L-ornithine phenyl acetate and methods of making thereof

Information

  • Patent Grant
  • 11161802
  • Patent Number
    11,161,802
  • Date Filed
    Friday, January 31, 2020
    4 years ago
  • Date Issued
    Tuesday, November 2, 2021
    2 years ago
Abstract
Disclosed herein are forms of L-ornithine phenyl acetate and methods of making the same. A crystalline form may, in some embodiments, be Forms I, II, III and V, or mixtures thereof. The crystalline forms may be formulated for treating subjects with liver disorders, such as hepatic encephalopathy. Accordingly, some embodiments include formulations and methods of administering L-ornithine phenyl acetate.
Description
BACKGROUND
Field

The present application relates to the fields of pharmaceutical chemistry, biochemistry, and medicine. In particular, it relates to L-ornithine phenyl acetate salts and methods of making and using the same.


Description

Hyperammonemia is a hallmark of liver disease and is characterized by an excess of ammonia in the bloodstream. Hepatic encephalopathy is a primary clinical consequence of progressive hyperammonemia and is a complex neuropsychiatric syndrome, which may complicate acute or chronic hepatic failure. It is characterized by changes in mental state including a wide range of neuropsychiatric symptoms ranging from minor signs of altered brain function to overt psychiatric and/or neurological symptoms, or even deep coma. The accumulation of unmetabolized ammonia has been considered as the main factor involved in the pathogenesis of hepatic encephalopathy, but additional mechanisms may be associated.


L-Ornithine monohydrochloride and other L-ornithine salts are available for their use in the treatment of hyperammonemia and hepatic encephalopathy. For example, U.S. Publication No. 2008/0119554, which is hereby incorporated by reference in its entirety, describes compositions of L-ornithine and phenyl acetate for the treatment of hepatic encephalopathy. L-ornithine has been prepared by enzymatic conversion methods. For example, U.S. Pat. Nos. 5,405,761 and 5,591,613, both of which are hereby incorporated by reference in their entirety, describe enzymatic conversion of arginine to form L-ornithine salts. Sodium phenyl acetate is commercially available, and also available as an injectable solution for the treatment of acute hyperammonemia. The injectable solution is marketed as AMMONUL.


Although salt forms may exhibit improved degradation properties, certain salts, particularly sodium or chloride salts, may be undesirable when treating patients having diseases associated with the liver disease, such as hepatic encephalopathy. For example, a high sodium intake may be dangerous for cirrhotic patients prone to ascites, fluid overload and electrolyte imbalances. Similarly, certain salts are difficult to administer intravenously because of an increased osmotic pressure, i.e., the solution is hypertonic. High concentrations of excess salt may require diluting large volumes of solution for intravenous administration which, in turn, leads to excessive fluid overload. Accordingly, there exists a need for the preparation of L-ornithine and phenyl acetate salts which are favorable for the treatment of hepatic encephalopathy or other conditions where fluid overload and electrolyte imbalance are prevalent.


SUMMARY

Some embodiments disclosed herein include a composition comprising a crystalline form of L-ornithine phenyl acetate.


In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising at least one characteristic peak, wherein said characteristic peak is selected from the group consisting of approximately 6.0°, 13.9°, 14.8°, 17.1°, 17.8° and 24.1° 2θ. In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising at least three characteristic peaks, wherein said characteristic peaks are selected from the group consisting of approximately 6.0°, 13.9°, 14.8°, 17.1°, 17.8° and 24.1° 2θ. In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising characteristic peaks at approximately 6.0°, 13.9°, 14.8°, 17.1°, 17.8° and 24.1° 2θ.


In some embodiments, the crystalline form has a melting point of about 202° C. In some embodiments, the crystalline form exhibits a single crystal X-ray crystallographic analysis with crystal parameters approximately equal to the following: unit cell dimensions: a=6.594(2) Å, b=6.5448(18) Å, c=31.632(8) Å, α=90°, β=91.12(3°), γ=90°; Crystal System: Monoclinic; and Space Group: P21. In some embodiments, the crystalline form is represented by the formula [C5H13N2O2][C8H7O2].


Some embodiments have the crystalline form exhibit an X-ray powder diffraction pattern comprising at least one characteristic peak, wherein said characteristic peak is selected from the group consisting of approximately 4.9°, 13.2°, 17.4°, 20.8° and 24.4° 2θ. In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising at least three characteristic peaks, wherein said characteristic peaks are selected from the group consisting of approximately 4.9°, 13.2°, 17.4°, 20.8° and 24.4° 2θ. In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising characteristic peaks at approximately 4.9°, 13.2°, 17.4°, 20.8° and 24.4° 2θ.


Some embodiments have the crystalline form comprising water and/or ethanol molecules. In some embodiments, the crystalline form comprises about 11% by weight of said molecules as determined by thermogravimetric analysis. In some embodiments, the crystalline form is characterized by differential scanning calorimetry as comprising an endotherm at about 35° C. In some embodiments, the crystalline has a melting point at about 203° C.


Some embodiments have the crystalline form exhibiting a single crystal X-ray crystallographic analysis with crystal parameters approximately equal to the following: unit cell dimensions: a=5.3652(4) Å, b=7.7136(6) Å, c=20.9602(18) Å, α=90°, β=94.986(6°), γ=90°; Crystal System: Monoclinic; and Space Group: P21. In some embodiments, the crystalline form is represented by the formula [C5H13N2O2][C8H7O2]EtOH.H2O.


Some embodiments have the crystalline form exhibiting an X-ray powder diffraction pattern comprising at least one characteristic peak, wherein said characteristic peak is selected from the group consisting of approximately 5.8°, 14.1°, 18.6°, 19.4°, 22.3° and 24.8° 2θ. In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising at least three characteristic peaks, wherein said characteristic peaks are selected from the group consisting of approximately 5.8°, 14.1°, 18.6°, 19.4°, 22.3° and 24.8° 2θ. In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising characteristic peaks at approximately 5.8°, 14.1°, 18.6°, 19.4°, 22.3° and 24.8° 2θ.


In some embodiments, the crystalline form is characterized by differential scanning calorimetry as comprising an endotherm at about 40° C. In some embodiments, the crystalline form comprises a melting point at about 203° C.


In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising at least one characteristic peak, wherein said characteristic peak is selected from the group consisting of approximately 13.7°, 17.4°, 19.8°, 20.6° and 23.7° 2θ. In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising at least three characteristic peaks, wherein said characteristic peak is selected from the group consisting of approximately 13.7°, 17.4°, 19.8°, 20.6° and 23.7° 2θ. In some embodiments, the crystalline form exhibits an X-ray powder diffraction pattern comprising characteristic peaks at approximately 13.7°, 17.4°, 19.8°, 20.6° and 23.7° 2θ.


In some embodiments, the crystalline form is characterized by differential scanning calorimetry as comprising an endotherm at about 174° C. In some embodiments, the crystalline form has a melting point of about 196° C. In some embodiments, the crystalline form comprises a pharmaceutically acceptable carrier.


Some embodiments disclosed herein have a composition comprising: at least about 50% by weight of a crystalline form of L-ornithine phenyl acetate salt and at least about 0.01% by weight benzoic acid or a salt thereof.


In some embodiments, the composition comprises at least about 0.10% by weight benzoic acid or a salt thereof. In some embodiments, the composition comprises no more than 5% by weight benzoic acid or a salt thereof. In some embodiments, the composition comprises no more than 1% by weight benzoic acid or a salt thereof.


In some embodiments, the composition further comprises at least 10 ppm silver. In some embodiments, comprises at least 20 ppm silver. In some embodiments, the composition further comprises at least 25 ppm silver. In some embodiments, comprises no more than 600 ppm silver. In some embodiments, composition comprises no more than 100 ppm silver. In some embodiments, the composition comprises no more than 65 ppm silver.


In some embodiments, about 50 mg/mL of the composition in water is isotonic with body fluids. In some embodiments, the isotonic solution has an osmolality in the range of about 280 to about 330 mOsm/kg.


In some embodiments, the composition has a density in the range of about 1.1 to about 1.3 kg/m3.


Some embodiments disclosed herein include a process for making L-ornithine phenyl acetate salt comprising: intermixing an L-ornithine salt, a benzoate salt and a solvent to form an intermediate solution; intermixing phenyl acetate with said intermediate solution; and isolating a composition comprising at least 70% crystalline L-ornithine phenyl acetate by weight.


In some embodiments, the process comprises removing at least a portion of a salt from said intermediate solution before intermixing the phenyl acetate, wherein said salt is not an L-ornithine salt. In some embodiments, the process comprises adding hydrochloric acid before removing at least a portion of the salt.


In some embodiments, intermixing the L-ornithine, the benzoate salt and the solvent comprises: dispersing the L-ornithine salt in water to form a first solution; dispersing the benzoate salt in DMSO to form a second solution; and intermixing said first solution and said second solution to form said solution.


In some embodiments, the composition comprises at least about 0.10% by weight benzoate salt. In some embodiments, composition comprises no more than 5% by weight benzoate salt. In some embodiments, composition comprises no more than 1% by weight benzoate salt.


In some embodiments, the L-ornithine salt is L-ornithine hydrochloride. In some embodiments, the benzoate salt is silver benzoate.


In some embodiments, the composition comprises at least 10 ppm silver. In some embodiments, composition comprises at least 20 ppm silver. In some embodiments, the composition comprises at least 25 ppm silver. In some embodiments, the composition comprises no more than 600 ppm silver. In some embodiments, the composition comprises no more than 100 ppm silver. In some embodiments, the composition comprises no more than 65 ppm silver.


In some embodiments, the phenyl acetate is in an alkali metal salt. In some embodiments, the alkali metal salt is sodium phenyl acetate.


In some embodiments, the composition comprises no more than 100 ppm sodium. In some embodiments, the composition comprises no more than 20 ppm sodium.


In some embodiments, the L-ornithine is in a halide salt. In some embodiments, the halide salt is L-ornithine hydrochloride.


In some embodiments, the composition comprises no more than 0.1% by weight chloride. In some embodiments, the composition comprises no more than 0.01% by weight chloride.


Some embodiments disclosed herein include a composition obtained by any of the processes disclosed herein.


Some embodiments disclosed herein include a process for making L-ornithine phenyl acetate salt comprising: increasing the pH value of a solution comprising an L-ornithine salt at least until an intermediate salt precipitates, wherein said intermediate salt is not an L-ornithine salt; isolating the intermediate salt from said solution; intermixing phenyl acetic acid with said solution; and isolating L-ornithine phenyl acetate salt from said solution.


In some embodiments, the pH value is increased to at least 8.0. In some embodiments, the pH value is increased to at least 9.0. In some embodiments, increasing the pH value comprises adding a pH modifier selected from the group consisting of sodium hydroxide, potassium hydroxide, sodium methoxide, potassium t-butoxide, sodium carbonate, calcium carbonate, dibutylamine, tryptamine, sodium hydride, calcium hydride, butyllithium, ethylmagnesium bromide or combinations thereof.


Some embodiments disclosed herein include a method of treating or ameliorating hyperammonemia in a subject by administering a therapeutically effective amount of a crystalline form of L-ornithine phenyl acetate salt.


In some embodiments, the crystalline form is administered orally.


In some embodiments, the crystalline form is selected from the group consisting of Form I, Form II, Form III, Form V, wherein: Form I exhibits an X-ray powder diffraction pattern having characteristic peaks at approximately 4.9°, 13.2°, 17.4°, 20.8° and 24.4° 2θ; Form II exhibits an X-ray powder diffraction pattern having characteristic peaks at approximately 6.0°, 13.9°, 14.8°, 17.1°, 17.8° and 24.1° 2θ; Form III exhibits an X-ray powder diffraction pattern having characteristic peaks at approximately 5.8°, 14.1°, 18.6°, 19.4°, 22.3° and 24.8° 2θ; and Form V exhibits an X-ray powder diffraction pattern having characteristic peaks at approximately 13.7°, 17.4°, 19.8°, 20.6° and 23.7° 2θ.


In some embodiments, the crystalline form is Form I. In some embodiments, the crystalline form is Form II. In some embodiments, the crystalline form is Form III. In some embodiments, the crystalline form is Form V.


In some embodiments, the at least two crystalline forms selected from the group consisting of Form I, Form II, Form III and Form V, are administered. In some embodiments, the at least two crystalline forms are administered at about the same time.


In some embodiments, the crystalline form is administered from 1 to 3 times daily. In some embodiments, the therapeutically effective amount is in the range of about 500 mg to about 50 g.


In some embodiments, the subject is identified as having hepatic encephalopathy. In some embodiments, the subject is identified as having hyperammonemia.


Some embodiments disclosed herein include a process for making L-ornithine phenyl acetate salt comprising: intermixing an L-ornithine salt, silver phenyl acetate and a solvent to form a solution, wherein the L-ornithine salt is a halide salt; and isolating L-ornithine phenyl acetate from said solution.


Some embodiments disclosed herein include a method of treating or ameliorating hyperammonemia comprising intravenously administering a therapeutically effective amount of a solution comprising L-ornithine phenyl acetate, wherein said therapeutically effective amount comprises no more than 500 mL of said solution.


In some embodiments, the solution comprises at least about 25 mg/mL of L-ornithine phenyl acetate. In some embodiments, the solution comprises at least about 40 mg/mL of L-ornithine phenyl acetate. In some embodiments, the solution comprises no more than 300 mg/mL. In some embodiments, the solution is isotonic with body fluid.


Some embodiments disclosed herein include a method of compressing L-ornithine phenyl acetate, the method comprising applying pressure to a metastable form of L-ornithine phenyl acetate to induce a phase change.


In some embodiments, the metastable form is amorphous. In some embodiments, the metastable form exhibits an X-ray powder diffraction pattern comprising at least one characteristic peak, wherein said characteristic peak is selected from the group consisting of approximately 4.9°, 13.2°, 20.8° and 24.4° 2θ.


In some embodiments, the pressure is applied for a predetermined time. In some embodiments, the predetermined time is about 1 second or less. In some embodiments, the pressure is at least about 500 psi.


In some embodiments, the phase change yields a composition having a density in the range of about 1.1 to about 1.3 kg/m3 after applying the pressure.


In some embodiments, the phase change yields a composition exhibiting an X-ray powder diffraction pattern comprising at least one characteristic peak, wherein said characteristic peak is selected from the group consisting of approximately 6.0°, 13.9°, 14.8°, 17.1°, 17.8° and 24.1° 2θ.


Some embodiments disclosed herein include a composition obtained by applying pressure to a metastable form of L-ornithine phenyl acetate to induce a phase change.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is an X-ray powder diffraction pattern of Form I.



FIG. 2 shows differential scanning calorimetry results for Form I.



FIG. 3 shows thermogravimetric gravimetric/differential thermal analysis of Form I.



FIG. 4 shows the 1H nuclear magnetic resonance spectrum obtained from a sample of Form I.



FIG. 5 shows dynamic vapor sorption results for Form I.



FIG. 6 is an X-ray powder diffraction pattern of Form II.



FIG. 7 shows differential scanning calorimetry results for Form II.



FIG. 8 shows thermogravimetric gravimetric/differential thermal analysis of Form II.



FIG. 9 shows the 1H nuclear magnetic resonance spectrum obtained from a sample of Form II.



FIG. 10 shows dynamic vapor sorption results for Form II.



FIG. 11 is an X-ray powder diffraction pattern of Form III.



FIG. 12 shows differential scanning calorimetry results for Form III.



FIG. 13 shows thermogravimetric gravimetric/differential thermal analysis of Form III.



FIG. 14 shows the 1H nuclear magnetic resonance spectrum obtained from a sample of Form III.



FIG. 15 shows dynamic vapor sorption results for Form III.



FIG. 16 is an X-ray powder diffraction pattern of Form V.



FIG. 17 shows differential scanning calorimetry results for Form V.



FIG. 18 shows thermogravimetric gravimetric/differential thermal analysis of Form V.



FIG. 19 shows the 1H nuclear magnetic resonance spectrum obtained from a sample of Form V.



FIG. 20 shows dynamic vapor sorption results for Form V.



FIG. 21 shows the 1H nuclear magnetic resonance spectrum obtained from a sample of L-ornithine benzoate.



FIG. 22 shows the 1H nuclear magnetic resonance spectrum obtained from a sample of L-ornithine phenyl acetate.





DETAILED DESCRIPTION

Disclosed herein are methods of making L-ornithine phenyl acetate salts, and in particular, crystalline forms of said salt. These methods permit large-scale production of pharmaceutically acceptable forms of L-ornithine phenyl acetate using economical processes. Moreover, crystalline forms of L-ornithine phenyl acetate, including Forms I, II, III and V are also disclosed. The L-ornithine phenyl acetate salts permit intravenous administration with negligible concomitant sodium load, and therefore minimize the amount of i.v. fluid that is required.


The present application relates to new crystalline forms of L-ornithine phenyl acetate salts, as well as methods of making and using L-ornithine phenyl acetate salts. The salt advantageously exhibits long-term stability without significant amounts of sodium or chloride. As a result, L-ornithine phenyl acetate is expected to provide an improved safety profile compared to other salts of L-ornithine and phenyl acetate. Also, L-ornithine phenyl acetate exhibits lower tonicity compared to other salts, and therefore can be administered intravenously at higher concentrations. Accordingly, L-ornithine phenyl acetate is expected to provide significant clinical improvements for the treatment of hepatic encephalopathy.


The present application also relates to various polymorphs of L-ornithine phenyl acetate. The occurrence of different crystal forms (polymorphism) is a property of some molecules and molecular complexes. Salt complexes, such as L-ornithine phenyl acetate, may give rise to a variety of solids having distinct physical properties like melting point, X-ray diffraction pattern, infrared absorption fingerprint and NMR spectrum. The differences in the physical properties of polymorphs result from the orientation and intermolecular interactions of adjacent molecules (complexes) in the bulk solid. Accordingly, polymorphs can be distinct solids sharing the same active pharmaceutical ingredient yet having distinct advantageous and/or disadvantageous physico-chemical properties compared to other forms in the polymorph family.


Method of Making L-Ornithine Phenyl Acetate Salt

Some embodiments disclosed herein include a method of making L-ornithine phenyl acetate salt. L-Ornithine phenyl acetate may be produced, for example, through an intermediate salt, such as L-ornithine benzoate. As shown in Scheme 1, an L-ornithine salt of Formula I can be reacted with a benzoate salt of Formula II to obtain the intermediate L-ornithine benzoate.




embedded image


Various salts of L-ornithine may be used in the compound of Formula I, and therefore X in Formula I can be any ion capable of forming a salt with L-ornithine other than benzoic acid or phenyl acetic acid. X can be a monoatomic anion, such as, but not limited to, a halide (e.g., fluoride, chloride, bromide, and iodide). X can also be a polyatomic anion, such as, but not limited to, acetate, aspartate, formate, oxalate, bicarbonate, carbonate, bitrate, sulfate, nitrate, isonicotinate, salicylate, citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, glutamate, methanesulfonate, ethanesulfonate, benzensulfonate, p-toluenesulfonate, pamoate (i.e., 1,1′-methylene-bis-(2-hydroxy-3-naphthoate), phosphate and the like. In some embodiments, X is a monovalent ion. In some embodiments, X is chloride.


Similarly, the benzoate salt of Formula II is not particularly limited, and therefore Y in Formula II can be any appropriate ion capable of forming a salt with benzoic acid. In some embodiments, Y can be a monoatomic cation, such as an alkali metal ion (e.g., Li+, Na+, and K+) and other monovalent ions (e.g., Ag+). Y may also be a polyatomic cation, such as ammonium, L-arginine, diethylamine, choline, ethanolamine, 1H-imidazole, trolamine, and the like. In some embodiments, Y is an inorganic ion. In some embodiments, Y is silver.


Many other possible salts of L-ornithine and benzoic acid may be used for the compounds of Formulae I and II, respectively, and can readily be prepared by those skilled in the art. See, for example, Bighley L. D., et al., “Salt forms of drugs and absorption,” In: Swarbrick J., Horlan J. C., eds. Encyclopedia of pharmaceutical technology, Vol. 12. New York: Marcel Dekker, Inc. pp. 452-499, which is hereby incorporated by reference in its entirety.


The intermediate L-ornithine benzoate (i.e., Formula III) can be prepared by intermixing solutions including compounds of Formulae I and II. As an example, the compounds of Formulae I and II may be separately dissolved in water and dimethyl sulfoxide (DMSO), respectively. The two solutions may then be intermixed so that the L-ornithine and benzoic acid react to form the salt of Formula III. Alternatively, the two salt compounds can be directly dissolved into a single solution. In some embodiments, L-ornithine and benzoic acid are dissolved in separate solvents, and subsequently intermixed. In some embodiments, L-ornithine is dissolved in an aqueous solution, benzoic acid is dissolved in an organic solvent, and the L-ornithine and benzoic acid solutions are subsequently intermixed.


Non-limiting examples of solvents which may be used when intermixing L-ornithine and benzoate salts include acetonitrile, dimethylsulfoxide (DMSO), cyclohexane, ethanol, acetone, acetic acid, 1-propanol, dimethylcarbonate, N-methyl-2-pyrrolidone (NMP), ethyl acetate (EtOAc), toluene, isopropyl alcohol (IPA), diisopropoyl ether, nitromethane, water, 1,4 dioxane, tdiethyl ether, ethylene glycol, methyl acetate (MeOAc), methanol, 2-butanol, cumene, ethyl formate, isobutyl acetate, 3-methyl-1-butanol, anisole, and combinations thereof. In some embodiments, the L-ornithine benzoate solution includes water. In some embodiments, the L-ornithine benzoate solution includes DMSO.


Upon intermixing L-ornithine and benzoate salts, counterions X and Y may form a precipitate that can be removed from the intermixed solution using known methods, such as filtration, centrifugation, and the like. In some embodiments, X is chloride, Y is silver, and the reaction produces a precipitate having AgCl. Although Scheme 1 shows the compounds of Formulae I and II as salts, it is also within the scope of the present application to intermix the free base of L-ornithine and benzoic acid to form the intermediate of L-ornithine benzoate. Consequently, forming and isolating the precipitate is optional.


The relative amount of L-ornithine and benzoate salts that are intermixed is not limited; however the molar ratio of L-ornithine to benzoic acid may optionally be in the range of about 10:90 and 90:10. In some embodiments, the molar ratio of L-ornithine benzoate can be in the range of about 30:70 and 30:70. In some embodiments, the molar ratio of L-ornithine to benzoate can be in the range of about 40:60 and 60:40. In some embodiments, the molar ratio of L-ornithine to benzoate is about 1:1.


In embodiments where X and Y are both inorganic ions (e.g., X and Y are chloride and silver, respectively), additional amounts of X-containing salt may be added to encourage further precipitation of the counterion Y. For example, if X is chloride and Y is silver, the molar ratio of L-ornithine hydrochloride to silver benzoate may be greater than 1:1 so that an excess of chloride is present relative to silver. Accordingly, in some embodiments, the molar ratio of L-ornithine to benzoic acid is greater than about 1:1. Nevertheless, the additional chloride salt is not required to be derived from an L-ornithine salt (e.g., L-ornithine hydrochloride). For example, dilute solutions of hydrochloric acid may be added to the solution to further remove silver. Although it is not particularly limited when the additional X-containing salt is added, it is preferably added before the AgCl is initially isolated.


As shown in Scheme 2, the L-ornithine benzoate can be reacted with a phenyl acetate salt of Formula IV to form L-ornithine phenyl acetate. For example, sodium phenyl acetate can be intermixed with a solution of L-ornithine benzoate to form L-ornithine phenyl acetate. Various salts of phenyl acetate may be used, and therefore Z in Formula IV can be any cation capable of forming a salt with phenyl acetate other than benzoic acid or L-ornithine. In some embodiments, Z can be a monoatomic cation, such as an alkali metal ion (e.g., Li+, Na+, and K+) and other monovalent ions (e.g., Ag+). Z may also be a polyatomic cation, such as ammonium, L-arginine, diethylamine, choline, ethanolamine, 1H-imidazole, trolamine, and the like. In some embodiments, Z is an inorganic ion. In some embodiments, Z is sodium.


The relative amount of L-ornithine and phenyl acetate salts that are intermixed is also not limited; however the molar ratio of L-ornithine to phenyl acetate may optionally be in the range of about 10:90 and 90:10. In some embodiments, the molar ratio of L-ornithine to phenyl acetate can be in the range of about 30:70 and 30:70. In some embodiments, the molar ratio of L-ornithine to phenyl acetate can be in the range of about 40:60 and 60:40. In some embodiments, the molar ratio of L-ornithine to benzoic acid is about 1:1.




embedded image


The L-ornithine phenyl acetate of Formula V may then be isolated from solution using known techniques. For example, by evaporating any solvent until the L-ornithine phenyl acetate crystallizes, or alternatively by the adding an anti-solvent miscible in the L-ornithine phenyl acetate solution until the L-ornithine phenyl acetate precipitates from solution. Another possible means for isolating the L-ornithine phenyl acetate is to adjust the temperature of the solution (e.g., lower the temperature) until the L-ornithine phenyl acetate precipitates. As will be discussed in further detail in a later section, the method of isolating the L-ornithine phenyl acetate affects the crystalline form that is obtained.


