Compositions of peptides and processes of preparation thereof

-

The present invention provides composition of comprising a therapeutically effective amount of at least one peptide, polypeptide, analog or derivative thereof and a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, an analog or derivative thereof, wherein at least one stabilizing agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent. The method for preparation of a composition of a peptide, polypeptide, protein, an analog and/or derivative thereof is also provided. The process comprises mixing the peptide, polypeptide, protein, an analog or derivative thereof with a sufficient amount of at least one stabilizing agents to improve the stability of the peptide, polypeptide, protein, an analog or derivative thereof, and the agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
RELATED APPLICATIONS

This application claims the benefit under 35 U.S.C. §119(e) of U.S. Provisional Patent Application Ser. No. 61/051,038, filed May 7, 2008, the disclosures of which is incorporated herein by reference in its entirety.

FIELD OF THE INVENTION

The present invention generally relates to compositions of peptides, polypeptides, proteins, analogues or derivatives thereof, and the process of preparation thereof.

BACKGROUND OF THE INVENTION

A variety of biologically active peptides, polypeptides and proteins have been widely used for medical treatment. For a majority of medical treatments, there is a need to deliver a sustained level of biologically active peptides or proteins to animals or humans to provide a stable therapeutic effect. Additionally, the biologically active peptides or proteins also need to be stored for a prolonged period of time and still maintain their activity. However, many naturally occurring and synthetic peptides and proteins as well as their analogs have exhibited a tendency to form gels, aggregates, fibrils, dimers, other polymers, coagulates, etc. In some cases, the formed material may be able to revert back to the active monomer form. In other cases, the altered state may be permanent and represent a degraded state. Some exemplary biologically active peptides include insulin, glucagon-like peptide-1 (GLP-1) and Gonadotropin-releasing hormone (GnRH) analogs. The instability of the peptides has become a significant barrier for the preparation, process, storage and delivery of these peptides.

To date, a few methods have been discovered that may be used to stabilize solution of peptides or proteins. For example, U.S. Pat. No. 6,124,261 describes a non aqueous polar aprotic peptide formulation that may be used to stabilize the peptide. In another example, a citrate buffering agent may be used to reduce the gelation of fatty acid-acylated protein. See U.S. Pat. No. 5,631,347.

However, due to the diversity of biologically active peptides and proteins, there is a continuing need to find new compositions of peptides or proteins with improved stability and processes of preparing compositions of peptides or proteins.

SUMMARY OF THE INVENTION

The present invention provides compositions comprising a therapeutically effective amount of at least one peptide, polypeptide, protein, an analog or derivative thereof and a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof in a solution, wherein at least one stabilizing agent is a medium chain fatty acid salt, or an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent. In some embodiments, at least one peptide, polypeptide, protein, an analog or derivative thereof is insulin, or an analog or derivative thereof. In one embodiment, at least one peptide, polypeptide, protein, analog or derivative thereof is Glucagon-Like Peptide (GLP), or an analog or derivatives thereof. In another embodiment, the stabilizing agent is selected from the group consisting of sodium caprylate, sodium caprate and sodium laurate.

According to another aspect of the present invention, methods for preparation of a composition of at least one peptide, polypeptide, protein, analog or derivative thereof are provided. In some embodiments, the methods comprises mixing the peptide, polypeptide, protein, analog or derivative thereof with a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof, and the stabilizing agent is a medium chain fatty acid salt, or an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms. In one embodiment, the stabilizing agent is a medium chain fatty acid salt has a carbon chain length of from about 8 to about 14 carbon atoms.

BRIEF DESCRIPTION OF THE DRAWINGS

The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.

FIG. 1 is a table describing the preparation of different acyline batches.

FIG. 2 shows appearance of 9% and 16.7% acyline samples of different acyline batches in propylene glycol.

FIG. 3 shows the appearance of 0.5% and 1% acyline samples of different acyline batches in water.

FIG. 4(a) shows the appearance of 5 mg and 10 mg doses acyline microemulsions of different acyline batches.

FIG. 4(b) shows the formulation of the microemulsion.

FIG. 5 graphically demonstrates the comparison results of the gelation of 0.1 mg/mL acyline sample with 1% and 0% Tween 80.

FIG. 6 graphically demonstrates the comparison results of the gelation of 0.1 mg/mL acyline sample having 5 mg/mL, 1 mg/mL, 0.1 mg/mL and 1% Tween 80.

FIG. 7 graphically demonstrates the comparison results of the gelation of 0.1 and 0.01 mg/mL acyline sample having 1% and 1 mg/mL Tween 80.

FIG. 8 graphically demonstrates the comparison results of the gelation of 0.01 mg/mL acyline sample having 0.1%, 0.5% and 1% Tween 80.

FIG. 9 graphically demonstrates the correlation between the concentration of sodium caprate and the gelation of acyline.

FIG. 10(a) illustrates the formulation of microemulsion 1—55% Capmul MCM. FIG. 10(b) illustrates the formulation of microemulsion 2—45% Capmul PG-8. FIG. 10(c) illustrates the formulation of microemulsion 3—55% Capmul MCM C10.

FIG. 11 graphically demonstrates the comparison results of relative bioavailability of different formulations of acyline.

FIG. 12 graphically demonstrates the comparison results of relative bioavailability of (1) enteric tablets 10 mg acyline versus acyline sample with no surface active agent and (2) enteric tablets 10 mg acyline versus 5 mg acyline with sodium caprate sample.

FIG. 13(a) illustrates the correlation of % of recovery of insulin for insulin formulations with or without sodium caprate. FIG. 13(b) illustrates % of recovery of insulin from insulin formulations with capric acid.

DETAILED DESCRIPTION

The foregoing and other aspects of the present invention will now be described in more detail with respect to the description and methodologies provided herein. It should be appreciated that the invention can be embodied in different forms and should not be construed as limited to the embodiments set forth herein. Rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the invention to those skilled in the art.

All patents, patent applications and publications referred to herein are incorporated by reference in their entirety. In case of a conflict in terminology, the present specification is controlling.

The terminology used in the description of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used in the description of the embodiments of the invention and the appended claims, the singular forms “a”, “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. Also, as used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items. Furthermore, the term “about,” as used herein when referring to a measurable value such as an amount of a compound, dose, time, temperature, and the like, is meant to encompass variations of 20%, 10%, 5%, 1%, 0.5%, or even 0.1% of the specified amount. Unless otherwise defined, all terms, including technical and scientific terms used in the description, have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.

As used herein, “alcohol” means an organic compound in which one or more hydroxyl (OH) groups are attached to carbon (C) atoms in place of hydrogen (H) atoms. In some embodiments, the alcohol contains 1-6 carbon atoms. Yet, in other embodiments, the alcohol contains 1-4 carbon atoms. Exemplary alcohols include, but are not limited to, methanol, ethanol, n-propanol, iso-propanol, butanol, tert-butanol, pentanol, hexanol.

As used herein, “gelation” means that a compound of interest undergoes aggregation to form fibrils, dimers, longer polymers, coagulates, or structures that may result in formation of a colloid structure or gel. The viscosity of the mixture, used in the present application, may be applied to a compound of interest in aqueous solution or a solid mass.

“Gelation” may occur to varying extents, and may occur in such a maimer as to be non-detectable by ordinary means. For example, there may be no increased viscosity or changes in the flow characteristics of the solution. However, the material formed from the gelation (hereinafter “gel”) may be removed by physical means such as filtration and thus detected by analytical techniques known to one skilled in the art. The presence of gels may cause significant problems in the development of different administration forms. For example, when the gelled system is processed to obtain powders of the drug as part of drug substance manufacturing techniques, a xerogel may be formed during the process. As used herein, ‘xerogel’ is a solid formed from the gel after the liquid is removed from a gelled system. The powder obtained from the gelled system containing xerogel may have substantially different characteristics from powders obtained from solutions that contain no gelled material. In some situations, obtained power loses the biological activity. Therefore, the formation of gels may potentially cause deleterious effects at either the drug substance processing stage, or at the stage of preparing the dosage form or storage stage.

