NZ750950B2 - Omecamtiv Mecarbil and Salts Thereof and Their Uses - Google Patents

Omecamtiv Mecarbil and Salts Thereof and Their Uses Download PDF

Info

Publication number
NZ750950B2
NZ750950B2 NZ750950A NZ75095014A NZ750950B2 NZ 750950 B2 NZ750950 B2 NZ 750950B2 NZ 750950 A NZ750950 A NZ 750950A NZ 75095014 A NZ75095014 A NZ 75095014A NZ 750950 B2 NZ750950 B2 NZ 750950B2
Authority
NZ
New Zealand
Prior art keywords
acid
cardiac
omecamtiv mecarbil
cardiac muscle
pharmaceutically acceptable
Prior art date
Application number
NZ750950A
Other versions
NZ750950A (en
Inventor
Mingda Bi
Robert Kuehl
Original Assignee
Amgen Inc
Cytokinetics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc, Cytokinetics Inc filed Critical Amgen Inc
Priority claimed from NZ711225A external-priority patent/NZ711225A/en
Publication of NZ750950A publication Critical patent/NZ750950A/en
Publication of NZ750950B2 publication Critical patent/NZ750950B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/38Cellulose; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates

Abstract

The cardiac sarcomere is the basic unit of muscle contraction in the heart, and is a highly ordered cytoskeletal structure composed of cardiac muscle myosin, actin and a set of regulatory proteins. The discovery and development of small molecule cardiac muscle myosin activators would lead to promising treatments for acute and chronic heart failure. Cardiac muscle myosin is the cytoskeletal motor protein in the cardiac muscle cell. It is directly responsible for converting chemical energy into the mechanical force, resulting in cardiac muscle contraction. Current positive inotropic agents, such as beta-adrenergic receptor agonists or inhibitors of phosphodiesterase activity, increase the concentration of intracellular calcium, thereby increasing cardiac sarcomere contractility. However, the increase in calcium levels increase the velocity of cardiac muscle contraction and shortens systolic ejection time, which has been linked to potentially life-threatening side effects. In contrast, cardiac muscle myosin activators work by a mechanism that directly stimulates the activity of the cardiac muscle myosin motor protein, without increasing the intracellular calcium concentration. They accelerate the rate-limiting step of the myosin enzymatic cycle and shift it in favour of the force-producing state. Rather than increasing the velocity of cardiac contraction, this mechanism instead lengthens the systolic ejection time, which results in increased cardiac muscle contractility and cardiac output in a potentially more oxygen-efficient manner. Omecamtiv mecarbil is a first in class direct activator of cardiac myosin, the motor protein that causes cardiac contraction. It is being evaluated as a potential treatment of heart failure in both intravenous and oral formulations with the goal of establishing a new continuum of care for patients in both the in-hospital and outpatient settings. Clinical trials providing an I.V. delivery of omecamtiv mecarbil have shown that plasma levels of the drug can be delivered safely and effectively. However, standard release formulations and some extended release formulations gave peak to trough ratios that may be too great to provide a safe and effective amount of omecamtiv mecarbil to patients who need the drug in a chronic or preventative setting. Accordingly, an effective sustained release formulation would be desirable for increased patient safety and effectiveness. Therefore, in one embodiment, there is provided a pharmaceutical formulation comprising omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically acceptable hydrate, or a pharmaceutically acceptable hydrate of a pharmaceutically acceptable salt thereof, such as a dihydrochloride monohydrate salt of omecamtiv mecarbil. ing treatments for acute and chronic heart failure. Cardiac muscle myosin is the cytoskeletal motor protein in the cardiac muscle cell. It is directly responsible for converting chemical energy into the mechanical force, resulting in cardiac muscle contraction. Current positive inotropic agents, such as beta-adrenergic receptor agonists or inhibitors of phosphodiesterase activity, increase the concentration of intracellular calcium, thereby increasing cardiac sarcomere contractility. However, the increase in calcium levels increase the velocity of cardiac muscle contraction and shortens systolic ejection time, which has been linked to potentially life-threatening side effects. In contrast, cardiac muscle myosin activators work by a mechanism that directly stimulates the activity of the cardiac muscle myosin motor protein, without increasing the intracellular calcium concentration. They accelerate the rate-limiting step of the myosin enzymatic cycle and shift it in favour of the force-producing state. Rather than increasing the velocity of cardiac contraction, this mechanism instead lengthens the systolic ejection time, which results in increased cardiac muscle contractility and cardiac output in a potentially more oxygen-efficient manner. Omecamtiv mecarbil is a first in class direct activator of cardiac myosin, the motor protein that causes cardiac contraction. It is being evaluated as a potential treatment of heart failure in both intravenous and oral formulations with the goal of establishing a new continuum of care for patients in both the in-hospital and outpatient settings. Clinical trials providing an I.V. delivery of omecamtiv mecarbil have shown that plasma levels of the drug can be delivered safely and effectively. However, standard release formulations and some extended release formulations gave peak to trough ratios that may be too great to provide a safe and effective amount of omecamtiv mecarbil to patients who need the drug in a chronic or preventative setting. Accordingly, an effective sustained release formulation would be desirable for increased patient safety and effectiveness. Therefore, in one embodiment, there is provided a pharmaceutical formulation comprising omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically acceptable hydrate, or a pharmaceutically acceptable hydrate of a pharmaceutically acceptable salt thereof, such as a dihydrochloride monohydrate salt of omecamtiv mecarbil.