The isolated L-ornithine phenyl acetate may be subjected to various additional processing, such as drying and the like. In some embodiments, L-ornithine phenyl acetate may be subsequently intermixed with a dilute HCl solution to precipitate residual silver. The L-ornithine phenyl acetate may again be isolated from solution using similar methods disclosed above.


As would be appreciated by a person of ordinary, guided by the teachings of the present application, L-ornithine phenyl acetate may similarly be prepared using an intermediate salt other than L-ornithine benzoate. Thus, for example, L-ornithine, or a salt thereof (e.g., L-ornithine hydrochloride), can be intermixed with a solution having acetic acid. L-Ornithine acetate may then be intermixed with phenyl acetic acid, or a salt thereof (e.g., sodium phenyl acetate), to obtain L-ornithine phenyl acetate. Scheme 4 illustrates an exemplary process of forming L-ornithine phenyl acetate using L-ornithine acetate as an intermediate salt. In some embodiments, the intermediate salt can be a pharmaceutically acceptable salt of L-ornithine. For example, the intermediate L-ornithine salt can be an acetate, aspartate, formate, oxalate, bicarbonate, carbonate, bitrate, sulfate, nitrate, isonicotinate, salicylate, citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzensulfonate, p-toluenesulfonate, pamoate (i.e., 1,1′-methylene-bis-(2-hydroxy-3-naphthoate) or phosphate. The free acid of the intermediate is preferably a weaker acid relative to phenyl acetic acid. In some embodiments, the intermediate is an L-ornithine salt with an anion component that exhibits a pKa value that is higher than the pKa value of phenyl acetic acid. As an example, for L-ornithine acetate, acetic acid and phenyl acetic acid exhibit pKa values of about 4.76 and 4.28, respectively.




embedded image


L-Ornithine phenyl acetate may also be prepared, in some embodiments, without forming an intermediate salt, such as L-ornithine benzoate. Scheme 4 illustrates an exemplary process for preparing L-ornithine phenyl acetate without an intermediate salt. A pH modifier may be added to a solution of L-ornithine salt (e.g., as illustrated in Scheme 4 by the compound of Formula I) until a salt precipitates from solution, where the salt is not an L-ornithine salt. As an example, sodium methoxide (NaOMe) can be added to a solution of L-ornithine hydrochloride until sodium chloride precipitates from solution to leave a free base of L-ornithine. The precipitate may optionally be isolated from solution using known techniques, such as filtration, centrifugation, and the like. The free base of L-ornithine (e.g., as illustrated in Scheme 4 by the compound of Formula I-a) may be intermixed with phenyl acetic acid, or a salt thereof (e.g., as illustrated in Scheme 4 by the compound of Formula IV), to obtain L-ornithine phenyl acetate. The L-ornithine phenyl acetate of Formula V may then be isolated as previously described.




embedded image


A pH modifier can include a basic compound, or anhydrous precursor thereof, and/or a chemically protected base. Non-limiting examples of pH modifiers include sodium hydroxide, potassium hydroxide, sodium methoxide, potassium t-butoxide, sodium carbonate, calcium carbonate, dibutylamine, tryptamine, sodium hydride, calcium hydride, butyllithium, ethylmagnesium bromide and combinations thereof. Also, the amount of pH modifier to be added is not particularly limited; however the molar ratio of L-ornithine to pH modifier may optionally be in the range of about 10:90 and 90:10. In some embodiments, the molar ratio of L-ornithine to pH modifier can be in the range of about 30:70 and 30:70. In some embodiments, the molar ratio of L-ornithine to pH modifier can be in the range of about 40:60 and 60:40. In some embodiments, the molar ratio of L-ornithine to pH modifier is about 1:1. The pH modifier may, in some embodiments be added to adjust the pH value to at least about 8.0; at least about 9.0; or at least about 9.5.


Another process for forming L-ornithine phenyl acetate, in some embodiments, includes reacting an alkali metal salt of L-ornithine with a phenyl acetate salt. As an example, L-ornithine hydrochloride may be intermixed with silver phenyl acetate and a solvent. AgCl may then precipitate and is optionally isolated from the solution. The remaining L-ornithine phenyl acetate can also be isolated using known methods. This process can be completed using generally the same procedures and conditions outlined above. For example, the relative molar amounts of L-ornithine to phenyl acetate can be 10:90 to 90:10; 30:70 to 70:30; 40:60 to 60:40; or about 1:1. Also, the L-ornithine phenyl acetate may be isolated by evaporating the solvent, adding an anti-solvent, and/or reducing the temperature.


Compositions of L-Ornithine Phenyl Acetate

Also disclosed herein are compositions of L-ornithine phenyl acetate. The compositions of the present application advantageously have low amounts of inorganic salts, particularly alkali metal salts and/or halide salts, and therefore are particularly suited for oral and/or intravenous administration to patients with hepatic encephalopathy. Meanwhile, these compositions may exhibit similar stability profiles compared to other salts (e.g., mixtures of L-ornithine hydrochloride and sodium phenyl acetate). The compositions may, in some embodiments, be obtained by one of the processes disclosed in the present application. For example, any of the disclosed processes using L-ornithine benzoate as an intermediate may yield the compositions of the present application.


The compositions, in some embodiments, can include a crystalline form of L-ornithine phenyl acetate (e.g., Forms I, II, III and/or V disclosed herein). In some embodiments, the composition may include at least about 20% by weight of a crystalline form of L-ornithine phenyl acetate (preferably at least about 50% by weight, and more preferably at least about 80% by weight). In some embodiments, the composition consists essentially of a crystalline form of L-ornithine phenyl acetate. In some embodiments, the composition includes a mixture of at least two (e.g., two, three or four forms) of Forms I, II, III, and V.


The compositions, in some embodiments, include Form II. For example, the compositions may include at least about 20%; at least about 50%; at least about 90%; at least about 95%; or at least about 99% of Form II. Similarly, the compositions may also include, for example, Forms I, III or V. The compositions may optionally include at least about 20%; at least about 50%; at least about 90%; at least about 95%; or at least about 99% of Forms I, II, III and/or V.


Also within the scope of the present application are amorphous forms of L-ornithine phenyl acetate. Various methods are known in the art for preparing amorphous forms. For example, a solution of L-ornithine phenyl acetate may be dried under vacuum by lyophilization to obtain an amorphous composition. See P.C.T. Application WO 2007/058634, which published in English and designates the U.S., and is hereby incorporated by reference for disclosing methods of lyophilization.


It is preferred that the composition have low amounts (if any) of alkali and halogen ions or salts, particular sodium and chloride. In some embodiments, the composition comprises no more than about 100 ppm of alkali metals (preferably no more than about 20 ppm, and most preferably no more than about 10 ppm). In some embodiments, the composition comprises no more than about 100 ppm of sodium (preferably no more than about 20 ppm, and most preferably no more than about 10 ppm). In some embodiments, the composition comprises no more than about 0.1% by weight of halides (preferably no more than about 0.01% by weight). In some embodiments, the composition comprises no more than about 0.1% by weight of chloride (preferably no more than about 0.01% by weight).


The reduced content of alkali metals and halides provides a composition suitable for preparing concentrated isotonic solutions. As such, these compositions can be more easily administered intravenously compared to, for example, administering mixtures of L-ornithine hydrochloride and sodium phenyl acetate. In some embodiments, an about 45 to about 55 mg/mL solution of L-ornithine phenyl acetate in water (preferably about 50 mg/mL) is isotonic with body fluids (e.g., the solution exhibits an osmolality in the range of about 280 to about 330 mOsm/kg).


The compositions may also include residual amounts of the anion from an intermediate salt formed during the process of making the L-ornithine phenyl acetate composition. For example, some of the processes disclosed herein yield compositions having benzoic acid or a salt thereof. In some embodiments, the composition comprises at least about 0.01% by weight benzoic acid or a salt thereof (preferably at least about 0.05% by weight, and more preferably about 0.1% by weight). In some embodiments, the composition comprises no more than about 3% by weight benzoic acid or a salt thereof (preferably no more than about 1% by weight, and more preferably no more than about 0.5% by weight). In some embodiments, the composition includes a salt, or an acid thereof, in the range of about 0.01% to about 3% by weight (preferably about 0.1% to about 1%), wherein the salt is selected from acetate, aspartate, formate, oxalate, bicarbonate, carbonate, bitrate, sulfate, nitrate, isonicotinate, salicylate, citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzensulfonate, p-toluenesulfonate, pamoate (i.e., 1,1′-methylene-bis-(2-hydroxy-3-naphthoate) or phosphate.


Similarly, a composition prepared using an acetate intermediate may have residual amounts of acetic acid or acetate. In some embodiments, the composition includes at least about 0.01% by weight acetic acid or acetate (preferably at least about 0.05% by weight, and more preferably about 0.1% by weight). In some embodiments, the composition includes no more than about 3% by weight acetic acid or acetate (preferably no more than about 1% by weight, and more preferably no more than about 0.5% by weight).


The compositions may also include low amounts of silver. Exemplary processes disclosed herein utilize, for example, silver benzoate, but still yield compositions with surprisingly low amounts of silver. Thus, in some embodiments, the composition includes no more than about 600 ppm silver (preferably no more than about 100 ppm, and more preferably no more than about 65 ppm). In some embodiments, the composition includes at least about 10 ppm silver (alternatively at least about 20 or 25 ppm silver).


Pharmaceutical Compositions

The compositions of L-ornithine phenyl acetate of the present application may also be formulated for administration to a subject (e.g., a human). L-Ornithine phenyl acetate, and accordingly the compositions disclosed herein, may be formulated for administration with a pharmaceutically acceptable carrier or diluent. L-ornithine phenyl acetate may thus be formulated as a medicament with a standard pharmaceutically acceptable carrier(s) and/or excipient(s) as is routine in the pharmaceutical art. The exact nature of the formulation will depend upon several factors including the desired route of administration. Typically, L-ornithine phenyl acetate is formulated for oral, intravenous, intragastric, subcutaneous, intravascular or intraperitoneal administration.


The pharmaceutical carrier or diluent may be, for example, water or an isotonic solution, such as 5% dextrose in water or normal saline. Solid oral forms may contain, together with the active compound, diluents, e.g. lactose, dextrose, saccharose, cellulose, corn starch or potato starch; lubricants, e.g. silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols; binding agents, e.g. starches, gum arabic, gelatin, methylcellulose, carboxymethylcellulose or polyvinyl pyrrolidone; disaggregating agents, e.g. starch, alginic acid, alginates or sodium starch glycolate; effervescing mixtures; dyestuffs; sweeteners; wetting agents, such as lecithin, polysorbates, laurylsulphates; and, in general, non-toxic and pharmacologically inactive substances used in pharmaceutical formulations. Such pharmaceutical preparations may be manufactured in known manners, for example, by means of mixing, granulating, tabletting, sugar-coating, or film-coating processes.


Liquid dispersions for oral administration may be syrups, emulsions or suspensions. The syrups may contain as carriers, for example, saccharose or saccharose with glycerine and/or mannitol and/or sorbitol.


Suspensions and emulsions may contain a carrier, for example a natural gum, agar, sodium alginate, pectin, methylcellulose, carboxymethylcellulose, or polyvinyl alcohol. The suspensions or solutions for intramuscular injections may contain, together with L-ornithine phenyl acetate, a pharmaceutically acceptable carrier, e.g. sterile water, olive oil, ethyl oleate, glycols, e.g. propylene glycol, and if desired, a suitable amount of lidocaine hydrochloride.


The medicament may consist essentially of L-ornithine phenyl acetate and a pharmaceutically acceptable carrier. Such a medicament therefore contains substantially no other amino acids in addition to L-ornithine and phenyl acetate. Furthermore, such a medicament contains insubstantial amounts of other salts in addition to L-ornithine phenyl acetate.


Oral formulations may generally include dosages of L-ornithine phenyl acetate in the range of about 500 mg to about 100 g. Accordingly, in some embodiments, the oral formulation includes the L-ornithine phenyl acetate compositions disclosed herein in the range of about 500 mg to about 50 g. In some embodiments, the oral formulation is substantially free of alkali metal salts and halides (e.g., contains no more than trace amounts of alkali metal salts and halides).


Intravenous formulations may also generally include dosages of L-ornithine phenyl acetate in the range of about 500 mg to about 100 g (preferably about 1 g to about 50 g). In some embodiments, the intravenous formulation is substantially free of alkali metal salts and halides (e.g., contains no more than trace amounts of alkali metal salts and halides). In some embodiments, the intravenous formulation has a concentration of about 5 to about 300 mg/mL of L-ornithine phenyl acetate (preferably about 25 to about 200 mg/mL, and more preferably about 40 to about 60 mg/mL).


The composition, or medicament containing said composition, may optionally be placed is sealed packaging. The sealed packaging may reduce or prevent moisture and/or ambient air from contacting the composition or medicament. In some embodiments, the packaging includes a hermetic seal. In some embodiments, the packaging sealed under vacuum or with an inert gas (e.g., argon) within the sealed package. Accordingly, the packaging can inhibit or reduce the rate of degradation for the composition or medicament stored within the packaging. Various types of sealed packaging are known in the art. For example, U.S. Pat. No. 5,560,490, is hereby incorporate by reference in its entirety, discloses an exemplary sealed package for medicaments.


Compositions with Improved Density

Applicants have surprisingly found that compositions with greater density may be obtained by applying sufficient pressure to compositions having Form I (described below) to induce a transition to Form II (described below). For example, applying 3 tons of force for 90 minutes to Forms I and II yield densities of 1.197 kg/m3 and 1.001 kg/m3, respectively. Surprisingly, Form I converted to Form II under these conditions; therefore the greater density appears to be explained by the different crystalline form as the starting material.


Accordingly, disclosed herein are methods of increasing the density of an L-ornithine phenyl acetate composition having Form I by applying pressure to the composition sufficient to induce a transition to Form II. The appropriate amount of force or pressure to induce the phase change may vary with the amount of time the force or pressure is applied. Thus, a person of ordinary skill, guided by the teachings of the present application, can determine appropriate amounts of pressure and time to induce the phase change. In some embodiments, at least about 1 ton of force is applied (preferably at least about 2 tons, and more preferably about 3 tons). In some embodiments, at least about 500 psi of pressure is applied (preferably at least about 1000 psi, and more preferably at least about 2000 psi).


The amount of time for applying pressure is not particularly limited, and as discussed above, will vary depending upon the amount time. For example, when applying large forces (e.g., 10 tons) to a typical tablet-sized punch, the time may be about 1 second or less. In some embodiments, the time for apply pressure is a predetermined time. The time may be, for example, about 0.1 seconds; about 1 second; at least about 1 minute; at least about 5 minutes; or at least about 20 minutes.


In some embodiments, the composition includes at least about 10% by weight of Form I. In some embodiments, the composition includes at least about 30% by weight of Form I.


Without being bound to any particular theory, Applicants believe the greater density may result, at least in part, from ethanol solvate component present in Form I. Applying pressure to the solvate may facilitate forming a denser structure with fewer defects (e.g., grain boundaries). Consequently, in some embodiments, methods of increasing the density of an L-ornithine phenyl acetate composition having solvate components include applying pressure to the composition sufficient to induce a transition to Form II. In some embodiments, the pressure is at least about 500 psi (preferably at least about 1000 psi, and more preferably at least about 2000 psi). In some embodiments, the time for apply pressure is a predetermined time. In some embodiments, the composition includes at least about 10% of the solvate form (preferably at least about 30%, and more preferably at least about 50%).


The compositions of L-ornithine phenyl acetate disclosed herein may therefore have higher densities compared to compositions obtain by, for example, precipitating a crystalline form. In some embodiments, the composition has a density of at least about 1.1 kg/m3 (preferably at least about 1.15 kg/m3, and more preferably at least about 1.18 kg/m3). In some embodiments, the composition has a density of no more than about 1.3 kg/m3 (preferably no more than about 1.25 kg/m3, and more preferably no more than about 1.22 kg/m3). In some embodiments, the composition has a density of about 1.2 kg/m3.


Crystalline Forms of L-Ornithine Phenyl Acetate

Also disclosed herein are crystalline forms of L-ornithine phenyl acetate, and in particular, crystalline Form I, Form II, Form III, and Form V. L-Ornithine phenyl acetate may, in some embodiments, be obtained using the processes disclosed above and then crystallized using any of the methods disclosed herein.


Form I


The precise conditions for forming crystalline Form I may be empirically determined and it is only possible to give a number of methods which have been found to be suitable in practice.


Thus, for example, crystalline Form I may generally be obtained by crystallizing L-ornithine phenyl acetate under controlled conditions. As an example, precipitating L-ornithine phenyl acetate from a saturated solution by adding ethanol at reduced temperatures (e.g., 4° or −21° C.). Exemplary solvents for the solution that yield crystalline Form I upon adding ethanol include, but are not limited to, cyclohexanone, 1-propanol, dimethylcarbonate, N-methylpyrrolidine (NMP), diethyl ether, 2-butanol, cumene, ethyl formate, isobutyl acetate, 3-methyl-1-butanol, and anisole.


Accordingly, in the context of the processes for making L-ornithine phenyl acetate disclosed above, the process can yield Form I by utilizing particular isolation methods. For example, L-ornithine phenyl acetate may be isolated by adding ethanol at reduced temperature to yield Form I.


Crystalline Form I was characterized using various techniques which are described in further detail in the experimental methods section. FIG. 1 shows the crystalline structure of Form I as determined by X-ray powder diffraction (XRPD). Form I, which may be obtained by the methods disclosed above, exhibits characteristic peaks at approximately 4.9°, 13.2°, 17.4°, 20.8° and 24.4° 2θ. Thus, in some embodiments, a crystalline form of L-ornithine phenyl acetate has one or more characteristic peaks (e.g., one, two, three, four or five characteristic peaks) selected from approximately 4.9°, 13.2°, 17.4°, 20.8°, and 24.4° 2θ.


As is well understood in the art, because of the experimental variability when X-ray diffraction patterns are measured on different instruments, the peak positions are assumed to be equal if the two theta (2θ) values agree to within 0.2° (i.e., ±0.2°). For example, the United States Pharmacopeia states that if the angular setting of the 10 strongest diffraction peaks agree to within ±0.2° with that of a reference material, and the relative intensities of the peaks do not vary by more than 20%, the identity is confirmed. Accordingly, peak positions within 0.2° of the positions recited herein are assumed to be identical.



FIG. 2 shows results obtained by differential scanning calorimetry (DSC) for Form I. These results indicate an endotherm at 35° C., which is possibly associated with a desolvation and/or dehydration to Form II. A second transition at about 203° C. indicates the melting point for the crystal. To explore the possible existence of a desolvation and/or dehydration transition, Form I was analyzed by thermogravimetric gravimetric/differential thermal analysis (TG/DTA), which is shown in FIG. 3. Form I exhibits a 11.28% weight loss at about 35° C., and therefore these results further suggest that Form I exhibits a desolvation and/or dehydration transition at about 35° C. The melting point of about 203° C. could also be observed by TGA testing. Accordingly, in some embodiments, the crystalline form of L-ornithine phenyl acetate is characterized by differential scanning calorimetry as having an endotherm at about at about 35° C. In some embodiments, a crystalline form of L-ornithine phenyl acetate exhibits a weight loss of about 11% at about 35° C., as determined by TGA. In some embodiments, a crystalline form of L-ornithine phenyl acetate exhibits a melting point of about 203° C.



FIG. 4 shows nuclear magnetic resonance (NMR) integrals and chemical shifts for Form I. The integrals confirm the presence of L-ornithine phenyl acetate: 7.5 (aromatic CH), 3.8 (CH adjacent to NH2), 3.6 (CH2 unit of phenyl acetate), 3.15 (CH2 adjacent to NH2) and 1.9 (aliphatic CH2 units) ppm (integrals: 5:1:2:2:4 protons; 1.2, 0.25, 0.5, 0.5, 1.0). Amine protons and hydroxyl protons were not observed due to proton exchange at both the zwitterion and site of salt formation. Meanwhile, FIG. 5 shows dynamic vapor sorption (DVS) results for Form I, and show a water uptake of about 0.2% by weight. XRPD results following DVA analysis (not shown) confirm that Form I did not transition to a different polymorph. Form I can therefore be characterized as non-hygroscopic and stable over a wide range of humidity.


A 7-day stability study of Form I at 40° C./75% RH indicated that a transformation to Form II occurred under these conditions. Form I also converts to Form II at elevated temperatures (e.g., 80° or 120° C.), with or without applying a vacuum, after 7 or 14 days. Accordingly, Form I is metastable.


Single crystal x-ray diffraction (SXRD) was also used to determine the structure of Form I at −20° and −123° C., and the results are summarized in TABLES 1 and 2. The results confirm that Form I is a solvate having ethanol and water molecules within the unit cell. In some embodiments, a crystalline form of L-ornithine phenyl acetate can be represented by the formula C15H28N2O6. In some embodiments, a crystalline form of L-ornithine phenyl acetate can be represented by the formula [C5H13N2O2][C8H7O2]EtOH.H2O. In some embodiments, a crystalline form of L-ornithine phenyl acetate exhibits a single crystal X-ray crystallographic analysis with crystal parameters approximately equal to the following: unit cell dimensions of a=5.3652(4) Å, b=7.7136(6) Å, c=20.9602(18) Å, α=90°, β=94.986(6°), γ=90°; a monoclinic crystal system, and a P21 space group.









TABLE 1





Crystallographic Data of Form I Collected at −20° C.


















Empirical Formula
C15 H28 N2 O6 or




[C5H13N2O2][C8H7O2]EtOH•H2O



Formula Weight
332.39



Crystal System
Monoclinic



Space Group
P21



Unit Cell Dimensions
a = 5.3652(4) Å α = 90°




b = 7.7136(6) Å β = 94.986(6)°




c = 20.9602(18) Å γ = 90°



Volume
864.16(12) Å3



Number of Reflections
1516 (2.5° < θ < 28°)



Density (calculated)
1.277 mg/cm3

















TABLE 2





Crystallographic Data of Form I Collected at −123° C.


















Empirical Formula
C15 H28 N2 O6 or




[C5H13N2O2][C8H7O2]EtOH•H2O



Formula Weight
332.39



Crystal System
Monoclinic



Space Group
P21



Unit Cell Dimensions
a = 5.3840(9) Å α = 90°




b = 7.7460(12) Å β = 95.050(12)°




c = 21.104(4) Å γ = 90°



Volume
876.7(3) Å3



Number of Reflections
1477 (2.5° < θ < 18°)



Density (calculated)
1.259 mg/cm3











Form II


The precise conditions for forming crystalline Form II may be empirically determined and it is only possible to give a number of methods which have been found to be suitable in practice.


Thus, for example, crystalline Form II may be prepared by crystallization under controlled conditions. Crystalline Form II can be prepared by, for example, evaporating a saturated organic solution of L-ornithine phenyl acetate. Non-limiting examples of organic solutions that may be used to obtain Form II include ethanol, acetone, benzonitrile, dichloromethane (DCM), dimethyl sulfoxide (DMSO), ethyl acetate (EtOAc), acetonitrile (MeCN), methyl acetate (MeOAc), nitromethane, tert-butyl methyl ether (TBME), tetrahydrofuran, and toluene. Other solvents may yield a mixture of Form I and II, such as, but not limited to, 1,4 dioxane, 1-butanol, cyclohexane, IPA, THF, MEK, MeOAc and water.


Form II can also be obtained by precipitating L-ornithine phenyl acetate from a saturated organic solution by adding an anti-solvent for L-ornithine phenyl acetate, such as IPA. Form II may be precipitated over a broad range of temperatures (e.g., room temperature, 4° C., and −21° C.). Non-limiting examples of suitable solvents for the saturated organic solution include cyclohexanone, 1-propanol, dimethyl carbonate, N-methylpyrrolidone (NMP), diisopropyl ether, diethyl ether, ethylene glycol, dimethylformamide (DMF), 2-butanol, cumene, isobutyl acetate, 3-methyl-1-butanol, and anisole. Alternatively, the same listed solvents (e.g., cyclohexanone) can be used to form a solution of L-ornithine phenyl acetate, and Form II may be precipitated by adding ethanol at ambient conditions. As another example, Form II may also be obtained by forming a slurry of L-ornithine phenyl acetate with the listed organic solvents and cycling the temperature between 25° and 40° C. every 4 hours for about 18 cycles (or 72 hours).


Accordingly, in the context of the processes for making L-ornithine phenyl acetate disclosed above, the process can yield Form II by utilizing particular isolation methods. For example, L-ornithine phenyl acetate may by isolated by adding IPA, or evaporating the organic solvent, to yield Form II.



FIG. 6 shows the crystalline structure of Form II as determined by XRPD. Form II, which may be obtained by the methods disclosed above, exhibits characteristic peaks at approximately 6.0°, 13.9°, 14.8°, 17.1°, 17.8° and 24.1° 2θ. Thus, in some embodiments, a crystalline form of L-ornithine phenyl acetate has one or more characteristic peaks (e.g., one, two, three, four, five or six characteristic peaks) selected from approximately 6.0°, 13.9°, 14.8°, 17.1°, 17.8° and 24.12° θ.



