US20200046713A1 - Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin - Google Patents

Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin Download PDF

Info

Publication number
US20200046713A1
US20200046713A1 US16/528,934 US201916528934A US2020046713A1 US 20200046713 A1 US20200046713 A1 US 20200046713A1 US 201916528934 A US201916528934 A US 201916528934A US 2020046713 A1 US2020046713 A1 US 2020046713A1
Authority
US
United States
Prior art keywords
dpp
patients
inhibitor
patient
combination
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/528,934
Inventor
Klaus Dugi
Eva Ulrike Graefe-Mody
Michael Mark
Hans-Juergen Woerle
Heike Zimdahl-Gelling
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boehringer Ingelheim International GmbH
Original Assignee
Boehringer Ingelheim International GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=43466576&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20200046713(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Boehringer Ingelheim International GmbH filed Critical Boehringer Ingelheim International GmbH
Priority to US16/528,934 priority Critical patent/US20200046713A1/en
Publication of US20200046713A1 publication Critical patent/US20200046713A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/155Amidines (), e.g. guanidine (H2N—C(=NH)—NH2), isourea (N=C(OH)—NH2), isothiourea (—N=C(SH)—NH2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/64Sulfonylureas, e.g. glibenclamide, tolbutamide, chlorpropamide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/26Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the invention describes DPP-4 inhibitors, pharmaceutical compositions or combinations comprising a DPP-4 inhibitor as defined herein and optionally one or more other active substances, for use in methods of treatment or prevention as described herein, such as e.g. of one or more conditions selected from type 1 diabetes mellitus, type 2 diabetes mellitus, impaired glucose tolerance, impaired fasting blood glucose and hyperglycemia inter alia.
  • the therapeutic and/or preventive methods of this invention comprise the step of identifying a patient being susceptible to the treatment and/or prevention, said identifying comprising testing whether the patient has variation(s) in one or more genes associated with metabolic diseases (e.g.
  • a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament each as described herein for a therapeutic and/or preventive method or use according this invention in a patient who has variation(s) in one or more genes associated with metabolic diseases is contemplated.
  • TCF7L2 risk genotype patients include, without being limited, patients (particularly type 2 diabetes patients) harboring genetic risk variants in the gene TCF7L2 and suffering often from the pathological influences thereof, particularly associated with the risk T-allele of TCF7L2 rs7903146, such as patients harboring the TCF7L2 rs7903146 CT heterozygous risk genotype or patients harboring the TCF7L2 rs7903146 TT homozygous high risk genotype.
  • the usability of a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament each as described herein for a therapeutic and/or preventive method or use according this invention in a patient who carries the TCF7L2 wild genotype, particularly the TCF7L2 rs7903146 CC wild genotype, is contemplated.
  • the present invention provides a diagnostic method for identifying a subject (particularly a type 2 diabetes patient) statistically more likely to have a favorable response (e.g. in achieving glycemic control, such as change in HbA1c) to the administration of a therapeutically effective amount of a DPP-4 inhibitor, optionally in combination with one or more other active substances (e.g.
  • said method comprising determining whether the subject is either of TCF7L2 risk genotype (particularly TCF7L2 rs7903146 CT or TT risk genotype) or of TCF7L2 wild genotype (particularly TCF7L2 rs7903146 CC wild genotype), wherein the subject being of TCF7L2 rs7903146 CC homozygous wild genotype (and, to a lesser extent, the subject being of TCF7L2 rs7903146 CT heterozygous risk genotype) has an increased likelihood of favorable response to the administered DPP-4 inhibitor relative to a subject of TCF7L2 rs7903146 TT homozygous risk genotype.
  • DPP-4 inhibitor as defined hereinafter (preferably linagliptin), optionally in combination with one or more other active substances.
  • the present invention describes the use of a DPP-4 inhibitor for the manufacture of a medicament for use in a method as described hereinbefore and hereinafter.
  • the present invention describes a DPP-4 inhibitor for use in a therapy of a patient (particularly human type 2 diabetes patient) as described hereinbefore and hereinafter.
  • the present invention describes a DPP-4 inhibitor for use in a treatment or prevention of a (particularly metabolic) disease, disorder or condition (particularly diabetes, especially type 2 diabetes, and conditions related thereto, such as e.g. diabetic complications) as described hereinbefore and hereinafter.
  • the invention also describes a use of a pharmaceutical composition or combination according to this invention for the manufacture of a medicament for use in a method as described hereinbefore and hereinafter.
  • the invention also relates to the DPP-4 inhibitors as defined herein for use in a method as described hereinbefore and hereinafter, said method comprising administering the DPP-4 inhibitor, optionally in combination with one or more other active substances (e.g. which may selected from those mentioned herein), to the patient.
  • DPP-4 inhibitors as defined herein for use in a method as described hereinbefore and hereinafter, said method comprising administering the DPP-4 inhibitor, optionally in combination with one or more other active substances (e.g. which may selected from those mentioned herein), to the patient.
  • Type 2 diabetes is an increasingly prevalent disease that due to a high frequency of complications leads to a significant reduction of life expectancy. Because of diabetes-associated microvascular complications, type 2 diabetes is currently the most frequent cause of adult-onset loss of vision, renal failure, and amputations in the industrialized world. In addition, the presence of type 2 diabetes is associated with a two to five fold increase in cardiovascular disease risk.
  • Oral and non-oral antidiabetic drugs conventionally used in therapy include, without being restricted thereto, metformin, sulphonylureas, thiazolidinediones, glinides, a-glucosidase inhibitors, GLP-1 or GLP-1 analogues, and insulin or insulin analogues.
  • the high incidence of therapeutic failure is a major contributor to the high rate of long-term hyperglycemia-associated complications or chronic damages (including micro- and makrovascular complications such as e.g. diabetic nephrophathy, retinopathy or neuropathy, or cardiovascular complications) in patients with type 2 diabetes.
  • chronic damages including micro- and makrovascular complications such as e.g. diabetic nephrophathy, retinopathy or neuropathy, or cardiovascular complications
  • TCF7L2 transcription factor 7-like 2
  • SNPs single nucleotid polymorphisms
  • rs12255372 and, particularly, rs7903146 are strongly associated with diabetes.
  • the risk of developing type 2 diabetes is increased by roughly 45% (Odds ratio 1.45) among carriers of one risk T-allele of TCF7L2 rs7903146 (CT heterozygotes), and is at least doubled (Odds ratio of 2.41) among TT homozygotes compared to CC homozygotes wild genotypes (Grant et al, Nature Genetics, Vol. 38, 2006, p 320-323).
  • TCF7L2 risk genotypes are associated with increased TCF7L2 expression in pancreatic beta cells, impaired (glucose-stimulated) insulin secretion, incretin effects and enhanced rate of hepatic glucose production as well as predisposition to and prediction of future type 2 diabetes (cf. Lyssenko et al., The Journal of Clinical Investigation, Vol. 117, No 8, 2007, p. 2155-2163).
  • TCF7L2 rs7903146 risk variants are associated with lower incretin effect on insulin secretion, which may be based, at least in parts, on an impaired sensitivity of the beta cells to incretins.
  • diabetes patients harboring TCF7L2 risk variants particularly carriers of the at risk T-allele of TCF7L2 rs7903146, such as patients harboring the TCF7L2 rs7903146 CT genotype or, particularly, patients harboring the TCF7L2 rs7903146 TT genotype, are expected to be difficult to treat in antidiabetic therapy.
  • DPP-4 inhibitors represent another novel class of agents that are being developed for the treatment or improvement in glycemic control in patients with type 2 diabetes.
  • DPP-4 inhibitors and their uses are disclosed in WO 2002/068420, WO 2004/018467, WO 2004/018468, WO 2004/018469, WO 2004/041820, WO 2004/046148, WO 2005/051950, WO 2005/082906, WO 2005/063750, WO 2005/085246, WO 2006/027204, WO 2006/029769, W02007/014886; WO 2004/050658, WO 2004/111051, WO 2005/058901, WO 2005/097798; WO 2006/068163, WO 2007/071738, WO 2008/017670; WO 2007/128721, WO 2007/128724, WO 2007/128761, or WO 2009/121945.
  • the aim of the present invention is to provide a medication and/or method for preventing, slowing progression of, delaying or treating a metabolic disorder, in particular of type 2 diabetes mellitus.
  • a further aim of the present invention is to provide a medication and/or method for improving glycemic control in a patient in need thereof, in particular in patients with type 2 diabetes mellitus, for example in those patients who have variation(s) in one or more genes associated with metabolic diseases (such as e.g. a TCF7L2 risk genotype patient as described herein) or in those patients who are of respective wild-type genotype.
  • metabolic diseases such as e.g. a TCF7L2 risk genotype patient as described herein
  • Another aim of the present invention is to provide a medication and/or method for improving glycemic control in a patient with insufficient glycemic control despite monotherapy with an antidiabetic drug, for example metformin, or despite combination therapy with two or three antidiabetic drugs, for example in such a patient who has variation(s) in one or more genes associated with metabolic diseases (such as e.g. a TCF7L2 risk genotype patient as described herein) or in such a patient who is of respective wild-type genotype.
  • an antidiabetic drug for example metformin
  • combination therapy with two or three antidiabetic drugs for example in such a patient who has variation(s) in one or more genes associated with metabolic diseases (such as e.g. a TCF7L2 risk genotype patient as described herein) or in such a patient who is of respective wild-type genotype.
  • Another aim of the present invention is to provide a medication and/or method for preventing, slowing or delaying progression from impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), insulin resistance and/or metabolic syndrome to type 2 diabetes mellitus.
  • ITT impaired glucose tolerance
  • IGF impaired fasting blood glucose
  • Yet another aim of the present invention is to provide a medication and/or method for preventing, slowing progression of, delaying or treating of a condition or disorder from the group consisting of complications of diabetes mellitus.
  • a further aim of the present invention is to provide a medication and/or method for reducing the weight or preventing an increase of the weight in a patient in need thereof, for example in such a patient who has variation(s) in one or more genes associated with metabolic diseases (such as e.g. a TCF7L2 risk genotype patient as described herein) or in such a patient who is of respective wild-type genotype.
  • metabolic diseases such as e.g. a TCF7L2 risk genotype patient as described herein
  • Another aim of the present invention is to provide a medication with a high efficacy for the treatment of metabolic disorders, in particular of diabetes mellitus, impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), and/or hyperglycemia, which has good to very good pharmacological and/or pharmacokinetic and/or physicochemical properties.
  • metabolic disorders in particular of diabetes mellitus, impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), and/or hyperglycemia, which has good to very good pharmacological and/or pharmacokinetic and/or physicochemical properties.
  • a DPP-4 inhibitor preferably linagliptin, as well as a pharmaceutical composition or combination comprising the DPP-4 inhibitor and optionally one or more other active substances (e.g. antidiabetics), is therapeutically effective for improving glycemic control and treating type 2 diabetes mellitus in TCF7L2 rs7903146 CT or TT risk genotype patients and in TCF7L2 rs7903146 CC wild genotype patients.
  • active substances e.g. antidiabetics
  • TCF7L2 genotype patients have a clinically meaningful response to the administered DPP-4 inhibitor, preferably linagliptin.
  • certain subgroups of diabetes patients amenable to antidiabetic therapy according to this invention include for example, without being limited to, those patients harboring TCF7L2 rs7903146 CC or CT or TT genotype, respectively.
  • DPP-4 inhibitors as defined herein as well as pharmaceutical compositions or combinations comprising a DPP-4 inhibitor as defined herein and optionally one or more other active substances can be used in a method of preventing, slowing progression of, delaying (e.g. delaying the onset of) or treating a metabolic disorder (particularly diabetes, especially type 2 diabetes mellitus and conditions related thereto, e.g. diabetic complications), in particular a method for improving glycemic control in a patient, such as in a patient who has variation(s) in one or more genes associated with metabolic diseases (such as e.g. in TCF7L2 risk genotype patients as described herein).
  • DPP-4 inhibitors as defined herein as well as pharmaceutical compositions or combinations comprising a DPP-4 inhibitor as defined herein and optionally one or more other active substances can be used in a method of preventing, slowing progression of, delaying (e.g. delaying the onset of) or treating a metabolic disorder (particularly diabetes, especially type 2 diabetes mellitus and conditions related thereto), in particular a method for improving glycemic control in a patient, such as in a patient who is of TCF7L2 wild genotype, particularly of TCF7L2 rs7903146 CC wild genotype.
  • the method comprises the step of of identifying a patient being susceptible to the method being used, e.g. comprising testing whether the patient has variation(s) in one or more genes associated with metabolic diseases (e.g. whether the patient is of a TCF7L2 risk genotype as described herein) or whether the patient is of TCF7L2 wild genotype as described herein, and the step of administering such a DPP-4 inhibitor, pharmaceutical composition or combination to the patient determined as being susceptible.
  • metabolic diseases e.g. whether the patient is of a TCF7L2 risk genotype as described herein
  • TCF7L2 wild genotype as described herein
  • the present invention provides a method for determining of a probability of the likelihood of a favorable response (e.g. in providing glycemic control) or the magnitude of a favorable change in HbA1c of an individual resulting from treating the individual with a DPP-4 inhibitor, preferably linagliptin, or the DPP-4 inhibitor in combination with one or more other active substances (e.g.
  • said method comprising determining whether the subject is either of TCF7L2 risk genotype (particularly TCF7L2 rs7903146 TT risk genotype) or of TCF7L2 wild genotype (particularly TCF7L2 rs7903146 CC wild genotype), wherein the probability of likelihood of a favorable response or the significantly high magnitude of a favorable change in HbA1c response to administration of the DPP-4 inhibitor, preferably linagliptin, or the DPP-4 inhibitor in combination with one or more other active substances (e.g. antidiabetics) is
  • TCF7L2 rs7903146 CC homozygous wild genotype, and lower in an individual of TCF7L2 rs7903146 TT homozygous risk genotype (e.g. but still clinically significant or meaningful).
  • composition or combination comprising
  • a second antidiabetic agent selected from the group G3 consisting of biguanides (particularly metformin), thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues and insulin or insulin analogues, and, optionally,
  • a third antidiabetic agent being different from (b) selected from the group G3 consisting of biguanides (particularly metformin), thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues and insulin or insulin analogues,
  • a second antidiabetic agent selected from the group G3 consisting of biguanides (particularly metformin), thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues and insulin or insulin analogues, and, optionally,
  • a third antidiabetic agent being different from (b) selected from the group consisting of metformin, a sulfonylurea, pioglitazone, rosiglitazone, repaglinide, nateglinide, acarbose, voglibose, miglitol, GLP-1 or a GLP-1 analogue and insulin or an insulin analogue,
  • a second antidiabetic agent selected from the group consisting of metformin, a sulfonylurea, pioglitazone, rosiglitazone, repaglinide, nateglinide, acarbose, voglibose, miglitol, GLP-1 or a GLP-1 analogue and insulin or an insulin analogue, and, optionally,
  • a third antidiabetic agent being different from (b) selected from the group G3 consisting of biguanides (particularly metformin), thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues and insulin or insulin analogues,
  • composition or combination comprising
  • a second antidiabetic agent selected from the group consisting of metformin, a sulfonylurea and pioglitazone, and, optionally,
  • a third antidiabetic agent being different from (b) selected from the group consisting of metformin, a sulfonylurea, pioglitazone, rosiglitazone, repaglinide, nateglinide, acarbose, voglibose, miglitol, GLP-1 or GLP-1 analogue and insulin or insulin analogue,
  • composition or combination comprising
  • a second antidiabetic agent selected from the group consisting of metformin, a sulfonylurea, pioglitazone, rosiglitazone, repaglinide, nateglinide, acarbose, voglibose, miglitol, GLP-1 or GLP-1 analogue and insulin or insulin analogue, and, optionally,
  • a third antidiabetic agent being different from (b) selected from the group consisting of metformin, a sulfonylurea and pioglitazone,
  • a second antidiabetic agent selected from the group consisting of metformin and pioglitazone, and, optionally,
  • a third antidiabetic agent being different from (b) selected from the group consisting of metformin, a sulfonylurea and pioglitazone,
  • a second antidiabetic agent selected from the group consisting of metformin, a sulfonylurea and pioglitazone, and, optionally,
  • a third antidiabetic agent being different from (b) selected from the group consisting of metformin and pioglitazone,
  • said third antidiabetic agent is preferably chosen from another class than the second antidiabetic agent.
  • the second and the third antidiabetic agent are different, and preferably they are from different classes (e.g. when the second antidiabetic agent is chosen from the biguanide class, the third antidiabetic agent is preferably chosen from another class).
  • Classes of antidiabetic agents are mentioned above, e.g. biguanide class, thiazolidindione class, sulfonylurea class, glinide class, alpha-glucosidase inhibitor class, GLP-1 analogue class, insulin class, etc.
  • a particular embodiment of this invention refers to monotherapy with a DPP-4 inhibitor as defined herein and/or to pharmaceutical compositions comprising a DPP-4 inhibitor as sole active ingredient.
  • a particular embodiment refers to dual combinations and/or dual therapy; another embodiment refers to triple combinations and/or triple therapy.
  • a method for preventing, slowing the progression of, delaying or treating a metabolic disorder selected from the group consisting of type 1 diabetes mellitus, type 2 diabetes mellitus, impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), hyperglycemia, postprandial hyperglycemia, overweight, obesity and metabolic syndrome in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • a metabolic disorder selected from the group consisting of insulin resistance, hyperlipidemia, hypercholesterolemia, dyslipidemia, hypertension, chronic systemic inflammation, retinopathy, neuropathy, nephropathy, atherosclerosis, endothelial dysfunction, non-alcoholic fatty liver disease (NAFLD) and osteoporosis
  • 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • the pharmaceutical composition of 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.
  • ITT impaired glucose tolerance
  • IGF impaired fasting blood glucose
  • 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • ITT impaired glucose tolerance
  • IGF impaired fasting blood glucose
  • 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.
  • a condition or disorder selected from the group consisting of complications of diabetes mellitus such as cataracts
  • tissue ischaemia particularly comprises diabetic macroangiopathy, diabetic microangiopathy, impaired wound healing and diabetic ulcer.
  • micro- and macrovascular diseases and “micro- and macrovascular complications” are used interchangeably in this application.
  • 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • a beta-cell degeneration and a decline of beta-cell functionality such as for example apoptosis or necrosis of pancreatic beta cells can be delayed or prevented.
  • the functionality of pancreatic cells can be improved or restored, and the number and size of pancreatic beta cells increased. It may be shown that the differentiation status and hyperplasia of pancreatic beta-cells disturbed by hyperglycemia can be normalized by treatment with a pharmaceutical composition or combination of this invention.
  • a method for preventing, slowing, delaying or treating the degeneration of pancreatic beta cells and/or the decline of the functionality of pancreatic beta cells and/or for improving and/or restoring the functionality of pancreatic beta cells and/or restoring the functionality of pancreatic insulin secretion in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • an abnormal accumulation of (ectopic) fat, in particular in the liver may be reduced or inhibited.
  • a method for preventing, slowing, delaying or treating diseases or conditions attributed to an abnormal accumulation of liver or ectopic fat in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • liver or ectopic 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, particularly non-alcoholic fatty liver disease (NAFLD), including hepatic steatosis, non-alcoholic steatohepatitis (NASH) and/or liver fibrosis.
  • NAFL non-alcoholic fatty liver
  • NASH non-alcoholic steatohepatitis
  • NASH non-alcoholic steatohepatitis
  • liver fibrosis 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, particularly non-alcoholic fatty liver disease (NAFLD), including hepatic ste
  • a method for preventing, slowing the progression, delaying, attenuating, treating or reversing hepatic steatosis, (hepatic) inflammation and/or an abnormal accumulation of liver fat in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • NODAT new onset diabetes after transplantation
  • PTMS post-transplant metabolic syndrome
  • 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • a patient in need thereof comprising administering the DPP-4 inhibitor alone or, optionally, in combination with a second and, optionally, with a third antidiabetic agent as defined hereinbefore and hereinafter to the patient.
  • a patient in need thereof comprising administering the second antidiabetic agent in combination with a DPP-4 inhibitor and, optionally, with a third antidiabetic agent as defined hereinbefore and hereinafter to the patient.
  • TCF7L2 risk genotype patients within the meaning of this invention refer to those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially a SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146; in more particular, those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype; especially those who carry two T alleles of SNP rs7903146 of TCF7L2, i.e. the TT genotype, are at high-risk and are expected to be difficult to treat (e.g. to achieve adequate glycemic control).
  • SNPs single nucleotide polymorphisms
  • the present invention provides a DPP-4 inhibitor (preferably linagliptin), pharmaceutical composition, combination or medicament according to the present invention for use in a therapeutic and/or preventive method as described hereinbefore and hereinafter (e.g. treating type 2 diabetes) in one or more of the following patient groups:
  • DPP-4 inhibitor preferably linagliptin
  • pharmaceutical composition, combination or medicament according to the present invention for use in a therapeutic and/or preventive method as described hereinbefore and hereinafter (e.g. treating type 2 diabetes) in one or more of the following patient groups:
  • the invention relates to a DPP-4 inhibitor, a pharmaceutical composition or combination of the present invention for a therapeutic and/or preventive method or use as described hereinbefore and hereinafter (e.g. treating type 2 diabetes), said method or use comprising
  • the invention relates to a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament of the present invention for a therapeutic and/or preventive method or use as described hereinbefore and hereinafter (e.g. treating type 2 diabetes) in TCF7L2 risk genotype patients, e.g. in those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially a SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146; in more particular, in those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype.
  • SNPs single nucleotide polymorphisms
  • the invention relates to a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament of the present invention for a therapeutic and/or preventive method or use as described hereinbefore and hereinafter (e.g. treating type 2 diabetes) in TCF7L2 wild genotype patients, e.g. in those patients who carry two C alleles of SNP rs7903146 of TCF7L2, i.e. the CC genotype.
  • a particular sub-population of the patients described hereinbefore and hereinafter refers to those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially at least one SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146, in more particular, those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype.
  • SNPs single nucleotide polymorphisms
  • those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype, especially who carry two T alleles of SNP rs7903146 of TCF7L2, i.e. the TT genotype, are strongly susceptible to increased TCF7L2 expression in pancreatic beta cells, impaired insulin secretion, incretine effects, enhanced rate of hepatic glucose production and/or diabetes.
  • the T allele of rs7903146 TCF7L2 is associated with impaired insulinotropic action of incretin hormones, reduced 24 h profiles of plasma insulin and glucagon, and increased hepatic glucose production.
  • Another particular sub-population of the patients described hereinbefore and hereinafter refers to those patients who are of TCF7L2 wild genotype, particularly those who are of the TCF7L2 rs7903146 CC wild genotype.
  • a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament according to the present invention for a therapeutic and/or preventive method or use as described hereinbefore and hereinafter (particularly for treating and/or preventing type 2 diabetes and/or obesity), in patients with reduced (glucose-stimulated) insulin secretion, increased hepatic gluconeogenesis and/or reduced insulinotropic effect or action of incretin hormones (e.g. GLP-1 and/or GIP), e.g. impaired incretin sensitivity, associated with a TCF7L2 risk genotype, particularly with such a TCF7L2 risk genotype as mentioned above.
  • incretin hormones e.g. GLP-1 and/or GIP
  • impaired incretin sensitivity associated with a TCF7L2 risk genotype, particularly with such a TCF7L2 risk genotype as mentioned above.
  • a method of determining patient's treatment response to a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament according to the present invention comprising the step of determining whether the patient is of TCF7L2 risk genotype as described herein, e.g. testing whether the patient belongs to the particular subpopulation of TCF7L2 risk genotype carriers, or determining whether the patient is of TCF7L2 wild genotype, e.g. testing whether the patient carries the wild-type CC allele at rs7903146 in TCF7L2.
  • a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament according to the present invention for use in a therapeutic and/or preventive method as described hereinbefore and hereinafter (particularly for treating and/or preventing type 2 diabetes and/or obesity) in a patient in need thereof, said method comprising testing whether the patient is of any TCF7L2 risk genotype as described herein.
  • a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament according to the present invention for use in a therapeutic and/or preventive method as described hereinbefore and hereinafter (particularly for treating and/or preventing type 2 diabetes and/or obesity) in a patient in need thereof, said method comprising testing whether the patient is of TCF7L2 wild genotype as described herein.
  • the testing for TCF7L2 risk genotypes may be used for patient stratification, e.g. to enrich patient population in clinical trials to test the efficacy of the DPP-4 inhibitor.
  • the method of determining the treatment susceptibility of an individual may be used for determination whether the patient may respond to a lower level or may require a higher level of administered DPP-4 inhibitor, optionally in combination with one or more other active substances.
  • determining the treatment susceptibility of an individual comprising the testing for TCF7L2 risk or wild genotypes as described herein may be used for determination whether the patient may be treated in monotherapy or in combination therapy with one or more additional antidiabetics according to this invention, e.g. to provide adequate glycemic control. For example, those patients with decreased likelihood of favorable response may require combination treatment, e.g. to achieve adequate glycemic control.
  • FIG. 1 shows mean values and 95% confidence intervals for baseline HbA1c values for the whole patient population of the studies (full analysis set, FAS), for the subpopulation for which genetic analyses are performed (full analysis set for pharmacogenetic analyses, FASG), as well as for the subgroups defined by genotype (CC, CT, TT) of this subpopulation.
  • the numbers of patients for placebo control and linagliptin treatment are given in braces.
  • FIG. 2 shows a statistical association between TCF7L2 SNP rs7903146 genotypes with a likelihood of a favorable response in CC/CT genotype carriers to the administration of a therapeutically-effective amount of linagliptin or linagliptin in combination with other oral antidiabetic therapy.
  • active ingredient of a pharmaceutical composition or combination of the present invention means the DPP-4 inhibitor and/or, if present, the second antidiabetic agent and/or, if present, the third antidiabetic agent of the present invention.
  • 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/m 2 .
  • weight is defined as the condition wherein the individual has a BMI greater than or 25 kg/m 2 and less than 30 kg/m 2 .
  • 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/m 2 .
  • 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/m 2 but lower than 35 kg/m 2 ; the term “class II obesity” is the condition wherein the BMI is equal to or greater than 35 kg/m 2 but lower than 40 kg/m 2 ; the term “class III obesity” is the condition wherein the BMI is equal to or greater than 40 kg/m 2 .
  • 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.
  • 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).
  • 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 110 mg/dL (6.11 mmol/L) or 100 mg mg/dL (5.6 mmol/L).
  • fasting has the usual meaning as a medical term.
  • hypoglycemia is defined as the condition in which a subject has a fasting blood glucose concentration above the normal range, greater than 110 mg/dL (6.11 mmol/L) or 100 mg mg/dL (5.6 mmol/L).
  • fasting has the usual meaning as a medical term.
  • hypoglycemia is defined as the condition in which a subject has a blood glucose concentration below the normal range of 60 to 115 mg/dL (3.3 to 6.3 mmol/L), in particular below 70 mg/dL (3.89 mmol/L).
  • 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 (11.11 mmol/L).
  • IGF paired fasting blood glucose
  • a subject with “normal fasting glucose” has a fasting glucose concentration smaller than 100 mg/dl, i.e. smaller than 5.6 mmol/l.
  • ITT paired glucose tolerance
  • 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 (7.78 mmol/L).
  • 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).
  • Insulin-sensitizing As insulin-sensitizing, “insulin resistance-improving” or “insulin resistance-lowering” are synonymous and used interchangeably.
  • 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).
  • 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.
  • HOMA-IR homeostasis model assessment to insulin resistance
  • HOMA-IR score is calculated with the formula (Galvin P, et al. Diabet Med 1992;9:921-8):
  • HOMA-IR [fasting serum insulin ( ⁇ U/mL)] ⁇ [fasting plasma glucose(mmol/L)/22.5]
  • 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. 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 needs 2-3 times as much insulin as a healthy person, without this resulting in any clinical symptoms.
  • pancreatic beta-cells 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.
  • 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).
  • 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.
  • 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.
  • 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.
  • late stage type 2 diabetes mellitus includes type 2 diabetes patients with a secondary antidiabetic drug failure, indication for insulin therapy and progression to micro- and macrovascular complications e.g. diabetic nephropathy, or coronary heart disease (CHD).
  • CHD coronary heart disease
  • 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.
  • metformin on its own achieves an average improvement in the HbA1c value in the diabetic of the order of 1.0 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.
  • 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).
  • diagnosis of the metabolic syndrome is made when three or more of the following risk factors are present:
  • 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.
  • 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.
  • SBP systolic blood pressure
  • DBP diastolic blood pressure
  • NODAT new onset diabetes after transplantation
  • PTMS post-transplant metabolic syndrome
  • IDF International Diabetes Federation
  • PTMS post-transplant 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.
  • hyperuricemia denotes a condition of high serum total urate levels.
  • uric acid concentrations between 3.6 mg/dL (ca. 214 ⁇ mol/L) and 8.3 mg/dL (ca. 494 ⁇ mol/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.
  • uric acid 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.
  • DPP-4 inhibitor in the scope of the present invention relates to a compound that exhibits inhibitory activity on the enzyme dipeptidyl peptidase IV (DPP-4). Such inhibitory activity can be characterised by the IC50 value.
  • a DPP-4 inhibitor preferably exhibits an IC50 value below 10000 nM, preferably below 1000 nM.
  • Certain DPP-4 inhibitors exhibit an IC50 value below 100 nM, or even ⁇ 50 nM.
  • IC50 values of DPP-4 inhibitors are usually above 0.01 nM, or even above 0.1 nM.
  • DPP-IV inhibitors may include biologic and non-biologic, in particular non-peptidic compounds.
  • DPP-4 inhibitor also comprises any pharmaceutically acceptable salts thereof, hydrates and solvates thereof, including the respective crystalline forms.
  • treatment and “treating” or ananlogous terms comprise particularly 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.
  • 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.
  • prophylactically treating “preventive treating” and “preventing” or ananlogous terms are used interchangeably and comprise a treatment of patients at risk to develop a condition mentioned hereinbefore, thus reducing said risk.
  • the aspects of the present invention in particular the pharmaceutical compounds, compositions, combinations, methods and uses, refer to DPP-4 inhibitors, second and/or third antidiabetic agents as defined hereinbefore and hereinafter.
  • a DPP-4 inhibitor in the context of the present invention is any DPP-4 inhibitor of
  • R1 denotes ([1,5]naphthyridin-2-yl)methyl, (quinazolin-2-yl)methyl, (quinoxalin-6-yl)methyl, (4-methyl-quinazolin-2-yl)methyl, 2-cyano-benzyl, (3-cyano-quinolin-2-yl)methyl, (3-cyano-pyridin-2-yl)methyl, (4-methyl-pyrimidin-2-yl)methyl, or (4,6-dimethyl-pyrimidin-2-yl)methyl and R2 denotes 3-(R)-amino-piperidin-1-yl, (2-amino-2-methyl-propyl)-methylamino or (2-(S)-amino-propyl)-methylamino,
  • a DPP-4 inhibitor in the context of the present invention is a DPP-4 inhibitor selected from the group consisting of
  • sitagliptin sitagliptin, vildagliptin, saxagliptin, alogliptin, gemigliptin,
  • preferred DPP-4 inhibitors are any or all of the following compounds and their pharmaceutically acceptable salts:
  • a more preferred DPP-4 inhibitor among the abovementioned DPP-4 inhibitors of embodiment A of this invention is 1-[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-(3-(R)-amino-piperidin-1-yl)-xanthine, particularly the free base thereof (which is also known as linagliptin or BI 1356).
  • DPP-4 inhibitors As further DPP-4 inhibitors the following compounds can be mentioned:
  • sitagliptin is in the form of its dihydrogenphosphate salt, i.e. sitagliptin phosphate.
  • sitagliptin phosphate is in the form of a crystalline anhydrate or monohydrate.
  • a class of this embodiment refers to sitagliptin phosphate monohydrate.
  • Sitagliptin free base and pharmaceutically acceptable salts thereof are disclosed in U.S. Pat. No. 6,699,871 and in Example 7 of WO 03/004498. Crystalline sitagliptin phosphate monohydrate is disclosed in WO 2005/003135 and in WO 2007/050485.
  • a tablet formulation for sitagliptin is commercially available under the trade name Januvia®.
  • a tablet formulation for sitagliptin/metformin combination is commercially available under the trade name Janumet®.
  • Vildagliptin is specifically disclosed in U.S. Pat. No. 6,166,063 and in Example 1 of WO 00/34241. Specific salts of vildagliptin are disclosed in WO 2007/019255. A crystalline form of vildagliptin as well as a vildagliptin tablet formulation are disclosed in WO 2006/078593.
  • Vildagliptin can be formulated as described in WO 00/34241 or in WO 2005/067976.
  • a modified release vildagliptin formulation is described in WO 2006/135723.
  • a tablet formulation for vildagliptin is commercially available under the trade name Galvus®.
  • a tablet formulation for vildagliptin/metformin combination is commercially available under the trade name Eucreas®.
  • Saxagliptin is specifically disclosed in U.S. Pat. No. 6,395,767 and in Example 60 of WO 01/68603.
  • saxagliptin is in the form of its HCl salt or its mono-benzoate salt as disclosed in WO 2004/052850.
  • saxagliptin is in the form of the free base.
  • saxagliptin is in the form of the monohydrate of the free base as disclosed in WO 2004/052850.
  • Crystalline forms of the HCl salt and of the free base of saxagliptin are disclosed in WO 2008/131149.
  • a process for preparing saxagliptin is also disclosed in WO 2005/106011 and WO 2005/115982. Saxagliptin can be formulated in a tablet as described in WO 2005/117841.
  • Alogliptin is specifically disclosed in US 2005/261271, EP 1586571 and in WO 2005/095381.
  • alogliptin is in the form of its benzoate salt, its hydrochloride salt or its tosylate salt each as disclosed in WO 2007/035629.
  • a class of this embodiment refers to alogliptin benzoate.
  • Polymorphs of alogliptin benzoate are disclosed in WO 2007/035372.
  • a process for preparing alogliptin is disclosed in WO 2007/112368 and, specifically, in WO 2007/035629.
  • Alogliptin (namely its benzoate salt) can be formulated in a tablet and administered as described in WO 2007/033266.
  • a solid preparation of alogliptin/pioglitazone and its preparation and use is described in WO 2008/093882.
  • a solid preparation of alogliptin/metformin and its preparation and use is described in WO 2009
  • the mesylate salt of the former compound as well as crystalline polymorphs thereof are disclosed in WO 2006/100181.
  • the fumarate salt of the latter compound as well as crystalline polymorphs thereof are disclosed in WO 2007/071576.
  • These compounds can be formulated in a pharmaceutical composition as described in WO 2007/017423.
  • This compound and methods for its preparation are disclosed in WO 2005/000848.
  • a process for preparing this compound is also disclosed in WO 2008/031749, WO 2008/031750 and WO 2008/055814.
  • This compound can be formulated in a pharmaceutical composition as described in WO 2007/017423.
  • the DPP-4 inhibitor is selected from the group G2 consisting of linagliptin, sitagliptin, vildagliptin, alogliptin, saxagliptin, carmegliptin, gosogliptin, teneligliptin, melogliptin and dutogliptin, or a pharmaceutically acceptable salt of one of the hereinmentioned DPP-4 inhibitors, or a prodrug thereof.
  • the DPP-4 inhibitor is selected from the group G2 consisting of linagliptin, sitagliptin, vildagliptin, alogliptin, saxagliptin, teneligliptin and dutogliptin, or a pharmaceutically acceptable salt of one of the hereinmentioned DPP-4 inhibitors, or a prodrug thereof.
  • a particularly preferred DPP-4 inhibitor within the present invention is linagliptin.
  • linagliptin refers to linagliptin and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. Crystalline forms are described in WO 2007/128721. Methods for the manufacture of linagliptin are described in the patent applications WO 2004/018468 and WO 2006/048427 for example.
  • Linagliptin is distinguished from structurally comparable DPP-4 inhibitors, as it combines exceptional potency and a long-lasting effect with favourable pharmacological properties, receptor selectivity and a favourable side-effect profile or bring about unexpected therapeutic advantages or improvements in monotherapy and/or when used in combination with a second and, optionally, a third antidiabetic agent according to this invention.
  • compositions, methods and uses according to this invention relate to those compositions which comprise the DPP-4 inhibitor as sole active ingredient (i.e. the second and third antidiabetic agent are both absent) and/or, respectively, to monotherapy using the DPP-4 inhibitor alone.
  • compositions, combinations, methods and uses according to this invention relate to those compositions or combinations which comprise the DPP-4 inhibitor and the second antidiabetic agent as sole active ingredients (i.e. the third antidiabetic agent is absent) and/or, respectively, to dual combination therapy using the DPP-4 inhibitor and the second antidiabetic agent.
  • compositions, combinations, methods and uses according to this invention relate to those compositions or combinations which comprise the DPP-4 inhibitor, the second and the third antidiabetic agent and/or, respectively, to triple combination therapy using the DPP-4 inhibitor, the second and the third antidiabetic agent.
  • a DPP-4 inhibitor according to this invention may be further characterized in that said DPP-4 inhibitor does not significantly impair glomerular and/or tubular function of a type 2 diabetes patient with chronic renal insufficiency (e.g. mild, moderate or severe renal impairment or end stage renal disease), and/or
  • said DPP-4 inhibitor does not require to be dose-adjusted in a type 2 diabetes patient with impaired renal function (e.g. mild, moderate or severe renal impairment or end stage renal disease).
  • impaired renal function e.g. mild, moderate or severe renal impairment or end stage renal disease.
  • the second antidiabetic agent and, if present, the third antidiabetic agent is selected from the group G3 consisting of biguanides, thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues, and insulin or insulin analogues, or a pharmaceutically acceptable salt thereof.
  • G3 consisting of biguanides, thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues, and insulin or insulin analogues, or a pharmaceutically acceptable salt thereof.
  • the group G3 comprises biguanides.
  • biguanides are metformin, phenformin and buformin.
  • a preferred biguanide is metformin.
  • a DPP-4 inhibitor in combination with a biguanide, in particular metformin, can provide more efficacious glycemic control and/or may act together with the biguanide, for example to reduce weight, that has e.g. overall beneficial effects on the metabolic syndrome which is commonly associated with type 2 diabetes mellitus.
  • metformin refers to metformin or a pharmaceutically acceptable salt thereof such as the hydrochloride salt, the metformin (2:1) fumarate salt, and the metformin (2:1) succinate salt, the hydrobromide salt, the p-chlorophenoxy acetate or the embonate, and other known metformin salts of mono and dibasic carboxylic acids. It is preferred that the metformin employed herein is the metformin hydrochloride salt.
  • the group G3 comprises thiazolidindiones.
  • thiazolidindiones are pioglitazone and rosiglitazone.
  • TZD therapy is associated with weight gain and fat redistribution.
  • TZD cause fluid retention and are not indicated in patients with congestive heart failure.
  • Long term treatment with TZD are further associated with an increased risk of bone fractures.
  • a DPP-4 inhibitor in combination with a thiazolidindione, in particular pioglitazone can provide more efficacious glycemic control and/or can minimize side effects of the treatment with TZD.
  • pioglitazone refers to pioglitazone, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salt thereof such as the hydrochloride salt.
  • rosiglitazone refers to rosiglitazone, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salt thereof such as the maleate salt.
  • the group G3 comprises sulfonylureas.
  • sulfonylureas are glibenclamide, tolbutamide, glimepiride, glipizide, gliquidone, glibornuride, glyburide, glisoxepide and gliclazide.
  • Preferred sulfonylureas are tolbutamide, gliquidone, glibenclamide and glimepiride, in particular glibenclamide and glimepiride.
  • a combination of a DPP-4 inhibitor with a sulfonylurea may offer additional benefit to the patient in terms of better glycemic control.
  • treatment with sulfonylureas is normally associated with gradual weight gain over the course of treatment and a DPP-4 inhibitor may minimize this side effect of the treatment with an sulfonylurea and/or improve the metabolic syndrome.
  • a DPP-4 inhibitor in combination with a sulfonylurea may minimize hypoglycemia which is another undesirable side effect of sulfonylureas. This combination may also allow a reduction in the dose of sulfonylureas, which may also translate into less hypoglycemia.
  • glibenclamide refers to the respective active drug or a pharmaceutically acceptable salt thereof.
  • the group G3 comprises glinides.
  • glinides are nateglinide, repaglinide and mitiglinide.
  • a combination of a DPP-4 inhibitor with a meglitinide may offer additional benefit to the patient in terms of better glycemic control.
  • treatment with meglitinides is normally associated with gradual weight gain over the course of treatment and a DPP-4 inhibitor may minimize this side effect of the treatment with an meglitinide and/or improve the metabolic syndrome.
  • a DPP-4 inhibitor in combination with a meglitinide may minimize hypoglycemia which is another undesirable side effect of meglitinides. This combination may also allow a reduction in the dose of meglitinides, which may also translate into less hypoglycemia.
  • nateglinide refers to nateglinide, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salts and esters thereof.
  • repaglinide refers to repaglinide, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salts and esters thereof.
  • the group G3 comprises inhibitors of alpha-glucosidase.
  • inhibitors of alpha-glucosidase are acarbose, voglibose and miglitol. Additional benefits from the combination of a DPP-4 inhibitor and an alpha-glucosidase inhibitor may relate to more efficacious glycemic control, e.g. at lower doses of the individual drugs, and/or reducement of undesirable gastrointestinal side effects of alpha-glucosidase inhibitors.
  • acarbose refers to the respective active drug or a pharmaceutically acceptable salt thereof.
  • the group G3 comprises inhibitors of GLP-1 analogues.
  • GLP-1 analogues are exenatide, liraglutide, taspoglutide, semaglutide, albiglutide, and lixisenatide.
  • the combination of a DPP-4 inhibitor and a GLP-1 analogue may achieve a superior glycemic control, e.g. at lower doses of the individual drugs.
  • the body weight reducing capability of the GLP-1 analogue may be positively act together with the properties of the DPP-4 inhibitor.
  • a reduction of side effects e.g. nausea, gastrointestinal side effects like vomiting
  • a reduced dose of the GLP-1 analogue is applied in the combination with a DPP-4 inhibitor.
  • compositions, combinations, methods and uses according to this invention relate to those combinations wherein the DPP-4 inhibitor and the second antidiabetic agent are preferably selected according to the entries in the Table 1.
  • the pharmaceutical compositions, combinations, methods and uses according to this invention relate to those combinations wherein the DPP-4 inhibitor is linagliptin.
  • the second antidiabetic agent is preferably selected according to the entries in the Table 2.
  • the combination of a DPP-4 inhibitor and a second and, optionally, a third antidiabetic agent according to this invention can be found to improve the glycemic control, in particular in patients as described herein, compared with a monotherapy using either a DPP-4 inhibitor or the second or third antidiabetic agent alone, for example with a monotherapy of metformin, or with a dual therapy using the second and third antidiabetic agent.
  • the triple combination of a DPP-4 inhibitor and a second and a third antidiabetic agent according to this invention can be found to improve the glycemic control, in particular in patients as described herein, compared with a combination therapy using a DPP-4 inhibitor and either the second or third antidiabetic agent, or using the second and the third antidiabetic agent.
  • the improved glycemic control is determined as an increased lowering of blood glucose and an increased reduction of HbA1c.
  • the glycemic control may not be further improved significantly by an administration of the drug above a certain highest dose.
  • a long term treatment using a highest dose may be unwanted in view of potential side effects.
  • a satisfying glycemic control may not be achievable in all patients via a monotherapy using either the DPP-4 inhibitor or the second or the third antidiabetic agent alone.
  • monotherapy do not yield in full glycemic control
  • dual therapy may become necessary.
  • triple therapy may become necessary.
  • a progression of the diabetes mellitus may continue and complications associated with diabetes mellitus may occur, such as macrovascular complications.
  • the pharmaceutical composition or combination 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, e.g. in the case of dual or triple combination therapy compared with a monotherapy using one of or, respectively, a dual therapy using two of the combination partners.
  • the combination of a DPP-4 inhibitor and the second and, optionally, the third therapeutic agent according to this invention can be found to allow a reduction in the dose of either the DPP-4 inhibitor or the second or third antidiabetic agent or even of two or three of the active ingredients.
  • a dose reduction is beneficial for patients which otherwise would potentially suffer from side effects in a therapy using a higher dose of one or more of the active ingredients, in particular with regard to side effect caused by the second and/or third antidiabetic agent. Therefore, the pharmaceutical combination as well as the methods according to the present invention, may show less side effects, thereby making the therapy more tolerable and improving the patients compliance with the treatment.
  • a DPP-4 inhibitor according to the present invention is able—via the increases in active GLP-1 levels—to reduce the glucagon secretion in a patient. This will therefore limit the hepatic glucose production. Furthermore, the elevated active GLP-1 levels produced by the DPP-4 inhibitor will have beneficial effects on beta-cell regeneration and neogenesis. All these features of DPP-4 inhibitors may render a pharmaceutical composition or combination or method of this invention quite useful and therapeutically relevant.
  • this invention refers to patients requiring treatment or prevention, it relates primarily to treatment and prevention in humans, but the pharmaceutical composition may also be used accordingly in veterinary medicine in mammals.
  • adult patients are preferably humans of the age of 18 years or older.
  • patients are adolescent humans, i.e. humans of age 10 to less than 18 years, preferably of age 13 to less than 18 years.
  • patients in need of treatment or prevention as described herein can be identified by determining whether they have variation(s) (e.g. polymorphisms) in one or more genes associated with metabolic diseases and/or whether they have variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, in particular whether they are of TCF7L2 risk genotype as described herein.
  • variation(s) e.g. polymorphisms
  • genes associated with metabolic diseases e.g. polymorphisms
  • variation(s) e.g. polymorphisms
  • patients in need of treatment or prevention as described herein can be identified by determining whether they are of respective wild-type genotype, in particular whether they are of TCF7L2 wild genotype as described herein.
  • a particular sub-population of the patients in need of treatment or prevention as described herein refers to those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially a SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146, in more particular, those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype.
  • SNPs single nucleotide polymorphisms
  • Another particular sub-population of the patients in need of treatment or prevention as described herein refers to those patients who carry TCF7L2 rs7903146 CC wild genotype.
  • a treatment or prophylaxis according to this invention is suitable in those patients in need of such treatment or prophylaxis who are diagnosed of having variation(s) (e.g. polymorphisms) in one or more genes associated with metabolic diseases and/or variation(s) (e.g. SNPs) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, in particular of TCF7L2 risk genotype as described herein.
  • variation(s) e.g. polymorphisms
  • genes associated with metabolic diseases and/or variation(s) e.g. SNPs
  • a treatment or prophylaxis according to this invention is particular suitable in those patients in need of such treatment or prophylaxis who are diagnosed of having TCF7L2 wild genotype as described herein.
  • a treatment or prophylaxis according to this invention is suitable in those patients in need of such treatment or prophylaxis who are diagnosed of having one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, e.g. at least one SNP selected from rs7903146, rs12255372 and rs10885406, for example rs7903146, in particular, carrying at least one T allele of rs7903146, (i.e. of CT or TT genotype), among them, in more particular, those carrying one T allele of rs7903146 (i.e. of CT risk genotype) or, in less particular, those carrying two T alleles of rs7903146 (i.e. of TT high risk genotype).
  • SNPs single nucleotide polymorphisms
  • a treatment or prophylaxis according to this invention is particular favorable in those patients in need of such treatment or prophylaxis who are diagnosed of carrying wild-type two C alleles of rs7903146 in TCF7L2 (i.e. of CC genotype).
  • a treatment or prophylaxis according to this invention is 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.
  • a treatment or prophylaxis according to this invention is advantageously suitable in those patients in which a weight increase is contraindicated. Any weight increasing effect in the therapy, for example due to the administration of the second and/or third antidiabetic agent, may be attenuated or even avoided thereby.
  • the pharmaceutical composition or combination of this invention 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).
  • HbA1c fasting plasma glucose, postprandial plasma glucose and/or glycosylated hemoglobin
  • the present invention also discloses the use of the pharmaceutical composition or combination for improving glycemic control in patients having type 2 diabetes or showing first signs of pre-diabetes.
  • the invention also includes diabetes prevention. If therefore a pharmaceutical composition or combination of 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.
  • the pharmaceutical composition or combination of 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.
  • patients include patients with diabetes type 2 and patients with diabetes type 1.
  • ITT impaired glucose tolerance
  • IGF impaired fasting blood glucose
  • a method for improving gycemic control in patients, in particular in adult patients, with type 2 diabetes mellitus as an adjunct to diet and exercise is provided.
  • a therapeutic or preventive method and/or use according to this invention is suitable in those patients who have variation(s) (e.g. polymorphisms) in one or more genes associated with metabolic diseases and/or who have variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R.
  • variation(s) e.g. polymorphisms
  • genes associated with metabolic diseases e.g. polymorphisms
  • a sub-population of the patients described hereinbefore and hereinafter refers to TCF7L2 risk genotype patients, such as e.g. to those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially at least one SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146.
  • SNPs single nucleotide polymorphisms
  • the TT genotype are strongly susceptible to increased TCF7L2 expression in pancreatic beta cells, impaired insulin secretion, incretine effects, enhanced rate of hepatic glucose production and/or diabetes.
  • the T allele of rs7903146 TCF7L2 is associated with impaired insulinotropic action of incretin hormones, reduced 24 h profiles of plasma insulin and glucagon, and increased hepatic glucose production.
  • the present invention also includes the compounds, pharmaceutical compositions or combinations according to this invention for use in the treatment and/or prevention of those diseases, disorders or conditions mentioned herein in those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially at least one SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146; in more particular, in those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype, particularly in those patients who carry one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype, or who carry two T alleles of SNP rs7903146 of TCF7L2, i.e. the TT genotype.
  • SNPs single nucleotide polymorphisms
  • TCF7L2 risk genotype patients as described herein include, without being limited, patients of Caucasian, North European, East Asian, Indian and/or African descent.
  • the present invention further includes a therapeutic and/or preventive method or use according to this invention for application in a patient in need thereof, said method or use comprising the step of determining whether the patient has variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, particularly whether the patient is of a TCF7L2 risk genotype as described herein.
  • variation(s) e.g. polymorphisms
  • the determination or diagnosis whether the patient has variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, particularly whether the patient is of a TCF7L2 risk genotype as described herein, or whether the patient is of wild genotype, particularly whether the patient is of TCF7L2 wild genotype as described herein, may be used for determining the likelihood (e.g., increased, decreased, or no likelihood) of a favourable therapeutic and/or preventive response of the patient to the treatment with a DPP-4 inhibitor (or with a combination of a DPP-4 inhibitor with the second and/or third antidiabetic agent as defined herein) in a therapeutic and/or preventive method or use as described hereinabove or hereinbelow (e.g. in treating diabetes or in improving glycemic control), and thus for identifying a subject being susceptible to such treatment.
  • variation(s) e.g. polymorphisms
  • polymorphisms in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, particularly whether the subject is of a TCF7L2 risk genotype as described herein, or determining whether the subject is of TCF7L2 wild genotype, particularly testing whether the subject is of the TCF7L2 rs7903146 CC wild genotype.
  • the present invention provides a DPP-4 inhibitor, a pharmaceutical composition or combination according to the present invention for use in a therapeutic or preventive method as described hereinbefore or hereinafter (particularly for treating or preventing type 2 diabetes and/or obesity), said method comprising
  • identifying a subject being susceptible to the therapeutic or preventive method comprising testing whether the subject has variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, in particular whether the subject is of any TCF7L2 risk genotype as described herein, in more particular whether he/she has one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially at least one SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146, for example whether the subject carries at least one T allele of SNP rs7903146 of TCF7L2, e.g.
  • SNPs single nucleotide polymorphisms
  • the present invention further provides a therapeutic and/or preventive method or use of this invention for application in a patient in need thereof, said method or use comprising the steps of
  • an improvement of the glycemic control can be achieved even in those patients who have insufficient glycemic control in particular despite treatment with the second or third antidiabetic agent or a combination of the second with the third antidiabetic agent, for example despite maximal tolerated dose of oral monotherapy with metformin, a thiazolidinedione (e.g. pioglitazone) or a sulfonylurea, or a combination of metformin with a thiazolidinedione (e.g. pioglitazone), of metformin with a sulfonylurea, or of a thiazolidinedione (e.g. pioglitazone) with a sulfonylurea.
  • metformin e.g. pioglitazone
  • a thiazolidinedione e.g. pioglitazone
  • metformin with a thiazolidinedione e
  • an improvement of the glycemic control can be achieved even in those patients who have insufficient glycemic control in particular despite treatment with a DPP-4 inhibitor or a combination of a DPP-4 inhibitor with the second or third antidiabetic agent, for example despite maximal tolerated dose of oral monotherapy with a DPP-4 inhibitor or a dual combination of a DPP-4 inhibitor with the second or third antidiabetic agent.
  • a maximal tolerated dose with regard to metformin is for example 2000 mg per day, 1500 mg per day (for example in asian countries) or 850 mg three times a day or any equivalent thereof.
  • the method and/or use according to this invention is applicable in those patients who show one, two or more of the following conditions:
  • the dual or triple combination method and/or use according to this invention is further applicable in those patients who show the following conditions (e) or (f), respectively:
  • a pharmaceutical composition or combination is suitable in the treatment of patients who are diagnosed having one or more of the following conditions
  • compositions or combination according to this invention is particularly suitable in the treatment of patients who are diagnosed having one or more of the following conditions
  • ITT impaired glucose tolerance
  • IGF impaired fasting blood glucose
  • 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.
  • compositions and the methods according to this invention are particularly suitable in the treatment of patients after organ transplantation, in particular those patients who are diagnosed having one or more of the following conditions
  • a pharmaceutical composition or combination according to this invention in particular due to the DPP-4 inhibitor therein, exhibits a good safety profile. Therefore, a treatment or prophylaxis according to this invention is possible in those patients for which the mono-therapy with another antidiabetic drug, such as for example metformin, is contraindicated and/or who have an intolerance against such drugs at therapeutic doses.
  • 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.
  • a pharmaceutical composition or combination 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 or combination 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.
  • long term 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.
  • a particular 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.
  • 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.
  • DPP-4 inhibitor and the second and, optionally, third antidiabetic agent are administered together, for example simultaneously in one single or two or three separate formulations, and/or when they are administered in alternation, for example successively in two or three separate formulations.
  • the amount of the pharmaceutical composition according to this invention to be administered to the patient and required for use in treatment or prophylaxis according to the present invention will vary with the route of administration, the nature and severity of the condition for which treatment or prophylaxis is required, the age, weight and condition of the patient, concomitant medication and will be ultimately at the discretion of the attendant physician.
  • the DPP-4 inhibitor and, optionally, the second and/or third antidiabetic agent according to this invention are included in the pharmaceutical composition, combination or dosage form in an amount sufficient that by their administration the glycemic control in the patient to be treated is improved.
  • the amount of the DPP-4 inhibitor, the second and/or third antidiabetic agent to be employed in the pharmaceutical composition and the methods and uses according to this invention are described. These ranges refer to the amounts to be administered per day with respect to an adult patient, in particular to a human being, for example of approximately 70 kg body weight, and can be adapted accordingly with regard to an administration 2, 3, 4 or more times daily and with regard to other routes of administration and with regard to the age of the patient. The ranges of the dosage and amounts are calculated for the inidividual active moiety.
  • the combination therapy of the present invention utilizes lower dosages of the individual DPP-4 inhibitor and/or of the individual second and/or third antidiabetic agent used in monotherapy or used in conventional therapeutics, thus avoiding possible toxicity and adverse side effects incurred when those agents are used as monotherapies.
  • the pharmaceutical composition or combination is preferably administered orally.
  • Other forms of administration are possible and described hereinafter.