Description

OMECAMTIV MECARBIL AND SALTS THEREOF AND THEIR USES CROSS-REFERENCE TO D APPLICATIONS The benefit of U.S. Provisional ation No. 61/785,763, filed March 14, 2014 is claimed, the disclosure of which is incorporated by reference in its entirety.
FIELD Provided is a ceutical formulation comprising omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically acceptable hydrate, or a pharmaceutically acceptable e of a pharmaceutically acceptable salt f, such as omecamtiv mecarbil dihydrochloride hydrate.
BACKGROUND The cardiac sarcomere is the basic unit of muscle contraction in the heart. The cardiac sarcomere is a highly ordered cytoskeletal structure ed of cardiac muscle myosin, actin and a set of regulatory proteins. The discovery and development of small molecule cardiac muscle myosin activators would lead to promising treatments for acute and chronic heart failure. Cardiac muscle myosin is the cytoskeletal motor protein in the cardiac muscle cell. It is directly responsible for converting chemical energy into the mechanical force, resulting in c muscle contraction.
Current positive inotropic agents, such as drenergic receptor agonists or inhibitors of phosphodiesterase activity, increase the concentration of intracellular calcium, thereby increasing cardiac sarcomere contractility. However, the increase in calcium levels increase the velocity of cardiac muscle contraction and shortens systolic ejection time, which has been linked to potentially life-threatening side effects. In contrast, cardiac muscle myosin activators work by a mechanism that directly stimulates the activity of the cardiac muscle myosin motor protein, without sing the intracellular calcium concentration. They accelerate the rate-limiting step of the myosin enzymatic cycle and shift it in favor of the producing state.
Rather than increasing the velocity of cardiac contraction, this mechanism instead lengthens the systolic ejection time, which s in increased cardiac muscle contractility and cardiac output in a potentially more -efficient manner.
US. Patent No. 7,507,735, herein incorporated by nce, discloses a genus of compounds, including tiv mecarbil (AMG 423, CK—1827452), having the structure: Me02C\ Me V ; ‘N N H H Omecamtiv mecarbil is a first in class direct activator of cardiac myosin, the motor protein that causes cardiac contraction. It is being evaluated as a potential treatment of heart failure in both intravenous and oral formulations with the goal of establishing a new continuum of care for patients in both the in—hospital and outpatient gs.
Clinical trials providing an I.V. delivery of omecamtiv mecarbil have shown that plasma levels of the drug can be delivered safely and effectively. r, standard release formulations and some extended release formulations gave peak to trough ratios that may be too great to provide a safe and effective amount of omecamtiv mecarbil to patients who need the drug in a chronic or preventative setting (See, Figure 4). Accordingly, an effective sustained release formulation would be desirable for increased patient safety and effectiveness.
SUMMARY Provided is a ceutical formulation comprising: omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically acceptable hydrate, or a pharmaceutically acceptable hydrate of a pharmaceutically acceptable salt f; a control release agent; a pH modifying agent; a ; and a lubricant.
Also provided is a process for making a pharmaceutical formulation sing: blending a mixture comprising omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically able hydrate, or a pharmaceutically able hydrate of a ceutically acceptable salt thereof, a control release agent, a pH modifying agent, and a filler; lubricating the blended mixture using a lubricant; granulating the lubricated blend; WO 52236 lubricating the resultant ation using the lubricant; and compressing the lubricated granulation into desired form.
Also provided is a method of treating a e selected from acute heart failure and chronic heart failure, comprising administering a pharmaceutical formulation described herein to a patient in need thereof.
DESCRIPTION OF THE FIGURES Figure l is a ?ow diagram for the preparation of immediate release (IR) tablets of omecamtiv mecarbil (25 mg); see Example 1.
Figure 2 is a ?ow diagram for the preparation of matrix modified release itions; see Example 2.
Figure 3 is a ?ow diagram for the ation of matrix modified release compositions; see, Examples 3—5.
Figure 4 shows the exposure of healthy volunteers (plasma concentration (ng/ml) V. time (h)), fasted (top) and fed (bottom) for an immediate release composition (IR) and two matrix modified release compositions (MTX—Fl and MTX—F2); the study was a ized, open—label, 4—way crossover lete block design study in healthy adult subjects: 0 60 subjects; 1 site in the US 0 12 total ents (each treatment taken 20 times) 0 Various formulations; Each taken fasted or fed 0 Each subject will be randomized to 1 sequence 0 Each subject receives 4 out of 12 possible treatments 0 Each period ~7 days; Study duration: 27 days (Period 4: 5 days).
Figure 5 is a table with data for an immediate release composition (IR) and two matrix modified e compositions (MTX—Fl and MTX—F2).
Figure 6 shows drug release at two pHs (2 and 6.8) for a matrix formulation of omecamtiv mecarbil free base (top) and for a omecamtiv mecarbil dihydrochloride hydrate salt form, Form A (bottom).
Figure 7 shows an X—ray powder diffraction pattern (XRPD) for Form A.
Figure 8 shows an XRPD of a omecamtiv mecarbil dihydrochloride hydrate salt form at varying relative ty conditions.
Figure 9 shows an XRPD of a omecamtiv mecarbil dihydrochloride hydrate salt form at varying atures.
Figure 10 shows an overlay of XRPD patterns for Forms A, B and C of omecamtiv mecarbil dihydrochloride salt.
DETAILED DESCRIPTION Unless otherwise specified, the following definitions apply to terms found in the specification and claims: “Treatment” or “treating” means any treatment of a disease in a patient, including: a) preventing the disease, that is, causing the clinical ms of the disease not to develop; b) inhibiting the disease; c) slowing or arresting the development of clinical symptoms; and/or d) relieving the disease, that is, causing the regression of clinical symptoms. Treatment of diseases and disorders herein is intended to also include the prophylactic administration of a pharmaceutical formulation described herein to a subject (i.e., an animal, ably a mammal, most preferably a human) believed to be in need of preventative treatment, such as, for example, c heart failure.
The term peutically effective amount” means an amount effective, when administered to a human or non—human patient, to treat a disease, e.g., a therapeutically effective amount may be an amount sufficient to treat a disease or disorder responsive to myosin activation. The therapeutically effective amount may be ascertained experimentally, for example by assaying blood concentration of the chemical , or theoretically, by ating bioavailability.
“Pharmaceutically acceptable salts” include, but are not d to salts with inorganic acids, such as hlorate (i.e., hydrochloride), phosphate, diphosphate, hydrobromate, sulfate, sulfinate, nitrate, and like salts; as well as salts with an organic acid, such as malate, maleate, te, tartrate, succinate, citrate, acetate, lactate, methanesulfonate, enesulfonate, 2—hydroxyethylsulfonate, benzoate, salicylate, stearate, and alkanoate such as acetate, HOOC——(CH2)n——COOH where n is 0—4, and like salts.
Similarly, pharmaceutically acceptable cations include, but are not limited to sodium, potassium, calcium, aluminum, lithium, and ammonium. Those skilled in the art will recognize various synthetic methodologies that may be used to prepare xic pharmaceutically acceptable addition salts.
The term “hydrate” refers to the chemical entity formed by the interaction of water and a compound, including, for example, hemi—hydrates, monohydrates, dihydrates, trihydrates, etc.
“Crystalline form,” “polymorph,” and “novel form” may be used interchangeably herein, and are meant to include all crystalline and amorphous forms of the compound, ing, for example, polymorphs, pseudopolymorphs, solvates, hydrates, unsolvated polymorphs (including anhydrates), conformational polymorphs, and amorphous forms, as well as mixtures thereof, unless a particular crystalline or amorphous form is referred to.
The specification and claims contain listing of species using the language “selected from . . . and . . .” and “is . . . or . . .” imes referred to as Markush groups). When this ge is used in this application, unless otherwise stated it is meant to include the group as a whole, or any single members thereof, or any subgroups thereof. The use of this language is merely for shorthand purposes and is not meant in any way to limit the removal of individual ts or subgroups as needed. ed is a pharmaceutical formulation comprising omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically able hydrate, or a pharmaceutically acceptable e of a pharmaceutically acceptable salt thereof, such as omecamtiv mecarbil dihydochloride hydrate.
The pharmaceutical formulations described herein are e of releasing omecamtiv mecarbil evenly at a pace controlled by the diffusion of omecamtiv mecarbil through a gel layer formed by the hydration of the control release agents in the s. In some embodiments, in conjunction with other above or below ments, the present modified release matrix tablets demonstrate a minimal pH—dependent release in—vitro. In some embodiments, in conjunction with other above or below embodiments, te release of omecamtiv mecarbil is achieved in both pH 2 and 6.8 dissolution medium within 24 hours, possibly resulting in less inter— and intra— subject ility and food effect. It is found that the present modified release matrix tablet dosage form is or to the former immediate release dosage form in minimizing the plasma peak—trough ratio. As a result, the t modified e matrix tablets reduce plasma concentration ?uctuation, leading to reduced side effects, and improved safety and efficacy. It is also expected that the present modified release matrix tablets will improve patient compliance by reducing the dosing frequency.
Additionally, the present modified release matrix tablets are ochemically stable— resulting in no al attribute, assay, impurity, or dissolution profile changes after storage at 40 OC/75%RH for 6 months.
In some embodiments, in ction with other above or below embodiments, the re of omecamtiv mecarbil from two to twelve hours after dosing in humans is between 40 and 70 ng/ml.
In some embodiments, in conjunction with other above or below embodiments, the exposure of omecamtiv mecarbilfrom two to twelve hours after dosing in humans remains between 40 and 55 ng/ml.
In some embodiments, in conjunction with other above or below embodiments, the omecamtiv mecarbil is released in the following intervals: S 30% dose dissolved at 1 hour; —75% dose dissolved at 3 hours; and Z 80% dose dissolved at 12 hours.
In some embodiments, in conjunction with other above or below embodiments, the omecamtiv il is released in the following intervals: S 30% dose dissolved at 2 hours; —75% dose dissolved at 6 hours; and Z 80% dose dissolved at 16 hours.
Provided is a pharmaceutical formulation sing: omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically able hydrate, or a ceutically acceptable hydrate of a ceutically acceptable salt thereof; a control release agent; a pH modifying agent; a filler; and a lubricant.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about 3—30% w/w of omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically acceptable e, or a pharmaceutically acceptable hydrate of a pharmaceutically acceptable salt thereof; —35% w/w control release agent; —45% w/w pH modifying agent; —65% w/w filler; and 0.1—1.0% w/w lubricant.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about 12—25 (w/w%) omecamtiv mecarbil Di—HCl hydrate; 25—35 (w/w%) methocelTM K100 M Prem CR; 20—30 (w/w%) microcrystalline cellulose, PH 102; 5—10 (w/w%) e monohydrate, FF 316; 12—25(w/w%) fumaric acid; 0.1—2 (w/w%) intra—granular magnesium stearate; and 0.1—2 (w/w%) extra—granular magnesium stearate.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about: 3—10 (w/w%) omecamtiv mecarbil Di—HCl hydrate; 20—40 (w/w%) methocelTM K100 M Prem CR; 30—42 (w/w%) microcrystalline cellulose, PH 102; 12—25 (w/w%) lactose monohydrate, FF 316; 4—11 (w/w%) c acid; 0.1—2 (w/w%) intra—granular magnesium stearate; and 0.1—2 (w/w%) extra—granular magnesium stearate.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about: 12—25 (w/w%) omecamtiv mecarbil Di—HCl hydrate; 1—10 (w/w%) elTM K100 M Prem CR; 12—27 (w/w%) methocelTM K100 LV Prem CR; 20—35 (w/w%) rystalline cellulose, PH 102; 4—15 (w/w%) lactose monohydrate, FF 316; 12—25 (w/w%) c acid; 0.1—2 (w/w%) intra—granular magnesium stearate; and 0.1—2 (w/w%) extra—granular magnesium stearate.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about: 3—10 (w/w%) omecamtiv mecarbil Di—HCl hydrate; 1—10 (w/w%) methocelTM K100 M Prem CR; 12—27 (w/w%) methocelTM K100 LV Prem CR; 30—50 (w/w%) rystalline cellulose, PH 102; 15—25 (w/w%) lactose monohydrate, FF 316; 3—11 (w/w%) fumaric acid; 0.1—2 (w/w%) intra—granular magnesium stearate; and 0.1—2 (w/w%) extra—granular ium te.
In some ments, in conjunction with other above or below embodiments, the pharmaceutical formulation ses about: 18—19 (w/w%) omecamtiv mecarbil Di—HCl hydrate; 28—32 (w/w%) methocelTM K100 M Prem CR; 23—26 (w/w%) microcrystalline cellulose, PH 102; 7—9 (w/w%) lactose monohydrate, FF 316; 17—20 (w/w%) fumaric acid; 0.1—1 (w/w%) intra—granular ium stearate; and 0.1—1 (w/w%) extra—granular magnesium stearate.
In some ments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about: —7 (w/w%) omecamtiv mecarbil Di—HCl hydrate; 27—33 (w/w%) methocelTM K100 M Prem CR;35—38 (w/w%) microcrystalline cellulose, PH 102;17—20 (w/w%) lactose monohydrate, FF 316;6—9 (w/w%) fumaric acid;0.1—1 (w/w%) intra—granular magnesium stearate; and0.1—l (w/w%) granular magnesium te.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation ses about: 17—20 (w/w%) omecamtiv mecarbil Di—HCl hydrate;3—7 (w/w%) methocelTM K100 M Prem CR;18—22 (w/w%) methocelTM K100 LV Prem CR; 26—30 (w/w%) microcrystalline cellulose, PH 102;8—1l (w/w%) lactose monohydrate, FF 316;17—20 (w/w%) fumaric acid;0.1—1 (w/w%) intra—granular magnesium stearate; and0.1—l (w/w%) extra—granular magnesium stearate.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about: —7 (w/w%) omecamtiv mecarbil Di—HCl hydrate;3—7 (w/w%) methocelTM K100 M Prem CR;18—22 (w/w%) methocelTM K100 LV Prem CR; 37—43 (w/w%) microcrystalline ose, PH 102;18—22 (w/w%) lactose monohydrate, FF 316;6—9 (w/w%) fumaric .1— 1 (w/w%) intra—granular magnesium stearate; and0.1—1 (w/w%) extra—granular magnesium stearate.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about: 18.37 (w/w%) tiv mecarbil Di—HCl e;30 (w/w%) methocelTM K100 M Prem CR;24.20 (w/w%) rystalline ose, PH 102;8.07 (w/w%) lactose monohydrate, FF 316;18.37 (w/w%) fumaric acid;0.5 (w/w%) intra—granular magnesium stearate; and0.5 (w/w%) extra—granular ium stearate.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about: 6.13 (w/w%) omecamtiv mecarbil Di—HCl hydrate;30 (w/w%) methocelTM K100 M Prem CR;36.81 (w/w%) microcrystalline cellulose, PH 102;18.40 (w/w%) lactose monohydrate, FF 3l6;7.66 (w/w%) fumaric acid;0.5 (w/w%) intra—granular magnesium stearate; and0.5 (w/w%) extra—granular magnesium te.
In some embodiments, in ction with other above or below embodiments, the pharmaceutical formulation comprises about: 18.37 (w/w%) omecamtiv il Di—HCl hydrate;5 (w/w%) methocelTM K100 M Prem CR;20 (w/w%) elTM K100 LV Prem CR; 27.95 (w/w%) microcrystalline cellulose, PH 31 (w/w%) lactose monohydrate, FF 316;18.37 (w/w%) fumaric acid;0.5 (w/w%) intra—granular magnesium stearate; and0.5 (w/w%) extra—granular magnesium stearate.
In some embodiments, in conjunction with other above or below embodiments, the pharmaceutical formulation comprises about: 6.13 (w/w%) tiv mecarbil Di—HCl hydrate;5 (w/w%) methocelTM K100 M Prem CR;20 (w/w%) methocelTM K100 LV Prem CR; 40.14 (w/w%) microcrystalline cellulose, PH 102;20.07 (w/w%) lactose monohydrate, FF 3l6;7.66 (w/w%) fumaric acid;0.5 (w/w%) granular magnesium stearate; and0.5 (w/w%) extra—granular magnesium Omecamtiv Mecarbil In some embodiments, in conjunction with other above or below embodiments, the drug formulation comprises omecamtiv mecarbil dihydrochloride salt. In some embodiments, in conjunction with other above or below embodiments, the drug formulation comprises tiv mecarbil ochloride hydrate. In some embodiments, in conjunction with other above or below embodiments, the drug formulation comprises omecamtiv mecarbil dihydrochloride hydrate Form A.
In some embodiments, in conjunction with other above or below embodiments, Form A can be characterized by an X—ray powder diffraction pattern, obtained as set forth in the Examples, having peaks at about 6.6, 14.9, 20.1, 21.4, and 26.8 i 0.20 20 using Cu KOL radiation. Form A optionally can be further characterized by an X—ray powder diffraction pattern having additional peaks at about 8.4, 24.2, 26.0, 33.3 i 0.20 20 using Cu Ka radiation.
Form A optionally can be even further characterized by an X—ray powder diffraction pattern having additional peaks at about 6.2, 9.7, 13.2, 14.3, 15.4, 16.3, 16.9, 18.9, 19.5, 20.7, 21.8, 22.8, 23.6, 25.1, 27.3, 27.7, 28.4, 29.4, 30.2, 31.2, 31.5, 31.9, 33.9, 34.5, 34.9, 36.1, 36.8, 2014/027104 37.7, 38.5, and 39.7i 0.20 20 using Cu Ka radiation. In various cases, Form A can be characterized by an XRPD pattern having peaks at about 6.2, 6.6, 8.4, 9.7, 13.2, 14.3, 14.9, .4, 16.3, 16.9, 18.9, 19.5, 20.1, 20.7, 21.4, 21.8, 22.8, 23.6, 24.3, 25.1, 26.0, 26.8, 27.3, 27.7, 28.4, 29.4, 30.2, 31.2, 31.5, 31.9, 33.3, 33.9, 34.5, 34.9, 36.1, 36.8, 37.7, 38.5, and 39.7i 0.20 20 using Cu Ka radiation. In some embodiments, in conjunction with other above or below ments, Form A can be characterized by an X—ray powder diffraction pattern substantially as depicted in Figure 7 wherein by “substantially” is meant that the reported peaks can vary by about i0.2°. It is well known in the field of XRPD that while relative peak heights in spectra are dependent on a number of factors, such as sample preparation and instrument geometry, peak positions are relatively insensitive to experimental details.
Form B and Form C polymorphs of omecamtiv mecarbil, are metastable anhydrous dihydrochloride forms, and can be formed under varied hydration conditions and temperatures, as noted in Figure 8 and 9. Characteristic Form B 2—theta values include 6.8, 8.8, 14.7, 17.7, and 22.3i 0.20 20 using Cu Ka radiation, and can additionally e peaks at 9.6, 13.5, 19.2, 26.2i 0.20 20 using Cu Ka radiation. Form B can be characterized with XRPD pattern peaks at 6.2, 6.8, 8.8, 9.6, 13.5, 14.4, 14.7, 15.4, 16.3, 17.0, 17.7, 18.3, 19.2, 19.9, 20.5, 20.8, 21.8, 22.3, 22.7, 23.0, 24.8, 25.1, 25.5, 26.2, 26.4, 26.8, 27.5, 28.5, 30.2, .6, 31.1, 31.5, 32.1, 32.7, 34.1, 34.4, 35.5, 35.9, 38.1, 38.9i 0.20 20 using Cu Ka radiation. teristic Form C 2—theta values include 6.7, 14.8, 17.4, 20.6, and 26.2i 0.20 20 using Cu Ka radiation, and can additionally include peaks at 8.7, 22.0, 27.1, and 27.7i 0.20 20 using Cu Ka radiation. Form C can be characterized with XRPD pattern peaks at 6.2, 6.7, 8.7, 9.6, 13.5, 14.5, 14.8, 15.4, 16.4, 17.1, 17.4, 18.4, 19.3, 19.5, 19.9, 20.6, 20.8, 21.8, 22.0, 22.5, 22.8, 24.3, 24.7, 25.1, 25.6, 26.2, 26.5, 27.1, 27.3, 27.7, 28.5, 30.0, 30.5, 31.0, 31.5, 32.2, 32.8, 34.1, 35.2, 36.0, 36.9, and 38.8i 0.20 20 using Cu Ka radiation.
See, also, Figure 9 (variable temperature XRPD data), Figure 8 ble relative humidity XRPD data), and Figure 10 (overlay) Control e Agent As used herein, the term “control release agents” refer to agents that facilitate the release of the active ingredient from the present composition in a controlled n. In some embodiments, in ction with other above or below embodiments, the control release agents form a gel upon hydration. Control release agents include pulluan, dextrin, sodium and calcium acid, polyacrylic acid, polymethacrylic acid, thylvinylether co—maleic WO 52236 anhydride, polyvinylpyrrolidone, polyethylene oxide, polyethylene glycol, hydroxypropylcellulose, hydroxypropylmethylcellulose, yethylcellulose, hydroxymethyl methacrylate, sodium carboxymethylcellulose, calcium carboxymethylcellulose, methylcellulose, maltodextrin, xanthan gum, tragacanth gum, agar, gellan gum, kayara gum, alginic acids, pectins, latinized starch, polyvinyl alcohol, carboxymethylethylcellulose, cellulose acetate phthalate, cellulose acetate succinate, methylcellulose phthate, hydroxymethylethylcellulosephthate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose acetate succinate, polyvinyl alcohol phthalate, polyvinyl butylate phthalate, polyvinyl actal ate, a copolymer of vinyl e/maleic anhydride, a copolymer of styrene/maleic acid monoester, a copolymer of methyl acryl— ate/methacrylic acid, a copolymer of styrene/acrylic acid, a copolymer of methyl acrylate/methacrylic acid/octyl acrylate, a mer of methacrylic ethyl methacrylate, benzylaminomethylcellulose, laminomethylcellulose, piperidylethylhydroxyethylcellulose, cellulose acetate dimethylaminoacetate, a copolymer of vinyl diethylamine/vinyl acetate, a copolymer of vinyl benzylamine/vinyl acetate, nyl acetaldiethylamino acetate, a copolymer of iperidylacetoacetal/vinyl acetate, polydiethylaminomethylstyrene, a copolymer of methyl methacrylate/butyl methacrylate/dimethylaminoethyl methacrylate and polydimethylaminoethylmethacrylate, a copolymer of 2—methyl—5—vinylpyridine/methylmethacrylate/methacrylic acid, a copolymer of 2—methyl—5—vinylpyridine/methyl acrylate/methacrylic acid, a copolymer of 2—vinyl—5— ethylpyridine/methacrylic acid/methy acrylate, a copolymer of 2—vinylpyrid—ine/methacrylic acid/acrylonitrile, carboxymethylpiperidyl starch, carboxy—methylbenzylaminocellulose, a copolymer of N—vinylglycine/styrene, chitosan, poly(vinyl alcohol), maleic anhydride copolymer, poly (vinyl pyrolidone), starch and starch—based polymers, poly (2—ehtyl—2— oxazoline), thyleneimine), polyurethane hydrogels, welan gum, rhamsan gum, polyvinyl acetates, ethylcellulose, eudragit RL, RS, NE 30D, oat EMM 30D, or combinations f.
In some embodiments, in conjunction with other above or below embodiments, the control release agent is a polymer.
In some embodiments, in conjunction with other above or below embodiments, the control release agent is selected from pulluan, dextrin, sodium and calcium acid, polyacrylic acid, polymethacrylic acid, polymethylvinylether co—maleic anhydride, polyvinylpyrrolidone, polyethylene oxide, polyethylene glycol, hydroxypropylcellulose, hydroxypropylmethylcellulose, yethylcellulose, ymethyl methacrylate, sodium carboxymethylcellulose, calcium carboxymethylcellulose, methylcellulose, maltodextrin, xanthan gum, tragacanth gum, agar, gellan gum, kayara gum, alginic acids, pectins, pre— gelatinized starch, polyvinyl alcohol, carboxymethylethylcellulose, cellulose acetate phthalate, cellulose acetate succinate, methylcellulose phthate, hydroxymethylethylcellulosephthate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose acetate ate, nyl l phthalate, polyvinyl butylate phthalate, polyvinyl actal phthalate, a copolymer of vinyl acetate/maleic anhydride, a copolymer of styrene/maleic acid monoester, a copolymer of methyl acryl—ate/methacrylic acid, a copolymer of styrene/acrylic acid, a copolymer of methyl te/methacrylic acid/octyl acrylate, a copolymer of methacrylic acid/methyl methacrylate, benzylaminomethylcellulose, diethylaminomethylcellulose, piperidylethylhydroxyethylcellulose, cellulose e dimethylaminoacetate, a copolymer of vinyl diethylamine/vinyl acetate, a copolymer of vinyl benzylamine/vinyl acetate, polyvinyl acetaldiethylamino acetate, a mer of vinylpiperidylacetoacetal/vinyl acetate, polydiethylaminomethylstyrene, a copolymer of methyl methacrylate/butyl methacrylate/dimethylaminoethyl methacrylate and polydimethylaminoethyl methacrylate, a copolymer of y—5vinylpyrid-Iine/methylmethacryl-Iate/methacrylic acid, a copolymer of 2—methyl—5—vinylpyridine/methyl acrylate/methacrylic acid, a copolymer of l—5— ethylpyridine/methacrylic acid/methy acrylate, a copolymer of 2—vinylpyrid—ine/methacrylic acid/acrylonitrile, ymethylpiperidyl starch, carboxy—methylbenzylaminocellulose, a copolymer of N—vinylglycine/styrene, chitosan, poly(vinyl alcohol), maleic anhydride copolymer, poly (vinyl pyrolidone), starch and starch—based polymers, poly (2—ehtyl—2— oxazoline), poly(ethyleneimine), polyurethane hydrogels, welan gum, rhamsan gum, polyvinyl acetates, ellulose, eudragit RL, RS, NE 30D, and Kollicoat EMM 30D, or any combination thereof. pH Modifying Agent As used herein, the term “pH modifying agent” refers to an agent capable of ting the pH to a desired range. In some embodiments, in conjunction with other above or below embodiments, the pH modifying agent is an acidifying agent. In some embodiments, in conjunction with other above or below embodiments, the pH modifying agent is present in an amount sufficient to lower the pH. pH Modulation agents e maleic acid, citric acid, tartaric acid, pamoic acid, fumaric acid, salicylic acid, 2,6— diaminohexanoic acid, camphorsulfonic acid, glycerophosphoric acid, 2— hydroxyethanesulfonic acid, onic acid, succinic acid, carbonic acid, p—toluenesulfonic acid, aspartic acid, 8—chloro-Itheophylline, benezenesulfonic acid, malic acid, orotic acid, oxalic acid, benzoic acid, 2—naphthalenesulfonic acid, c acid, adipic acid, p— amino-Isalicylic acid, 5—aminoslicylic acid, ascorbic acid, ic acid, cyclamic acid, sodium lauryl e, glucoheptonic acid, glucuronic acid, glycine, sulfuric acid, mandelic acid, l,5—naphthalenedisulfonic acid, nicotinic acid, oleic acid, 2—oxoglutaric acid, pyridoxal —phosphate, undecanoic acid, p—acetamidobenzoic acid, o—acetamido—benzoic acid, m— acetamidobenzoic acid, N—acetyl—L—aspartic acid, camphoric acid, dehydrocholic acid, malonic acid, edetic acid, ethylenediainetetraacetic acid, ethylsulfuric acid, hydroxyphenylbenzoylbenzoic acid, glutamic acid, glycyrrhizic acid, 4—hexylresorcinol, hippuric acid, p—phenolsulfonic acid, 4—hydroxybenzoic acid, 3—hydroxybenzoic acid, 3— hydroxy—2—naphthoic acid, l—hydroxy—2naphthoic acid, lactobionic acid, nylic acid, 5’— adenylic acid, mucic acid, galactaric acid, pantothenic acid, pectic acid, lacturonic acid, 5—sulfosalicylic acid, l,2,3,6—tetrahydro—l,3—dimethyl—2,6—dioxopurine—7— propanesulfonic acid, terephthalic acid, l—hydroxy—2naphthoic acid, and combinations thereof. In some embodiments, in conjunction with other above or below embodiments, acidic excipients include, for example, maleic acid, citric acid, malic acid, fumaric acid, sulfuric acid, tartaric acid, lactoic acid, salicylic acid, aspartic acid, aminosalicylic acid, malonic acid, glutamic acid, and combinations f.