FIG. 7 shows results obtained by differential scanning calorimetry (DSC) for Form II. These results indicate a melting point of about 202° C., which is approximately the same as the melting point for Form I. This suggests that Form I transitions to Form II upon heating above about 35° C. Form II was also analyzed using TG/DTA, as shown in FIG. 8, and exhibits an about 9.7% weight loss associated with residual solvent. The melting point of about 202° C. could also be observed by TGA testing. Accordingly, in some embodiments, a crystalline form of L-ornithine phenyl acetate exhibits a melting point of about 202° C.


A 7-day stability study of Form II at 40° C./75% RH failed to produce an observable phase change. In fact, Form II was stable over 14 days when exposed to elevated temperatures, varying pHs, UV light or oxygen. Accordingly, Form II is considered stable.



FIG. 9 shows nuclear magnetic resonance (NMR) integrals and chemical shifts for Form II. The integrals confirm the presence of L-ornithine phenyl acetate: 7.5 (aromatic CH), 3.8 (CH adjacent to NH2), 3.6 (CH2 unit of phenylacetate), 3.15 (CH2 adjacent to NH2) and 1.9 (aliphatic CH2 units) ppm (integrals: 5:1:2:2:4 protons; 7.0, 1.4, 2.9, 3.0, 5.9). Amine protons and hydroxyl protons were not observed due to proton exchange at both the zwitterion and site of salt formation. Meanwhile, FIG. 10 shows dynamic vapor sorption (DVS) results for Form II, and show a water uptake of about 0.3% by weight. XRPD results following DVA analysis (not shown) confirm that Form II did not transition to a different polymorph. Form II can therefore be characterized as non-hygroscopic and stable over a wide range of humidity.


Single crystal x-ray diffraction (SXRD) was also used to determine the structure of Form II at 23° and −123° C., and the results are summarized in TABLES 3 and 4. The results demonstrate that Form II is anhydrous and therefore structurally different from Form I. In some embodiments, a crystalline form of L-ornithine phenyl acetate can be represented by the formula C13H20N2O4. In some embodiments, a crystalline form of L-ornithine phenyl acetate can be represented by the formula [C5H13N2O2][C8H7O2]. In some embodiments, a crystalline form of L-ornithine phenyl acetate exhibits a single crystal X-ray crystallographic analysis with crystal parameters approximately equal to the following: unit cell dimensions of a=6.594(2) Å, α=90°, b=6.5448(18) Å, β=91.12(3°), c=31.632(8) Å, γ=90°; a monoclinic crystal system; and a P21 space group.









TABLE 3





Crystallographic Data of Form II Collected at 23° C.


















Empirical Formula
C13H20N2O4 or [C5H13N2O2][C8H7O2]



Formula Weight
268.31



Crystal System
Monoclinic



Space Group
P21



Unit Cell Dimensions
a = 6.594(2) Å α = 90°




b = 6.5448(18) Å β = 91.12(3)°




c = 31.632(8) Å γ = 90°



Volume
1364.9(7) Å3



Number of Reflections
3890 (3° < θ < 20.5°)



Density (calculated)
1.306 mg/cm3

















TABLE 4





Crystallographic Data of Form II Collected at −123° C.
















Empirical Formula
C15 H28 N2 O6 or [C5H13N2O2][C8H7O2]


Formula Weight
332.39


Crystal System
Monoclinic


Space Group
P21


Unit Cell Dimensions
a = 5.3652(4) Å α = 90°



b = 7.7136(6) Å β = 94.986(6)°



c = 20.9602(18) Å γ = 90°


Volume
864.16(12) Å3


Number of Reflections
1516 (2.5° < θ < 28°)


Density (calculated)
1.277 mg/cm3










Form III


The precise conditions for forming crystalline Form III may be empirically determined and it is only possible to give a number of methods which have been found to be suitable in practice.


Thus, for example, Form III may be obtained by placing a saturated solution of L-ornithine phenyl acetate in a cooled temperature environment of about −21° C., where the solution is a mixture of acetone and water (e.g., equal parts volume of acetone and water). As another example, adding IPA to a saturated solution of L-ornithine phenyl acetate in 2-butanol can yield Form III when completed at ambient conditions. Furthermore, Form III may be obtained, for example, by adding IPA to a saturated solution of L-ornithine phenyl acetate in isobutyl acetate when completed at reduced temperatures of about −21° C.


Accordingly, in the context of the processes for making L-ornithine phenyl acetate disclosed above, the process can yield Form III by utilizing particular solvents and isolation methods. For example, L-ornithine phenyl acetate may be formed within a mixture of acetone and water, and subsequently placed in a cool environment of about −21° C. to yield Form III.



FIG. 11 shows the crystalline structure of Form III as determined by XRPD. Form III, which may be obtained by the methods disclosed above, exhibits characteristic peaks at approximately 5.8°, 14.1°, 18.6°, 19.4°, 22.3° and 24.8° 2θ. Thus, in some embodiments, a crystalline form of L-ornithine phenyl acetate has one or more characteristic peaks (e.g., one, two, three, four, five or six characteristic peaks) selected from approximately 5.8°, 14.1°, 18.6°, 19.4°, 22.3° and 24.8° 2θ.



FIG. 12 shows results obtained by differential scanning calorimetry (DSC) for Form III. These results indicate a melting point of about 203° C., which is approximately the same as the melting points for Form I and Form II. Additionally, Form III exhibits an endotherm at about 40° C. Form III was also analyzed using TG/DTA, as shown in FIG. 13, and exhibits no significant weight loss before the melting point. Form III may therefore be characterized as anhydrous. The melting point of about 203° C. could also be observed by TGA testing. Accordingly, in some embodiments, a crystalline form of L-ornithine phenyl acetate exhibits a melting point of about 203° C. In some embodiments, a crystalline form of L-ornithine phenyl acetate is characterized by differential scanning calorimetry as having an endotherm at about 40° C. In some embodiments, a crystalline form of L-ornithine phenyl acetate is anhydrous.


A 7-day stability study of Form III at 40° C./75% RH indicated that a transformation to Form II occurred under these conditions. In contrast, Form II is stable at elevated temperatures, with or without vacuum, for periods of 7 or 10 days. Accordingly, Form III is most likely metastable, but more stable than Form I.



FIG. 14 shows nuclear magnetic resonance (NMR) integrals and chemical shifts for Form III. The integrals confirm the presence of L-ornithine phenyl acetate: 7.5 (aromatic CH), 3.8 (CH adjacent to NH2), 3.6 (CH2 unit of phenyl acetate), 3.15 (CH2 adjacent to NH2) and 1.9 (aliphatic CH2 units) ppm (integrals: 5:1:2:2:4 protons; 4.2, 0.8, 1.7, 1.7, 3.0). Amine protons and hydroxyl protons were not observed due to proton exchange at both the zwitterion and site of salt formation. Meanwhile, FIG. 15 shows dynamic vapor sorption (DVS) results for Form III, and show a water uptake of about 2.0% by weight. XRPD results following DVS analysis (not shown) confirm that Form III did not transition to a different polymorph. Form III therefore exhibits greater water uptake compared to Forms I and II; however Form III is still characterized as non-hygroscopic and stable over a wide range of humidity at room temperature.


Form V


The precise conditions for forming crystalline Form V may be empirically determined and it is only possible to give a number of methods which have been found to be suitable in practice.


Thus, for example, Form V may be obtained by placing a saturated solution of L-ornithine phenyl acetate in a cooled temperature environment of about −21° C., where the solution is cyclohexanone. As another example, the same saturated solution may yield Form V when evaporating the solvent.


Form V also forms from saturated solutions of L-ornithine phenyl acetate having diisopropyl ether as a solvent. For example, a saturated solution having a solvent ratio of about 1 to 2 of diisopropyl ether and IPA will yield Form V when placed in a cooled temperature environment of about 4° C. Similarly, a solution having only the solvent diisopropyl ether can yield Form V when placed in a cooled temperature environment of about −21° C.



FIG. 16 shows the crystalline structure of Form V as determined by XRPD.


Form V, which may be obtained by the methods disclosed above, exhibits characteristic peaks at approximately 13.7°, 17.4°, 19.8°, 20.6° and 23.7° 2θ. Thus, in some embodiments, a crystalline form of L-ornithine phenyl acetate has one or more characteristic peaks (e.g., one, two, three, four, or five characteristic peaks) selected from approximately 13.7°, 17.4°, 19.8°, 20.6° and 23.7° 2θ.



FIG. 17 shows results obtained by differential scanning calorimetry (DSC) for Form V. These results indicate a melting point of about 196° C., which is below the melting point of other forms. Form V also exhibits an endotherm at about 174° C. Form V was also analyzed using thermal gravimetric analysis (TGA), as shown in FIG. 18, and exhibits no significant weight loss before the melting point. Form V may therefore be characterized as anhydrous. The melting point of about 196° C. could also be observed by TGA testing. Accordingly, in some embodiments, a crystalline form of L-ornithine phenyl acetate exhibits a melting point of about 196° C. In some embodiments, a crystalline form of L-ornithine phenyl acetate is characterized by differential scanning calorimetry as having an endotherm at about 174° C. In some embodiments, a crystalline form of L-ornithine phenyl acetate is anhydrous.



FIG. 19 shows nuclear magnetic resonance (NMR) integrals and chemical shifts for Form V. The integrals confirm the presence of L-ornithine phenyl acetate: 7.5 (aromatic CH), 3.8 (CH adjacent to NH2), 3.6 (CH2 unit of phenyl acetate), 3.15 (CH2 adjacent to NH2) and 1.9 (aliphatic CH2 units) ppm (integrals: 5:1:2:2:4 protons; 4.2, 0.8, 1.7, 1.7, 3.0). Amine protons and hydroxyl protons were not observed due to proton exchange at both the zwitterion and site of salt formation. Meanwhile, FIG. 19 shows dynamic vapor sorption (DVS) results for Form V, and show a water uptake of about 0.75% by weight. XRPD results following DVS analysis (not shown) suggest that Form V transitioned to Form II, but the chemical composition was unchanged. Form V is therefore characterized as non-hygroscopic, but not stable over a wide range of humidity.


A 7-day stability study of Form V at 40° C./75% RH indicated that a transformation to Form II occurred under these conditions; however the chemical composition was unchanged. Accordingly, Form V is most likely metastable.


Methods of Treating Liver Decompensation or Hepatic Encephalopathy


L-Ornithine phenyl acetate, and accordingly any of the compositions of L-ornithine phenyl acetate disclosed herein, may be administered to a subject for treating or ameliorating the onset of liver decompensation or hepatic encephalopathy. L-Ornithine phenyl acetate can thus be administered to improve the condition of a subject, for example a subject suffering from chronic liver disease following a precipitating event. As another example, L-ornithine phenyl acetate may be administered to combat or delay the onset of liver decompensation or hepatic encephalopathy.


L-Ornithine phenyl acetate may be administered in combination to a subject for treatment of hepatic encephalopathy. L-Ornithine phenyl acetate may be administered to improve the condition of a patient suffering from hepatic encephalopathy. L-Ornithine phenyl acetate may be administered to alleviate the symptoms associated with hepatic encephalopathy. L-Ornithine phenyl acetate may be administered to combat hepatic encephalopathy. L-Ornithine phenyl acetate may be administered to prevent or reduce the likelihood of an initial hepatic encephalopathic episode in a person at risk for hepatic encephalopathic episodes. L-Ornithine phenyl acetate may be administered to lessen the severity of an initial hepatic encephalopathic episode in a person at risk for hepatic encephalopathic episodes. L-Ornithine phenyl acetate may be administered to delay an initial hepatic encephalopathic episode in a person at risk for hepatic encephalopathic episodes.


Development of liver decompensation and hepatic encephalopathy commonly involves “precipitating events” (or “acute attacks”). Such precipitating events include gastrointestinal bleeding, infection (sepsis), portal vein thrombosis and dehydration. The onset of such an acute attack is likely to lead to hospitalization. The patient may suffer one of these acute attacks or a combination of these acute attacks.


A subject who has had or is suspected of having had an acute attack is treated according to the invention with L-ornithine phenyl acetate to prevent or reduce the likelihood of progression of the liver to the decompensated state. Consequently, L-ornithine phenyl acetate can prevent or reduce the likelihood of the medical consequences of liver decompensation such as hepatic encephalopathy. L-Ornithine phenyl acetate may be used to preserve liver function. Use of L-ornithine phenyl acetate may thus extend the life of a patient with liver disease. In one embodiment, the metabolic consequences of a gastrointestinal bleed such as hyperammonemia, hypoisoleucemia and reduced protein synthesis in the post-bleeding period are prevented.


Typically, treatment of subjects may begin as soon as possible after the onset or the suspected onset of a precipitating event (acute attack). Preferably, treatment of the subject begins prior to repeated acute attacks. More preferably, treatment of the subject begins following the first acute attack. Thus, in some embodiments, the subject treated with L-ornithine phenyl acetate is identified as having the onset or the suspected onset of a precipitating event (acute attack).


Treatment is typically given promptly after the start of an acute attack. Treatment may begin after the symptom(s) of an acute attack or suspected acute attack have been detected e.g. by a medic such as a physician, a paramedic or a nurse. Treatment may begin upon hospitalization of the subject. Treatment may thus begin within 6 hours, within 3 hours, within 2 hours or within 1 hour after the symptom(s) of an acute attack or suspected acute attack have been detected. Treatment of the subject may therefore begin from 1 to 48 hours, for example from 1 to 36 hours or from 1 to 24 hours after the symptom(s) of an acute attack or suspected acute attack have been detected.


Treatment may occur for up to 8 weeks, for example up to 6 weeks, up to 4 weeks or up to 2 weeks after the symptom(s) of an acute attack or suspected acute attack have been detected. Treatment may therefore occur for up to 48 hours, for example for up to 36 hours or for up to 24 hours after the symptom(s) of an acute attack or suspected acute attack have been detected. Typically, treatment occurs to the time when recovery from the acute precipitating event is evident.


L-Ornithine phenyl acetate may also be used to treat or ameliorate hyperammonemia. Thus, L-ornithine phenyl acetate may be administered to patients identified as having excess ammonia levels in the blood, or patients exhibiting symptoms of excess ammonia in the blood. L-Ornithine phenyl acetate may also be administered to reduce the risk of hyperammonemia. In some embodiments, L-ornithine phenyl acetate can be administered daily, for an indefinite period of time. For example, daily dosages may be administered for the life of the patient, or until a physician determines the patient no longer exhibits a risk for hyperammonemia. In some embodiments, a therapeutically effective amount of L-ornithine phenyl acetate is administered to reduce the risk of hyperammonemia. In some embodiments, a therapeutically effective amount of L-ornithine phenyl acetate is administered orally for the prophylaxis of hyperammonemia.


A therapeutically effective amount of L-ornithine phenyl acetate is administered to the subject. As will be readily apparent to one skilled in the art, the useful in vivo dosage to be administered and the particular mode of administration will vary depending upon the age, weight, the severity of the affliction, and mammalian species treated, the particular compounds employed, and the specific use for which these compounds are employed. (See e.g., Fingl et al. 1975, in “The Pharmacological Basis of Therapeutics”, which is hereby incorporated herein by reference in its entirety, with particular reference to Ch. 1, p. 1). The determination of effective dosage levels, that is the dosage levels necessary to achieve the desired result, can be accomplished by one skilled in the art using routine pharmacological methods. Typically, human clinical applications of products are commenced at lower dosage levels, with dosage level being increased until the desired effect is achieved. Alternatively, acceptable in vitro studies can be used to establish useful doses and routes of administration of the compositions identified by the present methods using established pharmacological methods.


A typical dose of L-ornithine phenyl acetate may be from about 0.02 to about 1.25 g/kg of bodyweight (preferably from about 0.1 to about 0.6 g/kg of bodyweight). A dosage may therefore be from about 500 mg to about 50 g (preferably about 5 g to about 40 g, and more preferably about 10 g to about 30 g).


A single daily dose may be administered. Alternatively, multiple doses, for example two, three, four or five doses may be administered. Such multiple doses may be administered over a period of one month or two weeks or one week. In some embodiments, a single dose or multiple doses such as two, three, four or five doses may be administered daily.


EXAMPLES AND EXPERIMENTAL METHODS

Additional embodiments are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the claims.


X-Ray Powder Diffraction (XRPD)


XRPD analysis was carried out on a Bruker D8 advance or Seimens D5000, scanning the samples between 4° and 50° 2θ. In embodiments using the Bruker D8 device, approximately 5 mg of a sample was gently compressed on the XRPD zero back ground single 96 well plate sample holder. The sample was then loaded into a Bruker D8-Discover diffractometer in transmission mode and analysed using the following experimental conditions.















Operator
D8-Discover


Raw Data Origin
BRUKER-binary V3 (.RAW)


Scan Axis
Gonio


Start Position [°2θ.]
4.0000


End Position [°2θ.]
49.9800


Step Size [°2θ.]
0.0200


Scan Step Time [s]
39.1393


Scan Type
Continuous


Offset [°2θ.]
0.0000


Divergence Slit Type
Fixed


Divergence Slit Size [°]
2.0000


Specimen Length [mm]
10.00


Receiving Slit Size [mm]
0.1000


Measurement Temperature [° C.]
25.00


Anode Material
Cu


K-Alpha1 [Å]
1.54060


K-Alpha2 [Å]
1.54443


K-Beta [Å]
1.39225


K-A2/K-A1 Ratio
0.50000


Generator Settings
40 mA, 40 kV


Diffractometer Type
Unknown


Diffractometer Number
0


Goniometer Radius [mm]
250.00


Dist. Focus-Diverg. Slit [mm]
91.00


Incident Beam Monochromator
No


Spinning
No









In embodiments using the Seimens D5000 device, approximately 5 mg of sample was gently compressed on glass slide containing a thin layer of holding grease. The sample was then loaded into a Seimens D5000 diffractometer running in reflection mode and analysed, whilst spinning, using the following experimental conditions.


















Raw Data Origin
Siemens-binary V2 (.RAW)



Start Position [°2θ.]
3.0000



End Position [°2θ.]
50.000



Step Size [°2θ.]
0.0200



Scan Step Time [s]
0.8



Scan Type
Continuous



Offset [°2θ.]
0.0000



Divergence Slit Type
Fixed



Divergence Slit Size [°]
1.0000



Specimen Length [mm]
various



Receiving Slit Size [mm]
0.2000



Measurement Temperature [° C.]
20.00



Anode Material
Cu



K-Alpha1 [Å]
1.54060



K-Alpha2 [Å]
1.54443



K-Beta [Å]
1.39225



K-A2/K-A1 Ratio
0.50000 (nominal)



Generator Settings
40 mA, 40 kV



Diffractometer Type
d5000



Diffractometer Number
0



Goniometer Radius [mm]
217.50



Incident Beam Monochromator
No



Diffracted Beam Monochromator
(Graphite)



Spinning
Yes











Single Crystal X-Ray Diffraction (SXRD)


All measurements were carried out using a Bruker Smart Apex diffractometer operating with Mo-Kα radiation. Unless otherwise specified the data were obtained in 60 ω-scan 10 s images collected in three separate settings of 2θ and φ.


Differential Scanning Calorimetry (DSC)


Approximately 5 mg of sample was weighed into an aluminium DSC pan and sealed with a pierced aluminium lid (non-hermetically). The sample pan was then loaded into a Seiko DSC6200 (equipped with a cooler), cooled, and held at 25° C. Once a stable heat-flow response was obtained, the sample and reference were then heated to about 250° C. at a scan rate of 10° C./min and the resulting heat flow response monitored. Prior to analysis, the instrument was temperature and heat-flow calibrated using an indium reference standard. Sample analysis was carried out by Muse measurement software where the temperatures of thermal events were quoted as the onset temperature, measured according to the manufacturer's specifications.


Thermogravimetric Gravimetric/Differential Thermal Analysis (TG/DTA)


Approximately 5 mg of sample was weighed into an aluminium pan and loaded into a simultaneous thermogravimetric/differential thermal analyser (DTA) and held at room temperature. The sample was then heated at a rate of 10° C./min from 25° C. to 300° C. during which time the change in sample weight was monitored along with any thermal events (DTA). Nitrogen was used as the purge gas, at a flow rate of 20 cm3/min. Prior to analysis the instrument was weight and temperature calibrated using a 100 mg reference weight and an indium reference standard, respectively.


Dynamic Vapor Sorption (DVS)


Approximately 10 mg of sample was placed into a wire-mesh vapor sorption balance pan and loaded into a DVS-1 dynamic vapor sorption balance supplied by Scientific and Medical Systems (SMS). The sample was then dried by maintaining a 0% humidity environment until no further weight change was recorded. The sample was then subjected to a ramping profile from 0-90% relative humidity (RH) at 10% increments, maintaining the sample at each step until a stable weight had been achieved (99.5% step completion). After completion of the sorption cycle, the sample was then dried using the same procedure. The weight change during the sorption/desorption cycles were then plotted, allowing for the hygroscopic nature of the sample to be determined.



1H Nuclear Magnetic Resonance (NMR)



1H NMR was performed on a Bruker AC200. An NMR of each sample was performed in d-H2O and each sample was prepared to about 5 mg concentration. The NMR spectra for L-ornithine benzoate and L-ornithine phenyl acetate are provided in FIGS. 21 and 22, respectively.


Solubility Approximations


Approximately, 25 mg portions of the sample were placed in vials 5 volume increments of the appropriate solvent system were added. Between each addition, the mixture was checked for dissolution and if no dissolution was apparent, the mixture was warmed to 50° C., and checked again. The procedure was continued until dissolution was observed or when 100 volumes of solvent had been added.


HPLC Solubility Determinations


Slurries of each solvent were prepared and the samples shaken for about 48 hrs at 25° C. Each sample was then drawn through a filter, and the filtrate transferred to an HPLC vial for analysis. From the data the solubility of L-ornithine phenyl acetate for each solvent was determined.


Temperature Cycling Experiments


Using the information gathered from the solubility approximations, slurries of the sample were prepared in 24 selected solvent systems. The slurries were temperature cycled at 40° C. or 25° C. in 4 hour cycles for a period of 72 hours. The solids were visually checked for any obvious signs of degradation (i.e. color changes) and then, if not degraded, isolated by filtration. The solids were allowed to dry at ambient conditions for about 24 hours prior to analysis.


Crash Cooling Experiments


Crash cooling experiments were performed by placing saturated solutions of the sample, in the 24 selected solvent systems, in environments of 4° C. and −21° C. for about 48 hours. Any solid material was recovered and the solids were allowed to dry at ambient conditions for about 24 hours prior to analysis.


Evaporation Experiments


Evaporation experiments were conducted by allowing saturated solutions of the sample to evaporate freely at ambient conditions. The solid material was then recovered after the material had evaporated to dryness and analyzed.


Anti-Solvent Addition Experiments


Anti-solvent addition experiments were conducted by adding anti-solvent to saturated solutions of the sample. The addition was continued until there was no further precipitation and the samples adjusted to various temperature for 24 hours: elevated, ambient, 4° C. or −21°. The solid was then isolated and dried at ambient conditions for about 24 hours prior to analysis.


Polarized Light Microscopy (PLM)


The presence of crystallinity (birefringence) was determined using a Leica Leitz DMRB polarised optical microscope equipped with a high resolution Leica camera and image capture software (Firecam V.1.0). All images were recorded using a 10× objective, unless otherwise stated.


Silver Analysis


All silver analysis was carried out on an Agilent 7500ce ICP-MS.


Intrinsic Dissolution Rates


Approximately 100 mg of each form was compressed into discs by placing the material into a die (diameter 12 mm) and compressing the die under 5 tons of pressure in a hydraulic press for about 2 minutes. The dissolution instrument, Sotax AT7 conforms to EP2 and USP2 in which paddles were used to stir the media. Each form was tested under the following pH conditions; 1.0, 4.5 and 6.7, in the stationary disc mode (i.e. discs were added at time=0 seconds and allowed to sink to the bottom of the media). 1 cm3 aliquots of media were extracted from the dissolution pots at times 10, 20, 30, 40, 50, 60, 70, 80 and 120 seconds and tested for API concentration by HPLC. Dissolution curves were plotted and from the first 6 or 7 points on the curves the intrinsic dissolution rate curves were calculated. All tests were carried out at 37° C. and a paddle speed of 150 rpm.


HPLC-UV Instrument Details

    • Instrument: Agilent 1200
    • Column: Gemini C18, 5 μm, 150.0×4.6 mm
    • Column Temperature: 40° C.
    • Mobile Phase A: Phosphate Buffer
    • Mobile Phase B: Acetonitrile
    • Elution: Gradient
    • λ: 210 nm
    • Injection Volume: 10 μL
    • Flow Rate: 1 mL/min


      Thin Layer Chromatography (TLC)


A small spot of solution containing the sample was applied to a plate, about one centimeter from the base. The plate is then dipped into the TLC tank (sealed container) containing methanol:ethyl acetate (95:5) solvent mixture. The solvent moves up the plate by capillary action and meets the sample mixture, which is dissolved and is carried up the plate by the solvent mixture. The number of spots was noted and the Rf values were calculated for each spot.