  • the one or more dosage forms comprising the DPP-4 inhibitor and/or the second and/or the third antidiabetic agent is oral or usually well known.
  • the amount of the DPP-4 inhibitor in the combinations, combination methods or combined uses of this invention is preferably in the range from 1/5 to 1/1 of the amount usually recommended for a monotherapy using said DPP-4 inhibitor.
  • a preferred dosage range of linagliptin when administered orally is 0.5 mg to 10 mg per day, preferably 2.5 mg to 10 mg, most preferably 1 mg to 5 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 0.5 to 10 mg, in particular 1 to 5 mg. Examples of particular dosage strengths are are 1, 2.5, 5 or 10 mg.
  • the application of the active ingredient may occur up to three times a day, preferably one or two times a day.
  • Suitable formulations for linagliptin may be those formulations disclosed in the application WO 2007/128724, the disclosure of which is incorporated herein in its entirety.
  • Typical dosage strengths of the dual fixed dose combination (tablet) of linagliptin/metformin IR (immediate release) are 2.5/500 mg, 2.5/850 mg and 2.5/1000 mg, which may be administered 1-3 times a day, particularly twice a day.
  • Typical dosage strengths of the dual fixed dose combination (tablet) of linagliptin/metformin XR (extended release) are 5/500 mg, 5/1000 mg and 5/1500 mg, which may be administered 1-2 times a day, particularly once a day, preferably to be taken in the evening with meal, or 2.5/500 mg, 2.5/750 mg and 2.5/1000 mg, which may be administered 1-2 times a day, particularly once a day two tablets, preferably to be taken in the evening with meal.
  • a preferred dosage range of sitagliptin when administered orally is from 10 to 200 mg, in particular 25 to 150 mg per day.
  • a recommended dose of sitagliptin is 100 mg calculated for the active moiety (free base anhydrate) once daily or 50 mg twice daily.
  • the preferred range of amounts in the pharmaceutical composition is 10 to 150 mg, in particular 25 to 100 mg. Examples are 25, 50, 75 or 100 mg.
  • the application of the active ingredient may occur up to three times a day, preferably one or two times a day. Equivalent amounts of salts of sitagliptin, in particular of the phosphate monohydrate can be calculated accordingly. Adjusted dosages of sitagliptin, for example 25 and 50 mg, are preferably used for patients with renal failure.
  • a preferred dosage range of vildagliptin when administered orally is from 10 to 150 mg daily, in particular from 25 to 150 mg, 25 and 100 mg or 25 and 50 mg or 50 and 100 mg daily.
  • the daily administration of vildagliptin is 50 or 100 mg.
  • the preferred range of amounts in the pharmaceutical composition is 10 to 150 mg, in particular 25 to 100 mg. Examples are 25, 50, 75 or 100 mg.
  • the application of the active ingredient may occur up to three times a day, preferably one or two times a day.
  • a preferred dosage range of alogliptin when administered orally is from 5 to 250 mg daily, in particular from 10 to 150 mg daily.
  • the preferred range of amounts in the pharmaceutical composition is 5 to 150 mg, in particular 10 to 100 mg. Examples are 10, 12.5, 20, 25, 50, 75 and 100 mg.
  • the application of the active ingredient may occur up to three times a day, preferably one or two times a day.
  • a preferred dosage range of saxagliptin when administered orally is from 2.5 to 100 mg daily, in particular from 2.5 to 50 mg daily.
  • the preferred range of amounts in the pharmaceutical composition is from 2.5 to 100 mg, in particular from 2.5 and 50 mg. Examples are 2.5, 5, 10, 15, 20, 30, 40, 50 and 100 mg.
  • the application of the active ingredient may occur up to three times a day, preferably one or two times a day.
  • a preferred dosage range of dutogliptin when administered orally is from 50 to 400 mg daily, in particular from 100 to 400 mg daily.
  • the preferred range of amounts in the pharmaceutical composition is from 50 to 400 mg. Examples are 50, 100, 200, 300 and 400 mg.
  • the application of the active ingredient may occur up to three times a day, preferably one or two times a day.
  • DPP-4 inhibitors of this invention refers to those orally administered DPP-4 inhibitors which are therapeutically efficacious at low dose levels, e.g. at dose levels ⁇ 100 mg or ⁇ 70 mg per patient per day, preferably ⁇ 50 mg, more preferably ⁇ 30 mg or ⁇ 20 mg, even more preferably from 1 mg to 10 mg (if required, divided into 1 to 4 single doses, particularly 1 or 2 single doses, which may be of the same size), particularly from 1 mg to 5 mg (more particularly 5 mg), per patient per day, preferentially, administered orally once-daily, more preferentially, at any time of day, administered with or without food.
  • the daily oral amount 5 mg BI 1356 can be given in a once daily dosing regimen (i.e. 5 mg BI 1356 once daily) or in a twice daily dosing regimen (i.e. 2.5 mg BI 1356 twice daily), at any time of day, with or without food.
  • the amount of the second and/or third antidiabetic agent in the combinations, combination methods and/or combined uses of this invention is preferably in the range from 1/5 to 1/1 of the amount usually recommended for a monotherapy using said antidiabetic agent.
  • Using lower dosages of the individual second and/or third antidiabetic agent compared with monotherapy could avoid or minimize possible toxicity and adverse side effects incurred when those agents are used as monotherapies.
  • a preferred dosage range of metformin when administered orally is 250 to 3000 mg, in particular 500 to 2000 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 250 to 1000, in particular 500 to 1000 mg or 250 to 850 mg respectively. Examples are 500, 750, 850 or 1000 mg.
  • the administration of said amounts is once, twice or three times daily.
  • the amounts of 500, 750 and 850 mg preferably require once-daily, twice-daily or three-times daily dosing and the amount of 1000 mg preferably requires once-daily or twice-daily dosing.
  • Certain controlled or sustained release formulations allow a once-daily dosing.
  • Metformin can be administered for example in the form as marketed under the trademarks GLUCOPHAGETM, GLUCOPHAGE-DTM or GLUCOPHAGE-XRTM.
  • a preferred dosage range of pioglitazone when administered orally is 5 to 50 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 5 to 50 mg, 10 to 45 mg and 15 to 45 mg respectively. Examples are 15, 30 or 45 mg.
  • Preferably the administration of said amounts is once or twice daily, in particular once daily.
  • Pioglitazone can be administered in the form as it is marketed for example under the trademark ACTOSTM.
  • a preferred dosage range of rosiglitazone when administered orally is 1 mg to 10 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 1 to 10 mg, 2 to 8 mg, 4 to 8 mg and 1 to 4 mg. Examples are 1, 2, 4 or 8 mg.
  • Preferably the administration of said amounts is once or twice daily.
  • Preferably the dose should not exceed 8 mg daily.
  • Rosiglitazone can be administered in the form as it is marketed for example under the trademark AVANDIATM.
  • a preferred dosage range of a thiazolidindione (other than pioglitazone or rosiglitazone as described above) when administered orally is 2 to 100 mg per day.
  • the preferred range of amounts in the pharmaceutical composition for an administration once, twice or three times daily is 2 to 100, 1 to 50 and 1 to 33 mg respectively.
  • a preferred dosage range of glibenclamide when administered orally is 0.5 to 15 mg, in particular 1 to 10 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 0.5 to 5 mg, in particular 1 to 4 mg. Examples are 1.0, 1.75 and 3.5 mg.
  • Preferably the administration of said amounts is once, twice or three-times daily.
  • Glibenclamide can be administered in the form as it is marketed for example under the trademark EUGLUCONTM.
  • a preferred dosage range of glimepiride when administered orally is 0.5 to 10 mg, in particular 1 to 6 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 0.5 to 10 mg, in particular 1 to 6 mg. Examples are 1, 2, 3, 4, and 6 mg.
  • Preferably the administration of said amounts is once, twice or three-times daily, preferably once daily.
  • Glimepiride can be administered in the form as it is marketed for example under the trademark AMARYLTM.
  • a preferred dosage range of gliquidone when administered orally is 5 to 150 mg, in particular 15 to 120 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 5 to 120 mg, in particular 5 to 30 mg. Examples are 10, 20, 30 mg.
  • Preferably the administration of said amounts is once, twice, three-times or four-times daily.
  • Gliquidone can be administered in the form as it is marketed for example under the trademark GLURENORMTM.
  • a preferred dosage range of glibornuride when administered orally is 5 to 75 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 5 to 75 mg, in particular 10 to 50 mg.
  • Preferably the administration of said amounts is once, twice or three-times daily.
  • a preferred dosage range of gliclazide when administered orally is 20 to 300 mg, in particular 40 to 240 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 20 to 240 mg, in particular 20 to 80 mg. Examples are 20, 30, 40 and 50 mg.
  • Preferably the administration of said amounts is once, twice or three-times daily.
  • a preferred dosage range of glisoxepide when administered orally is 1 to 20 mg, in particular 1 to 16 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 1 to 8 mg, in particular 1 to 4 mg.
  • Preferably the administration of said amounts is once, twice, three-times or four-times daily.
  • a preferred dosage range of tolbutamide when administered orally is 100 to 3000 mg, preferably 500 to 2000 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 100 to 1000 mg.
  • the administration of said amounts is once or twice daily.
  • a preferred dosage range of glipizide when administered orally is 1 to 50 mg, in particular 2.5 to 40 mg per day.
  • the preferred range of amounts in the pharmaceutical composition for an administration once, twice or three times daily is 1 to 50, 0.5 to 25 and 0.3 to 17 mg respectively.
  • a preferred dosage range of nateglinide when administered orally is 30 to 500 mg, in particular 60 to 360 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 30 to 120 mg. Examples are 30, 60 and 120 mg.
  • Preferably the administration of said amounts is once, twice or three-times daily.
  • Nateglinide can be administered in the form as it is marketed for example under the trademark STARLIXTM.
  • a preferred dosage range of repaglinide when administered orally is 0.1 to 16 mg, in particular 0.5 to 6 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 0.5 to 4 mg. Examples are 0.5, 1, 2 or 4 mg. Preferably the administration of said amounts is once, twice, three-times or four-times daily.
  • Repaglinide can be administered in the form as it is marketed for example under the trademark NOVONORMTM.
  • a preferred dosage range of acarbose when administered orally is 50 to 1000 mg, in particular 50 to 600 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 50 to 150 mg. Examples are 50 and 100 mg.
  • Preferably the administration of said amounts is once, twice, three-times or four-times daily.
  • Acarbose can be administered in the form as it is marketed for example under the trademark GlucobayTM.
  • a preferred dosage range of voglibose when administered orally is 100 to 1000 mg, in particular 200 to 600 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 50 to 300 mg. Examples are 50, 100, 150, 200 and 300 mg.
  • Preferably the administration of said amounts is once, twice, three-times or four-times daily.
  • Voglibose can be administered in the form as it is marketed for example under the trademark BasenTM or VoglisanTM.
  • a preferred dosage range of miglitol when administered orally is 25 to 500 mg, in particular 25 to 300 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 25 to 100 mg. Examples are 25, 50 and 100 mg.
  • Preferably the administration of said amounts is once, twice, three-times or four-times daily.
  • Miglitol can be administered in the form as it is marketed for example under the trademark GlysetTM.
  • a preferred dosage range of GLP-1 analogues, in particular of exenatide is 5 to 30 ⁇ g, in particular 5 to 20 ⁇ g per day.
  • the preferred range of amounts in the pharmaceutical composition is 5 to 10 ⁇ g. Examples are 5 and 10 ⁇ g.
  • Preferably the administration of said amounts is once, twice, three-times or four-times daily by subcutaneous injection.
  • Exenatide can be administered in the form as it is marketed for example under the trademark ByettaTM.
  • a long acting formulation, preferably for a once weekly subcutaneous injection comprises an amount from 0.1 to 3.0 mg, preferably 0.5 to 2.0 mg exenatide. Examples are 0.8 mg and 2.0 mg.
  • An example of a long acting formulation of exenatide is Byetta LARTM.
  • a preferred dosage range of liraglutide is 0.5 to 3 mg, in particular 0.5 to 2 mg per day.
  • the preferred range of amounts in the pharmaceutical composition is 0.5 to 2 mg. Examples are 0.6, 1.2 and 1.8 mg.
  • Preferably the administration of said amounts is once or twice daily by subcutaneous injection.
  • the amount of the DPP-4 inhibitor and the second and/or third therapeutic agent in the pharmaceutical composition and in the methods and uses of this invention correspond to the respective dosage ranges as provided hereinbefore.
  • preferred dosage ranges in a pharmaceutical composition, combination, method and use according to this invention are an amount of 0.5 to 10 mg (in particular 1 to 5 mg, especially 2.5 mg or 5 mg) of linagliptin and/or, optionally, an amount of 250 to 1000 mg (especially 500 mg, 850 mg or 1000 mg) of metformin.
  • An oral administration once or twice daily is preferred.
  • the DPP-4 inhibitor and the second and/or third therapeutic agent are administered in combination including, without being limited, the active ingredients are administered at the same time, i.e. simultaneously, or essentially at the same time, or the active ingredients are administered in alternation, i.e. that at first one or two active ingredients are administered and after a period of time the other two or one active ingredients are administered, i.e. at least two of the three active ingredients are administered sequentially.
  • the period of time may be in the range from 30 min to 12 hours.
  • the administration which is in combination or in alternation may be once, twice, three times or four times daily, preferably once or twice daily.
  • all three active ingredients may be present in one single dosage form, for example in one tablet or capsule, or one or two of the active ingredients may be present in a separate dosage form, for example in two different or identical dosage forms.
  • one or two of the active ingredients are present in a separate dosage form, for example in two different or identical dosage forms.
  • a pharmaceutical combination of this invention may be present as single dosage forms which comprise the DPP-4 inhibitor and the second and, optionally, the third antidiabetic agent.
  • a pharmaceutical combination of this invention may be present as two separate dosage forms wherein one dosage form comprises the DPP-4 inhibitor and the other dosage form comprises the second plus, optionally, the third antidiabetic agent, or, in case of a triple combination, one dosage form comprises the DPP-4 inhibitor inhibitor plus either the second or the third antidiabetic agent and the other dosage form comprises the third or the second antidiabetic agent, respectively.
  • a pharmaceutical combination of this invention may be present as three separate dosage forms wherein one dosage form comprises the DPP-4 inhibitor and a second dosage form comprises the second antidiabetic agent and the third dosage form comprises the third antidiabetic agent.
  • a pharmaceutical combination of this invention may be present as two separate dosage forms wherein one dosage form comprises the DPP-4 inhibitor and the second dosage form comprises the second antidiabetic agent.
  • “administration in combination” also includes an administration scheme in which first all active ingredients are administered in combination and after a period of time an active ingredient is administered again or vice versa.
  • the present invention also includes pharmaceutical combinations which are present in separate dosage forms wherein one dosage form comprises the DPP-4 inhibitor and the second and, optionally, the third, therapeutic agent and the other dosage form comprises the second and/or the third therapeutic agent only.
  • the present invention also includes pharmaceutical compositions or combinations for separate, sequential, simultaneous, concurrent, alternate or chronologically staggered use of the active ingredients or components.
  • a further aspect of the present invention is a manufacture comprising the pharmaceutical combination being present as separate dosage forms according to the present invention and a label or package insert comprising instructions that the separate dosage forms are to be administered in combination.
  • a manufacture comprises (a) a pharmaceutical composition comprising a DPP-4 inhibitor according to the present invention and (b) a label or package insert which comprises instructions that the medicament may or is to be administered, for example in combination, with a medicament comprising a second antidiabetic agent according to the present invention or with a fixed or free combination (e.g. a medicament) comprising a second antidiabetic agent and a third antidiabetic agent according to the present invention.
  • a manufacture comprises (a) a second antidiabetic agent according to the present invention and (b) a label or package insert which comprises instructions that the medicament may or is to be administered, for example in combination, with a medicament comprising a DPP-4 inhibitor according to the present invention or with a a fixed or free-combination (e.g. a medicament) comprising a DPP-4 inhibitor and a third antidiabetic agent according to the present invention.
  • a manufacture comprises (a) a pharmaceutical composition comprising a DPP-4 inhibitor and a second antidiabetic agent according to the present invention and (b) a label or package insert which comprises instructions that the medicament may or is to be administered, for example in combination, with a medicament comprising a third antidiabetic agent according to the present invention.
  • the desired dose of the pharmaceutical composition according to this invention may conveniently be presented in a once daily or as divided dose administered at appropriate intervals, for example as two, three or more doses per day.
  • the pharmaceutical composition may be formulated for oral, rectal, nasal, topical (including buccal and sublingual), transdermal, vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration in liquid or solid form or in a form suitable for administration by inhalation or insufflation. Oral administration is preferred.
  • the formulations may, where appropriate, be conveniently presented in discrete dosage units and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing into association the active ingredient with one or more pharmaceutically acceptable carriers, like liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired formulation.
  • the pharmaceutical composition may be formulated in the form of tablets, granules, fine granules, powders, capsules, caplets, soft capsules, pills, oral solutions, syrups, dry syrups, chewable tablets, troches, effervescent tablets, drops, suspension, fast dissolving tablets, oral fast-dispersing tablets, etc.
  • the pharmaceutical composition and the dosage forms preferably comprises one or more pharmaceutical acceptable carriers.
  • Preferred carriers must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Examples of pharmaceutically acceptable carriers are known to the one skilled in the art.
  • compositions suitable for oral administration may conveniently be presented as discrete units such as capsules, including soft gelatin capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution, a suspension or as an emulsion, for example as syrups, elixirs or self-emulsifying delivery systems (SEDDS).
  • the active ingredients may also be presented as a bolus, electuary or paste.
  • Tablets and capsules for oral administration may contain conventional excipients such as binding agents, fillers, lubricants, disintegrants, or wetting agents.
  • the tablets may be coated according to methods well known in the art.
  • Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous vehicles (which may include edible oils), or preservatives.
  • compositions according to the invention may also be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative.
  • the compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredients may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilisation from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • compositions suitable for rectal administration wherein the carrier is a solid are most preferably presented as unit dose suppositories.
  • suitable carriers include cocoa butter and other materials commonly used in the art, and the suppositories may be conveniently formed by admixture of the active compound(s) with the softened or melted carrier(s) followed by chilling and shaping in moulds.
  • the compounds of this invention are usually used in dosages from 0.001 to 100 mg/kg body weight, preferably at 0.1-15 mg/kg, in each case 1 to 4 times a day.
  • the compounds, optionally combined with other active substances may be incorporated together with one or more inert conventional carriers and/or diluents, e.g.
  • compositions according to this invention comprising the DPP-4 inhibitors as defined herein are thus prepared by the skilled person using pharmaceutically acceptable formulation excipients as described in the art.
  • excipients include, without being restricted to diluents, binders, carriers, fillers, lubricants, flow promoters, crystallisation retardants, disintegrants, solubilizers, colorants, pH regulators, surfactants and emulsifiers.
  • Suitable diluents for compounds according to embodiment A include cellulose powder, calcium hydrogen phosphate, erythritol, low substituted hydroxypropyl cellulose, mannitol, pregelatinized starch or xylitol. Among those diluents mannitol, low substituted hydroxypropyl cellulose and pregelatinized starch are to be emphasized.
  • Suitable lubricants for compounds according to embodiment A include talc, polyethyleneglycol, calcium behenate, calcium stearate, hydrogenated castor oil or magnesium stearate. Among those lubricants magnesium stearate is to be emphasized.
  • Suitable binders for compounds according to embodiment A include copovidone (copolymerisates of vinylpyrrolidon with other vinylderivates), hydroxypropyl methylcellulose (HPMC), hydroxypropylcellulose (HPC), polyvinylpyrrolidon (povidone), pregelatinized starch, or low-substituted hydroxypropylcellulose (L-HPC).
  • copovidone copolymerisates of vinylpyrrolidon with other vinylderivates
  • HPMC hydroxypropyl methylcellulose
  • HPC hydroxypropylcellulose
  • polyvinylpyrrolidon povidone
  • pregelatinized starch or low-substituted hydroxypropylcellulose (L-HPC).
  • L-HPC low-substituted hydroxypropylcellulose
  • Suitable disintegrants for compounds according to embodiment A include corn starch or crospovidone.
  • corn starch is to be emphasized.
  • An exemplary composition of a DPP-4 inhibitor according to embodiment A of the invention comprises the first diluent mannitol, pregelatinized starch as a second diluent with additional binder properties, the binder copovidone, the disintegrant corn starch, and magnesium stearate as lubricant; wherein copovidone and/or corn starch may be optional.
  • compositions may be packaged in a variety of ways.
  • an article for distribution includes a container that contains the pharmaceutical composition in an appropriate form. Tablets are typically packed in an appropriate primary package for easy handling, distribution and storage and for assurance of proper stability of the composition at prolonged contact with the environment during storage.
  • Primary containers for tablets may be bottles or blister packs.
  • a suitable bottle e.g. for a pharmaceutical composition or combination comprising a DPP-4 inhibitor according to embodiment A of the invention, may be made from glass or polymer (preferably polypropylene (PP) or high density polyethylene (HD-PE)) and sealed with a screw cap.
  • the screw cap may be provided with a child resistant safety closure (e.g. press-and-twist closure) for preventing or hampering access to the contents by children.
  • a desiccant such as e.g. bentonite clay, molecular sieves, or, preferably, silica gel
  • the shelf life of the packaged composition can be prolonged.
  • a suitable blister pack e.g. for a pharmaceutical composition or combination comprising a DPP-4 inhibitor according to embodiment A of the invention, comprises or is formed of a top foil (which is breachable by the tablets) and a bottom part (which contains pockets for the tablets).
  • the top foil may contain a metalic foil, particularly an aluminium or aluminium alloy foil (e.g. having a thickness of 20 ⁇ m to 45 ⁇ m, preferably 20 ⁇ m to 25 ⁇ m) that is coated with a heat-sealing polymer layer on its inner side (sealing side).
  • the bottom part may contain a multi-layer polymer foil (such as e.g.
  • PVDC poly(vinyl choride) coated with poly(vinylidene choride)
  • PCTFE poly(chlorotriflouroethylene)
  • multi-layer polymer-metal-polymer foil such as e.g. a cold-formable laminated PVC/aluminium/polyamide composition
  • the article may further comprise a label or package insert, which refer to instructions customarily included in commercial packages of therapeutic products, that may contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • the label or package inserts indicates that the composition can be used for any of the purposes described herein.
  • compositions and methods according to this invention show advantageous effects in the treatment and prevention of those diseases and conditions as described hereinbefore.
  • the dual combinations show advantageous effects compared with monotherapy with an active ingredient.
  • the triple combinations show advantageous effects compared with dual therapy with one or two of the three active ingredients.
  • Advantageous effects may be seen for example with respect to efficacy, dosage strength, dosage frequency, pharmacodynamic properties, pharmacokinetic properties, fewer adverse effects, convenience, compliance, etc.
  • the active ingredients may be present in the form of a pharmaceutically acceptable salt.
  • Pharmaceutically acceptable salts include, without being restricted thereto, such as salts of inorganic acid like hydrochloric acid, sulfuric acid and phosphoric acid; salts of organic carboxylic acid like oxalic acid, acetic acid, citric acid, malic acid, benzoic acid, maleic acid, fumaric acid, tartaric acid, succinic acid and glutamic acid and salts of organic sulfonic acid like methanesulfonic acid and p-toluenesulfonic acid.
  • the salts can be formed by combining the compound and an acid in the appropriate amount and ratio in a solvent and decomposer. They can be also obtained by the cation or anion exchange from the form of other salts.
  • the active ingredients or a pharmaceutically acceptable salt thereof may be present in the form of a solvate such as a hydrate or alcohol adduct.
  • a DPP-4 inhibitor is combined with active substances customary for the respective disorders, such as e.g. one or more active substances selected from among the other antidiabetic substances, especially active substances that lower the blood sugar level or the lipid level in the blood, raise the HDL level in the blood, lower blood pressure or are indicated in the treatment of atherosclerosis or obesity.
  • Such a combined treatment may be given as a free combination of the substances or in the form of a fixed combination, for example in a tablet or capsule.
  • Pharmaceutical formulations of the combination partner needed for this may either be obtained commercially as pharmaceutical compositions or may be formulated by the skilled man using conventional methods.
  • the active substances which may be obtained commercially as pharmaceutical compositions are described in numerous places in the prior art, for example in the list of drugs that appears annually, the “Rote Liste®” of the federal association of the pharmaceutical industry, or in the annually updated compilation of manufacturers' information on prescription drugs known as the “Physicians' Desk Reference”.
  • Examples of antidiabetic combination partners are metformin; sulphonylureas such as glibenclamide, tolbutamide, glimepiride, glipizide, gliquidon, glibornuride and gliclazide; nateglinide; repaglinide; thiazolidinediones such as rosiglitazone and pioglitazone; PPAR gamma modulators such as metaglidases; PPAR-gamma agonists such as rivoglitazone, mitoglitazone, INT-131 or balaglitazone; PPAR-gamma antagonists; PPAR-gamma/alpha modulators such as tesaglitazar, muraglitazar, aleglitazar, indeglitazar and KRP297; PPAR-gamma/alpha/delta modulators such as e.g.
  • AMPK-activators such as AICAR; acetyl-CoA carboxylase (ACC1 and ACC2) inhibitors; diacylglycerol-acetyltransferase (DGAT) inhibitors; pancreatic beta cell GCRP agonists such as SMT3-receptor-agonists and GPR119, such as the GPR119 agonists 5-ethyl-2- ⁇ 4-[4-(4-tetrazol-1-yl-phenoxymethyl)-thiazol-2-yl]-piperidin-1-yl ⁇ -pyrimidine or 5-[1-(3-isopropyl-[1,2,4]oxadiazol-5-yl)-piperidin-4-ylmethoxy]-2-(4-methanesulfonyl-phenyl)-pyridine; 11B-HSD-inhibitors; FGF19 agonists or analogues; alpha-glucosidase blockers such as acarb
  • GLP-1 and GLP-1 analogues such as Exendin-4, e.g. exenatide, exenatide LAR, liraglutide, taspoglutide, lixisenatide (AVE-0010), LY-2428757, dulaglutide (LY-2189265), semaglutide or albiglutide; SGLT2-inhibitors such as e.g. dapagliflozin, sergliflozin (KGT-1251), atigliflozin, canagliflozin, ipragliflozin or tofogliflozin; inhibitors of protein tyrosine-phosphatase (e.g.
  • trodusquemine inhibitors of glucose-6-phosphatase; fructose-1,6-bisphosphatase modulators; glycogen phosphorylase modulators; glucagon receptor antagonists; phosphoenolpyruvatecarboxykinase (PEPCK) inhibitors; pyruvate dehydrogenasekinase (PDK) inhibitors; inhibitors of tyrosine-kinases (50 mg to 600 mg) such as PDGF-receptor-kinase (cf. EP-A-564409, WO 98/35958, U.S. Pat. No.
  • PEPCK phosphoenolpyruvatecarboxykinase
  • PDK pyruvate dehydrogenasekinase
  • inhibitors of tyrosine-kinases 50 mg to 600 mg
  • PDGF-receptor-kinase cf. EP-A-564409, WO 98/35958, U.S. Pat
  • Metformin is usually given in doses varying from about 500 mg to 2000 mg up to 2500 mg per day using various dosing regimens from about 100 mg to 500 mg or 200 mg to 850 mg (1-3 times a day), or about 300 mg to 1000 mg once or twice a day, or delayed-release metformin in doses of about 100 mg to 1000 mg or preferably 500 mg to 1000 mg once or twice a day or about 500 mg to 2000 mg once a day.
  • Particular dosage strengths may be 250, 500, 625, 750, 850 and 1000 mg of metformin hydrochloride.
  • metformin For children 10 to 16 years of age, the recommended starting dose of metformin is 500 mg given once daily. If this dose fails to produce adequate results, the dose may be increased to 500 mg twice daily. Further increases may be made in increments of 500 mg weekly to a maximum daily dose of 2000 mg, given in divided doses (e.g. 2 or 3 divided doses). Metformin may be administered with food to decrease nausea.
  • a dosage of pioglitazone is usually of about 1-10 mg, 15 mg, 30 mg, or 45 mg once a day.
  • Rosiglitazone is usually given in doses from 4 to 8 mg once (or divided twice) a day (typical dosage strengths are 2, 4 and 8 mg).
  • Glibenclamide is usually given in doses from 2.5-5 to 20 mg once (or divided twice) a day (typical dosage strengths are 1.25, 2.5 and 5 mg), or micronized glibenclamide in doses from 0.75-3 to 12 mg once (or divided twice) a day (typical dosage strengths are 1.5, 3, 4.5 and 6 mg).
  • Glipizide is usually given in doses from 2.5 to 10-20 mg once (or up to 40 mg divided twice) a day (typical dosage strengths are 5 and 10 mg), or extended-release glibenclamide in doses from 5 to 10 mg (up to 20 mg) once a day (typical dosage strengths are 2.5, 5 and 10 mg).
  • Glimepiride is usually given in doses from 1-2 to 4 mg (up to 8 mg) once a day (typical dosage strengths are 1, 2 and 4 mg).
  • a dual combination of glibenclamide/metformin is usually given in doses from 1.25/250 once daily to 10/1000 mg twice daily. (typical dosage strengths are 1.25/250, 2.5/500 and 5/500 mg).
  • a dual combination of glipizide/metformin is usually given in doses from 2.5/250 to 10/1000 mg twice daily (typical dosage strengths are 2.5/250, 2.5/500 and 5/500 mg).
  • a dual combination of glimepiride/metformin is usually given in doses from 1/250 to 4/1000 mg twice daily.
  • a dual combination of rosiglitazone/glimepiride is usually given in doses from 4/1 once or twice daily to 4/2 mg twice daily (typical dosage strengths are 4/1, 4/2, 4/4, 8/2 and 8/4 mg).
  • a dual combination of pioglitazone/glimepiride is usually given in doses from 30/2 to 30/4 mg once daily (typical dosage strengths are 30/4 and 45/4 mg).
  • a dual combination of rosiglitazone/metformin is usually given in doses from 1/500 to 4/1000 mg twice daily (typical dosage strengths are 1/500, 2/500, 4/500, 2/1000 and 4/1000 mg).
  • a dual combination of pioglitazone/metformin is usually given in doses from 15/500 once or twice daily to 15/850 mg thrice daily (typical dosage strengths are 15/500 and 15/850 mg).
  • the non-sulphonylurea insulin secretagogue nateglinide is usually given in doses from 60 to 120 mg with meals (up to 360 mg/day, typical dosage strengths are 60 and 120 mg); repaglinide is usually given in doses from 0.5 to 4 mg with meals (up to 16 mg/day, typical dosage strengths are 0.5, 1 and 2 mg).
  • a dual combination of repaglinide/metformin is available in dosage strengths of 1/500 and 2/850 mg.
  • Acarbose is usually given in doses from 25 to 100 mg with meals.
  • Miglitol is usually given in doses from 25 to 100 mg with meals.
  • HMG-CoA-reductase inhibitors such as simvastatin, atorvastatin, lovastatin, fluvastatin, pravastatin, pitavastatin and rosuvastatin; fibrates such as bezafibrate, fenofibrate, clofibrate, gemfibrozil, etofibrate and etofyllinclofibrate; nicotinic acid and the derivatives thereof such as acipimox; PPAR-alpha agonists; PPAR-delta agonists; inhibitors of acyl-coenzyme
  • A:cholesterolacyltransferase such as avasimibe; cholesterol resorption inhibitors such as ezetimib; substances that bind to bile acid, such as cholestyramine, colestipol and colesevelam; inhibitors of bile acid transport; HDL modulating active substances such as D4F, reverse D4F, LXR modulating active substances and FXR modulating active substances; CETP inhibitors such as torcetrapib, JTT-705 (dalcetrapib) or compound 12 from WO 2007/005572 (anacetrapib); LDL receptor modulators; MTP inhibitors (e.g. lomitapide); and ApoB100 antisense RNA.
  • ACAT cholesterolacyltransferase
  • avasimibe such as avasimibe
  • cholesterol resorption inhibitors such as ezetimib
  • substances that bind to bile acid such as cholestyramine, colestipol and colesevelam
  • a dosage of atorvastatin is usually from 1 mg to 40 mg or 10 mg to 80 mg once a day.
  • beta-blockers such as atenolol, bisoprolol, celiprolol, metoprolol and carvedilol
  • diuretics such as hydrochlorothiazide, chlortalidon, xipamide, furosemide, piretanide, torasemide, spironolactone, eplerenone, amiloride and triamterene
  • calcium channel blockers such as amlodipine, nifedipine, nitrendipine, nisoldipine, nicardipine, felodipine, lacidipine, lercanipidine, manidipine, isradipine, nilvadipine, verapamil, gallopamil and diltiazem
  • ACE inhibitors such as ramipril, lisinopril, cilazapril, quinapril, captopril, enalapril, ben
  • a dosage of telmisartan is usually from 20 mg to 320 mg or 40 mg to 160 mg per day.
  • combination partners which increase the HDL level in the blood are Cholesteryl Ester Transfer Protein (CETP) inhibitors; inhibitors of endothelial lipase; regulators of ABC1; LXRalpha antagonists; LXRbeta agonists; PPAR-delta agonists; LXRalpha/beta regulators, and substances that increase the expression and/or plasma concentration of apolipoprotein A-I.
  • CETP Cholesteryl Ester Transfer Protein
  • combination partners for the treatment of obesity are sibutramine; tetrahydrolipstatin (orlistat); alizyme (cetilistat); dexfenfluramine; axokine; cannabinoid receptor 1 antagonists such as the CB1 antagonist rimonobant; MCH-1 receptor antagonists; MC4 receptor agonists; NPY5 as well as NPY2 antagonists (e.g.
  • beta3-AR agonists such as SB-418790 and AD-9677
  • 5HT2c receptor agonists such as APD 356 (lorcaserin); myostatin inhibitors; Acrp30 and adiponectin; steroyl CoA desaturase (SCD1) inhibitors; fatty acid synthase (FAS) inhibitors; CCK receptor agonists; Ghrelin receptor modulators; Pyy 3-36; orexin receptor antagonists; and tesofensine; as well as the dual combinations bupropion/naltrexone, bupropion/zonisamide, topiramate/phentermine and pramlintide/metreleptin.
  • SCD1 steroyl CoA desaturase
  • FES fatty acid synthase
  • CCK receptor agonists Ghrelin receptor modulators
  • Pyy 3-36 orexin receptor antagonists
  • tesofensine as well as the dual combinations bupropion/naltrexone, bupropion/zonisamide,
  • combination partners for the treatment of atherosclerosis are phospholipase A2 inhibitors; inhibitors of tyrosine-kinases (50 mg to 600 mg) such as PDGF-receptor-kinase (cf. EP-A-564409, WO 98/35958, U.S. Pat. No. 5,093,330, WO 2004/005281, and WO 2006/041976); oxLDL antibodies and oxLDL vaccines; apoA-1 Milano; ASA; and VCAM-1 inhibitors.
  • phospholipase A2 inhibitors inhibitors of tyrosine-kinases (50 mg to 600 mg) such as PDGF-receptor-kinase (cf. EP-A-564409, WO 98/35958, U.S. Pat. No. 5,093,330, WO 2004/005281, and WO 2006/041976); oxLDL antibodies and oxLDL vaccines; apoA-1 Milano; ASA
  • BI 1356 a Potent and Selective DPP-4 Inhibitor, is Safe and Efficacious in Patients With Inadequately Controlled Type 2 Diabetes Despite Metformin Therapy
  • BI 1356 (1, 5, or 10 mg qd), a potent and selective dipeptidyl peptidase-4 (DPP-4) inhibitor
  • DPP-4 dipeptidyl peptidase-4
  • T2DM type 2 diabetic patients
  • Effects were compared to add-on of placebo (PBO) or of open label glimepiride (GLIM; 1 to 3 mg qd) in a 12-week randomized, double-blind study.
  • Antidiabetic medication other than metformin was washed out for 6 weeks (34.7% of the patients).
  • the primary endpoint was change from baseline in HbA1c, adjusted for prior antidiabetic medication.
  • 333 patients (mean baseline HbA1c 8.3%; fasting plasma glucose [FPG] 185 mg/dL) were randomized to BI 1356, PBO or open-label GLIM.
  • DPP-4 inhibitor or combination according to this invention for the purpose of the present invention (e.g. the beneficial effect on glycemic control) can be tested using clinical trials.
  • a DPP-4 inhibitor according to the invention e.g. 5 mg of linagliptin administered orally once daily
  • HbA1c insufficient glycemic control
  • one or two conventional antihyperglycemic agents e.g. selected from metformin, thiazolidindiones (e.g. pioglitazone), sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues, and insulin or insulin analogues.
  • the success of the treatment is tested by determining the HbA1c value, by comparison with the initial value and/or with the value of the placebo group. A significant change in the HbA1c value compared with the initial value and/or the placebo value demonstrates the efficacy of the DPP-4 inhibitor for the treatment.
  • the success of the treatment can be also tested by determining the fasting plasma glucose values, by comparison with the initial values and/or with the values of the placebo group. A significant drop in the fasting glucose levels demonstrates the efficacy of the treatment. Also, the occurrence of a treat to target response (i.e. an HbA1c under treatment ⁇ 7%) demonstrates the efficacy of the treatment.
  • the safety and tolerability of the treatment is investigated by assessing patient's condition and relevant changes from baseline, e.g. incidence and intensity of adverse events (such as e.g. hypoglycaemic episodes or the like) or weight gain.
  • adverse events such as e.g. hypoglycaemic episodes or the like
  • weight gain e.g. weight gain
  • the efficacy of a pharmaceutical composition or combination according to the invention in the treatment of pre-diabetes characterised by pathological fasting glucose and/or impaired glucose tolerance can be tested using clinical studies. In studies over a shorter period (e.g. 2-4 weeks) the success of the treatment is examined by determining the fasting glucose values and/or the glucose values after a meal or after a loading test (oral glucose tolerance test or food tolerance test after a defined meal) after the end of the period of therapy for the study and comparing them with the values before the start of the study and/or with those of a placebo group. In addition, the fructosamine value can be determined before and after therapy and compared with the initial value and/or the placebo value.
  • a significant drop in the fasting or non-fasting glucose levels demonstrates the efficacy of the treatment.
  • the success of the treatment is tested by determining the HbA1c value, by comparison with the initial value and/or with the value of the placebo group.
  • a significant change in the HbA1c value compared with the initial value and/or the placebo value demonstrates the efficacy of the DPP-4 inhibitors or combinations according to the present invention for treating pre-diabetes.
  • Treating patients with pathological fasting glucose and/or impaired glucose tolerance is also in pursuit of the goal of preventing the transition to manifest type 2 diabetes.
  • the efficacy of a treatment can be investigated in a comparative clinical study in which pre-diabetes patients are treated over a lengthy period (e.g. 1-5 years) with either a pharmaceutical composition or combination according to this invention or with placebo or with a non-drug therapy or other medicaments.
  • a loading test e.g. oGTT
  • a fasting glucose level of >125 mg/dl and/or a 2 h value according to oGTT of >199 mg/dl A significant reduction in the number of patients who exhibit manifest type 2 diabetes when treated with a DPP-4 inhibitor or combination according to the present invention as compared to one of the other forms of treatment, demonstrates the efficacy in preventing a transition from pre-diabetes to manifest diabetes.
  • Treating patients with type 2 diabetes with the pharmaceutical composition or combination according to the invention in addition to producing an acute improvement in the glucose metabolic situation, prevents a deterioration in the metabolic situation in the long term. This can be observed is patients are treated for a longer period, e.g. 3 months to 1 year or even 1 to 6 years, with the pharmaceutical composition or combination according to the invention and are compared with patients who have been treated with other antidiabetic medicaments. There is evidence of therapeutic success compared with patients treated with other antidiabetic medicaments if no or only a slight increase in the fasting glucose and/or HbA1c value is observed.
  • type 2 diabetes or pre-diabetes patients with a DPP-4 inhibitor, pharmaceutical composition or combination according to the invention prevents or reduces or reduces the risk of developing microvascular complications (e.g. diabetic neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic foot, diabetic ulcer) or macrovascular complications (e.g. myocardial infarct, acute coronary syndrome, unstable angina pectoris, stable angina pectoris, stroke, peripheral arterial occlusive disease, cardiomyopathy, heart failure, heart rhythm disorders, vascular restenosis).
  • microvascular complications e.g. diabetic neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic foot, diabetic ulcer
  • macrovascular complications e.g. myocardial infarct, acute coronary syndrome, unstable angina pectoris, stable angina pectoris, stroke, peripheral arterial occlusive disease, cardiomyopathy, heart failure, heart rhythm disorders, vascular restenosis.
  • diabetic nephropathy With regard to diabetic nephropathy the following parameters may be investigated before the start, during and at the end of the study: secretion of albumin, creatinine clearance, serum creatinin values, time taken for the serum creatinine values to double, time taken until dialysis becomes necessary.
  • the efficacy of a DPP-4 inhibitor, pharmaceutical composition or combination according to the present invention according to the invention can be tested in clinical studies with varying run times (e.g. 12 weeks to 6 years) by determining the fasting glucose or non-fasting glucose (e.g. after a meal or a loading test with oGTT or a defined meal) or the HbA1c value.
  • a significant fall in these glucose values or HbA1c values during or at the end of the study, compared with the initial value or compared with a placebo group, or a group given a different therapy, proves the efficacy of an active substance or combination of active substances in the treatment of Metabolic Syndrome.
  • Examples of this are a reduction in systolic and/or diastolic blood pressure, a lowering of the plasma triglycerides, a reduction in total or LDL cholesterol, an increase in HDL cholesterol or a reduction in weight, either compared with the starting value at the beginning of the study or in comparison with a group of patients treated with placebo or a different therapy.
  • Genomic DNA samples from individual patients enrolled in a clinical trial e.g. a clinical study as described herein
  • a DPP-4 inhibitor e.g. linagliptin, e.g. in a daily oral amount of 5 mg, optionally in combination with one or more other antidiabetic agents
  • genotyped for variation(s) e.g. polymorphisms
  • candidate genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, particularly for a TCF7L2 risk genotype as described herein, and evaluated relative to each patients response in the clinical trial (cf., e.g., Example 21).
  • the association between the likelihood (e.g., increased, decreased, or no likelihood) of a favorable DPP-4 inhibitor therapy response (e.g. favorable change in HbA1c value) and genetic variations (e.g. TCF7L2 risk genotypes) or references can be investigated by applying statistical analysis to the results of genotyping.
  • the probablility of the likelihood of a favorable response of an individual resulting from treating said individual with the DPP-4 inhibitor may be thus determined by such genotyping a nucleic acid sample of the individual, for example by detecting one or more single nucleotide polymorphisms within the TCF7L2 gene, for example one SNP selected from rs7903146, rs12255372 and rs10885406, or by detecting the respective wild-type genotype (cf., e.g., Example 21).
  • Methods for genotyping i.e. determining genetic variations (e.g. polymorphisms, particularly those described herein) from patients' nucleic acid samples are known in the art.
  • molecular genetic methods to detect single nucleotide polymorphisms may be based on genetic sequencing, microarray or PCR analysis.
  • Linagliptin Monotherapy Improves Glycemic Control and Measures of ⁇ -Cell Function in Type 2 Diabetes
  • HbA1c Mean baseline demographics (HbA1c, 8.0% [SD 0.87]; fasting plasma glucose (FPG), 166.0 mg/dL [41.1]; body mass index (BMI), 29.05 kg/m2 [4.81]; age, 55.7 yrs [10.2]) are similar in both groups.
  • the primary endpoint is the change from baseline in HbA1c after 24 wks of treatment.
  • LI shows a PBO-adjusted change in HbA1c from baseline of ⁇ 0.69% (p ⁇ 0.0001) with a continuous HbA1c reduction over time of ⁇ 0.46% at 6 weeks to ⁇ 0.69% at 24 weeks (both p ⁇ 0.0001).
  • LI patients are >4-fold more likely to achieve a reduction in HbA1c of ⁇ 0.5% at 24 weeks than PBO (47.1% vs 19.0%; p ⁇ 0.0001).
  • Patients with baseline HbA1c levels of ⁇ 9.0% show the greatest reduction in HbA1c ( ⁇ 0.86%) from baseline.
  • FPG improves by ⁇ 23.3 mg/dL (p ⁇ 0.0001) vs. PBO.
  • the LI patiens show a greater reduction in the adjusted mean change from baseline at week 24 for 2-hr postprandial glucose (PPG) ( ⁇ 58.4 mg/dL; p ⁇ 0.0001) vs. PBO.
  • LI improves insulin secretion (p ⁇ 0.05), as shown by changes in HOMA-% B index (LI, 5.02 vs PBO, ⁇ 17.2 [(mU/L)/(mmol/L)]), proinsulin/insulin ratio (LI, ⁇ 0.015 vs PBO, 0.024) and the disposition index (LI, 3.05 vs PBO, ⁇ 0.68).
  • the proportion of patients reporting at least one adverse event (AE) is similar for both groups (52.4% LI; 58.7% PBO).
  • Linagliptin trough levels in patients with mild and moderate renal impairment are comparable to patients with normal renal function.
  • Linagliptin monotherapy shows a significant, clinically meaningful and sustained improvement in glycemic control reflected in changes in FPG and HbA1c, and accompanied by ⁇ -cell function improvements. Linagliptin is safe and well tolerated with no clinically significant changes in body weight or waist circumference. Linagliptin trough levels in patients with mild and moderate renal impairment are comparable to patients with normal renal function, supporting that no dose adjustment is required in renally impaired patients.
  • a multi-center, 24-week, randomized, placebo-controlled, double-blind, parallel group study examines the efficacy and safety of linagliptin (LI) administered as add-on therapy to metformin (MET) in type 2 diabetes mellitus (T2DM) hyperglycemic patients with insufficient glycemic control (HbA1c to 0.0% for patients previously treated only with metformin, or ⁇ 6.5 to ⁇ 9.0% for patients previously treated with additional oral antihyperglycemic drugs).
  • LI linagliptin
  • MET metformin
  • T2DM type 2 diabetes mellitus
  • Mean baseline characteristics and demographics HbA1c, 8.1%; fasting plasma glucose [FPG], 168.8 mg/dL; age, 56.5 yrs; BMI, 29.9 kg/m2 are similar between groups.
  • the primary endpoint is the change from baseline HbA1c after 24 weeks of treatment, evaluated with an analysis of covariance (ANCOVA) adjusted for baseline HbA1c and prior antidiabetic medication.
  • the adjusted mean treatment difference between LI+MET and PBO+MET is ⁇ 0.64% (p ⁇ 0.0001) in favor of LI+MET for change in HbA1c (%).
  • LI+MET is superior to PBO+MET in reducing the mean fasting plasma glucose (FPG) from baseline ( ⁇ 21.1 mg/dL; p ⁇ 0.0001).
  • Linagliptin Improves Glycemic Control in Type 2 Diabetes Patients Inadequately Controlled by Metformin and Sulfonylurea Without Weight Gain or Hypoglycemia
  • LI DPP-4 inhibitor linagliptin
  • HbA1c HbA1c, 8.14% (SD 0.8); fasting plasma glucose (FPG), 160.1 mg/dL (36.6); age, 58.1 yrs (9.8); BMI, 28.3 kg/m2 (4.7). Most of the pts (73.3%) have T2DM for >5 years before enrollment.
  • the primary endpoint is the change from baseline in HbA1c after 24 weeks of treatment, adjusted for baseline HbA1c. After 24 weeks of treatment, the mean HbA1c for LI+MET+SU is ⁇ 0.62% lower (p ⁇ 0.0001) relative to PBO+MET+SU. The maximum mean HbA1c reduction with LI+MET+SU is seen at week 12 ( ⁇ 0.84%).
  • Linagliptin may provide an additional option prior to insulin therapy in many patients for whom glycemia is insufficiently controlled with metformin plus a sulfonylurea agent. Linagliptin is shown to have a favorable safety and tolerablility profile. However, when linagliptin is added on pre-existing sulfonylurea therapy, hypoglycemia may occur.
  • a multi-center, 24-week, randomized, double-blind, placebo-controlled, parallel group study investigates the efficacy and safety of initial combination therapy with the DPP-4 inhibitor linagliptin (LI) and pioglitazone (PIO).
  • T2DM type 2 diabetes mellitus
  • HbA1c 7.5-11.0% insufficient glycemic control
  • OAD oral antihyperglycemic drug
  • HbA1c 8.6%; fasting plasma glucose [FPG] 190 mg/dL; age 57.5 yrs; BMI 29.0 kg/m2) are similar between the groups.
  • the primary endpoint is the change from baseline in HbA1c after 24 weeks of treatment, adjusted for baseline HbA1c and prior antidiabetic medication.
  • the adjusted mean change in HbA1c for the patients in the LI+PIO group is ⁇ 1.06% (standard error (SE) ⁇ 0.06).
  • the difference in the adjusted mean HbA1c for the LI+PIO group compared with PBO+PIO is ⁇ 0.51% (p ⁇ 0.0001; 95% confidence interval (CI), ⁇ 0.71, ⁇ 0.30).
  • Reductions in FPG are also significantly greater for the LI+PIO group compared with PBO+PIO with a treatment difference of ⁇ 14.2 mg/dL (p ⁇ 0.0001; 95% confidence interval (CI), ⁇ 21.1, ⁇ 7.3) at 24 weeks.
  • Patients in the LI+PIO group are more likely to achieve a target HbA1c of ⁇ 7% vs. those on PBO+PIO (42.9% vs.
  • Linagliptin Monotherapy Improves Glycemic Control in Japanese Patients With Type 2 Diabetes Mellitus Over 12 Weeks
  • Linagliptin demonstrates a significant and clinically meaningful improvement in glycemic control, reflected in changes in HbA1c and FPG in Japanese patients with T2DM. Both linagliptin 5 and 10 mg doses have similar efficacy in lowering HbA1c and are well tolerated within this population. 5 mg linagliptin is the therapeutic dose in Japanese patients, which is identical to the therapeutic dose in Caucasians.
  • Linagliptin Provides Superior Glycemic Control Compared to Voglibose as Monotherapy in Japanese Patients With Type 2 Diabetes
  • a multi-center, 26-week, randomized, double-blind, active-controlled, parallel group Study compares the efficacy and safety of the DPP-4 inhibitor linagliptin (LI) vs. the ⁇ -glucosidase inhibitor voglibose (VB) in drug na ⁇ ve or previously treated Japanese patients (pts) with Type 2 diabetes mellitus (T2DM) (baseline HbA1c 7.0-10.0% if drug na ⁇ ve, 7.0-9.0% if previously treated with an oral antihyperglycemic drug (OAD)).
  • LI DPP-4 inhibitor linagliptin
  • VB ⁇ -glucosidase inhibitor voglibose
  • T2DM Type 2 diabetes mellitus
  • HbA1c 8.01% [0.68]; fasting plasma glucose (FPG), 163.5 mg/dL [32.4]; BMI, 24.97 kg/m2 [3.86]; age, 60.0 yrs [9.7]
  • FPG fasting plasma glucose
  • BMI 24.97 kg/m2 [3.86]
  • age, 60.0 yrs [9.7] are similar across groups.
  • the primary endpoint is the change from baseline in HbA1c after 26 weeks.
  • the differences of adjusted mean changes from baseline in HbA1c at week 26 are ⁇ 0.32% for LI 5 mg vs.
  • the occurrence of ⁇ 1 adverse event (AE) is comparable between groups (72.3% LI 5 mg, 77.5% LI 10 mg and 71.6% VB). Of the AEs, 11.3%, 10.6% and 18.5%, respectively, are assessed as drug related. Drug-related gatsrointstinal disorders are more common in the VB (14.2%) than LI (8.2% 5 mg; 8.1% 10 mg) groups.ln the VB group, 1 hypoglycemic episode is reported vs. none in the LI groups.
  • Linagliptin monotherapy demonstrates greater efficacy than VB for improving glycemic control in Japanese patients with T2DM.
  • Both linagliptin 5 mg and linagliptin 10 mg have comparable efficacy and show statistically significant decreases in HbA1c and FPG from baseline compared with VB after 26 weeks.
  • Linagliptin is well tolerated in Japanese patients with T2DM compared to VB, with less gastrointestinal AEs, and may provide a valuable addition to the therapies available to this population.
  • 5 mg linagliptin is the therapeutic dose in Japanese patients, which is identical to the therapeutic dose in Caucasians.
  • DPP-4 dipeptidyl peptidase-4
  • linagliptin a DPP-4 inhibitor on human ⁇ -cell function
  • Human isolated islets are exposed to increased glucose concentrations (5.5-33.3 mM), 0.5 mM palmitic acid, the mixture of 2 ng/mL IL-1 ⁇ or 1,000 U/mL IFN- ⁇ for 4 days or 50 ⁇ M H2O2 for 8 h.
  • Islets are pre-treated with 500 ng/mL Interleukin-1 Receptor Antagonist (IL-1Ra, which has been shown to restore ⁇ -cell function), 100 nM linagliptin or solvent for 1 h before exposure to the diabetic stimuli and during the whole 4-day treatment period.
  • IL-1Ra Interleukin-1 Receptor Antagonist
  • islets secrete 3.8-fold more insulin at 16.7 mM than at 2.8 mM glucose.
  • stimulatory index is 1.9- and 2.4-fold decreased when islets are exposed to 11.1 mM and 33.3 mM glucose (P ⁇ 0.05).
  • IL-1Ra is similarly effective in restoring ⁇ -cell function at conditions of high glucose, palmitic acid and cytokines, but IL-1Ra fails to restore ⁇ -cell function at oxidative stress conditions induced by H2O2 treatment.
  • Nitrotyrosine levels are highly elevated in human islets under all diabetic conditions (13-, 14-, 6-, 14- and 8-fold increased at 11.1 or 33.3 mM glucose, palmitic acid, cytokines or H2O2, P ⁇ 0.05), while no elevated nitrotyrosine production is observed in islets treated with linagliptin.
  • the DPP-4 inhibitor linagliptin has comparable protective effects on gluco-, lipo- and cytokinetoxicity as IL-1Ra and, in addition, could improve ⁇ -cell function under oxidative stress conditions and blocks apoptosis (induced by H2O2 treatment).
  • the study provides evidence of a direct protective effect of linagliptin on ⁇ -cell survival and insulin secretion.
  • Renal impairment is a frequent complication of T2DM.
  • the effect of chronic renal disease on the pharmacokinetics of dipeptidyl peptidase-4 inhibitors (linagliptin, sitagliptin, alogliptin) in a rat model of chronic renal insufficiency (5/6 nephrectomy, 5/6N) is investigated: Eight weeks after surgery rats are treated orally with inhibitors for 4 days.
  • 5/6N causes a highly significantly (P ⁇ 0.001) decrease of glomerular filtration rate measured by creatinin clearance (sham: 2510 ⁇ 210 mL/24 h; 5/6N: 1665 ⁇ 104.3 mL/24 h) and increases cystatin C levels (sham: 700 ⁇ 35.7 ng/mL; 5/6N: 1434 ⁇ 77.6 ng/mL).
  • Tubular function is significantly (P ⁇ 0.001) impaired as evidenced by plasma neutrophil gelatinase-associated lipocalin (NGAL), (sham: 286 ⁇ 23 ng/ml; 5/6N: 680 ⁇ 56.3 ng/ml) and ⁇ 2 microglobulin (sham: 20.4 ⁇ 2.4 ⁇ g/mL; 5/6N: 33.3 ⁇ 1.34 ⁇ g/mL).
  • DPP-4 activity is comparable among groups.
  • linagliptin may not have to be dose-adjusted in patients with T2DM and renal impairment or diabetic nephopathy.
  • linagliptin significantly inhibits mRNA expression of profibrotic factors, such as TGF-p ⁇ 1, T1MP-1 and collagen (Col3alpha1) in the heart of uremic rats, which factors are tissue fibrosis markers of cardiac fibrosis and are increased in uremic heart. Characteristic cardiomyopathy with intestinal expansion and fibrosis develops often in uremia.
  • these antifibrotic properties of DPP-4 inhibitors may be used for the treatment of cardiac and renal injury, uremic heart, cardiac fibrosis and/or cardiomyopathy with intestinal expansion and fibrosis associated with uremia in patients with type 2 diabetes.
  • the antifibrotic action of linagliptin can be an additional benefit for patients with chronic kidney and/or heart diseases that often accompany type 2 diabetes.
  • Linagliptin is a selective and non-renal excreted inhibitor of dipeptidyl peptidase-4 (DPP-4).
  • liver fat content is significantly reduced. Changes in liver fat content are visible as early as 2 weeks on treatment.
  • linagliptin significantly reduces liver fat content and histological NAFLD in a high fat diet model.
  • Linagliptin reverses liver triglyceride content and hepatic steatosis (with greater therapeutic impact when hepatic steatosis is more pronounced).
  • the reversal of hepatic steatosis supports the use of linagliptin in patients with Type 2 diabetes as well as liver-associated diseases (NAFLD).
  • DPP-4 dipeptidyl peptidase-4
  • linagliptin improves wound healing (as shown in ob/ob mice).
  • the impact of linagliptin on inflammatory markers in wounded skin is examined and a rationale for the beneficial action of linagliptin on wound healing is provided:
  • linagliptin Wounds of linagliptin (3 mg/kg/day) and mock-treated ob/ob mice for the inflammatory markers COX-2 and MIP by RNase protection assays are investigated with no significant differences. Furthermore, linagliptin does not increase the number of apoptotic infiltrating F4/80-positive macrophages. Therefore, the expression of DPP-4 in the skin of diabetic and non-diabetic animals is assessed. Immunohisto-chemistry (IHC) and immunoblots reveal a strong expression of DPP-4 in skin from healthy and diabetic (ob/ob) mice and keratinocytes as the major cellular source of the enzyme.
  • IHC Immunohisto-chemistry
  • immunoblots reveal a strong expression of DPP-4 in skin from healthy and diabetic (ob/ob) mice and keratinocytes as the major cellular source of the enzyme.
  • a DPP-4 inhibitor being suitable for improving wound healing is such a DPP-4 inhibitor which can effectively bind to DPP-4 in the skin, e.g. to dysregulated DPP-4 in diabetic wounds, preferably in its therapeutic dose level.
  • a DPP-4 inhibitor being suitable for improving wound healing, particularly in a type 2 diabetes patient, is such a DPP-4 inhibitor which can be applied topically to wounds, e.g. comprised in wound dressings or patches or creams or ointments.
  • wounds e.g. comprised in wound dressings or patches or creams or ointments.
  • topical devices for wounds such as e.g. wound dressings or patches, comprising linagliptin and, optionally, one or more pharmaceutically acceptable carriers and/or excipients.
  • locus specific DNA fragments are amplified by polymerase chain reaction (PCR).
  • PCR is carried out using an ABI BioRad® Tetrad PCR System. Quality of the PCR products is analyzed by agarose gel electrophoresis The purified PCR-products are used as templates in sequencing reactions According to the chain terminating methodology of Sanger et al. (1977), the analysis of DNA sequence is based on the termination of a growing DNA strand due to incorporation of a dye-labeled 2′,3′-Dideoxyribonucleotidetriphosphate (ddNTP) by the DNA polymerase. Purified sequencing products are analyzed using an ABI PRISM® 3730 Genetic Analyzer.
  • ddNTP dye-labeled 2′,3′-Dideoxyribonucleotidetriphosphate
  • Sequencing data are generated using the original ABI Software. The subsequent KB-basecalling as well as the assembly is performed using the Staden Software Package. KB-basecalling assigns quality values to all called bases of automated sequencer traces using KB-basecaller error probabilities. These quality values are used during assembling the single reads and are the basic requirement for calculating the sequence accuracy (Applied Biosystems, 3730/3730xl/DNA Analyzer Sequencing Analysis Software Training).
  • a quality value (q) of 20 corresponds to an error probability (ep) of 1/100, a value of 30 to an ep of 1/1000 and so on. In the assembly phase those values are set against each other. In general sequencing is continued until each consensus base has a quality value (q) of 50 or more. This corresponds to an error probability (ep) of 1/100000. Due to the fact that most of the consensus bases have an even higher quality score than the minimal one, the calculated cumulative error probability for the finished sequence is again significantly lower. Sequencing data are uploaded and analyzed using the software seqpatient from jsi-medical systems (version Seq Pilot 3.3.2, JSI medical systems GmbH, Friedhofstr. 5, 77971 Kippenheim, Germany).
  • genotyping results refer only to the variant positions depicted in Table i.
  • the TaqMan® technology comprises amplification of a PCR fragment with simultaneous detection of the degradation of a labelled probe.
  • Probes are labelled at both ends with an allele-specific dye and a quencher.
  • the specifically hybridized probe is displaced by the DNA polymerase. This displacement occurs either as degradation through the 5′ exonuclease activity of the polymerase in the case of a perfect match with the probe, or without degradation in the case of a mismatch.
  • the quencher and dye are separated and the fluorescence signal increased. An increase in the fluorescence signal is indicative for the presence of the respective allele. Fluorescence signals are recorded with the ABI PRISM 7700 system (Applied Biosystems).
  • a master mix is prepared containing all components for PCR reaction and aliquoted in the appropriate number of wells. Subsequently, DNA is added to each well according to the plate layout; except for no-template control (NTC).
  • NTC no-template control
  • the mastermix per sample contains:
  • the cycling conditions are:
  • the TaqMan® pre- and post-reads of the AD are performed on the TaqMan® 7900HT Fast Real System.
  • the SDS software V2.3 calculates the fluorescence measurements made during the plate read and plots Rn values based on the signals from each well. Using the software, it is determined which SNP alleles are present in each sample. NTC should be given as not determined.
  • ANCOVA covariance model
  • the statistical model includes ‘Treatment’, ‘Genotype’, ‘Study’, ‘Wash-Out-Period for prior oral antidiabetic drugs (yes/no)’, ‘Race’, as well as the interaction term ‘Treatment*Genotype’ as fixed effects and ‘HbA1c baseline’ as a linear covariate.
  • the ANCOVA model provides estimates for the mean change from baseline in HbA1c after 24 weeks of therapy for the different genotypes taking baseline clinical and demographic information into account.
  • Model based pair-wise comparisons between wild-type homozygous (genotype CC) and heterozygous (genotype CT) or rare homozygous (genotype TT) individuals on linagliptin or combination treatment are performed.
  • the statistical evaluation is prepared using the software packages SAS Version 9.2 (SAS Institute Inc., Cary, N.C., USA) and S-PLUS® 8.0 (Insightful Corp., Seattle, Wash., USA).
  • Results are shown as point estimates and 95% confidence intervals for the mean change in HbA1c from baseline [%] after 24 weeks as estimated by ANCOVA models.
  • the results are given for the whole patient population of the studies (full analysis set, FAS), for the subpopulation for which genetic analyses are performed (full analysis set for pharmacogenetic analyses, FASG), as well as for the subgroups defined by genotype (CC, CT, TT) of this subpopulation.
  • the numbers of patients for placebo control and linagliptin treatment are given in braces.
  • active substance denotes one or more compounds according to the invention, i.e. denotes a DPP-4 inhibitor or a second or third antidiabetic compound according to this invention or a combination of two or three of said active ingredients, for example selected from the combinations as listed in the Table 1 or 2.
  • Additional suitable formulations for the DPP-4 inhibitor linagliptin may be those formulations disclosed in the application WO 2007/128724, the disclosure of which is incorporated herein in its entirety.
  • Additional suitable formulations for the other DPP-4 inhibitors may be those formulations which are available on the market, or formulations described in the patent applications cited above in paragraph “background of the invention”, or those described in the literature, for example as disclosed in current issues of “Rote Liste®” (Germany) or of “Physician's Desk Reference”.
  • Active substance and mannitol are dissolved in water. After packaging the solution is freeze-dried. To produce the solution ready for use, the product is dissolved in water for injections.
  • Active substance and mannitol are dissolved in water. After packaging, the solution is freeze-dried.
  • the product is dissolved in water for injections.
  • Diameter of the tablets 9 mm.
  • Diameter of the tablets 12 mm.
  • (1) is triturated with (3). This trituration is added to the mixture of (2) and (4) with vigorous mixing. This powder mixture is packed into size 3 hard gelatin capsules in a capsule filling machine.
  • (1) is triturated with (3). This trituration is added to the mixture of (2) and (4) with vigorous mixing. This powder mixture is packed into size 0 hard gelatin capsules in a capsule filling machine.

