Pharmaceutical compositions

Information

  • Patent Grant
  • 11564886
  • Patent Number
    11,564,886
  • Date Filed
    Wednesday, December 16, 2020
    3 years ago
  • Date Issued
    Tuesday, January 31, 2023
    a year ago
Abstract
The present invention relates to pharmaceutical compositions comprising fixed dose combinations of a DPP-4 inhibitor drug and/or a SGLT-2 inhibitor drug, and metformin XR, processes for the preparation thereof, and their use to treat certain diseases.
Description

The present invention relates to pharmaceutical compositions containing a fixed dose combination (FDC) comprising a DPP-4 inhibitor drug (particularly 1-[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-(3-(R)-amino-piperidin-1-yl)-xanthine, also named linagliptin) and/or a SGLT-2 inhibitor drug (particularly 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, also named Compound “A” herein), and metformin (particularly metformin hydrochloride) in extended release form (metformin XR); processes for the preparation thereof, and their use to treat certain diseases.


In particular, the present invention relates to a pharmaceutical composition comprising a fixed dose combination of an extended release form of metformin hydrochloride, optionally seal coated, which is further coated with an immediate release form of 1-[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-(3-(R)-amino-piperidin-1-yl)-xanthine (linagliptin) and/or 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene (Compound “A”).


Further, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as e.g. a tablet), comprising or consisting essentially of

    • a) an inner extended release core comprising metformin (particularly metformin hydrochloride) and one or more excipients;
    • b) an optional intermediate seal coating; and
    • c) an outer immediate release coating comprising at least one active pharmaceutical ingredient selected from
      • a DPP-4 inhibitor, preferably linagliptin, and
      • a SGLT-2 inhibitor, preferably Compound “A”,
      • and one or more excipients.


In a more detailed aspect, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as a tablet) of a selected dipeptidyl peptidase-4 (DPP-4) inhibitor (preferably linagliptin, particularly in immediate release form) and metformin (particularly metformin hydrochloride) in extended release form (metformin XR). In one embodiment of this aspect, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as a tablet), comprising a fixed dose combination of an extended release form of metformin hydrochloride, optionally seal coated, and further coated with an immediate release form of linagliptin.


In another more detailed aspect, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as a tablet) of a selected SGLT-2 inhibitor (preferably 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, particularly in immediate release form) and metformin (particularly metformin hydrochloride) in extended release form (metformin XR). In one embodiment of this aspect, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as a tablet), comprising a fixed dose combination of an extended release form of metformin hydrochloride, optionally seal coated, and further coated with an immediate release form of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene.


In a further more detailed aspect, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as e.g. a tablet), comprising


a first component, part or composition comprising metformin (particularly metformin hydrochloride) in extended release form and one or more excipients, and


a second component, part or composition comprising a selected dipeptidyl peptidase-4 (DPP-4) inhibitor (preferably linagliptin), particularly in immediate release form, and one or more excipients.


In particular, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as a tablet), comprising an extended release form of metformin hydrochloride, optionally seal coated, and further coated with an immediate release form of linagliptin.


In another further more detailed aspect, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as e.g. a tablet), comprising


a first component, part or composition comprising metformin (particularly metformin hydrochloride) in extended release form and one or more excipients, and


a second component, part or composition comprising a selected SGLT-2 inhibitor (preferably 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene), particularly in immediate release form, and one or more excipients.


In particular, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as a tablet), comprising an extended release form of metformin hydrochloride, optionally seal coated, and further coated with an immediate release form of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene.


In a yet further more detailed aspect, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as e.g. a tablet), comprising

    • a) an inner extended release core comprising metformin (particularly metformin hydrochloride) and one or more excipients,
    • b) an optional seal coating, and
    • c) an outer immediate release coating comprising a selected dipeptidyl peptidase-4 (DPP-4) inhibitor (preferably linagliptin) and one or more excipients.


In another yet further more detailed aspect, the present invention relates to a pharmaceutical composition, particularly a solid preparation (e.g. an oral solid dosage form, such as e.g. a tablet), comprising

    • a) an inner extended release core comprising metformin (particularly metformin hydrochloride) and one or more excipients,
    • b) an optional seal coating, and
    • c) an outer immediate release coating comprising a selected SGLT-2 inhibitor (preferably 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene) and one or more excipients.


Particularly, the pharmaceutical compositions of this invention comprise an inner core formulation of metformin hydrochloride comprising a swellable and/or extended release material.


In an embodiment, the pharmaceutical compositions of this invention comprise an inner extended release core which is a formulation (e.g. matrix formulation) comprising metformin hydrochloride, a swellable and/or extended release material, and one or more further excipients.


Particularly, the pharmaceutical compositions of this invention comprise an outer coat of active pharmaceutical ingredient (API) (linagliptin and/or 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene) in an immediate release polymer film.


Further, the present invention relates to a coating process (e.g. coating technology and processing conditions) and immediate release coating formulations of active pharmaceutical ingredients (API) in low doses (typically in doses of 0.5 to 25 mg) on top of tablet cores comprising active pharmaceutical ingredients (API) in high doses (typically in doses of 500-1500 mg) preferably, but not exclusively on extended release tablets. Anyhow, essential parts of the formulation and the process of this invention may be also applicable to any other fixed dose combination with the described setting.


An aim of the present invention is to provide a pharmaceutical composition comprising a combination of a selected DPP-4 inhibitor (preferably linagliptin, particularly in immediate release form), and metformin (particularly metformin hydrochloride) in extended release form. Another aim of the present invention is to provide a pharmaceutical composition comprising a combination of a selected SGLT-2 inhibitor (preferably 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, particularly in immediate release form), and metformin (particularly metformin hydrochloride) in extended release form.


The objectives of are to identify suitable formulations and processing conditions, such as e.g. of a coat of linagliptin or of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene on top metformin XR cores, providing adequate:

    • Chemical stability of the API (particularly linagliptin) in the API film coat,
    • Assay of linagliptin or 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene in the API film-coat (e.g. 95-105%),
    • Content uniformity of linagliptin or 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene (e.g. RSD<3%) in the API film-coat,
    • Low defect rate of the API-film during film coating process,
    • Fast dissolution of the API from the API film-coat and no changes of XR Metformin HCl dissolution, due to the API coating with immediate release of linagliptin or 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene,
    • Processing aspects of coating process/technology, processing conditions and immediate release API (linagliptin or Compound “A”) coating formulations (API film coat),
    • Processing aspects of coating process/technology, processing conditions and immediate release API (linagliptin or Compound “A”) coating formulations on top of metformin extended release tablets.


A particular objective of the present invention is to provide a pharmaceutical composition and suitable coating process with very broad range of drug substance (linagliptin or Compound “A”)/drug substance (metformin) ratio: 1:400-1:40. And the ratio of very low dosed API, e.g. linagliptin with 1 mg or 2.5 mg to very high dosed metformin with 1000 mg and more. And the suitable immediate release dissolution of the low dosed API with high dosed extended release metformin.


The unit dosage strength of the metformin hydrochloride for incorporation into the fixed-dose combination of the present invention is 500, 750, 850 or 1000 milligrams, or even more (e.g. 1500 mg).


These unit dosage strengths of metformin hydrochloride represent the dosage strengths approved in the U.S. for marketing to treat Type 2 diabetes.


The unit dosage strength of linagliptin for incorporation into the fixed-dose combination of the present invention is 2.5 or 5 milligrams, or even less (e.g. 0.5 mg or 1 mg).


The unit dosage strength of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene for incorporation into the fixed-dose combination of the present invention is 5, 10, 12.5 or 25 milligrams.


Specific embodiments of dosage strengths for linagliptin and metformin hydrochloride in the fixed-dose combinations of the present invention are the following:


(1) 5 milligrams of linagliptin and 1000 milligrams metformin hydrochloride;


(2) 2.5 milligrams of linagliptin and 1000 milligrams metformin hydrochloride;


(3) 2.5 milligrams of linagliptin and 750 milligrams metformin hydrochloride.


Specific embodiments of dosage strengths for 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and metformin hydrochloride in the fixed-dose combinations of the present invention are the following:


(1) 25 milligrams of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 1000 milligrams metformin hydrochloride;


(2) 12.5 milligrams of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 1000 milligrams metformin hydrochloride;


(3) 12.5 milligrams of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 750 milligrams metformin hydrochloride;


(4) 10 milligrams of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 1000 milligrams metformin hydrochloride;


(5) 10 milligrams of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 750 milligrams metformin hydrochloride;


(6) 5 milligrams of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 1000 milligrams metformin hydrochloride;


(7) 5 milligrams of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 750 milligrams metformin hydrochloride.


(a) Metformin Part:


The first part in the present invention is a part (composition, particularly solid composition, e.g. a solid pharmaceutical composition for oral administration, e.g. tablet) comprising metformin (particularly metformin hydrochloride) in extended release form, particularly an extended release formulation of metformin.


Exemplary extended release formulations of metformin are disclosed in U.S. Pat. Nos. 6,340,475; 6,488,962; 6,635,280; 6,723,340; 7,780,987; 6,866,866; 6,495,162; 6,790,459; 6,866,866; 6,475,521; and 6,660,300; the disclosures of which are incorporated herein in their entireties.


A particular extended release formulation of metformin is described in U.S. Pat. No. 6,723,340, the disclosure of which is incorporated herein in its entirety.


In an embodiment, the fixed-dose combination products of the present invention comprise—as first part—an inner core matrix formulation with metformin hydrochloride dispersed therein, said matrix formulation containing an extended release material. The matrix formulation is compressed into a tablet form.


In particular, the fixed-dose combination products of the present invention comprise—as first part—an inner core extended release formulation comprising metformin hydrochloride, hydroxypropyl methylcellulose (hypromellose), polyethylene oxide, microcrystalline cellulose, and magnesium stearate.


A particular extended release formulation of metformin is described in U.S. Pat. No. 6,723,340 as follows:


In an embodiment, the extended release material of the matrix comprises poly(ethylene oxide) and/or hydroxypropyl methylcellulose (HPMC), preferably a combination of poly(ethylene oxide) and hydroxypropyl methylcellulose (HPMC), preferably at a weight ratio that causes the matrix to swell upon contact with gastric fluid to a size large enough to provide gastric retention.


The poly(ethylene oxide) component of the matrix may limit initial release of the drug and may impart gastric retention through swelling. The hydroxypropyl methylcellulose (HPMC) component may lower the amount of poly(ethylene oxide) required while still allowing the swelling to occur.


Preferably, the poly(ethylene oxide) has a viscosity average molecular weight of from about 2,000,000 to about 10,000,000 daltons, more preferably from about 4,000,000 to about 7,000,000 daltons.


Preferably, the hydroxypropyl methylcellulose (HPMC) has a viscosity of from about 4,000 centipoise to about 200,000 centipoise, more preferably from about 50,000 to about 200,000 centipoise, even more preferably 80,000 centipoise to about 120,000 centipoise, measured as a 2% solution in water.


More preferably, the poly(ethylene oxide) has a viscosity average molecular weight of from about 4,000,000 to about 7,000,000 daltons, and the hydroxypropyl methylcellulose (HPMC) has a viscosity of from about 80,000 centipoise to about 120,000 centipoise, measured as a 2% solution in water.


In an embodiment, the weight ratio of the poly(ethylene oxide) to hydroxypropyl methylcellulose (HPMC) is within the range from about 1:3 to 3:1, preferably 1:2 to 2:1.


In a further embodiment, the weight ratio of the poly(ethylene oxide) and hydroxypropyl methylcellulose (HPMC) in combination constitutes from about 15% to about 90%, or from about 30% to about 65%, or from about 40% to about 50%, by weight of the metformin part.


Tablet cores in accordance with this invention can be prepared by common tabletting methods that involve mixing, comminution, and fabrication steps commonly practiced by and well known to those skilled in the art of manufacturing drug formulations. Examples of such techniques are:


(1) Direct compression using appropriate punches and dies, typically fitted to a suitable rotary tabletting press;


(2) Injection or compression molding;


(3) Granulation by fluid bed, by low or high shear granulation, or by roller compaction, followed by compression; and


(4) Extrusion of a paste into a mold or to an extrudate to be cut into lengths.


When tablets are made by direct compression, the addition of lubricants may be helpful and is sometimes important to promote powder flow and to prevent breaking of the tablet when the pressure is relieved. Examples of typical lubricants are magnesium stearate (in a concentration of from 0.25% to 3% by weight, preferably about 1% or less by weight, in the powder mix), stearic acid (0.5% to 3% by weight), and hydrogenated vegetable oil (preferably hydrogenated and refined triglycerides of stearic and palmitic acids at about 1% to 5% by weight, most preferably about 2% by weight).


Additional excipients may be added, such as e.g. granulating aids (e.g. low molecular weight HPMC at 2-5% by weight), binders (e.g. microcrystalline cellulose), and additives to enhance powder flowability, tablet hardness, and tablet friability and to reduce adherence to the die wall.


An exemplary extended release metformin tablet core comprises metformin hydrochloride, a combination of poly(ethylene oxide) and hydroxypropyl methylcellulose (e.g. Methocel K100M) as a matrix for a swellable extended release tablet, microcrystalline cellulose as binder, low molecular weight hydroxypropyl methylcellulose (e.g. Methocel E5) as granulating aid, and magnesium stearate as lubricant.


The composition of a representative metformin core tablet is provided as follows:


metformin hydrochloride, e.g. 49.97% by weight of the first part,


poly(ethylene oxide), e.g. 26.50% by weight of the first part,


hydroxypropyl methylcellulose (e.g. Methocel K100M), e.g. 16.08% by weight of the first part,


microcrystalline cellulose, e.g. 4.99% by weight of the first part,


low molecular weight hydroxypropyl methylcellulose (e.g. Methocel E5), e.g. 1.70% by weight of the first part, and


magnesium stearate, e.g. 0.75% by weight of the first part.


Tablets may be formulated by dry blending a granulation comprising metformin hydrochloride and low molecular weight HPMC (e.g. Methocel E5) and the remaining excipients listed above, followed by pressing on a tablet press.


Such an extended release matrix formulation of metformin is disclosed in U.S. Pat. No. 6,723,340 (e.g. Example 3), the disclosure of which is incorporated herein in its entirety.


As further example of a lubricant sodium stearyl fumarate may be mentioned (e.g. at about 0.25-3% by weight).


In a further embodiment, the metformin extended release formulation allows for targeted, controlled delivery of metformin to the upper gastrointestinal (GI) tract. In a further embodiment, the metformin extended release formulation is a hydrogel matrix system and contains a swelling hydrophilic polymer and further excipients, which may allow the metformin tablet core to be retained in the stomach (‘gastric retention’) for approximately eight to nine hours. During this time, the tablet core's metformin is steadily delivered to the upper GI tract at the desired rate and time, without potentially irritating ‘burst’ of drug. This gradual, extended release typically allows for more of the metformin drug to be absorbed in the upper GI tract and minimizes the amount of drug that passes through to the lower GI tract.


(b1) Linagliptin Part:


In one variant, the second part in the present invention is a part (composition, particularly film coat) comprising linagliptin in immediate release form.


In a particular embodiment, the fixed-dose combination products of the present invention comprise—as second part—a film coat formulation of linagliptin, said film coat formulation comprising linagliptin, a stabilizer for stabilizing linagliptin (e.g. a basic and/or nucleophilic excipient, preferably L-arginine as stabilizer), a film-coating agent (such as e.g. hydroxypropyl methylcellulose, e.g. Hypromellose 2910, Methocel E5, or Methocel E15), a plasticizer (such as e.g. polyethylene glycol, e.g. Macrogol 400, 6000 or 8000, or propylene glycol), and, optionally, a glidant (such as e.g. talc).


In an embodiment, the weight ratio of the L-arginine to linagliptin is within the range from about 2:1 to about 1:1, up to about 0.2:1.


The composition of a representative linagliptin containing film coat is provided as follows:

    • linagliptin, e.g. 2.5 mg or 5 mg;
    • L-arginine, e.g. depending from need of stabilizer amount, e.g. in the range from about 0.5 mg to about 10 mg (e.g. 5 mg);
    • hydroxypropyl methylcellulose (e.g. Methocel E5, Methocel E15, or Pharmacoat 603 or 606), e.g. from about 25 mg to about 40 mg (especially from 34.5 mg to 38 mg, or 34.5 mg);
    • polyethylene glycol (e.g. Macrogol 400, 6000 or 8000), e.g. from about 0 to about 12 mg;
    • propylene glycol, e.g. from about 0 mg to about 15 mg (especially 9 mg); and
    • talc, e.g. from about 0 mg to about 15 mg (especially 9 mg).


Depending from need of stabilizer the amount of L-arginine may be in the range from 0.5 mg to 10 mg. With different dose and different arginine amount, the arginine amount may be substituted by hydroxypropyl methylcellulose (HPMC).


In an embodiment, polyethylene glycol and propylene glycol are mutually exclusive in above composition, i.e. if polyethylene glycol is present then propylene glycol is absent, or if propylene glycol is present then polyethylene glycol is absent.


The composition of a representative linagliptin containing film coat suspension further comprises water, e.g. from about 240 mg to about 1440 mg, especially in the range from 904 mg to 1440 mg. The total solids concentration of the suspension is from about 4% to about 12.5% w/w, especially from 4% to 6% w/w. Viscosity may be from about 10 mPas to 110 mPas (e.g. 46-56 mPas).