Infrared (IR)


Infrared spectroscopy was carried out on a Bruker ALPHA P spectrometer. Sufficient material was placed onto the centre of the plate of the spectrometer and the spectra were obtained using the following parameters:

    • Resolution: 4 cm-1
    • Background Scan Time: 16 scans
    • Sample Scan Time: 16 scans
    • Data Collection: 4000 to 400 cm-1
    • Result Spectrum: Transmittance
    • Software: OPUS version 6


      Stabilities Studies: pH 1, 4, 7, 10 and 14 Environments


Slurries (supersaturated solution: about 250 μl of pH solution and solid was added until dissolution was no longer observed and ca. 100 mg of solid was in the slurry) were prepared for each form in a variety of pH environments; 1, 4, 7, 10 and 13.2. The slurries were shaken constantly for a period of 14 days and measurements taken at 7 and 14 day time points. Appropriate buffers were prepared for each pH and are detailed further below.


A buffer having a pH value of 1 was prepared by dissolving 372.75 mg of potassium chloride in 25 ml of deionized water to give a 0.2 M solution. Subsequently, 67 ml of 0.2 M hydrochloric acid was added (this was prepared from a 5 M solution; 10 ml was added to 40 ml of deionized water giving a 1 M solution which was diluted further; 20 ml was added to 80 ml of deionized water giving the required 0.2 M solution) to achieve the desired pH.


A buffer having a pH value of 4 was prepared by dissolving 1.02 g of potassium hydrogen phthalate in 50 ml of deionized water to give a 0.1 M solution.


A buffer having a pH value of 7 was prepared by dissolving 680.00 mg of potassium phosphate monobasic in 50 ml of deionized water to give a 0.1 M solution. Subsequently, 29.1 ml of 0.1 M sodium hydroxide was added (this was prepared from a 1 M solution; 5 ml was added to 45 ml of deionized water giving the required 0.1 M solution) to achieve the desired pH.


A buffer having a pH value of 10 was prepared by dissolving 210.00 mg of sodium bicarbonate in 50 ml of deionized water to give a 0.05 M solution. Subsequently, 10.7 ml of 0.1 M sodium hydroxide was added (this was prepared from a 1 M solution; 5 ml was added to 45 ml of deionized water giving the required 0.1 M solution) to achieve the desired pH.


A buffer having a pH value of pH 13.2 by dissolving 372.75 mg of potassium chloride in 25 ml of deionized water to give a 0.2 M solution. Subsequently, 66 ml of 0.2 M sodium hydroxide was added (this was prepared from a 1 M solution; 20 ml was added to 80 ml of deionized water giving the required 0.2 M solution) taking the pH to 13. 1M sodium hydroxide was then added drop wise to achieve the desired pH.


Example 1: Precipitating Crystalline Forms

Saturated solutions of L-ornithine phenyl acetate were subjected to temperature cycling, crash cooling, evaporation, or anti-solvent addition as described above. The precipitate was analyzed by PLM and XRPD to determine the crystalline form (if any). The results are summarized in TABLE 5.


Six unique crystalline forms were identified from the precipitation studies, Forms I-VI. However, Forms IV and VI were obtained from solutions of acetic acid, and NMR results confirmed these forms to be L-ornithine acetate. Meanwhile, Tests 540-611 utilized samples of L-ornithine phenyl acetate originally isolated by the addition of ethanol anti-solvent. Many of these example produced Form I, which is an ethanol solvate, and therefore it is believed these samples originally included residual ethanol. Consequently, Form I may not be reproduced for certain conditions if the original sample does not include residual ethanol.









TABLE 5







Examples of Preparing Crystalline Forms











Crystallization




Test
Method
Solvent
Results













1
Temp. Cycling
cyclohexanone
Form II


2
Controlled Cool (4° C.)
cyclohexanone
No Solid


3
Controlled Cool (−21° C.)
cyclohexanone
Form V


4
Evaporation
cyclohexanone
Form V


5
Anti-Solvent (IPA)
cyclohexanone
No Solid



Addition Elevated



Temperature


6
Anti-Solvent (IPA)
cyclohexanone
Form II



Addition Ambient



Temperature


7
Anti-Solvent (IPA)
cyclohexanone
Form II



Addition (4° C.)


8
Anti-Solvent (IPA)
cyclohexanone
Form II



Addition (−21° C.)


9
Anti-Solvent (Ethanol)
cyclohexanone
Form II



Addition Ambient



Temperature


10
Anti-Solvent (Ethanol)
cyclohexanone
Form I



Addition (4° C.)


11
Anti-Solvent (Ethanol)
cyclohexanone
Form I



Addition (−21° C.)


12
Temp. Cycling
ethanol/acetone
Form II




(50:50)


13
Controlled Cool (4° C.)
ethanol/acetone
No Solid




(50:50)


14
Controlled Cool (−21° C.)
ethanol/acetone
Form III




(50:50)


15
Evaporation
ethanol/acetone
Form II




(50:50)


16
Anti-Solvent (IPA)
ethanol/acetone
Form II



Addition Elevated
(50:50)



Temperature


17
Anti-Solvent (IPA)
ethanol/acetone
Form II



Addition Ambient
(50:50)



Temperature


18
Anti-Solvent (IPA)
ethanol/acetone
Form II



Addition (4° C.)
(50:50)


19
Anti-Solvent (IPA)
ethanol/acetone
Form II



Addition (−21° C.)
(50:50)


20
Anti-Solvent (Ethanol)
ethanol/acetone
Form II



Addition Ambient
(50:50)



Temperature


21
Anti-Solvent (Ethanol)
ethanol/acetone
Form I



Addition (4° C.)
(50:50)


22
Anti-Solvent (Ethanol)
ethanol/acetone
Form I



Addition (−21° C.)
(50:50)


23
Temp. Cycling
acetic acid
Form IV


24
Controlled Cool (4° C.)
acetic acid
No Solid


25
Controlled Cool (−21° C.)
acetic acid
No Solid


26
Evaporation
acetic acid
Form II


27
Anti-Solvent (IPA)
acetic acid
Form VI



Addition Elevated



Temperature


28
Anti-Solvent (IPA)
acetic acid
Form IV



Addition Ambient



Temperature


29
Anti-Solvent (IPA)
acetic acid
Form IV



Addition (4° C.)


30
Anti-Solvent (IPA)
acetic acid
Form IV



Addition (−21° C.)


31
Anti-Solvent (Ethanol)
acetic acid
Form IV



Addition Ambient



Temperature


32
Anti-Solvent (Ethanol)
acetic acid
Form IV



Addition (4° C.)


33
Anti-Solvent (Ethanol)
acetic acid
Form IV



Addition (−21° C.)


34
Temp. Cycling
1-propanol
Form II


35
Controlled Cool (4° C.)
1-propanol
Form II


36
Controlled Cool (−21° C.)
1-propanol
Form II


37
Evaporation
1-propanol
Form II


38
Anti-Solvent (IPA)
1-propanol
Form II



Addition Elevated



Temperature


39
Anti-Solvent (IPA)
1-propanol
Form II



Addition Ambient



Temperature


40
Anti-Solvent (IPA)
1-propanol
Form II



Addition (4° C.)


41
Anti-Solvent (IPA)
1-propanol
Form II



Addition (−21° C.)


42
Anti-Solvent (Ethanol)
1-propanol
Form II



Addition Ambient



Temperature


43
Anti-Solvent (Ethanol)
1-propanol
Form I/II



Addition (4° C.)


44
Anti-Solvent (Ethanol)
1-propanol
Form I



Addition (−21° C.)


45
Temp. Cycling
dimethylcarbonate
Form II


46
Controlled Cool (4° C.)
dimethylcarbonate
No Solid


47
Controlled Cool (−21° C.)
dimethylcarbonate
Form II


48
Evaporation
dimethylcarbonate
Form II


49
Anti-Solvent (IPA)
dimethylcarbonate
Form II



Addition Elevated



Temperature


50
Anti-Solvent (IPA)
dimethylcarbonate
Form II



Addition Ambient



Temperature


51
Anti-Solvent (IPA)
dimethylcarbonate
Form II



Addition (4° C.)


52
Anti-Solvent (IPA)
dimethylcarbonate
Form II



Addition (−21° C.)


53
Anti-Solvent (Ethanol)
dimethylcarbonate
Form II



Addition Ambient



Temperature


54
Anti-Solvent (Ethanol)
dimethylcarbonate
Form I



Addition (4° C.)


55
Anti-Solvent (Ethanol)
dimethylcarbonate
Form II



Addition (−21° C.)


56
Temp. Cycling
NMP
Form II


57
Controlled Cool (4° C.)
NMP
Form II


58
Controlled Cool (−21° C.)
NMP
Form II


59
Evaporation
NMP
Form II


60
Anti-Solvent (IPA)
NMP
Form II



Addition Elevated



Temperature


61
Anti-Solvent (IPA)
NMP
Form II



Addition Ambient



Temperature


62
Anti-Solvent (IPA)
NMP
Form II



Addition (4° C.)


63
Anti-Solvent (IPA)
NMP
Form II



Addition (−21° C.)


64
Anti-Solvent (Ethanol)
NMP
Form II



Addition Ambient



Temperature


65
Anti-Solvent (Ethanol)
NMP
Form I/II



Addition (4° C.)


66
Anti-Solvent (Ethanol)
NMP
Form II



Addition (−21° C.)


67
Temp. Cycling
EtOAc/cyclohexane
Form II




(1:2)


68
Controlled Cool (4° C.)
EtOAc/cyclohexane
No Solid




(1:2)


69
Controlled Cool (−21° C.)
EtOAc/cyclohexane
No Solid




(1:2)


70
Evaporation
etOAc/cyclohexane
Form II




(1:2)


71
Anti-Solvent (IPA)
etOAc/cyclohexane
Form II



Addition Elevated
(1:2)



Temperature


72
Anti-Solvent (IPA)
etOAc/cyclohexane
Form II



Addition Ambient
(1:2)



Temperature


73
Anti-Solvent (IPA)
etOAc/cyclohexane
Form II



Addition (4° C.)
(1:2)


74
Anti-Solvent (IPA)
etOAc/cyclohexane
Form II



Addition (−21° C.)
(1:2)


75
Anti-Solvent (Ethanol)
etOAc/cyclohexane
Form II



Addition Ambient
(1:2)



Temperature


76
Anti-Solvent (Ethanol)
etOAc/cyclohexane
Form I



Addition (4° C.)
(1:2)


77
Anti-Solvent (Ethanol)
etOAc/cyclohexane
Form I/II



Addition (−21° C.)
(1:2)


78
Temp. Cycling
etOAc/toluene (1:2)
Form II


79
Controlled Cool (4° C.)
etOAc/toluene (1:2)
No Solid


80
Controlled Cool (−21° C.)
etOAc/toluene (1:2)
Form II


81
Evaporation
etOAc/toluene (1:2)
Form II


82
Anti-Solvent (IPA)
etOAc/toluene (1:2)
Form II



Addition Elevated



Temperature


83
Anti-Solvent (IPA)
etOAc/toluene (1:2)
Form II



Addition Ambient



Temperature


84
Anti-Solvent (IPA)
etOAc/toluene (1:2)
Form II



Addition (4° C.)


85
Anti-Solvent (IPA)
etOAc/toluene (1:2)
Form II



Addition (−21° C.)


86
Anti-Solvent (Ethanol)
etOAc/toluene (1:2)
Form II



Addition Ambient



Temperature


87
Anti-Solvent (Ethanol)
etOAc/toluene (1:2)
Form I



Addition (4° C.)


88
Anti-Solvent (Ethanol)
etOAc/toluene (1:2)
Form II



Addition (−21° C.)


89
Temp. Cycling
IPA/diisopropyl ether
Form II




(1:2)


90
Controlled Cool (4° C.)
IPA/diisopropyl ether
Form V




(1:2)


91
Controlled Cool (−21° C.)
IPA/diisopropyl ether
Form II




(1:2)


92
Evaporation
IPA/diisopropyl ether
Form II




(1:2)


93
Anti-Solvent (IPA)
IPA/diisopropyl ether
Form II



Addition Elevated
(1:2)



Temperature


94
Anti-Solvent (IPA)
IPA/diisopropyl ether
Form II



Addition Ambient
(1:2)



Temperature


95
Anti-Solvent (IPA)
IPA/diisopropyl ether
Form II



Addition (4° C.)
(1:2)


96
Anti-Solvent (IPA)
IPA/diisopropyl ether
Form II



Addition (−21° C.)
(1:2)


97
Anti-Solvent (Ethanol)
IPA/diisopropyl ether
Form II



Addition Ambient
(1:2)



Temperature


98
Anti-Solvent (Ethanol)
IPA/diisopropyl ether
Form I



Addition (4° C.)
(1:2)


99
Anti-Solvent (Ethanol)
IPA/diisopropyl ether
Form I/II



Addition (−21° C.)
(1:2)


100
Temp. Cycling
DIPE
Form II


101
Controlled Cool (4° C.)
DIPE
No Solid


102
Controlled Cool (−21° C.)
DIPE
Form V


103
Evaporation
DIPE
Form II


104
Anti-Solvent (IPA)
DIPE
Form II



Addition Elevated



Temperature


105
Anti-Solvent (IPA)
DIPE
Form II



Addition Ambient



Temperature


106
Anti-Solvent (IPA)
DIPE
Form II



Addition (4° C.)


107
Anti-Solvent (IPA)
DIPE
Form II



Addition (−21° C.)


108
Anti-Solvent (Ethanol)
DIPE
Form II



Addition Ambient



Temperature


109
Anti-Solvent (Ethanol)
DIPE
Form I



Addition (4° C.)


110
Anti-Solvent (Ethanol)
DIPE
Form II



Addition (−21° C.)


111
Temp. Cycling
nitromethane/water
No Solid




(20%)


112
Controlled Cool (4° C.)
nitromethane/water
No Solid




(20%)


113
Controlled Cool (−21° C.)
nitromethane/water
No Solid




(20%)


114
Evaporation
nitromethane/water
Form II




(20%)


115
Anti-Solvent (IPA)
nitromethane/water
No Solid



Addition Elevated
(20%)



Temperature


116
Anti-Solvent (IPA)
nitromethane/water
Form II



Addition Ambient
(20%)



Temperature


117
Anti-Solvent (IPA)
nitromethane/water
Form II



Addition (4° C.)
(20%)


118
Anti-Solvent (IPA)
nitromethane/water
Form II



Addition (−21° C.)
(20%)


119
Anti-Solvent (Ethanol)
nitromethane/water
Form II



Addition Ambient
(20%)



Temperature


120
Anti-Solvent (Ethanol)
nitromethane/water
Form I



Addition (4° C.)
(20%)


121
Anti-Solvent (Ethanol)
nitromethane/water
Form I/II



Addition (−21° C.)
(20%)


122
Temp. Cycling
acetone/water (20%)
No Solid


123
Controlled Cool (4° C.)
acetone/water (20%)
Form II


124
Controlled Cool (−21° C.)
acetone/water (20%)
Form II


125
Evaporation
acetone/water (20%)
Form II


126
Anti-Solvent (IPA)
acetone/water (20%)
Form II



Addition Elevated



Temperature


127
Anti-Solvent (IPA)
acetone/water (20%)
Form II



Addition Ambient



Temperature


128
Anti-Solvent (IPA)
acetone/water (20%)
Form II



Addition (4° C.)


129
Anti-Solvent (IPA)
acetone/water (20%)
Form II



Addition (−21° C.)


130
Anti-Solvent (Ethanol)
acetone/water (20%)
Form II



Addition Ambient



Temperature


131
Anti-Solvent (Ethanol)
acetone/water (20%)
Form I



Addition (4° C.)


132
Anti-Solvent (Ethanol)
acetone/water (20%)
Form II



Addition (−21° C.)


133
Temp. Cycling
1,4 dioxane/water
Form II




(20%)


134
Controlled Cool (4° C.)
1,4 dioxane/water
Form II




(20%)


135
Controlled Cool (−21° C.)
1,4 dioxane/water
No Solid




(20%)


136
Evaporation
1,4 dioxane/water
Form II




(20%)


137
Anti-Solvent (IPA)
1,4 dioxane/water
Form II



Addition Elevated
(20%)



Temperature


138
Anti-Solvent (IPA)
1,4 dioxane/water
Form II



Addition Ambient
(20%)



Temperature


139
Anti-Solvent (IPA)
1,4 dioxane/water
Form II



Addition (4° C.)
(20%)


140
Anti-Solvent (IPA)
1,4 dioxane/water
Form II



Addition (−21° C.)
(20%)


141
Anti-Solvent (Ethanol)
1,4 dioxane/water
Form II



Addition Ambient
(20%)



Temperature


142
Anti-Solvent (Ethanol)
1,4 dioxane/water
Form I



Addition (4° C.)
(20%)


143
Anti-Solvent (Ethanol)
1,4 dioxane/water
Form II



Addition (−21° C.)
(20%)


144
Temp. Cycling
diethyl ether
Form II


145
Controlled Cool (4° C.)
diethyl ether
No Solid


146
Controlled Cool (−21° C.)
diethyl ether
No Solid


147
Evaporation
diethyl ether
Form II


148
Anti-Solvent (IPA)
diethyl ether
Form II



Addition Elevated



Temperature


149
Anti-Solvent (IPA)
diethyl ether
Form II



Addition Ambient



Temperature


150
Anti-Solvent (IPA)
diethyl ether
Form II



Addition (4° C.)


151
Anti-Solvent (IPA)
diethyl ether
Form II



Addition (−21° C.)


152
Anti-Solvent (Ethanol)
diethyl ether
Form II



Addition Ambient



Temperature


153
Anti-Solvent (Ethanol)
diethyl ether
Form I



Addition (4° C.)


154
Anti-Solvent (Ethanol)
diethyl ether
Form I/II



Addition (−21° C.)


155
Temp. Cycling
ethylene glycol
Form II


156
Controlled Cool (4° C.)
ethylene glycol
No Solid


157
Controlled Cool (−21° C.)
ethylene glycol
No Solid


158
Evaporation
ethylene glycol
No Solid


159
Anti-Solvent (IPA)
ethylene glycol
Form II



Addition Elevated



Temperature


160
Anti-Solvent (IPA)
ethylene glycol
Form II



Addition Ambient



Temperature


161
Anti-Solvent (IPA)
ethylene glycol
Form II



Addition (4° C.)


162
Anti-Solvent (IPA)
ethylene glycol
Form II



Addition (−21° C.)


163
Anti-Solvent (Ethanol)
ethylene glycol
Form II



Addition Ambient



Temperature


164
Anti-Solvent (Ethanol)
ethylene glycol
Form II



Addition (4° C.)


165
Anti-Solvent (Ethanol)
ethylene glycol
Form II



Addition (−21° C.)


166
Temp. Cycling
meOAc/water (20%)
No Solid


167
Controlled Cool (4° C.)
meOAc/water (20%)
No Solid


168
Controlled Cool (−21° C.)
meOAc/water (20%)
No Solid


169
Evaporation
meOAc/water (20%)
Form II


170
Anti-Solvent (IPA)
meOAc/water (20%)
Form II



Addition Elevated



Temperature


171
Anti-Solvent (IPA)
meOAc/water (20%)
Form II



Addition Ambient



Temperature


172
Anti-Solvent (IPA)
meOAc/water (20%)
Form II



Addition (4° C.)


173
Anti-Solvent (IPA)
meOAc/water (20%)
Form II



Addition (−21° C.)


174
Anti-Solvent (Ethanol)
meOAc/water (20%)
Form II



Addition Ambient



Temperature


175
Anti-Solvent (Ethanol)
meOAc/water (20%)
Form I/II



Addition (4° C.)


176
Anti-Solvent (Ethanol)
meOAc/water (20%)
Form II



Addition (−21° C.)


177
Temp. Cycling
meOH/acetone
Form II




(50:50)


178
Controlled Cool (4° C.)
meOH/acetone
No Solid




(50:50)


179
Controlled Cool (−21° C.)
meOH/acetone
No Solid




(50:50)


180
Evaporation
meOH/acetone
Form II




(50:50)


181
Anti-Solvent (IPA)
meOH/acetone
Form II



Addition Elevated
(50:50)



Temperature


182
Anti-Solvent (IPA)
meOH/acetone
Form II



Addition Ambient
(50:50)



Temperature


183
Anti-Solvent (IPA)
meOH/acetone
Form II



Addition (4° C.)
(50:50)


184
Anti-Solvent (IPA)
meOH/acetone
Form II



Addition (−21° C.)
(50:50)


185
Anti-Solvent (Ethanol)
meOH/acetone
Form II



Addition Ambient
(50:50)



Temperature


186
Anti-Solvent (Ethanol)
meOH/acetone
Form I



Addition (4° C.)
(50:50)


187
Anti-Solvent (Ethanol)
meOH/acetone
Form I/II



Addition (−21° C.)
(50:50)


188
Temp. Cycling
DMF
Form II


189
Controlled Cool (4° C.)
DMF
Form II


190
Controlled Cool (−21° C.)
DMF
Form II


191
Evaporation
DMF
Form II


192
Anti-Solvent (IPA)
DMF
Form II



Addition Elevated



Temperature


193
Anti-Solvent (IPA)
DMF
Form II



Addition Ambient



Temperature


194
Anti-Solvent (IPA)
DMF
Form II



Addition (4° C.)


195
Anti-Solvent (IPA)
DMF
Form II



Addition (−21° C.)


196
Anti-Solvent (Ethanol)
DMF
Form II



Addition Ambient



Temperature


197
Anti-Solvent (Ethanol)
DMF
Form I/II



Addition (4° C.)


198
Anti-Solvent (Ethanol)
DMF
Form II



Addition (−21° C.)


199
Temp. Cycling
2-butanol
Form II


200
Controlled Cool (4° C.)
2-butanol
No Solid


201
Controlled Cool (−21° C.)
2-butanol
No Solid


202
Evaporation
2-butanol
Form II


203
Anti-Solvent (IPA)
2-butanol
Form III



Addition Elevated



Temperature


204
Anti-Solvent (IPA)
2-butanol
Form II



Addition Ambient



Temperature


205
Anti-Solvent (IPA)
2-butanol
Form II



Addition (4° C.)


206
Anti-Solvent (IPA)
2-butanol
Form II



Addition (−21° C.)


207
Anti-Solvent (Ethanol)
2-butanol
Form II



Addition Ambient



Temperature


208
Anti-Solvent (Ethanol)
2-butanol
Form I/II



Addition (4° C.)


209
Anti-Solvent (Ethanol)
2-butanol
Form I/II



Addition (−21° C.)


210
Temp. Cycling
cumene
Form II


211
Controlled Cool (4° C.)
cumene
No Solid


212
Controlled Cool (−21° C.)
cumene
No Solid


213
Evaporation
cumene
Form II


214
Anti-Solvent (IPA)
cumene
Form II



Addition Elevated



Temperature


215
Anti-Solvent (IPA)
cumene
Form II



Addition Ambient



Temperature


216
Anti-Solvent (IPA)
cumene
Form II



Addition (4° C.)


217
Anti-Solvent (IPA)
cumene
Form II



Addition (−21° C.)


218
Anti-Solvent (Ethanol)
cumene
Form II



Addition Ambient



Temperature


219
Anti-Solvent (Ethanol)
cumene
Form II



Addition (4° C.)


220
Anti-Solvent (Ethanol)
cumene
Form I/II



Addition (−21° C.)


221
Temp. Cycling
ethyl formate
Form II


222
Controlled Cool (4° C.)
ethyl formate
No Solid


223
Controlled Cool (−21° C.)
ethyl formate
Form II


224
Evaporation
ethyl formate
Form II


225
Anti-Solvent (IPA)
ethyl formate
Form II



Addition Elevated



Temperature


226
Anti-Solvent (IPA)
ethyl formate
Form II



Addition Ambient



Temperature


227
Anti-Solvent (IPA)
ethyl formate
Form II



Addition (4° C.)


228
Anti-Solvent (IPA)
ethyl formate
Form II



Addition (−21° C.)


229
Anti-Solvent (Ethanol)
ethyl formate
Form II



Addition Ambient



Temperature


230
Anti-Solvent (Ethanol)
ethyl formate
Form I



Addition (4° C.)


231
Anti-Solvent (Ethanol)
ethyl formate
Form I/II



Addition (−21° C.)


232
Temp. Cycling
isobutyl acetate
Form II


233
Controlled Cool (4° C.)
isobutyl acetate
No Solid


234
Controlled Cool (−21° C.)
isobutyl acetate
Form II


235
Evaporation
isobutyl acetate
No Solid


236
Anti-Solvent (IPA)
isobutyl acetate
Form II



Addition Elevated



Temperature


237
Anti-Solvent (IPA)
isobutyl acetate
Form II



Addition Ambient



Temperature


238
Anti-Solvent (IPA)
isobutyl acetate
Form II



Addition (4° C.)


239
Anti-Solvent (IPA)
isobutyl acetate
Form III



Addition (−21° C.)