Abstract

The present invention relates to methods for preventing or treating of metabolic disorders and related conditions, such as in certain patient groups.

Description

    TECHNICAL FIELD OF THE INVENTION
  • The invention describes DPP-4 inhibitors, pharmaceutical compositions or combinations comprising a DPP-4 inhibitor as defined herein and optionally one or more other active substances, for use in methods of treatment or prevention as described herein, such as e.g. of one or more conditions selected from type 1 diabetes mellitus, type 2 diabetes mellitus, impaired glucose tolerance, impaired fasting blood glucose and hyperglycemia inter alia. In a particular embodiment, the therapeutic and/or preventive methods of this invention comprise the step of identifying a patient being susceptible to the treatment and/or prevention, said identifying comprising testing whether the patient has variation(s) in one or more genes associated with metabolic diseases (e.g. whether the patient is of a TCF7L2 risk genotype as described herein) or whether the patient is of respective wild-type genotype (e.g. whether the patient is of TCF7L2 wild genotype as described herein), and the further step of administering such DPP-4 inhibitor, pharmaceutical composition or combination to the patient determined as being susceptible.
  • Further, in one embodiment, the usability of a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament each as described herein for a therapeutic and/or preventive method or use according this invention in a patient who has variation(s) in one or more genes associated with metabolic diseases (such as e.g. a TCF7L2 risk genotype patient as described herein) is contemplated.
  • TCF7L2 risk genotype patients according to this invention include, without being limited, patients (particularly type 2 diabetes patients) harboring genetic risk variants in the gene TCF7L2 and suffering often from the pathological influences thereof, particularly associated with the risk T-allele of TCF7L2 rs7903146, such as patients harboring the TCF7L2 rs7903146 CT heterozygous risk genotype or patients harboring the TCF7L2 rs7903146 TT homozygous high risk genotype.
  • Further, in another embodiment, the usability of a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament each as described herein for a therapeutic and/or preventive method or use according this invention in a patient who carries the TCF7L2 wild genotype, particularly the TCF7L2 rs7903146 CC wild genotype, is contemplated.
  • Moreover, the present invention provides a diagnostic method for identifying a subject (particularly a type 2 diabetes patient) statistically more likely to have a favorable response (e.g. in achieving glycemic control, such as change in HbA1c) to the administration of a therapeutically effective amount of a DPP-4 inhibitor, optionally in combination with one or more other active substances (e.g. antidiabetics), said method comprising determining whether the subject is either of TCF7L2 risk genotype (particularly TCF7L2 rs7903146 CT or TT risk genotype) or of TCF7L2 wild genotype (particularly TCF7L2 rs7903146 CC wild genotype), wherein the subject being of TCF7L2 rs7903146 CC homozygous wild genotype (and, to a lesser extent, the subject being of TCF7L2 rs7903146 CT heterozygous risk genotype) has an increased likelihood of favorable response to the administered DPP-4 inhibitor relative to a subject of TCF7L2 rs7903146 TT homozygous risk genotype.
  • Furthermore the invention describes a method
      • for preventing, slowing progression of, delaying, or treating a metabolic disorder;
      • for improving glycemic control and/or for reducing of fasting plasma glucose, of postprandial plasma glucose and/or of glycosylated hemoglobin HbA1c;
      • for preventing, slowing, delaying or reversing progression from impaired glucose tolerance, impaired fasting blood glucose, insulin resistance and/or from metabolic syndrome to type 2 diabetes mellitus;
      • for preventing, slowing progression of, delaying or treating of a condition or disorder selected from the group consisting of complications of diabetes mellitus;
      • 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;
      • for preventing or treating the degeneration of pancreatic beta cells and/or for improving and/or restoring or protecting the functionality of pancreatic beta cells and/or restoring the functionality of pancreatic insulin secretion;
      • for preventing, slowing, delaying or treating diseases or conditions attributed to an abnormal accumulation of liver or ectopic fat; or
      • for maintaining and/or improving the insulin sensitivity and/or for treating or preventing hyperinsulinemia and/or insulin resistance;
      • for preventing, slowing progression of, delaying, or treating new onset diabetes after transplantation (NODAT) and/or post-transplant metabolic syndrome (PTMS);
      • for preventing, delaying, or reducing NODAT and/or PTMS associated complications including micro- and macrovascular diseases and events, graft rejection, infection, and death;
      • for treating hyperuricemia and hyperuricemia associated conditions;
  • in patients in need thereof, for example in those patients (particularly type 2 diabetes mellitus patients) who have variation(s) in one or more genes associated with metabolic diseases (such as e.g. in a TCF7L2 risk genotype patient as described herein) or in those patients which are of respective wild-type genotype (such as e.g. in a TCF7L2 wild genotype as described herein), wherein said method comprises
  • testing the patient whether he/she has variation(s) in one or more genes associated with metabolic diseases (e.g. whether he/she is of a TCF7L2 risk genotype as described herein) or whether the patient is of respective wild-type genotype (e.g. whether the patient is of TCF7L2 wild genotype as described herein), and
  • administering a DPP-4 inhibitor as defined hereinafter (preferably linagliptin), optionally in combination with one or more other active substances.
  • In addition, the present invention describes the use of a DPP-4 inhibitor for the manufacture of a medicament for use in a method as described hereinbefore and hereinafter.
  • In addition, the present invention describes a DPP-4 inhibitor for use in a therapy of a patient (particularly human type 2 diabetes patient) as described hereinbefore and hereinafter.
  • In addition, the present invention describes a DPP-4 inhibitor for use in a treatment or prevention of a (particularly metabolic) disease, disorder or condition (particularly diabetes, especially type 2 diabetes, and conditions related thereto, such as e.g. diabetic complications) as described hereinbefore and hereinafter.
  • The invention also describes a use of a pharmaceutical composition or combination according to this invention for the manufacture of a medicament for use in a method as described hereinbefore and hereinafter.
  • The invention also relates to the DPP-4 inhibitors as defined herein for use in a method as described hereinbefore and hereinafter, said method comprising administering the DPP-4 inhibitor, optionally in combination with one or more other active substances (e.g. which may selected from those mentioned herein), to the patient.
  • BACKGROUND OF THE INVENTION
  • Type 2 diabetes is an increasingly prevalent disease that due to a high frequency of complications leads to a significant reduction of life expectancy. Because of diabetes-associated microvascular complications, type 2 diabetes is currently the most frequent cause of adult-onset loss of vision, renal failure, and amputations in the industrialized world. In addition, the presence of type 2 diabetes is associated with a two to five fold increase in cardiovascular disease risk.
  • After long duration of disease, most patients with type 2 diabetes will eventually fail on oral therapy and become insulin dependent with the necessity for daily injections and multiple daily glucose measurements.
  • The UKPDS (United Kingdom Prospective Diabetes Study) demonstrated that intensive treatment with metformin, sulfonylureas or insulin resulted in only a limited improvement of glycemic control (difference in HbA1c ˜0.9%). In addition, even in patients within the intensive treatment arm glycemic control deteriorated significantly over time and this was attributed to deterioration of β-cell function. Importantly, intensive treatment was not associated with a significant reduction in macrovascular complications, i.e. cardiovascular events. Therefore many patients with type 2 diabetes remain inadequately treated, partly because of limitations in long term efficacy, tolerability and dosing inconvenience of existing antihyperglycemic therapies.
  • Oral and non-oral antidiabetic drugs conventionally used in therapy (such as e.g. first- or second-line, and/or mono- or (initial or add-on) combination therapy) include, without being restricted thereto, metformin, sulphonylureas, thiazolidinediones, glinides, a-glucosidase inhibitors, GLP-1 or GLP-1 analogues, and insulin or insulin analogues.
  • The high incidence of therapeutic failure is a major contributor to the high rate of long-term hyperglycemia-associated complications or chronic damages (including micro- and makrovascular complications such as e.g. diabetic nephrophathy, retinopathy or neuropathy, or cardiovascular complications) in patients with type 2 diabetes.
  • Genetic association studies have identified genetic variations in several genes which are associated with increased risk of type 2 diabetes mellitus. E.g. variations in the genes TCF7L2, KCNJ11 and PPARG independently and interactively increase the risk of progression from impaired fasting glucose and impaired glucose tolerance to overt diabetes. While variation in KCNJ11 may alter insulin secretion and variation in PPARG may alter insulin action, TCF7L2 (transcription factor 7-like 2) is the major susceptibility gene identified to date for type 2 diabetes in various ethnic groups (e.g. Europeans, Indian and Japanese people, Mexican Americans and West Africans). Polymorphisms (single nucleotid polymorphisms, so called SNPs) in TCF7L2, such as e.g. rs12255372 and, particularly, rs7903146, are strongly associated with diabetes. The risk of developing type 2 diabetes is increased by roughly 45% (Odds ratio 1.45) among carriers of one risk T-allele of TCF7L2 rs7903146 (CT heterozygotes), and is at least doubled (Odds ratio of 2.41) among TT homozygotes compared to CC homozygotes wild genotypes (Grant et al, Nature Genetics, Vol. 38, 2006, p 320-323). TCF7L2 risk genotypes are associated with increased TCF7L2 expression in pancreatic beta cells, impaired (glucose-stimulated) insulin secretion, incretin effects and enhanced rate of hepatic glucose production as well as predisposition to and prediction of future type 2 diabetes (cf. Lyssenko et al., The Journal of Clinical Investigation, Vol. 117, No 8, 2007, p. 2155-2163). There is evidence that the TCF7L2 rs7903146 risk variants are associated with lower incretin effect on insulin secretion, which may be based, at least in parts, on an impaired sensitivity of the beta cells to incretins.
  • Thus, diabetes patients harboring TCF7L2 risk variants, particularly carriers of the at risk T-allele of TCF7L2 rs7903146, such as patients harboring the TCF7L2 rs7903146 CT genotype or, particularly, patients harboring the TCF7L2 rs7903146 TT genotype, are expected to be difficult to treat in antidiabetic therapy.
  • Therefore, there is an unmet medical need for methods, medicaments and pharmaceutical compositions or combinations with a good efficacy with regard to glycemic control, with regard to disease-modifying properties and with regard to reduction of cardiovascular morbidity and mortality while at the same time showing an improved safety profile.
  • DPP-4 inhibitors represent another novel class of agents that are being developed for the treatment or improvement in glycemic control in patients with type 2 diabetes.
  • For example, DPP-4 inhibitors and their uses are disclosed in WO 2002/068420, WO 2004/018467, WO 2004/018468, WO 2004/018469, WO 2004/041820, WO 2004/046148, WO 2005/051950, WO 2005/082906, WO 2005/063750, WO 2005/085246, WO 2006/027204, WO 2006/029769, W02007/014886; WO 2004/050658, WO 2004/111051, WO 2005/058901, WO 2005/097798; WO 2006/068163, WO 2007/071738, WO 2008/017670; WO 2007/128721, WO 2007/128724, WO 2007/128761, or WO 2009/121945.
  • AIM OF THE PRESENT INVENTION
  • The aim of the present invention is to provide a medication and/or method for preventing, slowing progression of, delaying or treating a metabolic disorder, in particular of type 2 diabetes mellitus.
  • A further aim of the present invention is to provide a medication and/or method for improving glycemic control in a patient in need thereof, in particular in patients with type 2 diabetes mellitus, for example in those patients who have variation(s) in one or more genes associated with metabolic diseases (such as e.g. a TCF7L2 risk genotype patient as described herein) or in those patients who are of respective wild-type genotype.
  • Another aim of the present invention is to provide a medication and/or method for improving glycemic control in a patient with insufficient glycemic control despite monotherapy with an antidiabetic drug, for example metformin, or despite combination therapy with two or three antidiabetic drugs, for example in such a patient who has variation(s) in one or more genes associated with metabolic diseases (such as e.g. a TCF7L2 risk genotype patient as described herein) or in such a patient who is of respective wild-type genotype.
  • Another aim of the present invention is to provide a medication and/or method for preventing, slowing or delaying progression from impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), insulin resistance and/or metabolic syndrome to type 2 diabetes mellitus.
  • Yet another aim of the present invention is to provide a medication and/or method for preventing, slowing progression of, delaying or treating of a condition or disorder from the group consisting of complications of diabetes mellitus.
  • A further aim of the present invention is to provide a medication and/or method for reducing the weight or preventing an increase of the weight in a patient in need thereof, for example in such a patient who has variation(s) in one or more genes associated with metabolic diseases (such as e.g. a TCF7L2 risk genotype patient as described herein) or in such a patient who is of respective wild-type genotype.
  • Another aim of the present invention is to provide a medication with a high efficacy for the treatment of metabolic disorders, in particular of diabetes mellitus, impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), and/or hyperglycemia, which has good to very good pharmacological and/or pharmacokinetic and/or physicochemical properties.
  • Further aims of the present invention become apparent to the one skilled in the art by description hereinbefore and in the following and by the examples.
  • SUMMARY OF THE INVENTION
  • Within the scope of the present invention it has now been found that a DPP-4 inhibitor, preferably linagliptin, as well as a pharmaceutical composition or combination comprising the DPP-4 inhibitor and optionally one or more other active substances (e.g. antidiabetics), is therapeutically effective for improving glycemic control and treating type 2 diabetes mellitus in TCF7L2 rs7903146 CT or TT risk genotype patients and in TCF7L2 rs7903146 CC wild genotype patients.
  • In particular, it has been found that all investigated TCF7L2 genotype patients (patients with TCF7L2 rs7903146 CT or TT risk genotype or with TCF7L2 rs7903146 CC wild genotype) have a clinically meaningful response to the administered DPP-4 inhibitor, preferably linagliptin.
  • Thus, within the scope of the present invention, certain subgroups of diabetes patients amenable to antidiabetic therapy according to this invention (comprising using preferably linagliptin, optionally in combination with one or more other active substances such as e.g. other antidiabetics as described herein), include for example, without being limited to, those patients harboring TCF7L2 rs7903146 CC or CT or TT genotype, respectively.
  • Within the scope of the present invention it has further been found that DPP-4 inhibitors as defined herein as well as pharmaceutical compositions or combinations comprising a DPP-4 inhibitor as defined herein and optionally one or more other active substances can be used in a method of preventing, slowing progression of, delaying (e.g. delaying the onset of) or treating a metabolic disorder (particularly diabetes, especially type 2 diabetes mellitus and conditions related thereto, e.g. diabetic complications), in particular a method for improving glycemic control in a patient, such as in a patient who has variation(s) in one or more genes associated with metabolic diseases (such as e.g. in TCF7L2 risk genotype patients as described herein).
  • Within the scope of the present invention it has further been found that DPP-4 inhibitors as defined herein as well as pharmaceutical compositions or combinations comprising a DPP-4 inhibitor as defined herein and optionally one or more other active substances can be used in a method of preventing, slowing progression of, delaying (e.g. delaying the onset of) or treating a metabolic disorder (particularly diabetes, especially type 2 diabetes mellitus and conditions related thereto), in particular a method for improving glycemic control in a patient, such as in a patient who is of TCF7L2 wild genotype, particularly of TCF7L2 rs7903146 CC wild genotype.
  • In an embodiment the method comprises the step of of identifying a patient being susceptible to the method being used, e.g. comprising testing whether the patient has variation(s) in one or more genes associated with metabolic diseases (e.g. whether the patient is of a TCF7L2 risk genotype as described herein) or whether the patient is of TCF7L2 wild genotype as described herein, and the step of administering such a DPP-4 inhibitor, pharmaceutical composition or combination to the patient determined as being susceptible.
  • This opens up new therapeutic possibilities in the treatment and prevention of type 2 diabetes mellitus, overweight, obesity, complications of diabetes mellitus and of neighboring disease states, including such patients who have variation(s) in one or more genes associated with metabolic diseases (such as e.g. in TCF7L2 risk genotype patients as described herein) and such patients who are of respective wild-type genotype (such as e.g. TCF7L2 wild genotype patients as described herein).
  • Moreover, the present invention provides a method for determining of a probability of the likelihood of a favorable response (e.g. in providing glycemic control) or the magnitude of a favorable change in HbA1c of an individual resulting from treating the individual with a DPP-4 inhibitor, preferably linagliptin, or the DPP-4 inhibitor in combination with one or more other active substances (e.g. antidiabetics), said method comprising determining whether the subject is either of TCF7L2 risk genotype (particularly TCF7L2 rs7903146 TT risk genotype) or of TCF7L2 wild genotype (particularly TCF7L2 rs7903146 CC wild genotype), wherein the probability of likelihood of a favorable response or the significantly high magnitude of a favorable change in HbA1c response to administration of the DPP-4 inhibitor, preferably linagliptin, or the DPP-4 inhibitor in combination with one or more other active substances (e.g. antidiabetics) is
  • greater in an individual being of TCF7L2 rs7903146 CC homozygous wild genotype, and lower in an individual of TCF7L2 rs7903146 TT homozygous risk genotype (e.g. but still clinically significant or meaningful).
  • Therefore, in a one aspect there is provided a pharmaceutical composition or combination comprising
  • (a) a DPP-4 inhibitor, and, optionally,
  • (b) a second antidiabetic agent selected from the group G3 consisting of biguanides (particularly metformin), thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues and insulin or insulin analogues, and, optionally,
  • (c) a third antidiabetic agent being different from (b) selected from the group G3 consisting of biguanides (particularly metformin), thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues and insulin or insulin analogues,
  • or a pharmaceutically acceptable salt thereof.
  • In a subaspect there is provided a pharmaceutical composition or combination comprising
  • (a) a DPP-4 inhibitor, and, optionally,
  • (b) a second antidiabetic agent selected from the group G3 consisting of biguanides (particularly metformin), thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues and insulin or insulin analogues, and, optionally,
  • (c) a third antidiabetic agent being different from (b) selected from the group consisting of metformin, a sulfonylurea, pioglitazone, rosiglitazone, repaglinide, nateglinide, acarbose, voglibose, miglitol, GLP-1 or a GLP-1 analogue and insulin or an insulin analogue,
  • or a pharmaceutically acceptable salt thereof.
  • In another subaspect there is provided a pharmaceutical composition or combination comprising
  • (a) a DPP-4 inhibitor, and, optionally,
  • (b) a second antidiabetic agent selected from the group consisting of metformin, a sulfonylurea, pioglitazone, rosiglitazone, repaglinide, nateglinide, acarbose, voglibose, miglitol, GLP-1 or a GLP-1 analogue and insulin or an insulin analogue, and, optionally,
  • (c) a third antidiabetic agent being different from (b) selected from the group G3 consisting of biguanides (particularly metformin), thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues and insulin or insulin analogues,
  • or a pharmaceutically acceptable salt thereof.
  • In a further subaspect there is provided a pharmaceutical composition or combination comprising
  • (a) a DPP-4 inhibitor, and, optionally,
  • (b) a second antidiabetic agent selected from the group consisting of metformin, a sulfonylurea and pioglitazone, and, optionally,
  • (c) a third antidiabetic agent being different from (b) selected from the group consisting of metformin, a sulfonylurea, pioglitazone, rosiglitazone, repaglinide, nateglinide, acarbose, voglibose, miglitol, GLP-1 or GLP-1 analogue and insulin or insulin analogue,
  • or a pharmaceutically acceptable salt thereof.
  • In a further subaspect there is provided a pharmaceutical composition or combination comprising
  • (a) a DPP-4 inhibitor, and, optionally,
  • (b) a second antidiabetic agent selected from the group consisting of metformin, a sulfonylurea, pioglitazone, rosiglitazone, repaglinide, nateglinide, acarbose, voglibose, miglitol, GLP-1 or GLP-1 analogue and insulin or insulin analogue, and, optionally,
  • (c) a third antidiabetic agent being different from (b) selected from the group consisting of metformin, a sulfonylurea and pioglitazone,
  • or a pharmaceutically acceptable salt thereof.
  • In a yet further subaspect there is provided a pharmaceutical composition or combination comprising
  • (a) a DPP-4 inhibitor, and, optionally,
  • (b) a second antidiabetic agent selected from the group consisting of metformin and pioglitazone, and, optionally,
  • (c) a third antidiabetic agent being different from (b) selected from the group consisting of metformin, a sulfonylurea and pioglitazone,
  • or a pharmaceutically acceptable salt thereof.
  • In a yet further subaspect there is provided a pharmaceutical composition or combination comprising
  • (a) a DPP-4 inhibitor, and, optionally,
  • (b) a second antidiabetic agent selected from the group consisting of metformin, a sulfonylurea and pioglitazone, and, optionally,
  • (c) a third antidiabetic agent being different from (b) selected from the group consisting of metformin and pioglitazone,
  • or a pharmaceutically acceptable salt thereof.
  • When—besides the second anidiabetic agent—a third antidiabetic agent is chosen, said third antidiabetic agent is preferably chosen from another class than the second antidiabetic agent. Thus, it is to be understood that the second and the third antidiabetic agent are different, and preferably they are from different classes (e.g. when the second antidiabetic agent is chosen from the biguanide class, the third antidiabetic agent is preferably chosen from another class). Classes of antidiabetic agents are mentioned above, e.g. biguanide class, thiazolidindione class, sulfonylurea class, glinide class, alpha-glucosidase inhibitor class, GLP-1 analogue class, insulin class, etc.
  • A particular embodiment of this invention refers to monotherapy with a DPP-4 inhibitor as defined herein and/or to pharmaceutical compositions comprising a DPP-4 inhibitor as sole active ingredient.
  • Within combinations and/or combination therapy of this invention, a particular embodiment refers to dual combinations and/or dual therapy; another embodiment refers to triple combinations and/or triple therapy.
  • According to another aspect there is provided a method for preventing, slowing the progression of, delaying or treating a metabolic disorder selected from the group consisting of type 1 diabetes mellitus, type 2 diabetes mellitus, impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), hyperglycemia, postprandial hyperglycemia, overweight, obesity and metabolic syndrome in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • According to another aspect there is provided a method for preventing, slowing the progression of, delaying or treating a metabolic disorder selected from the group consisting of insulin resistance, hyperlipidemia, hypercholesterolemia, dyslipidemia, hypertension, chronic systemic inflammation, retinopathy, neuropathy, nephropathy, atherosclerosis, endothelial dysfunction, non-alcoholic fatty liver disease (NAFLD) and osteoporosis in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • According to another aspect 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • The pharmaceutical composition of 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 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • As by the use of a pharmaceutical composition or combination of 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.
  • According to another aspect 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 nephropathy, retinopathy, neuropathy, learning and memory impairment, neurodegenerative or cognitive disorders, cardio- or cerebrovascular diseases, arteriosclerosis, hypertension, endothelial dysfunction, myocardial infarction, accute coronary syndrome, unstable angina pectoris, stable angina pectoris, cardiomyopathy, heart failure, heart rhythm disorders, vascular restenosis, peripheral arterial occlusive disease, stroke, tissue ischaemia or diabetic foot or ulcus, in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient. In particular one or more aspects of diabetic nephropathy such as hyperperfusion, proteinuria and albuminuria (including micro- or macroalbuminuria) 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.
  • In an embodiment, by the administration of a pharmaceutical composition or combination of this invention no gain in weight or even a reduction in body weight is the result.
  • According to another aspect 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • In an embodiment, by an administration of a pharmaceutical composition or combination according to this invention a beta-cell degeneration and a decline of beta-cell functionality such as for example apoptosis or necrosis of pancreatic beta cells can be delayed or prevented. Furthermore, the functionality of pancreatic cells can be improved or restored, and the number and size of pancreatic beta cells increased. It may be shown that the differentiation status and hyperplasia of pancreatic beta-cells disturbed by hyperglycemia can be normalized by treatment with a pharmaceutical composition or combination of this invention.
  • According to another aspect there is provided a method for preventing, slowing, delaying or treating the degeneration of pancreatic beta cells and/or the decline of the functionality of pancreatic beta cells and/or for improving and/or restoring the functionality of pancreatic beta cells and/or restoring the functionality of pancreatic insulin secretion in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • In an embodiment, by the administration of a pharmaceutical composition or combination of the present invention, an abnormal accumulation of (ectopic) fat, in particular in the liver, may be reduced or inhibited.
  • According to another aspect there is provided a method for preventing, slowing, delaying or treating diseases or conditions attributed to an abnormal accumulation of liver or ectopic fat in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient. Diseases or conditions which are attributed to an abnormal accumulation of liver or ectopic 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, particularly non-alcoholic fatty liver disease (NAFLD), including hepatic steatosis, non-alcoholic steatohepatitis (NASH) and/or liver fibrosis.
  • According to a further aspect of the present invention, there is provided a method for preventing, slowing the progression, delaying, attenuating, treating or reversing hepatic steatosis, (hepatic) inflammation and/or an abnormal accumulation of liver fat in a patient in need thereof characterized in that a DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • According to another aspect there is provided 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • According to another aspect there is provided the use of a DPP-4 inhibitor for the manufacture of a medicament for use in a method of
      • preventing, slowing the progression of, delaying or treating a metabolic disorder selected from the group consisting of type 1 diabetes mellitus, type 2 diabetes mellitus, impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), hyperglycemia, postprandial hyperglycemia, overweight, obesity and metabolic syndrome; or
      • improving glycemic control and/or for reducing of fasting plasma glucose, of postprandial plasma glucose and/or of glycosylated hemoglobin HbA1c; or
      • 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; or
      • 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 nephropathy, retinopathy, neuropathy, tissue ischaemia, arteriosclerosis, myocardial infarction, stroke and peripheral arterial occlusive disease; or
      • 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; or
      • preventing, slowing, delaying or treating the degeneration of pancreatic beta cells and/or the decline of the functionality of pancreatic beta cells and/or for improving and/or restoring or protecting the functionality of pancreatic beta cells and/or restoring the functionality of pancreatic insulin secretion; or
      • preventing, slowing, delaying or treating diseases or conditions attributed to an abnormal accumulation of liver or ectopic fat; or
      • maintaining and/or improving the insulin sensitivity and/or for treating or preventing hyperinsulinemia and/or insulin resistance; or
      • preventing, slowing progression of, delaying, or treating new onset diabetes after transplantation (NODAT) and/or post-transplant metabolic syndrome (PTMS); or
      • preventing, delaying, or reducing NODAT and/or PTMS associated complications including micro- and macrovascular diseases and events, graft rejection, infection, and death; or
      • treating hyperuricemia and hyperuricemia associated conditions;
  • in a patient in need thereof, comprising administering the DPP-4 inhibitor alone or, optionally, in combination with a second and, optionally, with a third antidiabetic agent as defined hereinbefore and hereinafter to the patient.
  • According to another aspect there is provided the use of a second antidiabetic agent as defined hereinbefore and hereinafter for the manufacture of a medicament for use in a method of
      • preventing, slowing the progression of, delaying or treating a metabolic disorder selected from the group consisting of type 1 diabetes mellitus, type 2 diabetes mellitus, impaired glucose tolerance (IGT), impaired fasting blood glucose (IFG), hyperglycemia, postprandial hyperglycemia, overweight, obesity and metabolic syndrome; or
      • improving glycemic control and/or for reducing of fasting plasma glucose, of postprandial plasma glucose and/or of glycosylated hemoglobin HbA1c; or
      • 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; or
      • 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 nephropathy, retinopathy, neuropathy, tissue ischaemia, arteriosclerosis, myocardial infarction, stroke and peripheral arterial occlusive disease; or
      • 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; or
      • preventing, slowing, delaying or treating the degeneration of pancreatic beta cells and/or the decline of the functionality of pancreatic beta cells and/or for improving and/or restoring the functionality of pancreatic beta cells and/or restoring the functionality of pancreatic insulin secretion; or
      • preventing, slowing, delaying or treating diseases or conditions attributed to an abnormal accumulation of liver or ectopic fat; or
      • maintaining and/or improving the insulin sensitivity and/or for treating or preventing hyperinsulinemia and/or insulin resistance;
  • in a patient in need thereof, comprising administering the second antidiabetic agent in combination with a DPP-4 inhibitor and, optionally, with a third antidiabetic agent as defined hereinbefore and hereinafter to the patient.
  • According to another aspect there is provided the use of a pharmaceutical composition according to the present invention for the manufacture of a medicament for a therapeutic and preventive method as described hereinbefore and hereinafter.
  • Patients of a TCF7L2 risk genotype (also referred to herein as TCF7L2 risk genotype patients) within the meaning of this invention refer to those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially a SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146; in more particular, those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype; especially those who carry two T alleles of SNP rs7903146 of TCF7L2, i.e. the TT genotype, are at high-risk and are expected to be difficult to treat (e.g. to achieve adequate glycemic control).
  • The present invention provides a DPP-4 inhibitor (preferably linagliptin), pharmaceutical composition, combination or medicament according to the present invention for use in a therapeutic and/or preventive method as described hereinbefore and hereinafter (e.g. treating type 2 diabetes) in one or more of the following patient groups:
      • TCF7L2 high risk genotype patients carrying two T alleles of SNP rs7903146 of TCF7L2, i.e. TT genotype (where clinically meaningful response e.g. in glycemic control is provided),
      • TCF7L2 risk genotype patients carrying one T allele of SNP rs7903146 of TCF7L2, i.e. CT genotype (where clinically favorable response e.g. in glycemic control is provided),
      • TCF7L2 wild genotype patients carrying two CC alleles of SNP rs7903146 of TCF7L2, i.e. CC genotype (where clinically more favorable response e.g. in glycemic control is provided).
  • Within a particular aspect of the invention, the invention relates to a DPP-4 inhibitor, a pharmaceutical composition or combination of the present invention for a therapeutic and/or preventive method or use as described hereinbefore and hereinafter (e.g. treating type 2 diabetes), said method or use comprising
      • (i) identifying a patient being susceptible to said therapeutic and/or preventive method or use comprising testing whether the patient is of any TCF7L2 risk genotype, particularly whether the patient has one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially a SNP selected from rs7903146, rs12255372 and rs10885406, for example whether the patient carries at least one T allele of SNP rs7903146 of TCF7L2, e.g. whether the patient is of CT genotype (i.e. whether the patient carries one T allele of SNP rs7903146 of TCF7L2) or, particularly, whether the patient is of TT genotype (i.e. whether the patient carries two T alleles of SNP rs7903146 of TCF7L2), or testing whether the patient is of TCF7L2 wild genotype, particularly whether the patient carries two C alleles of SNP rs7903146 of TCF7L2 (i.e. whether the patient is of CC wild genotype), and
      • (ii) administering an effective amount of the DPP-4 inhibitor, pharmaceutical composition or combination to the patient identified in step (i).
  • Within another particular aspect of the invention, the invention relates to a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament of the present invention for a therapeutic and/or preventive method or use as described hereinbefore and hereinafter (e.g. treating type 2 diabetes) in TCF7L2 risk genotype patients, e.g. in those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially a SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146; in more particular, in those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype.
  • Within another particular aspect of the invention, the invention relates to a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament of the present invention for a therapeutic and/or preventive method or use as described hereinbefore and hereinafter (e.g. treating type 2 diabetes) in TCF7L2 wild genotype patients, e.g. in those patients who carry two C alleles of SNP rs7903146 of TCF7L2, i.e. the CC genotype.
  • In this context, a particular sub-population of the patients described hereinbefore and hereinafter (e.g. of the patients in need of a therapeutic or preventive method as described herein), refers to those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially at least one SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146, in more particular, those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype.
  • In more particular, those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype, especially who carry two T alleles of SNP rs7903146 of TCF7L2, i.e. the TT genotype, are strongly susceptible to increased TCF7L2 expression in pancreatic beta cells, impaired insulin secretion, incretine effects, enhanced rate of hepatic glucose production and/or diabetes. The T allele of rs7903146 TCF7L2 is associated with impaired insulinotropic action of incretin hormones, reduced 24 h profiles of plasma insulin and glucagon, and increased hepatic glucose production.
  • Another particular sub-population of the patients described hereinbefore and hereinafter (e.g. of the patients in need of a therapeutic or preventive method as described herein), refers to those patients who are of TCF7L2 wild genotype, particularly those who are of the TCF7L2 rs7903146 CC wild genotype.
  • According to one embodiment of this aspect of the invention, there is provided a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament according to the present invention for a therapeutic and/or preventive method or use as described hereinbefore and hereinafter (particularly for treating and/or preventing type 2 diabetes and/or obesity), in patients with reduced (glucose-stimulated) insulin secretion, increased hepatic gluconeogenesis and/or reduced insulinotropic effect or action of incretin hormones (e.g. GLP-1 and/or GIP), e.g. impaired incretin sensitivity, associated with a TCF7L2 risk genotype, particularly with such a TCF7L2 risk genotype as mentioned above.
  • According to another embodiment of this aspect of the invention, there is provided a method of determining patient's treatment response to a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament according to the present invention, said method comprising the step of determining whether the patient is of TCF7L2 risk genotype as described herein, e.g. testing whether the patient belongs to the particular subpopulation of TCF7L2 risk genotype carriers, or determining whether the patient is of TCF7L2 wild genotype, e.g. testing whether the patient carries the wild-type CC allele at rs7903146 in TCF7L2.
  • According to another embodiment of this aspect of the invention, there is provided a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament according to the present invention for use in a therapeutic and/or preventive method as described hereinbefore and hereinafter (particularly for treating and/or preventing type 2 diabetes and/or obesity) in a patient in need thereof, said method comprising testing whether the patient is of any TCF7L2 risk genotype as described herein.
  • According to another embodiment of this aspect of the invention, there is provided a DPP-4 inhibitor, a pharmaceutical composition, combination or medicament according to the present invention for use in a therapeutic and/or preventive method as described hereinbefore and hereinafter (particularly for treating and/or preventing type 2 diabetes and/or obesity) in a patient in need thereof, said method comprising testing whether the patient is of TCF7L2 wild genotype as described herein.
  • According to another aspect of the invention, the testing for TCF7L2 risk genotypes may be used for patient stratification, e.g. to enrich patient population in clinical trials to test the efficacy of the DPP-4 inhibitor.
  • According to another aspect of the invention, the method of determining the treatment susceptibility of an individual (e.g. comprising the testing for TCF7L2 risk or wild genotypes as described herein) may be used for determination whether the patient may respond to a lower level or may require a higher level of administered DPP-4 inhibitor, optionally in combination with one or more other active substances.
  • According to another aspect of the invention, determining the treatment susceptibility of an individual comprising the testing for TCF7L2 risk or wild genotypes as described herein may be used for determination whether the patient may be treated in monotherapy or in combination therapy with one or more additional antidiabetics according to this invention, e.g. to provide adequate glycemic control. For example, those patients with decreased likelihood of favorable response may require combination treatment, e.g. to achieve adequate glycemic control.
  • BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
  • FIG. 1 shows mean values and 95% confidence intervals for baseline HbA1c values for the whole patient population of the studies (full analysis set, FAS), for the subpopulation for which genetic analyses are performed (full analysis set for pharmacogenetic analyses, FASG), as well as for the subgroups defined by genotype (CC, CT, TT) of this subpopulation. The numbers of patients for placebo control and linagliptin treatment are given in braces.
  • FIG. 2 shows a statistical association between TCF7L2 SNP rs7903146 genotypes with a likelihood of a favorable response in CC/CT genotype carriers to the administration of a therapeutically-effective amount of linagliptin or linagliptin in combination with other oral antidiabetic therapy.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Definitions
  • The term “active ingredient” of a pharmaceutical composition or combination of the present invention means the DPP-4 inhibitor and/or, if present, the second antidiabetic agent and/or, if present, the third antidiabetic agent of 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 110 mg/dL (6.11 mmol/L) or 100 mg 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 110 mg/dL (6.11 mmol/L) or 100 mg 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 of 60 to 115 mg/dL (3.3 to 6.3 mmol/L), in particular below 70 mg/dL (3.89 mmol/L).
  • 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 (11.11 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 (7.78 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 (7.78 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. 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 needs 2-3 times as much insulin 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 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 type 2 diabetes patients with a secondary antidiabetic 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 1.001.5%. 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 ≥110 mg/dL or ≥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 “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 “DPP-4 inhibitor” in the scope of the present invention relates to a compound that exhibits inhibitory activity on the enzyme dipeptidyl peptidase IV (DPP-4). Such inhibitory activity can be characterised by the IC50 value. A DPP-4 inhibitor preferably exhibits an IC50 value below 10000 nM, preferably below 1000 nM. Certain DPP-4 inhibitors exhibit an IC50 value below 100 nM, or even ≤50 nM. IC50 values of DPP-4 inhibitors are usually above 0.01 nM, or even above 0.1 nM. DPP-IV inhibitors may include biologic and non-biologic, in particular non-peptidic compounds. The inhibitory effect on DPP-4 can be determined by methods known in the literature, in particular as described in the application WO 02/068420 or WO 2004/018468 (page 34), which are incorporated herein by reference in its entirety. The term “DPP-4 inhibitor” also comprises any pharmaceutically acceptable salts thereof, hydrates and solvates thereof, including the respective crystalline forms.
  • The terms “treatment” and “treating” or ananlogous terms comprise particularly 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”, “preventive treating” and “preventing” or ananlogous terms are used interchangeably and comprise a treatment of patients at risk to develop a condition mentioned hereinbefore, thus reducing said risk.
  • DETAILED DESCRIPTION
  • The aspects of the present invention, in particular the pharmaceutical compounds, compositions, combinations, methods and uses, refer to DPP-4 inhibitors, second and/or third antidiabetic agents as defined hereinbefore and hereinafter.
  • In a first embodiment (embodiment A), a DPP-4 inhibitor in the context of the present invention is any DPP-4 inhibitor of
  • Figure US20200046713A1-20200213-C00001
  • wherein R1 denotes ([1,5]naphthyridin-2-yl)methyl, (quinazolin-2-yl)methyl, (quinoxalin-6-yl)methyl, (4-methyl-quinazolin-2-yl)methyl, 2-cyano-benzyl, (3-cyano-quinolin-2-yl)methyl, (3-cyano-pyridin-2-yl)methyl, (4-methyl-pyrimidin-2-yl)methyl, or (4,6-dimethyl-pyrimidin-2-yl)methyl and R2 denotes 3-(R)-amino-piperidin-1-yl, (2-amino-2-methyl-propyl)-methylamino or (2-(S)-amino-propyl)-methylamino,
  • or its pharmaceutically acceptable salt.
  • In a second embodiment (embodiment B), a DPP-4 inhibitor in the context of the present invention is a DPP-4 inhibitor selected from the group consisting of
  • sitagliptin, vildagliptin, saxagliptin, alogliptin, gemigliptin,
  • (2S)-1-{([2-(5-Methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2-carbonitrile,
  • (2S)-1-{[1,1,-Dimethyl-3-(4-pyridin-3-yl-imidazol-1-yl)-propylamino]-acetyl}-pyrrolidine-2-carbonitrile,
  • (S)-1-((2S,3S,11bS)-2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one,
  • (3,3-Difluoropyrrolidin-1-yl)-((2S,4S)-4-(4-(pyrimidin-2-yl)piperazin-1-yl)pyrrolidin-2-yl)methanone,
  • (1((3S,4S)-4-amino-1-(4-(3,3-difluoropyrrolidin-1-yl)-1,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5-difluoropiperidin-2-one,
  • (2S,4S)-1-{2-[(3S,1R)-3-(1H-1,2,4-Triazol-1-ylmethyl)cyclopentylamino]-acetyl}-4-fluoropyrrolidine-2-carbonitrile,
  • (R)-2-[6-(3-Amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl]-4-fluoro-benzonitrile,
  • 5-{(S)-2-[2-((S)-2-Cyano-pyrrolidin-1-yl)-2-oxo-ethylamino]-propyl}-5-(1 H-tetrazol-5-yl)-10,11-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis-dimethylamide,
  • 3-{(2S,4S)-4-[4-(3-Methyl-1-phenyl-1H-pyrazol-5-yl)piperazin-1-yl]pyrrolidin-2-ylcarbonyl}thiazolidine,
  • [(2R)-1-{[(3R)-pyrrolidin-3-ylamino]acetyl}pyrrolidin-2-yl]boronic acid,
  • (2S,4S)-1-[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct-1-yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile,
  • 2-({6-[(3R)-3-amino-3-methylpiperidin-1-yl]-1,3-dimethyl-2,4-dioxo-1,2,3,4-tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile,
  • 6-[(3R)-3-amino-piperidin-1-yl]-5-(2-chloro-5-fluoro-benzyl)-1,3-dimethyl-1,5-dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione, and
  • (S)-2-methylpyrazolo[1,5-a]primidine-6-carboxylic acid {(2-[(2-cyanopyrrolidin-1-yl)-2-oxoethylamino]-2-methylpropyl}amide,
  • or its pharmaceutically acceptable salt.
  • Regarding the first embodiment (embodiment A), preferred DPP-4 inhibitors are any or all of the following compounds and their pharmaceutically acceptable salts:
      • 1-[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-(3-(R)-amino-piperidin-1-yl)-xanthine (compare WO 2004/018468, example 2(142)):
  • Figure US20200046713A1-20200213-C00002
      • 1-[([1,5]naphthyridin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-((R)-3-amino-piperidin-1-yl)-xanthine (compare WO 2004/018468, example 2(252)):
  • Figure US20200046713A1-20200213-C00003
      • 1-[(Quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-((R)-3-amino-piperidin-1-yl)-xanthine (compare WO 2004/018468, example 2(80)):
  • Figure US20200046713A1-20200213-C00004
      • 2-((R)-3-Amino-piperidin-1-yl)-3-(but-2-yinyl)-5-(4-methyl-quinazolin-2-ylmethyl)-3,5-dihydro-imidazo[4,5-d]pyridazin-4-one (compare WO 2004/050658, example 136):
  • Figure US20200046713A1-20200213-C00005
      • 1-[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyin-1-yl)-8-[(2-amino-2-methyl-propyl)-methylamino]-xanthine (compare WO 2006/029769, example 2(1)):
  • Figure US20200046713A1-20200213-C00006
      • 1-[(3-Cyano-quinolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-((R)-3-amino-piperidin-1-yl)-xanthine (compare WO 2005/085246, example 1(30)):
  • Figure US20200046713A1-20200213-C00007
      • 1-(2-Cyano-benzyl)-3-methyl-7-(2-butyn-1-yl)-8-((R)-3-amino-piperidin-1-yl)-xanthine (compare WO 2005/085246, example 1(39)):
  • Figure US20200046713A1-20200213-C00008
      • 1-[(4-Methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-[(S)-(2-amino-propyl)-methylamino]-xanthine (compare WO 2006/029769, example 2(4)):
  • Figure US20200046713A1-20200213-C00009
      • 1-[(3-Cyano-pyridin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-((R)-3-amino-piperidin-1-yl)-xanthine (compare WO 2005/085246, example 1(52)):
  • Figure US20200046713A1-20200213-C00010
      • 1-[(4-Methyl-pyrimidin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-((R)-3-amino-piperidin-1-yl)-xanthine (compare WO 2005/085246, example 1(81)):
  • Figure US20200046713A1-20200213-C00011
      • 1-[(4,6-Dimethyl-pyrimidin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-((R)-3-amino-piperidin-1-yl)-xanthine (compare WO 2005/085246, example 1(82)):
  • Figure US20200046713A1-20200213-C00012
      • 1-[(Quinoxalin-6-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-((R)-3-amino-piperidin-1-yl)-xanthine (compare WO 2005/085246, example 1(83)):
  • Figure US20200046713A1-20200213-C00013
  • A more preferred DPP-4 inhibitor among the abovementioned DPP-4 inhibitors of embodiment A of this invention is 1-[(4-methyl-quinazolin-2-yl)methyl]-3-methyl-7-(2-butyn-1-yl)-8-(3-(R)-amino-piperidin-1-yl)-xanthine, particularly the free base thereof (which is also known as linagliptin or BI 1356).
  • As further DPP-4 inhibitors the following compounds can be mentioned:
      • Sitagliptin (MK-0431) having the structural formula A below is (3R)-3-amino-1-[3-(trifluoromethyl)-5,6,7,8-tetrahydro-5H-[1,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl)butan-1-one, also named (2R)-4-oxo-4-[3-(trifluoromethyl)-5,6-dihydro[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]-1-(2,4,5-trifluorophenyl)butan-2-amine,
  • Figure US20200046713A1-20200213-C00014
  • In one embodiment, sitagliptin is in the form of its dihydrogenphosphate salt, i.e. sitagliptin phosphate. In a further embodiment, sitagliptin phosphate is in the form of a crystalline anhydrate or monohydrate. A class of this embodiment refers to sitagliptin phosphate monohydrate. Sitagliptin free base and pharmaceutically acceptable salts thereof are disclosed in U.S. Pat. No. 6,699,871 and in Example 7 of WO 03/004498. Crystalline sitagliptin phosphate monohydrate is disclosed in WO 2005/003135 and in WO 2007/050485.
  • For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
  • A tablet formulation for sitagliptin is commercially available under the trade name Januvia®. A tablet formulation for sitagliptin/metformin combination is commercially available under the trade name Janumet®.
      • Vildagliptin (LAF-237) having the structural formula B below is (2S)-{[(3-hydroxyadamantan-1-yl)amino]acetyl}pyrrolidine-2-carbonitrile, also named (S)-1-[(3-hydroxy-1-adamantyl)amino]acetyl-2-cyano-pyrrolidine,
  • Figure US20200046713A1-20200213-C00015
  • Vildagliptin is specifically disclosed in U.S. Pat. No. 6,166,063 and in Example 1 of WO 00/34241. Specific salts of vildagliptin are disclosed in WO 2007/019255. A crystalline form of vildagliptin as well as a vildagliptin tablet formulation are disclosed in WO 2006/078593.
  • Vildagliptin can be formulated as described in WO 00/34241 or in WO 2005/067976. A modified release vildagliptin formulation is described in WO 2006/135723.
  • For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
  • A tablet formulation for vildagliptin is commercially available under the trade name Galvus®.
  • A tablet formulation for vildagliptin/metformin combination is commercially available under the trade name Eucreas®.
      • Saxagliptin (BMS-477118) having the structural formula C below is (1S,3S,5S)-2-{(2S)-2-amino-2-(3-hydroxyadamantan-1-yl)acetyl}-2-azabicyclo[3.1.0]hexane-3-carbonitrile, also named (S)-3-hydroxyadamantylglycine-L-cis-4,5-methanoprolinenitrile,
  • Figure US20200046713A1-20200213-C00016
  • Saxagliptin is specifically disclosed in U.S. Pat. No. 6,395,767 and in Example 60 of WO 01/68603.
  • In one embodiment, saxagliptin is in the form of its HCl salt or its mono-benzoate salt as disclosed in WO 2004/052850. In a further embodiment, saxagliptin is in the form of the free base. In a yet further embodiment, saxagliptin is in the form of the monohydrate of the free base as disclosed in WO 2004/052850. Crystalline forms of the HCl salt and of the free base of saxagliptin are disclosed in WO 2008/131149. A process for preparing saxagliptin is also disclosed in WO 2005/106011 and WO 2005/115982. Saxagliptin can be formulated in a tablet as described in WO 2005/117841.
  • For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • Alogliptin (SYR-322) having the structural formula E below is 2-({6-[(3R)-3-aminopiperidin-1-yl]-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-yl}methyl)benzonitrile
  • Figure US20200046713A1-20200213-C00017
  • Alogliptin is specifically disclosed in US 2005/261271, EP 1586571 and in WO 2005/095381. In one embodiment, alogliptin is in the form of its benzoate salt, its hydrochloride salt or its tosylate salt each as disclosed in WO 2007/035629. A class of this embodiment refers to alogliptin benzoate. Polymorphs of alogliptin benzoate are disclosed in WO 2007/035372. A process for preparing alogliptin is disclosed in WO 2007/112368 and, specifically, in WO 2007/035629. Alogliptin (namely its benzoate salt) can be formulated in a tablet and administered as described in WO 2007/033266. A solid preparation of alogliptin/pioglitazone and its preparation and use is described in WO 2008/093882. A solid preparation of alogliptin/metformin and its preparation and use is described in WO 2009/011451.
  • For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • (2S)-1-{([2-(5-Methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2-carbonitrile or a pharmaceutically acceptable salt thereof, preferably the mesylate, or
  • (2S)-1-{([1,1,-Dimethyl-3-(4-pyridin-3-yl-imidazol-1-yl)-propylamino]-acetyl}-pyrrolidine-2-carbonitrile or a pharmaceutically acceptable salt thereof:
  • These compounds and methods for their preparation are disclosed in WO 03/037327.
  • The mesylate salt of the former compound as well as crystalline polymorphs thereof are disclosed in WO 2006/100181. The fumarate salt of the latter compound as well as crystalline polymorphs thereof are disclosed in WO 2007/071576. These compounds can be formulated in a pharmaceutical composition as described in WO 2007/017423.
  • For details, e.g. on a process to manufacture, to formulate or to use these compounds or salts thereof, reference is thus made to these documents.
      • (S)-1-((2S,3S,11bS)-2-Amino-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-3-yl)-4-fluoromethyl-pyrrolidin-2-one (also named carmegliptin) or a pharmaceutically acceptable salt thereof:
  • Figure US20200046713A1-20200213-C00018
  • This compound and methods for its preparation are disclosed in WO 2005/000848. A process for preparing this compound (specifically its dihydrochloride salt) is also disclosed in WO 2008/031749, WO 2008/031750 and WO 2008/055814. This compound can be formulated in a pharmaceutical composition as described in WO 2007/017423.
  • For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • (3,3-Difluoropyrrolidin-1-yl)-((2S,4S)-4-(4-(pyrimidin-2-yl)piperazin-1-yl)pyrrolidin-2-yl)methanone (also named gosogliptin) or a pharmaceutically acceptable salt thereof:
  • This compound and methods for its preparation are disclosed in WO 2005/116014 and U.S. Pat. No. 7,291,618.
  • For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • (1((3S,4S)-4-amino-1-(4-(3,3-difluoropyrrolidin-1-yl)-1,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5-difluoropiperidin-2-one or a pharmaceutically acceptable salt thereof:
  • Figure US20200046713A1-20200213-C00019
  • This compound and methods for its preparation are disclosed in WO 2007/148185 and US 20070299076. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • (2S,4S)-1-{2-[(35,1R)-3-(1H-1,2,4-Triazol-1-ylmethyl)cyclopentylamino]-acetyl}-4-fluoropyrrolidine-2-carbonitrile (also named melogliptin) or a pharmaceutically acceptable salt thereof:
  • Figure US20200046713A1-20200213-C00020
  • This compound and methods for its preparation are disclosed in WO 2006/040625 and WO 2008/001195. Specifically claimed salts include the methanesulfonate and p-toluenesulfonate. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • (R)-2-[6-(3-Amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethyl]-4-fluoro-benzonitrile or a pharmaceutically acceptable salt thereof:
  • Figure US20200046713A1-20200213-C00021
  • This compound and methods for its preparation and use are disclosed in WO 2005/095381, US 2007060530, WO 2007/033350, WO 2007/035629, WO 2007/074884, WO 2007/112368, WO 2008/033851, WO 2008/114800 and WO 2008/114807. Specifically claimed salts include the succinate (WO 2008/067465), benzoate, benzenesulfonate, p-toluenesulfonate, (R)-mandelate and hydrochloride. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • 5-{(S)-2-[2-((S)-2-Cyano-pyrrolidin-1-yl)-2-oxo-ethylamino]-propyl}-5-(1H-tetrazol-5-yl)-10,11-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis-dimethylamide or a pharmaceutically acceptable salt thereof:
  • Figure US20200046713A1-20200213-C00022
  • This compound and methods for its preparation are disclosed in WO 2006/116157 and US 2006/270701. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • 3-{(2S,4S)-4-[4-(3-Methyl-1-phenyl-1H-pyrazol-5-yl)piperazin-1-yl]pyrrolidin-2-ylcarbonyl}thiazolidine (also named teneligliptin) or a pharmaceutically acceptable salt thereof:
  • This compound and methods for its preparation are disclosed in WO 02/14271. Specific salts are disclosed in WO 2006/088129 and WO 2006/118127 (including hydrochloride, hydrobromide, inter alia). Combination therapy using this compound is described in WO 2006/129785. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • [(2R)-1-{[(3R)-pyrrolidin-3-ylamino]acetyl}pyrrolidin-2-yl]boronic acid (also named dutogliptin) or a pharmaceutically acceptable salt thereof:
  • This compound and methods for its preparation are disclosed in WO 2005/047297, WO 2008/109681 and WO 2009/009751. Specific salts are disclosed in WO 2008/027273 (including citrate, tartrate). A formulation of this compound is described in WO 2008/144730. A formulation of dutogliptin (as its tartrate salt) with metformin is described in WO 2009/091663. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • (2S,4S)-1-[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct-1-yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile or a pharmaceutically acceptable salt thereof:
  • This compound and methods for its preparation are disclosed in WO 2005/075421, US 2008/146818 and WO 2008/114857. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
      • 2-({6-[(3R)-3-amino-3-methylpiperidin-1-yl]-1,3-dimethyl-2,4-dioxo-1,2,3,4-tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile or a pharmaceutically acceptable salt thereof, or 6-[(3R)-3-amino-piperidin-1-yl]-5-(2-chloro-5-fluoro-benzyl)-1,3-dimethyl-1,5-dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione or a pharmaceutically acceptable salt thereof:
  • These compounds and methods for their preparation are disclosed in WO 2009/084497 and WO 2006/068163, respectively. Combination therapy using the latter of these two compounds is described in WO 2009/128360. For details, e.g. on a process to manufacture, to formulate or to use these compounds or salts thereof, reference is thus made to these documents.
      • (S)-2-methylpyrazolo[1,5-a]primidine-6-carboxylic acid {(2-[(2-cyanopyrrolidin-1-yl)-2-oxoethylamino]-2-methylpropyl}amide (also named anagliptin) or a pharmaceutically acceptable salt:
  • This compound and methods for its preparation are disclosed in WO 2004/067509. Combination therapy using this compound is described in WO 2009/139362. For details, e.g. on a process to manufacture, to formulate or to use this compound or a salt thereof, reference is thus made to these documents.
  • Preferably the DPP-4 inhibitor is selected from the group G2 consisting of linagliptin, sitagliptin, vildagliptin, alogliptin, saxagliptin, carmegliptin, gosogliptin, teneligliptin, melogliptin and dutogliptin, or a pharmaceutically acceptable salt of one of the hereinmentioned DPP-4 inhibitors, or a prodrug thereof.
  • More preferably the DPP-4 inhibitor is selected from the group G2 consisting of linagliptin, sitagliptin, vildagliptin, alogliptin, saxagliptin, teneligliptin and dutogliptin, or a pharmaceutically acceptable salt of one of the hereinmentioned DPP-4 inhibitors, or a prodrug thereof.
  • A particularly preferred DPP-4 inhibitor within the present invention is linagliptin. The term “linagliptin” as employed herein refers to linagliptin and pharmaceutically acceptable salts thereof, including hydrates and solvates thereof, and crystalline forms thereof. Crystalline forms are described in WO 2007/128721. Methods for the manufacture of linagliptin are described in the patent applications WO 2004/018468 and WO 2006/048427 for example. Linagliptin is distinguished from structurally comparable DPP-4 inhibitors, as it combines exceptional potency and a long-lasting effect with favourable pharmacological properties, receptor selectivity and a favourable side-effect profile or bring about unexpected therapeutic advantages or improvements in monotherapy and/or when used in combination with a second and, optionally, a third antidiabetic agent according to this invention.
  • For avoidance of any doubt, the disclosure of each of the foregoing documents cited above in connection with the specified DPP-4 inhibitors is specifically incorporated herein by reference in its entirety.
  • In one aspect of the present invention, the pharmaceutical compositions, methods and uses according to this invention relate to those compositions which comprise the DPP-4 inhibitor as sole active ingredient (i.e. the second and third antidiabetic agent are both absent) and/or, respectively, to monotherapy using the DPP-4 inhibitor alone.
  • In another aspect of the present invention, the pharmaceutical compositions, combinations, methods and uses according to this invention relate to those compositions or combinations which comprise the DPP-4 inhibitor and the second antidiabetic agent as sole active ingredients (i.e. the third antidiabetic agent is absent) and/or, respectively, to dual combination therapy using the DPP-4 inhibitor and the second antidiabetic agent.
  • In another aspect of the present invention, the pharmaceutical compositions, combinations, methods and uses according to this invention relate to those compositions or combinations which comprise the DPP-4 inhibitor, the second and the third antidiabetic agent and/or, respectively, to triple combination therapy using the DPP-4 inhibitor, the second and the third antidiabetic agent.
  • Further, a DPP-4 inhibitor according to this invention may be further characterized in that said DPP-4 inhibitor does not significantly impair glomerular and/or tubular function of a type 2 diabetes patient with chronic renal insufficiency (e.g. mild, moderate or severe renal impairment or end stage renal disease), and/or
  • said DPP-4 inhibitor does not require to be dose-adjusted in a type 2 diabetes patient with impaired renal function (e.g. mild, moderate or severe renal impairment or end stage renal disease).
  • The second antidiabetic agent and, if present, the third antidiabetic agent is selected from the group G3 consisting of biguanides, thiazolidindiones, sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues, and insulin or insulin analogues, or a pharmaceutically acceptable salt thereof. In the following preferred embodiments regarding the second and/or the third antidiabetic agent are described.
  • The group G3 comprises biguanides. Examples of biguanides are metformin, phenformin and buformin. A preferred biguanide is metformin. A DPP-4 inhibitor in combination with a biguanide, in particular metformin, can provide more efficacious glycemic control and/or may act together with the biguanide, for example to reduce weight, that has e.g. overall beneficial effects on the metabolic syndrome which is commonly associated with type 2 diabetes mellitus.
  • The term “metformin” as employed herein refers to metformin or a pharmaceutically acceptable salt thereof such as the hydrochloride salt, the metformin (2:1) fumarate salt, and the metformin (2:1) succinate salt, the hydrobromide salt, the p-chlorophenoxy acetate or the embonate, and other known metformin salts of mono and dibasic carboxylic acids. It is preferred that the metformin employed herein is the metformin hydrochloride salt.