The degree of agglomeration or coalescence which results in the formation of the gel may be measured by various methods known to one skilled in the art, for example micro or macro filtration followed by assay of the filtrate, centrifugation followed by assay of the supernatant, or various optical absorption or diffraction methods including those utilizing visible or UV light methods, or laser based methods. The methods of measuring physical characteristics of the liquid may also be used such as surface tension, freezing point depression, viscosity, or measurement of other colligative properties. The degree of xerogel formation may be measured by dispersing the material in water and using the techniques described herein or by direct measurement on the powder by methods known to one skilled in the art including x-ray powder diffraction, IR spectroscopy, or other methods known to the art that may be carried out on powdered materials.

As used herein, the term “reduce the gelation” means to disrupt, retard or eliminate the gel formation of a compound. In situation where the gelation is reversible, the term “reduce the gelation” also means to reverse the gel back to monomeric form of the compound, which maintains it biological activity. As used herein, the term “anti-gelling agent” refers to an agent, a compound, a composition or a combination thereof which may inhibit the gelation of at least 50% of the compound of interest, when a sufficient amount of the anti-gelling agent is used. In some embodiments, the anti-gelling agent may inhibit the gelation of at least 80% of the compound of interest, when a sufficient amount of the anti-gelling agent is used. In some embodiments, the anti-gelling may inhibit the gelation of at least 90% of the compound of interest, when a sufficient amount of the anti-gelling agent is used.

As used herein, a “therapeutically effective” or “therapeutically acceptable” amount refers to an amount that will elicit a therapeutically useful response in a subject. The therapeutically useful response may provide some alleviation, mitigation, or decrease in at least one clinical symptom in the subject. Those skilled in the art will appreciate that the therapeutic useful response need not be complete or curative, as long as some benefit is provided to the subject. In some embodiments, the subject is an animal. In some embodiments, the subject is a human.

As used herein, the term “water miscible solvent” means a solvent that can be mixed with water to form a solution. Water miscible solvents may be used to create a hydrophilic phase.

The present invention provides compositions comprising, consisting essentially of, or consisting of, a therapeutically effective amount of at least one peptide, polypeptide, protein, an analog or derivative thereof and a sufficient amount of at least one stabilizing agents to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof. As used herein, a “stabilizing agent” is an agent that improves the stability of a peptide, polypeptide, protein, an analog or derivative thereof described herein.

In some embodiments, at least one stabilizing agent is a medium chain fatty acid salt, or an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms. In some embodiments, at least one stabilizing agent is a surface active agent.

The present invention provides compositions that maintain the stability of peptide, polypeptide, protein, an analog or derivative thereof described herein for a sufficient time to be stored, processed and/or administered for treatment. One way of measuring the stability is measuring the percentage of peptide, polypeptide, protein, an analog or derivative thereof is retained under a specific condition. As used herein, the term “peptide, polypeptide, protein, analog or derivative thereof is retained” means that the peptide, polypeptide, protein, an analogs or derivative thereof does not form gels, aggregates, fibrils, dimers, other polymers, coagulates, rather it maintains as a monomer or its biological activity after a period of time. In one embodiment, in the composition of the present invention, at least about 50% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, in the composition of the present invention, at least about 80% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, in the composition of the present invention, at least about 90% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, in the composition of the present invention, at least about 95% of the peptide, polypeptide protein, an analog or derivative thereof is retained after the composition stands at about 37° C. for at least about 24 hours. In some embodiments, the stability is improved when the gelation of peptide, polypeptide, protein, an analog or derivative thereof is reduced.

The compositions described herein may be used directly for storage, processing or administration. Alternatively, the composition may also be used to mix with a suitable medium (e.g. a solvent, a microemulsion or a solid mass) for storage, processing or administration. In the situations where a medium is used, the stability of peptide, polypeptide, protein, analogues or derivatives thereof in the medium is improved as described herein.

In some embodiments, the stabilizing agent is an anti-gelling agent. In one embodiment, the present invention provides compositions comprising, a therapeutically effective amount of one or more peptide, polypeptide, protein, an analog or derivative thereof, and a sufficient amount of at least one anti-gelling agents to reduce the gelation of the peptide, polypeptide, protein, an analog or derivative thereof.

In one embodiment, the dissolution rate of the stabilizing agent and the peptide, polypeptide, protein, analogs or derivatives thereof in the composition are substantially the same.

I. Peptide, Polypeptide, Protein, an Analog and Derivative Thereof

The present invention can be applied to any peptide, polypeptide, protein, analog or derivative that has a tendency to aggregates, dimerizes, polymerizes, coagulates, gels or fibrillates. Analogs, derivatives, and pharmaceutically acceptable salts of any of the peptides, polypeptides or proteins are included in these terms. As used herein, the “tendency to to aggregates, dimerize, polymerize, coagulate, gel or fibrillate” refers to that at least 50% of a compound of interest undergoes aggregation to form fibrils, dimers, polymers that coagulates, or structures that may result in formation of a colloid structure or gel in a system at a certain temperature (e.g. 37° C.) after the system stands for a period of time (e.g. at least about 24 hours).

In some embodiments, at least one peptide, polypeptide, protein, an analog or derivative thereof is an insulin or an analog or derivative thereof.

In one embodiment, at least one peptide, polypeptide, protein, analogs or derivatives thereof is Glucagon-Like Peptide (GLP), or an analog or derivative thereof. In another embodiment, at least one GLP is selected from the group consisting of GLP-1, GLP-1 analogs, derivatives thereof, GLP-2, GLP-2 analogs, derivatives thereof, and exendin-4, analogs and derivatives thereof.

More exemplary peptides, polypeptides, proteins, analogs or derivatives thereof include, but are not limited to, GnRH agonists and antagonists, somatostatin, ACTH, corticotropin-releasing factor, angiotensin, calcitonin, gastric inhibitory peptide, growth hormone, growth hormone-releasing factor, pituitary adenylate, exendin, exendin-3, cyclase activating peptide, secretin, enterogastrin, somatostatin, somatotropin, somatomedin, parathyroid hormone, thrombopoietin, erythropoietin, hypothalamic releasing factors, prolactin, thyroid stimulating hormones, endorphins, enkephalins, vasopressin, oxytocin, opioids and analogues thereof, superoxide dismutase, interferon, asparaginase, arginase, arginine deaminase, adenosine deaminase, ribonuclease, FVII, FXIII, a mixture of FVII and FXIII, IL-20, IL-21, IL-28a, IL-29, IL-31, analogs and derivatives thereof.

In some embodiments, at least one peptide, polypeptide, protein, analog or derivative comprises a variety of GnRH related compounds which have a tendency of gelation. As used herein, the “tendency of gelation” refers to that at least 50% of a compound undergoes aggregation to form fibrils, dimers, polymers that coagulates, or structures that may result in formation of a colloid structure or gel in a system at a certain temperature (e.g. 37° C.) after the system stands for a period of time (e.g. at least about 2 hours). As used herein, GnRH related compounds include both GnRH antagonists and GnRH agonists. In some embodiments, the present invention may be applied to GnRH antagonists. In some embodiments, the present invention include, but is not limited to, the following GnRH antagonists, acyline (Ac-D2Nal-D4Cpa-D3Pal-Ser4Aph(Ac)-D4Aph(Ac)-Leu-ILys-Pro-DAla-NH2), Acetyl-β-[2-Naphthyl]-D-Ala-D-p-Chloro-Phe-β-[3-Pyridyl]-D-Ala-Ser-Nε-[Nicotinoyl]-Lys-Nε-[Nicotinoyl]-D-Lys-Leu-Nε-[Isopropyl]-Lys-Pro-D-Ala-NH2 (also referred to herein as Antide), acetyl-D2Nal1, D4CIPhe2, D3Pal3, ARg5, Dglu6 (AA) (also referred to herein as NalGlu), acetyl-D2Nal-D4CIPhe-D3Pal-Ser-Aph(Ac)-D-Aph(Ac)-Leu-Lys(lpr)-Pro-D-Ala-NH2, Abarelix (Specialty European Pharma, Dusseldorf, Germany), Nal-Lys, Synarel, (Searle Peapack, N.J.), Ganirelix (Orgalutron/Antagon) (Organan, West Orange, N.J.), Cetrorelix I (Aeterna Zentaris Inc, Frankfurt, Germany), Cetrotide, Azaline B, new generation long-acting GnRH analogues incorporating p-ureido-phenylalanines at positions 5 and 6 (such as Degarelix), FE200486, Ac-D2Nal-D4Cpa-D3Pal-Ser-4Aph(L-hydroorotyl)-D4Aph(carbarnoyl)-Leu-ILys-Pro-DAla-NH2 (the acetate salt of which is FE200486), Ac-D2Nal-D4Cpa-D3Pal-Ser-4Aph(Atz)-D4Aph(Atz)-Leu-ILys-Pro-DAla-NH2 wherein Atz is 3′-amino-1H-1′,2′,4′-triazol-5′-yl, and the antagonists described in U.S. Pat. Nos. 5,506,207, 5,821,230, 5,998,432, 6,156,772, 6,156,767, 6,150,522, 6,150,352, 6,147,088, 6,077,858, 6,077,847, 6,025,366, 6,017,944, 6,004,984, 6,214,798, and 6,875,843. In some embodiments, the GnRH antagonists of the present invention have a tendency of gelation in the presence of ions. In some embodiments, at least one GnRH antagonist is selected from the group consisting of acyline, abarelix, azaline B, cetrorelix, ganirelix, teverelix, degarelix, antide, orntide and GnRH antagonists described in U.S. Pat. No. 7,098,305.