240
Anti-Solvent (Ethanol)
isobutyl acetate
Form II



Addition Ambient



Temperature


241
Anti-Solvent (Ethanol)
isobutyl acetate
Form II



Addition (4° C.)


242
Anti-Solvent (Ethanol)
isobutyl acetate
Form I/II



Addition (−21° C.)


243
Temp. Cycling
3-methyl-1-butanol
Form II


244
Controlled Cool (4° C.)
3-methyl-1-butanol
No Solid


245
Controlled Cool (−21° C.)
3-methyl-1-butanol
No Solid


246
Evaporation
3-methyl-1-butanol
Form II


247
Anti-Solvent (IPA)
3-methyl-1-butanol
Form II



Addition Elevated



Temperature


248
Anti-Solvent (IPA)
3-methyl-1-butanol
Form II



Addition Ambient



Temperature


249
Anti-Solvent (IPA)
3-methyl-1-butanol
Form II



Addition (4° C.)


250
Anti-Solvent (IPA)
3-methyl-1-butanol
Form II



Addition (−21° C.)


251
Anti-Solvent (Ethanol)
3-methyl-1-butanol
Form II



Addition Ambient



Temperature


252
Anti-Solvent (Ethanol)
3-methyl-1-butanol
Form I/II



Addition (4° C.)


253
Anti-Solvent (Ethanol)
3-methyl-1-butanol
Form I/II



Addition (−21° C.)


254
Temp. Cycling
anisole
Form II


255
Controlled Cool (4° C.)
anisole
No Solid


256
Controlled Cool (−21° C.)
anisole
Form II


257
Evaporation
anisole
Form II


258
Anti-Solvent (IPA)
anisole
Form II



Addition Elevated



Temperature


259
Anti-Solvent (IPA)
anisole
Form II



Addition Ambient



Temperature


260
Anti-Solvent (IPA)
anisole
Form II



Addition (4° C.)


261
Anti-Solvent (IPA)
anisole
Form II/IV



Addition (−21° C.)


262
Anti-Solvent (Ethanol)
anisole
Form II



Addition Ambient



Temperature


263
Anti-Solvent (Ethanol)
anisole
Form II



Addition (4° C.)


264
Anti-Solvent (Ethanol)
anisole
Form I



Addition (−21° C.)


265
Temp. Cycling
IPA/isopropyl acetate
Form II




(1:2)


266
Controlled Cool (4° C.)
IPA/isopropyl acetate
No Solid




(1:2)


267
Controlled Cool (−21° C.)
IPA/isopropyl acetate
No Solid




(1:2)


268
Evaporation
IPA/isopropyl acetate
Form II




(1:2)


269
Anti-Solvent (IPA)
IPA/isopropyl acetate
Form II



Addition Elevated
(1:2)



Temperature


270
Anti-Solvent (IPA)
IPA/isopropyl acetate
Form II



Addition Ambient
(1:2)



Temperature


271
Anti-Solvent (IPA)
IPA/isopropyl acetate
Form II



Addition (4° C.)
(1:2)


272
Anti-Solvent (IPA)
IPA/isopropyl acetate
Form II



Addition (−21° C.)
(1:2)


273
Anti-Solvent (Ethanol)
IPA/isopropyl acetate
Form II



Addition Ambient
(1:2)



Temperature


274
Anti-Solvent (Ethanol)
IPA/isopropyl acetate
Form II



Addition (4° C.)
(1:2)


275
Anti-Solvent (Ethanol)
IPA/isopropyl acetate
Form I



Addition (−21° C.)
(1:2)


276
Temp. Cycling
EtOH: 1% H20
Form II


277
Controlled Cool (4° C.)
EtOH: 1% H20
No Solid


278
Controlled Cool (−21° C.)
EtOH: 1% H20
No Solid


279
Evaporation
EtOH: 1% H20
No Solid


280
Anti-Solvent (IPA)
EtOH: 1% H20
No Solid



Addition Elevated



Temperature


281
Anti-Solvent (IPA)
EtOH: 1% H20
No Solid



Addition Ambient



Temperature


282
Anti-Solvent (IPA)
EtOH: 1% H20
No Solid



Addition (4° C.)


283
Anti-Solvent (IPA)
EtOH: 1% H20
No Solid



Addition (−21° C.)


284
Anti-Solvent (Ethanol)
EtOH: 1% H20
No Solid



Addition Ambient



Temperature


285
Anti-Solvent (Ethanol)
EtOH: 1% H20
No Solid



Addition (4° C.)


286
Anti-Solvent (Ethanol)
EtOH: 1% H20
No Solid



Addition (−21° C.)


287
Temp. Cycling
EtOH: 3% H20
Form II


288
Controlled Cool (4° C.)
EtOH: 3% H20
No Solid


289
Controlled Cool (−21° C.)
EtOH: 3% H20
No Solid


290
Evaporation
EtOH: 3% H20
No Solid


291
Anti-Solvent (IPA)
EtOH: 3% H20
No Solid



Addition Elevated



Temperature


292
Anti-Solvent (IPA)
EtOH: 3% H20
No Solid



Addition Ambient



Temperature


293
Anti-Solvent (IPA)
EtOH: 3% H20
No Solid



Addition (4° C.)


294
Anti-Solvent (IPA)
EtOH: 3% H20
No Solid



Addition (−21° C.)


295
Anti-Solvent (Ethanol)
EtOH: 3% H20
No Solid



Addition Ambient



Temperature


296
Anti-Solvent (Ethanol)
EtOH: 3% H20
No Solid



Addition (4° C.)


297
Anti-Solvent (Ethanol)
EtOH: 3% H20
No Solid



Addition (−21° C.)


298
Temp. Cycling
EtOH: 5% H20
Form II


299
Controlled Cool (4° C.)
EtOH: 5% H20
No Solid


300
Controlled Cool (−21° C.)
EtOH: 5% H20
No Solid


301
Evaporation
EtOH: 5% H20
Form II


302
Anti-Solvent (IPA)
EtOH: 5% H20
No Solid



Addition Elevated



Temperature


303
Anti-Solvent (IPA)
EtOH: 5% H20
Form II



Addition Ambient



Temperature


304
Anti-Solvent (IPA)
EtOH: 5% H20
No Solid



Addition (4° C.)


305
Anti-Solvent (IPA)
EtOH: 5% H20
No Solid



Addition (−21° C.)


306
Anti-Solvent (Ethanol)
EtOH: 5% H20
No Solid



Addition Ambient



Temperature


307
Anti-Solvent (Ethanol)
EtOH: 5% H20
No Solid



Addition (4° C.)


308
Anti-Solvent (Ethanol)
EtOH: 5% H20
No Solid



Addition (−21° C.)


309
Temp. Cycling
IPA: 1% H20
Form II


310
Controlled Cool (4° C.)
IPA: 1% H20
No Solid


311
Controlled Cool (−21° C.)
IPA: 1% H20
No Solid


312
Evaporation
IPA: 1% H20
No Solid


313
Anti-Solvent (IPA)
IPA: 1% H20
No Solid



Addition Elevated



Temperature


314
Anti-Solvent (IPA)
IPA: 1% H20
No Solid



Addition Ambient



Temperature


315
Anti-Solvent (IPA)
IPA: 1% H20
No Solid



Addition (4° C.)


316
Anti-Solvent (IPA)
IPA: 1% H20
No Solid



Addition (−21° C.)


317
Anti-Solvent (Ethanol)
IPA: 1% H20
No Solid



Addition Ambient



Temperature


318
Anti-Solvent (Ethanol)
IPA: 1% H20
No Solid



Addition (4° C.)


319
Anti-Solvent (Ethanol)
IPA: 1% H20
No Solid



Addition (−21° C.)


320
Temp. Cycling
IPA: 3% H20
Form II


321
Controlled Cool (4° C.)
IPA: 3% H20
No Solid


322
Controlled Cool (−21° C.)
IPA: 3% H20
No Solid


323
Evaporation
IPA: 3% H20
No Solid


324
Anti-Solvent (IPA)
IPA: 3% H20
No Solid



Addition Elevated



Temperature


325
Anti-Solvent (IPA)
IPA: 3% H20
No Solid



Addition Ambient



Temperature


326
Anti-Solvent (IPA)
IPA: 3% H20
No Solid



Addition (4° C.)


327
Anti-Solvent (IPA)
IPA: 3% H20
No Solid



Addition (−21° C.)


328
Anti-Solvent (Ethanol)
IPA: 3% H20
No Solid



Addition Ambient



Temperature


329
Anti-Solvent (Ethanol)
IPA: 3% H20
No Solid



Addition (4° C.)


330
Anti-Solvent (Ethanol)
IPA: 3% H20
No Solid



Addition (−21° C.)


331
Temp. Cycling
IPA: 5% H20
Form II


332
Controlled Cool (4° C.)
IPA: 5% H20
No Solid


333
Controlled Cool (−21° C.)
IPA: 5% H20
No Solid


334
Evaporation
IPA: 5% H20
Form II


335
Anti-Solvent (IPA)
IPA: 5% H20
No Solid



Addition Elevated



Temperature


336
Anti-Solvent (IPA)
IPA: 5% H20
No Solid



Addition Ambient



Temperature


337
Anti-Solvent (IPA)
IPA: 5% H20
No Solid



Addition (4° C.)


338
Anti-Solvent (IPA)
IPA: 5% H20
No Solid



Addition (−21° C.)


339
Anti-Solvent (Ethanol)
IPA: 5% H20
No Solid



Addition Ambient



Temperature


340
Anti-Solvent (Ethanol)
IPA: 5% H20
No Solid



Addition (4° C.)


341
Anti-Solvent (Ethanol)
IPA: 5% H20
No Solid



Addition (−21° C.)


342
Temp. Cycling
ACN: 1% H20
Form II


343
Controlled Cool (4° C.)
ACN: 1% H20
No Solid


344
Controlled Cool (−21° C.)
ACN: 1% H20
No Solid


345
Evaporation
ACN: 1% H20
Form II


346
Anti-Solvent (IPA)
ACN: 1% H20
No Solid



Addition Elevated



Temperature


347
Anti-Solvent (IPA)
ACN: 1% H20
No Solid



Addition Ambient



Temperature


348
Anti-Solvent (IPA)
ACN: 1% H20
No Solid



Addition (4° C.)


349
Anti-Solvent (IPA)
ACN: 1% H20
No Solid



Addition (−21° C.)


350
Anti-Solvent (Ethanol)
ACN: 1% H20
No Solid



Addition Ambient



Temperature


351
Anti-Solvent (Ethanol)
ACN: 1% H20
No Solid



Addition (4° C.)


352
Anti-Solvent (Ethanol)
ACN: 1% H20
No Solid



Addition (−21° C.)


353
Temp. Cycling
ACN: 6% H20
Form II


354
Controlled Cool (4° C.)
ACN: 6% H20
No Solid


355
Controlled Cool (−21° C.)
ACN: 6% H20
No Solid


356
Evaporation
ACN: 6% H20
Form II


357
Anti-Solvent (IPA)
ACN: 6% H20
No Solid



Addition Elevated



Temperature


358
Anti-Solvent (IPA)
ACN: 6% H20
No Solid



Addition Ambient



Temperature


359
Anti-Solvent (IPA)
ACN: 6% H20
No Solid



Addition (4° C.)


360
Anti-Solvent (IPA)
ACN: 6% H20
No Solid



Addition (−21° C.)


361
Anti-Solvent (Ethanol)
ACN: 6% H20
No Solid



Addition Ambient



Temperature


362
Anti-Solvent (Ethanol)
ACN: 6% H20
No Solid



Addition (4° C.)


363
Anti-Solvent (Ethanol)
ACN: 6% H20
No Solid



Addition (−21° C.)


364
Temp. Cycling
ACN: 12% H20
No Solid


365
Controlled Cool (4° C.)
ACN: 12% H20
No Solid


366
Controlled Cool (−21° C.)
ACN: 12% H20
No Solid


367
Evaporation
ACN: 12% H20
Form II


368
Anti-Solvent (IPA)
ACN: 12% H20
No Solid



Addition Elevated



Temperature


369
Anti-Solvent (IPA)
ACN: 12% H20
Form II



Addition Ambient



Temperature


370
Anti-Solvent (IPA)
ACN: 12% H20
No Solid



Addition (4° C.)


371
Anti-Solvent (IPA)
ACN: 12% H20
Form II



Addition (−21° C.)


372
Anti-Solvent (Ethanol)
ACN: 12% H20
No Solid



Addition Ambient



Temperature


373
Anti-Solvent (Ethanol)
ACN: 12% H20
No Solid



Addition (4° C.)


374
Anti-Solvent (Ethanol)
ACN: 12% H20
No Solid



Addition (−21° C.)


375
Temp. Cycling
DMF: 5% H20
Form II


376
Controlled Cool (4° C.)
DMF: 5% H20
No Solid


377
Controlled Cool (−21° C.)
DMF: 5% H20
No Solid


378
Evaporation
DMF: 5% H20
No Solid


379
Anti-Solvent (IPA)
DMF: 5% H20
No Solid



Addition Elevated



Temperature


380
Anti-Solvent (IPA)
DMF: 5% H20
No Solid



Addition Ambient



Temperature


381
Anti-Solvent (IPA)
DMF: 5% H20
No Solid



Addition (4° C.)


382
Anti-Solvent (IPA)
DMF: 5% H20
No Solid



Addition (−21° C.)


383
Anti-Solvent (Ethanol)
DMF: 5% H20
No Solid



Addition Ambient



Temperature


384
Anti-Solvent (Ethanol)
DMF: 5% H20
No Solid



Addition (4° C.)


385
Anti-Solvent (Ethanol)
DMF: 5% H20
No Solid



Addition (−21° C.)


386
Temp. Cycling
DMF: 15% H20
Form II


387
Controlled Cool (4° C.)
DMF: 15% H20
No Solid


388
Controlled Cool (−21° C.)
DMF: 15% H20
No Solid


389
Evaporation
DMF: 15% H20
No Solid


390
Anti-Solvent (IPA)
DMF: 15% H20
No Solid



Addition Elevated



Temperature


391
Anti-Solvent (IPA)
DMF: 15% H20
No Solid



Addition Ambient



Temperature


392
Anti-Solvent (IPA)
DMF: 15% H20
No Solid



Addition (4° C.)


393
Anti-Solvent (IPA)
DMF: 15% H20
No Solid



Addition (−21° C.)


394
Anti-Solvent (Ethanol)
DMF: 15% H20
No Solid



Addition Ambient



Temperature


395
Anti-Solvent (Ethanol)
DMF: 15% H20
No Solid



Addition (4° C.)


396
Anti-Solvent (Ethanol)
DMF: 15% H20
No Solid



Addition (−21° C.)


397
Temp. Cycling
DMF: 30% H20
Form II


398
Controlled Cool (4° C.)
DMF: 30% H20
Form II


399
Controlled Cool (−21° C.)
DMF: 30% H20
Form II


400
Evaporation
DMF: 30% H20
Form II


401
Anti-Solvent (IPA)
DMF: 30% H20
Form II



Addition Elevated



Temperature


402
Anti-Solvent (IPA)
DMF: 30% H20
Form II



Addition Ambient



Temperature


403
Anti-Solvent (IPA)
DMF: 30% H20
Form II



Addition (4° C.)


404
Anti-Solvent (IPA)
DMF: 30% H20
Form II



Addition (−21° C.)


405
Anti-Solvent (Ethanol)
DMF: 30% H20
Form II



Addition Ambient



Temperature


406
Anti-Solvent (Ethanol)
DMF: 30% H20
No Solid



Addition (4° C.)


407
Anti-Solvent (Ethanol)
DMF: 30% H20
Form II



Addition (−21° C.)


408
Temp. Cycling
1,4-dioxane: 1% H20
Form II


409
Controlled Cool (4° C.)
1,4-dioxane: 1% H20
No Solid


410
Controlled Cool (−21° C.)
1,4-dioxane: 1% H20
No Solid


411
Evaporation
1,4-dioxane: 1% H20
No Solid


412
Anti-Solvent (IPA)
1,4-dioxane: 1% H20
No Solid



Addition Elevated



Temperature


413
Anti-Solvent (IPA)
1,4-dioxane: 1% H20
No Solid



Addition Ambient



Temperature


414
Anti-Solvent (IPA)
1,4-dioxane: 1% H20
No Solid



Addition (4° C.)


415
Anti-Solvent (IPA)
1,4-dioxane: 1% H20
No Solid



Addition (−21° C.)


416
Anti-Solvent (Ethanol)
1,4-dioxane: 1% H20
No Solid



Addition Ambient



Temperature


417
Anti-Solvent (Ethanol)
1,4-dioxane: 1% H20
No Solid



Addition (4° C.)


418
Anti-Solvent (Ethanol)
1,4-dioxane: 1% H20
No Solid



Addition (−21° C.)


419
Temp. Cycling
1,4-dioxane: 3% H20
Form II


420
Controlled Cool (4° C.)
1,4-dioxane: 3% H20
No Solid


421
Controlled Cool (−21° C.)
1,4-dioxane: 3% H20
No Solid


422
Evaporation
1,4-dioxane: 3% H20
Form II


423
Anti-Solvent (IPA)
1,4-dioxane: 3% H20
No Solid



Addition Elevated



Temperature


424
Anti-Solvent (IPA)
1,4-dioxane: 3% H20
No Solid



Addition Ambient



Temperature


425
Anti-Solvent (IPA)
1,4-dioxane: 3% H20
No Solid



Addition (4° C.)


426
Anti-Solvent (IPA)
1,4-dioxane: 3% H20
No Solid



Addition (−21° C.)


427
Anti-Solvent (Ethanol)
1,4-dioxane: 3% H20
No Solid



Addition Ambient



Temperature


428
Anti-Solvent (Ethanol)
1,4-dioxane: 3% H20
No Solid



Addition (4° C.)


429
Anti-Solvent (Ethanol)
1,4-dioxane: 3% H20
No Solid



Addition (−21° C.)


430
Temp. Cycling
1,4-dioxane: 10% H20
Form II


431
Controlled Cool (4° C.)
1,4-dioxane: 10% H20
No Solid


432
Controlled Cool (−21° C.)
1,4-dioxane: 10% H20
No Solid


433
Evaporation
1,4-dioxane: 10% H20
Form II


434
Anti-Solvent (IPA)
1,4-dioxane: 10% H20
No Solid



Addition Elevated



Temperature


435
Anti-Solvent (IPA)
1,4-dioxane: 10% H20
No Solid



Addition Ambient



Temperature


436
Anti-Solvent (IPA)
1,4-dioxane: 10% H20
No Solid



Addition (4° C.)


437
Anti-Solvent (IPA)
1,4-dioxane: 10% H20
No Solid



Addition (−21° C.)


438
Anti-Solvent (Ethanol)
1,4-dioxane: 10% H20
No Solid



Addition Ambient



Temperature


439
Anti-Solvent (Ethanol)
1,4-dioxane: 10% H20
No Solid



Addition (4° C.)


440
Anti-Solvent (Ethanol)
1,4-dioxane: 10% H20
No Solid



Addition (−21° C.)


441
Temp. Cycling
MeOH: 5% H20
Form II


442
Controlled Cool (4° C.)
MeOH: 5% H20
No Solid


443
Controlled Cool (−21° C.)
MeOH: 5% H20
No Solid


444
Evaporation
MeOH: 5% H20
Form II


445
Anti-Solvent (IPA)
MeOH: 5% H20
Form II



Addition Elevated



Temperature


446
Anti-Solvent (IPA)
MeOH: 5% H20
Form II



Addition Ambient



Temperature


447
Anti-Solvent (IPA)
MeOH: 5% H20
Form II



Addition (4° C.)


448
Anti-Solvent (IPA)
MeOH: 5% H20
Form II



Addition (−21° C.)


449
Anti-Solvent (Ethanol)
MeOH: 5% H20
No Solid



Addition Ambient



Temperature


450
Anti-Solvent (Ethanol)
MeOH: 5% H20
Form II



Addition (4° C.)


451
Anti-Solvent (Ethanol)
MeOH: 5% H20
Form II



Addition (−21° C.)


452
Temp. Cycling
MeOH: 20% H20
No Solid


453
Controlled Cool (4° C.)
MeOH: 20% H20
Form II


454
Controlled Cool (−21° C.)
MeOH: 20% H20
Form II


455
Evaporation
MeOH: 20% H20
Form II


456
Anti-Solvent (IPA)
MeOH: 20% H20
Form II



Addition Elevated



Temperature


457
Anti-Solvent (IPA)
MeOH: 20% H20
Form II



Addition Ambient



Temperature


458
Anti-Solvent (IPA)
MeOH: 20% H20
Form II



Addition (4° C.)


459
Anti-Solvent (IPA)
MeOH: 20% H20
Form II



Addition (−21° C.)


460
Anti-Solvent (Ethanol)
MeOH: 20% H20
Form II



Addition Ambient



Temperature


461
Anti-Solvent (Ethanol)
MeOH: 20% H20
Form II



Addition (4° C.)


462
Anti-Solvent (Ethanol)
MeOH: 20% H20
Form I/II



Addition (−21° C.)


463
Temp. Cycling
MeOH: 50% H20
Form II


464
Controlled Cool (4° C.)
MeOH: 50% H20
Form II


465
Controlled Cool (−21° C.)
MeOH: 50% H20
Form II


466
Evaporation
MeOH: 50% H20
No Solid


467
Anti-Solvent (IPA)
MeOH: 50% H20
Form II



Addition Elevated



Temperature


468
Anti-Solvent (IPA)
MeOH: 50% H20
No Solid



Addition Ambient



Temperature


469
Anti-Solvent (IPA)
MeOH: 50% H20
Form II



Addition (4° C.)


470
Anti-Solvent (IPA)
MeOH: 50% H20
No Solid



Addition (−21° C.)


471
Anti-Solvent (Ethanol)
MeOH: 50% H20
Form II



Addition Ambient



Temperature


472
Anti-Solvent (Ethanol)
MeOH: 50% H20
Form I



Addition (4° C.)


473
Anti-Solvent (Ethanol)
MeOH: 50% H20
Form I/II



Addition (−21° C.)


474
Temp. Cycling
THF: 1% H20
Form II


475
Controlled Cool (4° C.)
THF: 1% H20
No Solid


476
Controlled Cool (−21° C.)
THF: 1% H20
No Solid


477
Evaporation
THF: 1% H20
Form II


478
Anti-Solvent (IPA)
THF: 1% H20
No Solid



Addition Elevated



Temperature


479
Anti-Solvent (IPA)
THF: 1% H20
No Solid



Addition Ambient



Temperature


480
Anti-Solvent (IPA)
THF: 1% H20
No Solid



Addition (4° C.)


481
Anti-Solvent (IPA)
THF: 1% H20
No Solid



Addition (−21° C.)


482
Anti-Solvent (Ethanol)
THF: 1% H20
No Solid



Addition Ambient



Temperature


483
Anti-Solvent (Ethanol)
THF: 1% H20
No Solid



Addition (4° C.)


484
Anti-Solvent (Ethanol)
THF: 1% H20
No Solid



Addition (−21° C.)


485
Temp. Cycling
THF: 3% H20
Form II


486
Controlled Cool (4° C.)
THF: 3% H20
No Solid


487
Controlled Cool (−21° C.)
THF: 3% H20
No Solid


488
Evaporation
THF: 3% H20
No Solid


489
Anti-Solvent (IPA)
THF: 3% H20
No Solid



Addition Elevated



Temperature


490
Anti-Solvent (IPA)
THF: 3% H20
Form II



Addition Ambient



Temperature


491
Anti-Solvent (IPA)
THF: 3% H20
No Solid



Addition (4° C.)


492
Anti-Solvent (IPA)
THF: 3% H20
No Solid



Addition (−21° C.)


493
Anti-Solvent (Ethanol)
THF: 3% H20
No Solid



Addition Ambient



Temperature


494
Anti-Solvent (Ethanol)
THF: 3% H20
No Solid



Addition (4° C.)


495
Anti-Solvent (Ethanol)
THF: 3% H20
No Solid



Addition (−21° C.)


496
Temp. Cycling
THF: 5% H20
No Solid


497
Controlled Cool (4° C.)
THF: 5% H20
No Solid


498
Controlled Cool (−21° C.)
THF: 5% H20
No Solid


499
Evaporation
THF: 5% H20
Form II


500
Anti-Solvent (IPA)
THF: 5% H20
No Solid



Addition Elevated



Temperature


501
Anti-Solvent (IPA)
THF: 5% H20
No Solid



Addition Ambient



Temperature


502
Anti-Solvent (IPA)
THF: 5% H20
Form II



Addition (4° C.)


503
Anti-Solvent (IPA)
THF: 5% H20
No Solid



Addition (−21° C.)


504
Anti-Solvent (Ethanol)
THF: 5% H20
No Solid



Addition Ambient



Temperature


505
Anti-Solvent (Ethanol)
THF: 5% H20
No Solid



Addition (4° C.)


506
Anti-Solvent (Ethanol)
THF: 5% H20
No Solid



Addition (−21° C.)