  • The group G3 comprises thiazolidindiones. Examples of thiazolidindiones (TZD) are pioglitazone and rosiglitazone. TZD therapy is associated with weight gain and fat redistribution. In addition, TZD cause fluid retention and are not indicated in patients with congestive heart failure. Long term treatment with TZD are further associated with an increased risk of bone fractures. A DPP-4 inhibitor in combination with a thiazolidindione, in particular pioglitazone, can provide more efficacious glycemic control and/or can minimize side effects of the treatment with TZD.
  • The term “pioglitazone” as employed herein refers to pioglitazone, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salt thereof such as the hydrochloride salt.
  • The term “rosiglitazone” as employed herein refers to rosiglitazone, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salt thereof such as the maleate salt.
  • The group G3 comprises sulfonylureas. Examples of sulfonylureas are glibenclamide, tolbutamide, glimepiride, glipizide, gliquidone, glibornuride, glyburide, glisoxepide and gliclazide. Preferred sulfonylureas are tolbutamide, gliquidone, glibenclamide and glimepiride, in particular glibenclamide and glimepiride. As the efficacy of sulfonylureas wears off over the course of treatment, a combination of a DPP-4 inhibitor with a sulfonylurea may offer additional benefit to the patient in terms of better glycemic control. Also, treatment with sulfonylureas is normally associated with gradual weight gain over the course of treatment and a DPP-4 inhibitor may minimize this side effect of the treatment with an sulfonylurea and/or improve the metabolic syndrome. Also, a DPP-4 inhibitor in combination with a sulfonylurea may minimize hypoglycemia which is another undesirable side effect of sulfonylureas. This combination may also allow a reduction in the dose of sulfonylureas, which may also translate into less hypoglycemia.
  • Each term of the group “glibenclamide”, “glimepiride”, “gliquidone”, “glibornuride”, “gliclazide”, “glisoxepide”, “tolbutamide” and “glipizide” as employed herein refers to the respective active drug or a pharmaceutically acceptable salt thereof.
  • The group G3 comprises glinides. Examples of glinides are nateglinide, repaglinide and mitiglinide. As their efficacy wears off over the course of treatment, a combination of a DPP-4 inhibitor with a meglitinide may offer additional benefit to the patient in terms of better glycemic control. Also, treatment with meglitinides is normally associated with gradual weight gain over the course of treatment and a DPP-4 inhibitor may minimize this side effect of the treatment with an meglitinide and/or improve the metabolic syndrome. Also, a DPP-4 inhibitor in combination with a meglitinide may minimize hypoglycemia which is another undesirable side effect of meglitinides. This combination may also allow a reduction in the dose of meglitinides, which may also translate into less hypoglycemia.
  • The term “nateglinide” as employed herein refers to nateglinide, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salts and esters thereof.
  • The term “repaglinide” as employed herein refers to repaglinide, including its enantiomers, mixtures thereof and its racemate, or a pharmaceutically acceptable salts and esters thereof.
  • The group G3 comprises inhibitors of alpha-glucosidase. Examples of inhibitors of alpha-glucosidase are acarbose, voglibose and miglitol. Additional benefits from the combination of a DPP-4 inhibitor and an alpha-glucosidase inhibitor may relate to more efficacious glycemic control, e.g. at lower doses of the individual drugs, and/or reducement of undesirable gastrointestinal side effects of alpha-glucosidase inhibitors.
  • Each term of the group “acarbose”, “voglibose” and “miglitol” as employed herein refers to the respective active drug or a pharmaceutically acceptable salt thereof.
  • The group G3 comprises inhibitors of GLP-1 analogues. Examples of GLP-1 analogues are exenatide, liraglutide, taspoglutide, semaglutide, albiglutide, and lixisenatide. The combination of a DPP-4 inhibitor and a GLP-1 analogue may achieve a superior glycemic control, e.g. at lower doses of the individual drugs. In addition, e.g. the body weight reducing capability of the GLP-1 analogue may be positively act together with the properties of the DPP-4 inhibitor. On the other hand, a reduction of side effects (e.g. nausea, gastrointestinal side effects like vomiting) may be obtained, e.g. when a reduced dose of the GLP-1 analogue is applied in the combination with a DPP-4 inhibitor.
  • Each term of the group “exenatide”, “liraglutide”, “taspoglutide”, “semaglutide”, “albiglutide”, and “lixisenatide” as employed herein refers to the respective active drug or a pharmaceutically acceptable salt thereof.
  • In an embodiment (embodiment E1) the pharmaceutical compositions, combinations, methods and uses according to this invention relate to those combinations wherein the DPP-4 inhibitor and the second antidiabetic agent are preferably selected according to the entries in the Table 1.
  • TABLE 1
    DPP-4 Inhibitor Second Antidiabetic Agent
    selected from embodiment B selected from the group G3
    selected from embodiment B Metformin
    selected from embodiment B Pioglitazone
    selected from embodiment B Rosiglitazone
    selected from embodiment B Glibenclamide
    selected from embodiment B Glimepiride
    selected from embodiment B Gliquidone
    selected from embodiment B Nateglinide
    selected from embodiment B Repaglinide
    selected from embodiment B Acarbose
    selected from embodiment B Voglibose
    selected from embodiment B Miglitol
    selected from embodiment B Exenatide
    selected from embodiment B Liraglutide
    selected from embodiment B Taspoglutide
    selected from embodiment B Semaglutide
    selected from embodiment B Albiglutide
    selected from embodiment B Lixisenatide
    Linagliptin selected from the group G3
    Linagliptin Metformin
    Linagliptin Pioglitazone
    Linagliptin Rosiglitazone
    Linagliptin Glibenclamide
    Linagliptin Glimepiride
    Linagliptin Gliquidone
    Linagliptin Nateglinide
    Linagliptin Repaglinide
    Linagliptin Acarbose
    Linagliptin Voglibose
    Linagliptin Miglitol
    Linagliptin Exenatide
    Linagliptin Liraglutide
    Linagliptin Taspoglutide
    Linagliptin Semaglutide
    Linagliptin Albiglutide
    Linagliptin Lixisenatide
    Sitagliptin selected from the group G3
    Sitagliptin Metformin
    Sitagliptin Pioglitazone
    Sitagliptin Rosiglitazone
    Sitagliptin Glibenclamide
    Sitagliptin Glimepiride
    Sitagliptin Gliquidone
    Sitagliptin Nateglinide
    Sitagliptin Repaglinide
    Sitagliptin Acarbose
    Sitagliptin Voglibose
    Sitagliptin Miglitol
    Sitagliptin Exenatide
    Sitagliptin Liraglutide
    Sitagliptin Taspoglutide
    Sitagliptin Semaglutide
    Sitagliptin Albiglutide
    Sitagliptin Lixisenatide
    Vildagliptin selected from the group G3
    Vildagliptin Metformin
    Vildagliptin Pioglitazone
    Vildagliptin Rosiglitazone
    Vildagliptin Glibenclamide
    Vildagliptin Glimepiride
    Vildagliptin Gliquidone
    Vildagliptin Nateglinide
    Vildagliptin Repaglinide
    Vildagliptin Acarbose
    Vildagliptin Voglibose
    Vildagliptin Miglitol
    Vildagliptin Exenatide
    Vildagliptin Liraglutide
    Vildagliptin Taspoglutide
    Vildagliptin Semaglutide
    Vildagliptin Albiglutide
    Vildagliptin Lixisenatide
    Alogliptin selected from the group G3
    Alogliptin Metformin
    Alogliptin Pioglitazone
    Alogliptin Rosiglitazone
    Alogliptin Glibenclamide
    Alogliptin Glimepiride
    Alogliptin Gliquidone
    Alogliptin Nateglinide
    Alogliptin Repaglinide
    Alogliptin Acarbose
    Alogliptin Voglibose
    Alogliptin Miglitol
    Alogliptin Exenatide
    Alogliptin Liraglutide
    Alogliptin Taspoglutide
    Alogliptin Semaglutide
    Alogliptin Albiglutide
    Alogliptin Lixisenatide
    Saxagliptin selected from the group G3
    Saxagliptin Metformin
    Saxagliptin Pioglitazone
    Saxagliptin Rosiglitazone
    Saxagliptin Glibenclamide
    Saxagliptin Glimepiride
    Saxagliptin Gliquidone
    Saxagliptin Nateglinide
    Saxagliptin Repaglinide
    Saxagliptin Acarbose
    Saxagliptin Voglibose
    Saxagliptin Miglitol
    Saxagliptin Exenatide
    Saxagliptin Liraglutide
    Saxagliptin Taspoglutide
    Saxagliptin Semaglutide
    Saxagliptin Albiglutide
    Saxagliptin Lixisenatide
    Carmegliptin selected from the group G3
    Carmegliptin Metformin
    Carmegliptin Pioglitazone
    Carmegliptin Rosiglitazone
    Carmegliptin Glibenclamide
    Carmegliptin Glimepiride
    Carmegliptin Gliquidone
    Carmegliptin Nateglinide
    Carmegliptin Repaglinide
    Carmegliptin Acarbose
    Carmegliptin Voglibose
    Carmegliptin Miglitol
    Carmegliptin Exenatide
    Carmegliptin Liraglutide
    Carmegliptin Taspoglutide
    Carmegliptin Semaglutide
    Carmegliptin Albiglutide
    Carmegliptin Lixisenatide
    Melogliptin selected from the group G3
    Melogliptin Metformin
    Melogliptin Pioglitazone
    Melogliptin Rosiglitazone
    Melogliptin Glibenclamide
    Melogliptin Glimepiride
    Melogliptin Gliquidone
    Melogliptin Nateglinide
    Melogliptin Repaglinide
    Melogliptin Acarbose
    Melogliptin Voglibose
    Melogliptin Miglitol
    Melogliptin Exenatide
    Melogliptin Liraglutide
    Melogliptin Taspoglutide
    Melogliptin Semaglutide
    Melogliptin Albiglutide
    Melogliptin Lixisenatide
    Dutogliptin selected from the group G3
    Dutogliptin Metformin
    Dutogliptin Pioglitazone
    Dutogliptin Rosiglitazone
    Dutogliptin Glibenclamide
    Dutogliptin Glimepiride
    Dutogliptin Gliquidone
    Dutogliptin Nateglinide
    Dutogliptin Repaglinide
    Dutogliptin Acarbose
    Dutogliptin Voglibose
    Dutogliptin Miglitol
    Dutogliptin Exenatide
    Dutogliptin Liraglutide
    Dutogliptin Taspoglutide
    Dutogliptin Semaglutide
    Dutogliptin Albiglutide
    Dutogliptin Lixisenatide
    Gosogliptin selected from the group G3
    Gosogliptin Metformin
    Gosogliptin Pioglitazone
    Gosogliptin Rosiglitazone
    Gosogliptin Glibenclamide
    Gosogliptin Glimepiride
    Gosogliptin Gliquidone
    Gosogliptin Nateglinide
    Gosogliptin Repaglinide
    Gosogliptin Acarbose
    Gosogliptin Voglibose
    Gosogliptin Miglitol
    Gosogliptin Exenatide
    Gosogliptin Liraglutide
    Gosogliptin Taspoglutide
    Gosogliptin Semaglutide
    Gosogliptin Albiglutide
    Gosogliptin Lixisenatide
    Teneligliptin selected from the group G3
    Teneligliptin Metformin
    Teneligliptin Pioglitazone
    Teneligliptin Rosiglitazone
    Teneligliptin Glibenclamide
    Teneligliptin Glimepiride
    Teneligliptin Gliquidone
    Teneligliptin Nateglinide
    Teneligliptin Repaglinide
    Teneligliptin Acarbose
    Teneligliptin Voglibose
    Teneligliptin Miglitol
    Teneligliptin Exenatide
    Teneligliptin Liraglutide
    Teneligliptin Taspoglutide
    Teneligliptin Semaglutide
    Teneligliptin Albiglutide
    Teneligliptin Lixisenatide
  • In a particular embodiment (embodiment E2) the pharmaceutical compositions, combinations, methods and uses according to this invention relate to those combinations wherein the DPP-4 inhibitor is linagliptin. According to embodiment E2 the second antidiabetic agent is preferably selected according to the entries in the Table 2.
  • TABLE 2
    Embodiment Second Antidiabetic Agent
    E2.1 selected from the group G3
    E2.2 Metformin
    E2.3 Pioglitazone
    E2.4 Rosiglitazone
    E2.5 Glibenclamide
    E2.6 Glimepiride
    E2.7 Gliquidone
    E2.8 Nateglinide
    E2.9 Repaglinide
    E2.10 Acarbose
    E2.11 Voglibose
    E2.12 Miglitol
    E2.13 Exenatide
    E2.14 Liraglutide
    E2.15 Taspoglutide
    E2.16 Semaglutide
    E2.17 Albiglutide
    E2.18 Lixisenatide
    E2.19 insulin or insulin analogue
    E2.20 GLP-1 or GLP-1 analogue
  • The combination of a DPP-4 inhibitor and a second and, optionally, a third antidiabetic agent according to this invention can be found to improve the glycemic control, in particular in patients as described herein, compared with a monotherapy using either a DPP-4 inhibitor or the second or third antidiabetic agent alone, for example with a monotherapy of metformin, or with a dual therapy using the second and third antidiabetic agent. Further, the triple combination of a DPP-4 inhibitor and a second and a third antidiabetic agent according to this invention can be found to improve the glycemic control, in particular in patients as described herein, compared with a combination therapy using a DPP-4 inhibitor and either the second or third antidiabetic agent, or using the second and the third antidiabetic agent. The improved glycemic control is determined as an increased lowering of blood glucose and an increased reduction of HbA1c. With monotherapy in a patient, in particular in patients as described herein, the glycemic control may not be further improved significantly by an administration of the drug above a certain highest dose. In addition, a long term treatment using a highest dose may be unwanted in view of potential side effects. Therefore, a satisfying glycemic control may not be achievable in all patients via a monotherapy using either the DPP-4 inhibitor or the second or the third antidiabetic agent alone. In the case that monotherapy do not yield in full glycemic control, dual therapy may become necessary. Even with combination therapy using two agents selected from the DPP-4 inhibitors and second and third antidiabetic agents may not yield in a full glycemic control in all patients and/or over a long time. In the case that dual therapy do not yield in full glycemic control, triple therapy may become necessary. In such patients with inadequate glycemic control a progression of the diabetes mellitus may continue and complications associated with diabetes mellitus may occur, such as macrovascular complications. The pharmaceutical composition or combination 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, e.g. in the case of dual or triple combination therapy compared with a monotherapy using one of or, respectively, a dual therapy using two of the combination partners.
  • In addition, the combination of a DPP-4 inhibitor and the second and, optionally, the third therapeutic agent according to this invention can be found to allow a reduction in the dose of either the DPP-4 inhibitor or the second or third antidiabetic agent or even of two or three of the active ingredients. A dose reduction is beneficial for patients which otherwise would potentially suffer from side effects in a therapy using a higher dose of one or more of the active ingredients, in particular with regard to side effect caused by the second and/or third antidiabetic agent. Therefore, the pharmaceutical combination as well as the methods according to the present invention, may show less side effects, thereby making the therapy more tolerable and improving the patients compliance with the treatment.
  • A DPP-4 inhibitor according to the present invention is able—via the increases in active GLP-1 levels—to reduce the glucagon secretion in a patient. This will therefore limit the hepatic glucose production. Furthermore, the elevated active GLP-1 levels produced by the DPP-4 inhibitor will have beneficial effects on beta-cell regeneration and neogenesis. All these features of DPP-4 inhibitors may render a pharmaceutical composition or combination or method of this invention quite useful and therapeutically relevant.
  • When this invention refers to patients requiring treatment or prevention, it relates primarily to treatment and prevention in humans, but the pharmaceutical composition may also be used accordingly in veterinary medicine in mammals. In the scope of this invention adult patients are preferably humans of the age of 18 years or older. Also in the scope of this invention, patients are adolescent humans, i.e. humans of age 10 to less than 18 years, preferably of age 13 to less than 18 years.
  • In one embodiment, patients in need of treatment or prevention as described herein can be identified by determining whether they have variation(s) (e.g. polymorphisms) in one or more genes associated with metabolic diseases and/or whether they have variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, in particular whether they are of TCF7L2 risk genotype as described herein.
  • In another embodiment, patients in need of treatment or prevention as described herein can be identified by determining whether they are of respective wild-type genotype, in particular whether they are of TCF7L2 wild genotype as described herein.
  • A particular sub-population of the patients in need of treatment or prevention as described herein, refers to those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially a SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146, in more particular, those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype.
  • Another particular sub-population of the patients in need of treatment or prevention as described herein, refers to those patients who carry TCF7L2 rs7903146 CC wild genotype.
  • Thus, in an aspect of this invention, a treatment or prophylaxis according to this invention is suitable in those patients in need of such treatment or prophylaxis who are diagnosed of having variation(s) (e.g. polymorphisms) in one or more genes associated with metabolic diseases and/or variation(s) (e.g. SNPs) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, in particular of TCF7L2 risk genotype as described herein.
  • In another aspect of this invention, a treatment or prophylaxis according to this invention is particular suitable in those patients in need of such treatment or prophylaxis who are diagnosed of having TCF7L2 wild genotype as described herein.
  • In an sub-aspect of this invention, a treatment or prophylaxis according to this invention is suitable in those patients in need of such treatment or prophylaxis who are diagnosed of having one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, e.g. at least one SNP selected from rs7903146, rs12255372 and rs10885406, for example rs7903146, in particular, carrying at least one T allele of rs7903146, (i.e. of CT or TT genotype), among them, in more particular, those carrying one T allele of rs7903146 (i.e. of CT risk genotype) or, in less particular, those carrying two T alleles of rs7903146 (i.e. of TT high risk genotype).
  • In another sub-aspect of this invention, a treatment or prophylaxis according to this invention is particular favorable in those patients in need of such treatment or prophylaxis who are diagnosed of carrying wild-type two C alleles of rs7903146 in TCF7L2 (i.e. of CC genotype).
  • In an embodiment of this invention, a treatment or prophylaxis according to this invention is 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. Any weight increasing effect in the therapy, for example due to the administration of the second and/or third antidiabetic agent, may be attenuated or even avoided thereby.
  • In a further embodiment of this invention, the pharmaceutical composition or combination of this invention 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 or combination according to this invention, a reduction of HbA1c equal to or greater than preferably 1.0%, more preferably equal to or greater than 2.0%, even more preferably equal to or greater than 3.0% can be achieved and the reduction is particularly in the range from 1.0% to 3.0%.
  • Furthermore, the method and/or use according to this invention is 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 or greater than 100 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 or combination 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 or combination of 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 or combination of 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 an 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 DPP-4 inhibitor and, optionally, a second and, optionally, a third antidiabetic agent as defined hereinbefore and hereinafter are administered, for example in combination, to the patient.
  • According to another embodiment of the present invention, there is provided a method for improving gycemic control in patients, in particular in adult patients, with type 2 diabetes mellitus as an adjunct to diet and exercise.
  • Moreover, in a particular embodiment of this invention, a therapeutic or preventive method and/or use according to this invention is suitable in those patients who have variation(s) (e.g. polymorphisms) in one or more genes associated with metabolic diseases and/or who have variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R.
  • In this context, a sub-population of the patients described hereinbefore and hereinafter refers to TCF7L2 risk genotype patients, such as e.g. to those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially at least one SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146. In more particular, those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype, especially who carry two T alleles of SNP rs7903146 of TCF7L2, i.e. the TT genotype, are strongly susceptible to increased TCF7L2 expression in pancreatic beta cells, impaired insulin secretion, incretine effects, enhanced rate of hepatic glucose production and/or diabetes. The T allele of rs7903146 TCF7L2 is associated with impaired insulinotropic action of incretin hormones, reduced 24 h profiles of plasma insulin and glucagon, and increased hepatic glucose production.
  • Therefore, the present invention also includes the compounds, pharmaceutical compositions or combinations according to this invention for use in the treatment and/or prevention of those diseases, disorders or conditions mentioned herein in those patients who have one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially at least one SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146; in more particular, in those patients who carry at least one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype or TT genotype, particularly in those patients who carry one T allele of SNP rs7903146 of TCF7L2, i.e. the CT genotype, or who carry two T alleles of SNP rs7903146 of TCF7L2, i.e. the TT genotype.
  • TCF7L2 risk genotype patients as described herein include, without being limited, patients of Caucasian, North European, East Asian, Indian and/or African descent.
  • The present invention further includes a therapeutic and/or preventive method or use according to this invention for application in a patient in need thereof, said method or use comprising the step of determining whether the patient has variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, particularly whether the patient is of a TCF7L2 risk genotype as described herein.
  • The determination or diagnosis whether the patient has variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, particularly whether the patient is of a TCF7L2 risk genotype as described herein, or whether the patient is of wild genotype, particularly whether the patient is of TCF7L2 wild genotype as described herein, may be used for determining the likelihood (e.g., increased, decreased, or no likelihood) of a favourable therapeutic and/or preventive response of the patient to the treatment with a DPP-4 inhibitor (or with a combination of a DPP-4 inhibitor with the second and/or third antidiabetic agent as defined herein) in a therapeutic and/or preventive method or use as described hereinabove or hereinbelow (e.g. in treating diabetes or in improving glycemic control), and thus for identifying a subject being susceptible to such treatment.
  • Thus, further on, in another embodiment of this invention, there is provided a method of determining the probability of likelihood (e.g., increased, decreased, or no likelihood) of a favourable response to the administration of a pharmaceutically acceptable amount of a DPP-4 inhibitor (or of a combination of a DPP-4 inhibitor with the second and/or third antidiabetic agent as described herein) in a subject (particularly diabetes patient), said method comprising the step of determining whether the subject has variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, particularly whether the subject is of a TCF7L2 risk genotype as described herein, or determining whether the subject is of TCF7L2 wild genotype, particularly testing whether the subject is of the TCF7L2 rs7903146 CC wild genotype.
  • According to another particular embodiment this invention, the present invention provides a DPP-4 inhibitor, a pharmaceutical composition or combination according to the present invention for use in a therapeutic or preventive method as described hereinbefore or hereinafter (particularly for treating or preventing type 2 diabetes and/or obesity), said method comprising
  • (i) identifying a subject being susceptible to the therapeutic or preventive method, said identifying comprising testing whether the subject has variation(s) (e.g. polymorphisms) in one or more of the genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, in particular whether the subject is of any TCF7L2 risk genotype as described herein, in more particular whether he/she has one or more single nucleotide polymorphisms (SNPs) in the gene coding for TCF7L2, especially at least one SNP selected from rs7903146, rs12255372 and rs10885406, especially rs7903146, for example whether the subject carries at least one T allele of SNP rs7903146 of TCF7L2, e.g. whether the subject is of CT genotype (i.e. whether the patient carries one T allele of SNP rs7903146 of TCF7L2) or whether the subject is of TT genotype (i.e. whether the patient carries two T alleles of SNP rs7903146 of TCF7L2), or testing whether the subject is of TCF7L2 wild genotype, in particular whether the subject is of the TCF7L2 rs7903146 CC wild genotype; and thus determining the probability of likelihood of a favourable response (e.g. favorable change in HbA1c) resulting from therapeutic or preventive treatment of the subject with the DPP-4 inhibitor, pharmaceutical composition or combination; and
  • (ii) administering an effective amount of the DPP-4 inhibitor, pharmaceutical composition or combination to the subject, where said subject is determined to have a high probability of likelihood of a favorable response (e.g. favorable change in HbA1c) resulting from therapeutic or preventive treatment with the DPP-4 inhibitor, pharmaceutical composition or combination.
  • The present invention further provides a therapeutic and/or preventive method or use of this invention for application in a patient in need thereof, said method or use comprising the steps of
      • obtaining and assaying a nucleic acid sample from an individual with type 2 diabetes mellitus,
      • determining the efficacy and/or, optionally, the probability of the likelihood of a favorable response (e.g. in providing glycemic control, such as favorable change in HbA1c) to a treatment with a DPP-4 inhibitor, preferably linagliptin, or the DPP-4 inhibitor in combination with one or more other active substances (e.g. antidiabetics), comprising detecting either TT or CT or CC allele genotype at rs7903146 of TCF7L2 gene in patient's sample,
        • wherein the presence of the TT, CT or CC genotype is indicative of the efficacy to the treatment, and/or, optionally,
        • wherein the presence of the TT genotype is indicative of a decreased likelihood of favorable response and/or presence of the CC genotype is indicative of an increased likelihood of favorable response to the treatment, and
      • administering a therapeutically effective amount of the DPP-4 inhibitor, preferably linagliptin, or the DPP-4 inhibitor in combination with one or more other active substances (e.g. antidiabetics) to the individual.
  • It can be further found that by using a pharmaceutical composition or combination according to this invention, an improvement of the glycemic control can be achieved even in those patients who have insufficient glycemic control in particular despite treatment with the second or third antidiabetic agent or a combination of the second with the third antidiabetic agent, for example despite maximal tolerated dose of oral monotherapy with metformin, a thiazolidinedione (e.g. pioglitazone) or a sulfonylurea, or a combination of metformin with a thiazolidinedione (e.g. pioglitazone), of metformin with a sulfonylurea, or of a thiazolidinedione (e.g. pioglitazone) with a sulfonylurea.
  • It can be also found that by using a combination according to this invention, an improvement of the glycemic control can be achieved even in those patients who have insufficient glycemic control in particular despite treatment with a DPP-4 inhibitor or a combination of a DPP-4 inhibitor with the second or third antidiabetic agent, for example despite maximal tolerated dose of oral monotherapy with a DPP-4 inhibitor or a dual combination of a DPP-4 inhibitor with the second or third antidiabetic agent.
  • A maximal tolerated dose with regard to metformin is for example 2000 mg per day, 1500 mg per day (for example in asian countries) or 850 mg three times a day or any equivalent thereof.
  • Therefore, the method and/or use according to this invention is 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 monotherapy with metformin, a thiazolidinedione (e.g. pioglitazone), a sulfonylurea, GLP-1 or GLP-1 analogue, or insulin or insulin analogue, in particular despite oral monotherapy at a maximal tolerated dose of metformin, a thiazolidinedione (e.g. pioglitazone) or a sulfonylurea;
      • (c) insufficient glycemic control despite combination therapy with two agents selected from the group consisting of metformin, a thiazolidinedione (e.g. pioglitazone), a sulfonylurea, GLP-1 or GLP-1 analogue, and insulin or insulin analogue, for example despite combination therapy with a dual combination selected from metformin/pioglitazone, metformin/sulphonylurea, metformin/insulin, sulphonylurea/pioglitazone, sulphonylurea/insulin and pioglitazone/insulin;
  • The dual or triple combination method and/or use according to this invention is further applicable in those patients who show the following conditions (e) or (f), respectively:
      • (d) insufficient glycemic control despite oral monotherapy with the DPP-4 inhibitor, in particular despite oral monotherapy at a maximal tolerated dose of the DPP-4 inhibitor;
      • (e) insufficient glycemic control despite (oral) combination therapy with the DPP-4 inhibitor and the second or third antidiabetic agent, in particular despite oral dual therapy at a maximal tolerated dose of at least one of the combination partners.
  • In an embodiment of this invention, a pharmaceutical composition or combination is 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 or combination 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 or ≥100 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.
  • Furthermore, the pharmaceutical composition and the methods according to this invention are particularly suitable in the treatment of patients after organ transplantation, in particular those patients who are diagnosed having one or more of the following conditions
      • (a) a higher age, in particular above 50 years,
      • (b) male gender;
      • (c) overweight, obesity (including class I, II and/or III obesity), visceral obesity and/or abdominal obesity,
      • (d) pre-transplant diabetes,
      • (e) immunosuppression therapy.
  • A pharmaceutical composition or combination according to this invention, in particular due to the DPP-4 inhibitor therein, exhibits a good safety profile. Therefore, a treatment or prophylaxis according to this invention is possible in those patients for which the mono-therapy with another antidiabetic drug, such as for example metformin, 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 or combination 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 or combination 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 particular 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.
  • The effects mentioned above are observed both, when the DPP-4 inhibitor and the second and, optionally, third antidiabetic agent are administered together, for example simultaneously in one single or two or three separate formulations, and/or when they are administered in alternation, for example successively in two or three separate formulations.
  • Within this invention it is to be understood that combinations or combined uses envisage the separate, sequential, simultaneous, concurrent, chronologically staggered or alternating administration of the components. It will be appreciated that the DPP-4 inhibitor and the other active substance(s) can be administered in a single dosage form or each in separate dosage forms.
  • In this context, “combination” or “combined” within the meaning of this invention also includes, without being limited, fixed and non-fixed forms and uses.
  • It will be appreciated that the amount of the pharmaceutical composition according to this invention to be administered to the patient and required for use in treatment or prophylaxis according to the present invention will vary with the route of administration, the nature and severity of the condition for which treatment or prophylaxis is required, the age, weight and condition of the patient, concomitant medication and will be ultimately at the discretion of the attendant physician. In general, however, the DPP-4 inhibitor and, optionally, the second and/or third antidiabetic agent according to this invention are included in the pharmaceutical composition, combination or dosage form in an amount sufficient that by their administration the glycemic control in the patient to be treated is improved.
  • In the following preferred ranges of the amount of the DPP-4 inhibitor, the second and/or third antidiabetic agent to be employed in the pharmaceutical composition and the methods and uses according to this invention are described. These ranges refer to the amounts to be administered per day with respect to an adult patient, in particular to a human being, for example of approximately 70 kg body weight, and can be adapted accordingly with regard to an administration 2, 3, 4 or more times daily and with regard to other routes of administration and with regard to the age of the patient. The ranges of the dosage and amounts are calculated for the inidividual active moiety. Advantageously, the combination therapy of the present invention utilizes lower dosages of the individual DPP-4 inhibitor and/or of the individual second and/or third antidiabetic agent used in monotherapy or used in conventional therapeutics, thus avoiding possible toxicity and adverse side effects incurred when those agents are used as monotherapies.
  • Within the scope of the present invention, the pharmaceutical composition or combination is preferably administered orally. Other forms of administration are possible and described hereinafter. Preferably the one or more dosage forms comprising the DPP-4 inhibitor and/or the second and/or the third antidiabetic agent is oral or usually well known.
  • In general, the amount of the DPP-4 inhibitor in the combinations, combination methods or combined uses of this invention is preferably in the range from 1/5 to 1/1 of the amount usually recommended for a monotherapy using said DPP-4 inhibitor.
  • A preferred dosage range of linagliptin when administered orally is 0.5 mg to 10 mg per day, preferably 2.5 mg to 10 mg, most preferably 1 mg to 5 mg per day. The preferred range of amounts in the pharmaceutical composition is 0.5 to 10 mg, in particular 1 to 5 mg. Examples of particular dosage strengths are are 1, 2.5, 5 or 10 mg. The application of the active ingredient may occur up to three times a day, preferably one or two times a day. Suitable formulations for linagliptin may be those formulations disclosed in the application WO 2007/128724, the disclosure of which is incorporated herein in its entirety.
  • Typical dosage strengths of the dual fixed dose combination (tablet) of linagliptin/metformin IR (immediate release) are 2.5/500 mg, 2.5/850 mg and 2.5/1000 mg, which may be administered 1-3 times a day, particularly twice a day.
  • Typical dosage strengths of the dual fixed dose combination (tablet) of linagliptin/metformin XR (extended release) are 5/500 mg, 5/1000 mg and 5/1500 mg, which may be administered 1-2 times a day, particularly once a day, preferably to be taken in the evening with meal, or 2.5/500 mg, 2.5/750 mg and 2.5/1000 mg, which may be administered 1-2 times a day, particularly once a day two tablets, preferably to be taken in the evening with meal.
  • A preferred dosage range of sitagliptin when administered orally is from 10 to 200 mg, in particular 25 to 150 mg per day. A recommended dose of sitagliptin is 100 mg calculated for the active moiety (free base anhydrate) once daily or 50 mg twice daily. The preferred range of amounts in the pharmaceutical composition is 10 to 150 mg, in particular 25 to 100 mg. Examples are 25, 50, 75 or 100 mg. The application of the active ingredient may occur up to three times a day, preferably one or two times a day. Equivalent amounts of salts of sitagliptin, in particular of the phosphate monohydrate can be calculated accordingly. Adjusted dosages of sitagliptin, for example 25 and 50 mg, are preferably used for patients with renal failure.
  • A preferred dosage range of vildagliptin when administered orally is from 10 to 150 mg daily, in particular from 25 to 150 mg, 25 and 100 mg or 25 and 50 mg or 50 and 100 mg daily. For example the daily administration of vildagliptin is 50 or 100 mg. The preferred range of amounts in the pharmaceutical composition is 10 to 150 mg, in particular 25 to 100 mg. Examples are 25, 50, 75 or 100 mg. The application of the active ingredient may occur up to three times a day, preferably one or two times a day.
  • A preferred dosage range of alogliptin when administered orally is from 5 to 250 mg daily, in particular from 10 to 150 mg daily. The preferred range of amounts in the pharmaceutical composition is 5 to 150 mg, in particular 10 to 100 mg. Examples are 10, 12.5, 20, 25, 50, 75 and 100 mg. The application of the active ingredient may occur up to three times a day, preferably one or two times a day.
  • A preferred dosage range of saxagliptin when administered orally is from 2.5 to 100 mg daily, in particular from 2.5 to 50 mg daily. The preferred range of amounts in the pharmaceutical composition is from 2.5 to 100 mg, in particular from 2.5 and 50 mg. Examples are 2.5, 5, 10, 15, 20, 30, 40, 50 and 100 mg. The application of the active ingredient may occur up to three times a day, preferably one or two times a day.
  • A preferred dosage range of dutogliptin when administered orally is from 50 to 400 mg daily, in particular from 100 to 400 mg daily. The preferred range of amounts in the pharmaceutical composition is from 50 to 400 mg. Examples are 50, 100, 200, 300 and 400 mg. The application of the active ingredient may occur up to three times a day, preferably one or two times a day.
  • A special embodiment of the DPP-4 inhibitors of this invention refers to those orally administered DPP-4 inhibitors which are therapeutically efficacious at low dose levels, e.g. at dose levels <100 mg or <70 mg per patient per day, preferably <50 mg, more preferably <30 mg or <20 mg, even more preferably from 1 mg to 10 mg (if required, divided into 1 to 4 single doses, particularly 1 or 2 single doses, which may be of the same size), particularly from 1 mg to 5 mg (more particularly 5 mg), per patient per day, preferentially, administered orally once-daily, more preferentially, at any time of day, administered with or without food. Thus, for example, the daily oral amount 5 mg BI 1356 can be given in a once daily dosing regimen (i.e. 5 mg BI 1356 once daily) or in a twice daily dosing regimen (i.e. 2.5 mg BI 1356 twice daily), at any time of day, with or without food.
  • In general, the amount of the the second and/or third antidiabetic agent in the combinations, combination methods and/or combined uses of this invention is preferably in the range from 1/5 to 1/1 of the amount usually recommended for a monotherapy using said antidiabetic agent. Using lower dosages of the individual second and/or third antidiabetic agent compared with monotherapy could avoid or minimize possible toxicity and adverse side effects incurred when those agents are used as monotherapies.
  • A preferred dosage range of metformin when administered orally is 250 to 3000 mg, in particular 500 to 2000 mg per day. The preferred range of amounts in the pharmaceutical composition is 250 to 1000, in particular 500 to 1000 mg or 250 to 850 mg respectively. Examples are 500, 750, 850 or 1000 mg. Preferably the administration of said amounts is once, twice or three times daily. For example the amounts of 500, 750 and 850 mg preferably require once-daily, twice-daily or three-times daily dosing and the amount of 1000 mg preferably requires once-daily or twice-daily dosing. Certain controlled or sustained release formulations allow a once-daily dosing. Metformin can be administered for example in the form as marketed under the trademarks GLUCOPHAGE™, GLUCOPHAGE-D™ or GLUCOPHAGE-XR™.
  • A preferred dosage range of pioglitazone when administered orally is 5 to 50 mg per day. The preferred range of amounts in the pharmaceutical composition is 5 to 50 mg, 10 to 45 mg and 15 to 45 mg respectively. Examples are 15, 30 or 45 mg. Preferably the administration of said amounts is once or twice daily, in particular once daily. Pioglitazone can be administered in the form as it is marketed for example under the trademark ACTOS™.
  • A preferred dosage range of rosiglitazone when administered orally is 1 mg to 10 mg per day. The preferred range of amounts in the pharmaceutical composition is 1 to 10 mg, 2 to 8 mg, 4 to 8 mg and 1 to 4 mg. Examples are 1, 2, 4 or 8 mg. Preferably the administration of said amounts is once or twice daily. Preferably the dose should not exceed 8 mg daily. Rosiglitazone can be administered in the form as it is marketed for example under the trademark AVANDIA™.
  • A preferred dosage range of a thiazolidindione (other than pioglitazone or rosiglitazone as described above) when administered orally is 2 to 100 mg per day. The preferred range of amounts in the pharmaceutical composition for an administration once, twice or three times daily is 2 to 100, 1 to 50 and 1 to 33 mg respectively.
  • A preferred dosage range of glibenclamide when administered orally is 0.5 to 15 mg, in particular 1 to 10 mg per day. The preferred range of amounts in the pharmaceutical composition is 0.5 to 5 mg, in particular 1 to 4 mg. Examples are 1.0, 1.75 and 3.5 mg. Preferably the administration of said amounts is once, twice or three-times daily. Glibenclamide can be administered in the form as it is marketed for example under the trademark EUGLUCON™.
  • A preferred dosage range of glimepiride when administered orally is 0.5 to 10 mg, in particular 1 to 6 mg per day. The preferred range of amounts in the pharmaceutical composition is 0.5 to 10 mg, in particular 1 to 6 mg. Examples are 1, 2, 3, 4, and 6 mg. Preferably the administration of said amounts is once, twice or three-times daily, preferably once daily. Glimepiride can be administered in the form as it is marketed for example under the trademark AMARYL™.
  • A preferred dosage range of gliquidone when administered orally is 5 to 150 mg, in particular 15 to 120 mg per day. The preferred range of amounts in the pharmaceutical composition is 5 to 120 mg, in particular 5 to 30 mg. Examples are 10, 20, 30 mg. Preferably the administration of said amounts is once, twice, three-times or four-times daily. Gliquidone can be administered in the form as it is marketed for example under the trademark GLURENORM™.
  • A preferred dosage range of glibornuride when administered orally is 5 to 75 mg per day. The preferred range of amounts in the pharmaceutical composition is 5 to 75 mg, in particular 10 to 50 mg. Preferably the administration of said amounts is once, twice or three-times daily.
  • A preferred dosage range of gliclazide when administered orally is 20 to 300 mg, in particular 40 to 240 mg per day. The preferred range of amounts in the pharmaceutical composition is 20 to 240 mg, in particular 20 to 80 mg. Examples are 20, 30, 40 and 50 mg. Preferably the administration of said amounts is once, twice or three-times daily.
  • A preferred dosage range of glisoxepide when administered orally is 1 to 20 mg, in particular 1 to 16 mg per day. The preferred range of amounts in the pharmaceutical composition is 1 to 8 mg, in particular 1 to 4 mg. Preferably the administration of said amounts is once, twice, three-times or four-times daily.
  • A preferred dosage range of tolbutamide when administered orally is 100 to 3000 mg, preferably 500 to 2000 mg per day. The preferred range of amounts in the pharmaceutical composition is 100 to 1000 mg. Preferably the administration of said amounts is once or twice daily.
  • A preferred dosage range of glipizide when administered orally is 1 to 50 mg, in particular 2.5 to 40 mg per day. The preferred range of amounts in the pharmaceutical composition for an administration once, twice or three times daily is 1 to 50, 0.5 to 25 and 0.3 to 17 mg respectively.
  • A preferred dosage range of nateglinide when administered orally is 30 to 500 mg, in particular 60 to 360 mg per day. The preferred range of amounts in the pharmaceutical composition is 30 to 120 mg. Examples are 30, 60 and 120 mg. Preferably the administration of said amounts is once, twice or three-times daily. Nateglinide can be administered in the form as it is marketed for example under the trademark STARLIX™.
  • A preferred dosage range of repaglinide when administered orally is 0.1 to 16 mg, in particular 0.5 to 6 mg per day.
  • The preferred range of amounts in the pharmaceutical composition is 0.5 to 4 mg. Examples are 0.5, 1, 2 or 4 mg. Preferably the administration of said amounts is once, twice, three-times or four-times daily. Repaglinide can be administered in the form as it is marketed for example under the trademark NOVONORM™.
  • A preferred dosage range of acarbose when administered orally is 50 to 1000 mg, in particular 50 to 600 mg per day. The preferred range of amounts in the pharmaceutical composition is 50 to 150 mg. Examples are 50 and 100 mg. Preferably the administration of said amounts is once, twice, three-times or four-times daily. Acarbose can be administered in the form as it is marketed for example under the trademark Glucobay™.
  • A preferred dosage range of voglibose when administered orally is 100 to 1000 mg, in particular 200 to 600 mg per day. The preferred range of amounts in the pharmaceutical composition is 50 to 300 mg. Examples are 50, 100, 150, 200 and 300 mg. Preferably the administration of said amounts is once, twice, three-times or four-times daily. Voglibose can be administered in the form as it is marketed for example under the trademark Basen™ or Voglisan™.
  • A preferred dosage range of miglitol when administered orally is 25 to 500 mg, in particular 25 to 300 mg per day. The preferred range of amounts in the pharmaceutical composition is 25 to 100 mg. Examples are 25, 50 and 100 mg. Preferably the administration of said amounts is once, twice, three-times or four-times daily. Miglitol can be administered in the form as it is marketed for example under the trademark Glyset™.
  • A preferred dosage range of GLP-1 analogues, in particular of exenatide is 5 to 30 μg, in particular 5 to 20 μg per day. The preferred range of amounts in the pharmaceutical composition is 5 to 10 μg. Examples are 5 and 10 μg. Preferably the administration of said amounts is once, twice, three-times or four-times daily by subcutaneous injection. Exenatide can be administered in the form as it is marketed for example under the trademark Byetta™. A long acting formulation, preferably for a once weekly subcutaneous injection, comprises an amount from 0.1 to 3.0 mg, preferably 0.5 to 2.0 mg exenatide. Examples are 0.8 mg and 2.0 mg. An example of a long acting formulation of exenatide is Byetta LAR™.
  • A preferred dosage range of liraglutide is 0.5 to 3 mg, in particular 0.5 to 2 mg per day. The preferred range of amounts in the pharmaceutical composition is 0.5 to 2 mg. Examples are 0.6, 1.2 and 1.8 mg. Preferably the administration of said amounts is once or twice daily by subcutaneous injection.
  • The amount of the DPP-4 inhibitor and the second and/or third therapeutic agent in the pharmaceutical composition and in the methods and uses of this invention correspond to the respective dosage ranges as provided hereinbefore. For example, preferred dosage ranges in a pharmaceutical composition, combination, method and use according to this invention are an amount of 0.5 to 10 mg (in particular 1 to 5 mg, especially 2.5 mg or 5 mg) of linagliptin and/or, optionally, an amount of 250 to 1000 mg (especially 500 mg, 850 mg or 1000 mg) of metformin. An oral administration once or twice daily is preferred.
  • In the combination methods and combined uses according to the present invention the DPP-4 inhibitor and the second and/or third therapeutic agent are administered in combination including, without being limited, the active ingredients are administered at the same time, i.e. simultaneously, or essentially at the same time, or the active ingredients are administered in alternation, i.e. that at first one or two active ingredients are administered and after a period of time the other two or one active ingredients are administered, i.e. at least two of the three active ingredients are administered sequentially. The period of time may be in the range from 30 min to 12 hours. The administration which is in combination or in alternation may be once, twice, three times or four times daily, preferably once or twice daily.
  • With regard to combined administration of the DPP-4 inhibitor and the second and/or third antidiabetic agent, all three active ingredients may be present in one single dosage form, for example in one tablet or capsule, or one or two of the active ingredients may be present in a separate dosage form, for example in two different or identical dosage forms.
  • With regard to their administration in alternation, one or two of the active ingredients are present in a separate dosage form, for example in two different or identical dosage forms.
  • Therefore, a pharmaceutical combination of this invention may be present as single dosage forms which comprise the DPP-4 inhibitor and the second and, optionally, the third antidiabetic agent. Alternatively a pharmaceutical combination of this invention may be present as two separate dosage forms wherein one dosage form comprises the DPP-4 inhibitor and the other dosage form comprises the second plus, optionally, the third antidiabetic agent, or, in case of a triple combination, one dosage form comprises the DPP-4 inhibitor inhibitor plus either the second or the third antidiabetic agent and the other dosage form comprises the third or the second antidiabetic agent, respectively. Alternatively, in case of a triple combination, a pharmaceutical combination of this invention may be present as three separate dosage forms wherein one dosage form comprises the DPP-4 inhibitor and a second dosage form comprises the second antidiabetic agent and the third dosage form comprises the third antidiabetic agent. Alternatively, in case of a dual combination, a pharmaceutical combination of this invention may be present as two separate dosage forms wherein one dosage form comprises the DPP-4 inhibitor and the second dosage form comprises the second antidiabetic agent.
  • The case may arise in which an active ingredient has to be administered more often, for example twice per day, than the other active ingredient(s), which for example needs administration once daily. Therefore “administration in combination” also includes an administration scheme in which first all active ingredients are administered in combination and after a period of time an active ingredient is administered again or vice versa.
  • Therefore, the present invention also includes pharmaceutical combinations which are present in separate dosage forms wherein one dosage form comprises the DPP-4 inhibitor and the second and, optionally, the third, therapeutic agent and the other dosage form comprises the second and/or the third therapeutic agent only.
  • Thus, the present invention also includes pharmaceutical compositions or combinations for separate, sequential, simultaneous, concurrent, alternate or chronologically staggered use of the active ingredients or components.
  • A pharmaceutical composition which is present as a separate or multiple dosage form, preferably as a kit of parts, is useful in combination therapy to flexibly suit the individual therapeutic needs of the patient.
  • According to a first embodiment a kit of parts comprises
      • (a) a first containment containing a dosage form comprising the DPP-4 inhibitor and at least one pharmaceutically acceptable carrier, and
      • (b) a second containment containing a dosage form comprising the second antidiabetic agent and at least one pharmaceutically acceptable carrier, and, optionally,
      • (c) a third containment containing a dosage form comprising the third antidiabetic agent and at least one pharmaceutically acceptable carrier.
  • According to a second embodiment a kit of parts comprises
      • (a) a first containment containing a dosage form comprising the DPP-4 inhibitor and the second or third antidiabetic agent and at least one pharmaceutically acceptable carrier, and
      • (b) a second containment containing a dosage form comprising the third or second antidiabetic agent, respectively, and at least one pharmaceutically acceptable carrier.
  • According to a third embodiment a kit of parts comprises
      • (a) a first containment containing a dosage form comprising the DPP-4 inhibitor and at least one pharmaceutically acceptable carrier, and
      • (b) a second containment containing a dosage form comprising the second and third antidiabetic agent and at least one pharmaceutically acceptable carrier.
  • A further aspect of the present invention is a manufacture comprising the pharmaceutical combination being present as separate dosage forms according to the present invention and a label or package insert comprising instructions that the separate dosage forms are to be administered in combination.
  • According to a first embodiment a manufacture comprises (a) a pharmaceutical composition comprising a DPP-4 inhibitor according to the present invention and (b) a label or package insert which comprises instructions that the medicament may or is to be administered, for example in combination, with a medicament comprising a second antidiabetic agent according to the present invention or with a fixed or free combination (e.g. a medicament) comprising a second antidiabetic agent and a third antidiabetic agent according to the present invention.
  • According to a second embodiment a manufacture comprises (a) a second antidiabetic agent according to the present invention and (b) a label or package insert which comprises instructions that the medicament may or is to be administered, for example in combination, with a medicament comprising a DPP-4 inhibitor according to the present invention or with a a fixed or free-combination (e.g. a medicament) comprising a DPP-4 inhibitor and a third antidiabetic agent according to the present invention.
  • According to a third embodiment a manufacture comprises (a) a pharmaceutical composition comprising a DPP-4 inhibitor and a second antidiabetic agent according to the present invention and (b) a label or package insert which comprises instructions that the medicament may or is to be administered, for example in combination, with a medicament comprising a third antidiabetic agent according to the present invention.
  • The desired dose of the pharmaceutical composition according to this invention may conveniently be presented in a once daily or as divided dose administered at appropriate intervals, for example as two, three or more doses per day.
  • The pharmaceutical composition may be formulated for oral, rectal, nasal, topical (including buccal and sublingual), transdermal, vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration in liquid or solid form or in a form suitable for administration by inhalation or insufflation. Oral administration is preferred. The formulations may, where appropriate, be conveniently presented in discrete dosage units and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing into association the active ingredient with one or more pharmaceutically acceptable carriers, like liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired formulation.
  • The pharmaceutical composition may be formulated in the form of tablets, granules, fine granules, powders, capsules, caplets, soft capsules, pills, oral solutions, syrups, dry syrups, chewable tablets, troches, effervescent tablets, drops, suspension, fast dissolving tablets, oral fast-dispersing tablets, etc.
  • The pharmaceutical composition and the dosage forms preferably comprises one or more pharmaceutical acceptable carriers. Preferred carriers must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Examples of pharmaceutically acceptable carriers are known to the one skilled in the art.
  • Pharmaceutical compositions suitable for oral administration may conveniently be presented as discrete units such as capsules, including soft gelatin capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution, a suspension or as an emulsion, for example as syrups, elixirs or self-emulsifying delivery systems (SEDDS). The active ingredients may also be presented as a bolus, electuary or paste. Tablets and capsules for oral administration may contain conventional excipients such as binding agents, fillers, lubricants, disintegrants, or wetting agents. The tablets may be coated according to methods well known in the art. Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous vehicles (which may include edible oils), or preservatives.
  • The pharmaceutical composition according to the invention may also be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative. The compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredients may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilisation from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • Pharmaceutical compositions suitable for rectal administration wherein the carrier is a solid are most preferably presented as unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art, and the suppositories may be conveniently formed by admixture of the active compound(s) with the softened or melted carrier(s) followed by chilling and shaping in moulds.
  • For pharmaceutical application in warm-blooded vertebrates, particularly humans, the compounds of this invention are usually used in dosages from 0.001 to 100 mg/kg body weight, preferably at 0.1-15 mg/kg, in each case 1 to 4 times a day. For this purpose, the compounds, optionally combined with other active substances, may be incorporated together with one or more inert conventional carriers and/or diluents, e.g. with corn starch, lactose, glucose, microcrystalline cellulose, magnesium stearate, polyvinylpyrrolidone, citric acid, tartaric acid, water, water/ethanol, water/glycerol, water/sorbitol, water/polyethylene glycol, propylene glycol, cetylstearyl alcohol, carboxymethylcellulose or fatty substances such as hard fat or suitable mixtures thereof into conventional galenic preparations such as plain or coated tablets, capsules, powders, suspensions or suppositories.
  • The pharmaceutical compositions according to this invention comprising the DPP-4 inhibitors as defined herein are thus prepared by the skilled person using pharmaceutically acceptable formulation excipients as described in the art. Examples of such excipients include, without being restricted to diluents, binders, carriers, fillers, lubricants, flow promoters, crystallisation retardants, disintegrants, solubilizers, colorants, pH regulators, surfactants and emulsifiers.
  • Examples of suitable diluents for compounds according to embodiment A include cellulose powder, calcium hydrogen phosphate, erythritol, low substituted hydroxypropyl cellulose, mannitol, pregelatinized starch or xylitol. Among those diluents mannitol, low substituted hydroxypropyl cellulose and pregelatinized starch are to be emphasized.
  • Examples of suitable lubricants for compounds according to embodiment A include talc, polyethyleneglycol, calcium behenate, calcium stearate, hydrogenated castor oil or magnesium stearate. Among those lubricants magnesium stearate is to be emphasized.
  • Examples of suitable binders for compounds according to embodiment A include copovidone (copolymerisates of vinylpyrrolidon with other vinylderivates), hydroxypropyl methylcellulose (HPMC), hydroxypropylcellulose (HPC), polyvinylpyrrolidon (povidone), pregelatinized starch, or low-substituted hydroxypropylcellulose (L-HPC). Among those binders copovidone and pregelatinized starch are to be emphasized.
  • Examples of suitable disintegrants for compounds according to embodiment A include corn starch or crospovidone. Among those disintegrants corn starch is to be emphasized.
  • Suitable methods of preparing pharmaceutical formulations of the DPP-4 inhibitors according to embodiment A of the invention are
      • direct tabletting of the active substance in powder mixtures with suitable tabletting excipients;
      • granulation with suitable excipients and subsequent mixing with suitable excipients and subsequent tabletting as well as film coating; or
      • packing of powder mixtures or granules into capsules.
  • Suitable granulation methods are
      • wet granulation in the intensive mixer followed by fluidised bed drying;
      • one-pot granulation;
      • fluidised bed granulation; or
      • dry granulation (e.g. by roller compaction) with suitable excipients and subsequent tabletting or packing into capsules.
  • An exemplary composition of a DPP-4 inhibitor according to embodiment A of the invention comprises the first diluent mannitol, pregelatinized starch as a second diluent with additional binder properties, the binder copovidone, the disintegrant corn starch, and magnesium stearate as lubricant; wherein copovidone and/or corn starch may be optional.
  • For details on dosage forms, formulations and administration of DPP-4 inhibitors of this invention, reference is made to scientific literature and/or published patent documents, particularly to those cited herein.
  • The pharmaceutical compositions (or formulations) may be packaged in a variety of ways. Generally, an article for distribution includes a container that contains the pharmaceutical composition in an appropriate form. Tablets are typically packed in an appropriate primary package for easy handling, distribution and storage and for assurance of proper stability of the composition at prolonged contact with the environment during storage. Primary containers for tablets may be bottles or blister packs.
  • A suitable bottle, e.g. for a pharmaceutical composition or combination comprising a DPP-4 inhibitor according to embodiment A of the invention, may be made from glass or polymer (preferably polypropylene (PP) or high density polyethylene (HD-PE)) and sealed with a screw cap. The screw cap may be provided with a child resistant safety closure (e.g. press-and-twist closure) for preventing or hampering access to the contents by children. If required (e.g. in regions with high humidity), by the additional use of a desiccant (such as e.g. bentonite clay, molecular sieves, or, preferably, silica gel) the shelf life of the packaged composition can be prolonged.
  • A suitable blister pack, e.g. for a pharmaceutical composition or combination comprising a DPP-4 inhibitor according to embodiment A of the invention, comprises or is formed of a top foil (which is breachable by the tablets) and a bottom part (which contains pockets for the tablets). The top foil may contain a metalic foil, particularly an aluminium or aluminium alloy foil (e.g. having a thickness of 20 μm to 45 μm, preferably 20 μm to 25 μm) that is coated with a heat-sealing polymer layer on its inner side (sealing side). The bottom part may contain a multi-layer polymer foil (such as e.g. poly(vinyl choride) (PVC) coated with poly(vinylidene choride) (PVDC); or a PVC foil laminated with poly(chlorotriflouroethylene) (PCTFE)) or a multi-layer polymer-metal-polymer foil (such as e.g. a cold-formable laminated PVC/aluminium/polyamide composition).
  • The article may further comprise a label or package insert, which refer to instructions customarily included in commercial packages of therapeutic products, that may contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products. In one embodiment, the label or package inserts indicates that the composition can be used for any of the purposes described herein.
  • The pharmaceutical compositions and methods according to this invention show advantageous effects in the treatment and prevention of those diseases and conditions as described hereinbefore. The dual combinations show advantageous effects compared with monotherapy with an active ingredient. The triple combinations show advantageous effects compared with dual therapy with one or two of the three active ingredients. Advantageous effects may be seen for example with respect to efficacy, dosage strength, dosage frequency, pharmacodynamic properties, pharmacokinetic properties, fewer adverse effects, convenience, compliance, etc.
  • With respect to linagliptin, the methods of synthesis are known to the skilled person and as described in the literature, in particular as described in WO 2002/068420, WO 2004/018468, or WO 2006/048427, the disclosures of which are incorporated herein. Polymorphous crystal modifications and formulations of particular DPP-4 inhibitors are disclosed in WO 2007/128721 and WO 2007/128724, respectively, the disclosures of which are incorporated herein in their entireties. Formulations of particular DPP-4 inhibitors with metformin or other combination partners are described in WO 2009/121945, the disclosure of which is incorporated herein in its entirety.