The sum solids of the linagliptin coating suspension is from about 50 mg to about 120 mg. For example, the sum solids is 60 mg of solid amount of the film coating suspension for 2.5 mg linagliptin, and 120 mg sum solid amount of the film coating suspension for 5 mg linagliptin. Therefore with the same formulation of linagliptin and double coating time (i.e. double amount of coating suspension) it is possible to prepare the higher dose range of linagliptin. Hence different dosage strengths can be achieved by altering coating (spraying) times.


(b2) 1-Chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene Part:


In another variant, the second part in the present invention is a part (composition, particularly film coat) comprising 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene in immediate release form.


In another particular embodiment, the fixed-dose combination products of the present invention comprise—as second part—a film coat formulation of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, said film coat formulation comprising 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, a film-coating agent (such as e.g. hydroxypropyl methylcellulose, e.g. Hypromellose 2910, Methocel E5, or Methocel E15), a plasticizer (such as e.g. polyethylene glycol, e.g. Macrogol 400, 6000 or 8000, or propylene glycol), and, optionally, a glidant (such as e.g. talc).


The composition of a representative 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene containing film coat is provided as follows:

    • 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, e.g. 5 mg, 10 mg, 12.5 mg or 25 mg;
    • optionally, L-arginine, e.g. from about 5 mg to about 25 mg;
    • hydroxypropyl methylcellulose (e.g. Methocel E5, Methocel E15, or Pharmacoat 603 or 606), e.g. from about 25 mg to about 40 mg (especially from 34.5 mg to 38 mg, or 34.5 mg);
    • polyethylene glycol (e.g. Macrogol 400, 6000 or 8000), e.g. from about 0 to about 12 mg;
    • propylene glycol, e.g. from about 0 mg to about 15 mg (especially 9 mg); and
    • talc, e.g. from about 0 mg to about 15 mg (especially 9 mg).


With different dose and different arginine amount, the arginine amount may be substituted by hydroxypropyl methylcellulose (HPMC).


In an embodiment, polyethylene glycol and propylene glycol are mutually exclusive in above composition, i.e. if polyethylene glycol is present then propylene glycol is absent, or if propylene glycol is present then polyethylene glycol is absent.


The composition of a representative 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene containing film coat suspension further comprises water, e.g. from about 240 mg to about 1440 mg, especially in the range from 904 mg to 1440 mg. The total solids concentration of the suspension is from about 4% to about 12.5% w/w, especially from 4% to 6% w/w.


The sum solids of the 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene coating suspension is from about 50 mg to about 120 mg. For example, the sum solids is 60 mg of solid amount of the film coating suspension for 12.5 mg 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, and 120 mg sum solid amount of the film coating suspension for 25 mg 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene. Therefore with the same formulation of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and double coating time (i.e. double amount of coating suspension) it is possible to prepare the higher dose range of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene. Hence different dosage strengths can be achieved by altering coating (spraying) times.


L-Arginine is preferably necessary for the stabilization of linagliptin. Alternatively, a seal coat may be used between the metformin XR core and the linagliptin-containing film coat. In one embodiment, a seal coat is present between the metformin XR core and the linagliptin-containing film coat (optionally further containing L-arginine). In another embodiment, the seal coat is absent between the metformin XR core and the linagliptin-containing film coat (preferably further containing L-arginine).


For Compound “A” preferably no arginine is necessary. For Compound “A” the seal coating of metformin XR cores is optional. In one embodiment, a seal coat is present between the metformin XR core and the Compound “A” containing film coat. In another embodiment, the seal coat is absent between the metformin XR core and the Compound “A” containing film coat.


Alternatively, for the API (linagliptin or Compound “A”) containing film coat, a film coat comprising a mixture of hydroxypropylcellulose and hydroxypropyl methylcellulose, or a mixture of polyvinyl alcohol (PVA) and polyethylene glycol (PEG); or a commercial film-coat such as Opadry®, Opadry II® or other Opardy IR film coat, which are formulated powder blends provided by Colorcon, may be used. With Opadry II or PVA based API coating higher solid concentrations and shorter coating time durations are possible, therefore it works in a range of 10-30%, especially 20% solid concentration. This higher solid concentration, e.g. 20%, typically results in a shorter coating time, e.g. 2-5 hours.


For example, further versions of API-containing film coat compositions comprising one or more of the following ingredients of Tables 1 or 2 may be provided, e.g. as follows from Tables 1 or 2:









TABLE 1







Example formulations for API-coating of linagliptin on top of metformin XR cores















PEG-








containing
PG-
PG-





PEG-
version
containing
containing





containing
(reduced
version
version
Further
Further



version
arginine)
(low DL)
(high DL)
version
version


Composition
(e.g. 2.5
(e.g. 5
(e.g. 2.5
(e.g. 2.5
(e.g. 2.5
(e.g. 5


(% w/w)
mg API)
mg API)
mg API)
mg API)
mg API)
mg API)
















Linagliptin
4.20
4.39
4.55
5.29
4.16
4.16


HPMC (e.g
67.23 
70.18 
72.73 
70.55 




Pharmacoat 615)*








HPMC (e.g




57.5
57.5


Methocel E5)








Polyethylene glycol
20.17 
21.05 


15
15


(e.g. PEG 6000)








Propylene glycol


3.64
3.53




L-Arginine
8.40
4.39
9.09
10.58 
8.33
8.33


Talc


10.00 
10.05 
15
15


Purified water
**
**
**
**
**
**


Total
100.00 
100.00 
100.00 
100.00 
100.00
100.00


Solid content of
5.95
5.70
5.50
5.67
4.0
4.0


suspension (%)





*Alternative Methocel E15


**Solvent is a volatile component, which does not remain in the final product






In one embodiment of the API coatings of this invention, the film-coating agent used is highly viscous.


In another embodiment of the API coatings of this invention, the film-coating agent used is low viscous.









TABLE 2







Further Example formulations for API-coating of


linagliptin on top of metformin XR cores:













PEG-






containing
PG-
PG-



PEG-
version
containing
containing



containing
(reduced
version
version



version
arginine)
(low DL)
(high DL)



(e.g.
(e.g.
(e.g.
(e.g.


Composition
2.5 mg
5 mg
2.5 mg
2.5 mg


(% w/w)
API)
API)
API)
API)














Linagliptin
4.20
4.39
4.55
5.29


HPMC (e.g.
67.23
70.18
72.73
70.55


Pharmacoat






615)






Polyethylene
20.17
21.05




glycol






(e.g. PEG






6000)






Propylene


3.64
3.53


glycol






L-Arginine
8.40
4.39
9.09
10.58


Talc


10.00
10.05


Purified water
**
**
**
**


Total
100.00
100.00
100.00
100.00


Solid content
5.95
5.70
5.50
5.67


of suspension






(%)





** Solvent is a volatile component, which does not remain in the final product






Film coating suspensions/solutions of API (linagliptin or Compound “A”) according to this invention can be prepared by common methods, such as follows:


The film-coating agent hydroxypropyl methylcellulose (HPMC), the plasticizer polyethylene glycol (PEG) (e.g. Macrogol 400, 6000 or 8000) or, as alternative plasticizer, propylene glycol (PG) and water are dissolved and mixed by a suitable mixer (e.g. by propeller mixer) to produce the API-free coating solution. Optionally, the glidant talc suspended in water is added and the obtained suspension is homogenized. Talc may be used optionally. The API (linagliptin or Compound “A”) and—preferably in case of linagliptin—the stabilizer L-arginine are dissolved or suspended in water and added to the aqueous solution of HPMC, PEG or PG, and, optional talc, and dispersed by a suitable mixer (e.g. by propeller mixer) to provide the API coating suspension.


Alternatively, the film-coating agent hydroxypropyl methylcellulose (HPMC) and water are dissolved and mixed by a suitable mixer (e.g. by Ultraturrax).


The stabilizer L-arginine (which is present in case of linagliptin, and may be absent in case of Compound “A”), the plasticizer polyethylene glycol (PEG) (e.g. Macrogol 400, 6000 or 8000) or propylene glycol (PG), optional talc, and water are dispersed, e.g. by homogenization using e.g. ultra turrax.


After degassing of the HPMC solution (or directly after manufacturing of the HPMC solution), the aqueous suspension of PEG or PG, optional L-arginine and optional talc are added to the aqueous HPMC solution and mixed/homogenized.


The API (linagliptin or Compound “A”) is dissolved or suspended in water and added to the aqueous solution of HPMC, PEG or PG, optional L-arginine and optional talc to provide the API coating suspension.


The film-coating operation is carried out in a conventional film coater. The API (linagliptin or Compound “A”) coating suspension/solution are coated at metformin XR cores via coating process.


Preliminary preheating of the cores may be necessary, due to need of equilibrium of water amount of the cores.


The spray rate and air flow through the coating pan is adjusted to produce a uniform coating and coverage of the entire width of the tablet bed. The amount of the coating suspension applied can be controlled by percent weight gain of tablet cores and typically ranges from about 4 to about 12.5%.


In one aspect, this range results in linagliptin drug assay close to the desired 2.5 mg or 5 mg with a standard deviation of about 2-4% for content uniformity assay of linagliptin. The duration of the coating step is about 4-10 hours. The duration of the coating step depends on batch size, process parameters like spray rate and solid concentrations of the coating suspension.


In another aspect, this range results in Compound “A” drug assay close to the desired 5 mg, 12.5 mg, 10 mg or 25 mg with a standard deviation of about 2-4% for content uniformity assay of Compound “A”. The duration of the coating step is about 4-10 hours. The duration of the coating step depends on batch size, process parameters like spray rate and solid concentrations of the coating suspension.


The API coating suspension is applied to the tablet cores containing the metformin XR formulation and the amount of solids deposited in the API film layer is controlled to achieve the desired API doses.


The weight of the cores and film coated tablets may be controlled by percent weight gain during the coating process. Instead of or in addition to weight gain method a PAT method, e.g. online NIR or Raman method for end point detection of assay of API may be used.


An optional seal coat may separate the metformin XR core from the API-containing film coat. Typically, for the preparation of film-coated tablets a coating suspension is prepared and the tablet cores may be coated with the seal coating suspension using standard film coater. The film coating solvent is a volatile component, which does not remain in the final product. A typical seal film-coat comprises a film coating agent, a plasticizer, and, optionally, a glidant, one or more pigments and/or colors.


The metformin XR core may be seal coated using a seal coating agent (and a plasticizer), such as with a mixture of hydroxypropylcellulose and hydroxypropyl methylcellulose, a mixture of polyvinyl alcohol (PVA) and polyethylene glycol (PEG), a mixture of hydroxypropyl methylcellulose and either polyethylene glycol (PEG) or propylene glycol (PG), or any other suitable immediate-release film-coating agent(s). A commercial film-coat is Opadry®, Opadry II® or other Opardy IR film coat, which are formulated powder blend provided by Colorcon. Optionally the seal coat may further comprise a glidant.


The final pharmaceutical compositions of the present invention are tablets. Such tablets may be further film-coated with a final film over-coat, such as with a mixture of hydroxypropylcellulose and hydroxypropyl methylcellulose containing titanium dioxide and/or other coloring agents, such as iron oxides, dyes, and lakes; a mixture of polyvinyl alcohol (PVA) and polyethylene glycol (PEG) containing titanium dioxide and/or other coloring agents, such as iron oxides, dyes, and lakes; a mixture of hydroxypropyl methylcellulose and either polyethylene glycol (PEG) or propylene glycol (PG) containing titanium dioxide and/or other coloring agents, such as iron oxides, dyes, and lakes; or any other suitable immediate-release film-coating agent(s). The coat may provide taste masking and additional stability to the final tablet. A commercial film-coat is Opadry®, Opadry II® or other Opardy IR film coat, which are formulated powder blend provided by Colorcon.


Preferably, for the preparation of film-coated tablets a coating suspension is prepared and the tablet cores are coated with the coating suspension, typically for the API-free film over-coat to a weight gain of about 2-4%, preferably about 3%, using standard film coater. The film coating solvent is a volatile component, which does not remain in the final product. A typical film-coat comprise a film coating agent, a plasticizer, and, optionally, a glidant, one or more pigments and/or colors. For example, the film coat may comprise hydroxypropylmethylcellulose (HPMC), propylene glycol or polyethylene glycol, talc and, optionally, titanium dioxide and/or iron oxide (e.g. iron oxide yellow and/or red).


The pharmaceutical tablet compositions of the present invention may also contain one or more additional formulation ingredients selected from a wide variety of excipients known in the pharmaceutical formulation art. According to the desired properties of the pharmaceutical composition, any number of ingredients may be selected, alone or in combination, based upon their known uses in preparing tablet compositions. Such ingredients include, but are not limited to, diluents, compression aids, glidants, disintegrants, lubricants, flavors, flavor enhancers, sweeteners, and preservatives.


The term “tablet” as used herein is intended to encompass compressed pharmaceutical dosage formulations of all shapes and sizes.


The present invention also provides methods particularly for treating Type 2 diabetes by orally administering to a host in need of such treatment a therapeutically effective amount of one of the fixed-dose combination pharmaceutical compositions of the present invention. In one embodiment the host in need of such treatment is a human. In another embodiment the pharmaceutical composition is in the dosage form of a tablet. The pharmaceutical compositions comprising the fixed-dose combination may be administered once-daily (QD), twice-daily (BID), thrice-daily (TID), or four-times daily.


Manufacture and Polymorph


The term “linagliptin” as employed herein refers to linagliptin, a pharmaceutically acceptable salt thereof, a hydrate or solvate thereof, or a polymorphic form thereof. Crystalline forms are described in WO 2007/128721. Preferred crystalline forms are the polymorphs A and B described therein. In particular, linagliptin is the free base 1-[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-(3-(R)-amino-piperidin-1-yl)-xanthine. As linagliptin or a pharmaceutically acceptable salt thereof, linagliptin is preferred. Methods for the manufacture of linagliptin are described in the patent applications WO 2004/018468 and WO 2006/048427 for example.


1-[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-(3-(R)-amino-piperidin-1-yl)-xanthine (Linagliptin)



embedded image


According to this invention, it is to be understood that the definition of the SGLT2 inhibitor, in particular 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene (Compound “A”), also comprises its hydrates, solvates and polymorphic forms thereof, and prodrugs thereof. With regard to the preferred 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene an advantageous crystalline form is described in the international patent application WO 2006/117359 which hereby is incorporated herein in its entirety. This crystalline form possesses good solubility properties which enable a good bioavailability of the SGLT2 inhibitor. Furthermore, the crystalline form is physico-chemically stable and thus provides a good shelf-life stability of the pharmaceutical composition.


1-Chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene (Compound “A”)



embedded image


Methods for the manufacture of SGLT2 inhibitors according to this invention and of prodrugs thereof are known to the one skilled in the art. Advantageously, the compounds according to this invention can be prepared using synthetic methods as described in the literature, including patent applications as cited hereinbefore. Preferred methods of manufacture, in particular of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, are described in the WO 2006/120208.


For avoidance of any doubt, the disclosure of each of the foregoing documents cited above in connection with the specified SGLT2 or DPP-4 inhibitors is specifically incorporated herein by reference in its entirety.


Indications


As described herein by the administration of the pharmaceutical composition according to this invention, therapeutic effects can be achieved, which make it useful for treating and/or preventing certain diseases, disorders or conditions, such as e.g. those described herein.


Therefore, a treatment or prophylaxis according to this invention is advantageously suitable in those patients in need of such treatment or prophylaxis who are diagnosed of one or more of the conditions selected from the group consisting of overweight and obesity, in particular class I obesity, class II obesity, class III obesity, visceral obesity and abdominal obesity. In addition a treatment or prophylaxis according to this invention is advantageously suitable in those patients in which a weight increase is contraindicated. The pharmaceutical composition as well as the methods according to the present invention allow a reduction of the HbA1c value to a desired target range, for example <7% and preferably <6.5%, for a higher number of patients and for a longer time of therapeutic treatment compared with a corresponding monotherapy.


The pharmaceutical composition according to this invention and in particular the active ingredients therein exhibits a very good efficacy with regard to glycemic control, in particular in view of a reduction of fasting plasma glucose, postprandial plasma glucose and/or glycosylated hemoglobin (HbA1c). By administering a pharmaceutical composition according to this invention, a reduction of HbA1c equal to or greater than preferably 0.5%, even more preferably equal to or greater than 1.0% can be achieved and the reduction is particularly in the range from 1.0% to 2.0%.


Furthermore, the method and/or use according to this invention is advantageously applicable in those patients who show one, two or more of the following conditions:

  • (a) a fasting blood glucose or serum glucose concentration greater than 110 mg/dL, in particular greater than 125 mg/dL;
  • (b) a postprandial plasma glucose equal to or greater than 140 mg/dL;
  • (c) an HbA1c value equal to or greater than 6.5%, in particular equal to or greater than 7.0%, especially equal to or greater than 7.5%, even more particularly equal to or greater than 8.0%.


The present invention also discloses the use of the pharmaceutical composition for improving glycemic control in patients having type 2 diabetes or showing first signs of pre-diabetes. Thus, the invention also includes diabetes prevention. If therefore a pharmaceutical composition according to this invention is used to improve the glycemic control as soon as one of the above-mentioned signs of pre-diabetes is present, the onset of manifest type 2 diabetes mellitus can be delayed or prevented.


Furthermore, the pharmaceutical composition according to this invention is particularly suitable in the treatment of patients with insulin dependency, i.e. in patients who are treated or otherwise would be treated or need treatment with an insulin or a derivative of insulin or a substitute of insulin or a formulation comprising an insulin or a derivative or substitute thereof. These patients include patients with diabetes type 2 and patients with diabetes type 1.


Therefore, according to a preferred embodiment of the present invention, there is provided a method for improving glycemic control and/or for reducing of fasting plasma glucose, of postprandial plasma glucose and/or of glycosylated hemoglobin HbA1c in a patient in need thereof who is diagnosed with impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG) with insulin resistance, with metabolic syndrome and/or with type 2 or type 1 diabetes mellitus characterized in that a pharmaceutical composition as defined hereinbefore and hereinafter is administered to the patient.


According to another preferred embodiment of the present invention, there is provided a method for improving glycemic control in patients, in particular in adult patients, with type 2 diabetes mellitus as an adjunct to diet and exercise.


Therefore, the method and/or use according to this invention is advantageously applicable in those patients who show one, two or more of the following conditions:

  • (a) insufficient glycemic control with diet and exercise alone;
  • (b) insufficient glycemic control despite oral monotherapy with metformin, in particular despite oral monotherapy at a maximal tolerated dose of metformin;
  • (c) insufficient glycemic control despite oral monotherapy with another antidiabetic agent, in particular despite oral monotherapy at a maximal tolerated dose of the other antidiabetic agent.


The lowering of the blood glucose level by the administration of a pharmaceutical composition according to this invention is insulin-independent. Therefore, a pharmaceutical composition according to this invention is particularly suitable in the treatment of patients who are diagnosed having one or more of the following conditions

    • insulin resistance,
    • hyperinsulinemia,
    • pre-diabetes,
    • type 2 diabetes mellitus, particular having a late stage type 2 diabetes mellitus,
    • type 1 diabetes mellitus.


Furthermore, a pharmaceutical composition according to this invention is particularly suitable in the treatment of patients who are diagnosed having one or more of the following conditions

  • (a) obesity (including class I, II and/or III obesity), visceral obesity and/or abdominal obesity,
  • (b) triglyceride blood level ≥150 mg/dL,
  • (c) HDL-cholesterol blood level <40 mg/dL in female patients and <50 mg/dL in male patients,
  • (d) a systolic blood pressure ≥130 mm Hg and a diastolic blood pressure 85 mm Hg,
  • (e) a fasting blood glucose level ≥110 mg/dL.


It is assumed that patients diagnosed with impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), with insulin resistance and/or with metabolic syndrome suffer from an increased risk of developing a cardiovascular disease, such as for example myocardial infarction, coronary heart disease, heart insufficiency, thromboembolic events. A glycemic control according to this invention may result in a reduction of the cardiovascular risks.


A pharmaceutical composition according to this invention exhibits a good safety profile. Therefore, a treatment or prophylaxis according to this invention is advantageously possible in those patients for which the mono-therapy with another antidiabetic drug is contraindicated and/or who have an intolerance against such drugs at therapeutic doses. In particular, a treatment or prophylaxis according to this invention may be advantageously possible in those patients showing or having an increased risk for one or more of the following disorders: renal insufficiency or diseases, cardiac diseases, cardiac failure, hepatic diseases, pulmonal diseases, catabolytic states and/or danger of lactate acidosis, or female patients being pregnant or during lactation.


Furthermore, it can be found that the administration of a pharmaceutical composition according to this invention results in no risk or in a low risk of hypoglycemia. Therefore, a treatment or prophylaxis according to this invention is also advantageously possible in those patients showing or having an increased risk for hypoglycemia.


A pharmaceutical composition according to this invention is particularly suitable in the long term treatment or prophylaxis of the diseases and/or conditions as described hereinbefore and hereinafter, in particular in the long term glycemic control in patients with type 2 diabetes mellitus.


The term “long term” as used hereinbefore and hereinafter indicates a treatment of or administration in a patient within a period of time longer than 12 weeks, preferably longer than 25 weeks, even more preferably longer than 1 year.


Therefore, a particularly preferred embodiment of the present invention provides a method for therapy, preferably oral therapy, for improvement, especially long term improvement, of glycemic control in patients with type 2 diabetes mellitus, especially in patients with late stage type 2 diabetes mellitus, in particular in patients additionally diagnosed of overweight, obesity (including class I, class II and/or class III obesity), visceral obesity and/or abdominal obesity.


According to another aspect of the invention, there is provided a method for preventing, slowing the progression of, delaying or treating of a condition or disorder selected from the group consisting of complications of diabetes mellitus such as cataracts and micro- and macrovascular diseases, such as dyslipidemia, nephropathy, retinopathy, neuropathy, tissue ischaemia, diabetic foot, arteriosclerosis, myocardial infarction, acute coronary syndrome, unstable angina pectoris, stable angina pectoris, stroke, peripheral arterial occlusive disease, cardiomyopathy, heart failure, heart rhythm disorders and vascular restenosis, in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient. In particular one or more aspects of diabetic nephropathy such as hyperperfusion, proteinuria and albuminuria may be treated, their progression slowed or their onset delayed or prevented. The term “tissue ischaemia” particularly comprises diabetic macroangiopathy, diabetic microangiopathy, impaired wound healing and diabetic ulcer. The terms “micro- and macrovascular diseases” and “micro- and macrovascular complications” are used interchangeably in this application.


According to another aspect of the invention, there is provided a method for preventing, slowing the progression of, delaying or treating a metabolic disorder selected from the group consisting of type 2 diabetes mellitus, impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), hyperglycemia, postprandial hyperglycemia, overweight, obesity, metabolic syndrome, gestational diabetes and diabetes related to cystic fibrosis in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient.


According to another aspect of the invention, there is provided a method for improving glycemic control and/or for reducing of fasting plasma glucose, of postprandial plasma glucose and/or of glycosylated hemoglobin HbA1c in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient.


The pharmaceutical composition according to this invention may also have valuable disease-modifying properties with respect to diseases or conditions related to impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), insulin resistance and/or metabolic syndrome.


According to another aspect of the invention, there is provided a method for preventing, slowing, delaying or reversing progression from impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), insulin resistance and/or from metabolic syndrome to type 2 diabetes mellitus in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient.


As by the use of a pharmaceutical composition according to this invention, an improvement of the glycemic control in patients in need thereof is obtainable, also those conditions and/or diseases related to or caused by an increased blood glucose level may be treated.


By the administration of a pharmaceutical composition according to this invention excessive blood glucose levels are not converted to insoluble storage forms, like fat, but excreted through the urine of the patient. It can be seen that loss of fat may account for the majority of the observed weight loss whereas no significant changes in body water or protein content are observed. Therefore, no gain in weight or even a reduction in body weight is the result.


According to another aspect of the invention, there is provided a method for reducing body weight and/or body fat or preventing an increase in body weight and/or body fat or facilitating a reduction in body weight and/or body fat in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient.


By the administration of a combination or pharmaceutical composition according to the present invention, an abnormal accumulation of ectopic fat, in particular of the liver, may be reduced or inhibited. Therefore, according to another aspect of the present invention, there is provided a method for preventing, slowing, delaying or treating diseases or conditions attributed to an abnormal accumulation of ectopic fat, in particular of the liver, in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient. Diseases or conditions which are attributed to an abnormal accumulation of liver fat are particularly selected from the group consisting of general fatty liver, non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), hyperalimentation-induced fatty liver, diabetic fatty liver, alcoholic-induced fatty liver or toxic fatty liver.


Another aspect of the invention provides a method for maintaining and/or improving the insulin sensitivity and/or for treating or preventing hyperinsulinemia and/or insulin resistance in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient.


According to another aspect of the invention, there is provided a method for preventing, slowing progression of, delaying, or treating new onset diabetes after transplantation (NODAT) and/or post-transplant metabolic syndrome (PTMS) in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient.


According to a further aspect of the invention, there is provided a method for preventing, delaying, or reducing NODAT and/or PTMS associated complications including micro- and macrovascular diseases and events, graft rejection, infection, and death in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient.


The pharmaceutical composition according to the invention is capable of facilitating the lowering of serum total urate levels in the patient. Therefore according to another aspect of the invention, there is provided a method for treating hyperuricemia and hyperuricemia-associated conditions, such as for example gout, hypertension and renal failure, in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient.


The administration of a pharmaceutical composition increases the urine excretion of glucose. This increase in osmotic excretion and water release and the lowering of urate levels are beneficial as a treatment or prevention for kidney stones. Therefore in a further aspect of the invention, there is provided a method for treating or preventing kidney stones in a patient in need thereof characterized in that a pharmaceutical composition according to the invention is administered to the patient.


The invention also relates to a pharmaceutical composition according to this invention for use in a method as described hereinbefore and hereinafter.


The invention also relates to a use of a pharmaceutical composition according to this invention for the manufacture of a medicament for use in a method as described hereinbefore and hereinafter.







DEFINITIONS

The term “active ingredient” of a pharmaceutical composition according to the present invention means the SGLT2 inhibitor, the DPP-4 inhibitor and/or metformin according to the present invention.


The term “body mass index” or “BMI” of a human patient is defined as the weight in kilograms divided by the square of the height in meters, such that BMI has units of kg/m2.


The term “overweight” is defined as the condition wherein the individual has a BMI greater than or 25 kg/m2 and less than 30 kg/m2. The terms “overweight” and “pre-obese” are used interchangeably.


The term “obesity” is defined as the condition wherein the individual has a BMI equal to or greater than 30 kg/m2. According to a WHO definition the term obesity may be categorized as follows: the term “class I obesity” is the condition wherein the BMI is equal to or greater than 30 kg/m2 but lower than 35 kg/m2; the term “class II obesity” is the condition wherein the BMI is equal to or greater than 35 kg/m2 but lower than 40 kg/m2; the term “class III obesity” is the condition wherein the BMI is equal to or greater than 40 kg/m2.


The term “visceral obesity” is defined as the condition wherein a waist-to-hip ratio of greater than or equal to 1.0 in men and 0.8 in women is measured. It defines the risk for insulin resistance and the development of pre-diabetes.


The term “abdominal obesity” is usually defined as the condition wherein the waist circumference is >40 inches or 102 cm in men, and is >35 inches or 94 cm in women. With regard to a Japanese ethnicity or Japanese patients abdominal obesity may be defined as waist circumference 85 cm in men and 90 cm in women (see e.g. investigating committee for the diagnosis of metabolic syndrome in Japan).


The term “euglycemia” is defined as the condition in which a subject has a fasting blood glucose concentration within the normal range, greater than 70 mg/dL (3.89 mmol/L) and less than 100 mg/dL (5.6 mmol/L). The word “fasting” has the usual meaning as a medical term.


The term “hyperglycemia” is defined as the condition in which a subject has a fasting blood glucose concentration above the normal range, greater than 100 mg/dL (5.6 mmol/L). The word “fasting” has the usual meaning as a medical term.


The term “hypoglycemia” is defined as the condition in which a subject has a blood glucose concentration below the normal range, in particular below 70 mg/dL (3.89 mmol/L) or even below 60 mg/dl.


The term “postprandial hyperglycemia” is defined as the condition in which a subject has a 2 hour postprandial blood glucose or serum glucose concentration greater than 200 mg/dL (11.1 mmol/L).


The term “impaired fasting blood glucose” or “IFG” is defined as the condition in which a subject has a fasting blood glucose concentration or fasting serum glucose concentration in a range from 100 to 125 mg/dl (i.e. from 5.6 to 6.9 mmol/l), in particular greater than 110 mg/dL and less than 126 mg/dl (7.00 mmol/L). A subject with “normal fasting glucose” has a fasting glucose concentration smaller than 100 mg/dl, i.e. smaller than 5.6 mmol/l.


The term “impaired glucose tolerance” or “IGT” is defined as the condition in which a subject has a 2 hour postprandial blood glucose or serum glucose concentration greater than 140 mg/dl (7.8 mmol/L) and less than 200 mg/dL (11.11 mmol/L). The abnormal glucose tolerance, i.e. the 2 hour postprandial blood glucose or serum glucose concentration can be measured as the blood sugar level in mg of glucose per dL of plasma 2 hours after taking 75 g of glucose after a fast. A subject with “normal glucose tolerance” has a 2 hour postprandial blood glucose or serum glucose concentration smaller than 140 mg/dl (7.8 mmol/L).


The term “hyperinsulinemia” is defined as the condition in which a subject with insulin resistance, with or without euglycemia, has fasting or postprandial serum or plasma insulin concentration elevated above that of normal, lean individuals without insulin resistance, having a waist-to-hip ratio <1.0 (for men) or <0.8 (for women).


The terms “insulin-sensitizing”, “insulin resistance-improving” or “insulin resistance-lowering” are synonymous and used interchangeably.


The term “insulin resistance” is defined as a state in which circulating insulin levels in excess of the normal response to a glucose load are required to maintain the euglycemic state (Ford E S, et al. JAMA. (2002) 287:356-9). A method of determining insulin resistance is the euglycaemic-hyperinsulinaemic clamp test. The ratio of insulin to glucose is determined within the scope of a combined insulin-glucose infusion technique. There is found to be insulin resistance if the glucose absorption is below the 25th percentile of the background population investigated (WHO definition). Rather less laborious than the clamp test are so called minimal models in which, during an intravenous glucose tolerance test, the insulin and glucose concentrations in the blood are measured at fixed time intervals and from these the insulin resistance is calculated. With this method, it is not possible to distinguish between hepatic and peripheral insulin resistance.


Furthermore, insulin resistance, the response of a patient with insulin resistance to therapy, insulin sensitivity and hyperinsulinemia may be quantified by assessing the “homeostasis model assessment to insulin resistance (HOMA-IR)” score, a reliable indicator of insulin resistance (Katsuki A, et al. Diabetes Care 2001; 24: 362-5). Further reference is made to methods for the determination of the HOMA-index for insulin sensitivity (Matthews et al., Diabetologia 1985, 28: 412-19), of the ratio of intact proinsulin to insulin (Forst et al., Diabetes 2003, 52(Suppl. 1): A459) and to an euglycemic clamp study. In addition, plasma adiponectin levels can be monitored as a potential surrogate of insulin sensitivity. The estimate of insulin resistance by the homeostasis assessment model (HOMA)-IR score is calculated with the formula (Galvin P, et al. Diabet Med 1992; 9:921-8):

HOMA-IR=[fasting serum insulin(μU/mL)]×[fasting plasma glucose(mmol/L)/22.5]


As a rule, other parameters are used in everyday clinical practice to assess insulin resistance. Preferably, the patient's triglyceride concentration is used, for example, as increased triglyceride levels correlate significantly with the presence of insulin resistance.


Patients with a predisposition for the development of IGT or IFG or type 2 diabetes are those having euglycemia with hyperinsulinemia and are by definition, insulin resistant. A typical patient with insulin resistance is usually overweight or obese, but this is not always the case. If insulin resistance can be detected, this is a particularly strong indication of the presence of pre-diabetes. Thus, it may be that in order to maintain glucose homoeostasis a person have e.g. 2-3 times as high endogenous insulin production as a healthy person, without this resulting in any clinical symptoms.


The methods to investigate the function of pancreatic beta-cells are similar to the above methods with regard to insulin sensitivity, hyperinsulinemia or insulin resistance: An improvement of beta-cell function can be measured for example by determining a HOMA-index for beta-cell function (Matthews et al., Diabetologia 1985, 28: 412-19), the ratio of intact proinsulin to insulin (Forst et al., Diabetes 2003, 52(Suppl. 1): A459), the insulin/C-peptide secretion after an oral glucose tolerance test or a meal tolerance test, or by employing a hyperglycemic clamp study and/or minimal modeling after a frequently sampled intravenous glucose tolerance test (Stumvoll et al., Eur J Clin Invest 2001, 31: 380-81).


The term “pre-diabetes” is the condition wherein an individual is pre-disposed to the development of type 2 diabetes. Pre-diabetes extends the definition of impaired glucose tolerance to include individuals with a fasting blood glucose within the high normal range 100 mg/dL (J. B. Meigs, et al. Diabetes 2003; 52:1475-1484) and fasting hyperinsulinemia (elevated plasma insulin concentration). The scientific and medical basis for identifying pre-diabetes as a serious health threat is laid out in a Position Statement entitled “The Prevention or Delay of Type 2 Diabetes” issued jointly by the American Diabetes Association and the National Institute of Diabetes and Digestive and Kidney Diseases (Diabetes Care 2002; 25:742-749).


Individuals likely to have insulin resistance are those who have two or more of the following attributes: 1) overweight or obese, 2) high blood pressure, 3) hyperlipidemia, 4) one or more 1st degree relative with a diagnosis of IGT or IFG or type 2 diabetes. Insulin resistance can be confirmed in these individuals by calculating the HOMA-IR score. For the purpose of this invention, insulin resistance is defined as the clinical condition in which an individual has a HOMA-IR score >4.0 or a HOMA-IR score above the upper limit of normal as defined for the laboratory performing the glucose and insulin assays.


The term “type 1 diabetes” is defined as the condition in which a subject has, in the presence of autoimmunity towards the pancreatic beta-cell or insulin, a fasting blood glucose or serum glucose concentration greater than 125 mg/dL (6.94 mmol/L). If a glucose tolerance test is carried out, the blood sugar level of a diabetic will be in excess of 200 mg of glucose per dL (11.1 mmol/l) of plasma 2 hours after 75 g of glucose have been taken on an empty stomach, in the presence of autoimmunity towards the pancreatic beta cell or insulin. In a glucose tolerance test 75 g of glucose are administered orally to the patient being tested after 10-12 hours of fasting and the blood sugar level is recorded immediately before taking the glucose and 1 and 2 hours after taking it. The presence of autoimmunity towards the pancreatic beta-cell may be observed by detection of circulating islet cell autoantibodies [“type 1A diabetes mellitus”], i.e., at least one of: GAD65 [glutamic acid decarboxylase-65], ICA [islet-cell cytoplasm], IA-2 [intracytoplasmatic domain of the tyrosine phosphatase-like protein IA-2], ZnT8 [zinc-transporter-8] or anti-insulin; or other signs of autoimmunity without the presence of typical circulating autoantibodies [type 1B diabetes], i.e. as detected through pancreatic biopsy or imaging). Typically a genetic predisposition is present (e.g. HLA, INS VNTR and PTPN22), but this is not always the case.


The term “type 2 diabetes” is defined as the condition in which a subject has a fasting blood glucose or serum glucose concentration greater than 125 mg/dL (6.94 mmol/L). The measurement of blood glucose values is a standard procedure in routine medical analysis. If a glucose tolerance test is carried out, the blood sugar level of a diabetic will be in excess of 200 mg of glucose per dL (11.1 mmol/l) of plasma 2 hours after 75 g of glucose have been taken on an empty stomach. In a glucose tolerance test 75 g of glucose are administered orally to the patient being tested after 10-12 hours of fasting and the blood sugar level is recorded immediately before taking the glucose and 1 and 2 hours after taking it. In a healthy subject, the blood sugar level before taking the glucose will be between 60 and 110 mg per dL of plasma, less than 200 mg per dL 1 hour after taking the glucose and less than 140 mg per dL after 2 hours. If after 2 hours the value is between 140 and 200 mg, this is regarded as abnormal glucose tolerance.


The term “late stage type 2 diabetes mellitus” includes patients with a secondary drug failure, indication for insulin therapy and progression to micro- and macrovascular complications e.g. diabetic nephropathy, or coronary heart disease (CHD).


The term “HbA1c” refers to the product of a non-enzymatic glycation of the haemoglobin B chain. Its determination is well known to one skilled in the art. In monitoring the treatment of diabetes mellitus the HbA1c value is of exceptional importance. As its production depends essentially on the blood sugar level and the life of the erythrocytes, the HbA1c in the sense of a “blood sugar memory” reflects the average blood sugar levels of the preceding 4-6 weeks. Diabetic patients whose HbA1c value is consistently well adjusted by intensive diabetes treatment (i.e. <6.5% of the total haemoglobin in the sample), are significantly better protected against diabetic microangiopathy. For example, metformin on its own achieves an average improvement in the HbA1c value in the diabetic of the order of 1.0-1.5%. This reduction of the HbA1C value is not sufficient in all diabetics to achieve the desired target range of <6.5% and preferably <6% HbA1c.


The term “insufficient glycemic control” or “inadequate glycemic control” in the scope of the present invention means a condition wherein patients show HbA1c values above 6.5%, in particular above 7.0%, even more preferably above 7.5%, especially above 8%.


The “metabolic syndrome”, also called “syndrome X” (when used in the context of a metabolic disorder), also called the “dysmetabolic syndrome” is a syndrome complex with the cardinal feature being insulin resistance (Laaksonen D E, et al. Am J Epidemiol 2002; 156:1070-7). According to the ATP III/NCEP guidelines (Executive Summary of the Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) JAMA: Journal of the American Medical Association (2001) 285:2486-2497), diagnosis of the metabolic syndrome is made when three or more of the following risk factors are present:

    • 1. Abdominal obesity, defined as waist circumference >40 inches or 102 cm in men, and >35 inches or 94 cm in women; or with regard to a Japanese ethnicity or Japanese patients defined as waist circumference 85 cm in men and 90 cm in women;
    • 2. Triglycerides: ≥150 mg/dL
    • 3. HDL-cholesterol <40 mg/dL in men
    • 4. Blood pressure ≥130/85 mm Hg (SBP≥130 or DBP≥85)
    • 5. Fasting blood glucose ≥100 mg/dL


The NCEP definitions have been validated (Laaksonen D E, et al. Am J Epidemiol. (2002) 156:1070-7). Triglycerides and HDL cholesterol in the blood can also be determined by standard methods in medical analysis and are described for example in Thomas L (Editor): “Labor and Diagnose”, TH-Books Verlagsgesellschaft mbH, Frankfurt/Main, 2000.


According to a commonly used definition, hypertension is diagnosed if the systolic blood pressure (SBP) exceeds a value of 140 mm Hg and diastolic blood pressure (DBP) exceeds a value of 90 mm Hg. If a patient is suffering from manifest diabetes it is currently recommended that the systolic blood pressure be reduced to a level below 130 mm Hg and the diastolic blood pressure be lowered to below 80 mm Hg.


The definitions of NODAT (new onset diabetes after transplantation) and PTMS (post-transplant metabolic syndrome) follow closely that of the American Diabetes Association diagnostic criteria for type 2 diabetes, and that of the International Diabetes Federation (IDF) and the American Heart Association/National Heart, Lung, and Blood Institute, for the metabolic syndrome. NODAT and/or PTMS are associated with an increased risk of micro- and macrovascular disease and events, graft rejection, infection, and death. A number of predictors have been identified as potential risk factors related to NODAT and/or PTMS including a higher age at transplant, male gender, the pre-transplant body mass index, pre-transplant diabetes, and immunosuppression.


The term “gestational diabetes” (diabetes of pregnancy) denotes a form of the diabetes which develops during pregnancy and usually ceases again immediately after the birth. Gestational diabetes is diagnosed by a screening test which is carried out between the 24th and 28th weeks of pregnancy. It is usually a simple test in which the blood sugar level is measured one hour after the administration of 50 g of glucose solution. If this 1 h level is above 140 mg/dl, gestational diabetes is suspected. Final confirmation may be obtained by a standard glucose tolerance test, for example with 75 g of glucose.


The term “hyperuricemia” denotes a condition of high serum total urate levels. In human blood, uric acid concentrations between 3.6 mg/dL (ca. 214 μmol/L) and 8.3 mg/dL (ca. 494 μmol/L) are considered normal by the American Medical Association. High serum total urate levels, or hyperuricemia, are often associated with several maladies. For example, high serum total urate levels can lead to a type of arthritis in the joints known as gout. Gout is a condition created by a build up of monosodium urate or uric acid crystals on the articular cartilage of joints, tendons and surrounding tissues due to elevated concentrations of total urate levels in the blood stream. The build up of urate or uric acid on these tissues provokes an inflammatory reaction of these tissues. Saturation levels of uric acid in urine may result in kidney stone formation when the uric acid or urate crystallizes in the kidney. Additionally, high serum total urate levels are often associated with the so-called metabolic syndrome, including cardiovascular disease and hypertension.


The term “hyponatremia” denotes a condition of a positive balance of water with or without a deficit of sodium, which is recognized when the plasma sodium falls below the level of 135 mml/L. Hyponatremia is a condition which can occur in isolation in individuals that over-consume water; however, more often hyponatremia is a complication of medication or other underlying medical condition that leas to a diminished excretion of water. Hyponatremia may lead to water intoxication, which occurs when the normal tonicity of extracellular fluid falls below the safe limit, due to retention of excess water. Water intoxication is a potentially fatal disturbance in brain function. Typical symptoms of water intoxication include nausea, vomiting, headache and malaise.


The terms “treatment” and “treating” comprise therapeutic treatment of patients having already developed said condition, in particular in manifest form. Therapeutic treatment may be symptomatic treatment in order to relieve the symptoms of the specific indication or causal treatment in order to reverse or partially reverse the conditions of the indication or to stop or slow down progression of the disease. Thus the compositions and methods of the present invention may be used for instance as therapeutic treatment over a period of time as well as for chronic therapy.


The terms “prophylactically treating”, “preventivally treating” and “preventing” are used interchangeably and comprise a treatment of patients at risk to develop a condition mentioned hereinbefore, thus reducing said risk.

Claims
  • 1. A pharmaceutical composition comprising: a) an inner extended release core, wherein the inner extended release core is a formulation comprising metformin hydrochloride, a swellable and/or extended release polymer, and one or more further excipients;b) an intermediate seal coating, wherein the seal coating comprises a film-coating agent, a plasticizer, and, optionally, a glidant, one or more pigments and/or colors, wherein the film coating agent and the plasticizer are a mixture of hydroxypropylcellulose and hydroxypropyl methylcellulose; andc) an outer immediate release coating, wherein the outer immediate release coating is a film coat formulation comprising 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, a film-coating agent, a plasticizer, and, optionally, a glidant.
  • 2. The pharmaceutical composition according to claim 1, wherein the film-coating agent is hydroxypropyl methylcellulose.
  • 3. The pharmaceutical composition according to claim 1, wherein the plastizicer is polyethylene glycol.
  • 4. The pharmaceutical composition according to claim 1, wherein the plastizicer is propylene glycol.
  • 5. The pharmaceutical composition according to claim 1, wherein the optional glidant is talc.
  • 6. The pharmaceutical composition according to claim 1, wherein the metformin hydrochloride is present in a unit dosage strength of 500, 750, 850, 1000 or 1500 mg.
  • 7. The pharmaceutical composition according to claim 1, wherein 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and is present in a unit dosage strength of 5, 10, 12.5 or 25 mg.
  • 8. The pharmaceutical composition according to claim 1, which is a tablet for oral administration.
  • 9. The tablet according to claim 8 further comprising an outer film over-coat.
  • 10. The tablet according to claim 9, wherein the outer film over-coat comprises a film-coating agent, a plasticizer, and, optionally, a glidant, one or more pigments and/or colors.
  • 11. A method for treating, preventing, slowing the progression, or delaying the onset of metabolic diseases either in type 2 diabetes patients who have not been previously treated with an antihyperglycemic agent, or In type 2 diabetes patients with insufficient glycemic control despite therapy with one or two conventional antihyperglycemic agents selected from metformin, sulphonylureas, thiazolidinediones, glinides, alpha-glucosidase blockers, GLP-1 or GLP-1 analogues, and insulin or insulin analogues comprising administering the pharmaceutical composition of claim 1 to a subject.
  • 12. The method of claim 11, wherein the metabolic disease is type 2 diabetes mellitus and conditions related thereto caused by an increased blood glucose level.
  • 13. The pharmaceutical composition according to claim 3, wherein the polyethylene glycol is Macrogol 400, 6000 or 8000.
  • 14. The pharmaceutical composition according to claim 1, wherein the dosage strength is 5 mg of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 1000 mg metformin hydrochloride.
  • 15. The pharmaceutical composition according to claim 1, wherein the dosage strength is 10 mg of 1-chloro-4-β-D-glucopyranos-1-yl)-2-[4((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 1000 mg metformin hydrochloride.
  • 16. The pharmaceutical composition according to claim 1, wherein the dosage strength is 12.5 mg of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 1000 mg metformin hydrochloride.
  • 17. The pharmaceutical composition according to claim 1, wherein the dosage strength is 25 mg of 1-chloro-4-(β-D-glucopyranos-1-yl)-2-[4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and 1000 mg metformin hydrochloride.
  • 18. The pharmaceutical composition according to claim 1, wherein the outer immediate release coating comprises 1-chloro-4-(β-D-glucopyranos-1-yl)-2[-4-((S)-tetrahydrofuran-3-yloxy)-benzyl]-benzene and linagliptin.
  • 19. The pharmaceutical composition according to claim 18, wherein the dosage strength is 2.5 mg of linagliptin.
  • 20. The pharmaceutical composition according to claim 18, wherein the dosage strength is 5 mg of linagliptin.
Priority Claims (2)
Number Date Country Kind
11157240 Mar 2011 EP regional
11158358 Mar 2011 EP regional
US Referenced Citations (255)
Number Name Date Kind
3174901 Sterne Mar 1965 A
3884906 Van Der Meer et al. May 1975 A
4379785 Weyer et al. Apr 1983 A
4602023 Kiely et al. Jul 1986 A
4639436 Junge et al. Jan 1987 A
4786023 Harris et al. Nov 1988 A
4786755 Kiely et al. Nov 1988 A
4802924 Woznicki et al. Feb 1989 A
5807580 Luber Sep 1998 A
6303661 Demuth et al. Oct 2001 B1
6348090 Grillo et al. Feb 2002 B1
6414126 Ellsworth et al. Jul 2002 B1
6448323 Jordan et al. Sep 2002 B1
6515117 Ellsworth et al. Feb 2003 B2
6613806 Aven et al. Sep 2003 B1
6627611 Tomiyama et al. Sep 2003 B2
6774112 Gougoutas Aug 2004 B2
6794480 Goto et al. Sep 2004 B2
6890898 Bachovchin et al. May 2005 B2
6936590 Washbum et al. Aug 2005 B2
6972283 Fujikura et al. Dec 2005 B2
6995183 Hamann et al. Feb 2006 B2
7109192 Hauel et al. Sep 2006 B2
7169761 Tomiyama et al. Jan 2007 B2
7173028 Dahmann et al. Feb 2007 B2
7202350 Imamura et al. Apr 2007 B2
7371732 Eickelmann et al. May 2008 B2
7375087 Teranishi et al. May 2008 B2
7375090 Himmelsbach et al. May 2008 B2
7375213 Deshpande et al. May 2008 B2
7393836 Eckhardt et al. Jul 2008 B2
7407955 Himmelsbach et al. Aug 2008 B2
7417032 Eckhardt et al. Aug 2008 B2
7419959 Eckhardt et al. Sep 2008 B2
7482337 Himmelsbach et al. Jan 2009 B2
7501426 Himmelsbach et al. Mar 2009 B2
7541341 Fushimi et al. Jun 2009 B2
7579449 Eckhardt et al. Aug 2009 B2
7589193 Washbur et al. Sep 2009 B2
7662790 Himmelsbach et al. Feb 2010 B2
7674486 Bhaskaran et al. Mar 2010 B2
7683160 Eckhardt et al. Mar 2010 B2
7687469 Eckhardt et al. Mar 2010 B2
7713938 Himmelsbach et al. May 2010 B2
7723309 Himmelsbach et al. May 2010 B2
7745414 Eckhardt et al. Jun 2010 B2
7772191 Eckhardt et al. Aug 2010 B2
7772192 Esko Aug 2010 B2
7772378 Himmelsbach et al. Aug 2010 B2
7772407 Imamura et al. Aug 2010 B2
7776830 Eckhardt et al. Aug 2010 B2
7820815 Pfrengle et al. Oct 2010 B2
7847074 Eckhardt et al. Dec 2010 B2
7851502 Bindra et al. Dec 2010 B2
7851602 Himmelsbach et al. Dec 2010 B2
7858587 Eckhardt et al. Dec 2010 B2
7879806 Himmelsbach et al. Feb 2011 B2
7879807 Himmelsbach et al. Feb 2011 B2
8039441 Himmelsbach et al. Oct 2011 B2
8071583 Himmelsbach Dec 2011 B2
8106060 Pfrengle et al. Jan 2012 B2
8119648 Himmelsbach et al. Feb 2012 B2
8178541 Himmelsbach et al. May 2012 B2
8232281 Dugi et al. Jul 2012 B2
8283326 Eckhardt et al. Oct 2012 B2
8507450 Eckhardt et al. Aug 2013 B2
8551957 Dugi et al. Oct 2013 B2
8557782 Eckhardt et al. Oct 2013 B2
8802842 Weber et al. Aug 2014 B2
9155705 Friedl et al. Oct 2015 B2
9555001 Ito et al. Jan 2017 B2
9949997 Broedl et al. Apr 2018 B2
9949998 Broedl et al. Apr 2018 B2
10258637 Broedl et al. Apr 2019 B2
10406172 Eickelmann et al. Sep 2019 B2
10596120 Ito et al. Mar 2020 B2
10610489 Schneider et al. Apr 2020 B2
20010041674 Tomiyama et al. Nov 2001 A1
20010044435 Himmelsbach et al. Nov 2001 A1
20010053791 Babcock et al. Dec 2001 A1
20020137903 Ellsworth et al. Sep 2002 A1
20020173509 Himmelsbach et al. Nov 2002 A1
20020198205 Himmelsbach et al. Dec 2002 A1
20030064935 Gougoutas Apr 2003 A1
20030087843 Washburn May 2003 A1
20030104053 Gusler et al. Jun 2003 A1
20030105077 Kanstrup et al. Jun 2003 A1
20030114390 Washburn et al. Jun 2003 A1
20040037883 Zhou et al. Feb 2004 A1
20040087587 Himmelsbach et al. May 2004 A1
20040097510 Himmelsbach et al. May 2004 A1
20040138148 Fushimi et al. Jul 2004 A1
20040138439 Deshpande et al. Jul 2004 A1
20040180925 Matsuno et al. Sep 2004 A1
20050065098 Fujikura et al. Mar 2005 A1
20050085680 Auerbach et al. Apr 2005 A1
20050124555 Tomiyama et al. Jun 2005 A1
20050130985 Himmelsbach et al. Jun 2005 A1
20050187168 Eickelmann et al. Aug 2005 A1
20050209166 Eckhardt et al. Sep 2005 A1
20050233982 Himmelsbach et al. Oct 2005 A1
20050234108 Himmelsbach et al. Oct 2005 A1
20050256310 Hulin et al. Nov 2005 A1
20060002998 Trehan et al. Jan 2006 A1
20060009400 Eckhardt et al. Jan 2006 A1
20060019948 Eckhardt et al. Jan 2006 A1
20060025349 Eckhardt et al. Feb 2006 A1
20060035841 Eckhardt et al. Feb 2006 A1
20060063722 Washburn et al. Mar 2006 A1
20060074031 Eckhardt et al. Apr 2006 A1
20060079541 Langkopf et al. Apr 2006 A1
20060142210 Eckhardt et al. Jun 2006 A1
20060142310 Pfrengle et al. Jun 2006 A1
20060154866 Chu et al. Jul 2006 A1
20060189548 Himmelsbach et al. Aug 2006 A1
20060205711 Himmelsbach et al. Sep 2006 A1
20060210627 Pfeffer et al. Sep 2006 A1
20060234953 Himmelsbach et al. Oct 2006 A1
20060247226 Himmelsbach et al. Nov 2006 A1
20060251728 Himmelsbach et al. Nov 2006 A1
20060258749 Eckhardt et al. Nov 2006 A1
20070004648 Himmelsbach et al. Jan 2007 A1
20070027092 Himmelsbach et al. Feb 2007 A1
20070027168 Pfrengle et al. Feb 2007 A1
20070049537 Eckhardt et al. Mar 2007 A1
20070054867 Eckhardt et al. Mar 2007 A1
20070060530 Christopher et al. Mar 2007 A1
20070072813 Himmelsbach et al. Mar 2007 A1
20070073046 Eckhardt et al. Mar 2007 A1
20070077296 Folger et al. Apr 2007 A1
20070212411 Fawzy Sep 2007 A1
20070249544 Himmelsbach et al. Oct 2007 A1
20070259821 Eckhardt et al. Nov 2007 A1
20070259900 Sieger et al. Nov 2007 A1
20070264370 Jeffers Nov 2007 A1
20070281940 Dugi et al. Dec 2007 A1
20070293690 Tomiyama et al. Dec 2007 A1
20070299076 Piotrowski et al. Dec 2007 A1
20080058379 Eckhardt et al. Mar 2008 A1
20080107731 Kohlrausch et al. May 2008 A1
20080108816 Zutter May 2008 A1
20080193529 Kowalski et al. Aug 2008 A1
20080207882 Derdau et al. Aug 2008 A1
20080221174 Grenier et al. Sep 2008 A1
20080234367 Washburn et al. Sep 2008 A1
20080249089 Himmelsbach et al. Oct 2008 A1
20080255159 Himmelsbach et al. Oct 2008 A1
20080287529 Deshpande et al. Nov 2008 A1
20090023913 Eckhardt et al. Jan 2009 A1
20090137801 Himmelsbach et al. May 2009 A1
20090192314 Pfrengle et al. Jul 2009 A1
20090318547 Eckhardt et al. Dec 2009 A1
20090326215 Eckhardt et al. Dec 2009 A1
20100069310 Himmelsbach et al. Mar 2010 A1
20100074950 Sesha Mar 2010 A1
20100081625 Wienrich et al. Apr 2010 A1
20100092124 Magnusson et al. Apr 2010 A1
20100093654 Himmelsbach et al. Apr 2010 A1
20100099641 Himmelsbach et al. Apr 2010 A1
20100173916 Himmelsbach et al. Jul 2010 A1
20100179191 Himmelsbach et al. Jul 2010 A1
20100204250 Himmelsbach et al. Aug 2010 A1
20100209506 Eisenreich Aug 2010 A1
20100210662 Baroni et al. Aug 2010 A1
20100240879 Eckhardt et al. Sep 2010 A1
20100249392 Eckhardt et al. Sep 2010 A1
20100298243 Manuchehri et al. Nov 2010 A1
20100317847 Eckhardt et al. Dec 2010 A1
20100330177 Pourkavoos Dec 2010 A1
20110014284 Eisenreich et al. Jan 2011 A1
20110046076 Eickelmann et al. Feb 2011 A1
20110046087 Eickelmann et al. Feb 2011 A1
20110065731 Dugi et al. Mar 2011 A1
20110092510 Klein et al. Apr 2011 A1
20110098240 Dugi et al. Apr 2011 A1
20110112069 Himmelsbach et al. May 2011 A1
20110144083 Himmelsbach et al. Jun 2011 A1
20110144095 Himmelsbach et al. Jun 2011 A1
20110178033 Eckhardt et al. Jul 2011 A1
20110190322 Klein et al. Aug 2011 A1
20110195917 Dugi et al. Aug 2011 A1
20110206766 Friedl et al. Aug 2011 A1
20110236477 Schneider et al. Sep 2011 A1
20110237526 Weber et al. Sep 2011 A1
20110237532 De Vries et al. Sep 2011 A1
20110237789 Weber et al. Sep 2011 A1
20110263493 Dugi et al. Oct 2011 A1
20110263617 Mark et al. Oct 2011 A1
20110275561 Graefe-Mody et al. Nov 2011 A1
20110301182 Dugi Dec 2011 A1
20120003313 Kohlrausch et al. Jan 2012 A1
20120035158 Himmelsbach et al. Feb 2012 A1
20120040982 Himmelsbach et al. Feb 2012 A1
20120041069 Sesha Feb 2012 A1
20120071403 Strumph et al. Mar 2012 A1
20120094894 Graefe-Mody et al. Apr 2012 A1
20120107398 Schneider et al. May 2012 A1
20120121530 Klein et al. May 2012 A1
20120122776 Graefe-Mody et al. May 2012 A1
20120129874 Sieger et al. May 2012 A1
20120142712 Pfrengle et al. Jun 2012 A1
20120165251 Klein et al. Jun 2012 A1
20120196812 Eickelmann et al. Aug 2012 A1
20120208831 Himmelsbach et al. Aug 2012 A1
20120219622 Kohlrausch et al. Aug 2012 A1
20120219623 Meinicke Aug 2012 A1
20120283169 Grempler et al. Nov 2012 A1
20120296080 Eckhardt et al. Nov 2012 A1
20120296091 Sieger et al. Nov 2012 A1
20130035281 Klein et al. Feb 2013 A1
20130035298 Broedl et al. Feb 2013 A1
20130064887 Ito et al. Mar 2013 A1
20130096076 Dugi et al. Apr 2013 A1
20130137646 Wienrich et al. May 2013 A1
20130236543 Ito et al. Sep 2013 A1
20130252908 Mayoux et al. Sep 2013 A1
20140031301 Eickelmann et al. Jan 2014 A1
20140038911 Eickelmann et al. Feb 2014 A1
20140046046 Eckhardt et al. Feb 2014 A1
20140087996 Klein et al. Mar 2014 A1
20140088027 Grempler et al. Mar 2014 A1
20140256624 Grempler et al. Sep 2014 A1
20140303097 Broedl et al. Oct 2014 A1
20140303098 Broedl et al. Oct 2014 A1
20140315832 Broedl et al. Oct 2014 A1
20160000816 Broedl et al. Jan 2016 A1
20160235680 Ito et al. Aug 2016 A1
20170020907 Eickelmann et al. Jan 2017 A1
20170095424 Ito et al. Apr 2017 A1
20170305952 Klein et al. Oct 2017 A1
20180104249 Eisenreich Apr 2018 A1
20180169126 Broedl et al. Jun 2018 A1
20180177794 Wienrich et al. Jun 2018 A1
20180185291 Ito et al. Jul 2018 A1
20180193427 Grempler et al. Jul 2018 A1
20180200278 Broedl et al. Jul 2018 A1
20180289678 Eisenreich et al. Oct 2018 A1
20180344647 Boeck et al. Dec 2018 A1
20190038654 Broedl et al. Feb 2019 A1
20190298749 Mayoux et al. Oct 2019 A1
20190309004 Wirth et al. Oct 2019 A1
20200069713 Eickelmann et al. Mar 2020 A1
20200085851 Eickelmann et al. Mar 2020 A1
20200138770 von Eynatten et al. May 2020 A1
20200138844 Broedl et al. May 2020 A1
20200188306 Schneider et al. Jun 2020 A1
20200222423 Wienrich et al. Jul 2020 A1
20200268777 Broedl et al. Aug 2020 A1
20200297639 Ito et al. Sep 2020 A1
20200360412 Broedl et al. Nov 2020 A1
20200368261 Broedl et al. Nov 2020 A1
20200397809 Mayoux Dec 2020 A1
20200397867 Grempler et al. Dec 2020 A1
20210059974 Broedl et al. Mar 2021 A1
20210267902 Ito et al. Sep 2021 A1
Foreign Referenced Citations (246)
Number Date Country
2382480 Mar 2001 CA
2388818 Apr 2001 CA
2437240 Aug 2002 CA
2435730 Sep 2002 CA
2463989 Apr 2003 CA
2494177 Feb 2004 CA
2496249 Mar 2004 CA
2505389 May 2004 CA
2470365 Jun 2004 CA
2508024 Jun 2004 CA
2508226 Jun 2004 CA
2508233 Jun 2004 CA
2526145 Sep 2004 CA
2539032 Mar 2005 CA
2544480 Jun 2005 CA
2548353 Jul 2005 CA
2555050 Sep 2005 CA
2557269 Sep 2005 CA
2557320 Sep 2005 CA
2557801 Oct 2005 CA
2569915 Jan 2006 CA
2572149 Jan 2006 CA
2572819 Jan 2006 CA
2573777 Feb 2006 CA
2574451 Feb 2006 CA
2576294 Mar 2006 CA
2574500 Apr 2006 CA
2586938 May 2006 CA
2617090 Feb 2007 CA
2649922 Nov 2007 CA
2651019 Nov 2007 CA
2651089 Nov 2007 CA
2696579 Feb 2009 CA
2720450 Oct 2009 CA
2726244 Dec 2009 CA
2732803 Feb 2010 CA
2735562 Feb 2010 CA
2736421 Mar 2010 CA
2738367 Apr 2010 CA
2745037 Jul 2010 CA
2745039 Jul 2010 CA
2750798 Aug 2010 CA
2752437 Aug 2010 CA
2776296 Apr 2011 CA
2782179 Jun 2011 CA
101234105 Aug 2008 CN
2758025 Jul 1979 DE
2951135 Jun 1981 DE
102004044221 Mar 2006 DE
0206567 Dec 1986 EP
1224195 Jul 2002 EP
1344780 Sep 2003 EP
1354888 Oct 2003 EP
1385856 Feb 2004 EP
1406873 Apr 2004 EP
1500403 Jan 2005 EP
1523994 Apr 2005 EP
1553094 Jul 2005 EP
1564210 Aug 2005 EP
1586571 Oct 2005 EP
1609785 Dec 2005 EP
1791852 Jun 2007 EP
1852108 Nov 2007 EP
1852439 Nov 2007 EP
2143443 Jan 2010 EP
2166007 Mar 2010 EP
55007256 Jan 1980 JP
56039056 Apr 1981 JP
58164502 Sep 1983 JP
62030750 Feb 1987 JP
11124392 May 1999 JP
2001288178 Oct 2001 JP
2003511458 Mar 2003 JP
2004196788 Jul 2004 JP
2004359630 Dec 2004 JP
2005002092 Jan 2005 JP
2005060625 Mar 2005 JP
20070111099 Nov 2007 KR
9605873 Feb 1996 WO
9718814 May 1997 WO
9831697 Jul 1998 WO
0116147 Mar 2001 WO
2001027128 Apr 2001 WO
200152825 Jul 2001 WO
2001074834 Oct 2001 WO
200197808 Dec 2001 WO
200202560 Jan 2002 WO
2002053573 Jul 2002 WO
2002064606 Aug 2002 WO
2002068420 Sep 2002 WO
2002083066 Oct 2002 WO
2003004496 Jan 2003 WO
2003020737 Mar 2003 WO
2003024965 Mar 2003 WO
2003031458 Apr 2003 WO
2003032997 Apr 2003 WO
2003035177 May 2003 WO
2003037327 May 2003 WO
2003057200 Jul 2003 WO
2003078404 Sep 2003 WO
2003099836 Dec 2003 WO
2004007517 Jan 2004 WO
2004013118 Feb 2004 WO
2004018468 Mar 2004 WO
2004041820 May 2004 WO
2004046115 Jun 2004 WO
2004050658 Jun 2004 WO
2004052902 Jun 2004 WO
2004052903 Jun 2004 WO
2004063209 Jul 2004 WO
2004065380 Aug 2004 WO
2004076470 Sep 2004 WO
2004080990 Sep 2004 WO
2005000848 Jan 2005 WO
2005011786 Feb 2005 WO
2005012318 Feb 2005 WO
2005012326 Feb 2005 WO
2005021566 Mar 2005 WO
2005049022 Jun 2005 WO
2005051950 Jun 2005 WO
2005063785 Jul 2005 WO
2005067976 Jul 2005 WO
2005085237 Sep 2005 WO
2005085246 Sep 2005 WO
2005085265 Sep 2005 WO
2005092877 Oct 2005 WO
2005116014 Dec 2005 WO
2005117861 Dec 2005 WO
2006002912 Jan 2006 WO
2006005613 Jan 2006 WO
2006006496 Jan 2006 WO
2006008038 Jan 2006 WO
2006010557 Feb 2006 WO
2006011469 Feb 2006 WO
2006018150 Feb 2006 WO
2006024024 Mar 2006 WO
2006029769 Mar 2006 WO
2006034489 Mar 2006 WO
2006037537 Apr 2006 WO
2006040625 Apr 2006 WO
2006047248 May 2006 WO
2006048427 May 2006 WO
2006064033 Jun 2006 WO
2006072334 Jul 2006 WO
2006076231 Jul 2006 WO
2006078593 Jul 2006 WO
2006089872 Aug 2006 WO
2006108842 Oct 2006 WO
2006117359 Nov 2006 WO
2006117360 Nov 2006 WO
2006120208 Nov 2006 WO
2006135693 Dec 2006 WO
2007000445 Jan 2007 WO
2007014886 Feb 2007 WO
2007014894 Feb 2007 WO
2007014895 Feb 2007 WO
2007025943 Mar 2007 WO
2007028814 Mar 2007 WO
2007031548 Mar 2007 WO
2007033350 Mar 2007 WO
2007035665 Mar 2007 WO
2007039417 Apr 2007 WO
2007041053 Apr 2007 WO
2007071738 Jun 2007 WO
2007078726 Jul 2007 WO
2007093610 Aug 2007 WO
2007120702 Oct 2007 WO
2007120936 Oct 2007 WO
2007128721 Nov 2007 WO
2007128724 Nov 2007 WO
2007128749 Nov 2007 WO
2007128761 Nov 2007 WO
2007136151 Nov 2007 WO
2007144175 Dec 2007 WO
2007149797 Dec 2007 WO
2008017670 Feb 2008 WO
2008020011 Feb 2008 WO
2008022267 Feb 2008 WO
2008034859 Mar 2008 WO
2008049923 May 2008 WO
2008055870 May 2008 WO
2008055940 May 2008 WO
2008062273 May 2008 WO
2008089892 Jul 2008 WO
2008090210 Jul 2008 WO
2008093878 Aug 2008 WO
2008093882 Aug 2008 WO
2008101938 Aug 2008 WO
2008101939 Aug 2008 WO
2008101943 Aug 2008 WO
2008113000 Sep 2008 WO
2008116179 Sep 2008 WO
2008131149 Oct 2008 WO
2009022007 Feb 2009 WO
2009022008 Feb 2009 WO
2009022009 Feb 2009 WO
2009022010 Feb 2009 WO
2009024542 Feb 2009 WO
2009035969 Mar 2009 WO
2009063072 May 2009 WO
2009091082 Jul 2009 WO
2009099734 Aug 2009 WO
2009111200 Sep 2009 WO
2009121945 Oct 2009 WO
2009123992 Oct 2009 WO
2009147125 Dec 2009 WO
2010015664 Feb 2010 WO
2010018217 Feb 2010 WO
2010029089 Mar 2010 WO
2010043688 Apr 2010 WO
2010045656 Apr 2010 WO
2010072776 Jul 2010 WO
2010079197 Jul 2010 WO
2010086411 Aug 2010 WO
2010092123 Aug 2010 WO
2010092124 Aug 2010 WO
2010092125 Aug 2010 WO
2010092126 Aug 2010 WO
2010092163 Aug 2010 WO
2010138535 Dec 2010 WO
2010140111 Dec 2010 WO
2010147768 Dec 2010 WO
2011039107 Apr 2011 WO
2011039108 Apr 2011 WO
2011039337 Apr 2011 WO
2011039367 Apr 2011 WO
2011060256 May 2011 WO
2011060290 May 2011 WO
2011064352 Jun 2011 WO
2011113947 Sep 2011 WO
2011117295 Sep 2011 WO
2011120923 Oct 2011 WO
2011138380 Nov 2011 WO
2011138421 Nov 2011 WO
2011161161 Dec 2011 WO
2011163206 Dec 2011 WO
2012031124 Mar 2012 WO
2012062698 May 2012 WO
2012065993 May 2012 WO
2012106303 Aug 2012 WO
2012107476 Aug 2012 WO
2012120040 Sep 2012 WO
2013131967 Sep 2013 WO
2013139777 Sep 2013 WO
2014011926 Jan 2014 WO
2016059219 Apr 2016 WO
Non-Patent Literature Citations (269)
Entry
Abdul-Ghani, Muhammad “Where does Combination Therapy with an SGLT2 Inhibitor Plus a DPP-4 Inhibitor Fit in the Management of Type 2 Diabetes?” (2015) Diabetes Care, 38, 373-375.
Abdul-Ghani, Muhammad A. et al. “Role of Sodium-Glucose Cotransporter 2 (SGLT 2) Inhibitors in the Treatment of Type 2 Diabetes” (2011) Endocrine Reviews, 32(4), 515-531.
Abstract in English for KR20070111099, Nov. 11, 2007.
Adachi, Tetsuya., et al; T-1095, A Renal Na+-Glucose Transporter Inhibitor, Improves Hyperglycemia in Streptozotocin-lnduced Diabetic Rats; Metabolism (2000) vol. 49 No. 8 pp. 990-995.
Ahren B: “DPP-4 inhibitors”, Best practice and research in clinical endocrinology and metabolism—New therapies for diabetes Dec. 2007 GB LNKD—DOI:10.1016/J. Beem.2007.07.005, vol. 21, No. 4, Dec. 2007, pp. 517-533.
Ahren, Bo “Dipeptidyl Peptidase-4 Inhibitors” (2007) Diabetes Care, vol. 30, No. 6, 1344-1350.
Ahren, Bo “Novel Combination Treatment of Type 2 Diabetes DPP-4 Inhibition + Metformin” (2008) Vascular Health and Risk Management, 4(2), 383-394.
Ahren, Bo et al. “Twelve- and 52-Week Efficacy of the Dipeptidyl Peptidase IV Inhibitor LAF237 in Metformin-Treated Patients with Type 2 Diabetes” (2004) Diabetes Care, vol. 27, No. 12, 2874-2880.
American Diabetes Association “Standards of Medical Care in Diabetes—2009” (2009) Diabetes Care, vol. 32, Supplement 1, S-13-S61.
Ashiya, Mona et al. “Non-insulin therapies for type 2 diabetes” (2007) Nature Reviews, Drug Discovery vol. 6, 777-778.
Augeri, David J. “Discovery and Preclinical Profile of Saxagliptin (BMS-477118): A Highly Potent, Long-Acting, Orally Active Dipeptidyl Peptidase IV Inhibitor for the Treatment of Type 2 Diabetes” J. Med. Chem. (2005) vol. 48, pp. 5025-5037.
Aulinger, B.A. et al., “Ex-4 and the DPP-IV Inhibitor Vildagliptin have Additive Effects to Suppress Food Intake in Rodents”. Abstract No. 1545-P, 2008.
AULT Addison, “Techniques and experiments for organic chemistry” University Science Books, 1998, pp. 59-60.
Aulton, Michael E. “Pharmaceutics, The Science of Dosage Form Design” (2002) 2nd Edition, 404-409.
Baati, Rachid et al. “A Convenient Synthesis of 2-Tetrahydrofuranyl Ethers” (2000) Organic Letters, vol. 2, No. 4, 185-487.
Baggio, Laurie L. et al. “Biology of Incretins: GLP-1 and GIP” Gastroenterology (2007) vol. 132, 2131-2157.
Benhaddou, Rachida., et al; Tetra-n-Propylammonium Tetra-Oxoruthenate(VII): A Reagent of Choice for the Oxidation of Diversely Protected Glycopyranoses and Glycofuranoses to Lactones; Carbohydrate Research (1994) vol. 260 pp. 243-250.
Boehringer Ingelheim, “BI Trial No. 1275.1 Final Analysis” Clinical Study Synopsis for Public Disclosure, (2013) UNo: U13-2755-01, 38 pgs.
Brazg, R et al. “Effect of Adding MK-0431 to Ongoing Metformin Therapy in Type 2” (2005) Diabetes, vol. 54, Suppl. 1, A3.
Brazg, R. et al: “Effect of adding sitagliptin, a dipeptidyll peptidase-4 inhibitor, to metformin on 24-h glycaemic control and beta-cell function in patients with type 2 diabetes.” Diabetetes, Obesity and Metabolism, Mar. 2007, vol. 9, No. 2, Mar. 2007 pp. 186-193.
Bristol-Myers Squibb Company, Label “Glucophage (metformin hydrochloride) Tablets, Glucophage XR (metformin hydrochloride) Extended-Release Tablets” Apr. 2017, 35 pgs.
British National Formulary (2008) “6. Endrocrine system” 2 pgs.
Byrn, Stephen et al. “Pharmaceutical Solids: A Strategic Approach to Regulatory Considerations” Pharmaceutical Research, vol. 12, No. 7, (1995) pp. 945-954.
Caira, Mino R. “Crystalline Polymorphism of Organic Compounds” (1998) Topics in Current Chemistry, vol. 198, 164-208.
Campbell, R. Keith “Rationale for Dipeptidyl Peptidase 4 Inhibitors: A New Class of Oral Agents for the Treatment of Type 2 Diabetes Mellitus” (2007) The Annals of Pharmacotherapy, vol. 41, 51-60.
CAS Registry No. 668270-12-0; STN database entered Mar. 28, 2004. 5 pgs.
Castaneda, Francisco et al. “Thioglycosides as inhibitors of hSGLTI and hSGLT2: Potential therapeutic agents for the control of hyperglycemia in diabetes” International Journal of Medical Sciences (2007) 4(3), pp. 131-139.
Charpentier, Guillaume “Oral combination therapy for type 2 diabetes” (2002) Diabetes Metab Res Rev,vol. 18, S70-S76.
Chyan, Yau-Jan, et al. “Dipeptidyl Peptidase-IV Inhibitors: An Evolving Treatment for Tyep 2 Diabetes from the Incretin Concept” (2007) Recent Patents on Endocrine, Metabolic & Immune Drug Discovery, vol. 1, No. 1, 15-24.
Clinical Trials. “View of NCT00601250 on Jan. 25, 2008: Efficacy and Safety of BI 1356 vs Placebo added to Metformin Background Therapy in Patients with Type 2 Diabetes” Clinical Trials. Gov Archive, [Online] Jan. 25, 2008 URL:http://clinicaltrials.gov/archive/NCTO0601250/2008_01_25 [retrieved on Feb. 27, 2009].
Clinical Trials. NCTO0622284. “Efficacy and safety of BI 1356 in combination with metformin in patients with type 2 diabetes” ClinicalTrials.gov (Online) No. NCT00622284, Feb. 13, 2008, p. 1-5, URL:http://clinicaltrial.gov/ct2/show/.
Clinical Trials: NCT00954447, View on Jun. 14, 2010. “Efficacy and Safety of Linagliptin in Combination with Insulin in Patients with Type 2 Diabetes”. <http://clinicaltrials.gov/archive/NCT00954447/2010_06_14>.
Clinical Trials: NCT00309608. Efficacy and safety of BI 1356 in combination with metformin in patients with type2 diabetes. Boehringer Ingelheim Pharmaceuticals, Jan. 27, 2009. Clinical Trials.gov . http://clinicaltrials.gov/archive/NCT00309608/2009_01_27.
Clinical Trials: NCT00328172 “Efficacy and Safety of 3 Doses of BI1356 (Linagliptin) in Type 2 Diabetes Patients” Sponsor: Boehringer Ingelheim, Last Update Posted Mar. 14, 2014, 4 pgs.
Clinical Trials: NCT00554450 “Renal Impairment in Type 2 Diabetic Subjects” Sponsor: AstraZeneca, Last Update Posted Oct. 17, 2016, 5 pgs.
Clinical Trials: NCT00602472. “BI 1356 in Combination with Metformin and a Sulphonylurea in Type 2 Diabetes”. ClinicalTrials.gov processed: Jan. 14, 2009 3 pgs.
Clinical Trials: NCT00602472. “BI 1356 in combination with metformin and a sulphonylurea in Type 2 Diabetes”. DrugLib.com, Nov. 3, 2008. http://www.druglib.com/trial/08/NCT00309608.html.
Clinical Trials: NCT00602472. “History of Changes for Study: NCT00602472. BI 1356 in Combination with Metformin and a Sulphonylurea in Type 2 Diabetes”. Submitted Date: Feb. 11, 2009 7 pgs.
Clinical Trials: NCT00622284. Efficacy and Safety of BI 1356 in Combination with Metformin in Patients with Type 2 Diabetes. Boehringer Ingelheim Pharmaceuticals, Aug. 2008. http://clinicaltrials.gov/archive/NCT00622284/2010_01_13.
Clinical Trials: NCT00798161. “Safety and efficacy of Bi 1356 Plus Metformin in Type 2 Diabetes, Factorial Design”. Clinical Trials.gov archive. A Service of the U.S> National Institutes of Health. Nov. 24, 2008, p. 1-3. http://clinicaltrials.gov/archive/NCT00798161/2008_11_24.
Clinical Trials: NCT01064414 “An Efficacy, Safety and Tolerability Stude of Canagliflozin in Patients with Type 2 Diabetes Mellitus who have Moderate Renal Impairment” Sponsor: Janssen Research & Development LLC, Last Update Posted Aug. 14, 2013, 7 pgs.
Clinical Trials: NCT01210001 “Efficacy and Safety of Empagliflozin (BI 10773) in Type 2 Diabetes Patients on a Background of Pioglitazone Alone or with Metformin” Sponsor: Boehringer Ingelheim, Last Update Posted Jun. 17, 2014, 7 pgs.
Clinical Trials: NCT01422876 “Efficacy and Safety of Empagliflozin (BI 10773) / Linagliptin (BI 1356) Fixed Dose Combination in Treatment naive and Metformin Treated Type 2 Diabetes Patients” Sponsor: Boehringer Ingelheim Pharmaceuticals, Apr. 1, 2015, 4 pgs.
Colorcon; Opadry II Aqueous Film Coating; http://www.colorcon.com/products-formulation/all-products/film-coatings/immediate-release/opadry-Il ; Dec. 31, 2015.
Conarello, S.L. et al., “Mice lacking dipeptidyl peptidase IV are protected against obesity and insulin resistance”. PNAS, May 27, 2003, vol. 100, No. 11, p. 6825-6830.
Consoli, A. et al. “Initiating oral glucose-lowering therapy with metformin in type 2 diabetic patients: an evidence-based strategy to reduce the burden of late-developing diabetes complications” (2004) Diabetes Metab, 30, 509-516.
Cornell, Susan “Vildagliptin (LAF 237): A Dipeptidyl Peptidase IV Inhibitor for the Treatment of Type 2 Diabetes Mellitus” (2006) J Pharm Technol, vol. 22, pp. 105-109.
Crepaldi, G. et al. “Dipeptidyl peptidase 4 (DPP-4) inhibitors and their role in Type 2 diabetes management” (2007) J. Endocrinol Invest, 30, 610-614.
Deacon, Carolyn F. “Perspectives in Diabetes Therapeutic Strategies Based on Glucagon-Like Peptide 1” Diabetes, (2004) vol. 53 pp. 2181-2189.
Defronzo, Ralph A. et al. “Combination of Empagliflozin and Linagliptin as Second-Line Therapy in Subjects with Type 2 Diabetes Inadequately Controlled on Metformin” (2015) Diabetes Care, 38, 384-393.
Ida, Takehiko., et al; Tributylmagnesium Ate Complex-Mediated Novel Bromine-Magnesium Exchange Reaction for Selective Monosubstitution of Dibromoarenes; Tetrahedron Letters (2001) vol. 42 pp. 4841-4844; Pergamon Press.
International Search Report and Written Opinion for PCT/EP2012/053910 dated May 14, 2012.
International Search Report for PCT/EP2005/002618 dated Jun. 30, 2005.
International Search Report for PCT/EP2005/056806 dated Dec. 27, 2006.
International Search Report for PCT/EP2006/061520 dated Jul. 26, 2006.
International Search Report for PCT/EP2006/061956 dated Jul. 5, 2006.
International Search report for PCT/EP2006/061957 dated Jul. 5, 2006.
International Search Report for PCT/EP2006/062191 dated Aug. 8, 2006.
International Search Report for PCT/EP2006/064702 dated Jul. 26, 2007.
International Search Report for PCT/EP2006/065710 dated Mar. 8, 2007.
International Search Report for PCT/EP2006/066107 dated Jan. 11, 2007.
International Search Report for PCT/EP2006/066347 dated Mar. 7, 2007.
International Search Report for PCT/EP2007/051411 dated May 2, 2007.
International Search Report for PCT/EP2007/054248 dated Jun. 18, 2007.
International Search Report for PCT/EP2007/062023 dated Sep. 17, 2008.
International Search Report for PCT/EP2008/060736 dated Nov. 28, 2008.
International Search Report for PCT/EP2008/060744 dated Dec. 5, 2008.
International Search Report for PCT/EP2010/051735 dated May 20, 2010.
International Search Report for PCT/EP2010/051736 dated May 7, 2010.
International Search Report for PCT/EP2011/054734 dated Aug. 12, 2011.
International Search Report for PCT/EP2011/069532 dated Dec. 15, 2011.
International Search Report for PCT/EP2012/052108 dated Mar. 8, 2012.
International Search Report for PCT/EP2012/053910 dated May 14, 2012.
International Search Report for PCT/EP2013/054524 dated May 6, 2013.
International Search Report PCT/EP2016/079465 filed on Dec. 1, 2016.
Inzucchi, Silvo E. “Oral Antihyperglycemic Therapy for Type 2 Diabetes” (2002) JAMA, vol. 287, No. 3, 360-372.
Isaji, Masayuki “Sodium-glucose cotransporter inhibitors for diabetes” Current Opinion in Investigational Drugs, (2007) vol. 8, No. 4, pp. 285-292.
Jabbour, S.A. et al. “Sodium glucose co-transporter 2 inhibitors: blocking renal tubular reabsorption of glucose to improve glycaemic control in patients with diabetes” (2008) Int J Clin Pract, 62, 8, 1279-1284.
Jagdmann Jr, G. Erik ; Synthesis of 5-(4-Substituted Benzyl)-2,4-Diaminoquinazolines as Inhibitors of Candida Albicans Dihydrofolate Reductase; Journal Heterocyclic Chemical (1995) vol. 32 pp. 1461-1465.
Johnson & Johnson “FDA Advisory Committee Recommends Approval of Canagliflozin for Treatment of Adults with Type 2 Diabetes” (2013) Press Release, 3 pgs.
Kadowaki, T et al. “PPAR gamma agonist and antagonist” Nihon Yakurigaku Zasshi (2001) vol. 118, No. 9, pp. 321-326 (English abstract).
Katsuno, Kenji et al. “Sergliflozin, a Novel Selective Inhibitor of Low-Affinity Sodium Glucose Cotransporter (SGLT2) Validates the Critical Role of SGLT2 in Renal Glucose Reabsorption and Modulates Plasma Glucose Level” The Journal of Pharmacology and Experimental Therapeutics (2007) vol. 320, No. 1, pp. 323-330.
Klepser, Teresa B. et al. “Metformin hydrochloride: An antihyperglycemic agent” (1997) Am J Health-Syst Pharm, vol. 54, 893-903.
Knochel, Paul et al. “Highly functionalized Organomagnesium Reagents Prepared through Halogen-Metal Exchange” Angew. Chem. INt. Ed. (2003) vol. 42, 4302-4320.
Koo, Ja Seo., et al; 2-Pyridyl Cyanate: A Useful Reagent for he Preparation of Nitriles; Synthetic Communications (1996) vol. 26 No. 20 pp. 3709-3713; Marcel Dekker, Inc.
Krasovskiy Arkady et al. “A LiCL-Mediated Br/Mg Exchange Reaction for the Preparation of Functionalized Aryl- and Heterarylmagnesium Compounds from Organic Bromides**” Angew. Chem. Int. Ed. (2004) vol. 43, pp. 3333-3336.
Kuribayashi, Takeshi., et al; Bis C-Glycosylated Diphenylmethanes for Stable Glycoepitope Mimetics; Syntletters (1999) vol. 6 pp. 737-740.
Kuribayashi, Takeshi., et al.; c-Glycosylated Aryl tins: Versatile Building Blocks for Aryl C-Glycoside Glycomimetics; J. Carbohydrate Chemistry (1999) vol. 18, No. 4 pp. 371-382.
Kuribayashi, Takeshi., et al; C-Glycosylated Diphenylmethanes and Benzophenones: The Stille Coupling Reaction of C-Glycosylated Aryl tins with Benzyl Bromides and Acid Chlorides; J. Carbohydrate Chemistry (1999) vol. 18, No. 4 pp. 393-401.
Langle, Sandrine., et al; Selective Double Suzuki Cross-Coupling Reactions. Synthesis of Unsymmetrical Diaryl (or Heteroaryl) Methanes; Tetrahedron Letters (2003) vol. 44 pp. 9255-9258; Pergamon Press.
Langley, Alissa K. et al. “Dipeptidyl Peptidase IV Inhibitors and the Incretin System in Type 2 Diabetes Mellitus” (2007) Pharmacotherapy, vol. 27, 1164-1180.
Larsen, Mogens Lytken et al. “Effect of Long-Term Monitoring of Glycosylated Hemoglobin Levels in Insulin-Dependent Diabetes Mellitus” (1990) The New England Journal of Medicine, vol. 323, No. 15, 1021-1025.
Lebovitz, Harold E. “Insulin secretagogues: old and new” (1999) Diabetes Review, vol. 7, 139-153.
Lehmann, Ule et al. “Palladium-Catalyzed Cross-Coupling Reactions between Dihydropyranylindium Reagents and Aryl Halides, Synthesis of C-Aryl Glycals” Organic Letters, 2003, vol. 5, No. 14, pp. 2405-2408.
Levetan, Claresa “Oral antidiabetic agents in type 2 diabetes” (2007) Current Medical Research and Opinion, vol. 23, No. 4, 945-952.
Levey, Andrew S. et al. “Definition and classification of chronic kidney disease: A position statement from Kidney Disease: Improving Global Outcomes (KDIGO)” (2005) Kidney International, vol. 67, 2089-2100.
Levien,T.L. et al, “New drugs in development for the treatment of diabetes”, Diabetes Spectrum, American Diabetes Association, US, vol. 22, No. 2, Jan. 1, 2009, pp. 92-106.
Lewin, Andrew et al “Initial Combination of Empagliflozin and Linagliptin in Subjects with Type 2 Diabetes” (2015) Diabetes Care, vol. 38, 394-402.
Li, T, et al. “Lack of Pharmacokinetic Interaction between Dapagliflozin and Pioglitazone in Healthy Subjects” Journal of Clinical Pharmacology, (2009) vol. 49, No. 9, pp. 1093.
Li, Yazhou, et al. “Glucagon-like Peptide-1 Receptor Signaling Modulates b Cell Apoptosis” (2003) The Journal of Biological Chemistry, vol. 278, No. 1, 471-478.
Derosa, Giuseppe et al. “Optimizing combination treatment in the management of type 2 diabetes” (2007) Vascular Health and Risk Management, 3(5), pp. 665-671.
Dohle, Wolfgang., et al; Copper-Mediated Cross-Coupling of Functionalized Arylmagnesium Reagents with Functionalized Alkyl and Benzylic Halides; Organic Letters (2001) vol. 3 No. 18 pp. 2871-2873.
Downes, Martin J. et al. “Triple therapy in type 2 diabetes; a systematic review and network meta-analysis” (2015) Peer J, 3:e1461, 21 pgs.
Drucker, Daniel J. et al. “Glucagon-like peptide I stimulates insulin gene expression and increases cyclic AMP levels in a rat islet cell line” (1987) Proc. Natl. Acad. Sci. USA, vol. 84, 3434-3438.
Drug Watch “Type 2 Diabetes Mellitus” Formulary vol. 43 Aug. 2008 p. 304.
DrugBank entries for Linagliptin (Accession No. DB08882), Sitagliptin (Accession No. DB01261) and Vitagliptin (Accession No. DB04876), downloaded Jan. 30, 2018, 12 pgs.
Eckhardt, M. et al., “3,5-dihydro-imidazo[4,5-d]pyridazin-4-ones: a class of potent DPP-4 inhibitors” Bioorganic & Medicinal Chemistry Letters, Pergamon, Elsevier Science, GB, vol. 18, No. 11, Jun. 1, 2008, pp. 3158-3162, XP022711188.
Eckhardt, M. et al., “8-(3-(R) Aminopiperidin-1-yl)-7-but-2-ynyl-3-methyl-1-(4 methyl-quinazolin-2-ylmethyl)-3,7-dihydropurine-2,6-dione (BI1356), a Highly Potent, Selective, Long-Acting, and Orally Bioavailable DPP-4 Inhibitor for the Treatment of Type 2 Diabetes” J. Med Chem (2007) vol. 50, pp. 6450-6453.
European Medicines Agency, Science Medicines Health, “Assessment Report Forxiga dapagliflozin” (2012) 170 pgs.
FDA Formal Comments on “Draft Guidance for Industry on Powder Blends and Finished Dosage Units—Stratified In-Process Dosage Unit Sampling and Assessment” Jan. 21, 2004, 54 pgs.
Ferrannini, Ele et al. “Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients” (2014) The Journal of Clinical Investigation vol. 124, No. 2, 499-508 and article amendment, p. 1868.
Ferrannini, Ele et al. “SGLT2 inhibition in diabetes mellitus: rationale and clinical prospects” (2012) Nat. Rev. Endocrinol. vol. 8, 495-502.
Fiese, Eugene F et al. “Preformulation” (1987) The Theory and Practice of Industrial Pharmacy, 28 pgs.
Final Office Action dated Dec. 10, 2012, U.S. Appl. No. 12/703,988, filed Feb. 11, 2010. Inventor: Peter Eickelmann.
Final Office Action dated Dec. 3, 2019, U.S. Appl. No. 15/380,272, filed Dec. 15, 2016. Inventor: Masanori Ito.
Fuerstner, Alois., et al; Practical Method for the Rhodium-Catalyzed Addition of Aryl- and Alkenylboronic Acids to Aldehydes; Advanced Synthesis and Catalysis (2001) vol. 343 No. 4 pp. 343-350.
Fujimori, Yoshikazu et al. “Remogliflozin Etabonate in a Novel Category of Selective Low-Affinity Sodium Glucose Cotransporter (SGLT2) Inhibitors, Exhibits Antidiabetic Efficacy in Rodent Models” (2008) Journal of Pharmacology and Experimental Therapeutics vol. 327 No 1, pp. 268-276.
Gallwitz, B. “Sitagliptin with Metformin: Profile of a Combination for the Treatment of Type 2 Diabetes”. Drugs of Today, Oct. 2007, 43(10), p. 681-689.
Gallwitz, Baptist “Saxagliptin, a dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes” IDrugs (2008) 11(12), pp. 906-917.
Garber, A.J. et al. “Vildagliptin in combination with pioglitazone improves glycaemic control in patients with type 2 diabetes failing thiazolidinedione monotherapy: a randomized, placebo-controlled study” (2007) Diabetes, Obesity and Metabolism, 9, 166-174.
Gennaro, Alfonso R. “Remington: The Science and Practice of Pharmacy” Twentieth Edition (2000) 4 pgs.
Gerich, John E. “Matching Treatment to Pathophysiology in Type 2 Diabetes” (2001) Clinical Therapeutics, vol. 23, No. 5, 646-659.
Ghassemi et al. “Synthesis and properties of new sulfonated poly(p-phenylene) derivatives for proton exchange membranes” Polymer (2004) pp. 5847-5854.
Ghosh, Raktim Kumar et al. “SGLT2 Inhibitors: A New Emerging Therapeutic Class in the Treatment of Type 2 Diabetes Mellitus” (2012) Journal of Clinical Pharmacology, 52, 457-463.
Golay A. et al. “Link Between Obesity and Type 2 Diabetes” (2005) Best Practice & Research Clinical Endocrinology & Metabolism, vol. 19, No. 4, 649-663.
Goldstein, Barry J. “Insulin Resistance as the Core Defect in Type 2 Diabetes Mellitus” (2002) Am J. Cardiol, 90(suppl): 3G-10G.
Goldstein, Barry J. et al. “Effect of Initial Combination Therapy with Sitagliptin, a Dipeptidyl Peptidase-4 Inhibitor and Metformin on Glycemic Control in Patients with Type 2 Diabetes” (2007) Diabetes Care, vol. 30, No. 8, 1979-1987.
Graefe-Mody et al., “The novel DPP-4 inhibitor . . . ” Diabetes, (online) 2008, XP002561421 http://professional.diabetes.org/content/posters/2008/p553-p.pdf.
Graefe-Mody, E.U., et al., “Evaluation of the potential for steady-state pharmacokinectic and pharmacodynamic interactions between the DPP-4 inhibitor linagliptin and metformin in healthy subjects”. Current Medical Research and Opinion, Informa Healthcare, GB, vol. 25. No. 8, Aug. 1, 2009, pp. 1963-1972.
Greco, Gary T. et al. “Segregation of Active Constituents from Tablet Formulations During Grinding: Theoretical Considerations” Drug Development and Industrial Pharmacy, (1982) 8(4), pp. 565-578.
Green, Brian D. et al. “Dipeptidyl peptidase IV (DPP IV) inhibitors: a newly emerging drug class for the treatment of type 2 diabetes” (2006) Diabetes and Vascular Disease Research, 159-165.
Guillory, J. Keith “Generation of Polymorphs, Hydrates, Solvates and Amorphous Solids” Polymorphism in Pharmaceutical Solids (1999) 46 pgs.
Gupta, Rajesh et al. “Emerging Drug Candidates of Dipeptidyl Peptidase IV (DPP IV) Inhibitor Class for the Treatment of Type 2 Diabetes” (2009) Current Drug Targets, vol. 10, No. 1, 71-87.
Gwaltney, S. “Medicinal Chemistry Approaches to the Inhibition of Dipeptidyl Peptidase IV”, Current Topics in Medicinal Chemistry, 2008, 8, p. 1545-1552.
Halimi, S. “Metformin: 50 years old, fit as a fiddle, and indispensable for its pivotal role in type 2 diabetes management” (2006) 32, 555-556.
Halimi, Serge, et al. “Combination treatment in the management of type 2 diabetes: focus on vildagliptin and metformin as a single tablet” (2008) Vascular Health and Risk Management, 4(3) 481-492.
Handlon, Anthony L. “Sodium glucose co-transporter 2 (SGLT2) inhibitors as potential antidiabetic agents” (2005) Expert Opinion on Therapeutic Patents, 15:11,1531-1540.
Hansch, C. “Search for New Drugs, Use of Quantitative Structure-Activity Relationships (QSAR) in Drug Design” (1980) Pomona College, Clermont, CA, Translated from Khimiko-Farmatsevticheskii Zhurnal, vol. 14, No. 10, 15-30.
Harris, Maureen I. “Classification, Diagnostic Criteria, and Screening for Diabetes” (1995) Diabetes in America, 2nd Edition, pp. 15-36.
Hatsuda, Asanori., et al; A Practical Synthesis of Highly Functionalized Aryl Nitriles Through Cyanation of Aryl Bromides Employing Heterogeneous Pd/C; Tetrahedron Letters (2005) vol. 46 pp. 1849-1853; Elsevier Ltd.
He, Y. L. et al., “Bioequivalence of Vildagliptin/Metformin Combination Tablets and Coadministration of Vildagliptin and Metformin as Free Combination in Healthy Subjects”. J. Clinical Pharmacology, 2007, vol. 47, No. 9, Abstracts of the 36th Annual Meeting of the American College of Clinical Pharmacology, San Francisco, CA, Abstract 116, p. 1210.
Heise, Tim et al. “Treatment with BI 1356, a Novel and Potent DPP-IV Inhibitor Significantly Reduces Glucose Excursions after an oGTT in Patients with Type 2 Diabetes” (2007) Diabetes, Supp 1, vol. 56, 4 pgs.
Henry Ford Health System, “Chronic Kidney Disease, Clinical Practice Recommendations for Primary Care Physicians and Healthcare Providers, A Collaborative Approach”, (Edition 6.0), 76 pgs.
Hermansen, K. et al. “Efficacy and safety of the dipeptidyl peptidase-4 inhibitor, sitagliptin, in patients with type 2 diabetes mellitus inadequately controlled on glimepiride alone or on glimepiride and metformin” (2007) Diabetes, Obesity and Metabolism, 9, 733-745.
Holst, Jens Juul et al. “Role of Incretin Hormones in the Regulaion of Insulin Secretion in Diabetic and Nondiabetic Humans” (2004) Am. J Physiol Endocrinol Metab, 287: E199-E206.
Hummel, Charles S. et al. “Glucose transport by human renal Na+/D-glucose co-transporters” (2010) Am J Physiol Cell Physiol, 34 pgs.
Hussey, Elizabeth K. et al. “Safety, Pharmacokinetics and Pharmacodynamics of Remogliflozin Etabonate (SGLT2 Inhibitor) and Metformin When Co-Administered in Type 2 Diabetes Mellitus (T2DM) Patients” Diabetes, American Diabetes Association, (2009) XP00913667, vol. 58, p. A157.
Huttner, S. et al. “Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of Single Oral Doses of BI 1356, and Inhibitor of Dipeptidyl Peptidase 4, in Healthy Male Volunteers” Journal of Clinical Pharmacology (2008) V 48, pp. 1171-1178.
Hutton, Craig A., et al; A Convenient Preparation of dityrosine via Miyaura Borylation-Suzuki Coupling of Iodotyrosine Derivatives; Tetrahedron Letters (2003) vol. 44 pp. 4895-4898; Pergamon Press.
Idris, Iskandar et al “Sodium-glucose co-transporter-2 inhibitors: an emerging new class of oral antidiabetic drug” (2009) Diabetes, Obesity and Metabolism, 11, 79-88.
US Department of Health and Human Services, FDA, Center for Drug Evaluation and Research “Application No. 204629Orig1s000 Summary Review (Jardiance)” 2014, 20 pages.
U.S. Appl. No. 15/380,272, filed Dec. 15, 2016. Non-Final Office Action dated Mar. 5, 2019. 18 pgs.
Valentine, Virginia “The Role of the Kidney and Sodium-Glucose Cotransporter-2 Inhibition in Diabetes Management” (2012) Clinical Diabetes, vol. 30, No. 4, 151-155.
Valk, Harold W. de “DPP-4 Inhibitors and Combined Treatment in Type 2 Diabetes: Re-evaluation of Clinical Success and Safety” (2007) The Review of Diabetic Studies, vol. 4, No. 3, 126-133.
Wallace, Debra J., et al; Cyclopropylboronic Acid: Synthesis and Suzuki Cross-Coupling Reactions; Tetrahedron Letters (2002) vol. 43 pp. 6987-6990; Pergamon Press.
Wang et al., “Modern diagnosis and treatment of common cardiovascular diseases”, Jul. 31, 2013, Shanxi Science and Technology Press, 1st Edition, p. 32 (English Abstract).
Wang Y et al: “BI-1356. Dipeptidyl-peptidase IV inhibitor, antidiabetic agent” Drugs of the Future, Prous Science, ES,vol. 33, No. 6, Jun. 1, 2008, pp. 473-477.
Weber, Ann E. “Dipeptidyl Peptidase IV Inhibitors for the Treatment of Diabetes” (2004) J. Med. Chem., 47, 4135-4141.
Wettergren, Andre et al. “Truncated GLP-1 (Proglucagon 78-107-Amide) Inhibits Gastric and Pancreatic Functions in Man” (1993) Digestive Diseases and Sciences, vol. 38, No. 4, 665-673.
Wielert-Badt, Susanne et al. “Probing the Conformation of the Sugar Transport Inhibitor Phlorizin by 2D-NMR, Molecular Dynamics Studies, and Pharmacophore Analysis” (2000) J. Med. Chem., vol. 43, 1692-1698.
Wielert-Badt, Susanne et al. “Single Molecule Recognition of Protein Binding Epitopes in Brush Border Membranes by Force Microscopy” (2002) Biophysical Journal, vol. 82, 2767-2774.
Williams-Herman, D. et al., “Efficacy and safety of initial combination therapy with sitagliptin and metformin in patients with type 2 diabetes: a 54-week study”. Current Medical Research and Opinion, Informa Healthcare, GB, vol. 25, No. 3, Jan. 2009, p. 569-583.
Woo, Vincent C. “Dapagliflozin: where does it fit in the treatment of type 2 diabetes” (2009) Expert Opinion on Pharmacotherapy, 10(15): 2527-2535.
www.who.int/medicinedocs/index/assoc/s14141e/s14141e.pdf “Addendum 1 to The use of stems in the selection of International Nonproprietary names (INN) for pharmaceutical substances” Worid Health Organization Jun. 19, 2007, pp. 1-3, XP007906327.
Xue, Song., et al; Zinc-mediated Synthesis of Alpha-C-Glycosided from 1,2-Anhydroglycosides; Synletters (2003) vol. 6 pp. 870-872.
Yamada, Yuichiro et al. “Clinic: Careful Progress in the Field and new Therapeutic Methods” Medical Online, (2007) vol. 220, No. 13, pp. 1219-1221.
Zhang, L. et al “Dapagliflozin treatment in patients with different stages of type 2 diabetes mellitus: effects on glycaemic control and body weight” Diabetes, Obesity and Metabolism (2010) vol. 12, No. 6, p. 510-515.
Zhang, Qiang et al. “Pharmaceuticals” Peking University Medical Press, Jan. 2005, first edition, partial English language translation, pp. 171-177.
Zimmermann, Grant R et al. “Multi-target therapeutics: when the whole is greater than the sum of the parts” (2007) Drug Discovery Today, vol. 12, 34-42.
Lieberman, Herbert A. et al. “Pharmaceutical Dosage Forms: Tablets, vol. 1” (1989) pp. 5-6.
Lipworth, Brian J. “Clinical pharmacology of b3-adrenoceptors” Br J Clin Pharmacol (1996) pp. 291-300.
Lu, Jiangqian et al. “Chapter 8, Treatment of heart failure iwth clinical conditions, Section II Treatment of heart failure complicated by arrhythmia” Feb. 28, 2015, Practical Handbook of Diagnosis and Treatment of Heart Failure, People's Military Medical Publishing House 1st Edition, p. 177 (English Abstract).
Matsuyama, Tatsuo et al. “Glucagon-like peptide-1 (7-36 amide): a potent glucagonostatic and insulinotropic hormone” Diabetes Research and Clincial Practice (1988) 5, 281-284.
Mchale, Mary “Grignard Reaction” Connexions module: m15245, (2007) pp. 1-18.
Mckinney, James D. et al. “The Practice of Structure Activity Relationships (SAR) in Toxicology” (2000) Toxicological Sciences, vol. 56, 8-17.
Mclaughlin, Mark., et al; Suzuki-Miyaura Cross-Coupling of Benzylic Phospahates with Arylboronic Acids; Organic Letters (2005) vol. 7 No. 22 pp. 4875-4878.
McMaster University, Chem2006 Lab Manual, 1997/98, Expt 1, Part B, pp. 1-9.
Meece, J. “When Oral Agents Fail: Optimizing Insulin Therapy in the Older Adult”. Consultant Pharmacist, The Society, Arlington, VA US. vol. 24, No. Suppl B, Jun. 1, 2009, p. 11-17.
Meng, Wei et al “Discovery of Dapagliflozin: A Potent, Selective Renal Sodium-Dependent Glucose Cotransporter 2 (SGLT2) Inhibitor for the Treatment of Type 2 Diabetes” J. Med. Chem. (2008) vol. 51, pp. 1145-1149.
Merck Manual of Diagnosis and Therapy, 17th Edition, (1999) Ch 13 / Disorders of Carohydrate Metabolism, Diabetes Mellitus. pp. 165-177.
Merck Manual Online Edition, “Diabetes Mellitus” http://www.merckmanuals.com/professional/endocrine_and_metabolic_disorders/diabetes_mellitus_and_disorders of carbohyrate_metabolism/diabetes_mellitus_dm.html#v987998. last revision Jun. 2008 by Preeti Kishore M.D.
Merck: “Initial Therapy with Janumet (sitagliptin/metformin) provided significantly greater blood sugar lowering compared to metformin alone in patients with type 2 diabetes”. Webwire.com, Jun. 8, 2009, p. 1-4. http://www.webwire.com/ViewPressRel.asp?ald=96695.
Merriam-Webster's Collegiate Dictionary, published 1998 by Merriam-Webster Inc. “prevent”.
Meyer, Timothy W. “Tubular injury in glomerular disease” (2003) Kidney International, vol. 63, p. 774-787.
Mojsov, Svetlana “Insulinotropin: Glucagon-like Peptide I (7-37) Co-encoded in the Glucagon Gene Is a Potent Stimulator of Insulin Release in the Perfused Rat Pancreas” J. Clin. Invest. (1987) vol. 79, 616-619.
Mooradian, Arsharg D. et al. “Narrative Review: A Rational Approach to Starting Insulin Therapy” (2006) Annals of Internal Medicine, vol. 145, pp. 125-134.
Munir, Kashif et al. “Differential pharmacology and clinical utility of empagliflozin in type 2 diabetes” (2016) Clinical Pharmacology: Advances and Applications, vol. 8, 19-34.
Nathan, D.M. et al. “Medical management of hyperglycaemia in type 2 diabetes mellitus: a consensus algorithm for the initiation and adjustment of therapy” Diabetologia (2009) 52, 17-30.
Nathan, David M. et al. “Management of Hyperglycemia in Type 2 Diabetes: A Consensus Algorithm for the Initiation and Adjustment of Therapy” (2006) Diabetes Care, vol. 29, No. 8, 1963-1972.
Nathan, David M. et al. “Medical Management of Hyperglycemia in Type 2 Diabetes: A Consensus Algorithm for the Initiation and Adjustment of Therapy” (2009) vol. 32, No. 1, pp. 193-203.
Nathan, Medical Management of hyperglycemia in type 2 diabetes mellitus, Diabetologia, vol. 52, 2009, 14 pages.
National Institute for Health Research, Horizon Scanning Centre, “Empagliflozin for type 2 diabetes mellitus” Apr. 2012, 10 pgs.
National Kidney Foundation, “Clinical Practice Guidelines, for Chronic Kidney Disease: Evaluation, Classification and Stratification” (2002) 356 pgs.
Nauck, Michael A. et al. “Cardiovascular Actions and Clincial Outcomes with Glucagon-Like Peptide-1 Receptor Agonistsand Dipeptidyl Peptidase-4 Inhibitors” Circulation (2017) vol. 136, 849-870.
Neamati, Ouri., et al;, “Depsides and Depsidones as Inhibiton of HIV-1 Integrase: Dimvery of Novel Inhibitors Through 3D Database Searclung”, J. Med. Chem., 1997, vol. 40, pp. 942-951.
Nobre, Sabrina M., et al; Synthesis of Diarylmethane Derivatives from Pd-Catalyzed Cross-Coupling Reactions of Benzylic Halides with Arylboronic Acids; Tetrahedron Letters (2004) vol. 45 8225-8228.
Office Action dated Feb. 16, 2012, U.S. Appl. No. 12/703,988, filed Feb. 11, 2010. Inventor: Peter Eickelmann.
Office Action dated Nov. 18, 2019, U.S. Appl. No. 15/778,294, filed May 23, 2018. Inventor: Georg Boeck.
Office Action dated Jun. 5, 2012. U.S. Appl. No. 12/673,319, filed Apr. 15, 2010. First named inventor: Klaus Dugi.
Office Action dated Sep. 28, 2012. U.S. Appl. No. 12/704,019, filed Feb. 11, 2010. First named inventor: Wolfram Eisenreich.
Oku, Akira., et al; T-1095, An Inhibitor or renal Na+-Glucose Cotransporters, May Provide a Novel Approach to Treating Diabetes; Diabetes (1999) vol. 48 pp. 1794-1800.
Pei, Z.: “From the bench to the bedside: Dipeptidyl peptidase IV inhibitors, a new class of oral antihyperglycemic agents” Current Opinion in Drug Discovery and Development, Current Drugs, London, GB vol. 11, No. 4, Jul. 1, 2008 pp. 512-532.
Perner, Richard J., et al; 5,6,7-Trisubstituted 4-Aminopyrido[2,3-d]pyrimidines as Novel inhibitors of Adenosime Kinase; Journal of Medicinal Chemistry (2003) vol. 46 pp. 5249-5257.
Piya, Milan K. et al. “Emerging treatment options for type 2 diabetes” British Journal of Clinical Pharmacology, (2010) vol. 70, No. 5, pp. 631-644.
Plosker, Greg L. “Dapagliflozin: A Review of Its Use in Patients with Type 2 Diabetes” (2014) Drugs, 74, 2191-2209.
Pratley, Richard E. et al. “Inhibition of DPP-4: a new therapeutic approach for the treatment of type 2 diabetes” (2007) Current Medical Research and Opinion, vol. 23, No. 4, 919-931.
Printz, RIchard L. et al. “Tweaking the Glucose Sensor: Adjusting Glucokinase Activity with Activator Compounds” Endocrinology, (2005) vol. 146, No. 9, pp. 3693-3695.
Profit, Louise et al. “Vildagliptin: the evidence for its place in the treatment of type 2 diabetes mellitus” (2008) Core Evidence, 3(1), 13-30.
Pschyrembel et al. Clinical Dictionary, 257th Edition, Diabetes Mellitus, (1993) 320-321.
Rainier, Jon D. et al. “Aluminum- and Boron-Mediated C-Glycoside Synthesis from 1,2-Anhydroglycosides” Organic Letters, (2000) vol. 2, No. 17, pp. 2707-2709.
Randzio, Stanislaw L. et al. “Metastability and Instability of Organic Crystalline Substances” J. Phys. Chem. (2008) 112, pp. 1435-1444.
Redenti, Enrico et al. “Drug/Cyclodextrin/Hydroxy Acid Multicomponent Systems. Properties and Pharmaceutical Applications” Journal of Pharmaceutical Sciences, (2000) vol. 89, No. 1, pp. 1-8.
Revesz, Lasslo., et al; SAR of Benzoylpylpyridines and Benzophenones as p38 Alpha MAP Kinase Inhibitors with Oral Activity; Bioorganic & Medicinal Chemistry Letters (2004) vol. 14 pp. 3601-3605.
Riddle, Matthew “Combining Sulfonylureas and Other Oral Agents” (2000) The American Journal of Medicine, vol. 108 (6A), 15S-22S.
Riddle, Matthew C. “Oral Pharmacologic Management of Type 2 Diabetes” (1999) American Family Physician, 60(9), 2613-2620.
Robinson, J.A. “Chemical and Biochemical Aspects of Polyether-lonophore Antibiotic Biosynthesis” (1991) Progress in the Chemistry of Organic Natural Products, 1-81.
Rosenstock, et al., “Efficacy and tolerability of initial combination therapy with vildagliptin and pioglitazone compared with component montherapy in patients with type 2 diabetes”. Diabetes, Obesity and Metabolism, Mar. 2007, vol. 9, No. 2, p. 175-185.
Rosenstock, J. et al., “Alogliptin added to insulin therapy in patients with type 2 diabetes reduces HbA1c without causing weight gain or increased hypoglycaemia”. Diabetes, Obesity and Metabolishm, Dec. 2009, vol. 11. No. 12, p. 1145-1152.
Rosenstock, Julio et al. “Dipeptidyl peptidase-4 inhibitors and the management of type 2 diabetes mellitus” (2007) Current Opinion in Endocrinology, Diabetes & Obesity, vol. 14: 98-107.
Rosenstock, Julio et al. “Dual Add-on Therapy in Type 2 Diabetes Poorly Controlled with Metformin Monotherapy: A Randomized Double-Blind Trial of Saxagliptin Plus Dapagliflozin Addition Versus Single Additon of Saxagliptin or Dapagliflozin to Metformin” (2015) Diabetes Care, vol. 38: 376-383.
Rudnic, Edward et al. “Oral Solid Dosage Forms” Remington—The Science and Practice of Pharmacy, 21th Ed, (2005) Chapter 45, Multiple Compressed Tablets, p. 890.
Rudnic, Edward et al. “Oral Solid Dosage Forms” Remington's Pharmaceutical Sciences, 18th Ed, Gennaro, A.R. Ed, Macie Pub. Co. (1990) pp. 1633-1665.
Salgado Junior, et al. “Nonalcoholic fatty liver disease and obesity” Acti Cirugica Brasiliera (2006) vol. 21, Supp. 1, pp. 72-78.
Scheen, Andre J. “Pharmacokinetic considerations for the treatment of diabetes in patients with chronic kidney disease” (2013) Expert Opinion on Drug Metabolism and Toxicology, 9:5, 529-550.
Schernthaner, G et al. “How attractive is the combination of a sodium glucose co-transporter 2 inhibitor with a dipeptidyl peptidase 4 inhibitor in the treatment of type 2 diabetes” (2015) Diabetes, Obesity and Metabolism, 17, 613-615.
Scientific Discussion: “Eucreas Scientific discussion”. Online Oct. 2007, p. 1-27, URL:http://www.emea.europa.eu/humandocs/PDFs/EPAR/eucreas/H-807-en6.pdf. see point 2. quality aspects pp. 2-4. (EMEA).
Scottish Medicines Consortium, Product Assessment “dapagliflozin 5mg and 10mg (Forxiga)” Sep. 2012, 14 pgs.
Setter, Stephen M. et al. “Metformin Hydrochloride in the Treatment of Type 2 Diabetes Mellitus: A Clincial Review with a Focus on Dual Therapy” (2003) Clincial Therapeutics, vol. 25, No. 12, 2991-3026.
Sherwin, Robert S. et al. “The Prevention or Delay ofType 2 Diabetes” Diabetes Care, (2002) vol. 25, No. 4, pp. 742-749.
Shin-Yakuzaigaku Souron. Edited by Sadasuke Okano, published by Nankodo. 1987, vol. 3, pp. 255-256.
Silverman, et al. “Handbook of Grignard Reagents” Marcel Dekker (1996) p. 82.
Singhal, Dharmendra et al. “Drug polymorphism and dosage form design: a practical perspective” Advanced Drug Delivery Reviews, 56, (2004) pp. 335-347.
Softeland, Eirik et al. “Empagliflozin as Add-on Therapy in Patients with Type 2 Diabetes Inadequately Controlled With Linagliptin and Metformin: A 24-Week Randomized, Double-Blind, Parallel-Group Trial” (2016) Diabetes Care, DOI:10.2337/dc16-1347, pp. 1-9.
Sommer, Michael Bech., et al; displacement of Halogen of 2-Halogeno-Substituted Benzonitriles with Carbonions. Preparation of (2-Cyanoaryl)arylacetonitriles; Journal of Organic Chemistry (1990) vol. 55 pp. 4817-4821.
Song, Fujian et al. “What is indirect comparsion?” (2009) Hayward Medical Communications, www.whatisseries.co.uk, 6 pgs.
Stazi, Federica., et al; Statistical Experimental Design-Driven Discovery of room-Temperature Conditions for Palladium-Catalyzed Cyanation of Aryl Bromides; Tetrahedron Letters (2005) vol. 46 1815-1818; Elsevier Ltd.
Stern, Judith et al. “Insulin Resistance and Pancreatic Insulin Release in Genetically Obese Zucker Rat” (1972) P.S.E.B.M., vol. 139, 66-69.
Strack, Thomas “Metformin: A Review” (2008) Drugs of Today, 44(4), 303-314.
Takebayashi, Kohzo et al. “Effect of Sodium Glucose Cotransporter 2 Inhibitors With Low SGLT2/SGLT1 Selectivity on Circulating Glucagon-Like Peptide 1 Levels in Type 2 Diabetes Mellitus” (2017) J Clin Med Res., vol. 9, (9) 745-753.
Tanaka, Chikako “Therapeutic Drugs for Metabolic Diseases, Chapter 2” (2002) New Yakurigaku (New Pharmacology) pp. 524-527.
Taskinen, M.-R. et al., “Safety and efficacy of linagliptin as add-on therapy to metformin in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled study.” Diabetes, Obesity and Metabolism, 2011, vol. 13, pp. 65-74.
The Merck Manual Diagnosis and Therapy; Seventeenth Edition; “13 / Disorders of Carbohyrate Metabolism” Merck Research Laboratories (1999) pp. 165-177.
Thomas, L., “Chronic treatment with the Dipeptidyl Peptidase-4 Inhibitor BI 1356[9R)-8-(3-Amino-piperidin-1-yl)-7-but-2-ynyl-3-methyl-1(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione] Increases Basal Glucagon-Like Peptide-1 and Improves Glycemic Control in Diabetic Rodent Models” The Journal of Pharmacology and Experimental Therapeutics, Feb. 2009, vol. 328, No. 2, pp. 556-563.
Thomas, Leo “Long-term treatment with empagliflozin, a novel, potent and selective SGLT-2 inhibitor, improves glycaemic control and features of metabolic syndrome in diabetic rats” (2012) Diabetes, Obesity and Metabolism, vol. 14, No. 1, 94-96.
Thomas, Leo et al. “(R)-8-(3-Amino-piperidin-1-yl)-7-but-2-ynyl-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydro-purine-2,6-dione (BI 1356), a Novel Xanthine-Based Dipeptidyl Peptidase 4 Inhibitor, Has a Superior Potency and Longer Duration of Action Compared with Other Dipeptidyl Peptidase-4 Inhibitors” Journal of Pharmacology and Experimental Therapeutics (2008) 325, pp. 175-182.
Threlfall, Terry “Structural and Thermodynamic Explanations of Ostwald's Rule” Organic Process Research & Development (2003) vol. 7, pp. 1017-1027.
Tinahones, Francisco J. et al. “Linagliptin as add-on to empagliflozin and metformin in patients with type 2 diabetes: Two 24-week randonmized double-blind, double-dummy parallel-group trials” (2017) Diabetes Obes Metab, 19(2): 266-274.
Torrance, Christopher J. et al. “Combinatorial chemoprevention of intestinal neoplasia” (2000) Nature Medicine, vol. 6, No. 8, 1024-1028.
Turner, Robert C. et al. “Glycemic Control with Diet, Sulfonylurea, Metformin or Insulin in Patients with Type 2 Diabetes Mellitus, Progressive Requirement for Multiple Therapies (UKPDS-49)” (1999) American Medical Association, vol. 281, No. 21, 8 pgs.
Tykwinski, Rik R; Evolution in the Palladium-Catalyzed Cross-Coupling of sp- and sp2-Hybridized Carbon Atoms; Angew Chemical International Edition (2003) vol. 42 pp. 1566-1568.
U.S. Appl. No. 13/287,216, filed Nov. 2, 2011. Inventor: Rolf Grempler.
U.S. Appl. No. 13/367,739, filed Feb. 7, 2012. Inventor: Thomas Klein.
U.S. Appl. No. 13/413,702, filed Mar. 7, 2012. Inventor: Masanori Ito.
U.S. Appl. No. 13/484,506, filed May 31, 2012. Inventor: Marion Wienrich.
U.S. Appl. No. 13/539,713, filed Jul. 2, 2012. Inventor: Uli Broedl.
U.S. Appl. No. 13/634,886, filed Sep. 14, 2012. Inventor: Peter Eickelmann.
U.S. Appl. No. 13/637,413, filed Sep. 26, 2012. Inventor: Rolf Grempler.
U.S. Appl. No. 13/693,239, filed Dec. 4, 2012. Inventor: Klaus Dugi.
U.S. Appl. No. 13/695,492, filed Oct. 31, 2012. Inventor: Thomas Klein.
U.S. Appl. No. 13/785,365, filed Mar. 5, 2013. Inventor: Masanori Ito.
U.S. Appl. No. 13/833,097, filed Mar. 15, 2013. Inventor: Eric Williams Mayoux.
U.S. Appl. No. 14/244,196, filed Apr. 3, 2014. Inventor: Uli Christian Broedl.
U.S. Appl. No. 14/244,208, filed Apr. 3, 2014. Inventor: Uli Christian Broedl.
U.S. Appl. No. 14/253,935, filed Apr. 16, 2014. Inventor: Uli Christian Broedl.
Ueta, Kiichiro., et al; Long-Term Treatment with the Na+-Glucose Cotransporter Inhibitor T-1095 Causes Sustained Improvement in Hyperglycemia and Prevents Diabetic Neuropathy in Goto-Kakizaki Rats; Life Sciences (2005) vol. 76 pp. 2655-2668.
US Department of Health and Human Services, CDER, FDA, “Guideline for Submitting Supporting Documentation in Drug Applications for the Manufacture of Drug Substances” Feb. 1987, 48 pages.
US Department of Health and Human Services, FDA, Endocrinologic and Metabolic Drugs Advisory Committee; Notice of Meeting, [Docket No. FDA-2011-N-002], Federal Register, vol. 76, No. 80, Apr. 26, 2011, 23324-23325.
US Department of Health and Human Services, FDA, “Guidance for Industry, Diabetes Mellitus—Evaluating Cardiovascular Risk in New Antidiabetic Therapies to Treat Type 2 Diabetes” Dec. 2008, 8 pages.
US Department of Health and Human Services, FDA, “Guidance for Industry, Pharmacokinetics in Patients with Impaired Renal Function—Study Design, Data Analysis, and Impact on Dosing and Labeling” May 1998, 19 pages.
Related Publications (1)
Number Date Country
20210267902 A1 Sep 2021 US
Continuations (4)
Number Date Country
Parent 16732635 Jan 2020 US
Child 17123366 US
Parent 15906414 Feb 2018 US
Child 16732635 US
Parent 15140865 Apr 2016 US
Child 15906414 US
Parent 13413702 Mar 2012 US
Child 15140865 US