507
Temp. Cycling
butan-1-ol: 1% H20
Form II


508
Controlled Cool (4° C.)
butan-1-ol: 1% H20
No Solid


509
Controlled Cool (−21° C.)
butan-1-ol: 1% H20
No Solid


510
Evaporation
butan-1-ol: 1% H20
Form II


511
Anti-Solvent (IPA)
butan-1-ol: 1% H20
No Solid



Addition Elevated



Temperature


512
Anti-Solvent (IPA)
butan-1-ol: 1% H20
No Solid



Addition Ambient



Temperature


513
Anti-Solvent (IPA)
butan-1-ol: 1% H20
No Solid



Addition (4° C.)


514
Anti-Solvent (IPA)
butan-1-ol: 1% H20
No Solid



Addition (−21° C.)


515
Anti-Solvent (Ethanol)
butan-1-ol: 1% H20
No Solid



Addition Ambient



Temperature


516
Anti-Solvent (Ethanol)
butan-1-ol: 1% H20
No Solid



Addition (4° C.)


517
Anti-Solvent (Ethanol)
butan-1-ol: 1% H20
No Solid



Addition (−21° C.)


518
Temp. Cycling
butan-1-ol: 3% H20
Form II


519
Controlled Cool (4° C.)
butan-1-ol: 3% H20
No Solid


520
Controlled Cool (−21° C.)
butan-1-ol: 3% H20
No Solid


521
Evaporation
butan-1-ol: 3% H20
Form II


522
Anti-Solvent (IPA)
butan-1-ol: 3% H20
No Solid



Addition Elevated



Temperature


523
Anti-Solvent (IPA)
butan-1-ol: 3% H20
No Solid



Addition Ambient



Temperature


524
Anti-Solvent (IPA)
butan-1-ol: 3% H20
No Solid



Addition (4° C.)


525
Anti-Solvent (IPA)
butan-1-ol: 3% H20
No Solid



Addition (−21° C.)


526
Anti-Solvent (Ethanol)
butan-1-ol: 3% H20
No Solid



Addition Ambient



Temperature


527
Anti-Solvent (Ethanol)
butan-1-ol: 3% H20
No Solid



Addition (4° C.)


528
Anti-Solvent (Ethanol)
butan-1-ol: 3% H20
No Solid



Addition (−21° C.)


529
Temp. Cycling
butan-1-ol: 5% H20
Form II


530
Controlled Cool (4° C.)
butan-1-ol: 5% H20
No Solid


531
Controlled Cool (−21° C.)
butan-1-ol: 5% H20
No Solid


532
Evaporation
butan-1-ol: 5% H20
Form II


533
Anti-Solvent (IPA)
butan-1-ol: 5% H20
No Solid



Addition Elevated



Temperature


534
Anti-Solvent (IPA)
butan-1-ol: 5% H20
Form II



Addition Ambient



Temperature


535
Anti-Solvent (IPA)
butan-1-ol: 5% H20
No Solid



Addition (4° C.)


536
Anti-Solvent (IPA)
butan-1-ol: 5% H20
No Solid



Addition (−21° C.)


537
Anti-Solvent (Ethanol)
butan-1-ol: 5% H20
No Solid



Addition Ambient



Temperature


538
Anti-Solvent (Ethanol)
butan-1-ol: 5% H20
No Solid



Addition (4° C.)


539
Anti-Solvent (Ethanol)
butan-1-ol: 5% H20
No Solid



Addition (−21° C.)


540
Temp. Cycling
1,4-dioxane
Form I


541
Evaporation
1,4-dioxane
Form I/II


542
Anti-Solvent Addition
1,4-dioxane
No Solid


543
Temp. Cycling
1-butanol
Form I


544
Evaporation
1-butanol
Form I/II


545
Anti-Solvent (Hexane)
1-butanol
Form III



Addition (4° C.)


546
Temp. Cycling
ethanol
Form I


547
Evaporation
ethanol
Form II


548
Anti-Solvent (Hexane)
ethanol
Form I



Addition (4° C.)


549
Temp. Cycling
acetone
Form I


550
Evaporation
acetone
Form II


551
Anti-Solvent (Hexane)
acetone
Form III



Addition (4° C.)


552
Temp. Cycling
benzonitrile
Form I


553
Evaporation
benzonitrile
Form II


554
Anti-Solvent (Hexane)
benzonitrile
Form II



Addition (4° C.)


555
Temp. Cycling
cyclohexane
Form I


556
Evaporation
cyclohexane
Form II


557
Anti-Solvent (Hexane)
cyclohexane
No Solid



Addition (4° C.)


558
Temp. Cycling
DCM
Form I


559
Evaporation
DCM
Form II


560
Anti-Solvent (Hexane)
DCM
Form III



Addition (4° C.)


561
Temp. Cycling
DMSO
Form I


562
Evaporation
DMSO
Form II/II


563
Anti-Solvent (Hexane)
DMSO
No Solid/No



Addition (4° C.)

Solid


564
Temp. Cycling
EtOAc
Form I


565
Evaporation
EtOAc
Form II


566
Anti-Solvent (Hexane)
EtOAc
Form III



Addition (4° C.)


567
Temp. Cycling
Heptane
Form I


568
Evaporation
Heptane
Form I/II


569
Anti-Solvent (Hexane)
Heptane
No Solid/No



Addition (4° C.)

Solid


570
Temp. Cycling
IPA
Form I


571
Evaporation
IPA
Form I/II


572
Anti-Solvent (Hexane)
IPA
No Solid



Addition (4° C.)


573
Temp. Cycling
IPA: Water (1%)
Form I


574
Evaporation
IPA: Water (1%)
Form II


575
Anti-Solvent (Hexane)
IPA: Water (1%)
No Solid/No



Addition (4° C.)

Solid


576
Temp. Cycling
MeCN
Form I


577
Evaporation
MeCN
Form II


578
Anti-Solvent (Hexane)
MeCN
Form I/III



Addition (4° C.)


579
Temp. Cycling
MeCN: Water (1%)
Form I


580
Evaporation
MeCN: Water (1%)
Form I/II


581
Anti-Solvent (Hexane)
MeCN: Water (1%)
No Solid



Addition (4° C.)


582
Temp. Cycling
MEK
Form I


583
Evaporation
MEK
Form I/II


584
Anti-Solvent (Hexane)
MEK
Form III



Addition (4° C.)


585
Temp. Cycling
MeOAc
Form I


586
Evaporation
MeOAc
Form II


587
Anti-Solvent (Hexane)
MeOAc
Form III



Addition (4° C.)


588
Temp. Cycling
MeOH
Form I


589
Evaporation
MeOH
Form I/II


590
Anti-Solvent (Hexane)
MeOH
Form III



Addition (4° C.)


591
Temp. Cycling
MIBK
Form I


592
Evaporation
MIBK
Form II


593
Anti-Solvent (Hexane)
MIBK
No Solid



Addition (4° C.)


594
Temp. Cycling
Nitromethane
Form I


595
Evaporation
Nitromethane
Form II


596
Anti-Solvent (Hexane)
Nitromethane
Form I



Addition (4° C.)


597
Temp. Cycling
TBME
Form I


598
Evaporation
TBME
Form II


599
Anti-Solvent (Hexane)
TBME
Form I



Addition (4° C.)


600
Temp. Cycling
THF
Form I


601
Evaporation
THF
Form II


602
Anti-Solvent (Hexane)
THF
Form I/III



Addition (4° C.)


603
Temp. Cycling
THF: water (1%)
Form I


604
Evaporation
THF: water (1%)
Form I/II


605
Anti-Solvent (Hexane)
THF: water (1%)
No Solid



Addition (4° C.)


606
Temp. Cycling
toluene
Form I


607
Evaporation
toluene
Form II


608
Anti-Solvent (Hexane)
toluene
Form III



Addition (4° C.)


609
Temp. Cycling
water
No Solid


610
Evaporation
water
Form I/II


611
Anti-Solvent (Hexane)
water
Form III



Addition (4° C.)









Example 2: Intrinsic Dissolution Studies

The intrinsic dissolution rates for Forms I, II, and III were measured at pH conditions of 1.0, 4.5 and 6.7. The results are reproduced below in TABLE 6. In each case, complete dissolution was achieved in less than 3 minutes. Surprisingly, a pH dependence was observed for Form II; with the intrinsic dissolution rate increasing with the pH. In contrast, Forms I and III appear to dissolve at rates independent of pH.









TABLE 6







Calculated Intrinsic Dissolution Rates (mg/cm2/s)











1.0
4.5
6.7
















Form I
0.41
0.44
0.37



Form II
0.26
0.34
0.62



Form III
0.49
0.44
0.45










Example 3: Solubility Studies

The solubility of L-ornithine phenyl acetate was approximated according to methods disclosed above. 24 solvents systems were tested: 1,4 dioxane, 1-butanol, ethanol, acetone, benzonitrile, cyclohexane, DCM, DMSO, EtOAc, Heptane, IPA, IPA (1% H2O), MeCN, MeCn (1% H2O), MEK, MeOAc, methanol, MIBK, Nitromethane, THF, THF (1% H2O), Toluene and water. L-ornithine phenyl acetate exhibited a solubility in water, whereas L-ornithine phenyl acetate was substantially insoluble in the remaining solvent systems.


Slurries of L-ornithine phenyl acetate in water were also prepared and the slurry was filtered. The filtrate concentration was analyzed by HPLC, and the results show the solubility of L-ornithine phenyl acetate to be about 1.072 mg/mL.


HPLC determinations of solubility were also completed for five solvents: ethanol, acetone, methanol, DMSO and IPA. These results are summarized in TABLE 7.









TABLE 7







HPLC Solubility Determinations











Solubility




Solvent
(mg/mL)
Peak Area
Comments













EtOH
<0.0033
N/A
Small peak


Acetone
0
0
API content beyond the lower limit





of quantification (LLOQ)


MeOH
0.0033
1906.75 
Resolved peak


DMSO
>0.0033
N/A
Shoulder on DMSO peak


IPA
0
0
API content beyond the LLOQ









These results indicate that both acetone and IPA are suitable as anti-solvents for precipitating L-ornithine phenyl acetate. In contrast, solvents with measurable solubility are less favorable for precipitating crystalline forms of L-ornithine phenyl acetate.


Finally, the solubility of L-ornithine phenyl acetate was determined in various mixtures of IPA and water using HPLC. The results are shown in TABLE 8.









TABLE 8







HPLC Solubility Determinations (IPA/Water)









% IPA
Peak Area
Solubility (mg/mL)












100
0
0


90
295
0.0054


80
2634
0.0455


70
8340
0.1433









Example 4: Small-Scale Batch Process to Produce L-Ornithine Phenyl Acetate

About 8.4 g (0.049 moles) of L-ornithine HCl was dissolved in 42 mL H2O and, separately, about 11.4 g of silver benzoate was dissolved in 57 mL DMSO. Subsequently, the silver benzoate solution was added to the L-ornithine HCl solution. Combining the two mixtures resulted in an immediate, exothermic precipitation of a creamy white solid (AgCl). The solid was removed by vacuum filtration and retaining the filtrate (L-ornithine benzoate in solution). 200 mL of IPA was added to the filtrate and the mixture was cooled to 4° C. A crystalline solid precipitated after about 3 hours (L-ornithine benzoate) which was isolated by vacuum filtration. Yield: 60%


7.6 g (0.03 moles) of the L-ornithine benzoate was dissolved in 38 mL H2O and about 4.4 g of sodium phenyl acetate was dissolved 22 mL H2O. Subsequently, the sodium phenyl acetate solution was added to the L-ornithine benzoate solution and left to stir for about 10 minutes About 240 mL of IPA (8:2 IPA:H2O) was added and the solution stirred for 30 minutes before cooling to 4° C. A crystalline solid precipitated after about 3 hrs at 4° C. (L-ornithine phenyl acetate). The precipitate was isolated by vacuum filtration and washed with 48-144 mL of IPA. Yield: 57%


Example 5: Large-Scale Batch Process to Produce L-Ornithine Phenyl Acetate

Two separate batch of L-ornithine phenyl acetate were prepared as follows:


About 75 Kg of L-Ornithine monohydrochloride was dissolved in 227 kg of water. To the resulting solution was added 102 Kg of silver benzoate dissolved in 266 kg of DMSO at room temperature within 2 hours. Initially, a strong exothermy was observed and the silver chloride precipitated. The receiver containing the solution was then washed with 14 Kg of DMSO that was added to the reaction mass. In order to remove the silver chloride formed, the reaction mass was filtered over a lens filter prepared with 10 kg of Celite and a GAF filter of 1 mm. After filtration, the filter was washed with an additional 75 kg of water. The reaction mass was then heated at 35±2° C. and 80 kg of sodium phenyl acetate was added. At this point the reaction mass was stirred at 35±2° C. for at least 30 minutes.


In order to precipitate the final API, 353 kg of isopropyl alcohol was added to the reaction mass. The reaction mass was then cooled to 0±3° C. within 6 hours, stirred for 1 hour and then the product isolated in a centrifuge.


About 86 kg of finished wet produce was obtained. The product was then dried at 40±5° C. for about 6.5 to 8 hours to provide about 75 kg of L-ornithine phenyl acetate. Yield: 63.25. TABLE 9 summarizes measurements relating to the final product.









TABLE 9







Analytical Results for Large-scale Batch Process











Test
Batch 1
Batch 2







Purity
98.80% 
98.74% 



Benzoate
0.17%
0.14%



Silver
28 ppm
157 ppm



Chloride
 0.006%
 0.005%



Sodium
 7 ppm
 26 ppm



Total Impurities
0.17%
0.14%



Physical Form
Form II
Form II










Example 6: Reducing Silver Content in L-Ornithine Phenyl Acetate

Batch 2 from Example 5 exhibited high amounts of silver (157 ppm), and therefore procedures were tested for reducing the silver content. Nine trials were completed; each generally including dissolving about 20 g of L-ornithine phenyl acetate from Batch 2 into 1.9 parts water, and then subsequently adding 10.8 parts IPA. A crystalline form was isolated at 0° C. by filtration.


For four trials, 8.0 mg or 80 mg of heavy metal scavengers SMOPEX 102 or SMOPEX 112 were added to the aqueous solution and stirred for 2 hours. The scavengers failed to reduce the silver content below 126 ppm. Meanwhile, another trial applied the general conditions disclosed above and reduced the silver content to 179 ppm. In still another trial, the L-ornithine phenyl acetate was slurried in a solution of IPA, rather than crystallized; however this trial also failed to reduce the silver content below 144 ppm.


The last three trials included adding diluted HCl to the solution to precipitate remaining amount of silver as AgCl. The precipitate was then removed by filtration before The three trials included adding: (1) 1.0 g of 0.33% HCl at 20° C.; (2) 1.0 g of 0.33% HCl at 30° C.; and (3) 0.1 g of 3.3% HCl at 20° C. The three trials reduced the silver content to 30 ppm, 42 ppm, and 33 ppm, respectively, and each trial yielding greater than 90% L-ornithine phenyl acetate. Accordingly, the addition of HCl was effective in reducing the amount of residual silver.


Example 6: Process for Preparing L-Ornithine Phenyl Acetate without an Intermediate Salt

As a general procedure, L-ornithine hydrochloride was suspended in a solvent. After that the reaction mass was heated and a base, sodium methoxide, was added. NaCl formed and was removed from the system by filtration. The reaction mass was cooled and a molar equivalent of phenyl acetic acid was added to the reaction mass in order to form L-ornithine phenyl acetate. The final product was isolated, washed and dried. A summary of the trial for this process is provided in TABLE 10.









TABLE 10







Process Trials










Trial
Base
Eq. of Base
Solvent





1
NaOMe 21% in MeOH
1.0 eq.
MeOH


2
NaOMe 21% in MeOH
0.95 eq. 
IPA


3
NaOMe 21% in EtOH
1.0 eq.
EtOH


4
NaOMe 21% in MeOH
1.0 eq.
MeOH


5
NaOMe 21% in MeOH
1.0 eq.
MeOH w/IPA





for precipitation


6
NaOMe 21% in MeOH
1.0 eq.
Acetonitrile


7
NaOMe 21% in MeOH
1.0 eq.
Water/IPA


8
NaOMe 21% in MeOH
1.0 eq.
Water/IPA


9
NaOMe 21% in MeOH
1.0 eq.
n-butanol









The resulting L-ornithine phenyl acetate was found to exhibit high amounts of chloride (at least about 1% by weight), and presumably include similar amounts of sodium. The yields were about 50% for Trials 2, 4, and 5.


Example 7: Thermal Stability Studies of Forms I, II, and III

Samples of Forms I, II and III were stored at increased temperatures and designated conditions as outlined in TABLE 11. The vacuum applied 600 psi to achieve the reduced pressure. The final compositions were tested by XRPD, NMR, IR and HPLC to determine any changes to the material.


Most notably, Form III does not transition to Form II under vacuum at 120° C., but rather exhibits greater chemical degradation compared to Forms I and II under these conditions. Meanwhile, Form III converts to Form II and exhibits substantial chemical degradation at 120° C. without a vacuum.


Form I converted to Form II in all the trials, but most interestingly, Form I exhibits substantial chemical degradation at 120° C. without a vacuum. Thus, the conversion from Form I does not exhibit the same chemical stability as Form II, which is surprising considering the material readily converts to Form II.


Form II was stable and did not chemically degrade in all of the trials. Thus, Form II is the most stable. Meanwhile, Form III is more stable than Form I, but both forms exhibit substantial chemical degradation at 120° C. without a vacuum.









TABLE 11







Thermal Stability Trials













Initial






Trial
Form
Temperature
Condition
Period
Results

















1
Form I
80°
C.
no
7
days
Form II, no






vacuum


degradation


2
Form I
80°
C.
vacuum
7
days
Form II, no









degradation


3
Form I
80°
C.
no
14
days
Form II, no






vacuum


degradation


4
Form I
80°
C.
vacuum
14
days
Form II, no









degradation


5
Form II
80°
C.
no
7
days
Form II, no






vacuum


degradation


6
Form II
80°
C.
vacuum
7
days
Form II, no









degradation


7
Form II
80°
C.
no
14
days
Form II, no






vacuum


degradation


8
Form II
80°
C.
vacuum
14
days
Form II, no









degradation


5
Form III
80°
C.
no
7
days
Form III, no






vacuum


degradation


5
Form III
80°
C.
no
14
days
Form III, no






vacuum


degradation


6
Form I
120°
C.
no
7
days
Form II






vacuum


(>96% API)


7
Form I
120°
C.
vacuum
7
days
Form II









(>99.9% API)


8
Form I
120°
C.
no
14
days
Form II






vacuum


(37% API)


9
Form I
120°
C.
vacuum
14
days
Form II









(>96% API)


8
Form II
120°
C.
no
7
days
Form II






vacuum


(98.6% API)


9
Form II
120°
C.
vacuum
7
days
Form II









(98.7% API)


10
Form II
120°
C.
no
14
days
Form II






vacuum


(>95% API)


11
Form II
120°
C.
vacuum
14
days
Form II









(>95% API)


10
Form III
120°
C.
no
7
days
Form II






vacuum


(<30% API)


11
Form III
120°
C.
vacuum
7
days
Form III









(>95% API)


12
Form III
120°
C.
no
14
days
Form II






vacuum


(<30% API)


14
Form III
120°
C.
vacuum
14
days
Form III









(88.8% API)









HPLC results for the trials exhibiting chemical degradation (e.g., Trial 10 from TABLE 11) are summarized in TABLE 12. Each degraded material exhibits common peaks at relative retention times (RRT) of 1.9, 2.2, 2.4, and 2.7, which suggests a common degradation pathway for different forms.









TABLE 12







HPLC Results for Degraded Samples










Main Peak Retention Time (min)
Degradation/Impuirty Peak(s)















HPLC

Form

Timepoint
Retention

Retention



ID
Sample ID
Tested
Stability Test
(day)
Time (min)
% Peak Area
Time (min)
% Peak Area


















39
W00045/45/3
III
120° C. ambient pressure
7
2.857
35.786
6.763
6.103









7.582
45.161


42
W00045/45/6
III
120° C. under vacuum (ca. 600 psi)
7
2.787
88.885
7.598
9.389


51
W00045/45/1
I
120° C. ambient pressure
14
3.499
37.826
6.766
3.948









7.569
42.525









9.707
3.628


53
W00045/45/3
III
120° C. ambient pressure
14
3.476
30.394
6.763
5.975









7.583
56.459


56
W00045/45/6
III
120° C. under vacuum (ca. 600 psi)
14
3.400
87.389
7.555
11.500









Example 8: Oxygen Stability Studies of Forms I, II, and III

Samples of Forms I, II and III were stored in 100% oxygen environments for 7 or 14 days and analyzed by NMR and IR. The results establish that Forms I and II show no signs of degradation after 14 days. Only IR results were completed for Form III at 7 days, and these results confirm there was no significant degradation. TLC results for all samples indicated a single spot with similar Rf values.


Example 9: UV Stability Studies of Forms I, II, and III

Samples of Forms I, II and III were exposed to ultraviolet (UV) radiation for 7 or 14 days. A CAMAG universal UV Lampe applied radiation to the samples with setting of 254 mμ. NMR and IR results show no degradation of Forms I and II after 14 days. Similarly, Form III exhibits no degradation after 7 days as determined by NMR and IR. TLC results for all samples indicated a single spot with similar Rf values.


Example 10: pH Stability Studies of Forms I, II, and III

A slurry of Forms I, II and III were formed with water and the pH value adjusted to either 1.0, 4.0, 7.0, 10.0, and 13.2. The slurries were stored for 7 or 14 days, and subsequently the solids were removed by filtration. Form I converted to Form II in all of the samples. NMR and IR results show Forms I and II did not degrade after 14 days in the varied pHs, and similarly HPLC results show about 98% purity or more for these samples. Form III also exhibited no degradation after 7 days according to NMR and IR results. HPLC tests show about 95% purity or more; however IR results show Form III converted to Form II over the 7-day test. TLC results for all samples indicated a single spot with similar Rf values.


Example 11: Compression Studies of Forms I, II, and III

Samples of Forms I, II and III were subjected to 3 tons of force using a Moore Hydraulic Press for about 90 minutes. The resultant tablet's mass, diameter and thickness were measured to determine the density. The tablets were also analyzed by NMR and IR. Form I transitioned to a composition of Form II with a density of 1.197 kg/m3. Form II did not exhibit a transition and had a final density of 1.001 kg/m3. Finally, Form III did not exhibit a transition and had a final density of 1.078 kg/m3.


Example 12: Process for Producing L-Ornithine Phenyl Acetate Via an Acetate Intermediate

Dissolve 25 mg of L-ornithine HCl 5 vols of H2O, and then add excess acetic acid (about 5 vols) to form a slurry. Subject the slurry to temperature cycling between 25° and 40° C. every 4 hours for about 3 days. Add 1 equivalent of phenylacetic acid (with respect to L-ornithine) and stir for about 4-6 hrs (possibly heat). Use IPA as an anti-solvent, add enough to obtain a ratio of 70:30 (IPA:H2O). Isolate by vacuum filtration and dry for about 4-8 hrs at 80° C. to remove any residual acetic acid.

Claims
  • 1. A process for preparing L-ornithine phenylacetate, comprising intermixing phenylacetic acid or a salt thereof with an L-ornithine acetate solution to form a reaction mixture; andisolating a composition comprising a crystalline form of L-ornithine phenylacetate from the reaction mixture.
  • 2. The process of claim 1, wherein the L-ornithine acetate solution is prepared by intermixing an L-ornithine salt with acetic acid in a first solvent.
  • 3. The process of claim 2, wherein the L-ornithine salt is L-ornithine halide salt.
  • 4. The process of claim 3, wherein the L-ornithine salt is L-ornithine hydrochloride.
  • 5. The process of claim 2, wherein the molar ratio of the L-ornithine salt to acetic acid is at least 1:2.
  • 6. The process of claim 2, wherein the first solvent comprises water.
  • 7. The process of claim 1, further comprising adding a second solvent to the reaction mixture before isolating the crystalline form of L-ornithine phenylacetate.
  • 8. The process of claim 7, wherein the second solvent comprises isopropyl alcohol (IPA).
  • 9. The process of claim 1, further comprising recrystallizing the isolated crystalline form of L-ornithine phenylacetate.
  • 10. The process of claim 1, wherein the crystalline form of L-ornithine phenylacetate exhibits an X-ray powder diffraction pattern comprising at least three characteristic peaks, wherein said characteristic peaks are selected from the group consisting of peaks at approximately 6.0°, 13.9°, 14.8°, 17.1°, 17.8° and 24.1° 2θ.
  • 11. The process of claim 10, wherein the crystalline form of L-ornithine phenylacetate exhibits an X-ray powder diffraction pattern comprising characteristic peaks selected from the group consisting of peaks at approximately 6.0°, 13.9°, 14.8°, 17.1°, 17.8° and 24.1° 2θ.
  • 12. The process of claim 1, wherein the crystalline form of L-ornithine phenylacetate exhibits an X-ray powder diffraction pattern comprising at least three characteristic peaks, wherein said characteristic peaks are selected from the group consisting of peaks at approximately 4.9°, 13.2°, 17.4°, 20.8° and 24.4° 2θ.
  • 13. The process of claim 12, wherein the crystalline form of L-ornithine phenylacetate exhibits an X-ray powder diffraction pattern comprising characteristic peaks selected from the group consisting of peaks at approximately 4.9°, 13.2°, 17.4°, 20.8° and 24.4° 2θ.
  • 14. The process of claim 1, wherein the crystalline form of L-ornithine phenylacetate exhibits an X-ray powder diffraction pattern comprising at least three characteristic peaks, wherein said characteristic peaks are selected from the group consisting of peaks at approximately 5.8°, 14.1°, 18.6°, 19.4°, 22.3° and 24.8° 2θ.
  • 15. The process of claim 14, wherein the crystalline form of L-ornithine phenylacetate exhibits an X-ray powder diffraction pattern comprising characteristic peaks selected from the group consisting of peaks at approximately 5.8°, 14.1°, 18.6°, 19.4°, 22.3° and 24.8° 2θ.
INCORPORATION BY REFERENCE TO ANY PRIORITY APPLICATIONS

This application is a continuation of U.S. application Ser. No. 16/037,655, filed Jul. 17, 2018 and to be issued as U.S. Pat. No. 10,550,069; which is a continuation of U.S. application Ser. No. 15/469,359, filed Mar. 24, 2017, now U.S. Pat. No. 10,173,964; which is a divisional of U.S. application Ser. No. 14/715,481, filed May 18, 2015, now U.S. Pat. No. 9,604,909; which is a divisional of U.S. application Ser. No. 14/299,940, filed Jun. 9, 2014, now U.S. Pat. No. 9,034,925; which is a divisional of U.S. application Ser. No. 13/937,107, filed Jul. 8, 2013, now U.S. Pat. No. 8,785,498; which is a divisional of U.S. application Ser. No. 13/436,642, filed Mar. 30, 2012, now U.S. Pat. No. 8,492,439; which is a divisional of U.S. application Ser. No. 12/753,763, filed Apr. 2, 2010, now U.S. Pat. No. 8,173,706; which claims the benefit of priority of U.S. Provisional Application No. 61/166,676, filed Apr. 3, 2009. The priority documents are hereby incorporated by reference in their entireties. Any and all applications for which a foreign or domestic priority claim is identified in the Application Data Sheet as filed with the present application, or any correction thereto, are hereby incorporated by reference under 37 CFR 1.57.

US Referenced Citations (56)
Number Name Date Kind
3950529 Fischer et al. Apr 1976 A
4100293 Walser Jul 1978 A
4228099 Walser Oct 1980 A
4284647 Brusilow et al. Aug 1981 A
4320146 Walser Mar 1982 A
4352814 Walser Oct 1982 A
4457942 Brusilow et al. Jul 1984 A
5139981 Kurland Aug 1992 A
5405761 Makryaleas et al. Apr 1995 A
5571783 Montagne et al. Nov 1996 A
5591613 Makryaleas et al. Jan 1997 A
5741524 Staniforth et al. Apr 1998 A
5767086 Kauvar et al. Jun 1998 A
6083953 Nestor et al. Jul 2000 A
6258849 Burzynski Jul 2001 B1
6451340 Arimilli et al. Sep 2002 B1
6503530 Kang et al. Jan 2003 B1
6514953 Armitage et al. Feb 2003 B1
6768024 Watson-Straughan et al. Jul 2004 B1
6943192 Burzynski Sep 2005 B2
8173706 Anderson et al. May 2012 B2
8389576 Jalan et al. Mar 2013 B2
8492439 Anderson et al. Jul 2013 B2
8785498 Anderson et al. Jul 2014 B2
8946473 Anderson et al. Feb 2015 B2
9034925 Anderson et al. May 2015 B2
9260379 Anderson et al. Feb 2016 B2
9566257 Jalan et al. Feb 2017 B2
9604909 Anderson et al. Mar 2017 B2
10039735 Jalan et al. Aug 2018 B2
10173964 Anderson et al. Jan 2019 B2
10525029 Jalan et al. Jan 2020 B2
10550069 Anderson et al. Feb 2020 B2
20030105104 Burzynski Jun 2003 A1
20030195255 Summar Oct 2003 A1
20040152784 March Aug 2004 A1
20040229948 Summar et al. Nov 2004 A1
20050059150 Guarino et al. Mar 2005 A1
20050182064 Burzynski Aug 2005 A1
20060045912 Truog Mar 2006 A1
20080119554 Jalan et al. May 2008 A1
20100280119 Anderson et al. Nov 2010 A1
20120157526 Jalan et al. Jun 2012 A1
20120208885 Anderson et al. Aug 2012 A1
20120259016 Jalan et al. Oct 2012 A1
20130211135 Anderson et al. Aug 2013 A1
20130296429 Anderson et al. Nov 2013 A1
20140142186 Scharschmidt et al. May 2014 A1
20140288327 Anderson et al. Sep 2014 A1
20150133684 Anderson et al. May 2015 A1
20150251990 Anderson et al. Sep 2015 A1
20160338982 Ruettimann et al. Nov 2016 A1
20170189364 Jalan et al. Jul 2017 A1
20180161293 Jalan et al. Jun 2018 A1
20180221320 Rose et al. Aug 2018 A1
20190070142 Jalan et al. Mar 2019 A1
Foreign Referenced Citations (35)
Number Date Country
2014250643 Nov 2014 AU
2015221466 Sep 2015 AU
2763894 Jan 2011 CA
1383815 Dec 2002 CN
101010087 Aug 2007 CN
1179347 Feb 2002 EP
1334722 Aug 2003 EP
1374863 Jan 2004 EP
1541141 Jun 2005 EP
2113774 Jun 1972 FR
965637 Aug 1964 GB
1067742 May 1967 GB
1080599 Aug 1967 GB
1310658 Mar 1973 GB
1507951 Apr 1978 GB
H05-221858 Aug 1993 JP
3273578 Apr 2002 JP
2011-236160 Nov 2011 JP
S54-163518 Dec 2011 JP
PA03009902 May 2005 MX
WO 198504805 Nov 1985 WO
WO 198705297 Sep 1987 WO
WO 199730167 Aug 1997 WO
WO 2000071151 Nov 2000 WO
WO 2002034255 May 2002 WO
WO 2002074302 Sep 2002 WO
WO 2003037378 May 2003 WO
WO 2003045372 Jun 2003 WO
WO 2003086074 Oct 2003 WO
WO 2004019928 Mar 2004 WO
WO 2005053607 Jun 2005 WO
WO 2005082023 Sep 2005 WO
WO 2006056794 Jun 2006 WO
WO 2006059237 Jun 2006 WO
WO 2014081977 May 2014 WO
Non-Patent Literature Citations (227)
Entry
Abraldes et al., “Hemodynamic Response to Pharmacological Treatment of Portal Hypertension and Long-Term Prognosis of Cirrhosis”, Hepatol. 2003, 37:902-908.
Aggarwal et al., “Predictors of Mortality and Resource Utilization in Cirrhotic Patients Admitted to the Medical ICU”, Chest, 2001, vol. 119, Issue 5, pp. 1489-1497.
Albrecht et al., “Contrasting effects of thioacetamide-induced liver damage on the brain uptake indices of ornithine, arginine and lysine: modulation by treatment with ornithine aspartate”, Metab Brain Dis., 1996, vol. 11, Issue 3, pp. 229-237.
Albrecht et al., “Increase of the brain uptake index for L-ornithine in rats with hepatic encephalopathy”, Neuroreport., 1994, vol. 5, Issue 6, pp. 671-673.
Al-Hassnan et al., “The relationship of plasma glutamine to ammonium and of glycine to acid-base balance in propionic acidaemia”, J. Inherit. Metab. Dis., 2003, vol. 26, pp. 89-91.
Al Sibae et al., “Current Trends in the Treatment of Hepatic Encephalopathy”, Ther Clin Risk Manag. Jun. 2009, 5(3): 617-626.
Als-Nielsen et al.,, Non-Absorbable Disaccharides for Hepatic Encephalopathy: Systematic Review of Randomised Trials, BMJ, 2004, p. 1-6.
Anadiotis et al., “Ornithine transcarbamylase deficiency and pancreatitis”, J Pediatr, 2001, vol. 138, pp. 123-124.
Anonymous,, “Sodium phenylbutyrate for urea cycle enzyme deficiencies.” [No authors listed], Med Lett Drugs Ther., Nov. 22, 1996, vol. 38, Issue 988, pp. 105-106.
Australian Examination Report dated Jun. 11, 2014 from Application No. 2010232521, filed Apr. 1, 2010.
Australian Examination Report dated Feb. 9, 2018 from Application No. 2017202081, filed Mar. 29, 2017.
Bachmann et al., “Ammonia toxicity to the brain and creatine”, Molecular Genetics and Metabolism, 2004, vol. 81, pp. S52-S57.
Balata et al., “Induced hyperammonemia alters neuropsychology, brain MR spectroscopy and magnetization transfer in cirrhosis,”, Hepatology, 2003, vol. 4, Issue 37, pp. 931-939.
Batshaw et al., “Alternative pathway therapy for urea cycle disorders: twenty years later”, J Pediatr. 2001, 138(1 Suppl): S46-S55.
Batshaw et al., “Effect of sodium benzoate and sodium phenylacetate on brain serotonin turnover in the Ornithine transcarbamylase-deficient sparse-fur mouse”, Pediatric Research, 1988, vol. 23, Issue 4, pp. 368-374.
Beale et al., “Early enteral supplementation with key pharmaconutrients improves sequential organ failure assessment score in critically ill patients with sepsis: outcome of a randomized, controlled, double blind trial,”, Crit Care Med., 2008, vol. 1, Issue 36, pp. 131-144.
Berg et al., “Pharmacokinetics and cerebrospinal fluid penetration of phenylacetate and phenylbutyrate in the non-human primate”, Cancer Chemother Pharmacol. May 2001, 47(5): 385-390. Abstract Only.
Berge et al., “Pharmaceutical Salts”, J Pharm Sci, 1977, vol. 66, pp. 1-19.
Berry et al., “Long-term management of patients with urea cycle disorders”, J Pediatri, 2001, vol. 138, Issue 1, pp. S56-S61.
Bighley et al., “Salt Forms of Drugs and Absorption” in Encyclopedia of Pharmaceutical Technology, Marcel Dekker, Inc. New York, 1996, pp. 453-499.
Blei et al., Pathophysiology of Cerebral Edema in Fulminant Hepatic Failure, Journal of Hepatology, 1999, p. 771-776, vol. 31, Denmark.
Bleichner, et al., “Frequency of infections in cirrhotic patients presenting with acute gastrointestinal haemorrhage”, British Journal of Surgery, 1986, vol. 73, Issue 9, pp. 724-726.
Bongers et al., “Exogenous glutamine: the clinical evidence,”, Crit Care Med., 2007, vol. 9 Suppl, Issue 35, pp. S545-S552.
Bosoi et al., Long term oral treatment of ornithine phenylacetate increases lean mass and attenuates brain edema in bile-duct ligated rats. Hepatology Oct. 2015, 62(Suppl 1):953A; Abstract 1523.
Bosoi et al., “Oral Ornithine Phenylacetate Attenuates Muscle Mass Loss and Prevents Hepatic Encephalopathy in BDL Rats”, Abstract 23; J Clin Exper Hepatol. (Feb. 2017) 7:S18-S19.
Braga et al., “Crystal Polymorphism and Multiple Crystal Forms”, Struct Bond, 2009, 132: 25-50 [pub online Feb. 25, 2009].
Briggs et al., “Effect of Ornithine and Lactate on Urea Synthesis in Isolated Hepatocytes”, Biochem J, 1976, vol. 160, pp. 205-209.
Bruha et al., “Effect of carvedilol on portal hypertension depends on the degree of endothelial activation and inflammatory changes”, Scand J Gastroenter. 2006, 41: 1454-1463.
Brunquell et al., “Electroencephalographic findings in ornithine transcarbamylase deficiency”, J Child Neurol, 1999, vol. 14, Issue 8, pp. 533-536.
Brusilow et al., “Amino acid acylation: A mechanism of nitrogen excretion in inborn errors of urea synthesis”, Science, 1980, vol. 207, pp. 659-661.
Brusilow et al., “Treatment of episodic hyperammonemia in children with inborn errors of urea synthesis”, The New England Journal of Medicine, 1984, vol. 310, Issue 25, pp. 1630-1634.
Burlina et al., “Long-term treatment with sodium phenylbutyrate in ornithine transcarbamylase-deficient patients”, Molecular Genetics and Metabolism, 2001, vol. 72, pp. 351-355.
Butterworth, “Pathophysiology of hepatic encephalopathy: a new look at ammonia”, Metab Brain Dis., 2002, vol. 17, Issue 4, pp. 221-227.
Butterworth, “Neuronal cell death in hepatic encephalopathy”, Metab Brain Dis. Dec. 2007, 22(3-4): 309-320.
Byrn et al., “Pharmaceutical Solids: A Strategic Approach to Regulatory Considerations”, Pharma Res. 1995, 12(7): 945-954.
Caira, M.R., “Crystalline Polymorphism of Organic Compounds”, Topic in Current Chemistry 1998, 198: 163-208.
Callado França, et al., Five Days of Ceftriaxone to Treat Spontaneous Bacterial Peritonitis in Cirrhotic Patients, Journal of Gastroenterology, Feb. 2002, p. 119-122 vol. 37, No. 2, Springer, Japan.
Canbay et al. “L-Ornithine L-Aspartate (LOLA) as a Novel Approach for Therapy of Non-alcoholic Fatty Liver Disease”, Drugs 2019, vol. 79(Suppl 1), pp. S39-S44 (first published online Jan. 31, 2019).
Cavarec et al., “Molecular cloning and characterization of a transcription factor for the copia retrotransposon with homology to the BTB-Containing Lola Neurogenic Factor”, Mol. Cell. Biol., 1997, vol. 17, Issue 1, pp. 482-494.
Chainuvati et al., “Ornicetil on encephalopathy. Effect of ornicetil (ornithine alpha-ketoglutarate) on encephalopathy in patients with acute and chronic liver disease”, Acta Hepatogastro., 1977, vol. 24, Issue 6, pp. 434-439.
Chawla et al., “Challenges in Polymorphism of Pharmaeuticals”, CRIPS Mar. 2004, 5(1): 9-12.
Chen et al., “Continuous arteriovenous hemodiafiltration in the acute treatment of hyperammonaemia due to ornithine transcarbamylase deficiency”, Renal Failure, 2000, vol. 22, Issue 6, pp. 823-836.
Chinese Office Action dated Feb. 19, 2013 in Application No. 201080021311.6, filed Apr. 1, 2010.
Chinese Office Action dated Sep. 18, 2012 in Application No. 201080021311.6, filed Apr. 1, 2010.
Chinese Office Action dated Sep. 23, 2013 in Application No. 201080021311.6, filed Apr. 1, 2010.
Chinese Office Action dated Jul. 9, 2015 for Application No. 201410392027.X, filed Aug. 11, 2014.
Chinese Office Action dated Apr. 5, 2016 for Application No. 201410392027.X, filed Aug. 11, 2014.
Chinese Office Action dated Feb. 12, 2018 for Application No. 201710053850.1, filed Jan. 22, 2017.
Chung et al., “Cirrhosis and its Complications”, Harrison's Principles of Internal Medicine (16th Edition) (2005) 289, pp. 1858-1869.
Clément et al., “Minimal hepatic encephalopathy leads to hypotension-induced neuronal cell loss in BDL rats”, Abstract 51; Hepatology (Oct. 2015) 62(Suppl 1):233A-234A.
Clemmesen, et al., Cerebral Herniation in Patients With Acute Liver Failure is Correlated with Arterial Ammonia Concentration, Hepatology, Mar. 1999, p. 648-653, Vo. 29, No. 3, American Association for the Study of Liver Diseases.
ClinicalTrails.gov; William Lee, Med. Uni. S.C.; “Safety Study of Ornithine Phenylacetate to Treat Patients with Acute Liver Failure (STOP-ALF)”, ID #NCT01548690; Feb. 2012; 7 pages.
Darmaun et al., “Phenylbutyrate-induced glutamine depletion in humans; effect on leucine metabolism”, Am J Physiol Endocrinol Metab., 1998, vol. 274, pp. E801-E807.
Database WPI, Section Ch, Week 200331, Derwent Publications Ltd., London, GB; XP002364873 & CN 1383815 A (LIU W), Dec. 11, 2002 (Abstract Only).
Davies, et al., “L-ornithine and phenylacetate synergistically produce sustained reduction in ammonia and brain water in cirrhotic rats”, Hepatology Jul. 2009, 50(1): 155-164.
Dejong et al., “Altered glutamine metabolism in rat portal drained viscera and hindquarter during hyperammonemia”, Gastroenterology, 1992, vol. 103, Issue 3, pp. 936-948.
Del Rosario et al., Hyperammonemic encephalopathy, J Clin Gastroenterol, 1997, vol. 25, Issue 4, pp. 682-684.
Demand with Article 34 amendments, filed Feb. 3, 2011 in International Application No. PCT/US2010/029708.
Desjardins et al., “Effect of portacaval anastomosis on glutamine synthetase protein and gene expression in brain, liver and skeletal muscle”, Metab Brain Dis., 1999, vol. 14, Issue 4, pp. 273-280.
Dewhirst et al., “Phylogeny of the defined murine microbiota: Altered Schaedler Flora”, Appl. Environ Microbiol. 1999, 65(8): 3287-3292.
Dunitz et al., “Disappearing Polymorphs”, Acc Chem Res. 1995, 28: 193-200.
Efrati et al., “Effect of sodium benzoate on blood ammonia response to oral glutamine challenge in cirrhotic patients: a note of caution”, Am J Gastroenterol. (2000) 95(12):3574-3578. (Abstract).
Enns et al., “Survival after treatment with phenylacetate and benzoate for urea-cycle disorders,”, N Engl J Med., 2007, vol. 22, Issue 356, pp. 2282-2292.
Eurasian Office Action dated May 24, 2016 for Application No. 201500650 filed Jul. 16, 2015.
Eurasian Office Action dated Sep. 27, 2017 for Application No. 201500650 filed Jul. 16, 2015.
European Extended Search Report dated Nov. 30, 2012 for Application No. 10759442.6.
European Extended Search Report dated Nov. 9, 2017 for Application No. 17185173.6.
Fabbri et al., Unresponsiveness of Hepatic Nitrogen Metabolism to Glucagon Infusion in Patients with Cirrhosis: Dependence on Liver Cell Failure, Hepatology 1993, 18(1): 28-35.
Feuerstein et al., Cytokines, Inflammation, and Brain Injury: Role of Tumor Necrosis Factor-α. Cerebrovasc Brain Metab Rev. 1994, 6(4):341-360.
Garcia-Tsao, MD, et al., Management and Treatment of Patients with Cirrhosis and Portal Hypertension: Recommendations from the Department of Veterans Affairs Hepatitis C Resource Center Program and the National Hepatitis C Program, Am J Gastroenterol, 2009, p. 1802-1829, Vo. 104.
Garden et al., “Prediction of outcome following acute variceal haemorrhage”, Br J Surg., 1985, vol. 72, pp. 91-95.
Gebhardt et al., “Treatment of cirrhotic rats with L-Ornithine-L-Aspartate enhances urea synthesis and lowers serum ammonia levels”, J Pharm Exp Thera., 1997, vol. 283, Issue 1, pp. 1-6.
Gonzalez-Navajas et al., “Bacterial DNA in patients with cirrhosis and sterile ascites. Its role as a marker of bacterial translocation and prognostic tool,”, Rev Esp Enferm Dig., 2007, vol. 10, Issue 99, pp. 599-603.
Gordon, “Ornithine transcarbamylase deficiency: a urea cycle defect”, European Journal of Paediatric Neurology, 2003, vol. 7, pp. 115-121.
Grace et al., “Prevention of initial variceal hemorrhage”, Gastroenter Clin North Am., 1992, vol. 21, Issue 1, pp. 149-161.
Grant, D.J.W., “Theory and Origin of Polymorphism” Chapter 1 from Polymorphism in Pharmaceutical Solids, Brittain, Harry G. [Ed.]; Marcel Dekker, Inc., 1999; pp. 1-11.
Greenstein et al., Studies on the Metabolism of Amino Acids and Related Compounds in Vivo. III. Prevention of Ammonia Toxicity by Arginine and Related Compounds, Arch Biochem Biophys, 1956, vol. 64, Issue (2):, pp. 342-354.
Grossi et al., “Amino acids mixtures in prevention of acute ammonia intoxication in dogs”, Arch Surg, 1967, vol. 94, pp. 261-266.
Guillory, J.K., “Generation of Polymorphs, Hydrates, Solvates, and Amorphous” Chapter 5 from Polymorphism in Pharmaceutical Solids, Brittain, Harry G. [Ed.]; Marcel Dekker, Inc., 1999, pp. 183-226.
Häberle et al., Hyperammonämie: Ursachen, Diagnostik, Therapie, Dtsch Med Wochenschr, 2004, vol. 129; pp. 1430-1433.
Hamberg, Ole et al., Effects of an Increase in Protein Intake on Hepatic Efficacy for Urea Synthesis in Healthy Subjects and in Patients with Cirrhosis, Journal of Hepatology, 1992, pp. 237-243, Elsevier Science Publishers B.V.
Hass et al., “Detection of subclinical and overt hepatic encephalopathy and treatment control after L-Ornithine-L-Aspartate medication by magnetic resonance spectroscopy (1H-MRS)”, Z Gastroenterol, 2005, vol. 43, pp. 373-378.
Häussinger et al., “Hepatic encephalopathy in chronic liver disease: a clinical manifestation of astrocyte swelling and low-grade cerebral edema?”, J Hepatol., 2000, vol. 32, Issue 6, pp. 1035-1038.
Herlong et al., “The use of ornithine salts of branched-chain ketoacids in portal-systemic encephalopathy”, Ann Intern Med., 1980, vol. 93, Issue 4, pp. 545-550.
Hirayama et al., [Eds], “Organic compound crystal produced handbook—Principles and know-how”, Maruzen Co., Ltd., Japan; (Jul. 2008), pp. 17-23, 37-40, 45-51 and 57-65; 31 pages.
Honda et al., “Successful treatment of severe hyperammonemia using sodium phenylacetate powder prepared in hospital pharmacy”, Biol. Pharm. Bull., Sep. 2002, 25(9): 1244-1246.
Hopkins Medicine (http://www.hopkinsmedicine.org/gastroenterology_hepatology/_pdfs/liver/portal_hypertension.pdf; accessed Jun. 22, 2016); 13 pages.
Hursthouse et al., “Why Do Organic Compounds Crystallise Well or Badly or Ever so Slowly? Why is Crystallisation Nevertheless Such a Good Purification Technique?”, Organic Process Research & Development, 2009, 13:1231-1240.
Igarashi et al., “Determination of ornithine conjugates of some carboxylic acids in birds by high-performance liquid chromatography”, Chem Pharm Bull, 1992, vol. 40, Issue 8, pp. 2196-2198.
India Office Action dated Jul. 26, 2017 for Application No. 7982/DELNP/2011, filed Apr. 1, 2010.
Inoue et al., “Biochemical analysis of decreased ornithine transport activity in the liver mitochondria from patients with hyperornithinemia, hyperammonemia and homocitrullinuria”, Biochim Biophys Acta., 1988, vol. 964, Issue 1, pp. 90-95.
International Preliminary Report on Patentability dated Jun. 2, 2011 for Application No. PCT/US2010/029708, filed Apr. 1, 2010.
International Search Report and Written Opinion dated Jun. 3, 2010 for PCT/US2010/029708, filed Apr. 1, 2010.
Israel Office Action dated Sep. 30, 2014 for Application No. 215449, filed Apr. 1, 2010.
Islam et al., “Sorbitol and lactitol reduce body fat and toxic ammonia levels in rats”, Nutrition Res. (2007) 27:440-447.
Iyer et al., “Mouse model for human arginase deficiency”, Mol Cell Biol., 2002, vol. 22, Issue 13, pp. 4491-4498.
Jalan et al., “Acute-on-chronic liver failure: pathophysiological basis of therapeutic options”, Blood Purif, 2002, vol. 20, pp. 252-261.
Jalan et al., “Moderate hypothermia in patients with acute liver failure and uncontrolled intracranial hypertension,”, Gastroenterology, 2004, vol. 5, Issue 127, pp. 1338-1346.
Jalan et al., “The molecular pathogenesis of hepatic encephalopathy”, The International Journal of Biochemistry & Cell Biology, 2003, vol. 35, pp. 1175-1181.
Jalan et al., L-Ornithine Phenylacetate (OP): A Novel Treatment for Hyperammonemia and Hepatic Encephalopathy, Medical Hypotheses, 2007, 69(5): 1064-1069, Elsevier Ltd.
Jalan et al., Treatment of Hyperammonemia in Liver Failure: A Tale of Two Enzymes, Gastroenterology, 2009, p. 2048-2051, vol. 1236.
Jalan,, Intracranial Hypertension in Acute Liver Failure: Pathophysiological Basis of Rational Management, Seminars in Liver Disease, 2003, p. 271-282, vol. 23, No. 3, Thieme Medical Publisheres, Inc., New York, NY, USA.
James et al., “The conjugation of phenylacetic acid in man, sub-human primates and some non-primate species”, Proc R Soc Lond B., 1972, vol. 182, pp. 25-35.
Japanese Office Action dated May 13, 2014 for Application No. 2012-503725, filed Apr. 1, 2010.
Japanese Office Action dated Oct. 21, 2014 for Application No. 2012-503725, filed Apr. 1, 2010.
Japanese Office Action dated Jun. 7, 2016 for Application No. 2012-503725, filed Apr. 1, 2010.
Japanese Office Action dated Sep. 5, 2017 for Application No. 2016-237164, filed Jul. 12, 2016.
Jeyamani et al., Hepatitis E virus and acute-on-chronic liver failure,, Indian J Gastroentero., 2004, vol. 23, Issue 2, pp. 45-46.
Jiang et al., “L-Ornithine-I-aspartate in the management of hepatic encephalopathy: a meta-analysis”, J Gastroenterol Hepatol. 2009, 24(1): 9-14; available online: Sep. 28, 2008.
Jover-Cobos et al., Ornithine phenylacetate revisited; Metabolic Brain Disease 2013, 28(2): 327-331.
Kaiser, S. et al., Ammonia and Glutamine Metabolism in Human Liver Slices: New Aspects on the Pathogenesis of Hyperammonaemia in Chronic Liver Disease, European journal of Clinical Investigation , 1988, vol. 18, pp. 535-542.
Kasumov et al., “New secondary metabolites of phenylbutyrate in humans and rats”, Drug Metab Dispos., 2004, vol. 32, Issue 1, pp. 10-19.
Katayama, “Ammonia metabolism and hepatic encephalopathy”, Hep. Research, 2004, vol. 30, Issue 1, pp. S71-S78.
Khan et al.,, Frequency of Spontaneous Bacterial Peritonitis in Cirrhotic Patients with Ascites Due to Hepatitis C Virus and Efficacy of Ciprofloxacin in its Treatment, Gomal Journal of Medical Sciences, Jul.-Dec. 2009, p. 149-154, vol. 7, No. 2.
Kircheis et al., “Therapeutic efficacy of L-ornithine-L-aspartate infusions in patients with cirrhosis and hepatic encephalopathy: results of a placebo-controlled, double blind study,”, Hepatology, 1997, vol. 6, Issue 25, pp. 1351-1360.
Kojima et al., “Effective Solid Form Selection for the Pharmaceutical Development”, J Pharma Science Tech. Sep. 2008, 68(5): 344-349.
Korean Office Action dated Jun. 8, 2016 for Application No. 10-2011-7026256, filed Apr. 1, 2010.
Korean Office Action dated Mar. 30, 2017 for Application No. 10-2011-7026256, filed Apr. 1, 2010.
Korean Office Action dated Aug. 1, 2017 for Application No. 10-2011-7026256, filed Nov. 3, 2011.
Larsen et al., “Alternative Pathway Therapy for Hyperammonemia in Liver Failure”; Hepatolory, Jul. 2009, 50(1): 3-5.
Lee et al., Acute Liver Failure: Summary of a Workshop, Hepatology, Apr. 2008, p. 1401-1415, vol. 47, No. 4.
Lee, W. M., Acetaminophen-Related Acute Liver Failure in the United States, Hepatology Research, 2008, p. S3-S8, vol. 38, Suppl. 1, The Japan Society of Hepatology.
Lee et al., “Phase 2 Comparison of a Novel Ammonia Scavenging Agent with Sodium Phenylbutyrate in Patients with Urea Cycle Disorders: Safety, Pharmacokinetics and Ammonia Control”, Mol Genet Metab. Mar. 2010, 100(3): 221-228.
Linderoth et al., “Short-term prognosis of community-acquired bacteremia in patients with liver cirrhosis or alcoholism: A population-based cohort study,”, Alcohol Clin Exp Res., 2006, Issue 30, pp. 636-641.
Lopez-Talavera et al., “Thalidomide Inhibits Tumor Necrosis Factor alpha, Decreases Nitric Oxide Synthesis, and Ameliorates the Hyperdynamic Circulatory Syndrome in Portal-Hypertensive Rats”, Hepatology, 1996, 23(6): 1616-1621.
Lukkarinen, M. et al., Effect of Lysine Infusion on Urea Cycle in Lysinuric Protein Intolerance, Metabolism, May 2000, 49(5): 621-625.
Lukkarinen, M. et al., Oral Supplementation Corrects Plasma Lysine Concentrations in Lysinuric Protein Intolerance, Metabolism, Jul. 2003, 52(7): 935-938.
MacArthur et al., “Pharmacokinetics of sodium phenylacetate and sodium benzoate following intravenous administration as both a bolus and continuous infusion to healthy adult volunteers”, Molecular Genetics and Metabolism, 2004, vol. 81, pp. S67-S73.
Maestri et al., “Long-term treatment of girls with ornithine transcarbamylase deficiency”, N Engl J Med., 1996, vol. 335, Issue 12, pp. 855-859.
Maestri et al., “Prospective treatment of urea cycle disorders”, J Pediatr., 1991, vol. 119, Issue 6, pp. 923-928.
Maev I.V. Application of L-ornitine-L-aspartate in complex therapy of hepatic encephalopathy in liver cirrhosis patients (Engl. Title) koloproktologii, 2002, No. 6, pp. 60-66.
Maier et al., Originalien Activities of Urea-Cycle Enzymes in Chronic Liver Disease, Klinische-Wochenschrift, 1979, vol. 67, pp. 661-665, Springer-Verlag.
Maier, “Therapie der hepatischen Enzephalopathie”, Dtsch med Wschr., 1988, vol. 113, pp. 1886-1889.
Maruzen Co., Ltd., “Jikken Kagaku Guide Book (Experimental Chemistry Guide Book),” The Chemical Society of Japan, 1992, 3rd Edition, pp. 130-131.
Maruzen Co., Ltd., “Jikken Kagaku Koza (Zoku) Experimental Chemistry Course (cont.)”, 2. Bunri to Seisei (Isolation and Purification), Jan. 25, 1967, pp. 159-162 and 184-193.
Matsuoka et al., “Advanced Crystallization Technology of Organic Materials—Control of Size, Morphology, Polymorph and Purity”, Pharm Tech, Japan (May 2003) 19(6): 91(955)-101(965).
Meijer et al., Nitrogen Metabolism and Ornithine Cycle Function, Physiological Reviews, Jul. 1990, vol. 70, No. 3, pp. 701-748, The American Physiological Society.
Mendenhall et al., “A new therapy for portal systemic encephalopathy”, The American Journal of Gastroenterology, 1986, vol. 81, Issue 7, pp. 540-543.
Mexican Office Action dated Dec. 10, 2013 for Application No. MX/a/2012/013961, filed Nov. 30, 2012.
Mexican Office Action dated Oct. 5, 2017 for Application No. MX/a/2014/008993, filed Jul. 24, 2014.
Mihm et al., “Effect of L-ornithine-L-aspartate (LOLA) on neurometabolites in hepatic encephalopathy (HE)”, Hepatology, 2001, vol. 34, Issue 4, pp. 543A.
Mizock et al., “Septic Encephalopathy—Evidence for altered phenylalanine metabolism and comparison with hepatic encephalopathy”, Arch Intern Med, 1990, vol. 150, pp. 443-449.
Mizock, Nutritional Support in Hepatic Encephalopathy, Nutrition, 1999, pp. 220-228, vol. 15, No. 3, Elsevier Science Inc.
Mizutani et al., “Hyperargininemia: Clinical course and treatment with sodium benzoate and phenylacetic acid”, Brain Dev., 1983, vol. 5, Issue 6, pp. 555-563.
Moinard et al.,, “Effects of Ornithine 2-Oxoglutarate on Neutrophils in Stressed Rates: Evidence for the Involvement of Nitric Oxide and Polyamines”, Clin Sci, 2002, vol. 102, Issue 3, pp. 287-295, London, England.
Mookerjee et al., “Neutrophil dysfunction in alcoholic hepatitis superimposed on cirrhosis is reversible and predicts the outcome,”, Hepatology, 2007, vol. 3, Issue 46, pp. 831-840.
Mookerjee et al., “Increased gene and protein expression of the novel eNOS regulatory protein NOSTRIN and a variant in alcoholic hepatitis”, Gastroenterology Jun. 2007, 132(7): 2533-2541.
Mouille et al., “Adaptative increase of ornithine production and decrease of ammonia metabolism in rat colonocytes after hyperproteic diet ingestion”, Am J Gastrointest Liver Physiol., 2004, 287(2), G344-G351.
Nance et al., “Ammonia production in germ-free Eck fistula dogs”, Surgery, 1971, vol. 70, Issue 2, pp. 169-174.
Navasa et al., “Bacterial infections in liver cirrhosis,”, Ital J Gastroenterol Hepatol., 1999, vol. 7, Issue 31, pp. 616-625.
New Zealand Office Action dated Jan. 8, 2014 for Application No. 619235, filed Dec. 20, 2013.
Newsholme et al., “Glutamine metabolism by lymphocytes, macrophages, and neutrophils: its importance in health and disease,”, J Nutr Biochem., 1999, vol. 6, Issue 10, pp. 316-324.
Newsholme, “Why is L-glutamine metabolism important to cells of the immune system in health, postinjury, surgery or infection?”, J Nutr., 2001, vol. 9 Suppl, Issue 131, pp. 2515S-2522S.
Ocera Therapeutics, Inc., News Release: Ocera Completes Interim Analysis of OCR-002 in Phase 2b STOP-HE Study for the Treatment of Acute Hepatic Encephalopathy; Globe Newswire; Apr. 1, 2015, 2 pages.
Ocera Therapeutics, Inc., News Release: Ocera Announces Positive Phase 1 Results for Oral OCR-002 in Development for the Prevention of Acute Hepatic Encephalopathy; Globe Newswire; Nov. 16, 2015, 2 pages.
Ocera Therapeutics, Inc., News Release: Ocera Completes Plasma Data from Pilot Phase 1 Study for Orally-available OCR-002 in Development for the Prevention of Acute Hepatic Encephalopathy; Globe Newswire; Jan. 8, 2016, 3 pages.
Olde Damink et al., “Decreased plasma and tissue isoleucine levels after simulated gastrointestinal bleeding by blood gavages in chronic portacaval shunted rats”, Gut, 1997, vol. 40, pp. 418-424.
Olde Damink et al., “Interorgan ammonia and amino acid metabolism in metabolically stable patients with cirrhosis and a TIPSS”, Hepatology, 2002, vol. 36, Issue 5, pp. 1163-1171.
Olde Damink et al., “Interorgan ammonia metabolism in liver failure”, Neurochemistry International, 2002, vol. 41, pp. 177-188.
Olde Damink et al., “The kidney plays a major role in the hyperammonemia seen after simulated or actual GI bleeding in patients with cirrhosis”, Hepatology, 2003, vol. 37, pp. 1277-1285.
Olde Damink et al., Stimulated Liver and Muscle Protein Synthesis by Intravenous Isoleucine Supplementation During a Simulated Variceal Bleed in Patients with Cirrhosis of the Liver, Hepatology, Oct. 2001, AASLD Abstracts #50.
Pahan et al., Lovastatin and Phenylacetate Inhibit the Induction of Nitric Oxide Synthase and Cytokines in Rat primary Astrocytes, Microglia, and Macrophages, J Clin Invest. BMJ Group GB, 1997, 100(11):2671-2679.
Pauwels et al., “Systemic antibiotic prophylaxis after gastrointestinal hemorrhage in cirrhotic patients with a high risk of infection”, Hepatology, 1996, vol. 24, Issue 4, pp. 802-806.
Petrowski et al., “Pharmacologic amino acid acylation in the acute hyperammonemia of propionic acidemia”, Journal of Neurogenetics, 1987, vol. 4, pp. 87-96.
Plecko et al., “Partial N-acetylglutamate synthetase deficiency in a 13-year-old girl: diagnosis and response to treatment with N-carbamylglutamate”, Eur J Pediatr., 1998, vol. 157, pp. 996-998.
Powell et al., “Compendium of Excipients for Parenteral Formulations”, PDA J Pharm Sci Technol. 1998, 52(5): 238-311.
Praphanphoj et al., “Three cases of intravenous sodium benzoate and sodium phenylacetate toxicity occurring in the treatment of acute hyperammonemia”, J Inherit Metab Dis., 2000, vol. 23, pp. 129-136.
Rajkovic et al., “Mechanisms of abnormalities in host defences against bacterial infection in liver disease,”, Clin Sci. (Lond.), 1985, vol. 3, Issue 68, pp. 247-253, London.
Ramaswamy et al., “Mouse model for human arginase deficiency”, Mol Cell Biol., Jul. 2002, vol. 22, Issue 13, pp. 4491-4498.
Rees et al., “Effect of L-Ornithine-L-Aspartate on patients with and without TIPS undergoing glutamine challenge: a double blind, placebo controlled trial”, Gut, 2000, vol. 47, pp. 571-574.
Riordan et al., “Treatment of hepatic encephalopathy”, Curr Concepts, 1997, vol. 337, Issue 7, pp. 473-479.
Rockey et al., “Randomized, Double-Blind, Controlled Study of Glycerol Phenylbutyrate in Hepatic Encephalopathy,” Hepatology (2014) 59(3): 1073-1083.
Rogers, Q. R. et al., Deficiency of Pyrroline-5-Carboxylate Synthase in the Intestinal Mucosa of the Cat, J Nutrition, 1985, 115(1): 146-150.
Romero-Gómez et al., “Intestinal glutaminase activity is increased in liver cirrhosis and correlates with minimal hepatic encephalopathy”, J Hepatol. 2004, 41: 49-54.
Roque et al., “32* Pro-inflammatory effects of sodium 4-phenylbutyrate in CF lung epithelial cells containing F508del-CFTR”, J Cystic Fibrosis 2007, 6: S7.
Rose et al., “L-Ornithine-L-Aspartate in experimental portal-systemic encephalopathy: therapeutic efficacy and mechanism of action”, Metabolic Brain Disease, 1998, vol. 13, Issue 2, pp. 147-157.
Rose et al., “L-Ornithine-L-Aspartate lowers plasma and cerebrospinal fluid ammonia and prevents brain edema in rats with acute liver failure”, Hepatology, 1999, vol. 30, Issue 3, pp. 636-640.
Rudman et al., Maximal Rates of Excretion and Synthesis of Urea in Normal and Cirrhotic Subjects, The Journal of Clinical Investigation, Sep. 1973, vol. 52, pp. 2241-2249.
Rukmini et al., “Region-specific changes in CNS muscarinic acetylcholine receptors in a rat model of hyperammonemia”, Biochem Pharmacol., 1998, vol. 56, Issue 2, pp. 237-241.
Sanyal et al., Portosystemic Encephalopathy After Transjugular Intrahepatic Portosystemic Shunt: Results of a Prospective Controlled Study, Hepatology, 1994, p. 46-55, vol. 20, No. 1, Pt. 1, The American Association for the Study of Liver Diseases.
Sanyal, A. J., Prediction of Variceal Hemorrhage in Patients with Cirrhosis, UpToDate, Inc., Website (www.uptodate.com), Jan. 2010, UpToDate.
Sarhan et al., “Effects of inhibition of ornithine aminotransferase on thioacetamide-induced hepatogenic encephalopathy”, Neurochem Res., 1993, vol. 18, Issue 4, pp. 539-549.
Scaglia et al., “Effect of alternative pathway therapy on branched chain amino acid metabolism in urea cycle disorder patients”, Mol Genet Metabolism, 2004, vol. 81, pp. S79-S85.
Sears et al., “Disruption of the blood-brain barrier in hyperammonaemic coma and the pharmacologic effects of dexamethasone and difluoromethyl ornithine”, J Neurosci Res., 1985, vol. 14, Issue 2, pp. 255-261.
Seiler et al., “Ornithine aminotransferase activity, liver ornithine concentration and acute ammonia intoxication”, Life Sciences, 1989, vol. 45, Issue 11, pp. 1009-1020.
Seiler, “Ornithine aminotransferase, a potential target for the treatment of hyperammonemias”, 'Curr Drug Targets., Sep. 2000, vol. 1, Issue 2, pp. 119-153.
Sen et al., “The pathophysiological basis of acute-on-chronic liver failure”, Liver, 2002, vol. 22, Issue Suppl. 2, pp. 5-13.
Shangraw et al., Effect of Liver Disease and Transplantation on Urea Synthesis in Humans: Relationship to Acid-Base Status, AM J Physiol Gastrointest Liver Physiol, 1999, vol. 276, pp. 1145-1152.
Shawcross et al., “Ammonia impairs neutrophil phagocytic function in liver disease,”, Hepatology, 2008, vol. 4, Issue 48, pp. 1202-1212.
Shawcross et al., “Dispelling myths in the treatment of hepatic encephalopathy,”, Lancet, 2005, vol. 9457, Issue 365, pp. 431-433.
Shawcross et al., “Hyperammonemia impairs neutrophil function”, Hepatology, 2005, vol. 42, pp. 537A.
Shriner et al., “Recrystallization”, Chapter 3.5 Preliminary Examination in the Systematic Identification of Organic Compounds, John Wiley & Sons, Inc. New York, 1998, Chapter 3, pp. 78-81.
Simell et al., “Waste nitrogen excretion via amino acid acylation: benzoate and phenylacetate in lysinuric protein intolerance”, Pediatric Research, 1986, vol. 20, Issue 11, pp. 1117-1121.
Singapore Examination Report dated Sep. 27, 2013 for Application No. 201107116-4, filed Apr. 1, 2010.
Singapore Examination Report & Written Opinion dated Mar. 8, 2018 for Application No. 10201406300Y, filed Oct. 2, 2014.
Singh, et al., Changing Epidemiology and Predictors of Mortality in Patients With Spontaneous Bacterial Peritonitis at a Liver Transplant Unit, Clinical Microbiology and Infection, Jun. 2003, p. 531-537, vol. 9, No. 6., European Society of Clinical Microbiology and Infectious Diseases.
Smirnov et al., “Ammonia Neutralization and Urea Synthesis in Cardiac Muscle”, Circ Res. 1974, 35(Suppl 3):58-73.
Smith et al., “The treatment of inborn errors of the urea cycle”, Nature, 1981, vol. 291, Issue 5814, pp. 378-380.
Soláini et al., “Variations in the plasma concentration of ornithine, citrulline and arginine in acute experimental liver failure” [Article in Italian], Boll Soc Ital Biol Sper., 1981, vol. 57, Issue 7, pp. 705-710.
Stedman's Medical Dictionary; “Encephalopathy”, 27th Edition, 2002; 1 page.
Stewart, P. M., et al., Effects of Arginine-Free Meals on Ureagenesis in Cats, American Journal of Physiological, 1981, pp. E310-E315, vol. 241, No. 4, The American Physiological Society.
Stravitz, MD, et al., Intensive Care of Patients with Acute Liver Failure: Recommendations of the U.S. Acute Liver Failure Study Group, Critical Care Medicine, 2007, p. 2498-2508, vol. 35, No. 11, Lippincott Williams & Wilkins.
Suchy et al., Clinical Manifestations and Complications—Typical Clinical Presentation;, Liver Disease in Children, 2nd Edition, 2001, pp. 74-77.
Sugarbaker et al., “The role of the small intestine in ammonia production after gastric blood administration”, Ann Surg., 1987, vol. 206, Issue 1, pp. 5-17.
Sukhotnik et al., “Oral glutamine prevents gut mucosal injury and improves mucosal recovery following lipopolysaccharide endotoxemia in a rat,”, J Surg Res., 2007, vol. 2, Issue 143, pp. 379-384.
Svanberg et al., “Effects of amino acids on synthesis and degradation of skeletal muscle proteins in humans”, Am J Physiol., 1996, vol. 271, Issue 4 Pt1, pp. E718-E724.
Timely Data Resource (TDR), IPD Printable Search Results, “Incidence and Prevalence Database, ICD-9 Code: 567. Peritonitis,” <URL:http://www.tdrdata.com/IPD/ipd_searchresultsdataprinter.aspx?SessionGUID=ac0c91d 8-7 . . . , Jul. 27, 2010, 7 pages.
Teran et al., “Primary prophylaxis of variceal bleeding in cirrhosis: A cost-effectiveness analysis”, Gastroenter., 1997, vol. 112, Issue 2, pp. 473-482.
Trebicka et al., Atorvastatin lowers portal pressure in cirrhotic rats by inhibition of RhoA/Roh-kinase and activation of endothelial nitric oxide synthase, Hepatology, 2007, 46(1): 242-253.
Tuchman, M., et al., Management of Inherited Disorders of Ureagenesis, The Endocrinologist, 2002, vol. 12, No. 2, p. 99-109.
Tuchman, MD et al., “Episodic hyperammonemia in adult siblings with hyperornithinemia, hyperammonemia, and homocitrullinuria syndrome”, Arch Neurol., 1990, vol. 47, pp. 1134-1137.
UK Search Report dated Feb. 21, 2005 for GB priority application No. 0426141.8.
Van Berlo et al., “Is increased ammonia liberation after bleeding in the digestive tract the consequence of complete absence of isoleucine in hemoglobin? A study in pigs”, Hepatology, 1989, vol. 10, Issue 3, pp. 315-323.
Van Den Berg et al., “The effect of glutamine-enriched enteral nutrition on intestinal microflora in very low birth weight infants: a randomized controlled trial,”, Clin Nutr., 2007, vol. 4, Issue 26, pp. 430-439.
Ventura-Cots et al., Safety of ornithine phenylacetate in cirrhotic decompensated patients: an open-label, dose-escalating, single-cohort study; J Clin Gastroenter. (2013) 47(10): 881-887.
Vilstrup, H. et al., Elimination of Infused Amino Acids From Plasma of Control Subjects and of Patients With Cirrhosis of the Liver, European Journal of Clinical Investigation, 1982, vol. 12, pp. 197-202, Blackwell Scientific Publications.
Vilatoba et al., Sodium 4-phenylbutyrate protects against liver ischemia reperfusion injury; Surgery, 2005, 138(2):342-351.
Vogels et al., “L-ornithine vs L-ornithine-L-aspartate as a treatment for hyperammonemia-induced encephalopathy in rats”, J Hepatology, 1997, vol. 26, Issue 1, pp. 174-182.
Wasmuth et al., “Patients with acute on chronic liver failure display ‘sepsis-like’ immune paralysis,”, J Hepatol., 2005, vol. 2, Issue 42, pp. 195-201.
Wright et al., “Reduction in Ammonia with L-Ornithine, Phenylacetate (OP) but not Anti-TNF Prevents LPS Induced Brain Edema in Bile-duct Ligated Cirrhotic Rats”, Abstract 773; J Hepatology 2009, 50: S283.
Yoneda et al., “Treatment for non-alcoholic steatohepatitis”, Separate Igakuno Ayumi, Digestive diseases-state of arts Ver. 3, Oct. 10, 2006, pp. 370-372.
Ytreb{acute over (ø)} et al., “Interorgan ammonia, glutamate, and glutamine trafficking in pigs with acute liver failure,”, Am J Physiol Gastrointest Liver Physiol., 2006, vol. 3, Issue 291, pp. G373-G381.
Ytreb{acute over (ø)} et al., “L-Ornithine Phenylacetate Attenuates Increased Arterial and Extracellular Brain Ammonia and Prevents Intracranial Hypertension in Pigs with Acute Liver Failure”, Hepatology, Jul. 2009, 50(1): 165-174.
Yudkoff et al., “In Vivo Nitrogen Metabolism in Ornithine Transcarbamylase Deficiency”, J Clin. Invest., Nov. 1996, 98(9): 2167-2173.
Zetterman, Rowen K., MD, “Complications of Portal Hypertension: Hepatic Encephalopathy”, Medscape (Jun. 2011) available online at www.medscape.com/viewarticle/744392; downloaded Dec. 3, 2014; 6 pages.
Zieve et al., “Ammonia toxicity: comparative protective effect of various arginine and ornithine derivatives, aspartate, benzoate, and carbamyl glutamate”, Metabo Brain Dis., 1986, vol. 1, Issue 1, pp. 25-35.
Zieve et al., “Conditional deficiencies of ornithine or ornithine or arginine”, J Am Coll Nutr., 1986, vol. 5, Issue 2, pp. 167-176.
Related Publications (1)
Number Date Country
20200239406 A1 Jul 2020 US
Provisional Applications (1)
Number Date Country
61166676 Apr 2009 US
Divisions (5)
Number Date Country
Parent 14715481 May 2015 US
Child 15469359 US
Parent 14299940 Jun 2014 US
Child 14715481 US
Parent 13937107 Jul 2013 US
Child 14299940 US
Parent 13436642 Mar 2012 US
Child 13937107 US
Parent 12753763 Apr 2010 US
Child 13436642 US
Continuations (2)
Number Date Country
Parent 16037655 Jul 2018 US
Child 16778696 US
Parent 15469359 Mar 2017 US
Child 16037655 US