  • The methods of synthesis for the further DPP-4 inhibitors are described in the scientific literature and/or in published patent documents, particularly in those cited hereinbefore.
  • The active ingredients, in particular the DPP-4 inhibitor and/or the second and/or the third antidiabetic agent, may be present in the form of a pharmaceutically acceptable salt. Pharmaceutically acceptable salts include, without being restricted thereto, such as salts of inorganic acid like hydrochloric acid, sulfuric acid and phosphoric acid; salts of organic carboxylic acid like oxalic acid, acetic acid, citric acid, malic acid, benzoic acid, maleic acid, fumaric acid, tartaric acid, succinic acid and glutamic acid and salts of organic sulfonic acid like methanesulfonic acid and p-toluenesulfonic acid. The salts can be formed by combining the compound and an acid in the appropriate amount and ratio in a solvent and decomposer. They can be also obtained by the cation or anion exchange from the form of other salts.
  • The active ingredients or a pharmaceutically acceptable salt thereof may be present in the form of a solvate such as a hydrate or alcohol adduct.
  • As different metabolic functional disorders often occur simultaneously, it is quite often indicated to combine a number of different active principles with one another. Thus, depending on the functional disorders diagnosed, improved treatment outcomes may be obtained if a DPP-4 inhibitor is combined with active substances customary for the respective disorders, such as e.g. one or more active substances selected from among the other antidiabetic substances, especially active substances that lower the blood sugar level or the lipid level in the blood, raise the HDL level in the blood, lower blood pressure or are indicated in the treatment of atherosclerosis or obesity.
  • The DPP-4 inhibitors mentioned above—besides their use in mono-therapy—may also be used in conjunction with other active substances, by means of which improved treatment results can be obtained. Such a combined treatment may be given as a free combination of the substances or in the form of a fixed combination, for example in a tablet or capsule. Pharmaceutical formulations of the combination partner needed for this may either be obtained commercially as pharmaceutical compositions or may be formulated by the skilled man using conventional methods. The active substances which may be obtained commercially as pharmaceutical compositions are described in numerous places in the prior art, for example in the list of drugs that appears annually, the “Rote Liste®” of the federal association of the pharmaceutical industry, or in the annually updated compilation of manufacturers' information on prescription drugs known as the “Physicians' Desk Reference”.
  • Examples of antidiabetic combination partners are metformin; sulphonylureas such as glibenclamide, tolbutamide, glimepiride, glipizide, gliquidon, glibornuride and gliclazide; nateglinide; repaglinide; thiazolidinediones such as rosiglitazone and pioglitazone; PPAR gamma modulators such as metaglidases; PPAR-gamma agonists such as rivoglitazone, mitoglitazone, INT-131 or balaglitazone; PPAR-gamma antagonists; PPAR-gamma/alpha modulators such as tesaglitazar, muraglitazar, aleglitazar, indeglitazar and KRP297; PPAR-gamma/alpha/delta modulators such as e.g. lobeglitazone; AMPK-activators such as AICAR; acetyl-CoA carboxylase (ACC1 and ACC2) inhibitors; diacylglycerol-acetyltransferase (DGAT) inhibitors; pancreatic beta cell GCRP agonists such as SMT3-receptor-agonists and GPR119, such as the GPR119 agonists 5-ethyl-2-{4-[4-(4-tetrazol-1-yl-phenoxymethyl)-thiazol-2-yl]-piperidin-1-yl}-pyrimidine or 5-[1-(3-isopropyl-[1,2,4]oxadiazol-5-yl)-piperidin-4-ylmethoxy]-2-(4-methanesulfonyl-phenyl)-pyridine; 11B-HSD-inhibitors; FGF19 agonists or analogues; alpha-glucosidase blockers such as acarbose, voglibose and miglitol; alpha2-antagonists; insulin and insulin analogues such as human insulin, insulin lispro, insulin glusilin, r-DNA-insulinaspart, NPH insulin, insulin detemir, insulin degludec, insulin tregopil, insulin zinc suspension and insulin glargin; Gastric inhibitory Peptide (GIP); amylin and amylin analogues (e.g. pramlintide or davalintide); GLP-1 and GLP-1 analogues such as Exendin-4, e.g. exenatide, exenatide LAR, liraglutide, taspoglutide, lixisenatide (AVE-0010), LY-2428757, dulaglutide (LY-2189265), semaglutide or albiglutide; SGLT2-inhibitors such as e.g. dapagliflozin, sergliflozin (KGT-1251), atigliflozin, canagliflozin, ipragliflozin or tofogliflozin; inhibitors of protein tyrosine-phosphatase (e.g. trodusquemine); inhibitors of glucose-6-phosphatase; fructose-1,6-bisphosphatase modulators; glycogen phosphorylase modulators; glucagon receptor antagonists; phosphoenolpyruvatecarboxykinase (PEPCK) inhibitors; pyruvate dehydrogenasekinase (PDK) inhibitors; inhibitors of tyrosine-kinases (50 mg to 600 mg) such as PDGF-receptor-kinase (cf. EP-A-564409, WO 98/35958, U.S. Pat. No. 5,093,330, WO 2004/005281, and WO 2006/041976) or of serine/threonine kinases; glucokinase/regulatory protein modulators incl. glucokinase activators; glycogen synthase kinase inhibitors; inhibitors of the SH2-domain-containing inositol 5-phosphatase type 2 (SHIP2); IKK inhibitors such as high-dose salicylate; JNK1 inhibitors; protein kinase C-theta inhibitors; beta 3 agonists such as ritobegron, YM 178, solabegron, talibegron, N-5984, GRC-1087, rafabegron, FMP825; aldosereductase inhibitors such as AS 3201, zenarestat, fidarestat, epalrestat, ranirestat, NZ-314, CP-744809, and CT-112; SGLT-1 or SGLT-2 inhibitors, such as e.g. dapagliflozin, sergliflozin, atigliflozin, canagliflozin or (1S)-1,5-anhydro-1-[3-(1-benzothiophen-2-ylmethyl)-4-fluorophenyl]-D-glucitol; KV 1.3 channel inhibitors; GPR40 modulators such as e.g. [(35)-6-({2′,6′-dimethyl-4′-[3-(methylsulfonyl)propoxy]biphenyl-3-yl}methoxy)-2,3-dihydro-1-benzofuran-3-yl]acetic acid; SCD-1 inhibitors; CCR-2 antagonists; dopamine receptor agonists (bromocriptine mesylate [Cycloset]); 4-(3-(2,6-dimethylbenzyloxy)phenyl)-4-oxobutanoic acid; sirtuin stimulants; and other DPP IV inhibitors.
  • Metformin is usually given in doses varying from about 500 mg to 2000 mg up to 2500 mg per day using various dosing regimens from about 100 mg to 500 mg or 200 mg to 850 mg (1-3 times a day), or about 300 mg to 1000 mg once or twice a day, or delayed-release metformin in doses of about 100 mg to 1000 mg or preferably 500 mg to 1000 mg once or twice a day or about 500 mg to 2000 mg once a day. Particular dosage strengths may be 250, 500, 625, 750, 850 and 1000 mg of metformin hydrochloride.
  • For children 10 to 16 years of age, the recommended starting dose of metformin is 500 mg given once daily. If this dose fails to produce adequate results, the dose may be increased to 500 mg twice daily. Further increases may be made in increments of 500 mg weekly to a maximum daily dose of 2000 mg, given in divided doses (e.g. 2 or 3 divided doses). Metformin may be administered with food to decrease nausea.
  • A dosage of pioglitazone is usually of about 1-10 mg, 15 mg, 30 mg, or 45 mg once a day.
  • Rosiglitazone is usually given in doses from 4 to 8 mg once (or divided twice) a day (typical dosage strengths are 2, 4 and 8 mg).
  • Glibenclamide (glyburide) is usually given in doses from 2.5-5 to 20 mg once (or divided twice) a day (typical dosage strengths are 1.25, 2.5 and 5 mg), or micronized glibenclamide in doses from 0.75-3 to 12 mg once (or divided twice) a day (typical dosage strengths are 1.5, 3, 4.5 and 6 mg).
  • Glipizide is usually given in doses from 2.5 to 10-20 mg once (or up to 40 mg divided twice) a day (typical dosage strengths are 5 and 10 mg), or extended-release glibenclamide in doses from 5 to 10 mg (up to 20 mg) once a day (typical dosage strengths are 2.5, 5 and 10 mg).
  • Glimepiride is usually given in doses from 1-2 to 4 mg (up to 8 mg) once a day (typical dosage strengths are 1, 2 and 4 mg).
  • A dual combination of glibenclamide/metformin is usually given in doses from 1.25/250 once daily to 10/1000 mg twice daily. (typical dosage strengths are 1.25/250, 2.5/500 and 5/500 mg).
  • A dual combination of glipizide/metformin is usually given in doses from 2.5/250 to 10/1000 mg twice daily (typical dosage strengths are 2.5/250, 2.5/500 and 5/500 mg).
  • A dual combination of glimepiride/metformin is usually given in doses from 1/250 to 4/1000 mg twice daily.
  • A dual combination of rosiglitazone/glimepiride is usually given in doses from 4/1 once or twice daily to 4/2 mg twice daily (typical dosage strengths are 4/1, 4/2, 4/4, 8/2 and 8/4 mg).
  • A dual combination of pioglitazone/glimepiride is usually given in doses from 30/2 to 30/4 mg once daily (typical dosage strengths are 30/4 and 45/4 mg).
  • A dual combination of rosiglitazone/metformin is usually given in doses from 1/500 to 4/1000 mg twice daily (typical dosage strengths are 1/500, 2/500, 4/500, 2/1000 and 4/1000 mg).
  • A dual combination of pioglitazone/metformin is usually given in doses from 15/500 once or twice daily to 15/850 mg thrice daily (typical dosage strengths are 15/500 and 15/850 mg).
  • The non-sulphonylurea insulin secretagogue nateglinide is usually given in doses from 60 to 120 mg with meals (up to 360 mg/day, typical dosage strengths are 60 and 120 mg); repaglinide is usually given in doses from 0.5 to 4 mg with meals (up to 16 mg/day, typical dosage strengths are 0.5, 1 and 2 mg). A dual combination of repaglinide/metformin is available in dosage strengths of 1/500 and 2/850 mg.
  • Acarbose is usually given in doses from 25 to 100 mg with meals. Miglitol is usually given in doses from 25 to 100 mg with meals.
  • Examples of combination partners that lower the lipid level in the blood are HMG-CoA-reductase inhibitors such as simvastatin, atorvastatin, lovastatin, fluvastatin, pravastatin, pitavastatin and rosuvastatin; fibrates such as bezafibrate, fenofibrate, clofibrate, gemfibrozil, etofibrate and etofyllinclofibrate; nicotinic acid and the derivatives thereof such as acipimox; PPAR-alpha agonists; PPAR-delta agonists; inhibitors of acyl-coenzyme
  • A:cholesterolacyltransferase (ACAT; EC 2.3.1.26) such as avasimibe; cholesterol resorption inhibitors such as ezetimib; substances that bind to bile acid, such as cholestyramine, colestipol and colesevelam; inhibitors of bile acid transport; HDL modulating active substances such as D4F, reverse D4F, LXR modulating active substances and FXR modulating active substances; CETP inhibitors such as torcetrapib, JTT-705 (dalcetrapib) or compound 12 from WO 2007/005572 (anacetrapib); LDL receptor modulators; MTP inhibitors (e.g. lomitapide); and ApoB100 antisense RNA.
  • A dosage of atorvastatin is usually from 1 mg to 40 mg or 10 mg to 80 mg once a day.
  • Examples of combination partners that lower blood pressure are beta-blockers such as atenolol, bisoprolol, celiprolol, metoprolol and carvedilol; diuretics such as hydrochlorothiazide, chlortalidon, xipamide, furosemide, piretanide, torasemide, spironolactone, eplerenone, amiloride and triamterene; calcium channel blockers such as amlodipine, nifedipine, nitrendipine, nisoldipine, nicardipine, felodipine, lacidipine, lercanipidine, manidipine, isradipine, nilvadipine, verapamil, gallopamil and diltiazem; ACE inhibitors such as ramipril, lisinopril, cilazapril, quinapril, captopril, enalapril, benazepril, perindopril, fosinopril and trandolapril; as well as angiotensin II receptor blockers (ARBs) such as telmisartan, candesartan, valsartan, losartan, irbesartan, olmesartan, azilsartan and eprosartan.
  • A dosage of telmisartan is usually from 20 mg to 320 mg or 40 mg to 160 mg per day.
  • Examples of combination partners which increase the HDL level in the blood are Cholesteryl Ester Transfer Protein (CETP) inhibitors; inhibitors of endothelial lipase; regulators of ABC1; LXRalpha antagonists; LXRbeta agonists; PPAR-delta agonists; LXRalpha/beta regulators, and substances that increase the expression and/or plasma concentration of apolipoprotein A-I.
  • Examples of combination partners for the treatment of obesity are sibutramine; tetrahydrolipstatin (orlistat); alizyme (cetilistat); dexfenfluramine; axokine; cannabinoid receptor 1 antagonists such as the CB1 antagonist rimonobant; MCH-1 receptor antagonists; MC4 receptor agonists; NPY5 as well as NPY2 antagonists (e.g. velneperit); beta3-AR agonists such as SB-418790 and AD-9677; 5HT2c receptor agonists such as APD 356 (lorcaserin); myostatin inhibitors; Acrp30 and adiponectin; steroyl CoA desaturase (SCD1) inhibitors; fatty acid synthase (FAS) inhibitors; CCK receptor agonists; Ghrelin receptor modulators; Pyy 3-36; orexin receptor antagonists; and tesofensine; as well as the dual combinations bupropion/naltrexone, bupropion/zonisamide, topiramate/phentermine and pramlintide/metreleptin.
  • Examples of combination partners for the treatment of atherosclerosis are phospholipase A2 inhibitors; inhibitors of tyrosine-kinases (50 mg to 600 mg) such as PDGF-receptor-kinase (cf. EP-A-564409, WO 98/35958, U.S. Pat. No. 5,093,330, WO 2004/005281, and WO 2006/041976); oxLDL antibodies and oxLDL vaccines; apoA-1 Milano; ASA; and VCAM-1 inhibitors.
  • The present invention is not to be limited in scope by the specific embodiments described herein. Various modifications of the invention in addition to those described herein may become apparent to those skilled in the art from the present disclosure. Such modifications are intended to fall within the scope of the appended claims.
  • All patent applications cited herein are hereby incorporated by reference in their entireties.
  • Further embodiments, features and advantages of the present invention may become apparent from the following examples. The following examples serve to illustrate, by way of example, the principles of the invention without restricting it.
  • EXAMPLES Example 1 BI 1356, a Potent and Selective DPP-4 Inhibitor, is Safe and Efficacious in Patients With Inadequately Controlled Type 2 Diabetes Despite Metformin Therapy
  • Efficacy and safety of BI 1356 (1, 5, or 10 mg qd), a potent and selective dipeptidyl peptidase-4 (DPP-4) inhibitor, was examined in inadequately controlled, metformin-treated (MET, g daily) type 2 diabetic patients (T2DM; HbA1c at baseline 7.5-10.0%). Effects were compared to add-on of placebo (PBO) or of open label glimepiride (GLIM; 1 to 3 mg qd) in a 12-week randomized, double-blind study. Antidiabetic medication other than metformin was washed out for 6 weeks (34.7% of the patients).
  • The primary endpoint was change from baseline in HbA1c, adjusted for prior antidiabetic medication. 333 patients (mean baseline HbA1c 8.3%; fasting plasma glucose [FPG] 185 mg/dL) were randomized to BI 1356, PBO or open-label GLIM. After 12 weeks, BI 1356 treatment resulted in significant placebo corrected mean reductions in HbA1c (BI 1356 1 mg, n=65, −0.39%; 5 mg, n=66, −0.75%; 10 mg, n=66, −0.73%). Patients receiving GLIM demonstrated a slightly greater mean PBO corrected reduction in HbA1c at Week 12 (n=64, −0.90%). Reductions in FPG from baseline to Week 12 with BI 1356 were statistically significant (1 mg, −19 mg/dL; 5 mg, −35 mg/dL; 10 mg, −30 mg/dL). Hence, a dose-response relationship was demonstrated for HbA1c and FPG, reaching an effect plateau at 5 mg of BI 1356. For this dose, >80% DPP-4 inhibition at trough in >80% of the patients at week 12 was achieved.
  • In total, 106 patients (43.1%) experienced adverse events (AEs) with similar incidences across all treatments. Most frequently reported episodes were nasopharyngitis (7.5%), diarrhoea (3.3%), and nausea (3.0%). Drug-related hypoglycaemia did not occur with BI 1356 or PBO but in 3 patients receiving GLIM. Ten patients (3.7%) experienced serious AEs but none of these events were considered drug-related.
  • The addition of BI 1356 to MET in patients with T2DM inadequately controlled on MET alone achieved clinically relevant and statistically significant reductions in HbA1c. Combination treatment with BI 1356 1, 5, and 10 mg and MET was well tolerated and no case of hypoglycaemia was reported. The incidence of AEs was comparable with BI 1356 and PBO.
  • Example 2
  • The usability of a DPP-4 inhibitor or combination according to this invention for the purpose of the present invention (e.g. the beneficial effect on glycemic control) can be tested using clinical trials.
  • For example, in a randomised, double-blind, placebo-controlled, parallel group trial, the safety and efficacy of a DPP-4 inhibitor according to the invention (e.g. 5 mg of linagliptin administered orally once daily) is tested in patients with type 2 diabetes with insufficient glycemic control (HbA1c from 7.0% to 10% or from 7.5% to 10% or from 7.5% to 11%) despite a therapy with one or two conventional antihyperglycemic agents, e.g. selected from metformin, thiazolidindiones (e.g. pioglitazone), sulfonylureas, glinides, inhibitors of alpha-glucosidase, GLP-1 or GLP-1 analogues, and insulin or insulin analogues.
  • In the study with the sulphonylurea drug the efficacy and safety of a DPP-4 inhibitor according to this invention versus placebo added to a background therapy of a sulphonylurea is investigated (2 week placebo run-in phase; 18 weeks double-blind treatment followed by 1 week follow up after study medication termination; background therapy with a sulphonylurea drug is administered throughout the entire trial duration, including placebo run-in phase, in an unchanged dosage).
  • The success of the treatment is tested by determining the HbA1c value, by comparison with the initial value and/or with the value of the placebo group. A significant change in the HbA1c value compared with the initial value and/or the placebo value demonstrates the efficacy of the DPP-4 inhibitor for the treatment. The success of the treatment can be also tested by determining the fasting plasma glucose values, by comparison with the initial values and/or with the values of the placebo group. A significant drop in the fasting glucose levels demonstrates the efficacy of the treatment. Also, the occurrence of a treat to target response (i.e. an HbA1c under treatment <7%) demonstrates the efficacy of the treatment.
  • The safety and tolerability of the treatment is investigated by assessing patient's condition and relevant changes from baseline, e.g. incidence and intensity of adverse events (such as e.g. hypoglycaemic episodes or the like) or weight gain.
  • Example 3 Treatment of Pre-Diabetes
  • The efficacy of a pharmaceutical composition or combination according to the invention in the treatment of pre-diabetes characterised by pathological fasting glucose and/or impaired glucose tolerance can be tested using clinical studies. In studies over a shorter period (e.g. 2-4 weeks) the success of the treatment is examined by determining the fasting glucose values and/or the glucose values after a meal or after a loading test (oral glucose tolerance test or food tolerance test after a defined meal) after the end of the period of therapy for the study and comparing them with the values before the start of the study and/or with those of a placebo group. In addition, the fructosamine value can be determined before and after therapy and compared with the initial value and/or the placebo value. A significant drop in the fasting or non-fasting glucose levels demonstrates the efficacy of the treatment. In studies over a longer period (12 weeks or more) the success of the treatment is tested by determining the HbA1c value, by comparison with the initial value and/or with the value of the placebo group. A significant change in the HbA1c value compared with the initial value and/or the placebo value demonstrates the efficacy of the DPP-4 inhibitors or combinations according to the present invention for treating pre-diabetes.
  • Example 4 Preventing Manifest Type 2 Diabetes
  • Treating patients with pathological fasting glucose and/or impaired glucose tolerance (pre-diabetes) is also in pursuit of the goal of preventing the transition to manifest type 2 diabetes. The efficacy of a treatment can be investigated in a comparative clinical study in which pre-diabetes patients are treated over a lengthy period (e.g. 1-5 years) with either a pharmaceutical composition or combination according to this invention or with placebo or with a non-drug therapy or other medicaments. During and at the end of the therapy, by determining the fasting glucose and/or a loading test (e.g. oGTT), a check is made to determine how many patients exhibit manifest type 2 diabetes, i.e. a fasting glucose level of >125 mg/dl and/or a 2 h value according to oGTT of >199 mg/dl. A significant reduction in the number of patients who exhibit manifest type 2 diabetes when treated with a DPP-4 inhibitor or combination according to the present invention as compared to one of the other forms of treatment, demonstrates the efficacy in preventing a transition from pre-diabetes to manifest diabetes.
  • Example 5 Treatment of Type 2 Diabetes
  • Treating patients with type 2 diabetes with the pharmaceutical composition or combination according to the invention, in addition to producing an acute improvement in the glucose metabolic situation, prevents a deterioration in the metabolic situation in the long term. This can be observed is patients are treated for a longer period, e.g. 3 months to 1 year or even 1 to 6 years, with the pharmaceutical composition or combination according to the invention and are compared with patients who have been treated with other antidiabetic medicaments. There is evidence of therapeutic success compared with patients treated with other antidiabetic medicaments if no or only a slight increase in the fasting glucose and/or HbA1c value is observed. Further evidence of therapeutic success is obtained if a significantly smaller percentage of the patients treated with a pharmaceutical composition or combination according to the invention, compared with patients who have been treated with other medicaments, undergo a deterioration in the glucose metabolic position (e.g. an increase in the HbA1c value to >6.5% or >7%) to the point where treatment with an additional oral antidiabetic medicament or with insulin or with an insulin analogue is indicated.
  • Example 6 Treatment of Insulin Resistance
  • In clinical studies running for different lengths of time (e.g. 2 weeks to 12 months) the success of the treatment is checked using a hyperinsulinaemic euglycaemic glucose clamp study. A significant rise in the glucose infusion rate at the end of the study, compared with the initial value or compared with a placebo group, or a group given a different therapy, proves the efficacy of a DPP-4 inhibitor, pharmaceutical composition or combination according to the present invention according to the invention in the treatment of insulin resistance.
  • Example 7 Treatment of Hyperglycaemia
  • In clinical studies running for different lengths of time (e.g. 1 day to 24 months) the success of the treatment in patients with hyperglycaemia is checked by determining the fasting glucose or non-fasting glucose (e.g. after a meal or a loading test with oGTT or a defined meal). A significant fall in these glucose values during or at the end of the study, compared with the initial value or compared with a placebo group, or a group given a different therapy, proves the efficacy of a DPP-4 inhibitor, pharmaceutical composition or combination according to the present invention according to the invention in the treatment of hyperglycaemia.
  • Example 8 Prevention of Micro- or Macrovascular Complications
  • The treatment of type 2 diabetes or pre-diabetes patients with a DPP-4 inhibitor, pharmaceutical composition or combination according to the invention prevents or reduces or reduces the risk of developing microvascular complications (e.g. diabetic neuropathy, diabetic retinopathy, diabetic nephropathy, diabetic foot, diabetic ulcer) or macrovascular complications (e.g. myocardial infarct, acute coronary syndrome, unstable angina pectoris, stable angina pectoris, stroke, peripheral arterial occlusive disease, cardiomyopathy, heart failure, heart rhythm disorders, vascular restenosis). Type 2 diabetes or patients with pre-diabetes are treated long-term, e.g. for 1-6 years, with a pharmaceutical composition or combination according to the invention and compared with patients who have been treated with other antidiabetic medicaments or with placebo. Evidence of the therapeutic success compared with patients who have been treated with other antidiabetic medicaments or with placebo can be found in the smaller number of single or multiple complications. In the case of macrovascular events, diabetic foot and/or diabetic ulcer, the numbers are counted by anamnesis and various test methods. In the case of diabetic retinopathy the success of the treatment is determined by computer-controlled illumination and evaluation of the background to the eye or other ophthalmic methods. In the case of diabetic neuropathy, in addition to anamnesis and clinical examination, the nerve conduction rate can be measured using a calibrated tuning fork, for example. With regard to diabetic nephropathy the following parameters may be investigated before the start, during and at the end of the study: secretion of albumin, creatinine clearance, serum creatinin values, time taken for the serum creatinine values to double, time taken until dialysis becomes necessary.
  • Example 9 Treatment of Metabolic Syndrome
  • The efficacy of a DPP-4 inhibitor, pharmaceutical composition or combination according to the present invention according to the invention can be tested in clinical studies with varying run times (e.g. 12 weeks to 6 years) by determining the fasting glucose or non-fasting glucose (e.g. after a meal or a loading test with oGTT or a defined meal) or the HbA1c value. A significant fall in these glucose values or HbA1c values during or at the end of the study, compared with the initial value or compared with a placebo group, or a group given a different therapy, proves the efficacy of an active substance or combination of active substances in the treatment of Metabolic Syndrome. Examples of this are a reduction in systolic and/or diastolic blood pressure, a lowering of the plasma triglycerides, a reduction in total or LDL cholesterol, an increase in HDL cholesterol or a reduction in weight, either compared with the starting value at the beginning of the study or in comparison with a group of patients treated with placebo or a different therapy.
  • Example 10 Therapeutic Response to DPP-4 Inhibitor Treatment
  • Genomic DNA samples from individual patients enrolled in a clinical trial (e.g. a clinical study as described herein) for a DPP-4 inhibitor (e.g. linagliptin, e.g. in a daily oral amount of 5 mg, optionally in combination with one or more other antidiabetic agents) are obtained and genotyped for variation(s) (e.g. polymorphisms) in one or more candidate genes selected from TCF7L2, KCNJ11, PPARG and GLP1 R, particularly for a TCF7L2 risk genotype as described herein, and evaluated relative to each patients response in the clinical trial (cf., e.g., Example 21). The association between the likelihood (e.g., increased, decreased, or no likelihood) of a favorable DPP-4 inhibitor therapy response (e.g. favorable change in HbA1c value) and genetic variations (e.g. TCF7L2 risk genotypes) or references can be investigated by applying statistical analysis to the results of genotyping.
  • The probablility of the likelihood of a favorable response of an individual resulting from treating said individual with the DPP-4 inhibitor may be thus determined by such genotyping a nucleic acid sample of the individual, for example by detecting one or more single nucleotide polymorphisms within the TCF7L2 gene, for example one SNP selected from rs7903146, rs12255372 and rs10885406, or by detecting the respective wild-type genotype (cf., e.g., Example 21).
  • Methods for genotyping, i.e. determining genetic variations (e.g. polymorphisms, particularly those described herein) from patients' nucleic acid samples are known in the art. For example, molecular genetic methods to detect single nucleotide polymorphisms, e.g. within the TCF7L2 gene, may be based on genetic sequencing, microarray or PCR analysis.
  • Example 11 Linagliptin Monotherapy Improves Glycemic Control and Measures of β-Cell Function in Type 2 Diabetes
  • In a multi-center, 24 week, randomized, double-blind, placebo-controlled, parallel group study, the effects of linagliptin (LI) monotherapy (5 mg qd) are compared with placebo (PBO) in drug naïve or previously treated patients (pts) with type 2 diabetes mellitus (T2DM) (baseline HbA1c 4.9-10.6%). Randomization to LI (n=336) or PBO (n=167) follows a 2-week PBO run-in (previously treated pts go without medication for 4 wks prior to this). Mean baseline demographics (HbA1c, 8.0% [SD 0.87]; fasting plasma glucose (FPG), 166.0 mg/dL [41.1]; body mass index (BMI), 29.05 kg/m2 [4.81]; age, 55.7 yrs [10.2]) are similar in both groups. The primary endpoint is the change from baseline in HbA1c after 24 wks of treatment. LI shows a PBO-adjusted change in HbA1c from baseline of −0.69% (p<0.0001) with a continuous HbA1c reduction over time of −0.46% at 6 weeks to −0.69% at 24 weeks (both p<0.0001). LI patients are >4-fold more likely to achieve a reduction in HbA1c of ≥0.5% at 24 weeks than PBO (47.1% vs 19.0%; p<0.0001). For patients with baseline HbA1c ≥7.0% a significant greater number of LI-treated compared to PBO-treated patients achieve a target reduction of HbA1c to <7.0% at 24 weeks (25.2% vs. 11.6%; odds ratio of 2.9, p=0.0006). Patients with baseline HbA1c levels of ≥9.0% show the greatest reduction in HbA1c (−0.86%) from baseline. FPG improves by −23.3 mg/dL (p<0.0001) vs. PBO. In a meal tolerance test, the LI patiens show a greater reduction in the adjusted mean change from baseline at week 24 for 2-hr postprandial glucose (PPG) (−58.4 mg/dL; p<0.0001) vs. PBO. LI improves insulin secretion (p<0.05), as shown by changes in HOMA-% B index (LI, 5.02 vs PBO, −17.2 [(mU/L)/(mmol/L)]), proinsulin/insulin ratio (LI, −0.015 vs PBO, 0.024) and the disposition index (LI, 3.05 vs PBO, −0.68). The proportion of patients reporting at least one adverse event (AE) is similar for both groups (52.4% LI; 58.7% PBO). Hypoglycemia is rare, occurring in 1 patients in each of the groups. Serious AEs are reported in both groups (LI, 3.0%; PBO, 4.2%) but are not considered drug-related. Linagliptin trough levels in patients with mild and moderate renal impairment are comparable to patients with normal renal function. Conclusion: Linagliptin monotherapy shows a significant, clinically meaningful and sustained improvement in glycemic control reflected in changes in FPG and HbA1c, and accompanied by β-cell function improvements. Linagliptin is safe and well tolerated with no clinically significant changes in body weight or waist circumference. Linagliptin trough levels in patients with mild and moderate renal impairment are comparable to patients with normal renal function, supporting that no dose adjustment is required in renally impaired patients.
  • Example 12 Efficacy and Safety of Linagliptin in Type 2 Diabetes Inadequately Controlled on Metformin Monotherapy
  • A multi-center, 24-week, randomized, placebo-controlled, double-blind, parallel group study examines the efficacy and safety of linagliptin (LI) administered as add-on therapy to metformin (MET) in type 2 diabetes mellitus (T2DM) hyperglycemic patients with insufficient glycemic control (HbA1c to 0.0% for patients previously treated only with metformin, or ≥6.5 to ≥9.0% for patients previously treated with additional oral antihyperglycemic drugs). Subjects who enter the screening period discontinue previous antidiabetic medication other than MET (≥1500 mg/day) for 6 weeks (including a placebo (PBO) run-in period during the last 2 weeks) prior to randomization to LI (n=524) or PBO (n=177). Mean baseline characteristics and demographics (HbA1c, 8.1%; fasting plasma glucose [FPG], 168.8 mg/dL; age, 56.5 yrs; BMI, 29.9 kg/m2) are similar between groups. The primary endpoint is the change from baseline HbA1c after 24 weeks of treatment, evaluated with an analysis of covariance (ANCOVA) adjusted for baseline HbA1c and prior antidiabetic medication. After 24 weeks of treatment, the adjusted mean treatment difference between LI+MET and PBO+MET is −0.64% (p<0.0001) in favor of LI+MET for change in HbA1c (%). Patients with a baseline HbA1c of ≥7.0% who receive LI+MET are more likely to achieve an HbA1c ≤7.0% relative to those receiving placebo+MET (26.2% vs. 9.2%, respectively; odds ratio, 4.4; p=0.0001). At week 24 LI+MET is superior to PBO+MET in reducing the mean fasting plasma glucose (FPG) from baseline (−21.1 mg/dL; p<0.0001). At study-end, 2 hr post-prandial glucose (PPG) analyzed in meal tolerance tests shows a significantly greater (p<0.0001) mean reduction from baseline for the LI+MET treated (−67.1 mg/dL) versus the PBO+MET group. The proportion of patients reporting at least one adverse event (AE) is comparable within the LI+MET and PBO+MET groups (52.8% and 55.4%, respectively). Hypoglycemia is rare, occurring in 5 PBO+MET patients (2.8%) and 3 LI+MET patients (0.6%), all episodes being of mild intensity. The change in the body weight from baseline to 24 weeks is similar between the 2 treatment groups (−0.5 kg PBO+MET; −0.4 kg LI+MET).
  • Conclusion, linagliptin 5 mg qd as add-on therapy in patients with T2DM inadequately controlled on metformin is well tolerated and produces significant and clinically meaningful improvements in glycemic control (reductions in HbA1c, FPG and 2 h PPG without weight gain). Linagliptin as add-on therapy to metformin in patients with T2DM and insufficient glycemic control is well tolerated with the incidence of adverse events comparable to placebo.
  • Example 13 Linagliptin Improves Glycemic Control in Type 2 Diabetes Patients Inadequately Controlled by Metformin and Sulfonylurea Without Weight Gain or Hypoglycemia
  • A multi-center, 24-week, randomized, double-blind, placebo-controlled, parallel group study examines the efficacy and safety of the DPP-4 inhibitor linagliptin (LI; 5 mg qd) in type 2 diabetes (T2DM) patients (pts) with insufficient glycemic control (HbA1c 7.0-10.0%) on the combination of metformin (MET) plus a sulfonylurea (SU). Effects of LI as add-on are compared with placebo (PBO). All pts have a 2-wk PBO run-in before being randomized to LI+MET+SU (n=793) or PBO+MET+SU (n=265). Mean baseline characteristics are: HbA1c, 8.14% (SD 0.8); fasting plasma glucose (FPG), 160.1 mg/dL (36.6); age, 58.1 yrs (9.8); BMI, 28.3 kg/m2 (4.7). Most of the pts (73.3%) have T2DM for >5 years before enrollment. The primary endpoint is the change from baseline in HbA1c after 24 weeks of treatment, adjusted for baseline HbA1c. After 24 weeks of treatment, the mean HbA1c for LI+MET+SU is −0.62% lower (p<0.0001) relative to PBO+MET+SU. The maximum mean HbA1c reduction with LI+MET+SU is seen at week 12 (−0.84%). Patients with baseline HbA1c ≥7.0% are >5-fold more likely to achieve a target HbA1c of <7.0% when treated with LI+MET+SU (29.2%) compared with PBO+MET+SU (8.1%, odds ratio 5.5, p<0.0001) at 24 weeks. For the change in FPG, a statistically significant (p<0.0001) adjusted mean difference of −12.7 mg/dL is observed between Li+MET+SU and BPBO+MET+SU from baseline at week 24. Measures relating to β-cell function (fasting plasma insulin and HOMA-% B) along with HOMA-IR are significantly (p≤0.05) improved with LI+MET+SU compared with PBO+MET+SU. The proportion of patients that reported a severe adverse event (AE) is low for both LI+MET+SU and PBO+MET+SU groups (2.4% vs. 1.5%, respectively). The most frequent AE reported more commonly in the LI+MET+SU group than in the PBO+MET+SU group is hypoglycemia (22.7% vs. 14.8%, respectively). This is expected due to the combination with SU. No significant changes in weight are noted for either treatment group. Conclusion: Therapy with linagliptin added to the combination of metformin and a sulfonylurea is efficacious and safe in producing significant and clinically meaningful improvements in glycemic control in T2DM patients. Linagliptin may provide an additional option prior to insulin therapy in many patients for whom glycemia is insufficiently controlled with metformin plus a sulfonylurea agent. Linagliptin is shown to have a favorable safety and tolerablility profile. However, when linagliptin is added on pre-existing sulfonylurea therapy, hypoglycemia may occur.
  • Example 14 Efficacy and Safety of Initial Combination Therapy With Linagliptin and Pioglitazone in Patients With Inadequately Controlled Type 2 Diabetes
  • A multi-center, 24-week, randomized, double-blind, placebo-controlled, parallel group study investigates the efficacy and safety of initial combination therapy with the DPP-4 inhibitor linagliptin (LI) and pioglitazone (PIO). Patients (pts) with type 2 diabetes mellitus (T2DM) and insufficient glycemic control (HbA1c 7.5-11.0%) who are drug naïve or previously treated with any oral antihyperglycemic drug (OAD), are randomized to receive 5 mg LI plus 30 mg PIO qd (n=259) or 30 mg PIO plus placebo (PBO) qd (n=130). Patients do not take any OAD for at least 6 weeks before randomization. Mean baseline characteristics (HbA1c 8.6%; fasting plasma glucose [FPG] 190 mg/dL; age 57.5 yrs; BMI 29.0 kg/m2) are similar between the groups. The primary endpoint is the change from baseline in HbA1c after 24 weeks of treatment, adjusted for baseline HbA1c and prior antidiabetic medication. After 24 weeks of treatment, the adjusted mean change in HbA1c for the patients in the LI+PIO group (full analysis set, last observation carried forward) is −1.06% (standard error (SE) ±0.06). The difference in the adjusted mean HbA1c for the LI+PIO group compared with PBO+PIO is −0.51% (p<0.0001; 95% confidence interval (CI), −0.71, −0.30). Reductions in FPG are also significantly greater for the LI+PIO group compared with PBO+PIO with a treatment difference of −14.2 mg/dL (p<0.0001; 95% confidence interval (CI), −21.1, −7.3) at 24 weeks. Patients in the LI+PIO group are more likely to achieve a target HbA1c of <7% vs. those on PBO+PIO (42.9% vs. 30.5%, respectively, odds ratio 2.1; p=0.0051), as well as a reduction in HbA1c of ≥0.5% (75% vs. 50.8%, respectively, odds ratio 3.8; p<0.001). The proportion of patients that experienced at least one adverse event (AE) is similar for both LI+PIO and PBO+PIO groups (136, 52.5% vs. 53.1%, respectively). Hypoglycemia is rare, occurring in 3 patients (1.2%) in the LI+PIO group and none in the PBO+PIO group. All hypoglycemic events are of mild intensity.
  • Conclusion: Initial combination therapy with linagliptin and pioglitazone shows significant and clinically meaningful improvements in FPG and HbA1c levels compared with PIO alone, along with a greater improvement in beta-cell function. Co-administration of linagliptin with pioglitazone is shown to be safe and well tolerated. Combination therapy with linagliptin and pioglitazone may provide an important synergistic initial treatment option for T2DM patients with inadequate glycemic control or those with renal impairment for whom metformin is contraindicated.
  • Example 15 Linagliptin Monotherapy Improves Glycemic Control in Japanese Patients With Type 2 Diabetes Mellitus Over 12 Weeks
  • A multi-center, 12-week, randomized, double-blind, placebo-controlled, parallel group study investigates the efficacy and safety of the DPP-4 inhibitor linagliptin (LI). Effects of LI monotherapy (5 mg qd and 10 mg qd) are compared to placebo (PBO) in drug naïve or previously treated Japanese patients (pts) with type 2 diabetes mellitus (T2DM) (baseline HbA1c 7.0-10.0%, if drug naïve; 7.0-9.0%, if previously treated). Before being randomized to LI 5 (n=159) or 10 mg (n=160), or PBO (n=80), all patients have a 2-week PBO run-in (patients on an oral antihyperglycemic drug have no medication for 2 weeks prior to run-in). Mean [SD] baseline characteristics and demographics (HbA1c, 8.0% [0.68]; fasting plasma glucose (FPG), 163.5 mg/dL [32.4]; BMI, 24.97 kg/m2 [3.86]; age, 60.0 yrs [9.7]) are similar in all groups. The primary endpoint is the change from baseline in HbA1c after 12 weeks. The differences of adjusted mean changes from baseline in HbA1c at week 12 are −0.87% for LI 5 mg vs. PBO (p<0.0001) and −0.88% for LI 10 mg vs. PBO (p<0.0001). Proportions of patients achieving HbA1c <7.0% after 12 wks are 26.4% for LI 5 mg and 35.7% for LI 10 mg vs. 10.0% for PBO. Proportions of patients whose HbA1c levels lower by at least 0.5% are 57.2% with LI 5 mg, 59.9% with LI 10 mg, and 8.8% with PBO. Both LI 5 mg and 10 mg show statistically significant difference compared with PBO (p<0.0001). FPG is significantly improved with both LI 5 and 10 mg compared to PBO: after 12 weeks, the differences of adjusted mean changes from baseline are −19.7 mg/dL for LI 5 mg vs. PBO (p<0.0001) and −20.4 mg/dL for LI 10 mg vs. PBO (p<0.0001). As indicated by changes in the proinsulin/insulin ratio (LI 5 mg, p=0.0065; LI 10 mg, p=0.0004), LI also significantly improves insulin secretion. The proportion of patients experiencing at least one adverse event (AE) is comparable among the three groups (56.0% LI 5 mg, 53.1% LI 10 mg and 56.3% PBO). Of those; 9.4%, 8.8% and 10.0%, respectively, are assessed as being drug-related. There are no investigator-defined hypoglycemic episodes. Body weight is unchanged with both LI 5 mg and 10 mg, −0.39 and −0.06 kg, respectively, which is not sigificantly different vs. PBO (−0.04 kg).
  • Conclusion: Linagliptin demonstrates a significant and clinically meaningful improvement in glycemic control, reflected in changes in HbA1c and FPG in Japanese patients with T2DM. Both linagliptin 5 and 10 mg doses have similar efficacy in lowering HbA1c and are well tolerated within this population. 5 mg linagliptin is the therapeutic dose in Japanese patients, which is identical to the therapeutic dose in Caucasians.
  • Example 16 Linagliptin Provides Superior Glycemic Control Compared to Voglibose as Monotherapy in Japanese Patients With Type 2 Diabetes
  • A multi-center, 26-week, randomized, double-blind, active-controlled, parallel group Study compares the efficacy and safety of the DPP-4 inhibitor linagliptin (LI) vs. the α-glucosidase inhibitor voglibose (VB) in drug naïve or previously treated Japanese patients (pts) with Type 2 diabetes mellitus (T2DM) (baseline HbA1c 7.0-10.0% if drug naïve, 7.0-9.0% if previously treated with an oral antihyperglycemic drug (OAD)).
  • Following a 2-week PBO run-in, patients are randomized to LI 5 (n=159) or 10 mg qd (n=160), or VB (0.2 mg tid; n=162). Any previous OAD treatment is stopped 2 weeks prior to run-in. Mean baseline [SD] characteristics and demographics (HbA1c, 8.01% [0.68]; fasting plasma glucose (FPG), 163.5 mg/dL [32.4]; BMI, 24.97 kg/m2 [3.86]; age, 60.0 yrs [9.7]) are similar across groups. The primary endpoint is the change from baseline in HbA1c after 26 weeks. The differences of adjusted mean changes from baseline in HbA1c at week 26 are −0.32% for LI 5 mg vs. VB (p=0.0003) and −0.39% for LI 10 mg vs. VB (p<0.0001). Proportions of patients achieving HbA1c <7.0% after 26 weeks are 30.2% for LI 5 mg and 34.4% for LI 10 mg vs. 22.2% for VB. Proportions of patients whose HbA1c level lowered by ≥0.5% are 57.2% and 53.5% for LI 5 and 10 mg, vs. 37.7% for VB. FPG is significantly improved with both LI 5 and 10 mg compared to VB: the differences of adjusted mean changes from baseline are −6.9 mg/dL for LI 5 mg vs. VB (p=0.02) and −9.8 mg/dL for LI 10 mg vs. VB (p=0.0015). Both LI 5 mg and 10 mg show a significant decrease of HbA1c in patients previously treated with 1 OAD compared with VB (p=0.003 and p=0.0011, respectively). The occurrence of ≥1 adverse event (AE) is comparable between groups (72.3% LI 5 mg, 77.5% LI 10 mg and 71.6% VB). Of the AEs, 11.3%, 10.6% and 18.5%, respectively, are assessed as drug related. Drug-related gatsrointstinal disorders are more common in the VB (14.2%) than LI (8.2% 5 mg; 8.1% 10 mg) groups.ln the VB group, 1 hypoglycemic episode is reported vs. none in the LI groups.
  • Conclusion: Linagliptin monotherapy demonstrates greater efficacy than VB for improving glycemic control in Japanese patients with T2DM. Both linagliptin 5 mg and linagliptin 10 mg have comparable efficacy and show statistically significant decreases in HbA1c and FPG from baseline compared with VB after 26 weeks. Linagliptin is well tolerated in Japanese patients with T2DM compared to VB, with less gastrointestinal AEs, and may provide a valuable addition to the therapies available to this population. 5 mg linagliptin is the therapeutic dose in Japanese patients, which is identical to the therapeutic dose in Caucasians.
  • Example 17 Linagliptin Restores β-Cell Function and Survival in Human Isolated Islets
  • Studies in diabetic animal models show that dipeptidyl peptidase-4 (DPP-4) inhibitors reverse hyperglycemia and increase β-cell mass. Here, the role of linagliptin, a DPP-4 inhibitor on human β-cell function is investigated: Human isolated islets are exposed to increased glucose concentrations (5.5-33.3 mM), 0.5 mM palmitic acid, the mixture of 2 ng/mL IL-1β or 1,000 U/mL IFN-γ for 4 days or 50 μM H2O2 for 8 h. Islets are pre-treated with 500 ng/mL Interleukin-1 Receptor Antagonist (IL-1Ra, which has been shown to restore β-cell function), 100 nM linagliptin or solvent for 1 h before exposure to the diabetic stimuli and during the whole 4-day treatment period. At control conditions, islets secrete 3.8-fold more insulin at 16.7 mM than at 2.8 mM glucose. In contrast, stimulatory index is 1.9- and 2.4-fold decreased when islets are exposed to 11.1 mM and 33.3 mM glucose (P<0.05). Exposure of the islets to palmitate, cytokine mixture or H2O2 resulted in a 2.1-, 2.2- and 1.9-fold reduction of glucose stimulated insulin secretion (GSIS), respectively (P<0.05). Linagliptin significantly restores β-cell function at all conditions (1.9-, 2.5-, 3.3-, 1.9- and 3.7-fold increase in GSIS at 11.1 or 33.3 mM glucose, palmitic acid, cytokines or H2O2, P<0.05). IL-1Ra is similarly effective in restoring β-cell function at conditions of high glucose, palmitic acid and cytokines, but IL-1Ra fails to restore β-cell function at oxidative stress conditions induced by H2O2 treatment. Since loss of function is mediated by oxidative stress, the nitrotyrosine concentration is measured in islet lysates. Nitrotyrosine levels are highly elevated in human islets under all diabetic conditions (13-, 14-, 6-, 14- and 8-fold increased at 11.1 or 33.3 mM glucose, palmitic acid, cytokines or H2O2, P<0.05), while no elevated nitrotyrosine production is observed in islets treated with linagliptin.
  • In summary, it is shown that the DPP-4 inhibitor linagliptin has comparable protective effects on gluco-, lipo- and cytokinetoxicity as IL-1Ra and, in addition, could improve β-cell function under oxidative stress conditions and blocks apoptosis (induced by H2O2 treatment). The study provides evidence of a direct protective effect of linagliptin on β-cell survival and insulin secretion.
  • Example 18 Chronic Renal Disease Does Not Change the Pharmacokinetics of Linagliptin But Increases Exposure of Sitagliptin and Alogliptin in Rats
  • Renal impairment is a frequent complication of T2DM. The effect of chronic renal disease on the pharmacokinetics of dipeptidyl peptidase-4 inhibitors (linagliptin, sitagliptin, alogliptin) in a rat model of chronic renal insufficiency (5/6 nephrectomy, 5/6N) is investigated: Eight weeks after surgery rats are treated orally with inhibitors for 4 days. 5/6N causes a highly significantly (P<0.001) decrease of glomerular filtration rate measured by creatinin clearance (sham: 2510±210 mL/24 h; 5/6N: 1665±104.3 mL/24 h) and increases cystatin C levels (sham: 700±35.7 ng/mL; 5/6N: 1434±77.6 ng/mL). Tubular function is significantly (P<0.001) impaired as evidenced by plasma neutrophil gelatinase-associated lipocalin (NGAL), (sham: 286±23 ng/ml; 5/6N: 680±56.3 ng/ml) and β2 microglobulin (sham: 20.4±2.4 μg/mL; 5/6N: 33.3±1.34 μg/mL). DPP-4 activity is comparable among groups.
  • Administration of linagliptin (0.5 and 7 μmol/kg) to 5/6N rats shows no significant change in AUC(0-∞): sham: 316±54.7 nmol*h/L; 5/6N: 257±21.54 nmol*h/L; P=0.771 and sham: 1252±372 nmol*h/L; 5/6N: 748±74.5 nmol*h/L; P=0.284, respectively. In contrast, both sitagliptin and alogliptin (7 μmol/kg) have significantly (P=0.0001 and P=0.039) higher (41% and 28%) AUC(0-∞): sitagliptin sham: 3690±103 nmol* h/L; 5/6N: 6238±423 nmol*h/L and alogliptin sham: 1772±225 nmol*h/L; 5/6N: 2445±166 nmol*h/L). Furthermore, no correlation of markers of tubular and glomerular functions with linagliptin AUC is observed. In contrast, sitagliptin significantly correlate with creatinin clearance (r2=0.374, P<0.05), cystatin C (r2=0.499, P<0.01), NGAL (r2=0.604, P<0.01) and β2 microglobulin (r2=0.543, P<0.01). Alogliptin correlates less significantly with cystatin C (r2=0.376, P<0.05) and β2 microglobulin (r2=0.391, P<0.05) but not with creatinin clearance and NGAL.
  • These results demonstrate that renal impairment does not affect the pharmacokinetics of linagliptin whereas it increases the exposure of sitagliptin and alogliptin. Therefore, in contrast to sitagliptin and alogliptin, linagliptin may not have to be dose-adjusted in patients with T2DM and renal impairment or diabetic nephopathy.
  • Further, linagliptin significantly inhibits mRNA expression of profibrotic factors, such as TGF-pβ1, T1MP-1 and collagen (Col3alpha1) in the heart of uremic rats, which factors are tissue fibrosis markers of cardiac fibrosis and are increased in uremic heart. Characteristic cardiomyopathy with intestinal expansion and fibrosis develops often in uremia. Thus, these antifibrotic properties of DPP-4 inhibitors may be used for the treatment of cardiac and renal injury, uremic heart, cardiac fibrosis and/or cardiomyopathy with intestinal expansion and fibrosis associated with uremia in patients with type 2 diabetes. The antifibrotic action of linagliptin can be an additional benefit for patients with chronic kidney and/or heart diseases that often accompany type 2 diabetes.
  • Example 19 Linagliptin Improves Hepatic Steatosis in Rodent Models
  • Hepatic steatosis is a hallmark of patients with Type 2 diabetes and non-alcoholic fatty liver disease (NAFLD). Linagliptin is a selective and non-renal excreted inhibitor of dipeptidyl peptidase-4 (DPP-4).
  • In a model of diet-induced obesity (D10, fed for 2 and 3 months), the effect of 4 weeks therapy with linagliptin (3 and 30 mg/kg/day, n=10) is investigated. Liver lipid content is detected by magnetic resonance spectroscopy (MRS) in vivo and by analysis of liver triglycerides ex vivo. Linagliptin inhibits DPP-4 activity significantly (P<0.001) by 67% to 80% and 79% to 89% (3 and 30 mg/kg/day, respectively) compared to controls. Blood glucose levels following an OGTT (AUC) are significantly (P<0.01) decreased ranging from 16% to 20% (3 mg/kg/day) and 20% to 26% (30 mg/kg/day). Likewise, liver fat content (MRS detection) is significantly reduced. Changes in liver fat content are visible as early as 2 weeks on treatment. The correlation between liver lipid content as measured by MRS and hepatic triglyceride levels as measured ex vivo is r2=0.565 (P<0.0001). Furthermore, ob/ob mice are analyzed after 14 days of linagliptin treatment (3 mg/kg/day or control) and blinded histological scoring is performed (severity and grade of fat content, markers of inflammation). DPP-4 activity is inhibited by 80% and blood glucose AUC reduction is 25% (P<0.05). The histological score reveals less hepatic steatosis and inflammation in the linagliptin group (2.2±0.13, n=9, P<0.01) versus control (3±0.18, n=10).
  • In conclusion, linagliptin significantly reduces liver fat content and histological NAFLD in a high fat diet model. Linagliptin reverses liver triglyceride content and hepatic steatosis (with greater therapeutic impact when hepatic steatosis is more pronounced). The reversal of hepatic steatosis supports the use of linagliptin in patients with Type 2 diabetes as well as liver-associated diseases (NAFLD).
  • Example 20 Linagliptin Functionally Counteracts a Dysregulation in DPP-4 Expression in Diabetes-Impaired Wounds
  • Impaired wound healing is a major complication of diabetes mellitus. The dipeptidyl peptidase-4 (DPP-4) inhibitor linagliptin improves wound healing (as shown in ob/ob mice). The impact of linagliptin on inflammatory markers in wounded skin is examined and a rationale for the beneficial action of linagliptin on wound healing is provided:
  • Wounds of linagliptin (3 mg/kg/day) and mock-treated ob/ob mice for the inflammatory markers COX-2 and MIP by RNase protection assays are investigated with no significant differences. Furthermore, linagliptin does not increase the number of apoptotic infiltrating F4/80-positive macrophages. Therefore, the expression of DPP-4 in the skin of diabetic and non-diabetic animals is assessed. Immunohisto-chemistry (IHC) and immunoblots reveal a strong expression of DPP-4 in skin from healthy and diabetic (ob/ob) mice and keratinocytes as the major cellular source of the enzyme. In line, the localization of DPP-4 protein in the skin nicely correlates with whole body autoradiography obtained after [3H]-labelled linagliptin treatment. Analyzing DPP-4 expression in mice upon full-thickness excisional wounding it is found that in healthy mice, DPP-4 protein expression declines over 3 days after injury and the enzyme remains absent in the late phase of repair. Interestingly, skin injury leads to a strong down-regulation of DPP-4 expression in proliferating wound margin keratinocytes (IHC). In contrast, in acute wounds of diabetic mice any DPP-4 expression can not be observed. DPP-4 protein, however, is expressed in the late phase of wound repair. The inverse regulation of DPP-4 protein in diabetic versus non-diabetic skin provides a functional basis of the positive action of linagliptin in wound healing processes. Thus, improvement of the wound healing process mediated by a suitable DPP-4 inhibitor, such as linagliptin, depends on the compensation (inhibition) of a dysregulated DPP-4 in diabetic wounds rather than on the anti-glycemic or immunomodulatory effects thereof. Thus, a DPP-4 inhibitor being suitable for improving wound healing is such a DPP-4 inhibitor which can effectively bind to DPP-4 in the skin, e.g. to dysregulated DPP-4 in diabetic wounds, preferably in its therapeutic dose level.
  • Furthermore in this context, a DPP-4 inhibitor being suitable for improving wound healing, particularly in a type 2 diabetes patient, is such a DPP-4 inhibitor which can be applied topically to wounds, e.g. comprised in wound dressings or patches or creams or ointments. Thus, the present invention further provides topical devices for wounds, such as e.g. wound dressings or patches, comprising linagliptin and, optionally, one or more pharmaceutically acceptable carriers and/or excipients.
  • Example 21 Association Study (Genotyping TCF7L2, Treatment Response)
  • The polymorphisms and variants of the gene TCF7L2 as depicted in the Table i can be analyzed as described in the following procedure:
  • TABLE i
    Gene, variant nucleotides and rs numbers.
    Gene variant nucleotide rs number
    TCF7L2 c.382-41435 C > T rs7903146
    c.483 + 9017 G > T, rs12255372
    c.382-22060 A > G rs10885406
    c.1102 C > G rs731788
  • Samples
  • Patients' DNA samples (conc.: 50 ng/μl) in 96-well-plates are used for the analytical methods applied.
  • Genotyping by Direct Sanger Sequencing
  • Using gDNA as a template, locus specific DNA fragments are amplified by polymerase chain reaction (PCR).
  • PCR is carried out using an ABI BioRad® Tetrad PCR System. Quality of the PCR products is analyzed by agarose gel electrophoresis The purified PCR-products are used as templates in sequencing reactions According to the chain terminating methodology of Sanger et al. (1977), the analysis of DNA sequence is based on the termination of a growing DNA strand due to incorporation of a dye-labeled 2′,3′-Dideoxyribonucleotidetriphosphate (ddNTP) by the DNA polymerase. Purified sequencing products are analyzed using an ABI PRISM® 3730 Genetic Analyzer.
  • Sequencing data are generated using the original ABI Software. The subsequent KB-basecalling as well as the assembly is performed using the Staden Software Package. KB-basecalling assigns quality values to all called bases of automated sequencer traces using KB-basecaller error probabilities. These quality values are used during assembling the single reads and are the basic requirement for calculating the sequence accuracy (Applied Biosystems, 3730/3730xl/DNA Analyzer Sequencing Analysis Software Training).
  • A quality value (q) of 20 corresponds to an error probability (ep) of 1/100, a value of 30 to an ep of 1/1000 and so on. In the assembly phase those values are set against each other. In general sequencing is continued until each consensus base has a quality value (q) of 50 or more. This corresponds to an error probability (ep) of 1/100000. Due to the fact that most of the consensus bases have an even higher quality score than the minimal one, the calculated cumulative error probability for the finished sequence is again significantly lower. Sequencing data are uploaded and analyzed using the software seqpatient from jsi-medical systems (version Seq Pilot 3.3.2, JSI medical systems GmbH, Friedhofstr. 5, 77971 Kippenheim, Germany).
  • Only traces that fulfill internal quality aspects are processed for further genotype analyses. Genotyping is carried out through the analysis of single polymorphisms rather than the analysis of the entire gene. Therefore genotyping results refer only to the variant positions depicted in Table i.
  • Genotyping by TaqMan PCR
  • The TaqMan® technology comprises amplification of a PCR fragment with simultaneous detection of the degradation of a labelled probe. Probes are labelled at both ends with an allele-specific dye and a quencher. During the amplification reaction, the specifically hybridized probe is displaced by the DNA polymerase. This displacement occurs either as degradation through the 5′ exonuclease activity of the polymerase in the case of a perfect match with the probe, or without degradation in the case of a mismatch. Upon degradation, the quencher and dye are separated and the fluorescence signal increased. An increase in the fluorescence signal is indicative for the presence of the respective allele. Fluorescence signals are recorded with the ABI PRISM 7700 system (Applied Biosystems).
  • In detail, a master mix is prepared containing all components for PCR reaction and aliquoted in the appropriate number of wells. Subsequently, DNA is added to each well according to the plate layout; except for no-template control (NTC).
  • AB assay ID (rs7903146) C_29347861_10
    SNP context sequence:
    (SEQ ID NO: 1)
    TAGAGAGCTAAGCACTTTTTAGATA[C/T]TATATAATTTAATTGCCG
    TATGAGG
  • The mastermix per sample contains:
  • Nuclease-free water 0.25 μl
    2x PCR MasterMix  2.5 μl
    20x Primer/Probe Mix 0.25 μl
    DNA [10 ng/μl]  2 μl
    In total:  5 μl
  • The cycling conditions are:
  • 95° C. 10 min.
    95° C. 15 sec. {close oversize brace} 50 cycles
    60° C. 90 sec.
  • The TaqMan® pre- and post-reads of the AD are performed on the TaqMan® 7900HT Fast Real System. The SDS software V2.3 calculates the fluorescence measurements made during the plate read and plots Rn values based on the signals from each well. Using the software, it is determined which SNP alleles are present in each sample. NTC should be given as not determined.
  • Statistical Analyses
  • To assess the homogeneity of the treatment effect on the change from baseline of HbA1c after 24 weeks in the genotype subgroups defined by TCF7L2 SNP rs7903146 genotypes an analysis of covariance (ANCOVA) model including the treatment interaction with the covariate genotype is applied for pooled data over four studies. The statistical model includes ‘Treatment’, ‘Genotype’, ‘Study’, ‘Wash-Out-Period for prior oral antidiabetic drugs (yes/no)’, ‘Race’, as well as the interaction term ‘Treatment*Genotype’ as fixed effects and ‘HbA1c baseline’ as a linear covariate. The ANCOVA model provides estimates for the mean change from baseline in HbA1c after 24 weeks of therapy for the different genotypes taking baseline clinical and demographic information into account.
  • Model based pair-wise comparisons between wild-type homozygous (genotype CC) and heterozygous (genotype CT) or rare homozygous (genotype TT) individuals on linagliptin or combination treatment (linagliptin+pioglitazone, linagliptin+metformin, linagliptin+metformin+a sulphonylurea) are performed.
  • Additionally the results of the corresponding ANCOVA models without ‘Genotype’ and ‘Treatment*Genotype’ fixed effects are given for the whole patient population of the studies (full analysis set, FAS) as well as for the subpopulation for which genetic analyses are performed (full analysis set for pharmacogenetic analyses, FASG) to demonstrate comparability of the observed effects.
  • The statistical evaluation is prepared using the software packages SAS Version 9.2 (SAS Institute Inc., Cary, N.C., USA) and S-PLUS® 8.0 (Insightful Corp., Seattle, Wash., USA).
  • Results are shown as point estimates and 95% confidence intervals for the mean change in HbA1c from baseline [%] after 24 weeks as estimated by ANCOVA models. The results are given for the whole patient population of the studies (full analysis set, FAS), for the subpopulation for which genetic analyses are performed (full analysis set for pharmacogenetic analyses, FASG), as well as for the subgroups defined by genotype (CC, CT, TT) of this subpopulation. The numbers of patients for placebo control and linagliptin treatment are given in braces.
  • Point estimates and 95% confidence intervals for the differences in changes in HbA1c from baseline [%] for the comparison of between wild-type homozygous (genotype CC) and heterozygous (genotype CT) or rare homozygous (genotype TT) individuals on linagliptin treatment or combination treatment (linagliptin+pioglitazone, linagliptin+metformin, linagliptin+metformin+a sulphonylurea) are shown as well. They result in a statistically significant difference between TT and CC (p value=0.0192). (Other pairwise comparisons: CT vs. CC: p=0.4359; CT vs. TT: p=0.0712).
  • This indicates a significant association between the wild-type homozygous genotype and lower HbA1c on treatment.
  • EXAMPLES OF FORMULATIONS
  • The following examples of formulations, which may be obtained analogously to methods known in the art, serve to illustrate the present invention more fully without restricting it to the contents of these examples. The term “active substance” denotes one or more compounds according to the invention, i.e. denotes a DPP-4 inhibitor or a second or third antidiabetic compound according to this invention or a combination of two or three of said active ingredients, for example selected from the combinations as listed in the Table 1 or 2. Additional suitable formulations for the DPP-4 inhibitor linagliptin may be those formulations disclosed in the application WO 2007/128724, the disclosure of which is incorporated herein in its entirety. Additional suitable formulations for the other DPP-4 inhibitors may be those formulations which are available on the market, or formulations described in the patent applications cited above in paragraph “background of the invention”, or those described in the literature, for example as disclosed in current issues of “Rote Liste®” (Germany) or of “Physician's Desk Reference”.
  • Example 1 Dry Ampoule Containing 75 mg of Active Substance per 10 ml
  • Composition:
  • Active substance 75.0 mg
    Mannitol 50.0 mg
    water for injections ad 10.0 ml
  • Preparation:
  • Active substance and mannitol are dissolved in water. After packaging the solution is freeze-dried. To produce the solution ready for use, the product is dissolved in water for injections.
  • Example 2 Dry Ampoule Containing 35 mg of Active Substance per 2 ml
  • Composition:
  • Active substance  35.0 mg
    Mannitol 100.0 mg
    water for injections ad 2.0 ml
  • Preparation:
  • Active substance and mannitol are dissolved in water. After packaging, the solution is freeze-dried.
  • To produce the solution ready for use, the product is dissolved in water for injections.
  • Example 3 Tablet Containing 50 mg of Active Substance
  • Composition:
  • (1) Active substance 50.0 mg
    (2) Mannitol 98.0 mg
    (3) Maize starch 50.0 mg
    (4) Polyvinylpyrrolidone 15.0 mg
    (5) Magnesium stearate  2.0 mg
    215.0 mg 
  • Preparation:
  • (1), (2) and (3) are mixed together and granulated with an aqueous solution of (4). (5) is added to the dried granulated material. From this mixture tablets are pressed, biplanar, faceted on both sides and with a dividing notch on one side.
  • Diameter of the tablets: 9 mm.
  • Example 4 Tablet Containing 350 mg of Active Substance
  • Preparation:
  • (1) Active substance 350.0 mg
    (2) Mannitol 136.0 mg
    (3) Maize starch  80.0 mg
    (4) Polyvinylpyrrolidone  30.0 mg
    (5) Magnesium stearate  4.0 mg
    600.0 mg
  • (1), (2) and (3) are mixed together and granulated with an aqueous solution of (4). (5) is added to the dried granulated material. From this mixture tablets are pressed, biplanar, faceted on both sides and with a dividing notch on one side.
  • Diameter of the tablets: 12 mm.
  • Example 5 Capsules Containing 50 mg of Active Substance
  • Composition:
  • (1) Active substance  50.0 mg
    (2) Dried maize starch  58.0 mg
    (3) Mannitol  50.0 mg
    (4) Magnesium stearate  2.0 mg
    160.0 mg
  • Preparation:
  • (1) is triturated with (3). This trituration is added to the mixture of (2) and (4) with vigorous mixing. This powder mixture is packed into size 3 hard gelatin capsules in a capsule filling machine.
  • Example 6 Capsules Containing 350 mg of Active Substance
  • Composition:
  • (1) Active substance 350.0 mg
    (2) Dried maize starch  46.0 mg
    (3) Mannitol  30.0 mg
    (4) Magnesium stearate  4.0 mg
    430.0 mg
  • Preparation:
  • (1) is triturated with (3). This trituration is added to the mixture of (2) and (4) with vigorous mixing. This powder mixture is packed into size 0 hard gelatin capsules in a capsule filling machine.

