.2%










Chlorobutanol
0.4%-0.6% w/v
0.5%
0.49% 


pH
2.5-4.5
3.8
3.7
















TABLE 66







Lot A, Upright, 5° C.










Test
Acceptance Criteria
Initial Amount
3 months





Assay
90%-110% LC
100.3% 
99.8% 











Related Substances
Gly9-AVP
NMT 6%
0.1%
0.2%



Glu4-AVP
NMT 6%
0.1%
0.2%



D-Asn-AVP
NMT 1%
ND
ND



Asp5-AVP
NMT 1.5%
ND
ND



AVP-Dimer
NMT 1%
ND
ND



Acetyl-AVP
NMT 1%
0.6%
0.6%



Unidentified
NMT 1%
RRT 0.84 = <0.1%
RRT 0.11 = <0.1%





RRT 1.03 = 0.2%
RRT 0.83 = <0.1%






RRT 1.03 = 0.2%



Total Impurities
NMT 17%
  1%
1.3%










Chlorobutanol
0.4%-0.6% w/v
0.5%
0.49% 


pH
2.5-4.5
3.8
3.8
















TABLE 67







Lot A, Inverted, 25° C./60% RH














Initial





Test
Acceptance Criteria
Amount
1 month
2 months
3 months





Assay
90%-110% LC
100.3% 
98.7% 
98.9% 
98.9% 













Related
Gly9-AVP
NMT 6%
0.1%
0.3%
0.4%
0.4%


Substances
Glu4-AVP
NMT 6%
0.1%
0.3%
0.4%
0.6%



D-Asn-AVP
NMT 1%
ND
ND
ND
<0.1% 



Asp5-AVP
NMT 1.5%
ND
ND
<0.1% 
<0.1% 



AVP-Dimer
NMT 1%
ND
ND
ND
ND



Acetyl-AVP
NMT 1%
0.6%
0.6%
0.6%
0.6%



Unidentified
NMT 1%
RRT 0.84 = <0.1%
RRT 0.84 = <0.1%
RRT 0.11 = <0.1%
RRT 0.8 = <0.1%





RRT 1.03 = 0.2% 
RRT 1.7 + <0.1% 
RRT 0.83 = <0.1%
1.6 = <0.1%






RRT 1.03 = 0.3% 
RRT 1.03 = 0.2% 
RRT 1.03 = 0.2%



Total
NMT 17%
  1%
1.5%
1.6%
1.8%



Impurities












Chlorobutanol
0.4%-0.6% w/v
0.5%
0.49% 
0.49% 
0.49% 


pH
2.5-4.5
3.8
3.7
3.7
3.7
















TABLE 68







Lot A, Upright, 25° C./60% RH














Initial





Test
Acceptance Criteria
Amount
1 month
2 months
3 months





Assay
90%-110% LC
100.3% 
99.4% 
98.8% 
99.1% 













Related
Gly9-AVP
NMT 6%
0.1%
0.3%
0.4%
0.4%


Substances
Glu4-AVP
NMT 6%
0.1%
0.3%
0.4%
0.5%



D-Asn-AVP
NMT 1%
ND
ND
ND
<0.1% 



Asp5-AVP
NMT 1.5%
ND
ND
<0.1% 
<0.1% 



AVP-Dimer
NMT 1%
ND
ND
ND
ND



Acetyl-AVP
NMT 1%
0.6%
0.6%
0.6%
0.6%



Unidentified
NMT 1%
RRT 0.84 = <0.1%
RRT 0.84 = <0.1%
RRT 0.11 = <0.1%
RRT 0.8 = <0.1%





RRT 1.03 = 0.2% 
RRT 1.7 + <0.1% 
RRT 0.83 = <0.1%
1.6 = <0.1%






RRT 1.03 = 0.3% 
RRT 1.03 = 0.2% 
RRT 1.03 = 0.2%



Total
NMT 17%
  1%
1.4%
1.6%
1.7%



Impurities












Chlorobutanol
0.4%-0.6% w/v
0.5%
0.49% 
0.49% 
0.49% 


pH
2.5-4.5
3.8
3.7
3.7
3.7
















TABLE 69







Lot B, Inverted, 5° C.










Test
Acceptance Criteria
Initial Amount
3 months





Assay
90%-110% LC
100% 
99.8% 











Related Substances
Gly9-AVP
NMT 6%
<0.1% 
0.2%



Glu4-AVP
NMT 6%
0.1%
0.2%



D-Asn-AVP
NMT 1%
0.1%
0.1%



Asp5-AVP
NMT 1.5%
ND
ND



AVP-Dimer
NMT 1%
ND
ND



Acetyl-AVP
NMT 1%
0.3%
0.4%



Unidentified
NMT 1%
RRT 0.84 = 0.4%
RRT 0.11 = <0.1%





RRT 1.03 = 0.3%
RRT 0.83 = 0.4%






RRT 1.03 = 0.3%



Total Impurities
NMT 17%
1.2%
1.5%










Chlorobutanol
0.4%-0.6% w/v
0.49% 
0.47% 


pH
2.5-4.5
3.8
3.8
















TABLE 70







Lot B, Upright, 5° C.










Test
Acceptance Criteria
Initial Amount
3 months





Assay
90%-110% LC
100% 
100% 











Related Substances
Gly9-AVP
NMT 6%
<0.1% 
0.1%



Glu4-AVP
NMT 6%
0.1%
0.1%



D-Asn-AVP
NMT 1%
0.1%
0.1%



Asp5-AVP
NMT 1.5%
ND
ND



AVP-Dimer
NMT 1%
ND
ND



Acetyl-AVP
NMT 1%
0.3%
0.4%



Unidentified
NMT 1%
RRT 0.84 = 0.4%
RRT 0.11 = <0.1%





RRT 1.03 = 0.3%
RRT 0.83 = 0.4%






RRT 1.03 = 0.4%



Total Impurities
NMT 17%
1.2%
1.5%










Chlorobutanol
0.4%-0.6% w/v
0.49% 
0.47% 


pH
2.5-4.5
3.8
3.8
















TABLE 71







Lot B, Inverted, 25° C./60% RH














Initial





Test
Acceptance Criteria
Amount
1 month
2 months
3 months





Assay
90%-110% LC
100% 
99.2% 
98.9% 
 98%













Related
Gly9-AVP
NMT 6%
<0.1% 
0.2%
0.3%
0.4%


Substances
Glu4-AVP
NMT 6%
0.1%
0.2%
0.4%
0.5%



D-Asn-AVP
NMT 1%
0.1%
ND
ND
0.1%



Asp5-AVP
NMT 1.5%
ND
ND
<0.1% 
<0.1% 



AVP-Dimer
NMT 1%
ND
0.2%
ND
ND



Acetyl-AVP
NMT 1%
0.3%
0.3%
0.4%
0.3%



Unidentified
NMT 1%
RRT 0.84 = 0.4%
RRT 0.84 = 0.4%
 RRT 0.11 = <0.1%
RRT 0.83 = 0.4%