In some embodiments, in conjunction with other above or below embodiments, pH modifying agent includes maleic acid, citric acid, tartaric acid, pamoic acid, fumaric acid, lic acid, 2,6—diaminohexanoic acid, camphorsulfonic acid, glycerophosphoric acid, 2— hydroxyethanesulfonic acid, isethionic acid, ic acid, carbonic acid, p—toluenesulfonic acid, ic acid, 8—chlorotheophylline, benezenesulfonic acid, malic acid, orotic acid, oxalic acid, benzoic acid, 2—naphthalenesulfonic acid, stearic acid, adipic acid, p—amino— salicylic acid, 5—aminoslicylic acid, ascorbic acid, sulfuric acid, cyclamic acid, sodium lauryl e, glucoheptonic acid, onic acid, glycine, ic acid, mandelic acid, 1,5— naphthalenedisulfonic acid, nicotinic acid, oleic acid, 2—oxoglutaric acid, pyridoxal 5— phosphate, undecanoic acid, p—acetamidobenzoic acid, o—acetamidobenzoic acid, m— acetamidobenzoic acid, N—acetyl—L—aspartic acid, camphoric acid, dehydrocholic acid, malonic acid, edetic acid, ethylenediainetetraacetic acid, ethylsulfuric acid, hydroxyphenylbenzoylbenzoic acid, glutamic acid, glycyrrhizic acid, 4—hexylresorcinol, hippuric acid, p—phenolsulfonic acid, 4—hydroxybenzoic acid, 3—hydroxybenzoic acid, 3— hydroxy—2—naphthoic acid, l—hydroxy—2naphthoic acid, lactobionic acid, 3’—adenylic acid, 5’— ic acid, mucic acid, aric acid, pantothenic acid, pectic acid, polygalacturonic acid, 5—sulfosalicylic acid, l,2,3,6—tetrahydro—l,3—dimethyl—2,6—dioxopurine—7— esulfonic acid, terephthalic acid, l—hydroxy—2naphthoic acid, and combinations thereof.
In some embodiments, in conjunction with other above or below embodiments, the pH modifying agent is selected from maleic acid, citric acid, malic acid, fumaric acid, sulfuric acid, tartaric acid, lactoic acid, salicylic acid, aspartic acid, alicylic acid, malonic acid, ic acid, and any combination thereof.
In some embodiments, in conjunction with other above or below embodiments, c acid was used as the pH modifying agent as it is less hygroscopic and more compatible with omecamtiv mecarbil dihydrochloride hydrate than citric acid, resulting in less or no active form transformation and no changes in tablet appearance when stored at 40 OC/75%RH for 6 months, leading to improved final product quality. Additionally, fumaric acid is more acidic (2—fold) than citric acid. Therefore, it is more efficient, i.e., l:l weight ratio to active instead of 2: l, to use fumaric acid to modulate the microenvironmental pH to enhance omecamtiv mecarbil release at neutral environment. Fumaric acid also has a very slow dissolution rate. As a result, fumaric acid will stay in the tablet longer and maintain the low micro—environmental pH better, resulting in more complete release of tiv mecarbil within 24 hours.
Filler As used herein, the term “fillers” refers to one or more substances that can be added to components of a pharmaceutical composition to increase bulk weight of the material to be formulated, e.g. tabletted, in order to achieve the desired weight. Fillers include but are not limited to starches, lactose, ol (such as PearlitolTM SD 200), cellulose derivatives, calcium phosphate, sugar and the like.
Different grades of lactose include, but are not d, to e monohydrate, lactose DT (direct tableting), lactose anhydrous, FlowlacTM (available from Meggle products), PharmatoseTM (available from DMV) and others. Different grades of starches e, but are not limited to, maize starch, potato , rice starch, wheat starch, pregelatinized starch (commercially available as PCS PClO from Signet Chemical ation) and Starch 1500, Starch 1500 LM grade (low moisture content grade) from Colorcon, fully pregelatinized starch (commercially available as National 78—1551 from Essex Grain Products) and others. Different cellulose compounds that can be used include crystalline cellulose and powdered cellulose. Examples of crystalline cellulose products include but are not limited to CEOLUSTM KG801, AvicelTM PH 101, PH102, PH301, PH302 and PH—F20, microcrystalline cellulose 114, and rystalline cellulose 112. Other useful fillers include, but are not limited to, carmellose, sugar alcohols such as mannitol, ol and xylitol, calcium carbonate, magnesium carbonate, dibasic calcium phosphate, and tribasic calcium phosphate.
In some embodiments, in conjunction with other above or below embodiments, the filler is selected from starch, lactose, ol (such as tolTM SD 200), cellulose derivatives, calcium phosphate, and a sugar.
In some embodiments, in conjunction with other above or below embodiments, the filler is lactose anhydrous or e drate. In some embodiments, in ction with other above or below embodiments, the filler is lactose DT, FlowlacTM, or PharmatoseTM.
In some embodiments, in conjunction with other above or below embodiments, the filler is maize starch, potato starch, rice starch, wheat starch, atinized starch (such as Starch 1500 or Starch 1500 LM grade (low moisture content grade)), or fully pregelatinized starch.
In some embodiments, in ction with other above or below embodiments, the filler is rystalline cellulose such as CEOLUSTM KG801, AvicelTM PH 101, PH102, PH301, PH302 and PH—F20, microcrystalline cellulose 114, or microcrystalline cellulose 112.
In In some embodiments, in conjunction with other above or below embodiments, the filler is carmellose, mannitol, ol, xylitol, calcium carbonate, magnesium carbonate, dibasic calcium phosphate, or tribasic calcium phosphate.
As used herein, the term “lubricants” refers to one or more substances that can be added to components of the present compositions to reduce sticking by a solid formulation to the equipment used for production of a unit doss form. Lubricants include stearic acid, hydrogenated vegetable oils, hydrogenated soybean oil and hydrogenated soybean oil & castor waX, stearyl alcohol, e, polyethylene glycol, magnesium stearate, WO 52236 glycerylmonostearate, c acid, glycerybehenate, polyethylene glycol, ethylene oxide polymers, sodium lauryl sulfate, magnesium lauryl sulfate, sodium oleate, sodium stearylFumarate, DL—leucine, colloidal silica, and mixtures thereof.
In some embodiments, in conjunction with other above or below embodiments, the lubricant is stearic acid, hydrogenated vegetable oil, enated n oil, hydrogenated soybean oil, castor wax, stearyl alcohol, leucine, polyethylene glycol, magnesium stearate, glycerylmonostearate, stearic acid, glycerybehenate, polyethylene glycol, ethylene oxide polymers, sodium lauryl sulfate, magnesium lauryl sulfate, sodium oleate, sodium stearylfumarate, DL—leucine, colloidal silica, or any mixture thereof. cturing Process Also provided is a process for making a pharmaceutical formulation bed herein, comprising: blending a mixture comprising omecamtiv il, or a pharmaceutically acceptable salt, a pharmaceutically able hydrate, or a pharmaceutically acceptable hydrate of a pharmaceutically acceptable salt thereof, a control release agent, a pH modifying agent, and a lubricating the blended mixture using a lubricant; granulating the lubricated blend; ating the resultant granulation using the lubricant; and compressing the lubricated granulation into desired form.
Also provided is a process for making a pharmaceutical formulation described herein, comprising: providing a blended mixture comprising omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically acceptable hydrate, or a pharmaceutically acceptable hydrate of a pharmaceutically acceptable salt f, a control release agent, a pH modifying agent, a filler, and a lubricant; granulating the d mixture; and compressing the lubricated granulation into desired form.
Also provided is a process for making a pharmaceutical formulation described herein, comprising: compressing a granulation of omecamtiv mecarbil, or a pharmaceutically acceptable salt, a pharmaceutically acceptable e, or a pharmaceutically acceptable hydrate of a WO 52236 pharmaceutically acceptable salt thereof, a control release agent, a pH modifying agent, a , and a lubricant into desired form.
In some embodiments, in conjunction with other above or below embodiments, the modified e matrix tablets are manufactured using dry granulation. The dry granulation s can help to avoid the active form transformation in the modified release matrix s. In addition, dry granulation process avoids issues observed in a high shear wet granulation process.
Also provided is a ceutical formulation prepared by any of the processes described herein.
Stability Forced degradation conditions (e.g., 40°C and 75% relative humidity) are used to evaluate the long—term storage stability of a pharmaceutical ingredient or composition. In general terms, a stable composition is one which, after being subjected to forced degradation conditions, comprises the pharmaceutically active ingredients in an amount, for example 95%, relative to the amount initially present in the particular composition. Stability may be determined, using forced degradation or other methods, for periods of 1 week, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 9 months, 12 , 15 months, 18 months, 24 months, 30 months, 36 months, longer.
Assays for evaluating the ity of a ceutical composition, such as those described herein, are known in the pharmaceutical arts. For example, one can determine the percentage of active pharmaceutical ingredients t in a given composition, as well as the presence and percentage of impurities, h the use of standard analytical techniques.
Methods of Treatment/ Use of Formulations Disclosed Also provided is a method for the use of such pharmaceutical formulations for the treatment of heart failure, including but not limited to: acute (or decompensated) congestive heart failure, and chronic congestive heart failure; particularly diseases associated with systolic heart dysfunction.
WO 52236 EXAMPLES Manufacture of Omecamtiv Mecarbil dih drochloride h drate S nthetic Route to Omecamtiv Mecarbil i) NaHC03 MeoJLN/? ii) H2, Pd/C, IPAC K/N\/©\NH2 F iii) Heptane F SM-1 Piperazme Ani'ine ____________E.i192£33.in§__ttl_i_!_r_9:_|:_|_9!________________ PhOJL \ N . HCI i) iPr2NEt, THF N ii) solvent swap to IPA H . iii) HCI, H20 g SM-2 i Phenyl Carbamate-HCI l H3COAN/? o / CH3 K/N NJLN \ I N - 2HC|o H20 H H omecamtiv mecarbil-ZHCl-Hzo S nthesis 0f the API SM Pi erazine Nitro-HCl NBS F BZZO FN-Bromide HPO(OEt)2 Br —> —> NO2 Me N02 AcOH MeOH F F PhMe FN-Bromide FN-Toluene Br F zine Carboxylate MeOJLN/? ii) HCI, IPA, PhMe Piperazine Nitro-HCI 88% overall General Methods ts and solvents were used as ed from commercial sources. 1H NMR spectra were recorded on a 400 MHz spectrometer. Chemical shifts are ed in ppm from tetramethylsilane with the solvent resonance as the internal standard (CDCl3, DMSO—d6).
Data are reported as follows: chemical shift, multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, br 2 broad, m = multiplet), ng constants (Hz) and integration. 13C NMR spectra were recorded on a 100 MHz spectrometer with complete proton decoupling.
Chemical shifts are reported in ppm from tetramethylsilane with the solvent as the internal reference (CDCl3, DMSO—d6). All solvent charges are made with respect to starting 2—Fluoro— 3—nitrotoluene.
X—Ray powder diffraction data (XRPD) were obtained using a PANalyticalX’Pert PRO diffractometer (PANalytical, Almelo, The lands) fitted with a real time multiple strip (RTMS) detector. The radiation used was CuKOL(l.54 A) and the voltage and t were set at 45 kV and 40 mA, respectively. Data were collected at room temperature from 5 to 45 s 2—theta with a step size of 0.0334 degrees. Samples were prepared on a low background sample holder and placed on the sample stage which was rotated with a 2 second tion time.
Alternatively, XRPD data were obtained using a PANalyticalX’Pert PRO diffractometer (PANalytical, Almelo, The Netherlands) fitted with a RTMS detector. The radiation used was l.54 A) and the voltage and current were set at 45 kV and 40 mA, respectively. Data were collected at room temperature from 5 to 40, degrees 2—theta with a step size of 0.0334 s. Samples were prepared on a low background sample holder and placed on the sample stage which was rotated with a 2 second revolution time.
Alternatively, XRPD data were obtained using a PANalyticalX’Pert PRO diffractometer (PANalytical, , The Netherlands) fitted with a RTMS detector. The radiation used was CuKOL(l.54 A) and the voltage and current were set at 45 kV and 40 mA, respectively. Data were collected at room temperature from 5 to 40, degrees 2—theta with a step size of 0.0167 degrees. Samples were prepared on a low background sample holder and placed on the sample stage which was d with a 2 second revolution time.
Alternatively, XRPD data were obtained using a PANalyticalX’Pert Pro diffractometer (PANalytical, Almelo, The Netherlands) fitted with a RTMS detector. The radiation used was CuKOL (1.54 A) and the voltage and current were set at 45 kV and 40 mA, respectively. Data were collected at room temperature from 3 to 40, degrees 2—theta with a step size of 0.008 degrees. Samples were prepared on a low background sample holder and placed on the sample stage with a 2 second revolution time.
Alternatively, XRPD data were obtained using a Bruker D8 Discover X—ray diffraction system (Bruker, Billerica, MA) fitted with a motorized xyz sample stage and a GADDS area detector. The radiation used was CuKOL (1.54 A) and the voltage and current were set at 45 kV and 40 mA, respectively. The solid samples on a ?at glass plate were mapped and for each sample an area of 1 mm2 was scanned in an oscillating mode for 3 minutes from 5 to 48 degrees 2—theta.
Differential Scanning Calorimetry (DSC) data was collected using standard DSC mode (DSC Q200, TA Instruments, New Castle, DE). A heating rate of 10°C/min was employed over a temperature range from 40°C to 300°C. is was run under nitrogen and samples were loaded in standard, hermetically—sealed aluminum pans. Indium was used as a calibration standard.
Alternatively, DSC data were ted using temperature—modulated DSC mode (DSC Q200, TA Instruments, New Castle, DE). After sample equilibration at 20°C for five minutes, the heating rate of 3 °C/min was employed with a tion of +/— /min over a temperature range from 20°C to 200°C. Analysis was run under nitrogen and samples were loaded in standard, uncrimped aluminum pans. Indium was used as a calibration FN-Bromide In a 60 L reactor (containing no exposed Stainless steel, Hastelloy®, or other metal parts) equipped with a re?ux/retum condenser and scrubber charged with a 5N NaOH solution, a mechanically stirred mixture of uene (2.0 kg, 12.89 mol, 1.0 equiv.), N— Bromosuccinimide (3.9 kg, 21.92 mol, l.70 equiv.), benzoyl peroxide (125.0 g, 0.03 equiv., 0.39 mol, containing 25 wt% , and acetic acid (7.0 L, 3.5 volumes) was heated to 85 °C under an atmosphere of nitrogen for 7 hours. A solution of H3PO3 (106.0 g, 1.29 mol, 0.1 equiv.) and acetic acid (200 mL, 01 volume), ed in separate vessel, was added. The reaction mixture was agitated for 0.5 h and analysis of an aliquot med complete decomposition of l peroxide (not detected, HPLC254 nm). The reaction e was cooled to 22 °C. DI Water (8.0 L, 4 volumes) and toluene (16.0 L, 8 volumes) were charged, the biphasic mixture was agitated (20 min), and the layers were ted. Aqueous l.6N NaOH (14.0 L, 7.0 s) was added to the organic layer at a rate allowing the batch ature to stay under 25 °C and the pH of the resultant aqueous phase was measured (2 11). The biphasic mixture was filtered through a 5 um Te?on® cartridge line and the layers were separated. The filter line was washed with another 2L of toluene.
The assay yields were 2.5 % of FN—Toluene, 62.3 % of FN—Bromide and 30.0 % of Di—Bromide. The toluene solution contained no benzoyl peroxide, succinimide, or OL— bromoacetic acid and water content by KF titration was 1030 ppm (This solution could be held under nitrogen at room temperature for > 12 h t any change in the assay yield).
To this solution at room temperature was added diisopropylethylamine (880.0 g, 6.63 mol, 0.53 equiv.) followed by methanol (460 mL, 11.28 mol, 0.88 equiv.) and heated to 40 0C. A solution of diethylphosphite (820.0 g, 5.63 mol, 0.46 equiv.) in methanol (460 mL, 11.28 mol, 0.88 equiv.) was prepared and added to the reaction e at 40 °C through an addition funnel over a period of 1 hour at such a rate that the batch temperature was within 40 i 5 OC. The contents were stirred for a period of 3h at 40 °C from the start of addition and cooled to room temperature and held under nitrogen atmosphere for 12 hours. The assay yield of the reaction mixture was 2.5 % FN—Toluene 92.0% FN—Bromide and 0.2% Di—Bromide.
This solution is used as such for the alkylation step.
Characterization for ents of final t mixture (collected for pure compounds). 2-Flu0r0Nitr0toluene (FN-Toluene): 1H NMR (400 MHz, CHLOROFORM-d) 8ppm 2.37 (s, 1 H), 7.13-7.20 (m, 1 H), .51 (m, 1 H), 7.79-7.85 (m, 1 H). 13C NMR (100 MHz, CHLOROFORM-d) 8 ppm 14.3 (d, J: 5 Hz), 123.3 (d, J: 3 Hz), 123.6 (d, J: 5 Hz), 128.2 (d, J: 16 Hz), 136.7 (d, J: 5 Hz), 137.5 (broad), 153.7 (d, J: 261 Hz); 1- (bromomethyl)flu0r0nitrobenzene (FN-Bromide): 1H NMR (400 MHz, CHLOROFORM-d) 8 ppm 4.56 (s, 1 H), 7.28-7.34 (m, 1 H), 7.69—7.76 (m, 1 H), 7.98—8.05 (m, 1 H). 13C NMR (100 MHz, CHLOROFORM-d) 8 ppm 23.6 (d, J: 5 Hz), 124.5 (d, J: 5 Hz), 126.1 (d, J: 3 Hz), 128.5 (d, J: 14 Hz), 136.5 (d, J: 4 Hz), 137.7 (broad), 153.3 (d, J : 265 Hz). DSC: single melt at 53.59 °C. Exact Mass [C7H5BrFN02 + H]+: calc. : 233.9566, measured : 233.9561; 1-(dibromomethyl)fluoronitrobenzene (Dibromide): 1H NMR (400 MHz, CHLOROFORM-d) 8ppm 6.97 (s, 1 H), 7.39-7.45 (m, 1 H), 8.03—8.10 (m, 1 H), 8.16—8.21 (m, 1 H). 13C NMR (100 MHz, CHLOROFORM-d) 8 ppm 29.2 (d, J: 7 Hz), 124.9 (d, J: 5 Hz), 127.1 (d, J: 2 Hz), 132.1 (d, J: 11 Hz), 135.7 (d, J: 2 Hz), 137.2 (broad), 149.8 (d, J = 266 Hz). DSC: single melt at 49.03 °C. Exact Mass [C7H4Br2FNOZ + H]+: calc. 2 311.8671, ed 2 311.8666.
Piperazine Nitro-HCl: To a mechanically stirred toluene on (9 volumes) of FN—Bromide (prepared from previous step) in a 60 L reactor at 22 °C under an atmosphere of nitrogen, diisopropylethylamine was charged (1.90 kg, 14.69 mol, 1.14 equiv.). To this e a solution of piperazine ylate methylester (Piperazine Carboxylate) (2.03 kg, 14.05 mol, 1.09 equiv.) in toluene (1.0 L, 0.5 volumes) was added at a rate allowing the batch temperature to stay under 30.0 °C (Exothermic. During the addition, jacket temperature was adjusted to 5 °C in order to maintain batch temperature below 30 OC. The mixture was agitated at 22 °C for 3 hours and analysis of an aliquot confirmed tion of the alkylation on (<1.0 LCAP FN—Bromide, HPLC254 nm). The reaction mixture was treated with aqueous NH4Cl (20 wt%, 10.0 L, 5 volumes; prepared from 2.0 kg of NH4Cl and 10.0 L of DI water), the biphasic e was agitated (30 min), and the layers were separated. The organic layer was sequentially washed with aqueous NaHCO3 (9 wt%, 10.0 L, 5 volumes; prepared from 0.90 kg of NaHCO3 and 10.0 L of DI . The organic layer was filtered through a 5 pm Te?on® cartridge line and transferred in a drum, washed the filter line with another 1.0 L toluene and the combined toluene solution (10.0 volumes) weighed, and assayed (HPLC) to quantify Piperazine Nitro free base. The assay yield for the Piperazine Nitro—freebase is 89.0%, FN—Toluene 2.5% and mide 0.2% with FN—Bromide undetected. The total loss of product to the aqueous washes is < 1.0 %. This solution under nitrogen here is stable for more than 12h.
To a mechanically stirred toluene solution of Piperazine Nitro free base, prepared as described above, at 22 °C in a 60 L reactor under an atmosphere of nitrogen, IPA (19.4 L, 9.7 volumes) and DI water (1.0 L, 0.5 volume) were charged. The mixture was heated to 55 °C and 20% of the 1.4 equiv. of conc. HCl (Titrated prior to use and charge based on titer value; 276.0 mL, 3.21 mol) was charged. The contents were agitated for 15 min and Piperazine HCl seed (130.0 g, 0.39 mol, 0.03 equiv.) was charged as slurry in IPA (400 mL, 0.2 volume). The mixture was agitated for 30 min and the remaining conc. HCl (80% of the charge, 1.10 L, 12.82 mol) was added over a period of 4 hours. The mixture was stirred at 55 °C for 1 h, cooled to 20 °C in a linear manner over 1.5 hours, and agitated at this temperature for 12 hours. The supernatant concentration of Piperazine Nitro—HCl was measured (2.8 mg/g). The mixture was filtered through an aurora filter equipped with a 5 pm Te?on® cloth. The mother liquor were transferred to a clean drum and assayed. The filter cake was washed twice with IPA (11.2 L, 5.6 volumes) and dried to constant weight (defined as S 1.0% weight loss for 2 consecutive TGA measurements over a period of 2 hours) on filter with vacuum and a en sweep (14 h). The combined losses of zine Nitro— HCl in the mother liquors and the washes were 2.5 %. Piperazine Nitro—HCl was isolated 3.59 kg in 87.6% corrected yield with >99.5 wt% and 99.0% LCAP purity.
Methyl 4—(2—?uoro—3—nitrobenzyl)piperazine—1—carboxylate hydrochloride (Piperazine Nitro—HCl): 1H NMR (300 MHz, DMSO—d) 5 ppm 3.25 (br. s, 3 H), .66 (m, 8 H), 4.47 (s, 2 H), 7.44-7.63 (t, 1 H, J: 8 Hz), .15 (m, 1 H), .34 (m, 1 H). 13C NMR (75 MHz, DMSO-d) 8 ppm 50.3, 51.4, 52.8, 119.6 (d, J: 14 Hz), 125.1 (d, J: 5 Hz), 127.9, 137.4 (d, J: 8 Hz), 139.8 (d, J: 3 Hz), 152.2, 154.7, 155.7. DSC: melt onset at 248.4 0C. Exact Mass [C13H16FN3O4 + H]+: calculated 2 298.1203, measured 2 298.1198.
Piperazine Nitro Freebase: In a 60 L reactor equipped with a re?ux/return condenser, a mixture of Piperazine Nitro—HCl (2.0 kg, 5.99 mol, 1.0 equiv.) and pyl acetate (6.0 L, 3.0 volumes) was mechanically agitated at ambient temperature under an atmosphere of nitrogen. A solution of sodium bicarbonate (629 g, 7.49 mol, 1.25 equiv.) and water (7.5 L, 3.75 volume), prepared in separate vessel, was added. The biphasic e was agitated (15 min), and the layers were ted. The upper organic layer (containing product) was transferred to a separate vessel while the reactor was rinsed with water and isopropanol. The organic layer was then transferred through an inline 5 pm Te?on® cartridge back into the clean 60 L reactor. The filter line was washed with 4.0 L (2.0 s) of isopropanol into the 60 L reactor. An additional 12.0 L (6.0 volumes) of isoproponal was added to the 60 L reactor and heated to 40 OC. Under reduced pressure (50 torr) the batch was concentrated down to approximately 6 L (3.0 volumes). The solution was cooled from 27 °C to 20 °C in a linear manner over 10 minutes. Water (4.0 L, 2.0 volumes) was added at 20 °C over 30 minutes followed by Piperazine Nitro se seed (18 g, 0.06 mol, 0.01 equiv). The mixture was aged for 5 s and the ing water (24.0 L, 12.0 volumes) was added over 90 minutes. After holding overnight at 20 0C, the supernatant concentration of Piperazine Nitro Freebase was measured (< 10 mg/mL). The mixture was filtered through an aurora filter equipped with a 12 um Te?on® cloth. The filter cake was washed with a mixture of water (3.3 L, 1.65 volumes) and isopropanol (700 mL, 0.35 volumes) and dried to constant weight (defined as S 1.0% weight loss for 2 utive TGA measurements over a period of 2 hours) on filter with vacuum and a nitrogen sweep (48 h). The combined losses of Piperazine Nitro Freebase in the mother s and the wash were aproximately 7.5 %. Piperazine Nitro Freebase was isolated 1.67 kg in 92.5% corrected yield with 100.0 wt% and 99.4% LCAP purity.