In one embodiment, at least one GnRH antagonist is selected from the group consisting of abarelix, cetrorelix, degarelix, ganirelix, and a pharmaceutically acceptable salt thereof.

As used throughout this specification and claims, the term “abarelix” refers to a compound having a structure of Formula I

The IUPAC name of Formula I is acetyl-D-β-naphthylalanyl-D-4-chlorophenylalanyl-D-3-pyridylalanyl-L-seryl-L-N-methyl-tyrosyl-D-asparagyl-L-leucyl-L-N(e)-isopropyl-lysyl-L-prolyl-D-alanyl-amide. The term “abarelix” includes the compound of Formula I, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these. The term “abarelix” also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula I and pharmaceutically acceptable salts thereof.

As used throughout this specification and claims, the term “cetrorelix” refers to a compound having a structure of Formula II.

The IUPAC name of Formula II is Acetyl-D-3-(2′-naphtyl)-alanine-D-4-chlorophenylalanine-D-3-(3′-pyridyl)-alanine-L-serine-L-tyrosine-D-citruline-L-leucine-L-arginine-L-proline-D-alanine-amide. The term “cetrorelix” includes the compound of Formula II, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these. The term “cetrorelix” also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula II and pharmaceutically acceptable salts thereof.

As used throughout this specification and claims, the term “degarelix” refers to a compound having a structure of Formula III.

The IUPAC name of Formula III is N-acetyl-3-(naphtalen-2-yl)-D-alanyl-4-chloro-D-phenylalanyl-3-(pyridin-3-yl)-D-alanyl-L-seryl-4-((((4S)-2,6-dioxohexahydropyrimidin-4-yl)carbonyl)amino)-L-phenylalanyl-4-(carbamoylamino)-D-phenylalanyl-L-leucyl-N6-(1-methylethyl)-L-lysyl-L-prolyl-D-alaninamide. It is also known as FE-200486. The term “degarelix” includes the compound of Formula III, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these. The term “degarelix” also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula III and pharmaceutically acceptable salts thereof.

As used throughout this specification and claims, the term “ganirelix” refers to a compound having a structure of Formula IV.

The IUPAC name of Formula IV is a (2S)-1-[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2R)-2-[[(2S)-2-[[(2R)-2-[[(2R)-2-[[(2R)-2-acetamido-3-naphthalen-2-ylpropanoyl]amino]-3-(4-chlorophenyl)propanoyl]amino]-3-pyridin-3-ylpropanoyl]amino]-3-hydroxypropanoyl]amino]-3-(4-hydroxyphenyl)propanoyl]amino]-6-[bis(ethylamino)methylideneamino]hexanoyl]amino]-4-methylpentanoyl]amino]-6-[bis(ethylamino)methylideneamino]hexanoyl]-N-[(2R)-1-amino-1-oxopropan-2-yl]pyrrolidine-2-carboxamide. The term “ganirelix” includes the compound of Formula IV, pharmaceutically acceptable salts thereof, and equilibrium mixtures of these. The term “ganirelix” also includes crystalline, hydrated or solvated crystalline, and amorphous forms of the compound of Formula IV and pharmaceutically acceptable salts thereof.

In some embodiments, the present invention may be applied to GnRH agonists that have a tendency of gelation. The exemplary GnRH agonists include, but are not limited to, histerelin, leuprolide and goserelin.

The terms “GnRH related compound”, “GnRH antagonist” and “GnRH agonist” include all forms thereof including stereoisomers, enantiomers, diastereomers, racemic mixtures, and derivatives thereof, for example, salts, acids, esters and the like. The compound may be provided in any suitable phase state including as solid, liquid, solution, suspension and the like. When provided in a solid particulate form, the particles may be of any suitable size or morphology and may assume one or more crystalline, semi-crystalline and/or amorphous forms.

The peptide, polypeptide, protein, analogs or derivatives there of used in the present invention may be present in any amount which is sufficient to elicit a therapeutic effect and, where applicable, may be present either substantially in the form of one optically pure enantiomer or as a mixture, racemic or otherwise, of enantiomers. As will be appreciated by those skilled in the art, the actual amount of peptide, polypeptide, protein, analogs or derivatives there of used in the composition will depend on the potency of the selected compound in question.

The peptides, polypeptides, proteins, analogs or derivatives described herein may be obtained through commercial resources or may be prepared according to methods known to one skill in the art. The GnRH antagonists applied in the present invention may be prepared using a method known to one of ordinary skill in the art or a process described in the present invention. For example, acyline can be prepared according to the method described in U.S. Pat. No. 5,506,207.

II. Stabilizing Agents

The amount of the stabilizing agent used in the present invention can vary considerably. For example, the amount can be dependent upon individual stabilizing agents, solvent systems and other components in the composition. Generally, the amount of stabilizing agent should be sufficient to improve the stability of peptide, polypeptide, protein, analogs or derivatives thereof in a system such that at least 50% of peptide, polypeptide, protein, an analog or derivative thereof is retained when the composition stands at 37° C. for at least about 24 hours. In some embodiments, the amount of stabilizing agent should be sufficient to improve the stability of peptide, polypeptide, protein, analogs or derivatives thereof in a system such that at least 80% of peptide, polypeptide, protein, analog or derivative thereof is retained when the composition stands at 37° C. for at least about 24 hours. In other embodiments, the amount of anti-gelling agent should be sufficient to be sufficient to improve the stability of peptide, polypeptide, protein, an analog or derivative thereof in a system such that at least 90% of peptide, polypeptide, protein, an analog or derivative thereof is retained when the composition stands at 37° C. for at least about 24 hours. In some embodiments, the concentration of the stabilizing agent in the composition is at least equal to or above the CMC (Critical Micelle Concentration) of the stabilizing agent in the composition.

A. Medium Chain Fatty Acid and Derivatives Thereof

In some embodiments, at least one stabilizing agent is a medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 4 to 20 carbon atoms. In some embodiments, at least one stabilizing agent is medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 6 to 20 carbon atoms. In some embodiments, the carbon chain length is from 8 to 14. In some embodiments, at least one stabilizing agent is a salt of medium chain fatty acid and has a carbon chain length of from 8 to 14 carbon atoms. In some embodiments, at least one stabilizing agent is a medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 6 to 20 carbon atoms; with the provisos that (i) where the stabilizing agent is an ester of a medium chain fatty acid, said chain length of from 6 to 20 carbon atoms relates to the chain length of the carboxylate moiety, and (ii) where the stabilizing agent is an ether of a medium chain fatty acid, at least one alkoxy group has a carbon chain length of from 6 to 20 carbon atoms. In another embodiment, at least one stabilizing agent is a medium chain fatty acid salt, or ester, ether or a derivative of a medium chain fatty acid which is solid at room temperature and which has a carbon chain length of from 8 to 14 carbon atoms; with the provisos that (i) where the stabilizing agent is an ester of a medium chain fatty acid, said chain length of from 8 to 14 carbon atoms relates to the chain length of the carboxylate moiety, and (ii) where the stabilizing agent is an ether of a medium chain fatty acid, at least one alkoxy group has a carbon chain length of from 8 to 14 carbon atoms. In some embodiments, at least one stabilizing agent is a sodium salt of a medium chain fatty acid, the medium chain fatty acid having a carbon chain length of from 8 to 14 carbon atoms. In some embodiments, the stabilizing agent is solid at room temperature. In another embodiment, at least one stabilizing agent is selected from the group consisting of sodium caprylate, sodium caprate, and sodium laurate. In one embodiment, at least one stabilizing agent is sodium caprate.