Claims (6)

1. A method for improving glycemic control compared with placebo in a type 2 diabetes patient, said method comprising administering linagliptin 5 mg qd for at least 24 weeks, as monotherapy wherein the patient is a drug naïve type 2 diabetes patient, or as add-on combination therapy with metformin wherein the patient is a type 2 diabetes patient with insufficient glycemic control on metformin, or
as add-on combination therapy with metformin and a sulfonylurea wherein the patient is a type 2 diabetes patient with insufficient glycemic control on the combination of metformin and a sulfonylurea.
2. The method according to claim 1, wherein improving glycemic control is reducing HbA1c, FPG and/or PPG.
3. The method according to claim 1, wherein linagliptin 5 mg qd is administered for at least 24 weeks as monotherapy wherein the patient is a drug naïve type 2 diabetes patient.
4. The method according to claim 1, wherein linagliptin 5 mg qd is administered for at least 24 weeks as add-on combination therapy with metformin wherein the patient is a type 2 diabetes patient with insufficient glycemic control on metformin.
5. The method according to claim 1, wherein linagliptin 5 mg qd is administered for at least 24 weeks as add-on combination therapy with metformin and a sulfonylurea wherein the patient is a type 2 diabetes patient with insufficient glycemic control on the combination of metformin and a sulfonylurea.
6. The method according to claim 1, wherein the patient is of TCF7L2 rs7903146 CC, CT or TT genotype.
US16/528,934 2009-11-27 2019-08-01 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin Abandoned US20200046713A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/528,934 US20200046713A1 (en) 2009-11-27 2019-08-01 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
EP09177418 2009-11-27
EP09177418.2 2009-11-27
EP10166714.5 2010-06-21
EP10166714 2010-06-21
PCT/EP2010/068349 WO2011064352A1 (en) 2009-11-27 2010-11-26 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin
US201213511771A 2012-08-15 2012-08-15
US15/235,575 US10092571B2 (en) 2009-11-27 2016-08-12 Treatment of genotyped diabetic patients with DPP-IV inhibitors such as linagliptin
US16/121,885 US20190000855A1 (en) 2009-11-27 2018-09-05 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin
US16/528,934 US20200046713A1 (en) 2009-11-27 2019-08-01 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/121,885 Continuation US20190000855A1 (en) 2009-11-27 2018-09-05 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin

Publications (1)

Publication Number Publication Date
US20200046713A1 true US20200046713A1 (en) 2020-02-13

Family

ID=43466576

Family Applications (4)

Application Number Title Priority Date Filing Date
US13/511,771 Active US9457029B2 (en) 2009-11-27 2010-11-26 Treatment of genotyped diabetic patients with DPP-IV inhibitors such as linagliptin
US15/235,575 Active US10092571B2 (en) 2009-11-27 2016-08-12 Treatment of genotyped diabetic patients with DPP-IV inhibitors such as linagliptin
US16/121,885 Abandoned US20190000855A1 (en) 2009-11-27 2018-09-05 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin
US16/528,934 Abandoned US20200046713A1 (en) 2009-11-27 2019-08-01 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US13/511,771 Active US9457029B2 (en) 2009-11-27 2010-11-26 Treatment of genotyped diabetic patients with DPP-IV inhibitors such as linagliptin
US15/235,575 Active US10092571B2 (en) 2009-11-27 2016-08-12 Treatment of genotyped diabetic patients with DPP-IV inhibitors such as linagliptin
US16/121,885 Abandoned US20190000855A1 (en) 2009-11-27 2018-09-05 Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin

Country Status (15)

Country Link
US (4) US9457029B2 (en)
EP (2) EP3646859A1 (en)
JP (2) JP2013512229A (en)
KR (5) KR20190071840A (en)
CN (2) CN107115530A (en)
AU (1) AU2010323068B2 (en)
BR (1) BR112012012641A2 (en)
CA (1) CA2782179C (en)
CL (1) CL2012001337A1 (en)
EA (1) EA034869B1 (en)
ES (1) ES2760917T3 (en)
IL (1) IL219014A0 (en)
MX (2) MX364651B (en)
NZ (1) NZ599298A (en)
WO (1) WO2011064352A1 (en)

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7407955B2 (en) 2002-08-21 2008-08-05 Boehringer Ingelheim Pharma Gmbh & Co., Kg 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
US7501426B2 (en) 2004-02-18 2009-03-10 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthines, their preparation and their use as pharmaceutical compositions
DE102004054054A1 (en) 2004-11-05 2006-05-11 Boehringer Ingelheim Pharma Gmbh & Co. Kg Process for preparing chiral 8- (3-amino-piperidin-1-yl) -xanthines
DE102005035891A1 (en) 2005-07-30 2007-02-08 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8- (3-amino-piperidin-1-yl) -xanthines, their preparation and their use as pharmaceuticals
EP1852108A1 (en) 2006-05-04 2007-11-07 Boehringer Ingelheim Pharma GmbH & Co.KG DPP IV inhibitor formulations
PE20080251A1 (en) 2006-05-04 2008-04-25 Boehringer Ingelheim Int USES OF DPP IV INHIBITORS
EP2540725A1 (en) 2006-05-04 2013-01-02 Boehringer Ingelheim International GmbH Polymorphs of 1-((4-Methyl-chinazolin-2-yl)methyl)-3-methyl-7-(2-butin-1-yl)-8-(3-(R)-amino-piperidin-1-yl)xanthin
PE20090938A1 (en) 2007-08-16 2009-08-08 Boehringer Ingelheim Int PHARMACEUTICAL COMPOSITION INCLUDING A BENZENE DERIVATIVE SUBSTITUTED WITH GLUCOPYRANOSIL
PE20140960A1 (en) 2008-04-03 2014-08-15 Boehringer Ingelheim Int FORMULATIONS INVOLVING A DPP4 INHIBITOR
KR20190016601A (en) 2008-08-06 2019-02-18 베링거 인겔하임 인터내셔날 게엠베하 Treatment for diabetes in patients inappropriate for metformin therapy
UY32030A (en) 2008-08-06 2010-03-26 Boehringer Ingelheim Int "TREATMENT FOR DIABETES IN INAPPROPRIATE PATIENTS FOR THERAPY WITH METFORMIN"
RU2011113823A (en) 2008-09-10 2012-10-20 БЕРИНГЕР ИНГЕЛЬХАЙМ ИНТЕРНАЦИОНАЛЬ ГмбХ (DE) COMBINED THERAPY FOR THE TREATMENT OF DIABETES AND RELATED CONDITIONS
US20200155558A1 (en) 2018-11-20 2020-05-21 Boehringer Ingelheim International Gmbh Treatment for diabetes in patients with insufficient glycemic control despite therapy with an oral antidiabetic drug
UY32177A (en) * 2008-10-16 2010-05-31 Boehringer Ingelheim Int TREATMENT OF DIABETES IN PATIENTS WITH INSUFFICIENT GLUCEMIC CONTROL TO WEIGHT THERAPY WITH DRUG, ORAL OR NOT, ANTIDIABÉTICO
JP2012512848A (en) 2008-12-23 2012-06-07 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Salt forms of organic compounds
TW201036975A (en) 2009-01-07 2010-10-16 Boehringer Ingelheim Int Treatment for diabetes in patients with inadequate glycemic control despite metformin therapy
NZ594487A (en) 2009-02-13 2013-11-29 Boehringer Ingelheim Int Pharmaceutical composition comprising a sglt2 inhibitor, a dpp-iv inhibitor and optionally a further antidiabetic agent and uses thereof
NZ599298A (en) 2009-11-27 2014-11-28 Boehringer Ingelheim Int Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin
SI2796457T1 (en) 2009-11-27 2016-10-28 Genzyme Corporation Genz 112638 zur behandlung von gaucher- oder fabry-erkrankung in kombinations-therapie
JP6034781B2 (en) * 2010-05-05 2016-11-30 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Combination therapy
EP3725325B1 (en) 2010-06-24 2023-05-31 Boehringer Ingelheim International GmbH Diabetes therapy
AU2013204252B2 (en) * 2010-11-15 2016-05-26 Boehringer Ingelheim International Gmbh Vasoprotective and cardioprotective antidiabetic therapy
US9034883B2 (en) * 2010-11-15 2015-05-19 Boehringer Ingelheim International Gmbh Vasoprotective and cardioprotective antidiabetic therapy
AU2016202261B2 (en) * 2010-11-15 2017-11-30 Boehringer Ingelheim International Gmbh Vasoprotective and cardioprotective antidiabetic therapy
TR201010683A1 (en) * 2010-12-21 2012-07-23 Sanovel İlaç San. Ve Ti̇c. A.Ş. Vildagliptin formulations.
UY33937A (en) 2011-03-07 2012-09-28 Boehringer Ingelheim Int PHARMACEUTICAL COMPOSITIONS CONTAINING DPP-4 AND / OR SGLT-2 AND METFORMIN INHIBITORS
EP2731610A4 (en) * 2011-07-12 2014-12-10 Ipca Lab Ltd Pharmaceutical combination
DK2731947T3 (en) 2011-07-15 2019-04-23 Boehringer Ingelheim Int SUBSTITUTED DIMERIC QUINAZOLINE DERIVATIVE, PREPARATION AND USE thereof IN PHARMACEUTICAL COMPOSITIONS FOR TREATMENT OF TYPE I AND TYPE II DIABETES
US20130172244A1 (en) * 2011-12-29 2013-07-04 Thomas Klein Subcutaneous therapeutic use of dpp-4 inhibitor
WO2013134546A1 (en) 2012-03-07 2013-09-12 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US9555001B2 (en) 2012-03-07 2017-01-31 Boehringer Ingelheim International Gmbh Pharmaceutical composition and uses thereof
EP2849755A1 (en) 2012-05-14 2015-03-25 Boehringer Ingelheim International GmbH A xanthine derivative as dpp -4 inhibitor for use in the treatment of podocytes related disorders and/or nephrotic syndrome
TWI588153B (en) * 2012-05-18 2017-06-21 中國醫藥大學 Polypeptides, nucleic acid molecule encoding polypeptides, and uses of polypeptides
WO2013174768A1 (en) * 2012-05-24 2013-11-28 Boehringer Ingelheim International Gmbh A xanthine derivative as dpp -4 inhibitor for use in the treatment of autoimmune diabetes, particularly lada
WO2013174767A1 (en) 2012-05-24 2013-11-28 Boehringer Ingelheim International Gmbh A xanthine derivative as dpp -4 inhibitor for use in modifying food intake and regulating food preference
EP2854824A1 (en) * 2012-05-25 2015-04-08 Boehringer Ingelheim International GmbH Use of keratinocytes as a biologically active substance in the treatment of wounds, such as diabetic wounds, optionally in combination with a dpp-4 inhibitor
WO2014140284A1 (en) * 2013-03-15 2014-09-18 Boehringer Ingelheim International Gmbh Use of linagliptin in cardio- and renoprotective antidiabetic therapy
KR101598612B1 (en) * 2013-08-29 2016-02-26 재단법인 아산사회복지재단 Composition for prophylaxis or treatment of vascular or cardiac valvular calcification comprising dipeptidyl peptidase-4 inhibitor
JP6615109B2 (en) 2014-02-28 2019-12-04 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Medical use of DPP-4 inhibitors
WO2016034710A1 (en) * 2014-09-05 2016-03-10 Sanovel Ilac Sanayi Ve Ticaret A.S. Pharmaceutical combinations of sitagliptin
US10821110B2 (en) * 2014-10-17 2020-11-03 Hyundai Pharm Co., Ltd. Composite preparation, containing novel 3-(4--(benzyloxy)phenyl)hex-4-inoic acid derivative and another active ingredient, for preventing or treating metabolic diseases
CN104894250B (en) * 2015-05-26 2018-02-02 北京大学人民医院 The detection method of rs492594 genotype and its application
CA3022202A1 (en) 2016-06-10 2017-12-14 Boehringer Ingelheim International Gmbh Combinations of linagliptin and metformin
CN109996545A (en) * 2016-11-15 2019-07-09 株式会社Lg化学 For treating the medicinal composition of diabetes B and diabetic keratopathy dyslipidemia
WO2018142334A1 (en) * 2017-02-03 2018-08-09 Glenmark Pharmaceuticals Limited Formulations comprising oxalate salts of teneligliptin and solvates thereof
AU2019211322A1 (en) 2018-01-23 2020-07-23 Gila Therapeutics, Inc. Peptide YY pharmaceutical formulations, compositions, and methods
US11730742B2 (en) 2018-03-09 2023-08-22 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences Pharmaceutical use of thiophene [3,2-d] pyrimidine-4-ketone compound
KR102020031B1 (en) * 2019-03-15 2019-09-09 한국생명공학연구원 Pharmaceutical composition for prevention and treatment of non-alcoholic fatty liver disease comprising transcription factor 7-like 2 as effective component
CN114423461A (en) * 2019-07-09 2022-04-29 杰尼西斯制药有限公司 Combination of
KR102289381B1 (en) * 2020-03-17 2021-08-17 주식회사 대웅테라퓨틱스 Pharmaceutical composition for prevention and treatment of hyperlipidemia and diabetes
AU2021362692A1 (en) * 2020-10-14 2023-05-25 Brightseed, Inc. Methods for reversing hepatic steatosis

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070281940A1 (en) * 2006-05-04 2007-12-06 Klaus Dugi Uses of dpp-iv inhibitors