S nthesis of the API SM Phen l Carbamate-HCI / CH3 CICOZPh H2N P Amino Pyridine Phenyl Carbamate-HCI A 60 L, glass—lined, jacketed reactor set at 20 °C under nitrogen here and vented h a scrubber (containing 5N NaOH) was charged with 2.5 kg of Amino Pyridine (1.0 equiv, 23.1 moles), followed by 25 L (19.6 kg, 10 vol) acetonitrile. After initiating agitation and (the endothermic) dissolution of the Amino Pyridine, the vessel was charged with 12.5 L of N—methyl—2—pyrolidinone (12.8 kg, 5 vol). An addition funnel was charged with 1.8 L (0.6 equiv, 13.9 moles) phenyl chloroformate which was then added over 68 minutes to the solution of the Amino Pyridine keeping the internal temperature S 30°C.
The reaction was agitated for > 30 minutes at an internal ature of 20 i 5 °C. The vessel was then charged with 61 i 1 g of seed as a slurry in 200 mL acetonitrile and aged for 2 30 min. The addition funnel was d with 1.25 L (0.45 equiv, 9.7 moles) of phenyl chloroformate which was then added over 53 minutes to the reaction suspension while again keeping the temperature S 30°C. The ts of the reactor were aged 2 30 hours at 20 i °C. After assaying the supernatant (S 15mg/g for both product and starting material), the solids were filtered using an Aurora filter equipped with a 12pm Te?on cloth. The mother liquor was forwarded to a 2Ild 60 L, glass—lined, jacketed reactor. The reactor and cake were rinsed with 1 X 10 L of 5:10 NMP/ACN and 1 X 10 L ACN. The washes were forwarded to the 2Ild reactor as well. The cake was dried under vacuum with a nitrogen bleed for 2 24 hours to afford 5.65 kg (90.2% yield) of the product, Phenyl ate—HCl as an off—white solid in 98.8 wt% with 99.2% LCAP purity.
Phenyl (6—methylpyridin—3—yl)carbamate hydrochloride (Phenyl Carbamate—HCl) 1H NMR (400 MHz, DMSO-d6) 8ppm 11.24 (s, 1 H), 8.81 (s, 1 H), 8.41 (d, 1 H, J: 8.8 Hz), 7.85 (d, 1 H, J: 8.8 Hz), 7.48 - 7.44 (m, 2 H), 7.32 - 7.26 (m, 3 H), 2.69 (s, 3 H); 13C NMR (100 MHz, DMSO-d6) 8ppm 151.66, , 147.51, 136.14, 133.79, 129.99, 129.49, 127.75, 125.87, 121.70, 18.55: HR-MS : Calclulated for N202: 228.0899, M + H“ 2 229.0972; Observed mass: 229.0961 GMP Steps Methyl mino—2—?uorobenzyl)piperazine—1—carboxylate (Piperazine Aniline) Neutralization O 0 JL NaHCO (1.25 equiv) i MeO U s MeO U .HCI N02 IPAc (3V); Water (3.75V) N02 F lPAc solution F Piperazine NitrooHCl + NaCl (1 equiv) + 002 (1 equiv) + H20 (1 equiv) + NaHCO3 (0.25 equiv) 1 wt% Pd/C Hydrogenation_ H2 (60 psig) °C 1 ) Azeotropic Dry ing(lPAc) Meoio 2) Heptane (anti-solvent) N lPAc solution F Piperazine Aniline + 2 To a 100—L jacketed glass—lined reactor were added methyl 4—(2—?uoro—3— nitrobenzyl)piperazine—1—carboxylate hydrochloride (2.00 kg, 1.00 equiv) and isopropyl acetate (6.00 L, 3.00 Vol with—respect to starting material). The resulting slurry was agitated under a nitrogen sweep. To the mixture was added se over 45 i 30 min: 7.7 % w/w aqueous sodium bicarbonate solution (629 g, 1.25 equiv of sodium bicarbonate dissolved in 7.50 L water), maintaining an internal temperature of 20 i 5 °C by jacket l (NOTE: addition is endothermic, and may evolve up to 1 equiv of carbon dioxide gas). The mixture was d for Z 15 min, resulting in a clear biphasic e. Agitation was stopped and the layers were allowed to settle.
The bottom (aqueous) layer was drained and analyzed by pH paper to ensure that the layer is pH > 6. Quantititative HPLC analysis of the upper (organic) layer revealed 97— 100% assay yield of the methyl 4—(2—?uoro—3—nitrobenzyl)piperazine—1—carboxylate freebase (1.73 — 1.78 kg). The upper (organic) layer was transferred through an in—line filter into a 20— L Hastelloy® hydrogenator, and the 100—L reactor and lines were rinsed with an additional aliquot of isopropyl acetate (2.00 L, 1.00 Vol). The hydrogenator was purged with nitrogen and vented to heric pressure. To the reaction mixture was added a slurry of 5.0 wt% palladium on carbon (20.0 g, Strem/BASF EscatTM 1421, approx 50% water) in isopropyl e (400 mL), ed by a 400 mL rinse. The resulting reaction mixture was diluted with an additional aliquot of isopropyl acetate (1.2 L; total isopropyl acetate amount is 10.0 L, 5.00 Vol). The hydrogenator was purged three times with nitrogen (pressurized to 60 i 10 psig, then vented to atmospheric pressure), then rized to 60 i 5 psig with hydrogen.
The reaction mixture was stirred at < 100 rpm at 30 i 5 °C while maintaining 60 i 5 psig hydrogen, for >2 hours until reaction was deemed complete. This temperature and pressure correspond to a measured kLa value of approx 0.40 in a 20—L Hydrogenator. End of reaction is determined by dramatic decrease in hydrogen ption anied by a relief in the heat evolution of the reaction. To control potential dimeric impurities, the reaction is continued for at least 30 minutes after this change in reaction profile, and HPLC analysis is performed to confirm that >99.5% conversion of the hydroxyl—amine to the e is achieved.
At the end of reaction, the enator was purged with nitrogen twice (pressurized to 60 i 10 psig, then vented to atmospheric pressure). The crude on mixture was filtered through a 5 pm filter ed by a 0.45 pm filter in series, into a 40—L glass—lined reactor. The hydrogenator and lines were washed with an additional aliquot of isopropyl acetate (2.00 L). Quantitative HPLC analysis of the crude reaction mixture revealed 95—100% assay yield (1.52 — 1.60 kg aniline product). The reaction e was distilled under reduced re (typically 250 — 300 mbar) at a batch temperature of 50 i 5 °C until the total reaction volume was approximately 8.00 L (4.00 Vol). The batch was subjected to a constant—volume distillation at 50 i 5 0C, 250 — 300 mbar, by adding heptane to control the total batch volume. After approximately 8.00 L (4.00 Vol) of heptane were added, GC analysis indicated that the solvent composition was approximately 50 % isopropyl acetate, 50% heptane. Vacuum was broken, and the internal batch temperature was maintained at 50 i 5 0C. To the reaction mixture was added a slurry of seed (20.0 grams of product methyl 4—(3—amino—2—?uorobenzyl)piperazine—1—carboxylate, in a solvent mixture of 80 mL e and 20 mL isopropyl acetate). The ing slurry was allowed to stir at 50 i °C for 2 i 1 hours, then cooled to 20 i 5 °C over 2.5 i 1.0 h. Additional heptane (24.0 L, 12.0 Vol) was added dropwise over 2 hours, and the batch was allowed to stir at 20 i 5 °C for Z 1 hours (typically ght). tative HPLC analysis of this filtered supernatant revealed < 5 mg/mL product in solution, and the product crystals were 50 — 400 um birefringent rods. The reaction slurry was filtered at 20 °C onto a filter cloth, and the cake was displacement—washed with heptane (6.00 L, 2.00 Vol). The cake was dried on the filter under nitrogen sweep at ambient temperature for > 4 hours, until sample dryness was confirmed by LOD analysis (indicated <1.0 wt% loss). The product methyl 4—(3—amino—2— enzyl)piperazine—1—carboxylate (1.56 kg) was isolated as a pale—yellow powder in 86% yield at 99.8 wt% by HPLC with 100.0 LCAP210. [Analysis of the combined filtrates and washes ed 108 grams (7.0%) of t lost to the mother liquors. The remaining mass balance is sed of product hold—up in the reactor (fouling).] 1H NMR (DMSO-dg, 400 MHz) 5: 6.81 (dd, J = 7.53, 7.82 Hz, 1H), 6.67 (m, 1H), 6.49 (m, 1H), 5.04 (s, 2H), 3.58 (s, 3H), 3.45 (m, 2H), 3.34 (m, 4H), 2.33 (m, 4H). 19F NMR (d6-DMSO, 376 MHz) 5: - 140.2. 13C NMR (d6-DMSO, 125 MHz) 5: 155.0, 150.5, 148.2, 136.2 (m), 123.7 (m), 117.6, 115.1, 73.7, 54.9 (m), 52.1 (m), 43.4. mp = 89.2 °C.
Omecamtiv Mecarbil Dih drochloride H drate rocedure r ; NH2 / Me | O / [:1 I N JL DIPEA (1.30 equiv) \ + \ N PhO N - HCI —’ H THF (4V) 65°C 8—24 h 0%OMe (1.2 equiv) [N1': Phenyl Carbamate-HCI (1.0 equiv) phenyl (6—methylpyridin—. . AOMe Piperazine Aniline 3-y|)carbaniate DIPEA-HCI (1.2 equiv) methyl 4-(3-amino hydrochloride + DIPEA (0.10 equiv) fluorobenzyl)piperazine Phenol (1.0 equiv) carboxylate 2539880 (0.2 equiv) 1) 2-PrOH (11 V) 2) Distill to 4V 3) Water (2.30 V) 4) 6N HCI (2.4 equiv) ) 2-PrOH (16.5V) 6) I [“11310H---2HC|H20 0AOMe To a 15L glass lined reactor were charged methyl 4—(3—amino—2—?uoro— benzyl)piperazine—1—carboxylate (1,202 g, 4.50 mol), phenyl (6—methylpyridin—3— yl)carbamate hydrochloride (1,444 g, 5.40 mol), and tetrahydrofuran (4.81 L). The resulting slurry was agitated under a nitrogen sweep and N,N—diisopropylethylamine (1,019 L, 5.85 mol) was then charged to the slurry which resulted in a brown solution. The temperature of the solution was increased to 65 °C and agitated for 22 h, until <1% AUC piperazine aniline remained by HPLC analysis.
The batch was cooled to 50 °C and distilled under reduced pressure while maintaining the internal ature of the vessel below 50 °C by adjusting vacuum pressure. 2—Propanol was added with residual vacuum at a rate to in a constant volume in the 15 L reactor. A total of 10.5 kg of 2—propanol was required to achieve <5% THF by GC. Water (2.77 kg) was then charged to the reactor followed by the addition of 6N HCl (1.98 kg) at a rate to maintain the internal temperature below 60 °C. The reactor was brought to ambient pressure under a en sweep. The solution was then heated to 60 °C, and transferred to a 60L glass lined reactor through an inline filter. The 15L reactor was then rinsed with 1:1 water/2—propanol (1.2L) which was sent through the inline filter to the 60L reactor.
The 60L reactor was adjusted to 45 °C and a slurry of seed (114 g, 0.23 mol) in 2— ol (0.35 L) was added to the reactor ing in a . The batch was aged at 45 °C for 1 h, followed by the addition of 2—propanol (3.97 kg) through an inline filter over 2 h. The batch was heated to 55°C over 1 h and held for 0.25 h, then cooled back to 45°C over 1 h and held overnight at 45 °C. 2—propanol (11.71 kg) was then added through an inline filter to the batch over 3 h. The batch was aged for 1 h and then cooled to 20°C over 2 h and held at 20 °C for 0.5 h. The batch was then recirculated though a wet mill affixed with 1—medium and 2— fine rotor—stators operating at 56 Hz for 2.15 h, until no further particle size reduction was observed by microscopy.
The batch was then filtered through a 20” loy® filter fitted with a 12 um filter cloth under 500 torr . A wash solution of 95:5 2—propanol:water (1.82 L) was charged through an inline filter to the 60L reactor, then onto the filter. A second wash of 2— propanol (2.85L) was charged through an inline filter to the 60L reactor, then onto the filter.
The batch was then dried under 5 psi humidified en pressure until <5,000 ppm 2— propanol, and 2.5—5% water ed. The final solid was discharged from the filter to afford 2.09 kg of methyl 4—(2—?uoro—3—(3—(6—methylpyridin—3—yl)ureido)benzyl)piperazine—1— carboxylate as an off—white crystalline solid in 89% yield at 99.88 wt% by HPLC, 100.0% AUC. Total losses to liquors was 0.10 kg (4.7%).
DSC: Tonset = 61.7 0C, Tmax = 95.0 0C; TGA = 2.2%, degradation onset = 222 0C; 1H HMR (D20, 500 MHz) 8 8.87 (s, 1H), 8.18 (d, J: 8.9 Hz, 1H), 7.83 (t, J: 7.5 Hz, 1H), 7.71 (d, J: 8.8 Hz, 1H), 7.35—7.29 (m, 2H), 4.48 (s, 2H), 4.24 (br s, 2H), 3.73 (s, 3H), 3.31 (br s, 6H), 2.68 (s, 3H); 13C HMR (D20, 150 MHz) 8 156.8, 154.2, 153.9 (J: 249 Hz), 147.8, 136.3, 136.1, 130.1, 129.4, 128.0, 127.2, 125.5 (J: 11.8 Hz), 125.1 (J: 4.2 Hz), 116.1 (J: 13.5 Hz), 53.54, 53.52, 53.49, 50.9, 40.5, 18.2.
Comparative Example 1: Immediate Release Formulation Table 1 Immediate release ation comprising the above ents were prepared according to the process outlined in Figure 1.
Example 1: Prototype Modified Release Formulation Omecamtiv mecarbil prototype modified e (“MR”) matrix tablet formulation consists of omecamtiv mecarbil anhydrate free base (active), MethocelTM K100 M CR ol release agent), citric acid monohydrate (pH modulation agent), microcrystalline cellulose and lactose monohydrate (filler), MethocelTM E5 LV (binder), and magnesium te (lubricant). Table 1 shows the prototype formulation compositions. The prototype MR matrix tablets are manufactured Via a conventional high shear wet granulation process.
This includes screening omecamtiV mecarbil anhydrate, lactose monohydrate FFL 316, microcrystalline cellulose, AVicel® PH 101, MethocelTM K100 M CR, and citric acid monohydrate through a #20 mesh US standard screen ed by charging the screened materials into an appropriate size of high shear granulator, where the materials are dry mixed for a specific time at the pre—determined er and chopper speeds (granulator size, dry mixing time, impeller and chopper speeds are scale—dependent parameters). The wet granulation process starts with the addition of pre—prepared 3% w/w MethocelTM E5 solution using a pre—selected spray nozzle at a pre—determined spray pressure and spray rate. The pre— determined impeller and chopper speeds are used during the wet granulation process (the nozzle size, spray rate, spray re, impeller, and chopper speeds are scale—dependent parameters). After wet granulation, the wet mass is dried using a ?uid bed drying process with a target of LCD (loss on drying) of <2.4% (?uid bed granulator is scale—dependent The dried granulation is then milled using a Fitzmill® using a termined speed and screen size (Fitzmill® model, speed and screen size are scale—dependent parameters). After milling, the milled dry ation is lubricated using the pre—screened (#30 mesh) ium stearate in a tumble blender at a pre—determined speed, time, and fill—volume (tumble blender model, blending speed, time, and fill—volume are dependent parameters). After the lubrication, the final blend is compressed into MR matrix tablets using a rotary tablet press at a target tablet hardness of 10—14 kp.
The following case study exemplifies an embodiment of a manufacturing process of omecamtiV mecarbil anhydrate 25 mg prototype MR matrix tablets. The target batch size is 60 kg. the raw materials billed for the batch is 4.30 kg of omecamtiV mecarbil ate (approximately 14.7% excess to compensate the de—lumping loss), 10.1 kg of microcrystalline cellulose, AVicel® PH101, 8.12 kg of lactose monohydrate FFL316, 7.50 kg of citric acid drate, 30.0 kg of MethocelTM K100 M CR, 0.6 kg MethocelTM E5 LV s binder solution prepared, but the exact amount is added during wet granulation process. The residual binder solution is discarded as the , 19.4 kg of ed water, and 0.30 kg of magnesium stearate.
Binder solution preparation: Filling 19.4 kg of purified water into a l9—gallon portable mixing kettle and then adding 0.6 kg of MethocelTM E5 LV slowly and steadily.
Loading the raw materials into the Diosna P—300 high shear granulator: ly loading the majority of screened lactose monohydrate and microcrystalline cellulose into granulator bowl. ly g citric acid monohydrate into the bowl. Manually loading milled omecamtiV mecarbil ate into the bowl. Manually loading screened MethocelTM K100 M CR into the bowl.
Transferring the binder solution: Transferring the binder solution into the solution tank.
Wet granulation: Transferring 6.60 kg of binder on into granulator bowl.
Fluid bed drying: Dry the granulation.
Dry milling: Manually charging the dried granulation and beginning to mill.
Lubrication: Loading approximately half of the milled granulation into a V—blender and then adding the magnesium stearate in, adding the remaining half of milled granulation Compression: The final blend is manually d into the hopper of rotary tablet press ed with 7/16” round, standard cup, concave, plain tooling. The target tablet weight is 400 mg with a range of 370—430 mg. the target hardness is 12 kp with a range of 10— 14 kp.
Prototype MR matrix tablet ation composition Component omecamtiV mecarbil anhydrate MCC, AVicel® PHlOl Lactose monohydrate FFL 316 Citric acid Monohydrate MethocelTM K100 M CR Methocel E5 LV Magnesium stearate Matrix Modified Release Tablet: General Method A process for modified release (“MR”) matrix tablet cturing via a dry granulation process is described herein. Omecamtiv mecarbil dihydrochloride hydrate, microcrystalline cellulose, lactose monohydrate, MethocelTM K100 M CR/MethocelTM K100 LV CR, and fumaric acid are screened and then charged into a tumble blender and blended there for a specific time at a pre—determined speed (blender size, ng speed, and blending time are scale—dependent parameters). The blended als are lubricated in the same blender using the pre—screened magnesium stearate. The ated blend is then roller compacted and milled. The ant granulation is lubricated in a tumble blender using the pre—screened magnesium stearate. The lubricated granulation is compressed into modified release matrix tablets using a rotary tablet press with a target tablet hardness of 10 kp.
Example 2: Omecamtiv mecarbil dihydrochloride hydrate 25 mg slow release MR matrix tablets gMTX-Fl ].
The target batch size is 5 kg the raw materials billed for the batch is 306.50 g of omecamtiv mecarbil dihydrochloride hydrate, 1840.50 g of microcrystalline ose, ® PH102, 920.0 g of lactose monohydrate, FFL316, 383.0 g of fumaric acid, 1500.0 g of MethocelTM K100 M CR, 35 g of granular magnesium te (10 g excess from theoretical batch size to accommodate the screening process loss), and 35 g of extra—granular magnesium stearate (10 g excess from theoretical batch size to accommodate the screening process loss).
Powder Screening: Step 1. Screening 1840.5 g of microcrystalline cellulose, ® PH102, 306.50 g of omecamtiv mecarbil dihydrochloride hydrate, 383.11 g of fumaric acid, 920.0 g of lactose monohydrate, FFL316, and 1500.0 g of MethocelTM K100 M CR through a 20 mesh US standard sieve into a double PE bag.
Powder Blending: Step 2. Charging the screened blend from Step 1 into a 20 L Bohle blender and blending for 30 s at a speed of 20 rpm.
Powder Lubrication: Step 3. Screening the entire amount of intra—granular magnesium stearate through a 60 mesh US rd sieve and weighing out the required amount of sieved magnesium te, 25.0 g, into a an appropriate container. Step 4.
Manually pre—mixing the required amount of sieved magnesium stearate with approximately 1 x to 3 x of powder blend from Step 2 in the same container for approximately 60 seconds.
Step 5. Charging the pre—mix blend from Step 4 back into the powder blend in Step 2. Step 6. ng the powder blend from Step 2 for 4 minutes at a blending speed of 20 rpm. Step 7.
Discharging the lubricated powder blend into an appropriate container.
Dry granulation: Step 8. Charging the lubricated powder blend from Step 7 into Gerteis roller compactor hopper and start dry granulation manufacturing using the following process parameters. Roll Surface: Knurl; Agitator speed: 15 rpm; Roll force: 7.0 kn/cm; Roll speed: 2 rpm; Roll gap: 2.5 mm; Gap control: ON; Screen size: 1 mm; Clearance n granulator and screen: 2.0 mm; Granulator speed: 80 rpm; and Granulator rotation angle: 200/230 . Step 9. Discharging the granulation into an appropriate container and weigh the net weight, which is 4844 g. ation lubrication: Step 10. Calculating the required amount of magnesium stearate needed for the granulation blend, which is 24.34 g. Step 11. Screening the entire amount of extra—granular magnesium stearate through a 60 mesh US standard sieve and weighing out the required amount of screened magnesium stearate in Step 10. Step 12.
Charging the granulation from Step 9 into a 20 liter Bohle blender. Step 13. Manually pre— miXing the screened extra—granular magnesium stearate from Step 11 with 1X to 3 X of ation from Step 12 in an appropriate container for about 60 seconds. Step 14. Charging the pre—mixed blend from Step 13 back to the r in Step 12. Step 15. Blending the granulation blend from Step 12 for 5 minutes at a blending speed of 20 rpm. Step 16. rging the granulation blend from Step 15 into an appropriate container.
Tablet compression: Step 17. The final granulation blend from Step 16 is manually charged into the hopper of rotary tablet press Korsch XL100 equipped with 7/16” round, standard cup, concave, plain tooling. Step 18. The compression starts at a speed of 25 rpm to dial in the target tablet weight and hardness. The target tablet weight is 500 mg with a range of 475—525 mg. the target hardness is 10 kp with a range of 6—14 kp. The total number of tablet manufactured is 9,115.
Table 2. Composition of tiv mecarbil dihydrochloride hydrate 25 mg slow release MR matrix tablets MTX-Fl in ance with the disclosure mg Slow release Material Theo. Theo.
W/W (%) t Intra—granular omecamtiV mecarbil Di—HCl hydrate 6.13 30.65 Methocel K100 M Prem CR 30.00 150.00 Microcrystalline ose, PH 102 36.81 184.05 Lactose monohydrate, FF 316 18.40 92.00 Fumaric acid 7.66 38.30 Magnesium stearate 0.50 2.50 Sub Total 99.50 497.50 Extra—granular Magnesium stearate 0.50 2.50 Total/batch weight 100.00 500.00 Matrix modified e s comprising the above components were prepared according to the process outlined in Figure 2. Note: In some embodiments, the concentration range is 15%—80% for MethocelTM K100 M CR, 0%—70% for microcrystalline cellulose, AVicel® PH102, 0%—70% for lactose monohydrate, FFL316, 3.83%—50% for fumaric acid, 0%—2% for intra—granular magnesium stearate, and 0%—2% for extra—granular magnesium Example 3 Table 3. Composition of omecamtiv mecarbil dihydrochloride hydrate 25 mg fast e MR matrix tablets MTX-F2 in accordance with the sure Material 25 mg Fast release Theo. Theo. w/w ( %) mg/unit Intra—granular omecamtiV mecarbil Di—HCl e 6.13 30.65 Methocel K100 M Prem CR 5.00 25.00 Methocel K100 LV Prem CR 20.00 100.00 Microcrystalline cellulose, PH 102 40.14 200.70 Lactose monohydrate, FF 316 20.07 100.35 Fumaric acid 7.66 38.30 Magnesium stearate 0.50 2.50 Sub Total 99.50 497.50 2014/027104 Total/batch weight 100.00 500.00 Matrix modified release tablets comprising the above components were prepared according to the process outlined in Figure 3. Note: In some embodiments, the concentration range is 0%—15% for MethocelTM K100 M CR, l5%—50% for MethocelTM K100 LV, 0%—75% for rystalline cellulose, AVicel® PH102, 0%—75% for e monohydrate, FFL3l6, 3.83%—50% for fumaric acid, 0%—2% for intra—granular ium stearate, and 0%—2% for extra—granular magnesium stearate.
Example 4 Table 4. Composition of omecamtiv mecarbil dihydrochloride hydrate 75 mg slow release MR matrix tablets MTX-F3 in accordance with the disclosure al 75 mg Theo. Theo. w/w (%) mg/unit Intra—granular omecamtiV mecarbil Di—HCl 18.37 91.85 hydrate MethocelTM K100 M Prem CR 30.00 150.00 Microcrystalline cellulose, PH 102 24.20 121.00 Lactose monohydrate, FF 316 8.07 40.35 Fumaric acid 18.37 91.85 Magnesium stearate 0.50 2.50 Sub Total 99.50 497.50 Extra—granular Magnesium stearate 0.50 2.50 Total/batch weight 100.00 500.00 Matrix modified e tablets comprising the above components were prepared according to the process outlined in Figure 3. Note: In some embodiments, the concentration range is % for MethocelTM K100 M CR, 0%—65% for microcrystalline ose, AVicel® PH102, 0%—65% for lactose monohydrate, FFL3l6, 3.83%—50% for fumaric acid, 0%—2% for intra—granular magnesium stearate, and 0%—2% for extra—granular magnesium stearate.
Example 5 Table 5. Composition of tiv mecarbil dihydrochloride hydrate 75 mg fast release MR matrix tablets MTX-F4 in accordance with the disclosure 75 mg Fast release Material Theo. mg/unit Intra—granular omecamtiv mecarbil Di—HCl 18.37 91.85 hydrate Methocel K100 M Prem CR 5.00 25.00 Methocel K100 LV Prem CR 2000 100.00 Microcrystalline cellulose, PH 102 27.95 200.70 Lactose monohydrate, FF 316 9.31 100.35 Fumaric acid 18.37 91.85 Magnesium stearate 0.50 2.50 Sub Total 99.50 497.50 Extra—granular Magnesium stearate 0.50 2.50 Total/batch weight 100.00 500.00 Matrix modified e tablets sing the above components were prepared according to the process outlined in Figure 3. Note: In some embodiments, the concentration range is 0%—15% for MethocelTM K100 M CR, 15%—50% for MethocelTM K100 LV, 0%— 65% for microcrystalline cellulose, Avicel® PH102, 0%—65% for lactose monohydrate, , 50% for c acid, 0%—2% for intra—granular magnesium stearate, and 0%—2% for extra—granular magnesium stearate. pH ent release profiles A formulation of omecamtiv mecarbil hemihydrate (free base) and dihydrochloride hydrate (Form A) were prepared having the following components, all components reported as a w/w%: Free Base(75 mg matrix tablet) Active granulation: 15.37% free base; 30% hypromellose, HPMC K100 MPrem CR; 10% citric acid drate; 11.88% microcrystalline cellulose, Avicel PH 101; 6.75% lactose monohydrate, FastFlo 316; 12.5% purified water; and Citric Acid granulation: 20% citric acid drate; 5% microcrystalline cellulose, Avicel PH 101; and 1% magnesium stearate, non—bovine.
Form A (75 mg matrix tablet) Intra—granulation: 18.37% Form A; 30% hypromellose, HPMC K100 MPrem CR; 0.50% magnesium te;; and Extra—granulation: 16.88% microcrystalline cellulose, Avicel PH 101; 18.37% citric acid anhydrous; and 0.5% magnesium stearate, vine.
The formulations were tested at pH 2 and pH 6.8 and the amount of drug released over time was measured. The results of this drug release profile are shown in Figure 6.
The foregoing is merely illustrative of the invention and is not intended to limit the invention to the sed compounds. Variations and changes which are obvious to one skilled in the art are intended to be within the scope and nature of the invention which are defined in the appended claims.

Claims (6)

What is Claimed:
1. A dihydrochloride drate salt of omecamtiv mecarbil.
2. The salt of claim 1, wherein the salt is lline.
3. The salt of claim 1 or claim 2, n the salt is characterized by an X-ray powder ction pattern comprising peaks at about 6.6, 14.9, 20.1, 21.4, and 26.8 ± 0.2° 2? using Cu Ka radiation.
4. The salt of claim 3, wherein the X-ray powder diffraction pattern further comprises peaks at about 8.4, 24.2, 26.0, and 33.3± 0.2° 2? using Cu Ka radiation.
5. The salt of claim 3 or 4, wherein the X-ray powder diffraction pattern further comprises peaks at about 6.2, 9.7, 13.2, 14.3, 15.4, 16.3, 16.9, 18.9, 19.5, 20.7, 21.8, 22.8, 23.6, 25.1, 27.3, 27.7, 28.4, 29.4, 30.2, 31.2, 31.5, 31.9, 33.9, 34.5, 34.9, 36.1, 36.8, 37.7, 38.5, and 39.7± 0.2° 2? using Cu Ka radiation.
6. The salt of any one of claims 1 to 5, having an X-ray powder diffraction pattern substantially as shown in
NZ750950A 2013-03-14 2014-03-14 Omecamtiv Mecarbil and Salts Thereof and Their Uses NZ750950B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361785763P 2013-03-14 2013-03-14
US61/785,763 2013-03-14
NZ711225A NZ711225A (en) 2013-03-14 2014-03-14 A pharmaceutical formulation comprising omecamtiv mecarbil, processess and uses

Publications (2)

Publication Number Publication Date
NZ750950A NZ750950A (en) 2020-09-25
NZ750950B2 true NZ750950B2 (en) 2021-01-06

Family

ID=

Similar Documents

Publication Publication Date Title
US11884630B2 (en) Heterocyclic compounds and their uses
MX2007000911A (en) Nebivolol and its pharmaceutically acceptable salts, process for preparation and pharmaceutical compositions of nebivolol.
AU2014240104B2 (en) Heterocyclic compounds and their uses
KR20090083918A (en) Crystalline forms of aliskiren hemifumarate
WO2019020706A1 (en) Pharmaceutical composition comprising sacubitril and valsartan
JP2018515566A (en) Pharmaceutical composition
US20090030057A1 (en) Pharmaceutical composition of telmisartan
NZ750950B2 (en) Omecamtiv Mecarbil and Salts Thereof and Their Uses
HUE034353T2 (en) Formulation comprising benzothiazolone compound
US20200002343A1 (en) Crystalline Form of Ribociclib Succinate
KR20240027792A (en) Pharmaceutical compositions and oral dosage forms of ketamine derivatives
CN116211814A (en) Compound medicine preparation containing hypertension
MXPA06005545A (en) Solid pharmaceutical preparation form.