B. Surface Active Agents

In some embodiments, at least one stabilizing agent is a surface active agent. As used herein, the term “surface active agent” refers to an agent that lowers the surface tension of the medium in which it is dissolved and/or the interfacial tension with other phases, and, accordingly, is positively adsorbed at the liquid/vapor and/or at other interfaces. The surface active agents employed in the present invention include both ionic agents, i.e., cationic, anionic or zwitterionic, and non-ionic agents, or a mixture thereof.

Examples of cationic surface active agents include, but are not limited to, benzalkonium chloride, dicetyl ammonium chloride, cetyldimethylethylammonium bromide, cetylpyridinium chloride and salts of the above surface active agents.

Examples of anionic surface active agents include, but are not limited to, sodium stearoyl lactylate, hydrogenated lecithin, sodium lauryl sulfate, C8-32 fatty acids and salts thereof, cholic acid and derivatives thereof such as deoxycholate, and its salts, ursodeoxycholic acid, and taurocholic acid; C8-56 diesters of tartaric acid; phospholipids such as phosphatidic acid and phosphatidyl serine; C5-29 monoesters of lactic acid; C8-20 sulfonates, including alkyl-, olefin-, and alkylaryl derivatives; tridecyl- and dodecylbenzene sulfonic acids; and C5-33 sarcosine and betaine derivatives.

Examples of zwitterionic surface active agents include, but are not limited to, phospholipids such as lecithin, phosphatidylethanolamine, sphingomyelins, dodecyl betaine, dodecyl dimethylamine oxide, cocamidopropyl betaine, and coco ampho glycinate.

Examples of non-ionic surface active agents include, but are not limited to, steareths; polyethylene glycol (PEGs); polysorbates (e.g. Tween 80); cetearyl glucoside; various commercially available sorbitans and their derivatives, for example, sorbitan hexastearate ethoxylate EO 6 mole, sorbitan isostearate, sorbitan laurate, sorbitan monoisostearate ethoxylate EO 20 mole, sorbitan monolaurate ethoxylate EO 20 mole, sorbitan monooleate ethoxylate EO 20 mole, sorbitan monopalmitate ethoxylate EO 20 mole, sorbitan monostearate ethoxylate EO 20 mole, sorbitan monstearate ethoxylate EO 6 mole, Sorbitan oleate, sorbitan palmitate, sorbitan sesquioleate, sorbitan stearate, sorbitan tetraoleate ethoxylate EO 30 mole, sorbitan tetraoleate ethoxylate EO 40 mole, sorbitan tetraoleate ethoxylate EO 6 mole, sorbitan tetrastearate ethoxylate EO 60 mole, sorbitan trioleate ethoxylate EO 20 mole, sorbitan trioleate, sorbitan tristearate ethoxylate EO 20 mole, and sorbitan tristearate; ethoxylated castor oil, C5-29 mono-glycerides and ethoxylated derivatives thereof; C15-60 diglycerides and polyoxyethylene derivatives thereof having 1 to 90 POE groups; C10-40 esters (10-40 carbon atoms in the alcohol) of long chain fatty acids (fatty acids having 16 carbon atoms and above); C10-40 alcohols; sterols such as cholesterol, ergosterol, and C2-24 esters thereof; C8-96 ethoxylated fatty esters; C14-130 sucrose fatty esters; and polyoxyethylene (POE) derivatives thereof having 0 to 90 POE groups, e.g., polyoxyethylene sorbitan monooleate, sorbitol hexaoleate POE (50).

In some embodiments, at least one surface active agent is selected from the group consisting of sodium lauryl sulfate, polysorbate surface active agents (such as polysorbate 20 (Tween 20), polysorbate 80 (Tween 80)), sorbitan surface active agents, sorbitan monolaurate and polyethoxylated castor oil and a combination thereof. In some embodiments, at least one surface active agent comprises polysorbate.

III. Additional Excipients

According to some aspects of the present invention, the composition of the present invention further comprises one or more excipients. As will be appreciated by those skilled in the art, the exact choice of excipients and their relative amounts will depend to some extent on the final dosage form. In some embodiments, the excipients are selected from the group consisting of rate-controlling polymeric materials, diluents, lubricants, disintegrants, plasticizers, anti-tack agents, opacifying agents, pigments, and flavorings.

As used herein, the term “rate controlling polymer material” comprises hydrophilic polymers, hydrophobic polymers and mixtures of hydrophilic and/or hydrophobic polymers that are capable of controlling or retarding the release of the drug compound such as a GnRH related compound from a solid oral dosage form of the present invention. Suitable rate controlling polymer materials include those selected from the group consisting of hydroxyalkyl cellulose such as hydroxypropyl cellulose and hydroxypropyl methyl cellulose; poly(ethylene) oxide; alkyl cellulose such as ethyl cellulose and methyl cellulose; carboxymethyl cellulose, hydrophilic cellulose derivatives; polyethylene glycol; polyvinylpyrrolidone; cellulose acetate; cellulose acetate butyrate; cellulose acetate phthalate; cellulose acetate trimellitate; polyvinyl acetate phthalate; hydroxypropylmethyl cellulose phthalate; hydroxypropylmethyl cellulose acetate succinate; polyvinyl acetaldiethylamino acetate; poly(alkylmethacrylate) and poly (vinyl acetate). Other suitable hydrophobic polymers include polymers and/or copolymers derived from acrylic or methacrylic acid and their respective esters, zein, waxes, shellac and hydrogenated vegetable oils. In one embodiment, the rate-controlling polymer comprises a polymer derived from acrylic or methacrylic acid and their respective esters or copolymers derived from acrylic or methacrylic acid and their respective esters. In another embodiment, the rate-controlling polymer comprises hydroxypropylmethylcellulose (HPMC).

In some embodiments, at least one rate controlling polymer material is selected from the group consisting of poly acrylic acid, poly acrylate, poly methacrylic acid and poly methacrylate polymers such as those sold under the Eudragit® trade name (Rohm GmbH, Darmstadt, Germany), for example, Eudragit® L, Eudragit® S, Eudragit® RL, Eudragit® RS coating materials and mixtures thereof. Some of these polymers can be used as delayed release polymers to control the site where the drug is released. In some embodiments, the rate controlling polymer include polymethacrylate polymers such as those sold under the Eudragit® trade name (Rohm GmbH, Darmstadt, Germany), for example, Eudragit® L, Eudragit® S, Eudragit® RL, Eudragit® RS coating materials and mixtures thereof.

In some embodiments, the present invention may further comprise diluents. Any suitable diluent may be used in the present invention. Exemplary diluents include, but are not limited to, pharmaceutically acceptable inert fillers such as microcrystalline cellulose, lactose, dibasic calcium phosphate, saccharides, and/or mixtures thereof. Examples of diluents include microcrystalline cellulose such as those sold under the Avicel trademark (FMC Corp., Philadelphia, Pa.), for example, Avicel™ pH101, Avicel™ pH102 and Avicel™ pH112; lactose such as lactose monohydrate, lactose anhydrous and Pharmatose DCL21; dibasic calcium phosphate such as Emeompress® (JRS Pharma, Patterson, N.Y.); mannitol; starch; sorbitol; sucrose; and glucose. In some embodiments, the inert filler comprises microcrystalline cellulose. In one embodiment, the inert filler comprises a lactose selected from the group consisting of lactose monohydrate and lactose anhydrous. In another embodiment, the inert filler comprises a saccharide selected from the group consisting of mannitol, starch, sorbitol, sucrose, and glucose. In one embodiment, the saccharide is sorbitol.

In some embodiments, the composition of the present invention may further comprise lubricants. Any suitable lubricant may be used in the present invention. In some embodiments, the lubricant comprises agents that act on the flowability of the powder to be compressed. Exemplary lubricants include, but are not limited to, colloidal silicon dioxide such as Aerosil™ 200, talc, stearic acid, magnesium stearate, and calcium stearate. In some embodiments, the lubricant is stearic acid.

In some embodiments, the composition of the present invention may further comprise disintegrants. Any suitable disintegrant may be used in the present invention. Exemplary disintegrants include, but are not limited to, lightly cross-linked polyvinyl pyrrolidone, corn starch, potato starch, maize starch and modified starches, croscarmellose sodium, cross-povidone, sodium starch glycolate and combinations and mixtures thereof. In some embodiments, the disintegrant is chosen from crospovidone and polyvinylpyrrolidone.

In some embodiments, the composition described above may further comprise an enhancer. The enhancer can be any suitable enhancer that is known to one of ordinary skill in the art. Exemplary enhancers include, but are not limited to, a medium chain fatty acid salt, ester, ether or a derivative of a medium chain fatty acid which has a carbon chain length of from 4 to 20 carbon atoms. In some embodiments, the enhancer is solid at room temperature. In some embodiments, the enhancer is medium chain fatty acid salt, ester, ether or a derivative of a medium chain fatty acid and which has a carbon chain length of from 6 to 20 carbon atoms. In some embodiments, the carbon chain length is from 8 to 14. In some embodiments, the enhancers are S-Cyclodextrins, vitamin E TPGS, gallic acid esters, crospovidones, sorbitan esters, poloxamers, or olyoxyethylene glycolated natural or hydrogenated castor oil (Cremophor®, BASF). In some embodiments, the enhancers are medium chain glycerides or a mixture of medium chain glycerides. Exemplary enhancers are further described in U.S. Patent Publication No. 2003/0091623, and U.S. Pat. No. 6,372,728 which are incorporated by reference in their entireties.

IV. Application of Co-Solvent System During the Preparation of a GnRH Related Compound

The GnRH related compound described herein may be prepared in the presence of a co-solvent system in a manner that the gelation of the GnRH compound is reduced. In some embodiments, the final step of the preparation of the GnRH related compound is conducted in the presence of a co-solvent system. In some embodiments, the final step is drying, using known techniques, such as lyophilization, tray drying, spray drying, fluid bed drying, or other similar techniques known to one skilled in the art.

In some embodiments, the co-solvent system comprises water and at least one water-miscible solvent. The water miscible solvent can be chosen from, but are not limited to, linear or branched C1-6 alcohol, tetrahydrofuran, acetone, ethyl methyl ketone, methyl isobutyl ketone, methyl isopropyl ketone, cyclohexanone, diethyl ketone, pentan-3-one, cyclohexane, acetonitrile, N,N-dimethylformamide, N,N-dimethylacetamide, dioxane, alcohol, ethylene glycol, diglyme, monoglyme, ethylene glycol monomethyl ether, diethylene glycol, triethylene glycol, polyethylene glycol and a mixture thereof. In certain embodiments, at least one water miscible solvent is a linear or branched C1-6 alcohol. Exemplary suitable alcohols include, but are not limited to, methanol, ethanol, propanol, iso-propanol, butanol, sec-butanol, iso-butanol, tert-butanol, 1-pentanol, 2-pentanol, and hexanol. In some embodiments, at least one water miscible solvent is selected from the group consisting of methanol, ethanol, n-propanol, iso-propanol and tert-butanol. In some embodiments, at least one water miscible solvent is selected from the group consisting of methanol, ethanol, iso-propanol, tert-butanol, acetoniltrile and methylene chloride. In some embodiments, the present invention may be carried out with two or more water miscible solvents. In some embodiments, the weight ratio of the water miscible solvent to water is in the range of about 1/1000 to about 99/1. In some embodiments, the weight ratio is about 3/97 to about 59/41.

In some embodiments, the process comprises further adding acid during the preparation of the GnRH related compound. Acids used in the present invention include, but are not limited to acetic acid, sulfuric acid, hydrochloride acid, trifluoracetate, citrate acid, tartaric acid, ascorbic acid, and boric acid, etc. Generally, the concentration of the acid is sufficient to prepare a salt of the GnRH related compound. In some embodiments, the concentration of the acid depends on the molecular weight of the GnRH related compound and the acid used herein. In some embodiments, the concentration is in the range of about 0.5% to about 20% of the solution.

V. Methods of Preparing the Compositions

According to some aspects of the present invention, methods for preparation of a composition of a peptide, polypeptide, protein, an analog or derivative thereof, are provided. In some embodiments, the method comprises mixing the peptide, polypeptide, protein, an analog or derivative thereof with a sufficient amount of at least one stabilizing agents to improve the stability of peptide, polypeptide, protein, an analog and/or derivative thereof. In some embodiments, at least one stabilizing agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent. As used herein, the term “mixing” means contacting, combining, reacting, and/or coating a peptide, polypeptide, protein, an analog and/or derivative thereof, with one or more stabilizing agents. In one embodiment, “mixing” means combining a peptide, polypeptide, protein, an analog or derivative thereof with a stabilizing agent in a composition. In one embodiment, the GnRH related compound is prepared in the presence of a co-solvent system described herein. In some embodiment, the concentration of the stabilizing agent in the composition is equal to or above the critical micelle concentration of the stabilizing agent.

VI. Pharmaceutical Compositions and Administration

In one embodiment, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of one or more peptide, polypeptide, protein, an analog or derivative thereof, and a sufficient amount of at least one stabilizing agents to improve the stability of the peptide, polypeptide, protein, an analog or derivative thereof, wherein at least one stabilizing agent is a medium chain fatty acid salt, an ester, an ether, or a derivative of a medium chain fatty acid and has a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent. In another embodiment, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier. The term “pharmaceutically acceptable carrier” as used herein refers to any substance, not itself a therapeutic agent, used as a vehicle for delivery of a therapeutic agent to a subject.

The compositions of the present invention may be suitable for formulation for oral, parenteral, inhalation spray, topical, rectal, nasal, sublingual, buccal, vaginal or implanted reservoir administration, etc. In one embodiment, the compositions are administered orally, topically, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.

In some embodiments, the composition used in the present invention is in an oral dosage form. In some embodiments, the oral dosage form is chosen from tablets, capsules, granules, powders, capsules filled with granules or powders, capsules filled with liquids or semi-solids, sachets filled with granules or powders, liquid, emulsions, microemulsions, and any composition that capable of forming emulsions. In one embodiment, the oral dosage form may be a tablet, a multiparticulate, or a capsule. In one embodiment, the medium chain fatty acid and derivatives thereof described herein may also function as an enhancer.

In some embodiments, the oral dosage form is a delayed release dosage form which minimizes the release of the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agents and/or the enhancer in the stomach, and hence the dilution of the local stabilizing agent or the concentration of the enhancer therein, and releases the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agent or the enhancer in the intestine. In some embodiments, the oral dosage form is a delayed release rapid onset dosage form. Such a dosage form minimizes the release of peptide, polypeptide, protein, an analog or derivative thereof and stabilizing agent or enhancer in the stomach, and hence the dilution of the local stabilizing agent or enhancer concentration therein, but releases the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agent or the enhancer rapidly once the appropriate site in the intestine has been reached, maximizing the delivery of the poorly permeable or soluble peptide, polypeptide, protein, an analog or derivative thereof by maximizing the local concentration of the peptide, polypeptide, protein, an analog or derivative thereof and the stabilizing agent at the site of absorption.

In the case of any of the embodiments described herein, a controlled release coating may be applied to the final dosage form (capsule, tablet, multilayer tablet etc.). In one embodiment, the controlled release coating may comprise a rate controlling polymer material described herein. The dissolution characteristics of such a coating material may be pH dependent or independent of pH.

In some embodiments, the composition of the present invention may have an enteric coating thereon. In one embodiment, the enteric coating comprises a polymer selected from the group consisting of poly(acrylic acid), polyacrylate, poly(methacrylic acid), polymethacrylate, and mixtures thereof. In some embodiments, the enteric coated composition may be in the form of a tablet or capsule.

The term “tablet” as used herein includes, but is not limited to, immediate release (IR) tablets, sustained release (SR) tablets, matrix tablets, multilayer tablets, multilayer matrix tablets, extended release tablets, delayed release tablets and pulsed release tablets any or all of which may optionally be coated with one or more coating materials, including polymer coating materials, such as enteric coatings, rate-controlling coatings, semi-permeable coatings and the like. The term “tablet” also includes osmotic delivery systems in which a drug compound is combined with an osmagent (and optionally other excipients) and coated with a semi-permeable membrane, the semi-permeable membrane defining an orifice through which the drug compound may be released. Tablet solid oral dosage forms may be useful in the practice of the invention include those selected from the group consisting of IR tablets, SR tablets, coated IR tablets, matrix tablets, coated matrix tablets, multilayer tablets, coated multilayer tablets, multilayer matrix tablets and coated multilayer matrix tablets. In some embodiments, a tablet dosage form is an enteric-coated tablet dosage form. In some embodiments, a tablet dosage form is an enteric-coated rapid onset tablet dosage form.

Capsule solid oral dosage forms may be useful in the practice of the present invention include those selected from the group consisting of instant release capsules, sustained release capsules, coated instant release capsules, and coated sustained release capsules including delayed release capsules. Capsules may be filled with powders, granules, multi particulates, tablets, semi-solids, or liquids. In some embodiments, a capsule dosage form is an enteric-coated capsule dosage form. In some embodiments, a capsule dosage form is an enteric-coated rapid onset capsule dosage form. Capsules may be made of hard gelatin, soft gelatin, starch, cellulose polymers, or other materials as known to the art.

The term “multiparticulate” as used herein means a plurality of discrete particles, pellets, mini-tablets and mixtures or combinations thereof. If the oral form is a multiparticulate capsule, such hard or soft gelatin capsules can suitably be used to contain the multiparticulate. In some embodiments, a sachet may suitably be used to contain the multiparticulate. In some embodiments, the multiparticulate may be coated with a layer containing rate controlling polymer material. In some embodiments, a multiparticulate oral dosage form according to the invention may comprise a blend of two or more populations of particles, pellets, or mini-tablets having different in vitro and/or in vivo release characteristics. For example, a multiparticulate oral dosage form may comprise a blend of an instant release component and a delayed release component contained in a suitable capsule.

In some embodiments, the multiparticulate and one or more auxiliary excipient materials can be compressed into tablet form such as a multilayer tablet. In some embodiments, a multilayer tablet may comprise two layers containing the same or different levels of the same active ingredient having the same or different release characteristics. In some embodiments, a multilayer tablet may contain different active ingredient in each layer. Such a tablet, either single layered or multilayered, can optionally be coated with a controlled release polymer so as to provide additional controlled release properties. In some embodiments, multiparticulate dosage form comprises a capsule containing delayed release rapid onset minitablets. In some embodiments, a multiparticulate dosage form comprises a delayed release capsule comprising instant release minitablets. In some embodiments, a multiparticulate dosage form comprises a capsule comprising delayed release granules. In some embodiments, a multiparticulate dosage form comprises a delayed release capsule comprising instant release granules.

The term “emulsion” used herein means a suspension or dispersion of one liquid within a second immiscible liquid. In some embodiments, the emulsion is an oil-in-water or water-in-oil-in-water emulsion.

The term, “microemulsion” used herein means a solution in which the hydrophobic (oil-like) phase and the hydrophilic (water-like) phase and a surface active agent form micelle structures. Such dispersions are clear and stable over time.

In addition, “emulsion” or “microemulsion”, used herein includes a hydrophilic or a hydrophobic liquid which, on dilution with a hydrophobic or a hydrophilic liquid respectively, form an emulsion or a microemulsion. In some embodiments, “emulsion”, or ‘microemulsion’, used herein may include solid or semi-solid materials which may be liquid at higher temperatures. For example, the material may be solid at room temperature. At about body temperature (about 37° C.), the material may be liquid.

Alternatively, pharmaceutically acceptable compositions of this invention may be in the form of a suppository for rectal administration. The suppositories can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.

Pharmaceutically acceptable compositions of the present invention may be in the form of a topical solution, ointment, or cream in which the active component is suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Where the topical formulation is in the form of an ointment or cream, suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2 octyldodecanol, benzyl alcohol and water.

When the pharmaceutically acceptable composition is an ophthalmic formulation, it may be a micronized suspension in isotonic, pH adjusted sterile aqueous solution, or as a solution in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in the form of an ointment.

The pharmaceutically acceptable compositions of this invention may also be administered by nasal, aerosol or by inhalation administration routes. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.

It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, gender, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.

VII. Methods of Treatment and Use

Another aspect of the present invention provides a method of treatment of a medical condition treatable by a peptide, polypeptide, protein, an analog or derivative thereof comprising administering to a patient suffering from said condition a pharmaceutical composition as described in the present application. As used herein, the medical condition includes, but is not limited to, diabetes mellitus, Alzheimer, autoimmune diseases, colon cancer, sex hormone dependent disease such as benign prostate hyperplasia, prostate cancer, estrogen-dependent breast cancer, endometrial cancer, ovarian cancer, endometriosis and precocious puberty, and contraception in a human or animal subject

Yet, another embodiment provides use of pharmaceutical composition as described in the present application in the manufacture of a medicament for the treatment of a medical condition treatable by said peptide, polypeptide, protein, analogs and/or derivatives thereof.

It is understood that the combinations of all embodiments described herein are also envisaged in the present invention.

The present invention will now be described in more detail with reference to the following examples. However, these examples are given for the purpose of illustration and are not to be construed as limiting the scope of the invention.

EXAMPLES 1. Application of Co-Solvent During the Preparation of Acyline (1) Preparation of Acyline Batches

The six different acyline batches are described in FIG. 1, which summarizes the variations in the lyophilization procedure, which is the last step of the preparation of acyline. For lots XF 173/315-125, XF 185/165-133 and XF 173/315-151A, a co-solvent system is used in the lyophilization step. Water is used as a solvent for other batches. The batches and the associated lyophilization solvents are also illustrated in FIG. 1. The acyline may be prepared according to methods known to one of skill in the art, for example U.S. Pat. No. 6,747,125, or processes described in the present application.

Example 1

The study of gelation for different acyline batches in propylene glycol is carried out and the result is summarized in FIG. 2. The 9% and 16.7% of acyline sampls are prepared by using the acyline batches listed in FIG. 1. The 9% acyline sample of XF 173/315-125, XF 185/165-133 and XF 173/315-151A are prepared by using acyline prepared via use of a co-solvent during the lyophilization step. The acyline batches of XF 173/315-125, XF 185/165-133 and XF 173/315-151A appear clear and non-viscous after 2 hours. Other batches which are lyophilized by using water as a solvent did not appear as clear and non-viscous solutions due to the presence of gelled acyline.

Example 2

The gelation of different acyline drug batches in water is investigated and the results are summarized in FIG. 3. The 0.5% and 1% acyline samples are prepared by using the acyline batches listed in FIG. 1. The 0.5% and 1% of acyline samples of XF 173/315-125, XF 185/165-133 and XF 173/315-151A which are prepared by using co-solvent during the lyophilization step appear clear and non viscous upon dissolving in water.

Example 3

The tendency of gelation of different acyline batches in standardized microemulsion (SM) is investigated. The result is summarized in FIG. 4(a). The formulation of the standard micro emulsion is shown in FIG. 4(b). The 5 mg and 10 mg formulations of acyline in a microemulsion are prepared by using the acyline lots synthesized in FIG. 1. 5 mg and 10 mg dose of acyline batches of XF 173/315-151A which are prepared by using co-solvent during the lyophilization step appeared clear and transparent after 2 hours.

2. Application of Anti-Gelling Agents in the Formulation of Acyline Compositions General Procedures of Comparison Experiments

Different concentrations of acyline composition are prepared by adding acyline to pH 6.8 buffer solution either at room temperature or 37° C. All acyline solutions contain 0.6 mg/mL sodium caprate (sodium caprate is referred as C10 in the figures). All samples are centrifuged and filtered prior to analysis. Samples are analyzed by reverse phase HPLC with UV detection.

Example 4

10 mg acyline is transferred into 200 mL volumetric flask. A pH 6.8 buffer solution is preheated to 37° C. Then, 100 mL pre-heated buffer solution and 0.6 mg/mL sodium caprate is added to the flask to prepare a 0.1 mg/mL acyline sample. In a similar manner, Tween 80 is added to prepare a 0.1 mg/mL acyline solution with 1% Tween 80. Samples are shaken in a temperature controlled water bath at 37° C. 5 mL samples are taken at 1, 5, 10, 15, 20, 30 and 120 minutes after mixing acyline with the buffer solution. Samples are filtered immediately through 0.45 μm filters and the first 3 mL is discarded. Filtered samples are analyzed, undiluted, by reverse phase HPLC with UV detection. All samples are prepared in duplicate and the concentration was obtained for the analysis from the mean of the duplicates. The result of comparison of 1% and 0% Tween 80 at 0.1 mg/mL acyline sample is graphically recorded in FIG. 5. The results show that sodium caprate alone at a concentration below the CMC is insufficient to reduce the gelation of acyline. The addition of 1% Tween 80 successfully reduces the gelation.

Example 5

A similar experimental procedure as Example 4 is used to prepare acyline sample for example 5. The result of comparison of 5 mg/mL, 1.0 mg/mL, 0.1 mg/mL, and 0.1% Tween in phosphate buffer containing 0.6 mg/mL of sodium caprate and 0.1 mg/mL acyline is graphically recorded in FIG. 6. The results show that only 1% Tween 80 solution may completely inhibit the gelation of acyline.

Example 6

A similar experimental procedure as Example 4 is used to prepare acyline sample for example 6. The result of comparison of 1% and 1 mg/mL Tween 80 in 0.1 and 0.01 mg/mL acyline sample is graphically recorded in FIG. 7. The results show that 1% Tween 80 reduces gelation for both 0.1 mg/mL and 0.01 mg/mL acyline sample.

Example 7

A similar experimental procedure as Example 4 is used to prepare acyline sample for example 7. The result of comparison of 0.01 mg/mL acyline samples having 0.1%, 0.5% and 1% Tween 80 is graphically recorded in FIG. 8. The results show that the acyline sample with 1% Tween 80 significantly reduces the gelation.

3. Impact of Sodium Caprate (C10) on the Tendency of Gelation of Acyline in Water Example 8

FIG. 9 graphically demonstrates the impact of different concentrations of sodium caprate (C10) on the gelation of acyline in water. When the concentration of sodium caprate is below 10 mg/mL, the recovery of acyline significantly decreases, which implies an increase of gelation of acyline. The investigators of the present invention believe that the increase of the gelation is due to an increased concentration of ions caused by the addition of sodium caprate. However, when the concentration of sodium caprate reaches and is above the CMC of sodium caprate (˜20 mg/mL), it is observed that there is a sudden and significant increase of the recovery of acyline, which indicates an effective reduction of gelation. The CMC of sodium caprate is known as 100 mM (˜20 mg/mL). (See “kinetic studies of the interaction of fatty acids with phosphatidylcholine vesicles (liposomes), Rogerson et al., Colloids and Surfaces B: Biointerfaces, 48, 24-34 (2006).) When the concentration of sodium caprate reaches about 50 mg/mL, the recovery of acyline is almost 100%, which indicates that the gelation is completely inhibited.

4. Application of Anti-Gelling Agents in Different Micoremulsion formulations of Acyline Samples

Example 9

FIG. 11 graphically demonstrates the relative bioavailability of various formulations of acyline in dogs. The relative bioavailability is measured by comparing the absolute bioavailability of various formulations of acyline with the absolute bioavailability of a standard formulation of acyline, which is a formulation without any anti-gelling agent. The formulation of microemulsion 1 (M1/55% Capmul MCM), microemulsion 2 (M2/45% Capmul PG-8), and microemulsion 3 (M3/55% Capmul MCM C10) are illustrated in FIG. 10(a) through (c). The formulation of the standard solution is 5 mg acyline, 550 mg sodium caprate and 5 mL purified water. “C10” in FIGS. 10 and 11 represents sodium caprate. “SLS” in FIGS. 10 and 11 represents sodium lauryl sulphate. The comparison results in FIG. 11 show that all formulations with anti-gelling agents such as sodium caprate and sodium lauryl sulphate, increase the bioavailability of acyline from 7.8 fold to 32.5 fold.

Example 10

FIG. 12 graphically demonstrates the relative bioavailability of (1) enteric tablets of 10 mg acyline versus unenhanced 5 mg acyline solution sample and (2) enteric tablets of 10 mg acyline versus 5 mg acyline standard sample. The formulation of acyline standard sample is 5 mg acyline, 550 mg sodium caprate and 5 mL purified water. The acyline in the tablets and the 5 mg acyline sample is prepared by using a lyophilization step with water as the solvent. The tablets contained the same amount of sodium caprate as the 5 mg acyline sample. It is observed that, the concentration of sodium caprate, 110 mg/mL, is sufficient to reduce or even inhibit gelation in the solution and enhance the bioavailability of acyline solution. FIG. 12 demonstrates that the bioavailability of enhanced tablets is significantly improved compared to unenhanced tablet.

5. Application of Sodium Caprate and Capric Acid in Insulin Samples Example 11

A study evaluating the effect of sodium caprate (C10) and capric acid on the stability of bovine insulin is carried out. In this study, bovine Insulin with a potency of 29 U/mg is used to prepare a 10 U/mL solution (344.83 mg/L of solution) in a phosphate buffer pH 8. Using 200 mL vessels, appropriate amounts of C10 is added to 100 mL aliquots of the insulin solution. Two bottles are used a controls, containing only the insulin solution. An addition of 3 g of C10 is added to bottles 3 and 4, 6 g of C10 to bottles 5 and 6, 3 g of Capric Acid to bottles 7 and 8, and 6 g of Capric Acid to bottles 9 and 10. The bottles are gently swirled and 6 mL aliquots are removed from each bottle for the T=ZERO sample. The samples are filtered (Millex-HV 0.45 μm), and the first 5 ml of filtrate discarded. The appropriate test solutions are stirred and additional samples are collected from all test solutions at 6, 24 hours etc. The amount of bovine insulin in solution after filtration is determined against standards (70 mg in 200 ml of 0.01 N HCl).

The % recovery of intact insulin after the insulin solution stands for a period of time is recorded in Tables 2 and 3 and graphically illustrated in FIGS. 13(a) and (b). It is observed that insulin solutions with sodium caprate are significantly more stable than the insulin samples without sodium caprate or the solutions with capric acid. For insulin samples that contain sodium caprate, at least 90% of insulin is recovered after the solutions stands at 37° C. for at least about 24 hours.