Family Cites Families (491)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2056046A (en) 1933-05-19 1936-09-29 Rhone Poulenc Sa Manufacture of bases derived from benz-dioxane
US2375138A (en) 1942-05-01 1945-05-01 American Cyanamid Co Alkamine esters of aryloxymethyl benzoic acid
US2629736A (en) 1951-02-24 1953-02-24 Searle & Co Basically substituted n-alkyl derivatives of alpha, beta, beta-triarylpropionamides
US2730544A (en) 1952-07-23 1956-01-10 Sahyun Lab Alkylaminoalkyl esters of hydroxycyclohexylbenzoic acid
US2750387A (en) 1953-11-25 1956-06-12 Searle & Co Basically substituted derivatives of diarylaminobenzamides
DE1211359B (en) 1955-11-29 1966-02-24 Oreal Oxidant-free cold dye for human hair
US2928833A (en) 1959-03-03 1960-03-15 S E Massengill Company Theophylline derivatives
US3174901A (en) 1963-01-31 1965-03-23 Jan Marcel Didier Aron Samuel Process for the oral treatment of diabetes
US3454635A (en) 1965-07-27 1969-07-08 Hoechst Ag Benzenesulfonyl-ureas and process for their manufacture
US3673241A (en) 1968-04-04 1972-06-27 Ciba Geigy Corp Substituted benzaldehyde guanylhydrazones
ES385302A1 (en) 1970-10-22 1973-04-16 Miquel S A Lab Procedure for the obtaining of trisused derivatives of etilendiamine. (Machine-translation by Google Translate, not legally binding)
DE2205815A1 (en) 1972-02-08 1973-08-16 Hoechst Ag N-(oxazolin-2-yl)-piperazine - with antitussive activity
JPS5512435B2 (en) 1972-07-01 1980-04-02
US4005208A (en) 1975-05-16 1977-01-25 Smithkline Corporation N-Heterocyclic-9-xanthenylamines
US4061753A (en) 1976-02-06 1977-12-06 Interx Research Corporation Treating psoriasis with transient pro-drug forms of xanthine derivatives
NO154918C (en) 1977-08-27 1987-01-14 Bayer Ag ANALOGUE PROCEDURE FOR THE PREPARATION OF THERAPEUTIC ACTIVE DERIVATIVES OF 3,4,5-TRIHYDROXYPIPERIDINE.
DE2758025A1 (en) 1977-12-24 1979-07-12 Bayer Ag Tri:hydroxy-piperidine derivs. - useful as glucosidase inhibitors for treating diabetes etc. and as animal feed additives
DE2929596A1 (en) 1979-07-21 1981-02-05 Hoechst Ag METHOD FOR PRODUCING OXOALKYL XANTHINES
CY1306A (en) 1980-10-01 1985-12-06 Glaxo Group Ltd Aminoalkyl furan derivative
US4382091A (en) 1981-04-30 1983-05-03 Syntex (U.S.A.) Inc. Stabilization of 1-substituted imidazole derivatives in talc
FR2558162B1 (en) 1984-01-17 1986-04-25 Adir NOVEL XANTHINE DERIVATIVES, PROCESSES FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
FI79107C (en) 1984-06-25 1989-11-10 Orion Yhtymae Oy Process for the preparation of stable form of prazosin hydrochloride.
JPS6130567A (en) 1984-07-23 1986-02-12 Shiseido Co Ltd Method of stabilizing urea
JPS61124383A (en) 1984-11-16 1986-06-12 Unitika Ltd Stabilization of immobilized fibrinolytic enzyme
AR240698A1 (en) 1985-01-19 1990-09-28 Takeda Chemical Industries Ltd Process for the preparation of 5-(4-(2-(5-ethyl-2-pyridil)-ethoxy)benzyl)-2,4-thiazolodinedione and their salts
CA1242699A (en) 1985-02-01 1988-10-04 Bristol-Myers Company Cefbuperazone and derivatives thereof
US4741898A (en) 1985-04-01 1988-05-03 Fisher Scientific Company Stabilized stain composition
GB8515934D0 (en) 1985-06-24 1985-07-24 Janssen Pharmaceutica Nv (4-piperidinomethyl and-hetero)purines
US5258380A (en) 1985-06-24 1993-11-02 Janssen Pharmaceutica N.V. (4-piperidinylmethyl and -hetero)purines
ES2058061T3 (en) 1985-10-25 1994-11-01 Beecham Group Plc DERIVED FROM PIPERIDINE, ITS PREPARATION AND ITS USE AS A MEDICINAL PRODUCT.
US5034225A (en) 1985-12-17 1991-07-23 Genentech Inc. Stabilized human tissue plasminogen activator compositions
US5433959A (en) 1986-02-13 1995-07-18 Takeda Chemical Industries, Ltd. Stabilized pharmaceutical composition
DE3683760D1 (en) 1986-03-21 1992-03-12 Heumann Pharma Gmbh & Co CRYSTALLINE, WATER-FREE SIGMA -FORM OF 2- (4- (2-FUROYL- (2-PIPERAZINE) -1-YL) -4-AMINO-6,7-DIMETHOXYCHINAZOLINE HYDROCHLORIDE AND METHOD FOR THE PRODUCTION THEREOF.
CA1341320C (en) 1986-05-05 2001-11-20 Joel Habener Insulinotropic hormone
US5120712A (en) 1986-05-05 1992-06-09 The General Hospital Corporation Insulinotropic hormone
AU619444B2 (en) 1986-06-02 1992-01-30 Nippon Chemiphar Co. Ltd. 2-(2-aminobenzylsulfinyl)- benzimidazole derivatives
US4968672A (en) 1987-01-02 1990-11-06 The United States Of America As Represented By The Department Of Health And Human Services Adenosine receptor prodrugs
US4743450A (en) 1987-02-24 1988-05-10 Warner-Lambert Company Stabilized compositions
US5093330A (en) 1987-06-15 1992-03-03 Ciba-Geigy Corporation Staurosporine derivatives substituted at methylamino nitrogen
JPS6440433A (en) 1987-08-05 1989-02-10 Green Cross Corp Aqueous liquid composition of thrombin
JPH0395177A (en) 1988-05-19 1991-04-19 Chugai Pharmaceut Co Ltd Novel quinolone carboxylic acid derivative
US5329025A (en) 1988-09-21 1994-07-12 G. D. Searle & Co. 3-azido compound
DE3926119A1 (en) 1989-08-08 1991-02-14 Bayer Ag 3-AMINO-5-AMINOCARBONYL-1,2,4-TRIAZOLE DERIVATIVES
US5234897A (en) 1989-03-15 1993-08-10 Bayer Aktiengesellschaft Herbicidal 3-amino-5-aminocarbonyl-1,2,4-triazoles
GB8906792D0 (en) 1989-03-23 1989-05-10 Beecham Wuelfing Gmbh & Co Kg Treatment and compounds
DE3916430A1 (en) 1989-05-20 1990-11-22 Bayer Ag METHOD FOR PRODUCING 3-AMINO-5-AMINOCARBONYL-1,2,4-TRIAZOLE DERIVATIVES
US5332744A (en) 1989-05-30 1994-07-26 Merck & Co., Inc. Substituted imidazo-fused 6-membered heterocycles as angiotensin II antagonists
US5223499A (en) 1989-05-30 1993-06-29 Merck & Co., Inc. 6-amino substituted imidazo[4,5-bipyridines as angiotensin II antagonists
IL94390A (en) 1989-05-30 1996-03-31 Merck & Co Inc Di-substituted imidazo fused 6-membered nitrogen-containing heterocycles and pharmaceutical compositions containing them
FI94339C (en) 1989-07-21 1995-08-25 Warner Lambert Co Process for the preparation of pharmaceutically acceptable [R- (R *, R *)] - 2- (4-fluorophenyl) -, - dihydroxy-5- (1-methylethyl) -3-phenyl-4 - [(phenylamino) carbonyl] -1H- for the preparation of pyrrole-1-heptanoic acid and its pharmaceutically acceptable salts
JPH0374895A (en) 1989-08-15 1991-03-29 Toshiba Chem Corp Multilayer copper coated laminated board
HU208115B (en) 1989-10-03 1993-08-30 Biochemie Gmbh New process for producting pleuromutilin derivatives
FR2654935B1 (en) 1989-11-28 1994-07-01 Lvmh Rech USE OF XANTHINES, WHICH MAY BE INCORPORATED IN LIPOSOMES, TO PROMOTE PIGMENTATION OF THE SKIN OR HAIR.
ATE134624T1 (en) 1990-02-19 1996-03-15 Ciba Geigy Ag ACYL COMPOUNDS
KR930000861B1 (en) 1990-02-27 1993-02-08 한미약품공업 주식회사 Omeprazole rectal composition
DK0475482T3 (en) 1990-09-13 1995-04-03 Akzo Nobel Nv Stabilized solid chemical agents
GB9020959D0 (en) 1990-09-26 1990-11-07 Beecham Group Plc Novel compounds
US5084460A (en) 1990-12-24 1992-01-28 A. H. Robins Company, Incorporated Methods of therapeutic treatment with N-(3-ouinuclidinyl)-2-hydroxybenzamides and thiobenzamides
US5594003A (en) 1991-02-06 1997-01-14 Dr. Karl Thomae Gmbh Tetrahydroimidazo[1,2-a]pyridin-2-yl-(benzimidazol-1-yl)-methyl-biphenyls useful as angiotensin-II antagonists
US5602127A (en) 1991-02-06 1997-02-11 Karl Thomae Gmbh (Alkanesultam-1-yl)-benzimidazol-1-yl)-1yl)-methyl-biphenyls useful as angiotensin-II antagonists
US5591762A (en) 1991-02-06 1997-01-07 Dr. Karl Thomae Gmbh Benzimidazoles useful as angiotensin-11 antagonists
GB9109862D0 (en) 1991-05-08 1991-07-03 Beecham Lab Sa Pharmaceutical formulations
DE4124150A1 (en) 1991-07-20 1993-01-21 Bayer Ag SUBSTITUTED TRIAZOLES
TW225528B (en) 1992-04-03 1994-06-21 Ciba Geigy Ag
US5300298A (en) 1992-05-06 1994-04-05 The Pennsylvania Research Corporation Methods of treating obesity with purine related compounds
GB9215633D0 (en) 1992-07-23 1992-09-09 Smithkline Beecham Plc Novel treatment
ES2115725T3 (en) 1992-07-31 1998-07-01 Shionogi & Co TRIAZOLYLTIOMETHYLETHEOPHALOSPORINE HYDROCHLORIDE, ITS CRYSTALLINE HYDRATE AND THE PREPARATION OF IT.
TW252044B (en) 1992-08-10 1995-07-21 Boehringer Ingelheim Kg
DE4242459A1 (en) 1992-12-16 1994-06-23 Merck Patent Gmbh imidazopyridines
CA2118117A1 (en) 1993-02-18 1994-08-19 Shigeki Fujiwara Adenosine uptake inhibitor
JP3726291B2 (en) 1993-07-05 2005-12-14 三菱ウェルファーマ株式会社 Benzoxazine compound having stable crystal structure and process for producing the same
FR2707641B1 (en) 1993-07-16 1995-08-25 Fournier Ind & Sante Compounds of imidazol-5-carboxamide, their process for preparing their intermediates and their use in therapy.
DE4339868A1 (en) 1993-11-23 1995-05-24 Merck Patent Gmbh imidazopyridazines
DE4404183A1 (en) 1994-02-10 1995-08-17 Merck Patent Gmbh 4-amino-1-piperidylbenzoylguanidine
US5545745A (en) 1994-05-23 1996-08-13 Sepracor, Inc. Enantioselective preparation of optically pure albuterol
CO4410191A1 (en) 1994-09-19 1997-01-09 Lilly Co Eli SYNTHESIS OF 3- [4- (2-AMINOETOXI) BENZOIL] -2-ARYL-6- HYDROXYBENZO [b] THIOPHENES
DE69531623T2 (en) 1994-10-12 2004-06-17 Euroceltique S.A. NEW BENZOXAZOLES
GB9501178D0 (en) 1995-01-20 1995-03-08 Wellcome Found Guanine derivative
EP0825993A1 (en) 1995-05-19 1998-03-04 Chiroscience Limited Xanthines and their therapeutic use
JPH08333339A (en) 1995-06-08 1996-12-17 Fujisawa Pharmaceut Co Ltd Production of optically active piperidineacetic acid derivative
GB9523752D0 (en) 1995-11-21 1996-01-24 Pfizer Ltd Pharmaceutical formulations
DE19543478A1 (en) 1995-11-22 1997-05-28 Bayer Ag Crystalline hydrochloride of {(R) - (-) - 2N- [4- (1,1-dioxido-3-oxo-2,3-dihydrobenzisothiazol-2-yl) -buytl] aminomethyl} -chroman
FR2742751B1 (en) 1995-12-22 1998-01-30 Rhone Poulenc Rorer Sa NOVEL TAXOIDS, THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
AU720796B2 (en) 1995-12-26 2000-06-15 Alteon Inc. N-acylaminoalkylhydrazinecarboximidamides
DE19616486C5 (en) 1996-04-25 2016-06-30 Royalty Pharma Collection Trust Method for lowering the blood glucose level in mammals
TW518219B (en) 1996-04-26 2003-01-21 Chugai Pharmaceutical Co Ltd Erythropoietin solution preparation
AU1153097A (en) 1996-06-07 1998-01-05 Eisai Co. Ltd. Stable polymorphs of donepezil (1-benzyl-4-{(5,6-dimethoxy-1-indanon)-2-yl}methylpiperidine ) hydrochloride and process for production
US5965555A (en) 1996-06-07 1999-10-12 Hoechst Aktiengesellschaft Xanthine compounds having terminally animated alkynol side chains
US5958951A (en) 1996-06-14 1999-09-28 Novo Nordiskials Modified form of the R(-)-N-(4,4-di(3-methylthien-2-yl)but-3-enyl)-nipecotic acid hydrochloride
WO2008109681A2 (en) 2007-03-08 2008-09-12 Phenomix Corporation Methods and intermediates for synthesis of selective dpp-iv inhibitors
US5753635A (en) 1996-08-16 1998-05-19 Berlex Laboratories, Inc. Purine derivatives and their use as anti-coagulants
KR100516088B1 (en) 1996-09-23 2005-09-22 일라이 릴리 앤드 캄파니 Olanzapine Dihydrate D
AU4699697A (en) 1996-10-28 1998-05-22 Novo Nordisk A/S A process for the preparation of (-)-3,4-trans-diarylchromans
UA65549C2 (en) 1996-11-05 2004-04-15 Елі Ліллі Енд Компані Use of glucagon-like peptides such as glp-1, glp-1 analog, or glp-1 derivative in methods and compositions for reducing body weight
JP2001504105A (en) 1996-11-12 2001-03-27 ノボ ノルディスク アクティーゼルスカブ Use of GLP-1 peptide
GB9623859D0 (en) 1996-11-15 1997-01-08 Chiroscience Ltd Novel compounds
DK0948358T4 (en) 1996-12-24 2012-03-19 Biogen Idec Inc Stable inteferon fluid formulations
DE19705233A1 (en) 1997-02-12 1998-08-13 Froelich Juergen C Preparation of stable, orally administered arginine solutions
CO4950519A1 (en) 1997-02-13 2000-09-01 Novartis Ag PHTHALAZINES, PHARMACEUTICAL PREPARATIONS THAT UNDERSTAND THEM AND THE PROCESS FOR THEIR PREPARATION
US6011049A (en) 1997-02-19 2000-01-04 Warner-Lambert Company Combinations for diabetes
CZ291842B6 (en) 1997-03-13 2003-06-18 Hexal Ag Pharmaceutical formulation
US5972332A (en) 1997-04-16 1999-10-26 The Regents Of The University Of Michigan Wound treatment with keratinocytes on a solid support enclosed in a porous material
CO4750643A1 (en) 1997-06-13 1999-03-31 Lilly Co Eli STABLE FORMULATION OF INSULIN CONTAINING L-ARGININ AND PROTAMINE
US6174548B1 (en) 1998-08-28 2001-01-16 Andrx Pharmaceuticals, Inc. Omeprazole formulation
SK8402000A3 (en) 1997-12-05 2001-03-12 Astrazeneca Uk Ltd Novel compounds
ID21411A (en) 1997-12-10 1999-06-10 Takeda Chemical Industries Ltd AGENTS TO TREAT GLUCOSE RESISTANCE THAT IS RISK OF HIGH DAMAGED
JPH11193270A (en) 1997-12-26 1999-07-21 Koei Chem Co Ltd Production of optically active 1-methyl-3-piperidinemethanol
USRE39112E1 (en) 1998-01-05 2006-05-30 Eisai Co., Ltd. Purine derivatives and adenosine A2 receptor antagonists serving as preventives/remedies for diabetes
WO1999038501A2 (en) 1998-02-02 1999-08-05 Trustees Of Tufts College Method of regulating glucose metabolism, and reagents related thereto
US6310065B1 (en) 1998-03-31 2001-10-30 Nissan Chemical Industries, Ltd. Pyridazinone hydrochloride compound and method for producing the same
CA2268621A1 (en) 1998-04-13 1999-10-13 Takeda Chemical Industries, Ltd. 2-pipirazinone-1-acetic acid derivative, production and use thereof
US6207207B1 (en) 1998-05-01 2001-03-27 Mars, Incorporated Coated confectionery having a crispy starch based center and method of preparation
DE19823831A1 (en) 1998-05-28 1999-12-02 Probiodrug Ges Fuer Arzneim New pharmaceutical use of isoleucyl thiazolidide and its salts
DE19828114A1 (en) 1998-06-24 2000-01-27 Probiodrug Ges Fuer Arzneim Produgs of unstable inhibitors of dipeptidyl peptidase IV
CN1185013C (en) 1998-07-15 2005-01-19 旭化成株式会社 Excipient
CO5150173A1 (en) 1998-12-10 2002-04-29 Novartis Ag COMPOUNDS N- (REPLACED GLYCLE) -2-DIPEPTIDYL-IV PEPTIDASE INHIBITING CYANOPIRROLIDINS (DPP-IV) WHICH ARE EFFECTIVE IN THE TREATMENT OF CONDITIONS MEDIATED BY DPP-IV INHIBITION
IT1312018B1 (en) 1999-03-19 2002-04-04 Fassi Aldo IMPROVED PROCEDURE FOR THE PRODUCTION OF NON HYGROSCOPICIDAL SALTS OF L (-) - CARNITINE.
AR023966A1 (en) 1999-05-12 2002-09-04 Fujisawa Pharmaceutical Co A PHARMACEUTICAL COMPOSITION FOR THE PREVENTION AND / OR TREATMENT OF PARKINSON'S DISEASE AND THE CONCOMITING SYMPTOMS OF THE SAME, AND THE USE OF A DUAL ADENOSINE A1 A2A RECEIVER UNANTAGONIST TO PREPARE SUCH COMPOSITION
US20040152659A1 (en) 1999-05-12 2004-08-05 Fujisawa Pharmaceutical Co. Ltd. Method for the treatment of parkinson's disease comprising administering an A1A2a receptor dual antagonist
WO2000072799A2 (en) 1999-05-27 2000-12-07 The University Of Virginia Patent Foundation Method and compositions for treating the inflammatory response
AU5103200A (en) 1999-05-31 2000-12-18 Mitsubishi Chemical Corporation Freeze dried hgf preparations
US6545002B1 (en) 1999-06-01 2003-04-08 University Of Virginia Patent Foundation Substituted 8-phenylxanthines useful as antagonists of A2B adenosine receptors
WO2000072973A1 (en) 1999-06-01 2000-12-07 Elan Pharma International Ltd. Small-scale mill and method thereof
EE04748B1 (en) 1999-06-21 2006-12-15 Boehringer Ingelheim Pharma Kg Bicyclic heterocyclic compounds, drugs containing these compounds, their use and methods for their preparation
US6448323B1 (en) 1999-07-09 2002-09-10 Bpsi Holdings, Inc. Film coatings and film coating compositions based on polyvinyl alcohol
ES2166270B1 (en) 1999-07-27 2003-04-01 Almirall Prodesfarma Sa DERIVATIVES OF 8-PHENYL-6,9-DIHIDRO- (1,2,4,) TRIAZOLO (3,4-I) PURIN-5-ONA.
US6515117B2 (en) 1999-10-12 2003-02-04 Bristol-Myers Squibb Company C-aryl glucoside SGLT2 inhibitors and method
US6586438B2 (en) 1999-11-03 2003-07-01 Bristol-Myers Squibb Co. Antidiabetic formulation and method
GB9928330D0 (en) 1999-11-30 2000-01-26 Ferring Bv Novel antidiabetic agents
NZ531929A (en) 1999-12-23 2006-01-27 Novartis Ag Use of nateglinide as a hypoglycemic agent for treating impaired glucose metabolism
CA2396079A1 (en) 2000-01-07 2001-07-19 Transform Pharmaceuticals, Inc. High-throughput formation, identification, and analysis of diverse solid-forms
US6362172B2 (en) 2000-01-20 2002-03-26 Bristol-Myers Squibb Company Water soluble prodrugs of azole compounds
DE60132723T2 (en) 2000-01-21 2009-01-29 Novartis Pharma Ag Compositions consisting of dipeptidyl peptidase IV inhibitors and antidiabetics
JP4621326B2 (en) 2000-02-01 2011-01-26 エーザイ・アール・アンド・ディー・マネジメント株式会社 Teprenone stabilized composition
WO2001056993A2 (en) 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Pyrazole compositions useful as inhibitors of erk
EP1295609A4 (en) 2000-02-24 2004-11-03 Takeda Chemical Industries Ltd Drugs containing combined active ingredients
EP1132389A1 (en) 2000-03-06 2001-09-12 Vernalis Research Limited New aza-indolyl derivatives for the treatment of obesity
US6395767B2 (en) 2000-03-10 2002-05-28 Bristol-Myers Squibb Company Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl peptidase IV and method
GB0006133D0 (en) 2000-03-14 2000-05-03 Smithkline Beecham Plc Novel pharmaceutical
JP2001278812A (en) 2000-03-27 2001-10-10 Kyoto Pharmaceutical Industries Ltd Disintegrant for tablet and tablet using the same
ATE419036T1 (en) 2000-03-31 2009-01-15 Prosidion Ltd IMPROVED ISLAND CELL ACTIVITY IN DIABETES MELLITUS AND ITS PREVENTION
WO2001074397A1 (en) 2000-03-31 2001-10-11 Kirin Beer Kabushiki Kaisha Powdery preparation for transmucosal administration containing a polymeric form of drug and exhibiting improved storage stability
JP2001292388A (en) 2000-04-05 2001-10-19 Sharp Corp Reproducing device
GB0008694D0 (en) 2000-04-07 2000-05-31 Novartis Ag Organic compounds
EP1295873A4 (en) 2000-06-14 2004-05-19 Processes for producing racemic piperidine derivative and for producing optically active piperidine derivative
GB0014969D0 (en) 2000-06-19 2000-08-09 Smithkline Beecham Plc Novel method of treatment
US7078397B2 (en) 2000-06-19 2006-07-18 Smithkline Beecham Corporation Combinations of dipeptidyl peptidase IV inhibitors and other antidiabetic agents for the treatment of diabetes mellitus
US6689353B1 (en) 2000-06-28 2004-02-10 Bayer Pharmaceuticals Corporation Stabilized interleukin 2
AU6895801A (en) 2000-07-04 2002-01-14 Novo Nordisk As Heterocyclic compounds, which are inhibitors of the enzyme dpp-iv
WO2002014271A1 (en) 2000-08-10 2002-02-21 Mitsubishi Pharma Corporation Proline derivatives and use thereof as drugs
US6821978B2 (en) 2000-09-19 2004-11-23 Schering Corporation Xanthine phosphodiesterase V inhibitors
US20060034922A1 (en) 2000-11-03 2006-02-16 Andrx Labs, Llc Controlled release metformin compositions
US20040180925A1 (en) 2000-12-27 2004-09-16 Kenji Matsuno Dipeptidylpeptidase-IV inhibitor
FR2818906B1 (en) 2000-12-29 2004-04-02 Dospharma DRUG ASSOCIATION OF A BIGUANINE AND A CARRIER, FOR EXAMPLE OF METFORMIN AND ARGININE
FR2819254B1 (en) 2001-01-08 2003-04-18 Fournier Lab Sa NOVEL N- (PHENYLSULFONYL) GLYCINE COMPOUNDS, PROCESS FOR THEIR PREPARATION AND THEIR USE FOR OBTAINING PHARMACEUTICAL COMPOSITIONS
DE10109021A1 (en) 2001-02-24 2002-09-05 Boehringer Ingelheim Pharma New 8-substituted-xanthine derivatives, useful e.g. for treating diabetes and arthritis, act by inhibiting dipeptidylpeptidase-IV
DE10117803A1 (en) 2001-04-10 2002-10-24 Boehringer Ingelheim Pharma New 8-substituted-xanthine derivatives, useful e.g. for treating diabetes and arthritis, act by inhibiting dipeptidylpeptidase-IV
BR0206831A (en) 2001-02-02 2004-07-06 Takeda Chemical Industries Ltd Compound, crystal, pharmaceutical agent, agents for the prophylaxis or treatment of diabetes, diabetic complications, impaired glucose tolerance and obesity, peptidase inhibitor, use of a compound, and method of producing a compound
WO2002066015A1 (en) 2001-02-16 2002-08-29 Bristol-Myers Squibb Pharma Company Use of polyalkylamine polymers in controlled release devices
DK1953162T3 (en) 2001-02-24 2012-09-10 Boehringer Ingelheim Pharma Xanthine derivatives, their preparation and their use as a drug.
US6936590B2 (en) 2001-03-13 2005-08-30 Bristol Myers Squibb Company C-aryl glucoside SGLT2 inhibitors and method
US6693094B2 (en) 2001-03-22 2004-02-17 Chrono Rx Llc Biguanide and sulfonylurea formulations for the prevention and treatment of insulin resistance and type 2 diabetes mellitus
JP2002348279A (en) 2001-05-25 2002-12-04 Nippon Kayaku Co Ltd Production method for optically active pyridylketone derivatives and optically active pyridylketone derivatives
DE10130371A1 (en) 2001-06-23 2003-01-02 Boehringer Ingelheim Pharma New drug compositions based on anticholinergics, corticosteroids and betamimetics
GB0115517D0 (en) 2001-06-25 2001-08-15 Ferring Bv Novel antidiabetic agents
ATE455759T1 (en) 2001-06-27 2010-02-15 Smithkline Beecham Corp FLUOROPYRROLIDINE AS A DIPEPTIDYLPEPTIDASE INHIBITOR
EP1399433B1 (en) 2001-06-27 2007-08-22 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
JP2005502624A (en) 2001-07-03 2005-01-27 ノボ ノルディスク アクティーゼルスカブ Purine derivatives inhibiting DPP-IV for the treatment of diabetes
US6869947B2 (en) 2001-07-03 2005-03-22 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme DPP-IV
UA74912C2 (en) 2001-07-06 2006-02-15 Merck & Co Inc Beta-aminotetrahydroimidazo-(1,2-a)-pyrazines and tetratriazolo-(4,3-a)-pyrazines as inhibitors of dipeptylpeptidase for the treatment or prevention of diabetes
WO2003006424A1 (en) 2001-07-10 2003-01-23 4Sc Ag Novel compounds as anti-inflammatory, immunomodulatory and anti-proliferatory agents
US7638522B2 (en) 2001-08-13 2009-12-29 Janssen Pharmaceutica N.V. Salt of 4-[[4-[[4-(2-cyanoethenyl)-2,6-dimethylphenyl]amino]-2-pyrimidinyl]amino] benzonitrile
JP2005509603A (en) 2001-09-19 2005-04-14 ノボ ノルディスク アクティーゼルスカブ Heterocyclic compounds that are inhibitors of the DPP-IV enzyme
CA2461454A1 (en) 2001-09-24 2003-04-03 Oregon Health And Science University Assessment of neurons in the arcuate nucleus to screen for agents that modify feeding behavior
WO2003034944A1 (en) 2001-10-15 2003-05-01 Hemoteq Gmbh Coating of stents for preventing restenosis
DE10151296A1 (en) 2001-10-17 2003-04-30 Boehringer Ingelheim Pharma Keratinocytes useful as a biologically active substance in the treatment of wounds
US6723340B2 (en) 2001-10-25 2004-04-20 Depomed, Inc. Optimal polymer mixtures for gastric retentive tablets
US6861440B2 (en) 2001-10-26 2005-03-01 Hoffmann-La Roche Inc. DPP IV inhibitors
US20030083354A1 (en) 2001-10-26 2003-05-01 Pediamed Pharmaceuticals, Inc. Phenylephrine tannate and pyrilamine tannate salts in pharmaceutical compositions
CA2363053C (en) 2001-11-09 2011-01-25 Bernard Charles Sherman Clopidogrel bisulfate tablet formulation
CA2466870A1 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
KR20040064687A (en) 2001-12-21 2004-07-19 도오레 화인케미칼 가부시키가이샤 Process for production of optically active cis-piperidine derivatives
US6727261B2 (en) 2001-12-27 2004-04-27 Hoffman-La Roche Inc. Pyrido[2,1-A]Isoquinoline derivatives
CA2471766C (en) 2001-12-28 2011-01-11 Nrl Pharma, Inc. Compositions for improving lipid metabolism
AU2003201998C1 (en) 2002-01-10 2012-10-25 Imperial Innovations Limited Modification of feeding behavior
US20070197552A1 (en) 2002-01-11 2007-08-23 Novo Nordisk A/S Method and composition for treatment of diabetes, hypertension, chronic heart failure and fluid retentive states
WO2003057200A2 (en) 2002-01-11 2003-07-17 Novo Nordisk A/S Compositions comprising inhibitors of dpp-iv and nep enzymes for the treatment of diabetes
WO2003059327A1 (en) 2002-01-16 2003-07-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Bilayer pharmaceutical tablet comprising telmisartan and a diuretic and preparation thereof
WO2003061688A1 (en) 2002-01-21 2003-07-31 Nrl Pharma, Inc. Novel analgesics
EP1333033A1 (en) 2002-01-30 2003-08-06 Boehringer Ingelheim Pharma GmbH & Co.KG FAP-activated anti-tumor compounds
CN1688291A (en) 2002-02-01 2005-10-26 辉瑞产品公司 Immediate release dosage forms containing solid drug dispersions
US7610153B2 (en) 2002-02-13 2009-10-27 Virginia Commonwealth University Multi-drug titration and evaluation
JP5069395B2 (en) 2002-02-21 2012-11-07 ヴァレアント インターナショナル (バルバドス) エスアールエル Pharmaceutical composition with improved release of at least one form of tramadol
DE60304911D1 (en) 2002-02-25 2006-06-08 Eisai Co Ltd Xanthine derivatives as DPP-IV inhibitors
HUP0200849A2 (en) 2002-03-06 2004-08-30 Sanofi-Synthelabo N-aminoacetyl-pyrrolidine-2-carbonitrile derivatives, pharmaceutical compositions containing them and process for producing them
JP4298212B2 (en) 2002-03-29 2009-07-15 大日本印刷株式会社 Method for producing high melting point type epinastine hydrochloride
JP2003300977A (en) 2002-04-10 2003-10-21 Sumitomo Pharmaceut Co Ltd Xanthine derivative
US20050113577A1 (en) 2002-04-16 2005-05-26 Karki Shyam B. Solid forms of slats with tyrosine kinase activity
JP4424203B2 (en) 2002-04-26 2010-03-03 味の素株式会社 Diabetes prevention and treatment
AU2003231252A1 (en) 2002-05-09 2003-11-11 Enos Pharmaceuticals, Inc. Methods and compositions for the treatment and prevention of intermittent claudication or alzheimer's disease
GB0212412D0 (en) 2002-05-29 2002-07-10 Novartis Ag Combination of organic compounds
AR040232A1 (en) 2002-05-31 2005-03-23 Schering Corp PROCESS TO PREPARE INHIBITORS OF XANTINA FOSFODIESTERASA V, AND PRECURSORS OF THE SAME
NZ536794A (en) 2002-06-06 2007-04-27 Eisai Co Ltd Condensed imidazole derivatives
ES2199061B1 (en) 2002-06-10 2005-02-16 Laboratorios Vita, S.A. TROUBLE-BASED TABLETS AND PROCEDURE FOR OBTAINING.
FR2840897B1 (en) 2002-06-14 2004-09-10 Fournier Lab Sa NOVEL ARYLSULFONAMIDE DERIVATIVES AND THEIR USE IN THERAPEUTICS
US20040002615A1 (en) 2002-06-28 2004-01-01 Allen David Robert Preparation of chiral amino-nitriles
GB0215676D0 (en) 2002-07-05 2002-08-14 Novartis Ag Organic compounds
US20040023981A1 (en) 2002-07-24 2004-02-05 Yu Ren Salt forms with tyrosine kinase activity
AR040661A1 (en) 2002-07-26 2005-04-13 Theravance Inc CRYSTAL DICHLORHYDRATE OF N- {2 - [- ((R) -2-HYDROXI-2-PHENYLETHYLAMINE) PHENYL] ETIL} - (R) -2 HYDROXY-2- (3-FORMAMIDE-4-HYDROXYPHENYL) ETHYLAMINE, RECEIVER AGONIST BETA 2 ADRENERGIC
TW200404796A (en) 2002-08-19 2004-04-01 Ono Pharmaceutical Co Nitrogen-containing compound
EP3424926A1 (en) 2002-08-21 2019-01-09 Boehringer Ingelheim Pharma GmbH & Co. KG 8-[3-amino-piperidin-1-yl]-xanthins, their production and utilisation as medicine
DE10238243A1 (en) 2002-08-21 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New 8-(3-amino-piperidin-1-yl)-xanthine derivatives are dipeptidylpeptidase-IV inhibitors useful for, e.g. treating diabetes mellitus, arthritis or obesity
US7407955B2 (en) 2002-08-21 2008-08-05 Boehringer Ingelheim Pharma Gmbh & Co., Kg 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
US7495005B2 (en) 2002-08-22 2009-02-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivatives, their preparation and their use in pharmaceutical compositions
DE10238477A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New purine derivatives, their production and their use as medicines
US7569574B2 (en) 2002-08-22 2009-08-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Purine derivatives, the preparation thereof and their use as pharmaceutical compositions
DE10238470A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New xanthine derivatives, their production and their use as medicines
DE10238723A1 (en) 2002-08-23 2004-03-11 Bayer Ag Phenyl substituted pyrazolyprimidines
DE10238724A1 (en) 2002-08-23 2004-03-04 Bayer Ag New 6-alkyl-1,5-dihydro-4H-pyrazolo-(3,4-d)-pyrimidin-4-ones useful as selective phosphodiesterase 9A inhibitors for improving attention, concentration, learning and/or memory performance
US20060039974A1 (en) 2002-09-11 2006-02-23 Takeda Pharmaceutical Company Limited Sustained release preparation
CN1688293A (en) 2002-09-16 2005-10-26 韦思公司 Delayed release formulations for oral administration of a polypeptide therapeutic agent and methods of using same
EP1557165A4 (en) 2002-09-26 2008-12-03 Eisai R&D Man Co Ltd Combination drug
WO2004033455A2 (en) 2002-10-08 2004-04-22 Novo Nordisk A/S Hemisuccinate salts of heterocyclic dpp-iv inhibitors
AU2003267732A1 (en) 2002-10-08 2004-05-04 Ranbaxy Laboratories Limited Gabapentin tablets and methods for their preparation
US20040122048A1 (en) 2002-10-11 2004-06-24 Wyeth Holdings Corporation Stabilized pharmaceutical composition containing basic excipients
US6861526B2 (en) 2002-10-16 2005-03-01 Pfizer Inc. Process for the preparation of (S,S)-cis-2-benzhydryl-3-benzylaminoquinuclidine
DE60320008T2 (en) 2002-10-18 2009-06-18 Merck & Co., Inc. HETEROCYCLIC BETA-AMINO COMPOUNDS AS INHIBITORS OF DIPEPTIDYLPEPTIDASE FOR TREATMENT OR PREVENTION OF DIABETES
JP2004161749A (en) 2002-10-24 2004-06-10 Toray Fine Chemicals Co Ltd Method for producing optically active, nitrogen-containing compound
AU2003280680A1 (en) 2002-11-01 2004-06-18 Sumitomo Pharmaceuticals Co., Ltd. Xanthine compound
MXPA05004890A (en) 2002-11-07 2005-07-22 Merck & Co Inc Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes.
DE10251927A1 (en) 2002-11-08 2004-05-19 Boehringer Ingelheim Pharma Gmbh & Co. Kg New 1,7,8-trisubstituted xanthine derivatives, are dipeptidylpeptidase-IV inhibitors useful e.g. for treating diabetes mellitus type I or II, arthritis or obesity
US7482337B2 (en) 2002-11-08 2009-01-27 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivatives, the preparation thereof and their use as pharmaceutical compositions
DE10254304A1 (en) 2002-11-21 2004-06-03 Boehringer Ingelheim Pharma Gmbh & Co. Kg New xanthine derivatives, their production and their use as medicines
UY28103A1 (en) 2002-12-03 2004-06-30 Boehringer Ingelheim Pharma NEW IMIDAZO-PIRIDINONAS REPLACED, ITS PREPARATION AND ITS EMPLOYMENT AS MEDICATIONS
US7109192B2 (en) 2002-12-03 2006-09-19 Boehringer Ingelheim Pharma Gmbh & Co Kg Substituted imidazo-pyridinones and imidazo-pyridazinones, the preparation thereof and their use as pharmaceutical compositions
US7420079B2 (en) 2002-12-09 2008-09-02 Bristol-Myers Squibb Company Methods and compounds for producing dipeptidyl peptidase IV inhibitors and intermediates thereof
US20060111428A1 (en) 2002-12-10 2006-05-25 Pei-Ran Wang Combination of an dpp-iv inhibitor and a ppar-alpha compound
US20040152720A1 (en) 2002-12-20 2004-08-05 Boehringer Ingelheim Pharma Gmbh & Co. Kg Powdered medicaments containing a tiotropium salt and salmeterol xinafoate
DE10351663A1 (en) 2002-12-20 2004-07-01 Boehringer Ingelheim Pharma Gmbh & Co. Kg Stable, accurately dosable inhalable powder medicament for treating asthma or chronic obstructive pulmonary disease, containing tiotropium, specific form of salmeterol xinafoate and auxiliary
SI1599222T1 (en) 2003-01-08 2009-08-31 Novartis Vaccines & Diagnostic Stabilized aqueous compositions comprising tissue factor pathway inhibitor (tfpi) or tissue factor pathway inhibitor variant
RS20050532A (en) 2003-01-14 2007-12-31 Arena Pharmaceuticals Inc., 1,2,3-trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prpphylaxis and treatment of disorders related thereto such as diabetes and hyperglycemia
DE10335027A1 (en) 2003-07-31 2005-02-17 Boehringer Ingelheim Pharma Gmbh & Co. Kg Use of telmisartan and simvastatin for treatment or prophylaxis of cardiovascular, cardiopulmonary and renal diseases e.g. hypertension combined with hyperlipidemia or atherosclerosis
AU2004207731B2 (en) 2003-01-31 2009-08-13 Sanwa Kagaku Kenkyusho Co., Ltd. Compound inhibiting dipeptidyl peptidase iv
PE20040950A1 (en) 2003-02-14 2005-01-01 Theravance Inc BIPHENYL DERIVATIVES AS AGONISTS OF ß2-ADRENERGIC RECEPTORS AND AS ANTAGONISTS OF MUSCARINAL RECEPTORS
JP2004250336A (en) 2003-02-18 2004-09-09 Kao Corp Method for producing coated tablet and sugar-coated tablet
US7135575B2 (en) 2003-03-03 2006-11-14 Array Biopharma, Inc. P38 inhibitors and methods of use thereof
US7442387B2 (en) 2003-03-06 2008-10-28 Astellas Pharma Inc. Pharmaceutical composition for controlled release of active substances and manufacturing method thereof
AU2004220053A1 (en) 2003-03-12 2004-09-23 Arizona Board Of Regents On Behalf Of The University Of Arizona Weak base salts
WO2004082402A1 (en) 2003-03-18 2004-09-30 Novartis Ag Compositions comprising fatty acids and amino acids
DK2368553T3 (en) 2003-04-08 2015-02-09 Progenics Pharm Inc Pharmaceutical preparation comprising methylnaltrexone
US20040220186A1 (en) 2003-04-30 2004-11-04 Pfizer Inc. PDE9 inhibitors for treating type 2 diabetes,metabolic syndrome, and cardiovascular disease
WO2004096806A1 (en) 2003-04-30 2004-11-11 Sumitomo Pharmaceuticals Co. Ltd. Fused imidazole derivative
TW200510277A (en) 2003-05-27 2005-03-16 Theravance Inc Crystalline form of β2-adrenergic receptor agonist
AU2003902828A0 (en) 2003-06-05 2003-06-26 Fujisawa Pharmaceutical Co., Ltd. Dpp-iv inhibitor
DE10327439A1 (en) 2003-06-18 2005-01-05 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel imidazopyridazinone and imidazopyridone derivatives, their production and their use as pharmaceuticals
US7566707B2 (en) 2003-06-18 2009-07-28 Boehringer Ingelheim International Gmbh Imidazopyridazinone and imidazopyridone derivatives, the preparation thereof and their use as pharmaceutical compositions
AU2004251829B2 (en) 2003-06-20 2009-12-17 F. Hoffmann-La Roche Ag Hexahydropyridoisoqinolines as DPP-IV inhibitors
BRPI0411713B8 (en) 2003-06-20 2021-05-25 Hoffmann La Roche compounds, process for their manufacture, pharmaceutical compositions comprising them, method for treatment and/or prophylaxis of diseases that are associated with dpp-iv and their use
JO2625B1 (en) 2003-06-24 2011-11-01 ميرك شارب اند دوم كوربوريشن Phosphoric acid salt of a dipeptidyl peptidase-IV inhibitor
AR045047A1 (en) 2003-07-11 2005-10-12 Arena Pharm Inc ARILO AND HETEROARILO DERIVATIVES TRISUSTITUIDOS AS MODULATORS OF METABOLISM AND PROFILAXIS AND TREATMENT OF DISORDERS RELATED TO THEMSELVES
MXPA06000554A (en) 2003-07-14 2006-07-03 Arena Pharm Inc Fused-aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto.
US20050027012A1 (en) 2003-07-16 2005-02-03 Boehringer Ingelheim International Gmbh Tablets containing ambroxol
ES2340997T3 (en) 2003-07-24 2010-06-14 Wockhardt Limited ORAL COMPOSITIONS FOR THE TREATMENT OF DIABETES.
KR20060054410A (en) 2003-08-01 2006-05-22 제네랩스 테크놀로지스, 인코포레이티드 Bicyclic imidazol derivatives against flaviviridae
US6995183B2 (en) 2003-08-01 2006-02-07 Bristol Myers Squibb Company Adamantylglycine-based inhibitors of dipeptidyl peptidase IV and methods
EP1656158B1 (en) 2003-08-14 2016-03-09 Novo Nordisk Health Care AG Liquid, aqueous pharmaceutical composition of factor vii polypeptides
US20070190022A1 (en) 2003-08-29 2007-08-16 Bacopoulos Nicholas G Combination methods of treating cancer
JP2007505121A (en) 2003-09-08 2007-03-08 武田薬品工業株式会社 Dipeptidyl peptidase inhibitor