RRT 1.03 = 0.3%
RRT 1.03 = 0.4%
RRT 0.83 = 0.4%
RRT 0.9 = <0.1%







RRT 1.03 = 0.3%
RRT 1.6 = 0.1% 








RRT 1.03 = 0.3%



Total
NMT 17%
1.2%
1.8%
1.7%
2.1%



Impurities












Chlorobutanol
0.4%-0.6% w/v
0.49% 
0.49% 
0.48% 
0.48% 


pH
2.5-4.5
3.8
3.7
3.7
3.8
















TABLE 72







Lot B, Upright, 25° C./60% RH














Initial





Test
Acceptance Criteria
Amount
1 month
2 months
3 months





Assay
90%-110% LC
100% 
99.1% 
99.2% 
98.5% 













Related
Gly9-AVP
NMT 6%
<0.1% 
0.2%
0.3%
0.4%


Substances
Glu4-AVP
NMT 6%
0.1%
0.3%
0.4%
0.5%



D-Asn-AVP
NMT 1%
0.1%
ND
<0.1% 
0.1%



Asp5-AVP
NMT 1.5%
ND
ND
<0.1% 
<0.1% 



AVP-Dimer
NMT 1%
ND
ND
ND
ND



Acetyl-AVP
NMT 1%
0.3%
0.3%
0.4%
0.3%



Unidentified
NMT 1%
RRT 0.84 = 0.4%
RRT 0.84 = 0.4%
RRT 0.11 = <0.1%
RRT 0.8 = 0.4%





RRT 1.03 = 0.3%
RRT 1.03 = 0.4%
RRT 0.83 = 0.4% 
 RRT 1.6 = <0.1%







RRT 1.03 = 0.3% 







RRT 1.56 = <0.1%



Total
NMT 17%
1.2%
1.6%
1.8%
  2%



Impurities












Chlorobutanol
0.4%-0.6% w/v
0.49% 
0.49% 
0.48% 
0.48% 


pH
2.5-4.5
3.8
3.8
3.7
3.8
















TABLE 73







Lot C, Inverted, 5° C.










Test
Acceptance Criteria
Initial Amount
3 months





Assay
90%-110% LC
100.3% 
100% 











Related Substances
Gly9-AVP
NMT 6%
<0.1% 
0.2%



Glu4-AVP
NMT 6%
0.1%
0.1%



D-Asn-AVP
NMT 1%
ND
0.1%



Asp5-AVP
NMT 1.5%
ND
ND



AVP-Dimer
NMT 1%
ND
ND



Acetyl-AVP
NMT 1%
0.3%
0.4%



Unidentified
NMT 1%
RRT 0.84 = 0.4%
RRT 0.11 = <0.1%





RRT 1.03 = 0.3%
RRT 0.59 = <0.1%






RRT 0.83 = 0.4%






RRT 1.03 = 0.4%



Total Impurities
NMT 17%
1.1%
1.5%










Chlorobutanol
0.4%-0.6% w/v
0.49% 
0.48% 


pH
2.5-4.5
3.8
3.8
















TABLE 74







Lot C, Upright, 5° C.










Test
Acceptance Criteria
Initial Amount
3 months





Assay
90%-110% LC
100.3% 
100.1% 











Related Substances
Gly9-AVP
NMT 6%
<0.1% 
0.2%



Glu4-AVP
NMT 6%
0.1%
0.1%



D-Asn-AVP
NMT 1%
ND
0.1%



Asp5-AVP
NMT 1.5%
ND
ND



AVP-Dimer
NMT 1%
ND
ND



Acetyl-AVP
NMT 1%
0.3%
0.4%



Unidentified
NMT 1%
RRT 0.84 = 0.4%
RRT 0.11 = <0.1%





RRT 1.03 = 0.3%
RRT 0.83 = 0.4%






RRT 1.03 = 0.4%



Total Impurities
NMT 17%
1.1%
1.5%










Chlorobutanol
0.4%-0.6% w/v
0.49% 
0.48% 


pH
2.5-4.5
3.8
3.7
















TABLE 75







Lot C, Inverted, 25° C./60% RH














Initial





Test
Acceptance Criteria
Amount
1 month
2 months
3 months





Assay
90%-110% LC
100.3% 
99.2% 
99.1% 
98.8% 













Related
Gly9-AVP
NMT 6%
<0.1% 
0.2%
0.3%
0.4%


Substances
Glu4-AVP
NMT 6%
0.1%
0.3%
0.3%
0.4%



D-Asn-AVP
NMT 1%
ND
0.1%
<0.1% 
0.1%



Asp5-AVP
NMT 1.5%
ND
ND
<0.1% 
<0.1% 



AVP-Dimer
NMT 1%
ND
ND
ND
ND



Acetyl-AVP
NMT 1%
0.3%
0.3%
0.4%
0.3%



Unidentified
NMT 1%
RRT 0.84 = 0.4%
RRT 0.84 = 0.4%
 RRT 0.11 = <0.1%
RRT 0.8 = 0.4%





RRT 1.03 = 0.3%
RRT 1.03 = 0.4%
RRT 0.83 = 0.4%
RRT 1 = 0.3%  







RRT 1.03 = 0.3%
 RRT 1.6 = <0.1%



Total
NMT 17%
1.1%
1.7%
1.8%
1.9%



Impurities












Chlorobutanol
0.4%-0.6% w/v
0.49% 
0.49% 
0.49% 
0.49% 


pH
2.5-4.5
3.8
3.7
3.7
3.8
















TABLE 76







Lot C, Upright, 25° C./60% RH














Initial





Test
Acceptance Criteria
Amount
1 month
2 months
3 months





Assay
90%-110% LC
100.3% 
99.1% 
99.3% 
98.9% 













Related
Gly9-AVP
NMT 6%
<0.1% 
0.2%
0.3%
0.4%


Substances
Glu4-AVP
NMT 6%
0.1%
0.3%
0.3%
0.5%



D-Asn-AVP
NMT 1%
ND
0.1%
<0.1% 
0.1%



Asp5-AVP
NMT 1.5%
ND
ND
<0.1% 
<0.1% 



AVP-Dimer
NMT 1%
ND
ND
ND
ND



Acetyl-AVP
NMT 1%
0.3%
0.3%
0.4%
0.3%



Unidentified
NMT 1%
RRT 0.84 = 0.4%
RRT 0.84 = 0.4%
 RRT 0.11 = <0.1%
RRT 0.8 = 0.4%





RRT 1.03 = 0.3%
RRT 1.03 = 0.4%
RRT 0.83 = 0.4%
RRT 1 = 0.3%  







RRT 1.03 = 0.3%
 RRT 1.6 = <0.1%



Total
NMT 17%
1.1%
1.7%
1.8%
  2%



Impurities












Chlorobutanol
0.4%-0.6% w/v
0.49% 
0.49% 
0.49% 
0.49% 


Ph
2.5-4.5
3.8
3.7
3.7
3.8









Example 19: Illustrative Regimen for Therapeutic Use of a 10 mL Vasopressin Formulation

Vasopressin is indicated to increase blood pressure in adults with vasodilatory shock (for example, adults who are post-cardiotomy or septic) who remain hypotensive despite fluids and catecholamines.


Preparation and Use of Vasopressin.

Dilute vasopressin with normal saline (0.9% sodium chloride) or 5% dextrose in water (D5W) to either 0.1 units/mL or 1 unit/mL for intravenous administration. Discard unused diluted solution after 18 hours at room temperature or 24 hours under refrigeration.


Vasopressin is prepared according to TABLE 77 below:











TABLE 77









Mix










Fluid Restriction?
Final Concentration
Vasopressin
Diluent





No
0.1 units/mL
2.5 mL (50 units)
500 mL


Yes
  1 unit/mL
  5 mL (100 units)
100 mL









Diluted vasopressin should be inspected for particulate matter and discoloration prior to use whenever solution and container permit.


The goal of treatment with vasopressin is optimization of perfusion to critical organs, but aggressive treatment can compromise perfusion of organs, like the gastrointestinal tract, for which function is difficult to monitor.


For post-cardiotomy shock, a dose of 0.03 units/minute is used as a starting point. For septic shock, a dose of 0.01 units/minute is used as a starting point. If the target blood pressure response is not achieved, the dose is titrated up by 0.005 units/minute at 10- to 15-minute intervals. The maximum dose for post-cardiotomy shock is 0.1 units/minute and for septic shock 0.07 units/minute. After target blood pressure has been maintained for 8 hours without the use of catecholamines, the vasopressin can be tapered by 0.005 units/minute every hour as tolerated to maintain target blood pressure.


Vasopressin is provided as an injection at 20 units/mL. Vasopressin is supplied in vials as follows:

    • A carton of 25 multiple dose vials each containing 1 mL vasopressin each at 20 units/mL of vasopressin.
    • A carton of 1 multiple dose vial containing 10 mL vasopressin at 20 units/mL.
    • A carton of 5 multiple dose vials each containing 10 mL vasopressin at 20 unit/mL of vasopressin.


Vasopressin is stored between 2-8° C., and is not to be frozen.


The vasopressin vials can be held up to 12 months upon removal from refrigeration to room temperature storage conditions (20-25° C.), anytime within the labeled shelf life. Once removed from refrigeration, unopened vials should be marked to indicate the revised 12 month expiration date. If the original expiration date is shorter than the revised expiration date, then the shorter date should be used.


The 1 mL vial should be discarded 48 hours after the first puncture. The 10 mL vial can be discarded 30 days after the first puncture. The storage conditions and expiration periods are summarized in TABLE 78 below:













TABLE 78







Unopened
Unopened Room
Opened



Refrigerated
Temperature (20-25° C.;
(after first



(2-8° C.)
do not store above 25° C.)
puncture)



















1 mL
Until printed
12 months or until printed
48 hours


vial
expiration date
expiration date, whichever is




earlier


10 mL
Until printed
12 months or until printed
30 days


vial
expiration date
expiration date, whichever is




earlier









Contraindications, Adverse Reactions, and Drug-Drug Interactions.

Vasopressin is contraindicated in patients with known allergy or hypersensitivity to 8-L-arginine vasopressin or chlorobutanol. Additionally, use of vasopressin in patients with impaired cardiac response can worsen cardiac output.


Adverse reactions have been observed with the use of vasopressin, which adverse reactions include bleeding/lymphatic system disorders, specifically, hemorrhagic shock, decreased platelets, intractable bleeding; cardiac disorders, specifically, right heart failure, atrial fibrillation, bradycardia, myocardial ischemia; gastrointestinal disorders, specifically, mesenteric ischemia; hepatobiliary disorders, specifically, increased bilirubin levels; renal/urinary disorders, specifically, acute renal insufficiency; vascular disorders, specifically, distal limb ischemia; metabolic disorders, specifically, hyponatremia; and skin disorders, specifically, and ischemic lesions.


These reactions are reported voluntarily from a population of uncertain size. Thus, reliable estimation of frequency or establishment of a causal relationship to drug exposure is unlikely.


Vasopressin has been observed to interact with other drugs. For example, use of vasopressin with catecholamines is expected to result in an additive effect on mean arterial blood pressure and other hemodynamic parameters. Use of vasopressin with indomethacin can prolong the effect of vasopressin on cardiac index and systemic vascular resistance. Indomethacin more than doubles the time to offset for vasopressin's effect on peripheral vascular resistance and cardiac output in healthy subjects.


Further, use of vasopressin with ganglionic blocking agents can increase the effect of vasopressin on mean arterial blood pressure. The ganglionic blocking agent tetra-ethylammonium increases the pressor effect of vasopressin by 20% in healthy subjects.


Use of vasopressin with furosemide increases the effect of vasopressin on osmolar clearance and urine flow. Furosemide increases osmolar clearance 4-fold and urine flow 9-fold when co-administered with exogenous vasopressin in healthy subjects.


Use of vasopressin with drugs suspected of causing SIADH (Syndrome of inappropriate antidiuretic hormone secretion), for example, SSRIs, tricyclic antidepressants, haloperidol, chlorpropamide, enalapril, methyldopa, pentamidine, vincristine, cyclophosphamide, ifosfamide, and felbamate can increase the pressor effect in addition to the antidiuretic effect of vasopressin. Additionally, use of vasopressin with drugs suspected of causing diabetes insipidus for example, demeclocycline, lithium, foscarnet, and clozapine can decrease the pressor effect in addition to the antidiuretic effect of vasopressin.


Indomethacin can more than double the time to offset for vasopressin's effect on peripheral vascular resistance and cardiac output in healthy subjects.


Halothane, morphine, fentanyl, alfentanyl and sufentanyl do not impact exposure to endogenous vasopressin.


Use of Vasopressin in Specific Populations.

Vasopressin is a Category C Drug for Pregnancy.


Due to a spillover into the blood of placental vasopressinase, the clearance of exogenous and endogenous vasopressin increases gradually over the course of a pregnancy. During the first trimester of pregnancy the clearance is only slightly increased. However, by the third trimester the clearance of vasopressin is increased about 4-fold and at term up to 5-fold. Due to the increased clearance of vasopressin in the second and third trimester, the dose of vasopressin can be up-titrated to doses exceeding 0.1 units/minute in post-cardiotomy shock and 0.07 units/minute in septic shock. Vasopressin can produce tonic uterine contractions that could threaten the continuation of pregnancy. After delivery, the clearance of vasopressin returns to preconception levels.


Overdosage.

Overdosage with vasopressin can be expected to manifest as a consequence of vasoconstriction of various vascular beds, for example, the peripheral, mesenteric, and coronary vascular beds, and as hyponatremia. In addition, overdosage of vasopressin can lead less commonly to ventricular tachyarrhythmias, including Torsade de Pointes, rhabdomyolysis, and non-specific gastrointestinal symptoms. Direct effects of vasopressin overdose can resolve within minutes of withdrawal of treatment.


Pharmacology of Vasopressin.

Vasopressin is a polypeptide hormone that causes contraction of vascular and other smooth muscles and antidiuresis, which can be formulated as a sterile, aqueous solution of synthetic arginine vasopressin for intravenous administration. The 1 mL solution contains vasopressin (20 units/mL), chlorobutanol, NF 0.5% as a preservative, water for injection, USP, and sodium acetate buffer adjusted to a pH of 3.8.


The chemical name of vasopressin is Cyclo (1-6) L-Cysteinyl-L-Tyrosyl-L-Phenylalanyl-L-Glutaminyl-L-Asparaginyl-L-Cysteinyl-L-Prolyl-L-Arginyl-L-Glycinamide. Vasopressin is a white to off-white amorphous powder, freely soluble in water. The structural formula of vasopressin is:




embedded image


Molecular Formula: C46H65N15O12S2; Molecular Weight: 1084.23


One mg of vasopressin is equivalent to 530 units. Alternatively, one mg of vasopressin is equivalent to 470 units.


The vasoconstrictive effects of vasopressin are mediated by vascular V1 receptors. Vascular V1 receptors are directly coupled to phospholipase C, resulting in release of calcium, leading to vasoconstriction. In addition, vasopressin stimulates antidiuresis via stimulation of V2 receptors, which are coupled to adenyl cyclase.


At therapeutic doses, exogenous vasopressin elicits a vasoconstrictive effect in most vascular beds including the splanchnic, renal, and cutaneous circulation. In addition, vasopressin at pressor doses triggers contractions of smooth muscles in the gastrointestinal tract mediated by muscular V1-receptors and release of prolactin and ACTH via V3 receptors. At lower concentrations typical for the antidiuretic hormone, vasopressin inhibits water diuresis via renal V2 receptors. In patients with vasodilatory shock, vasopressin in therapeutic doses increases systemic vascular resistance and mean arterial blood pressure, and reduces the dose requirements for norepinephrine.


Vasopressin tends to decrease heart rate and cardiac output. The pressor effect is proportional to the infusion rate of exogenous vasopressin. Onset of the pressor effect of vasopressin is rapid, and the peak effect occurs within 15 minutes. After stopping the infusion, the pressor effect fades within 20 minutes. There is no evidence for tachyphylaxis or tolerance to the pressor effect of vasopressin in patients.


At infusion rates used in vasodilatory shock (0.01-0.1 units/minute), the clearance of vasopressin is 9 to 25 mL/min/kg in patients with vasodilatory shock. The apparent half-life of vasopressin at these levels is ≤10 minutes. Vasopressin is predominantly metabolized and only about 6% of the dose is excreted unchanged in urine. Animal experiments suggest that the metabolism of vasopressin is primarily by liver and kidney. Serine protease, carboxipeptidase and disulfide oxido-reductase cleave vasopressin at sites relevant for the pharmacological activity of the hormone. Thus, the generated metabolites are not expected to retain important pharmacological activity.


Carcinogenesis, Mutagenesis, Impairment of Fertility.

Vasopressin was found to be negative in the in vitro bacterial mutagenicity (Ames) test and the in vitro Chinese hamster ovary (CHO) cell chromosome aberration test. In mice, vasopressin can have an effect on function and fertilizing ability of spermatozoa.


Clinical Studies.

Increases in systolic and mean blood pressure following administration of vasopressin were observed in seven studies in septic shock and eight studies in post-cardiotomy vasodilatory shock.


Embodiments

The following non-limiting embodiments provide illustrative examples of the invention, but do not limit the scope of the invention.


In some embodiments, the invention provides a pharmaceutical composition comprising, in a unit dosage form: a) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin, or a pharmaceutically-acceptable salt thereof; and b) a polymeric pharmaceutically-acceptable excipient in an amount that is from about 1% to about 10% by mass of the unit dosage form or the pharmaceutically-acceptable salt thereof, wherein the unit dosage form exhibits from about 5% to about 10% less degradation of the vasopressin or the pharmaceutically-acceptable salt thereof after storage for about 1 week at about 60° C. than does a corresponding unit dosage form, wherein the corresponding unit dosage form consists essentially of: A) vasopressin, or a pharmaceutically-acceptable salt thereof; and B) a buffer having acidic pH. In some embodiments, the polymeric pharmaceutically-acceptable excipient comprises a polyalkylene oxide moiety. In some embodiments, the polymeric pharmaceutically-acceptable excipient is a polyethylene oxide. In some embodiments, the polymeric pharmaceutically-acceptable excipient is a poloxamer. In some embodiments, the unit dosage form has an amount of the polymeric pharmaceutically-acceptable excipient that is about 1% the amount of the vasopressin or the pharmaceutically-acceptable salt thereof. In some embodiments, the first unit dosage form exhibits about 10% less degradation of the vasopressin or the pharmaceutically-acceptable salt thereof after storage for about 1 week at about 60° C. than does the corresponding unit dosage form. In some embodiments, the unit dosage form further comprises SEQ ID NO. 2. In some embodiments, the composition further comprises SEQ ID NO. 3. In some embodiments, the composition further comprises SEQ ID NO. 4. In some embodiments, the unit dosage form is an injectable of about 1 mL volume. In some embodiments, the unit dosage form consists essentially of: a) about 0.04 mg/mL of vasopressin, or the pharmaceutically-acceptable salt thereof; b) the polymeric pharmaceutically-acceptable excipient in an amount that is from about 1% to about 10% by mass of the vasopressin or the pharmaceutically-acceptable salt thereof; and c) a plurality of peptides, wherein each of the peptides has from 88% to 90% sequence homology to the vasopressin or the pharmaceutically-acceptable salt thereof. In some embodiments, one of the plurality of peptides is SEQ ID NO.: 2. In some embodiments, one of the plurality of peptides is SEQ ID NO.:3. In some embodiments, wherein one of the plurality of peptides is SEQ ID NO.: 4. In some embodiments, the buffer has a pH of about 3.5.

Claims
  • 1-15. (canceled)
  • 16. A method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; optionally chlorobutanol; acetic acid, acetate, or a combination thereof; iv) 0-2% vasopressin degradation products; and v) water;
  • 17. The method of claim 16, further comprising drawing from the dispensing region of the container a third time a third portion of the pharmaceutical composition.
  • 18. The method of claim 17, wherein the third time that the third portion of the pharmaceutical composition is drawn from the dispensing region of the container occurs after the second time that the second portion of the pharmaceutical composition is drawn from the dispensing region of the container, and wherein the third time that the third portion of the pharmaceutical composition is drawn from the dispending region of the container occurs within about 30 days of the first time that the first portion of the pharmaceutical composition is drawn from the dispensing region of the container.
  • 19. The method of claim 18, further comprising administering the third portion of the pharmaceutical composition to the human, wherein the administration of the third portion occurs after the administration of the second portion, wherein the third administration occurs within about 30 days of the first time that the first portion of the pharmaceutical composition is drawn from the dispensing region of the container.
  • 20. The method of claim 16, wherein the human's hypotension is associated with vasodilatory shock.
  • 21. The method of claim 20, wherein the vasodilatory shock is post-cardiotomy shock.
  • 22. The method of claim 21, wherein the vasodilatory shock is septic shock.
  • 23. The method of claim 22, wherein the administration provides to the human from about 0.01 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute to about 0.07 units of vasopressin or the pharmaceutically-acceptable salt thereof per minute.
  • 24. The method of claim 16, wherein the pharmaceutical composition further comprises chlorobutanol.
  • 25. The method of claim 16, wherein a shelf-life of the pharmaceutical composition is from about 10 to about 20 months at 25° C.
  • 26. The method of claim 16, further comprising diluting the first portion of the pharmaceutical composition with a diluent prior to the intravenous administration of the first portion of the pharmaceutical composition.
  • 27. A method of increasing blood pressure in a human in need thereof, the method comprising: a) providing a pharmaceutical composition for intravenous administration comprising: i) from about 0.01 mg/mL to about 0.07 mg/mL of vasopressin or a pharmaceutically-acceptable salt thereof; optionally chlorobutanol; acetic acid, acetate, or a combination thereof; iv) 0-2% vasopressin degradation products; and v) water;
  • 28. The method of claim 27, wherein the total impurities comprise SEQ ID NO.: 2, and SEQ ID NO.: 2 is present in the container in an amount of 0.1% to 0.5% at the second time that the second portion of the pharmaceutical composition is drawn from the dispensing region of the container.
  • 29. The method of claim 27, wherein the total impurities comprise SEQ ID NO.: 4, and SEQ ID NO.: 4 is present in the container in an amount of 0.1% to 0.5% at the second time that the second portion of the pharmaceutical composition is drawn from the dispensing region of the container.
  • 30. The method of claim 27, wherein the total impurities comprise SEQ ID NO.: 7, and SEQ ID NO.: 7 is present in the container in an amount of 0.1% to 1% at the second time that the second portion of the pharmaceutical composition is drawn from the dispensing region of the container.
  • 31. The method of claim 27, wherein room temperature is 22° C. to 28° C.
  • 32. The method of claim 27, wherein room temperature is 25° C.
  • 33. The method of claim 27, further comprising diluting the first portion of the pharmaceutical composition with a diluent prior to the intravenous administration of the first portion of the pharmaceutical composition.
CROSS REFERENCE

This Application is a continuation of U.S. application Ser. No. 15/606,442, filed on May 26, 2017, which is a continuation-in-part of U.S. application Ser. No. 15/426,693, filed Feb. 7, 2017, now U.S. Pat. No. 9,744,209, which is a continuation-in-part of U.S. application Ser. No. 15/289,640, filed Oct. 10, 2016, now U.S. Pat. No. 9,687,526, which is a continuation-in-part of U.S. application Ser. No. 14/717,877, filed May 20, 2015, now U.S. Pat. No. 9,744,239, which is a continuation of U.S. application Ser. No. 14/610,499, filed Jan. 30, 2015, each of which is incorporated herein by reference in its entirety.

Continuations (2)
Number Date Country
Parent 15606442 May 2017 US
Child 15901412 US
Parent 14610499 Jan 2015 US
Child 14717877 US
Continuation in Parts (3)
Number Date Country
Parent 15426693 Feb 2017 US
Child 15606442 US
Parent 15289640 Oct 2016 US
Child 15426693 US
Parent 14717877 May 2015 US
Child 15289640 US