TABLE 1 Formulation of insulin solutions for Example 11 10 U/mL Bovine Insulin in ‘blank’ simulated 10 U/mL Bovine Insulin in ‘blank’ 10 U/mL Bovine Insulin in ‘blank’ intestinal simulated intestinal buffer pH 8 simulated intestinal buffer pH 8 buffer pH 8 Plus Sodium Caprate Plus Capric Acid 1 3 5 7 9 Sample Control 2 Control 4 Control 6 Control 8 Control 10 Sodium 30 mg/ml 30 mg/ml 60 mg/ml 60 mg/ml Caprate Capric 30 mg/ml 30 mg/ml 60 mg/ml 60 mg/ml Acid Stirring No Yes No Yes No Yes No Yes No Yes

TABLE 2 Bovine insulin fibrillation results with or without C10 % Recovery % Recovery % Recovery % Recovery % Recovery % Recovery T = T = T = T = T = T = Sample 0 6 24 29 48 72 1. Insulin Standing 100.24 100.00 100.25 100.28 100.29 100.29 2. Insulin Stirring 100.11 100.08 99.42 1.50 0.29 0.22 3. 30 mg/ml C10 98.15 97.85 97.88 97.39 97.15 96.54 Standing 4. 30 mg/ml C10 98.06 97.66 97.74 97.44 97.58 97.05 Stirring 5. 60 mg/ml C10 94.52 94.24 93.99 93.61 93.24 92.12 Standing 6. 60 mg/ml C10 94.79 94.52 94.41 93.98 93.29 92.79 Stirring

TABLE 3 Bovine insulin fibrillation results with or without Capric acid Bovine Insulin Fibrillation Expt. Results Summary % Recovery % Recovery % Recovery % Recovery % Recovery % Recovery T = T = T = T = T = T = Sample 0 6 24 29 48 72 1. Insulin Standing 100.24 100.00 100.25 100.28 100.29 100.29 2. Insulin Stirring 100.11 100.08 99.42 1.50 0.29 0.22 7. 30 mg/ml CA 93.73 68.88 15.32 3.26 1.10 0.44 Standing 8. 30 mg/ml CA 95.06 0.27 0.20 0.24 0.23 0.16 Stirring 9. 60 mg/ml CA 91.96 67.73 19.75 2.59 1.43 2.24 Standing 10. 60 mg/ml CA 91.38 0.27 0.19 0.09 0.13 0.10 Stirring Note: Recovery values based on the theoretical amount spiked = 0.344591 mg/ml

The foregoing is illustrative of the present invention and is not to be construed as limiting thereof. Although a few exemplary embodiments of this invention have been described, those skilled in the art will readily appreciate that many modifications are possible in the exemplary embodiments without materially departing from the novel teachings and advantages of this invention. Accordingly, all such modifications are intended to be included within the scope of this invention as defined in the claims. Therefore, it is to be understood that the foregoing is illustrative of the present invention and is not to be construed as limited to the specific embodiments disclosed, and that modifications to the disclosed embodiments, as well as other embodiments, are intended to be included within the scope of the appended claims. The invention is defined by the following claims, with equivalents of the claims to be included therein.

Claims

1. A composition comprising a therapeutically effective amount of at least one peptide, polypeptide, protein, an analog or derivative thereof and a sufficient amount of at least one stabilizing agent to improve the stability of the peptide, polypeptide, protein, analog or derivative thereof,

wherein at least one stabilizing agent is a salt, an ester, an ether, or a derivative of a medium chain fatty acid and having a carbon chain length of from about 4 to about 20 carbon atoms or is a surface active agent.

2. The composition of claim 1, wherein at least 90% of the peptide, polypeptide, protein, analog or derivative thereof is retained after the composition stands at 37° C. for at least about 24 hours.

3. The composition of claim 1, wherein at least one peptide, polypeptide, protein, analog or derivative thereof is insulin, or an analog or derivative thereof.

4. The composition of claim 1, wherein at least one peptide, polypeptide, protein, analogs or derivative thereof is glucagon-Like Peptide (GLP), or an analog or derivative thereof.

5. The composition of claim 4, wherein at least one GLPs is selected from the group consisting of GLP-1, GLP-1 analogs and derivatives thereof, GLP-2, GLP-2 analogs and derivatives thereof, exendin-4, analogs and derivatives thereof.

6. The composition of claim 1, wherein at least one peptide, polypeptide, protein, an analog or derivative thereof is selected from the group consisting of GnRH agonists and antagonists, somatostatin, ACTH, corticotropin-releasing factor, angiotensin, calcitonin, gastric inhibitory peptide, growth hormone-releasing factor, pituitary adenylate, exendin, exendin-3, cyclase activating peptide, secretin, enterogastrin, somatostatin, somatotropin, somatomedin, parathyroid hormone, thrombopoietin, erythropoietin, hypothalamic releasing factors, prolactin, thyroid stimulating hormones, endorphins, enkephalins, vasopressin, oxytocin, opioids and analogues thereof, superoxide dismutase, interferon, asparaginase, arginase, arginine deaminase, adenosine deaminase, ribonuclease, FVII, FXIII, a mixture of FVII and FXIII, IL-20, IL-21, IL-28a, IL-29, IL-31, and analogs and derivatives thereof.

7. The composition of claim 1, which is solid at room temperature.

8. The composition of claim 1, which is in a solid oral dosage form.

9. The composition of claim 1, wherein the stabilizing agent is a salt of a medium chain fatty acid and has a carbon chain length of from about 8 to 14 carbon atoms.

10. The composition of claim 1, wherein the stabilizing agent is selected from the group consisting of sodium caprylate, sodium caprate and sodium laurate.

11. The composition of claim 1, wherein the concentration of the stabilizing agent in the composition is equal to or above the critical micelle concentration of the stabilizing agent in the composition.

12. The composition of claim 1, wherein the dissolution rate of the stabilizing agent and the peptide, polypeptide, protein, analogs or derivatives thereof in the composition are substantially the same.

13. The composition of claim 1, further comprising one or more excipients selected from the group consisting of rate-controlling polymeric materials, diluents, lubricants, disintegrants, plasticizers, anti-tack agents, opacifying agents, pigments, and flavorings.

14. The composition of claim 13, wherein at least one rate-controlling polymer is a polymer derived from acrylic or methacrylic acid, esters or copolymers derived from acrylic or methacrylic acid.

15. The composition of claim 1, further comprising an enteric coating.

16. The composition of claim 15, wherein the enteric coating comprises at least one polymer selected from the group consisting of poly(acrylic acid), polyacrylate, poly(methacrylic acid), polymethacrylate, and mixtures thereof.

17. The composition of claim 15, wherein the enteric coated composition is a tablet or capsule.

18. The composition of claim 1, wherein the composition is in a form selected from the group consisting of a multiparticulate form, a sustained-release form and an instant release form.

19. The composition of claim 1, further comprising at least one diluent which is an inert filler chosen from microcrystalline cellulose, lactose, dibasic calcium phosphate and saccharides.

20. The composition of claim 19, wherein the inert filler is at least one lactose which is lactose monohydrate or lactose anhydrous.

21. The composition of claim 20, wherein the inert filler is at least one saccharide selected from the group consisting of mannitol, starch, sorbitol, sucrose, and glucose.

22. The composition of claim 1, further comprising at least one lubricant selected from the group consisting of colloidal silicon dioxide, talc, magnesium stearate, calcium stearate, and stearic acid.

23. The composition of claim 1, further compriseing at least one disintegrant selected from the group consisting of lightly crosslinked polyvinylpyrrolidone, corn starch, potato starch, maize starch and modified starches, croscarmellose sodium, crospovidone, and sodium starch glycolate.

24. A method for preparation of a composition of a peptide, polypeptide, protein, an analog or derivative thereof, wherein the process comprises mixing the peptide, polypeptide, protein analogs or derivatives thereof with a sufficient amount of at least one stabilizing agents to improve the stability of peptide, polypeptide, protein, an analog or derivative thereof, and the agent is a salt, an ester, an ether, or a derivative of a medium chain fatty acid and having a carbon chain length of from about 4 to about 20 carbon atoms, or is a surface active agent.

25. The methods of claim 24, wherein at least 90% of the peptide, polypeptide, protein, analog or derivative thereof is retained after the composition stands at 37° C. for at least about 24 hours.

26. The method of claim 24, wherein the stabilizing agent is a medium chain fatty acid salt.

27. The method of claim 24, wherein the stabilizing agent is a medium chain fatty acid salt having a carbon chain length of from about 8 to about 14 carbon atoms.

28. The method of claim 24, wherein the concentration of the stabilizing agent in the composition is equal or above the critical micelle concentration of the stabilizing agent in the composition.

Patent History
Publication number: 20090280169
Type: Application
Filed: May 7, 2009
Publication Date: Nov 12, 2009
Applicant:
Inventor: Thomas W. Leonard (Wilmington, NC)
Application Number: 12/436,990
Classifications
Current U.S. Class: Coated Capsules (424/463); Containing Solid Synthetic Polymers (424/482); 514/2
International Classification: A61K 9/48 (20060101); A61K 9/32 (20060101); A61K 38/00 (20060101);