EP1671649B1 (en) 2003-10-03 2011-11-23 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase IV inhibitors for treating diabetic patients with sulfonylurea secondary failure
BR0304443B1 (en) 2003-10-28 2012-08-21 process for obtaining high thio2 and low radionuclide titanium concentrates from mechanical anatase concentrates.
US7107714B2 (en) 2003-11-10 2006-09-19 Marketing Displays, Inc. Portable snap-fit sign stand
US7674913B2 (en) 2003-11-12 2010-03-09 Phenomix Corporation Heterocyclic boronic acid compounds
CA2545641A1 (en) 2003-11-17 2005-06-02 Novartis Ag Use of organic compounds
DE10355304A1 (en) 2003-11-27 2005-06-23 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel 8- (piperazin-1-yl) and 8 - ([1,4] diazepan-1-yl) xanthines, their preparation and their use as pharmaceuticals
JPWO2005053695A1 (en) 2003-12-04 2007-12-06 エーザイ・アール・アンド・ディー・マネジメント株式会社 Agents for preventing or treating multiple sclerosis
US7217711B2 (en) 2003-12-17 2007-05-15 Boehringer Ingelheim International Gmbh Piperazin-1-yl and 2-([1,4]diazepan-1-yl)-imidazo[4,5-d]-pyridazin-4-ones, the preparation thereof and their use as pharmaceutical compositions
DE10359098A1 (en) 2003-12-17 2005-07-28 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel 2- (piperazin-1-yl) and 2 - ([1,4] diazepan-1-yl) imidazo [4,5-d] pyridazin-4-ones, their preparation and their use as pharmaceuticals
TWI349007B (en) 2003-12-18 2011-09-21 Tibotec Pharm Ltd Piperidine-amino-benzimidazole derivatives as inhibitors of respiratory syncytial virus replication
DE10360835A1 (en) 2003-12-23 2005-07-21 Boehringer Ingelheim Pharma Gmbh & Co. Kg New bicyclic imidazole derivatives are dipeptidylpeptidase-IV inhibitors useful to treat e.g. arthritis, obesity, allograft transplantation and calcitonin-induced osteoporosis
NZ547965A (en) 2003-12-24 2009-12-24 Prosidion Ltd 1,2,4-Oxadiazole derivatives as GPCR receptor agonists
LT3366283T (en) 2004-01-20 2021-12-10 Novartis Ag Direct compression formulation and process
JP4994043B2 (en) 2004-01-21 2012-08-08 エランコ・アニマル・ヘルス・アイルランド・リミテッド Mitraltaide oral solution
KR101130433B1 (en) 2004-02-05 2012-03-27 교린 세이야꾸 가부시키 가이샤 Bicycloester derivative
SE0400234D0 (en) 2004-02-06 2004-02-06 Active Biotech Ab New compounds, methods for their preparation and use thereof
EP2119717B1 (en) 2004-02-18 2018-01-17 Boehringer Ingelheim International GmbH 8-[3-amino-piperidin-1-yl]-xanthins, their production and utilisation as DPP IV inhibitors
US7501426B2 (en) 2004-02-18 2009-03-10 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthines, their preparation and their use as pharmaceutical compositions
DE102004019540A1 (en) 2004-04-22 2005-11-10 Boehringer Ingelheim Pharma Gmbh & Co. Kg Composition, useful for treatment of e.g. inflammatory and obstructive respiratory complaint, sinus rhythm in heart in atrioventricular block and circulatory shock, comprises 6-hydroxy-4H-benzo1,4oxazin-3-one derivatives and other actives
DE102004009039A1 (en) 2004-02-23 2005-09-08 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8- [3-Amino-piperidin-1-yl] xanthines, their preparation and use as pharmaceuticals
EP1593671A1 (en) 2004-03-05 2005-11-09 Graffinity Pharmaceuticals AG DPP-IV inhibitors
US7393847B2 (en) 2004-03-13 2008-07-01 Boehringer Ingleheim International Gmbh Imidazopyridazinediones, their preparation and their use as pharmaceutical compositions
CN102134229B (en) 2004-03-15 2020-08-04 武田药品工业株式会社 Dipeptidyl peptidase inhibitors
EP2295422A3 (en) 2004-03-16 2012-01-04 Boehringer Ingelheim International GmbH Glucopyranosyl substituted benzol derivatives, pharmaceutical compositions containing these compounds, use thereof and method for their production
EP1577306A1 (en) 2004-03-17 2005-09-21 Boehringer Ingelheim Pharma GmbH & Co.KG novel benzoxazinone derivatives as slow-acting betamimetics and use thereof in treatment of respiratory tract diseases
US7179809B2 (en) 2004-04-10 2007-02-20 Boehringer Ingelheim International Gmbh 2-Amino-imidazo[4,5-d]pyridazin-4-ones, their preparation and their use as pharmaceutical compositions
JP2007531780A (en) 2004-04-10 2007-11-08 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Novel 2-amino-imidazo [4,5-D] pyridazin-4-one and 2-amino-imidazo [4,5-C] pyridazin-4-one, their preparation and use as pharmaceuticals
US7741082B2 (en) 2004-04-14 2010-06-22 Bristol-Myers Squibb Company Process for preparing dipeptidyl peptidase IV inhibitors and intermediates therefor
US20050239778A1 (en) 2004-04-22 2005-10-27 Boehringer Ingelheim International Gmbh Novel medicament combinations for the treatment of respiratory diseases
US20050244502A1 (en) 2004-04-28 2005-11-03 Mathias Neil R Composition for enhancing absorption of a drug and method
US7439370B2 (en) 2004-05-10 2008-10-21 Boehringer Ingelheim International Gmbh Imidazole derivatives, their preparation and their use as intermediates for the preparation of pharmaceutical compositions and pesticides
RS51106B (en) 2004-05-12 2010-10-31 Pfizer Products Inc. Proline derivatives and their use as dipeptidyl peptidase iv inhibitors
DE102004024454A1 (en) 2004-05-14 2005-12-08 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel enantiomerically pure beta agonists, process for their preparation and their use as pharmaceuticals
PE20060315A1 (en) 2004-05-24 2006-05-15 Irm Llc THIAZOLE COMPOUNDS AS PPAR MODULATORS
US7214702B2 (en) 2004-05-25 2007-05-08 Bristol-Myers Squibb Company Process for producing a dipeptidyl peptidase IV inhibitor
TWI354569B (en) 2004-05-28 2011-12-21 Bristol Myers Squibb Co Coated tablet formulation and method
EP1750750B1 (en) 2004-06-01 2012-02-01 Ares Trading S.A. Method of stabilizing proteins
US7935723B2 (en) 2004-06-04 2011-05-03 Novartis Pharma Ag Use of organic compounds
US20050276794A1 (en) 2004-06-09 2005-12-15 Papas Klearchos K Composition and method for improving pancreatic islet cell survival
DE102004030502A1 (en) 2004-06-24 2006-01-12 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel imidazoles and triazoles, their preparation and use as medicines
US20080269311A1 (en) 2004-07-14 2008-10-30 Edwin Bernard Villhauer Combination of Dpp-Iv Inhibitors and Compounds Modulating 5-Ht3 and/or 5-Ht4 Receptors
JP2006045156A (en) 2004-08-06 2006-02-16 Sumitomo Pharmaceut Co Ltd Condensed pyrazole derivative
TW200613275A (en) 2004-08-24 2006-05-01 Recordati Ireland Ltd Lercanidipine salts
WO2006022428A1 (en) 2004-08-26 2006-03-02 Takeda Pharmaceutical Company Limited Remedy for diabetes
DE102004043944A1 (en) 2004-09-11 2006-03-30 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel 8- (3-amino-piperidin-1-yl) -7- (but-2-ynyl) -xanthines, their preparation and their use as pharmaceuticals
DE102004044221A1 (en) 2004-09-14 2006-03-16 Boehringer Ingelheim Pharma Gmbh & Co. Kg New 3-methyl-7-butynyl xanthines, their preparation and their use as pharmaceuticals
CN1759834B (en) 2004-09-17 2010-06-23 中国医学科学院医药生物技术研究所 Application of berberine or associated with Simvastatin in preparing product for preventing or curing disease or symptom related to blood fat
CA2580461A1 (en) 2004-09-23 2006-04-06 Amgen Inc. Substituted sulfonamidopropionamides and methods of use
RU2007116869A (en) 2004-10-08 2008-11-20 Новартис АГ (CH) COMBINATION OF ORGANIC COMPOUNDS
EP1799639B1 (en) 2004-10-12 2013-09-04 Glenmark Pharmaceuticals S.A. Novel dipeptidyl peptidase iv inhibitors, pharmaceutical compositions containing them, and process for their preparation
AU2005299808B2 (en) 2004-10-25 2009-08-20 Novartis Ag Combination of DPP-IV inhibitor, PPAR antidiabetic and metformin
DE102004054054A1 (en) 2004-11-05 2006-05-11 Boehringer Ingelheim Pharma Gmbh & Co. Kg Process for preparing chiral 8- (3-amino-piperidin-1-yl) -xanthines
DE102005013967A1 (en) 2004-11-05 2006-10-05 Boehringer Ingelheim Pharma Gmbh & Co. Kg New imidazole or pyrimidine derivatives are bradykinin B1 antagonists used for treating e.g. pain, stroke, peptic ulcers and other inflammatory disorders
JP2006137678A (en) 2004-11-10 2006-06-01 Shionogi & Co Ltd Interleukin-2 composition
BRPI0518651A2 (en) 2004-12-24 2008-12-02 Dainippon Sumitomo Pharma compound, a prodrug thereof, or a pharmaceutically acceptable compound or prodrug salt, pharmaceutical composition, dipeptidyl peptidase iv inhibitor, use of a compound, a prodrug thereof or a compound or prodrug salt pharmaceutically acceptable method of treating diabetes
KR100760430B1 (en) 2004-12-31 2007-10-04 한미약품 주식회사 Controlled release complex formulation for oral administration of medicine for diabetes and method for the preparation thereof
MY148521A (en) 2005-01-10 2013-04-30 Arena Pharm Inc Substituted pyridinyl and pyrimidinyl derivatives as modulators of metabolism and the treatment of disorders related thereto
DOP2006000008A (en) 2005-01-10 2006-08-31 Arena Pharm Inc COMBINED THERAPY FOR THE TREATMENT OF DIABETES AND RELATED AFFECTIONS AND FOR THE TREATMENT OF AFFECTIONS THAT IMPROVE THROUGH AN INCREASE IN THE BLOOD CONCENTRATION OF GLP-1
GT200600008A (en) 2005-01-18 2006-08-09 FORMULATION OF DIRECT COMPRESSION AND PROCESS
CN102372705A (en) 2005-02-18 2012-03-14 田边三菱制药株式会社 Salt of proline derivative, solvate thereof, and production method thereof
JP5043825B2 (en) 2005-03-22 2012-10-10 エフ.ホフマン−ラ ロシュ アーゲー Novel salts and polymorphs of DPP-IV inhibitors
JP2008536881A (en) 2005-04-21 2008-09-11 ガストロテック・ファルマ・アクティーゼルスカブ Pharmaceutical formulation of GLP-1 molecule and antiemetic
US7553861B2 (en) 2005-04-22 2009-06-30 Alantos Pharmaceuticals Holding, Inc. Dipeptidyl peptidase-IV inhibitors
WO2006114923A1 (en) 2005-04-25 2006-11-02 Hitachi, Ltd. Inspection equipment employing magnetic resonance
CA2605847C (en) 2005-04-26 2014-02-04 Mitsubishi Tanabe Pharma Corporation Ddp-iv inhibitors for the treatment of postprandial hyperlipidemia
UA91546C2 (en) 2005-05-03 2010-08-10 Бьорінгер Інгельхайм Інтернаціональ Гмбх Crystalline form of 1-chloro-4-(я-d-glucopyranos-1-yl)-2-[4-((s)-tetrahydrofuran-3-yloxy)-benzyl]-benzene, a method for its preparation and the use thereof for preparing medicaments
EP1883631A1 (en) 2005-05-25 2008-02-06 Wyeth Methods of synthesizing substituted 3-cyanoquinolines and intermediates thereof
KR101438234B1 (en) 2005-06-03 2014-09-04 미쓰비시 타나베 파마 코퍼레이션 Concomitant pharmaceutical agents and use thereof
MY152185A (en) 2005-06-10 2014-08-29 Novartis Ag Modified release 1-[(3-hydroxy-adamant-1-ylamino)-acetyl]-pyrrolidine-2(s)-carbonitrile formulation
GT200600218A (en) 2005-06-10 2007-03-28 FORMULATION AND PROCESS OF DIRECT COMPRESSION
BRPI0612301A2 (en) * 2005-06-20 2009-01-27 Decode Genetics Ehf method for diagnosing a susceptibility to type ii diabetes in an individual, kit, and, method for assessing an individual for the likelihood of response to a tcf7l2 therapeutic agent
CN101212966B (en) 2005-07-01 2012-03-14 默沙东公司 Process for synthesizing a CETP inhibitor
EP1904531B1 (en) 2005-07-08 2010-10-06 Pfizer Limited Madcam antibodies
UY29694A1 (en) 2005-07-28 2007-02-28 Boehringer Ingelheim Int METHODS TO PREVENT AND TREAT METABOLIC AND NEW DISORDERS DERIVED FROM PIRAZOL-O-GLUCOSIDO
DE102005035891A1 (en) 2005-07-30 2007-02-08 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8- (3-amino-piperidin-1-yl) -xanthines, their preparation and their use as pharmaceuticals
CA2617327A1 (en) 2005-08-04 2007-02-15 Novartis Ag Salts of vildagliptin
AU2006278039B2 (en) 2005-08-11 2010-10-21 F. Hoffmann-La Roche Ag Pharmaceutical composition comprising a DPP-lV inhibitor
EP1760076A1 (en) 2005-09-02 2007-03-07 Ferring B.V. FAP Inhibitors
PT1931350E (en) 2005-09-14 2014-02-12 Takeda Pharmaceutical Administration of dipeptidyl peptidase inhibitors
EA015169B1 (en) 2005-09-14 2011-06-30 Такеда Фармасьютикал Компани Лимитед Use of dipeptidyl peptidase inhibitors
TW200745079A (en) 2005-09-16 2007-12-16 Takeda Pharmaceuticals Co Polymorphs of benzoate salt of 2-[[6-[(3R)-3-amino-1-piperidinyl]-3,4-dihydro-3-methyl-2,4-dioxo-1(2H)-pyrimidinyl]methyl]-benzonitrile and methods of use therefor
CN102675221A (en) 2005-09-16 2012-09-19 武田药品工业株式会社 Intermediate in method for preparing pyrimidinedione derivative
ES2326391T3 (en) 2005-09-16 2009-10-08 Arena Pharmaceuticals, Inc. METABOLISM MODULATORS AND THE TREATMENT OF DISORDERS RELATED TO THE SAME.
WO2007035665A1 (en) 2005-09-20 2007-03-29 Novartis Ag Use of a dpp-iv inhibitor to reduce hypoglycemic events
WO2007038979A1 (en) 2005-09-22 2007-04-12 Swissco Development Ag Effervescent metformin composition and tablets and granules made therefrom
JOP20180109A1 (en) 2005-09-29 2019-01-30 Novartis Ag New Formulation
CA2625646A1 (en) 2005-10-25 2007-05-03 Merck & Co., Inc. Combination of a dipeptidyl peptidase-4 inhibitor and an anti-hypertensive agent for the treatment of diabetes and hypertension
KR100945632B1 (en) 2005-11-04 2010-03-04 엘에스전선 주식회사 Synthesis of MDH-polymer hybrid paticles
CN101365432B (en) 2005-12-16 2011-06-22 默沙东公司 Pharmaceutical compositions of combinations of dipeptidyl peptidase-4 inhibitors with metformin
CN101341148A (en) 2005-12-21 2009-01-07 霍夫曼-拉罗奇有限公司 New salt and polymorph of DPP-IV inhibitor
GB0526291D0 (en) 2005-12-23 2006-02-01 Prosidion Ltd Therapeutic method
EP1966215A1 (en) 2005-12-23 2008-09-10 Novartis AG Condensed heterocyclic compounds useful as dpp-iv inhibitors
GEP20105033B (en) 2005-12-28 2010-06-25 Takeda Pharmaceuticals Co Therapeutic agent for diabetes
CN101394848A (en) * 2006-01-06 2009-03-25 诺瓦提斯公司 Use of vildagliptin for the treatment of diabetes
BRPI0706423A2 (en) 2006-01-06 2011-03-29 Novartis Ag use of organic compounds
US7745414B2 (en) 2006-02-15 2010-06-29 Boehringer Ingelheim International Gmbh Glucopyranosyl-substituted benzonitrile derivatives, pharmaceutical compositions containing such compounds, their use and process for their manufacture
WO2007099345A1 (en) 2006-03-02 2007-09-07 Betagenon Ab Medical use of bmp-2 and/ or bmp-4
JP2009531456A (en) 2006-03-28 2009-09-03 武田薬品工業株式会社 Preparation of (R) -3-aminopiperidine dihydrochloride
PE20071221A1 (en) 2006-04-11 2007-12-14 Arena Pharm Inc GPR119 RECEPTOR AGONISTS IN METHODS TO INCREASE BONE MASS AND TO TREAT OSTEOPOROSIS AND OTHER CONDITIONS CHARACTERIZED BY LOW BONE MASS, AND COMBINED THERAPY RELATED TO THESE AGONISTS
US8455435B2 (en) 2006-04-19 2013-06-04 Ludwig-Maximilians-Universitat Munchen Remedies for ischemia
EP1852108A1 (en) 2006-05-04 2007-11-07 Boehringer Ingelheim Pharma GmbH & Co.KG DPP IV inhibitor formulations
EP2540725A1 (en) 2006-05-04 2013-01-02 Boehringer Ingelheim International GmbH Polymorphs of 1-((4-Methyl-chinazolin-2-yl)methyl)-3-methyl-7-(2-butin-1-yl)-8-(3-(R)-amino-piperidin-1-yl)xanthin
KR20070111099A (en) 2006-05-16 2007-11-21 영진약품공업주식회사 Novel crystalline form of sitagliptin hydrochloride
EP2020996B1 (en) 2006-05-16 2011-11-23 Gilead Sciences, Inc. Method and compositions for treating hematological malignancies
WO2007137107A2 (en) 2006-05-19 2007-11-29 Abbott Laboratories Inhibitors of diacylglycerol o-acyltransferase type 1 enzyme
KR100858848B1 (en) 2006-05-23 2008-09-17 한올제약주식회사 Pharmaceutical compositions and formulations of Metformin extended release tablets
WO2007149797A2 (en) 2006-06-19 2007-12-27 Novartis Ag Use of organic compounds
WO2007148185A2 (en) 2006-06-21 2007-12-27 Pfizer Products Inc. Substituted 3 -amino- pyrrolidino-4 -lactams as dpp inhibitors
AT503443B1 (en) 2006-06-23 2007-10-15 Leopold Franzens Uni Innsbruck Preparation of an ice surface, useful for ice rink, and ice sports cars and trains, comprises freezing water in which an inorganic substance e.g. ammonia, alkali hydroxide, hydrogen halide, nitric acid and sulfuric acid, is added
EP2035395A2 (en) 2006-06-27 2009-03-18 Glenmark Pharmaceuticals S.A. Novel processes for the preparation of dpp iv inhibitors
TW200811140A (en) 2006-07-06 2008-03-01 Arena Pharm Inc Modulators of metabolism and the treatment of disorders related thereto
TW200811147A (en) 2006-07-06 2008-03-01 Arena Pharm Inc Modulators of metabolism and the treatment of disorders related thereto
US8071583B2 (en) 2006-08-08 2011-12-06 Boehringer Ingelheim International Gmbh Pyrrolo[3,2-D] pyrimidines as DPP-IV inhibitors for the treatment of diabetes mellitus
US8039441B2 (en) 2006-08-15 2011-10-18 Boehringer Ingelheim International Gmbh Glucopyranosyl-substituted cyclopropylbenzene derivatives, pharmaceutical compositions containing such compounds, their use as SGLT inhibitors and process for their manufacture
AU2007285827A1 (en) 2006-08-17 2008-02-21 Wellstat Therapeutics Corporation Combination treatment for metabolic disorders
CL2007002499A1 (en) 2006-08-30 2008-03-14 Phenomix Corp SALES CITRATE AND TARTRATE OF COMPOUNDS DERIVED FROM PIRROLIDINILAMINOACETILPIRROLIDINBORONICO ACID, DPP-IV INHIBITORS; PREPARATION METHOD; SOLID FORM; PHARMACEUTICAL COMBINATION, USEFUL FOR THE TREATMENT OF DIABETES.
DE102006042586B4 (en) 2006-09-11 2014-01-16 Betanie B.V. International Trading Process for the microparticulate loading of high polymer carbohydrates with hydrophobic active fluids
GEP20125701B (en) 2006-09-13 2012-12-10 Takeda Pharmaceuticals Co Application of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2h-pyrimidin-1-ylmethyl]-4-fluoro-benzonitrile
WO2008031750A2 (en) 2006-09-15 2008-03-20 F. Hoffmann-La Roche Ag Process for the preparation of pyrido[2,1-a]isoquinoline derivatives by catalytic asymmetric hydrogenation of an enamine
JP5235018B2 (en) 2006-09-15 2013-07-10 エフ.ホフマン−ラ ロシュ アーゲー Process for producing pyrido [2,1-a] isoquinoline derivative comprising optical resolution of enamine
JP2010508371A (en) 2006-11-06 2010-03-18 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Glucopyranosyl-substituted phenyl derivatives, pharmaceuticals containing the compounds, and uses and production methods thereof
US7956201B2 (en) 2006-11-06 2011-06-07 Hoffman-La Roche Inc. Process for the preparation of (S)-4-fluoromethyl-dihydro-furan-2-one
BRPI0718596B8 (en) 2006-11-09 2021-05-25 Boehringer Ingelheim Int pharmaceutical compositions for combination therapy with sglt-2 and metformin inhibitors
TW200838536A (en) 2006-11-29 2008-10-01 Takeda Pharmaceutical Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2H-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
ES2374952T3 (en) 2006-12-06 2012-02-23 Glaxosmithkline Llc BICYCLIC COMPOUNDS AND USE AS ANTIDIABETICS.
ES2319596B1 (en) 2006-12-22 2010-02-08 Laboratorios Almirall S.A. NEW DERIVATIVES OF THE AMINO-NICOTINIC AND AMINO-ISONICOTINIC ACIDS.
US7638541B2 (en) 2006-12-28 2009-12-29 Metabolex Inc. 5-ethyl-2-{4-[4-(4-tetrazol-1-yl-phenoxymethyl)-thiazol-2-yl]-piperidin-1-yl}-pyrimidine
AR064736A1 (en) 2007-01-04 2009-04-22 Prosidion Ltd GPCR AGONISTS
CL2008000133A1 (en) 2007-01-19 2008-05-23 Boehringer Ingelheim Int PHARMACEUTICAL COMPOSITION THAT INCLUDES A COMPOUND DERIVED FROM PIRAZOL-O-GLUCOSIDE COMBINED WITH AT LEAST A SECOND THERAPEUTIC AGENT; AND USE OF THE COMPOSITION FOR THE TREATMENT OF MELLITUS DIABETES, CATARATS, NEUROPATHY, MYOCARDIAL INFARTS, AND
MY147596A (en) * 2007-02-01 2012-12-31 Takeda Pharmaceutical Solid preparation comprising alogliptin and pioglitazone
WO2008093878A1 (en) 2007-02-01 2008-08-07 Takeda Pharmaceutical Company Limited Tablet preparation without causing a tableting trouble
ES2366000T3 (en) 2007-02-06 2011-10-14 Chelsea Therapeutics, Inc. NEW COMPOUNDS, METHODS FOR THEIR PREPARATION AND USE OF THE SAME.
JP5432720B2 (en) 2007-03-13 2014-03-05 武田薬品工業株式会社 2-[[6-[(3R) -3-Amino-1-piperidinyl] -3,4-dihydro-3-methyl-2,4-dioxo-1 (2H) -pyrimidinyl] methyl] -4-fluorobenzo Solid formulation containing nitrile
US8093236B2 (en) 2007-03-13 2012-01-10 Takeda Pharmaceuticals Company Limited Weekly administration of dipeptidyl peptidase inhibitors
CA2681092A1 (en) 2007-03-15 2008-09-18 Nectid, Inc. Anti-diabetic combinations comprising a slow release biguanide composition and an immediate release dipeptidyl peptidase iv inhibitor composition
WO2008114857A1 (en) 2007-03-22 2008-09-25 Kyorin Pharmaceutical Co., Ltd. Method for producing aminoacetylpyrrolidinecarbonitrile derivative
KR20120030570A (en) 2007-04-03 2012-03-28 미쓰비시 타나베 파마 코퍼레이션 Combined use of dipeptidyl peptidase iv inhibitor compound and sweetener
EP2142113B1 (en) 2007-04-16 2023-01-11 Smith & Nephew, Inc. Powered surgical system
PE20090696A1 (en) 2007-04-20 2009-06-20 Bristol Myers Squibb Co CRYSTALLINE FORMS OF SAXAGLIPTIN AND PROCESSES FOR PREPARING THEM
US7910583B2 (en) 2007-05-04 2011-03-22 Bristol-Myers Squibb Company [6,6] and [6,7]-bicyclic GPR119 G protein-coupled receptor agonists
CA2688721A1 (en) 2007-05-21 2008-11-27 Phenomix Corporation Stable pharmaceutical formulation for a dpp-iv inhibitor
US8440172B2 (en) 2007-07-09 2013-05-14 Symrise Ag Stable soluble salts of phenylbenzimidazole sulfonic acid at PHS at or below 7.0
TW200927144A (en) 2007-07-12 2009-07-01 Phenomix Corp A crystalline synthetic intermediate for preparation of a DPP-IV inhibitor and method of purification thereof
WO2009011451A1 (en) 2007-07-19 2009-01-22 Takeda Pharmaceutical Company Limited Solid preparation comprising alogliptin and metformin hydrochloride
PE20090938A1 (en) 2007-08-16 2009-08-08 Boehringer Ingelheim Int PHARMACEUTICAL COMPOSITION INCLUDING A BENZENE DERIVATIVE SUBSTITUTED WITH GLUCOPYRANOSIL
CL2008002425A1 (en) 2007-08-16 2009-09-11 Boehringer Ingelheim Int Pharmaceutical composition comprising an inhibitor of sglt2 and 1- (4-methyl-quinazolin-2-yl) methyl-3-methyl-7 - (- 2-butin-1-yl) -8- (3- (r) -amino- Piperidin-1yl) -xanthine, an iv dpp inhibitor and its use for the treatment of obesity and type 1 and 2 diabetes and complications thereof.
UY31290A1 (en) 2007-08-16 2009-03-31 PHARMACEUTICAL COMPOSITION THAT INCLUDES A DERIVATIVE OF PIRAZOL-O-GLUCOSIDO
PE20090597A1 (en) 2007-08-16 2009-06-06 Boehringer Ingelheim Int PHARMACEUTICAL COMPOSITION INCLUDING A DERIVATIVE OF PIRAZOL-O-GLUCOSIDE
EP3542801A1 (en) 2007-08-17 2019-09-25 Boehringer Ingelheim International GmbH Purin derivatives for use in the treatment of fap-related diseases
GB2465132B (en) 2007-09-21 2012-06-06 Lupin Ltd Compounds as dipeptidyl peptidase IV (DPP IV) inhibitors
DK2597103T3 (en) 2007-11-16 2017-02-13 Novo Nordisk As Stable pharmaceutical compositions comprising liraglutide and degludec
TW200938200A (en) 2007-12-28 2009-09-16 Dainippon Sumitomo Pharma Co Methyl-substituted piperidine derivative
CN101234105A (en) 2008-01-09 2008-08-06 北京润德康医药技术有限公司 Pharmaceutical composition containing diabetosan and vildagliptin and preparation thereof
WO2009091663A1 (en) 2008-01-14 2009-07-23 Phenomix Corporation Stable pharmaceutical formulation of a dpp-iv inhibitor with metformin
CL2008003653A1 (en) 2008-01-17 2010-03-05 Mitsubishi Tanabe Pharma Corp Use of a glucopyranosyl-derived sglt inhibitor and a selected dppiv inhibitor to treat diabetes; and pharmaceutical composition.
US20090186086A1 (en) 2008-01-17 2009-07-23 Par Pharmaceutical, Inc. Solid multilayer oral dosage forms
TW200936136A (en) 2008-01-28 2009-09-01 Sanofi Aventis Tetrahydroquinoxaline urea derivatives, their preparation and their therapeutic application
JP2011510986A (en) 2008-02-05 2011-04-07 メルク・シャープ・エンド・ドーム・コーポレイション Combination pharmaceutical composition of metformin and dipeptidyl peptidase-IV inhibitor
WO2009111200A1 (en) 2008-03-04 2009-09-11 Merck & Co., Inc. Pharmaceutical compositions of a combination of metformin and a dipeptidyl peptidase-iv inhibitor
GEP20135844B (en) 2008-03-05 2013-06-10 Takeda Pharmaceutical Heterocyclic compound
US8551524B2 (en) 2008-03-14 2013-10-08 Iycus, Llc Anti-diabetic combinations
MX2010010562A (en) 2008-03-31 2010-12-07 Metabolex Inc Oxymethylene aryl compounds and uses thereof.
PE20140960A1 (en) * 2008-04-03 2014-08-15 Boehringer Ingelheim Int FORMULATIONS INVOLVING A DPP4 INHIBITOR
WO2009128360A1 (en) 2008-04-18 2009-10-22 大日本住友製薬株式会社 Therapeutic agent for diabetes
EP2275108B1 (en) 2008-05-14 2014-07-09 Sanwa Kagaku Kenkyusho Co., Ltd Pharmaceutical preparation comprising dpp-iv inhibitor and other diabetes therapeutic agent in concomitant or combined form
PE20100156A1 (en) 2008-06-03 2010-02-23 Boehringer Ingelheim Int NAFLD TREATMENT
KR20190016601A (en) 2008-08-06 2019-02-18 베링거 인겔하임 인터내셔날 게엠베하 Treatment for diabetes in patients inappropriate for metformin therapy
UY32030A (en) 2008-08-06 2010-03-26 Boehringer Ingelheim Int "TREATMENT FOR DIABETES IN INAPPROPRIATE PATIENTS FOR THERAPY WITH METFORMIN"
PE20110297A1 (en) 2008-08-15 2011-05-26 Boehringer Ingelheim Int DPP-4 INHIBITORS FOR WOUND HEALING
JP2010053576A (en) 2008-08-27 2010-03-11 Sumitomo Forestry Co Ltd Mat for paving
RU2011113823A (en) 2008-09-10 2012-10-20 БЕРИНГЕР ИНГЕЛЬХАЙМ ИНТЕРНАЦИОНАЛЬ ГмбХ (DE) COMBINED THERAPY FOR THE TREATMENT OF DIABETES AND RELATED CONDITIONS
UY32177A (en) 2008-10-16 2010-05-31 Boehringer Ingelheim Int TREATMENT OF DIABETES IN PATIENTS WITH INSUFFICIENT GLUCEMIC CONTROL TO WEIGHT THERAPY WITH DRUG, ORAL OR NOT, ANTIDIABÉTICO
WO2010045656A2 (en) 2008-10-17 2010-04-22 Nectid, Inc. Novel sglt2 inhibitor dosage forms
JP2012512848A (en) 2008-12-23 2012-06-07 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Salt forms of organic compounds
TW201036975A (en) 2009-01-07 2010-10-16 Boehringer Ingelheim Int Treatment for diabetes in patients with inadequate glycemic control despite metformin therapy
TWI466672B (en) 2009-01-29 2015-01-01 Boehringer Ingelheim Int Treatment for diabetes in paediatric patients
CA2752437C (en) 2009-02-13 2017-07-11 Boehringer Ingelheim International Gmbh Antidiabetic medications
NZ594487A (en) 2009-02-13 2013-11-29 Boehringer Ingelheim Int Pharmaceutical composition comprising a sglt2 inhibitor, a dpp-iv inhibitor and optionally a further antidiabetic agent and uses thereof
UY32427A (en) 2009-02-13 2010-09-30 Boheringer Ingelheim Internat Gmbh PHARMACEUTICAL COMPOSITION, PHARMACEUTICAL FORM, PROCEDURE FOR PREPARATION, METHODS OF TREATMENT AND USES OF THE SAME
TW201031661A (en) 2009-02-17 2010-09-01 Targacept Inc Fused benzazepines as neuronal nicotinic acetylcholine receptor ligands
CA2754523A1 (en) 2009-03-20 2010-09-23 Pfizer Inc. 3-oxa-7-azabicyclo[3.3.1]nonanes
US8815292B2 (en) 2009-04-27 2014-08-26 Revalesio Corporation Compositions and methods for treating insulin resistance and diabetes mellitus
US20120100221A1 (en) 2009-06-02 2012-04-26 Ranbaxy Laboratories Limited Pharmaceutical compositions containing a combination of an antihistamine and a decongestant
WO2010147768A1 (en) 2009-06-15 2010-12-23 Merck Sharp & Dohme Corp. Pharmaceutical compositions of combinations of dipeptidyl peptidase-4 inhibitors with pioglitazone
CA2768656C (en) 2009-07-21 2016-10-04 Keryx Biopharmaceuticals, Inc. Ferric citrate dosage forms
CN102596191B (en) 2009-10-02 2016-12-21 勃林格殷格翰国际有限公司 Comprise the pharmaceutical composition of BI 1356 and metformin
US10610489B2 (en) 2009-10-02 2020-04-07 Boehringer Ingelheim International Gmbh Pharmaceutical composition, pharmaceutical dosage form, process for their preparation, methods for treating and uses thereof
JP5446716B2 (en) 2009-10-21 2014-03-19 大正製薬株式会社 Method for producing tablets containing arginine and carnitine
NZ599298A (en) 2009-11-27 2014-11-28 Boehringer Ingelheim Int Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin
JP2010070576A (en) 2009-12-28 2010-04-02 Sato Pharmaceutical Co Ltd Rapidly soluble tablet
TWI562775B (en) 2010-03-02 2016-12-21 Lexicon Pharmaceuticals Inc Methods of using inhibitors of sodium-glucose cotransporters 1 and 2
WO2011113947A1 (en) 2010-03-18 2011-09-22 Boehringer Ingelheim International Gmbh Combination of a gpr119 agonist and the dpp-iv inhibitor linagliptin for use in the treatment of diabetes and related conditions
EA201201509A1 (en) 2010-05-05 2013-04-30 Бёрингер Ингельхайм Интернациональ Гмбх PHARMACEUTICAL COMPOSITIONS CONTAINING PIOGLITAZONE AND LINAGLIPTIN
JP6034781B2 (en) 2010-05-05 2016-11-30 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Combination therapy
EP2579879B1 (en) 2010-06-09 2016-03-23 Poxel Triazine derivatives for delaying the onset of type 1 diabetes
BR112012032816A2 (en) 2010-06-22 2016-11-08 Twi Pharmaceuticals controlled release pharmaceutical composition, and methods for reducing the dietary effect of a controlled release composition, reducing the amount of time required to achieve a stable state for metformin, to improve the bioavailability of a controlled release dosage form. matrix
EP3725325B1 (en) 2010-06-24 2023-05-31 Boehringer Ingelheim International GmbH Diabetes therapy
JP5837072B2 (en) 2010-09-03 2015-12-24 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Formulations that use water-soluble antioxidants
US9034883B2 (en) 2010-11-15 2015-05-19 Boehringer Ingelheim International Gmbh Vasoprotective and cardioprotective antidiabetic therapy
WO2012088682A1 (en) 2010-12-29 2012-07-05 Shanghai Fochon Pharmaceutical Co Ltd. 2-(3-aminopiperidin-1-yl)-[1,2,4]triazolo[1,5-c]pyrimidine-5,7(3h,6h)-dione derivates as dipeptidyl peptidase iv(dpp-iv) inhibitors
CN103442697A (en) 2011-02-01 2013-12-11 百时美施贵宝公司 Pharmaceutical formulations including an amine compound
UY33937A (en) 2011-03-07 2012-09-28 Boehringer Ingelheim Int PHARMACEUTICAL COMPOSITIONS CONTAINING DPP-4 AND / OR SGLT-2 AND METFORMIN INHIBITORS
JP6028016B2 (en) 2011-05-10 2016-11-16 サンド・アクチエンゲゼルシヤフト Polymorphs of linagliptin benzoate
DK2731947T3 (en) 2011-07-15 2019-04-23 Boehringer Ingelheim Int SUBSTITUTED DIMERIC QUINAZOLINE DERIVATIVE, PREPARATION AND USE thereof IN PHARMACEUTICAL COMPOSITIONS FOR TREATMENT OF TYPE I AND TYPE II DIABETES
US8849828B2 (en) 2011-09-30 2014-09-30 International Business Machines Corporation Refinement and calibration mechanism for improving classification of information assets
US20130172244A1 (en) 2011-12-29 2013-07-04 Thomas Klein Subcutaneous therapeutic use of dpp-4 inhibitor
EP2800803A1 (en) 2012-01-04 2014-11-12 The Procter and Gamble Company Active containing fibrous structures with multiple regions
US9555001B2 (en) 2012-03-07 2017-01-31 Boehringer Ingelheim International Gmbh Pharmaceutical composition and uses thereof
ES2929025T3 (en) 2012-05-14 2022-11-24 Boehringer Ingelheim Int Linagliptin, a xanthine derivative as a dpp-4 inhibitor, for use in the treatment of SIRS and/or sepsis
EP2849755A1 (en) 2012-05-14 2015-03-25 Boehringer Ingelheim International GmbH A xanthine derivative as dpp -4 inhibitor for use in the treatment of podocytes related disorders and/or nephrotic syndrome
WO2013174767A1 (en) 2012-05-24 2013-11-28 Boehringer Ingelheim International Gmbh A xanthine derivative as dpp -4 inhibitor for use in modifying food intake and regulating food preference
WO2013174768A1 (en) 2012-05-24 2013-11-28 Boehringer Ingelheim International Gmbh A xanthine derivative as dpp -4 inhibitor for use in the treatment of autoimmune diabetes, particularly lada
EP2854824A1 (en) 2012-05-25 2015-04-08 Boehringer Ingelheim International GmbH Use of keratinocytes as a biologically active substance in the treatment of wounds, such as diabetic wounds, optionally in combination with a dpp-4 inhibitor
WO2013179307A2 (en) 2012-05-29 2013-12-05 Mylan Laboratories Limited Stabilized pharmaceutical compositions of saxagliptin
EP2908863A1 (en) 2012-10-09 2015-08-26 Boehringer Ingelheim International GmbH Use of moisture-conditioned disintegrants in tablet manufacture
EP2908806A1 (en) 2012-10-09 2015-08-26 Boehringer Ingelheim International GmbH Use of selectively moisture-adjusted tabletting material in the production of mechanically stable tablets which contain at least one hydrate-forming active substance and/or adjuvant relevant to the mechanical stability of the tablets, particularly arginine-containing tablets
US9050302B2 (en) 2013-03-01 2015-06-09 Jazz Pharmaceuticals Ireland Limited Method of administration of gamma hydroxybutyrate with monocarboxylate transporters
WO2014140284A1 (en) 2013-03-15 2014-09-18 Boehringer Ingelheim International Gmbh Use of linagliptin in cardio- and renoprotective antidiabetic therapy
ES2906115T3 (en) 2013-04-18 2022-04-13 Boehringer Ingelheim Int Pharmaceutical composition, treatment methods and uses thereof
EP2996724A1 (en) 2013-05-17 2016-03-23 Boehringer Ingelheim International GmbH Combination of a dpp-4 inhibitor and an alpha-glucosidase inhibitor
US20140371243A1 (en) 2013-06-14 2014-12-18 Boehringer Ingelheim International Gmbh Medical use of a dpp-4 inhibitor
JP6615109B2 (en) 2014-02-28 2019-12-04 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Medical use of DPP-4 inhibitors
WO2016059219A1 (en) 2014-10-17 2016-04-21 Boehringer Ingelheim International Gmbh Pharmaceutical composition and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070281940A1 (en) * 2006-05-04 2007-12-06 Klaus Dugi Uses of dpp-iv inhibitors

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Elrishi Practical Diabetes International 24(9) 474-482 (2007)-cited in IDS filed 10/23/2019 *
Florez New England Journal of Medicine 355 241-250 (2006)-cited in IDS filed 10/23/2019 *
H ttner et al Journal of Clinical Pharmacology 48 1171-1178 (2008)-cited in IDS filed 10/23/2019 *

Also Published As

Publication number Publication date
CA2782179C (en) 2020-06-23
EP2504002B1 (en) 2019-10-09
ES2760917T3 (en) 2020-05-18
JP6104989B2 (en) 2017-03-29
AU2010323068B2 (en) 2015-09-03
MX2019005130A (en) 2020-11-12
JP2015164964A (en) 2015-09-17
KR102328772B1 (en) 2021-11-19
CN102753161A (en) 2012-10-24
CL2012001337A1 (en) 2012-08-17
MX2012006110A (en) 2012-06-14
EA201200793A1 (en) 2014-04-30
KR20190071840A (en) 2019-06-24
AU2010323068A1 (en) 2012-05-03
US9457029B2 (en) 2016-10-04
JP2013512229A (en) 2013-04-11
US20160354380A1 (en) 2016-12-08
WO2011064352A1 (en) 2011-06-03
EP2504002A1 (en) 2012-10-03
KR20170136017A (en) 2017-12-08
KR20210033559A (en) 2021-03-26
US20130196898A1 (en) 2013-08-01
EA034869B1 (en) 2020-03-31
EP3646859A1 (en) 2020-05-06
KR20120107080A (en) 2012-09-28
US10092571B2 (en) 2018-10-09
BR112012012641A2 (en) 2020-08-11
US20190000855A1 (en) 2019-01-03
MX364651B (en) 2019-05-03
IL219014A0 (en) 2012-06-28
CN107115530A (en) 2017-09-01
KR20230021159A (en) 2023-02-13
CA2782179A1 (en) 2011-06-03
NZ599298A (en) 2014-11-28

Similar Documents

Publication Publication Date Title
US10092571B2 (en) Treatment of genotyped diabetic patients with DPP-IV inhibitors such as linagliptin
US20220088023A1 (en) Antidiabetic medications
US10195203B2 (en) Use of a DPP-4 inhibitor in podocytes related disorders and/or nephrotic syndrome
EA042990B1 (en) TREATMENT OF GENOTYPED DIABETES PATIENTS WITH DPP-4 INHIBITORS SUCH AS LINAGLIPTIN

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION