US20110201623A1 - Crystalline Form Of A Cyclopropyl Benzamide Derivative - Google Patents

Crystalline Form Of A Cyclopropyl Benzamide Derivative Download PDF

Info

Publication number
US20110201623A1
US20110201623A1 US13/029,362 US201113029362A US2011201623A1 US 20110201623 A1 US20110201623 A1 US 20110201623A1 US 201113029362 A US201113029362 A US 201113029362A US 2011201623 A1 US2011201623 A1 US 2011201623A1
Authority
US
United States
Prior art keywords
compound
crystalline form
disorder
2theta
angstroms
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
US13/029,362
Inventor
Michael A. UCZYNSKI
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.)
AstraZeneca AB
Original Assignee
AstraZeneca AB
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=44370080&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110201623(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by AstraZeneca AB filed Critical AstraZeneca AB
Priority to US13/029,362 priority Critical patent/US20110201623A1/en
Assigned to ASTRAZENECA PHARMACEUTICALS LP reassignment ASTRAZENECA PHARMACEUTICALS LP ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UCZYNSKI, MICHAEL A.
Assigned to ASTRAZENECA AB reassignment ASTRAZENECA AB ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ASTRAZENECA PHARMACEUTICALS LP
Publication of US20110201623A1 publication Critical patent/US20110201623A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/64Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings
    • C07C233/65Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings having the nitrogen atoms of the carboxamide groups bound to hydrogen atoms or to carbon atoms of unsubstituted hydrocarbon radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/192Radicals derived from carboxylic acids from aromatic carboxylic acids
    • 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
    • A61K31/4965Non-condensed pyrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C231/00Preparation of carboxylic acid amides
    • C07C231/06Preparation of carboxylic acid amides from nitriles by transformation of cyano groups into carboxamide groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C231/00Preparation of carboxylic acid amides
    • C07C231/14Preparation of carboxylic acid amides by formation of carboxamide groups together with reactions not involving the carboxamide groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/70Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/84Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups and doubly-bound oxygen atoms bound to the same carbon skeleton with the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/04Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/185Radicals derived from carboxylic acids from aliphatic carboxylic acids

Definitions

  • the present invention relates to crystalline forms of a compound (I), 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide, particularly such as Form I, pharmaceutical formulations containing said compound and to the use of said active compounds in therapy.
  • the crystalline forms of 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide are useful to treat at least one histamine H3 receptor associated condition.
  • the histamine H3 receptor is of current interest in developing new medicaments.
  • the H3 receptor is a presynaptic autoreceptor located both in the central and peripheral nervous systems, the skin, and in organs, such as, for example, the lung, the intestine, probably the spleen, and the gastrointestinal tract.
  • the H3 receptor has intrinsic, constitutive activity in vitro as well as in vivo (i.e., it is active in the absence of an agonist). Compounds acting as inverse agonists can inhibit this activity.
  • the histamine H3 receptor has been shown to regulate the release of histamine and also of other neurotransmitters, such as, for example, serotonin and acetylcholine.
  • histamine H3 ligands such as, for example, a histamine H3 receptor antagonist or inverse agonist may increase the release of neurotransmitters in the brain, whereas other histamine H3 ligands, such as, for example, histamine H3 receptor agonists may inhibit the biosynthesis of histamine, as well as, inhibit the release of neurotransmitters. This suggests that histamine H3 receptor agonists, inverse agonists, and antagonists could mediate neuronal activity. As a result, efforts have been undertaken to develop new therapeutics that target the histamine H3 receptor.
  • WO2009/024823 describes the synthesis of a number of cyclopropyl amide derivatives, such as, for example, 4-((trans)-2-[(4-cyclobutylpiperazin-yl)carbonyl]-cyclopropyl ⁇ -benzamide (enantiomer 1; Example 43).
  • One object of the present invention is to provide crystalline forms of compound (I), 4- ⁇ (1S, 2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide.
  • Another object of the present invention is to provide a crystalline form of compound (I), 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide, as Form I.
  • Said compound (I) having a histamine receptor antagonist or inverse agonist effect at the H3 receptor which making them suitable to be formulated into pharmaceutical is formulations.
  • the present invention provides a crystalline form of compound (I), 4- ⁇ (1S, 2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide
  • One aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with at least one peak at about 18.3 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Another aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with at least two peaks at about 4.9 and about 18.3 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Yet another aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with at least three peaks at about 4.9, about 18.3 and about 20.4 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still another aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with at least four peaks at about 4.9, about 18.3, about 19.6 and about 20.4 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with peaks at selected ° 2 Theta-values described above and with additional peaks at about 16.4 and about 16.6, °2Theta when measured using radiation with a wavelength of about 1.54 angstroms.
  • Yet a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with peaks at selected °2Theta-values described above and with an additional peak at about 5.3 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with peaks at °2Theta-values selected from about 4.9, about 16.4, about 16.6, about 18.3, about 19.6 and about 20.4, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with peaks at °2Theta-values selected from about 4.9, about 5.3, about 16.4, about 16.6, about 18.3, about 19.6 and about 20.4, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with peaks at °2Theta-values selected from about 4.9, about 12.6, about 16.4, about 16.6, about 18.3, about 19.6, about 20.4, and about 23.2, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Cu K ⁇ XRDP pattern
  • Still a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu K ⁇ ) with peaks at °2Theta-values selected from about 4.9, about 5.3, about 9.0, about 12.6, about 16.4, about 16.6, about 18.3, about 19.6, about 20.4, and about 23.2, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Cu K ⁇ XRDP pattern
  • Still a further aspect of the invention relates to a crystalline form of compound (I), is characterized in that said form has an XRDP pattern (Cu K ⁇ ) with peaks at from about 4.9, about 5.3, about 9.0, about 12.6, about 16.4, about 16.6, about 18.3, about 19.6, about 20.4, about 21.2, about 23.2 and about 24.6 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Cu K ⁇ XRDP pattern
  • Yet still a further aspect of the invention relates to relates to a crystalline form of compound (I), characterized by the X-ray powder diffraction pattern essentially as shown in FIG. 1 .
  • Another embodiment relates to a crystalline form of compound (I) that has a DSC thermogram essentially as depicted in FIG. 2 .
  • a crystalline form of compound (I) has a DSC thermogram comprising an endothermic event with an onset temperature of about 225° C.
  • a crystalline form of compound (I) has a DSC thermogram comprising an endothermic event with a peak temperature of about 235° C.
  • DSC onset and peak temperatures as well as energy values may vary due to, for example, the purity of the sample and sample size and due to instrumental parameters, especially the temperature scan rate.
  • the DSC data presented are not to be taken as absolute values.
  • a person skilled in the art can set up instrumental parameters for a Differential scanning calorimeter so that data comparable to the data presented here can be collected according to standard methods, for example those described in Höhne, G. W. H. et al (1996), Differential Scanning calorimetry, Springer, Berlin.
  • the crystalline forms of compound (I) of the present invention may also exist as solvates, including hydrates.
  • the present invention also relates to the use of a crystalline form of compound (I), as hereinbefore defined.
  • a crystalline form of compound (I), as hereinbefore defined has one or more advantageous properties.
  • a crystalline form of compound (I) shows advantageous properties, such as, for example, a high melting point, a substantial lack of solvent (e.g., water) content, little or no weight loss on heating, and/or low hygroscopicity.
  • solvent e.g., water
  • such properties advantageously facilitate the manufacture, storage, formulation, and/or delivery of compound (I).
  • a crystalline form of compound (I), as described herein, for example Form I of compound (I) provide advantageous properties with regard to stability.
  • a substance can be expected to be more stable chemically in a crystalline state in comparison with the same substance in an amorphous state, as described in Haleblian and McCrone J. Pharm. Sci 1969, 58, pages 911-929, especially page 913.
  • This observation is common for small molecules (i.e. non-proteins) but not always true for macromolecules like proteins, as described in Pikal and Rigsbee, Pharm. Res. 1997, 14, pages 1379-1387, especially page 1379.
  • a crystalline state is thus beneficial for small molecules such as compound (I).
  • X-rays will be scattered by electrons in atoms in a substance. Crystalline material will diffract X-rays giving peaks in directions of constructive interference. The directions are determined by the crystal structure, including the size and shape of the unit cell. All diffraction peak °2Theta values disclosed and/or claimed herein refer to Cu K ⁇ -radiation. An amorphous (non-crystalline) material will not give such diffraction peaks. See e.g. Klug, H. P. & Alexander, L. E., X-Ray Diffraction Procedures For Polycrystalline and Amorphous Materials, 1974, John Wiley & Sons.
  • a pharmaceutical formulation comprising a crystalline form of the compound (I), such as Form I, for use in the prevention and/or treatment of conditions associated with the H3 receptor.
  • the formulation used in accordance with the present invention may be in a form suitable for oral administration, for example such as a tablet, pill, powder, granule or capsule, for parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion), for topical administration for example such as an ointment, patch or cream, for rectal administration for example such as a suppository and for other non-parenteral administration.
  • parenteral injection including intravenous, subcutaneous, intramuscular, intravascular or infusion
  • topical administration for example such as an ointment, patch or cream
  • rectal administration for example such as a suppository and for other non-parenteral administration.
  • Suitable daily doses a crystalline form of the compound (I), such as Form I, in the treatment of a mammal, including human, are approximately 0.01 to 250 mg/kg bodyweight at per oral administration and about 0.001 to 250 mg/kg bodyweight at parenteral administration.
  • the typical daily dose of the active ingredients varies within a wide range and will depend on various factors such as the relevant indication, the route of administration, the age, weight and sex of the patient and may be determined by a physician.
  • a crystalline form of compound (I), such as for example Form I, may be used on its own but will usually be administered in the form of a pharmaceutical formulation in which the active ingredient is in association with pharmaceutically acceptable diluents, excipients and/or inert carrier known to a person skilled in the art.
  • the pharmaceutical formulation may comprise from 0.05 to 99% w (percent by weight), for example from 0.10 to 50% w, of active ingredient, all percentages by weight being based on total composition.
  • the invention further provides a process for the preparation of a pharmaceutical formulation of the invention which comprises mixing of a crystalline form of the compound (I), such as form I, as hereinbefore defined, with pharmaceutically acceptable diluents, excipients and/or inert carriers.
  • At least one crystalline form of the compound (I) described herein may be administered to a mammal, including human, to be used to modulate at least one histamine H3 receptor.
  • modulate refers to, for example, the activation (e.g., agonist activity) or inhibition (e.g., antagonist and inverse agonist activity) of at least one histamine H3 receptor.
  • at least one crystalline form of the compound (I) described herein may be administered to a mammal, including human, to be used as an inverse agonist of at least one histamine H3 receptor.
  • At least one crystalline form of the compound (I) described herein may be administered to a mammal, including human, to be used as an antagonist of at least one histamine H3 receptor. In another embodiment, at least one crystalline form of the compound (I) described herein may be used as an antagonist of at least one histamine H3 receptor. In yet another embodiment, at least one crystalline form of the compound (I) described herein may be used an antagonist of at least one histamine H3 receptor.
  • At least one crystalline form of the compound (I) described herein may be administered to a mammal, including human, to be used to treat one or more of a wide range of conditions or disorders in which modulating the histamine H3 receptor is beneficial. At least one crystalline form of the compound (I) described herein may administered to a mammal, including human, to be, for example, be useful to treat at least one disease of the central nervous system, the peripheral nervous system, the cardiovascular system, the pulmonary system, the gastrointestinal system, or the endocrinological system.
  • Another embodiment provides a method for treating a disorder in which modulating the function of at least one histamine H3 receptor is beneficial comprising administering to a warm-blooded animal in need of such treatment a therapeutically effective amount of crystalline form of the compound (I).
  • One embodiment relates to the use of the crystalline form of a compound (I) in the manufacture of a medicament for the treatment of at least one disorder selected from schizophrenia, narcolepsy, excessive daytime sleepiness, obesity, attention deficit hyperactivity disorder, pain, neuropathic pain, Alzheimer's disease, cognition deficiency, and cognition deficiency associated with schizophrenia.
  • a further embodiment relates to a method for the therapy of at least one disorder selected from schizophrenia, narcolepsy, excessive daytime sleepiness, obesity, attention deficit hyperactivity disorder, pain, neuropathic pain, Alzheimer's disease, cognition deficiency, and cognition deficiency associated with schizophrenia, in a warm-blooded animal in need of such therapy, wherein the method comprises administering to the animal a therapeutically effective amount of a crystalline form of the compound (I).
  • a crystalline form of the compound (I) may be useful to treat at least one autoimmune disorder.
  • autoimmune disorders include, but are not limited to, for example, arthritis, skin grafts, organ transplants and similar surgical needs, collagen diseases, various allergies, tumors and viruses.
  • a crystalline form of the compound (I) may be useful to treat at least one psychiatric disorder.
  • Exemplary psychiatric disorders include, but are not limited to, for example, Psychotic Disorder(s) and Schizophrenia Disorder(s), such as, for example, Schizoaffective Disorder(s), Delusional Disorder(s), Brief Psychotic Disorder(s), Shared Psychotic Disorder(s), and Psychotic Disorder(s) Due to a General Medical Condition; Dementia and other Cognitive Disorder(s); Anxiety Disorder(s), such as, for example, Panic Disorder(s) Without Agoraphobia, Panic Disorder(s) With Agoraphobia, Agoraphobia Without History of Panic Disorder(s), Specific Phobia, Social Phobia, Obsessive-Compulsive Disorder(s), Stress related Disorder(s), Posttraumatic Stress Disorder(s), Acute Stress Disorder(s), Generalized Anxiety Disorder(s) and Generalized Anxiety Disorder(s) Due to a General Medical Condition; Mood Disorder(s), such as, for example, a) Depressive Disorder(s) (including but not limited to, for example, Major Depressive Disorder
  • a crystalline form of the compound (I) may be useful: i) to treat obesity or being overweight (e.g., promotion of weight loss and maintenance of weight loss), eating disorders (e.g., binge eating, anorexia, bulimia and compulsive), and/or cravings (for drugs, tobacco, alcohol, any appetizing macronutrients or non-essential food items); ii) to prevent weight gain (e.g., medication-induced or subsequent to cessation of smoking); and/or iii) to modulate appetite and/or satiety.
  • At least one solid form described herein may be suitable for treating obesity by reducing appetite and body weight and/or maintaining weight reduction and preventing rebound.
  • At least one solid form described herein may be used to prevent or reverse medication-induced weight gain, e.g., weight gain caused by antipsychotic (neuroleptic) treatment(s); and/or weight gain associated with smoking cessation.
  • a crystalline form of the compound (I) may be useful to treat at least one Neurodegenerative Disorder.
  • Exemplary Neurodegenerative Disorders include, but are not limited to, for example, conditions associated with cognitive disorder(s) or indications with deficit(s) in cognition is such as: dementia; incl. pre-senile dementia (early onset Alzheimer's Disease); senile dementia (dementia of the Alzheimer's type); Alzheimer's Disease (AD); Familial Alzheimer's disease; Early Alzheimer's disease; mild to moderate dementia of the Alzheimer's type; delay of disease progression of Alzheimer's Disease; neurodegeneration associated with Alzheimer's disease, Mild Cognitive Impairment (MCI); Amnestic Mild Cognitive Impairment (aMCI); Age-associated Memory Impairment (AAMI); Lewy body dementia; vascular dementia (VD); HIV-dementia; AIDS dementia complex; AIDS—Neurological Complications; Frontotemporal dementia (FTD); Frontotemporal dementia Parkinson's Type (FTDP); dementia pugilistica; dementia due to infectious agents or metabolic disturbances; dementia of degenerative origin; dementia-Multi-Infarc
  • a crystalline form of the compound (I) may be useful to treat at least one Neuroinflammatory Disorder including, but not limited to, for example, Multiple Sclerosis (MS), which includes, but is not limited to, for example, Relapse Remitting Multiple Sclerosis (RRMS), Secondary Progressive Multiple Sclerosis (SPMS), and Primary Progressive Multiple Sclerosis (PPMS); Parkinson's disease; Multiple System Atrophy is (MSA); Corticobasal Degeneration; Progressive Supranuclear Paresis; Guillain-Barré Syndrome (GBS); and chronic inflammatory demyelinating polyneuropathy (CIDP).
  • MS Multiple Sclerosis
  • RRMS Relapse Remitting Multiple Sclerosis
  • SPMS Secondary Progressive Multiple Sclerosis
  • PPMS Primary Progressive Multiple Sclerosis
  • Parkinson's disease Multiple System Atrophy is
  • MSA Corticobasal Degeneration
  • Progressive Supranuclear Paresis Progressive Supranuclear Paresis
  • GBS Guillain-Barré Syndrome
  • CIDP chronic inflammatory demyelinating polyn
  • a crystalline form of the compound (I) may be useful to treat at least one Attention-Deficit and Disruptive Behavior Disorder.
  • Attention-Deficit and Disruptive Behavior Disorders include, but are not limited to, for example, attention deficit disorder (ADD), attention deficit hyperactivity disorder (ADHD), and affective disorders.
  • ADD attention deficit disorder
  • ADHD attention deficit hyperactivity disorder
  • affective disorders include, but are not limited to, for example, attention deficit disorder (ADD), attention deficit hyperactivity disorder (ADHD), and affective disorders.
  • a crystalline form of the compound (I) may be useful to treat pain, including acute or chronic pain disorders including but not limited to, for example, Widespread pain, Localized pain, Nociceptive pain, Inflammatory pain, Central pain, Central and peripheral neuropathic pain, Diabetic neuropathic pain, Central and peripheral neurogenic pain, Central and peripheral neuralgia, Low back pain, Postoperative pain, Visceral pain, and Pelvic pain; Allodynia; Anesthesia dolorosa; Causalgia; Dysesthesia; Fibromyalgia; Hyperalgesia; Hyperesthesia; Hyperpathia; Ischemic pain; Sciatic pain; Burn-induced pain; Pain associated with cystitis including, but not limited to, interstitial cystitis; Pain associated with multiple sclerosis; Pain associated with arthritis; Pain associated with osteoarthritis; Pain associated with rheumatoid arthritis; Pain associated with pancreatitis; Pain associated with psoriasis; Pain associated with fibromyalgia; Pain
  • a crystalline form of the compound (I) may be useful to treat at least one of the following disorders Autism, Dyslexia, Jetlag, Hyperkinesias, Dystonias, Rage outbursts, Muscular Dystrophy, Neurofibromatosis, Spinal Cord Injury, Cerebral Palsy, Neurological Sequelae of Lupus and Post-Polio Syndrome.
  • a crystalline form of the compound (I) may be used for the manufacture of a medicament for the treatment of at least one autoimmune disorder, psychiatric disorder, obesity disorder, eating disorder, craving disorder, neurodegenerative disorder, neuroinflammatory disorder, Attention-Deficit and Disruptive Behaviour Disorder, and/or pain disorder described hereinabove.
  • a crystalline form of the compound (I) may be used for the treatment of at least one disorder selected from cognitive deficits in schizophrenia and Alzheimer's disease.
  • One embodiment of the invention relates to the prevention and/or treatment of Alzheimer's Disease, especially the use in symptomatic treatment of mild to moderate Alzheimer's Disease or in the treatment of mild to moderate dementia of Alzheimer type.
  • inventions relate to the prevention and/or treatment of disorders selected from the group consisting of attention deficit disorder (ADD), attention deficit hyperactivity disorder (ADHD) and affective disorders, wherein the affective disorders are Bipolar Disorder including acute mania, bipolar depression, bipolar maintenance, major depressive disorders (MDD) including depression, major depression, mood stabilization, schizoaffective disorders including schizophrenia, and dysthymia.
  • ADD attention deficit disorder
  • ADHD attention deficit hyperactivity disorder
  • affective disorders are Bipolar Disorder including acute mania, bipolar depression, bipolar maintenance, major depressive disorders (MDD) including depression, major depression, mood stabilization, schizoaffective disorders including schizophrenia, and dysthymia.
  • ADD attention deficit disorder
  • ADHD attention deficit hyperactivity disorder
  • affective disorders are Bipolar Disorder including acute mania, bipolar depression, bipolar maintenance, major depressive disorders (MDD) including depression, major depression, mood stabilization, schizoaffective disorders including schizophrenia, and dysthymia.
  • MDD major depressive disorders
  • Another aspect provides a method for treating at least one autoimmune disorder, psychiatric disorder, obesity disorder, eating disorder, craving disorder, neurodegenerative disorder, neuroinflammatory disorder, attention-deficit and disruptive behaviour disorder, and/or pain disorder in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating at least one disorder selected from cognitive deficits in schizophrenia, narcolepsy, excessive daytime sleepiness, obesity, attention deficit hyperactivity disorder, pain, neuropathic pain, and Alzheimer's disease in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating cognitive deficits in schizophrenia in a warm-blooded animal, comprising administering to said animal in need of such treatment a to therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating obesity in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating narcolepsy in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating excessive daytime sleepiness in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • One embodiment of the invention relates to the prevention and/or treatment of Alzheimer's Disease, especially the use in the delay of the disease progression of Alzheimer's Disease.
  • inventions relate to the prevention and/or treatment of disorders selected from the group consisting of attention deficit disorder (ADD), attention deficit hyperactivity disorder (ADHD) and affective disorders, wherein the affective disorders are Bipolar Disorder including acute mania, bipolar depression, bipolar maintenance, major depressive disorders (MDD) including depression, major depression, mood stabilization, schizoaffective disorders including schizophrenia, and dysthymia.
  • ADD attention deficit disorder
  • ADHD attention deficit hyperactivity disorder
  • affective disorders are Bipolar Disorder including acute mania, bipolar depression, bipolar maintenance, major depressive disorders (MDD) including depression, major depression, mood stabilization, schizoaffective disorders including schizophrenia, and dysthymia.
  • ADD attention deficit disorder
  • ADHD attention deficit hyperactivity disorder
  • affective disorders are Bipolar Disorder including acute mania, bipolar depression, bipolar maintenance, major depressive disorders (MDD) including depression, major depression, mood stabilization, schizoaffective disorders including schizophrenia, and dysthymia.
  • MDD major depressive disorders
  • Still another aspect provides a method for treating Alzheimer's disease in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Still yet another aspect provides a method for treating attention deficit hyperactivity disorder in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet still another aspect provides a method for treating a pain disorder in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet still another aspect provides a method for treating neuropathic pain in a warm-blooded is animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • the warm-blooded animal is a mammalian species including, but not limited to, for example, humans and domestic animals, such as, for example, dogs, cats, and horses. In one embodiment, the warm-blooded animal is a human.
  • Another aspect provides the use of a crystalline form of the compound (I) in therapy.
  • Another embodiment provides the use of a crystalline form of the compound (I) in the manufacture of a medicament for use in therapy.
  • Another aspect of the invention is wherein a compound of formula (I) as defined herein, or a pharmaceutical composition or formulation comprising a combination comprising such a compound of formula (I) is administered, concurrently, simultaneously, sequentially, separately or adjunct with another pharmaceutically active compound or compounds selected from the following:
  • the dose required for the therapeutic or preventive treatment of a particular disease will necessarily be varied depending on the host treated, the route of administration and the severity of the illness being treated.
  • the dosage form and the dose of the medicament may vary and will depend on various factors such as, for example the individual requirement of the animal treated.
  • the above other pharmaceutically active compound may be used, for example, in the amounts indicated in the Physicians' Desk Reference (PDR; e.g., 64th ed. 2010) or approved dosage ranges and/or dosage described in published references or as otherwise determined by one of ordinary skill in the art.
  • PDR Physicians' Desk Reference
  • Dosages can be readily ascertained by those skilled in the art based on this disclosure and the knowledge in the art. Thus, the skilled person can readily determine the amount of crystalline form and optional additives, vehicles, and/or carriers in compositions and to be administered in methods provided herein.
  • the specific dose level and frequency of dosage for any particular subject may vary and generally depends on a variety of factors, including, but not limited to, for example, the dissolution and/or bioavailability of the solid form(s) described herein; species, age, body weight, general health, sex, and diet of the subject; mode and time of administration; rate of excretion; drug combination; and severity of the particular condition.
  • the term “therapy” also includes “prevention” unless there are specific indications to the contrary.
  • the terms “therapeutic” and “therapeutically” should be construed accordingly.
  • disorder also includes “condition” unless there are specific indications to the contrary.
  • a first therapeutic agent which is a H3 inhibitor
  • a second therapeutic agent which is a NMDA-receptor antagonist selected from:
  • a first therapeutic agent which is a crystalline form of compound (I), 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide, such as for example Form I, and (b) a second therapeutic agent, which is memantine;
  • a first therapeutic agent which is a H3 antagonist or inverse agonist
  • a second therapeutic agent which is a acetyl choline esteras inhibitor
  • a first therapeutic agent which is a crystalline form of compound (I), 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide, such as for example Form I, and (b) a second therapeutic agent, which is a donepezil.
  • a first therapeutic agent which is a crystalline form of compound (I), 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide, such as for example Form I, and (b) a second therapeutic agent, which is a rivastigmine.
  • a first therapeutic agent which is a crystalline form of compound (I), 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide, such as for example Form I, and (b) a second therapeutic agent, which is a galantamine.
  • a first therapeutic agent which is a H3 inhibitor and (b) a second therapeutic agent, which is a voltage-gated calcium channel inhibitor selected from:
  • a first therapeutic agent which is a crystalline form of compound (I), 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide, such as for example Form I, and (b) a second therapeutic agent, which is pregabalin.
  • a first therapeutic agent which is a crystalline form of compound (I), 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide, such as for example Form I, and (b) a second therapeutic agent, which is gabapentin.
  • Such combination products employ the compound of this invention within the dosage range described herein and the other pharmaceutically active compound or compounds within approved dosage ranges and/or the dosage described in the publication reference.
  • the compound of formula (I), 4- ⁇ (1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl ⁇ -benzamide, dissolved or as a slurry of amorphous material may be crystallised in a suitable solvent, such as, for example dimethylsulfoxide (DMSO), water or mixtures thereof.
  • suitable solvents for crystallisation are lower alcohols, such as, for example, methanol, ethanol, 1-propanol, 2-propanol. 1-butanol and water or mixtures thereof.
  • Crystallization of the compound of formula (I) from an appropriate solvent system, containing at least one solvent may be achieved by attaining supersaturation in a solvent system by solvent evaporation, by temperature decrease, and/or via the addition of antisolvent (i.e. a solvent in which the compounds of the invention are poorly soluble) and/or by a chemical reaction.
  • antisolvent i.e. a solvent in which the compounds of the invention are poorly soluble
  • An example of a suitable solvent is DMSO and an example of a suitable antisolvent is water.
  • Crystallisation temperatures and times depend upon the concentration in the solution and the solvent system used.
  • Crystallisation may also be initiated and/or effected by way of standard techniques, for example with or without seeding with crystals of the appropriate crystalline compound of the invention.
  • the crystalline form of a compounds of formula (I) may be isolated using techniques, which are well known to those skilled in the art, for example decanting, filtering or centrifuging.
  • the crystalline form of a compounds of formula (I) may be dried using standard is techniques, which are well known to those skilled in the art.
  • a crystalline form of compound of formula (I) may be further purified by column chromatography on silica eluting with a suitable organic solvent or a mixture of solvents, such as for example mixtures of dichloromethane and methanol optionally containing ammonia in methanol.
  • FIG. 1 shows an X-ray powder diffractogram (XRDP) pattern for Form I of Compound (I).
  • FIG. 2 shows a differential scanning calorimetry (DSC) thermogram for Form I of Compound (I).
  • the chiral purity of the product was analyzed on a chiral column with UV-detection (250 nm) using isocratic method (mobile phase: Heptane/EtOH (80/20)+0.1% Diethylamine) on Chiralpak AD-H, 4.6 ⁇ 150 mm, giving an enantiomeric purity of >99% ee.
  • the solid product obtained in the first method was analysed by XRPD.
  • a representative XRPD pattern is shown in FIG. 1 . Selected peaks are provided in Table 1.
  • the XRPD pattern confirmed the solid material to be Crystalline Form I.
  • the solid product obtained in the second and third method, respectively, were analysed by XRPD and a representative XRPD pattern as shown in FIG. 1 and with selected peaks as are provided in Table 1 were obtained.
  • the XRPD pattern confirmed the solid material to be Crystalline Form I.
  • XRPD X-Ray Powder Diffraction
  • a thin flat sample was prepared on a flat zero background plate made of silicon using a spatula.
  • the plate was mounted in a sample holder and rotated in a horizontal position during measurement.
  • a diffraction pattern was collected between 2°2theta and 40°2theta in a continuous scan mode. Total time for the scan was approximately 10 minutes.
  • an X-ray powder diffraction pattern may be obtained which has one or more measurement errors depending on measurement conditions (such as is equipment, sample preparation or machine used).
  • intensities in an X-ray powder diffraction pattern may fluctuate depending on measurement conditions and sample preparation.
  • persons skilled in the art of X-ray powder diffraction will realize that the relative intensities of peaks may vary according to the orientation of the sample under test and on the type and setting of the instrument used.
  • the skilled person will also realize that the position of reflections can be affected by the precise height at which the sample sits in the diffractometer and the zero calibration of the diffractometer.
  • the surface planarity of the sample may also have a small effect.
  • the degree of crystallinity as determined by X-ray powder diffraction data is for example greater than about 10%, is for example greater than about 20%, is for example greater than about 30%, is for example greater than about 40%, is for example greater than about 50%, is for example greater than about 60%, such as greater than about 80%, particularly greater than about 90%, more particularly greater than about 95%.
  • the degree of crystallinity as determined by X-ray powder diffraction data is greater than about 98%, wherein the % crystallinity refers to the % by weight of the total sample mass which is crystalline.
  • a manual peak search was done preceded by an angle correction against NIST SRM 676 alumina ( ⁇ -Al 2 O 3 ) standard.
  • the measured relative intensities vs. the strongest peak are given as very strong (vs) above 50%, as strong (s) between 25 and 50%, as medium (m) between 10 and 25% and as weak (w) between 5 and 10% relative peak height.
  • DSC from 25° C. to 350° C. was performed under nitrogen in an aluminum sample cup with a perforated lid using a NETZSCH DSC 204 instrument.
  • the scan rate was 10° C. per is minute.
  • the sample size was less than 1 mg.
  • DSC onset and peak temperatures as well as energy values may vary due to, for example, the purity of the sample and sample size and due to instrumental parameters, especially the temperature scan rate.
  • the DSC data presented are not to be taken as absolute values.
  • a person skilled in the art can set up instrumental parameters for a Differential scanning calorimeter so that data comparable to the data presented here can be collected according to standard methods, for example those described in Höhne, G. W. H. et al (1996), Differential Scanning calorimetry, Springer, Berlin.
  • Hexyl lithium in hexane 21 L, 48.3 moles, 2.3 M
  • Intermediate B as a 2-MeTHF solution (33.6 moles, 30 L) was added during 20 min.
  • the scavenger was filtered off via a filter with activated charcoal or equivalent (pall-filter).
  • the vessel and the filter system were washed with 2-MeTHF (53 L).
  • a pale yellow liquid resulted.
  • NaCl (3.5 kg) in H 2 O (16.4 L) was added during 15 min. at such a rate so the inner temperature remained below 15° C.
  • the organic phase was washed with NaHSO 4 ⁇ H 2 O in H 2 O (2 ⁇ (2.87 kg+16.4 L)) and NaCl in H 2 O (3.5 kg+16.4 L).
  • 2-MeTHF and H 2 O were distilled off under vacuum until a volume 85-90% of the volume prior to distillation, approximately 8 L.
  • N-Boc-piperazine 46 g, 0.25 moles
  • EtOH 415 mL
  • Acetic acid 140 mL
  • cyclobutanone 26.5 g, 0.37 moles
  • NaBH(OAc) 3 80 g, 0.36 moles, 95% w/w was added in 20 portions is over 2 h.
  • EtOH 25 mL was used for rinsing.
  • the reaction mixture was left stirring for 2 h.
  • NAOH 296 g, 3.70 moles, 50% w/w
  • H 2 O 230 mL
  • 1 H NMR spectra were recorded in the indicated deuterated solvent at 400 MHz or 500 MHz.
  • the 400 MHz spectra were obtained using a Bruker av400 NMR spectrometer equipped with a 3 mm flow injection SEI 1 H/D- 13 C probe head with Z-gradients, using a BEST 215 liquid handler for sample injection, or using a Bruker DPX400 NMRor Bruker 500 MHz ultrashield spectrometer equipped with a 4-nucleus probehead with Z-gradients. Chemical shifts are given in ppm down- and upfield from TMS. Resonance multiplicities are denoted s, d, t, q, m and br for singlet, doublet, triplet, quartet, multiplet, and broad respectively.
  • Mass spectra were run using an automated system with electrospray (+ESI) ionization. Generally, only spectra where parent masses are observed are reported. The lowest mass major ion is reported for molecules where isotope splitting results in multiple mass spectral peaks (for example when chlorine or bromine is present).
  • LC-MS analyses were recorded on a Waters LCMS equipped with a Waters X-Terra MS, C8-column, (3.5 ⁇ m, 100 mm ⁇ 3.0 mm i.d.).
  • the mobile phase system consisted of A: 10 mM ammonium acetate in water/acetonitrile (95:5) and B: acetonitrile.
  • a linear gradient was applied running from 0% to 100% B in 4-5 minutes with a flow rate of 1.0 mL/min.
  • the mass spectrometer was equipped with an electrospray ion source (ESI) operated in a positive or negative ion mode.
  • the capillary voltage was 3 kV and the mass spectrometer was typically scanned between m/z 100-700.
  • LC-MS analyses were recorded on a Waters 2790 LCMS equipped with a Phenomenex Luna C18 (5 ⁇ m, 50 ⁇ 4.6 mm i.d.)
  • the mobile phase system consisted of A: 10 mM ammonium formate (pH 4) in water and B: acetonitrile.
  • a linear gradient was applied running from 95% to 5% B in 5 minutes with a flow rate of 2.0 mL/min.
  • the mass spectrometer was equipped with an electrospray ion source (ESI) operated in a positive or negative ion mode.
  • the capillary voltage was 3 kV and the mass spectrometer was typically scanned between m/z 100-700.
  • LC-MS analyses were recorded on a Agilent 1200 LCMS equipped with a Zorbax SB C8 (3.5 ⁇ m, 150 ⁇ 4.6 mm i.d.)
  • the mobile phase system consisted of A: 0.05% TFA in water and B: acetonitrile. A linear gradient was applied running from 10% to 90% B in 8 minutes with a flow rate of 1.0 mL/min.
  • the mass spectrometer was equipped with an electrospray ion source (ESI) operated in a positive or negative ion mode. The capillary voltage was 3 kV and the mass spectrometer was typically scanned between m/z 100-700.
  • ESI electrospray ion source
  • ACN acetonitrile; aq: aqueous; br: broad; Bu: butyl; calcd: calculated; Celite®: brand of diatomaceous earth filtering agent, registered trader of Celite Corporation; d: doublet; dd: doublet of doublet; ddd: doublet of doublet of doublet; dddd: doublet of doublet of doublet of doublet; DMF: N,N-dimethyl formamide; DMSO: dimethyl sulfoxide; dq: doublet of is quartet; DSC: differential scanning calorimetry; dt: doublet of triplet; DVS: dynamic vapour sorption; EDC: 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride; ESI: electrospray ionization; EtOAc: ethyl acetate; EtOH: ethanol; Et: ethyl; FT-IR: Fourier-transform inf
  • the H3 binding assay was used to evaluate the ability of a compound of formula (I) to inhibit [ 3 H]-N-a-methylhistamine under the conditions as described below: All binding assays were performed in a buffer consisting of 20 mmol/L Hepes, 100 mmol/L NaCl. The pH was set at 7.4 at room temperature. Liquid handling robots were used to prepare, in drug plates, 10 points dose response curves of the compound of formula (I) (3-fold serial dilutions, starting at 10 ⁇ mol/L).
  • the assay performed in 96-well plates, consisted of 100 ⁇ L, containing 20 ⁇ L of buffer alone for total binding (column #1), 20 ⁇ L of imetit (5 ⁇ ) for the non specific binding (column #2), 20 ⁇ L of the compound of formula (I) (5 ⁇ ) at varying concentrations (column #3 to #12), plus 20 ⁇ L of [3H]-N-a-Methylhistamine (25 000 dpm/well, 1.5 nmol/L) in all wells and finally 60 ⁇ L of membranes (20 ⁇ g of protein/well) mix is added to start the reaction.
  • Membranes and SPA beads (1000 ⁇ g/well) were mixed together and incubated for 30 minutes at room is temperature prior to the start of the assay. Eight wells (column #1) were used to define total binding and 1 ⁇ mol/L imetit (eight wells, column #2) define the non-specific binding. Plates were then mixed on an orbital mixer, incubated 90 minutes at room temperature, and then read on a Trilux 384TM counter.
  • [ 35 S]GTP ⁇ S binding assay was performed in a buffer consisting of 20 mmol/L Hepes, 100 mmol/L NaCl, 10 mmol/L MgCl 2 , 3 ⁇ g/mL saponine and 10 ⁇ mol/L GDP.
  • the pH was set at 7.4 at room temperature.
  • Liquid handling robots were used to prepare, in drug plates, 10 points dose response curves of the compound of formula (I) (3-fold serial dilutions, starting at 1 ⁇ mol/L for hH3 receptors and 10 ⁇ mol/L for rH3 receptors).
  • the assay performed in 96-well plates, consisted of 120 ⁇ L, containing 20 ⁇ L of R ⁇ MH (EC80, antagonist mode), 20 ⁇ L the compound of formula (I) at varying concentrations, 20 ⁇ L of the tracer [ 35 S]GTP ⁇ S (60000 dpm/well, 0.2 nmol/L) and finally 60 ⁇ L of membranes (10 ⁇ g of protein/well for hH3) mix is added to start the reaction. Membranes, SPA beads (125 ⁇ g/well) and GDP were mixed together and incubated for 30 minutes at room temperature prior to the start of the assay. Eight wells were used to define baseline and R ⁇ MH EC 80 (30 nmol/L for hH3 receptors) define the positive control. Plates were then mixed (2 minutes) on an orbital mixer, incubated 60 minutes at room temperature, and then read on a Trilux 384TM counter.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Psychiatry (AREA)
  • Epidemiology (AREA)
  • Addiction (AREA)
  • Pain & Pain Management (AREA)
  • Obesity (AREA)
  • Anesthesiology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Rheumatology (AREA)
  • Psychology (AREA)
  • Hospice & Palliative Care (AREA)
  • Child & Adolescent Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Hydrogenated Pyridines (AREA)
  • Low-Molecular Organic Synthesis Reactions Using Catalysts (AREA)
  • Epoxy Compounds (AREA)

Abstract

The present invention relates to a crystalline form of compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide,
Figure US20110201623A1-20110818-C00001
pharmaceutical formulations containing said compound and to the use of said active compound in therapy.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application No. 61/305,583, filed Feb. 18, 2010, the entirety of which is incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to crystalline forms of a compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, particularly such as Form I, pharmaceutical formulations containing said compound and to the use of said active compounds in therapy.
  • BACKGROUND OF THE INVENTION
  • The crystalline forms of 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide are useful to treat at least one histamine H3 receptor associated condition.
  • The histamine H3 receptor is of current interest in developing new medicaments. The H3 receptor is a presynaptic autoreceptor located both in the central and peripheral nervous systems, the skin, and in organs, such as, for example, the lung, the intestine, probably the spleen, and the gastrointestinal tract. Recent evidence suggests the H3 receptor has intrinsic, constitutive activity in vitro as well as in vivo (i.e., it is active in the absence of an agonist). Compounds acting as inverse agonists can inhibit this activity. The histamine H3 receptor has been shown to regulate the release of histamine and also of other neurotransmitters, such as, for example, serotonin and acetylcholine. Some histamine H3 ligands, such as, for example, a histamine H3 receptor antagonist or inverse agonist may increase the release of neurotransmitters in the brain, whereas other histamine H3 ligands, such as, for example, histamine H3 receptor agonists may inhibit the biosynthesis of histamine, as well as, inhibit the release of neurotransmitters. This suggests that histamine H3 receptor agonists, inverse agonists, and antagonists could mediate neuronal activity. As a result, efforts have been undertaken to develop new therapeutics that target the histamine H3 receptor.
  • WO2009/024823 describes the synthesis of a number of cyclopropyl amide derivatives, such as, for example, 4-((trans)-2-[(4-cyclobutylpiperazin-yl)carbonyl]-cyclopropyl}-benzamide (enantiomer 1; Example 43).
  • DETAILED DESCRIPTION OF THE INVENTION
  • One object of the present invention is to provide crystalline forms of compound (I), 4-{(1S, 2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide.
  • Another object of the present invention is to provide a crystalline form of compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, as Form I.
  • Said compound (I) having a histamine receptor antagonist or inverse agonist effect at the H3 receptor which making them suitable to be formulated into pharmaceutical is formulations.
  • Accordingly, the present invention provides a crystalline form of compound (I), 4-{(1S, 2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide
  • Figure US20110201623A1-20110818-C00002
  • One aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with at least one peak at about 18.3 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Another aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with at least two peaks at about 4.9 and about 18.3 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Yet another aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with at least three peaks at about 4.9, about 18.3 and about 20.4 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still another aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with at least four peaks at about 4.9, about 18.3, about 19.6 and about 20.4 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • A further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with peaks at selected °2Theta-values described above and with additional peaks at about 16.4 and about 16.6, °2Theta when measured using radiation with a wavelength of about 1.54 angstroms.
  • Yet a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with peaks at selected °2Theta-values described above and with an additional peak at about 5.3 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with peaks at °2Theta-values selected from about 4.9, about 16.4, about 16.6, about 18.3, about 19.6 and about 20.4, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with peaks at °2Theta-values selected from about 4.9, about 5.3, about 16.4, about 16.6, about 18.3, about 19.6 and about 20.4, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with peaks at °2Theta-values selected from about 4.9, about 12.6, about 16.4, about 16.6, about 18.3, about 19.6, about 20.4, and about 23.2, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still a further aspect of the invention relates to a crystalline form of compound (I), characterized in that said form has an XRDP pattern (Cu Kα) with peaks at °2Theta-values selected from about 4.9, about 5.3, about 9.0, about 12.6, about 16.4, about 16.6, about 18.3, about 19.6, about 20.4, and about 23.2, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Still a further aspect of the invention relates to a crystalline form of compound (I), is characterized in that said form has an XRDP pattern (Cu Kα) with peaks at from about 4.9, about 5.3, about 9.0, about 12.6, about 16.4, about 16.6, about 18.3, about 19.6, about 20.4, about 21.2, about 23.2 and about 24.6 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
  • Yet still a further aspect of the invention relates to relates to a crystalline form of compound (I), characterized by the X-ray powder diffraction pattern essentially as shown in FIG. 1.
  • Another embodiment relates to a crystalline form of compound (I) that has a DSC thermogram essentially as depicted in FIG. 2.
  • In another embodiment, a crystalline form of compound (I) has a DSC thermogram comprising an endothermic event with an onset temperature of about 225° C.
  • In still another embodiment, a crystalline form of compound (I) has a DSC thermogram comprising an endothermic event with a peak temperature of about 235° C.
  • It is well known that the DSC onset and peak temperatures as well as energy values may vary due to, for example, the purity of the sample and sample size and due to instrumental parameters, especially the temperature scan rate. Hence the DSC data presented are not to be taken as absolute values. A person skilled in the art can set up instrumental parameters for a Differential scanning calorimeter so that data comparable to the data presented here can be collected according to standard methods, for example those described in Höhne, G. W. H. et al (1996), Differential Scanning calorimetry, Springer, Berlin.
  • The crystalline forms of compound (I) of the present invention may also exist as solvates, including hydrates.
  • The present invention also relates to the use of a crystalline form of compound (I), as hereinbefore defined.
  • A crystalline form of compound (I), as hereinbefore defined has one or more advantageous properties. For example, in some embodiments, a crystalline form of compound (I) shows advantageous properties, such as, for example, a high melting point, a substantial lack of solvent (e.g., water) content, little or no weight loss on heating, and/or low hygroscopicity. In certain embodiments, such properties advantageously facilitate the manufacture, storage, formulation, and/or delivery of compound (I).
  • A crystalline form of compound (I), as described herein, for example Form I of compound (I) provide advantageous properties with regard to stability.
  • A substance can be expected to be more stable chemically in a crystalline state in comparison with the same substance in an amorphous state, as described in Haleblian and McCrone J. Pharm. Sci 1969, 58, pages 911-929, especially page 913. This observation is common for small molecules (i.e. non-proteins) but not always true for macromolecules like proteins, as described in Pikal and Rigsbee, Pharm. Res. 1997, 14, pages 1379-1387, especially page 1379. A crystalline state is thus beneficial for small molecules such as compound (I).
  • X-rays will be scattered by electrons in atoms in a substance. Crystalline material will diffract X-rays giving peaks in directions of constructive interference. The directions are determined by the crystal structure, including the size and shape of the unit cell. All diffraction peak °2Theta values disclosed and/or claimed herein refer to Cu Kα-radiation. An amorphous (non-crystalline) material will not give such diffraction peaks. See e.g. Klug, H. P. & Alexander, L. E., X-Ray Diffraction Procedures For Polycrystalline and Amorphous Materials, 1974, John Wiley & Sons.
  • The ability for a compound to lump together or cake without control will increase if the compound is heated to near its melting temperature. Lumps and cakes will have different flow and dissolution properties as compared with a powder. Mechanical treatment of a powder, such as during particle size reduction, will bring energy into the material and thus give a possibility to raise the temperature. Storage of a compound as well as transport of a compound can unintentionally also lead to an increased temperature. Melting is an endothermic event. Endothermic events can be measured by, e.g. differential scanning calorimetry (DSC).
  • It is thus beneficial for a compound of formula (I) or a pharmaceutically acceptable salt thereof salt thereof to have such endothermic events at a temperature higher than the highest temperature expected during normal use to prevent said compounds from forming an undesired lump or cake.
  • Pharmaceutical Formulations
  • According to one aspect of the present invention there is provided a pharmaceutical formulation comprising a crystalline form of the compound (I), such as Form I, for use in the prevention and/or treatment of conditions associated with the H3 receptor.
  • The formulation used in accordance with the present invention may be in a form suitable for oral administration, for example such as a tablet, pill, powder, granule or capsule, for parenteral injection (including intravenous, subcutaneous, intramuscular, intravascular or infusion), for topical administration for example such as an ointment, patch or cream, for rectal administration for example such as a suppository and for other non-parenteral administration.
  • Suitable daily doses a crystalline form of the compound (I), such as Form I, in the treatment of a mammal, including human, are approximately 0.01 to 250 mg/kg bodyweight at per oral administration and about 0.001 to 250 mg/kg bodyweight at parenteral administration. The typical daily dose of the active ingredients varies within a wide range and will depend on various factors such as the relevant indication, the route of administration, the age, weight and sex of the patient and may be determined by a physician.
  • A crystalline form of compound (I), such as for example Form I, may be used on its own but will usually be administered in the form of a pharmaceutical formulation in which the active ingredient is in association with pharmaceutically acceptable diluents, excipients and/or inert carrier known to a person skilled in the art. Dependent on the mode of administration, the pharmaceutical formulation may comprise from 0.05 to 99% w (percent by weight), for example from 0.10 to 50% w, of active ingredient, all percentages by weight being based on total composition.
  • The invention further provides a process for the preparation of a pharmaceutical formulation of the invention which comprises mixing of a crystalline form of the compound (I), such as form I, as hereinbefore defined, with pharmaceutically acceptable diluents, excipients and/or inert carriers.
  • Medical Uses
  • In one embodiment, at least one crystalline form of the compound (I) described herein may be administered to a mammal, including human, to be used to modulate at least one histamine H3 receptor. The terms “modulate”, “modulates”, “modulating”, or “modulation”, as used herein, refer to, for example, the activation (e.g., agonist activity) or inhibition (e.g., antagonist and inverse agonist activity) of at least one histamine H3 receptor. In one embodiment, at least one crystalline form of the compound (I) described herein may be administered to a mammal, including human, to be used as an inverse agonist of at least one histamine H3 receptor. In another embodiment, at least one crystalline form of the compound (I) described herein may be administered to a mammal, including human, to be used as an antagonist of at least one histamine H3 receptor. In another embodiment, at least one crystalline form of the compound (I) described herein may be used as an antagonist of at least one histamine H3 receptor. In yet another embodiment, at least one crystalline form of the compound (I) described herein may be used an antagonist of at least one histamine H3 receptor.
  • At least one crystalline form of the compound (I) described herein may be administered to a mammal, including human, to be used to treat one or more of a wide range of conditions or disorders in which modulating the histamine H3 receptor is beneficial. At least one crystalline form of the compound (I) described herein may administered to a mammal, including human, to be, for example, be useful to treat at least one disease of the central nervous system, the peripheral nervous system, the cardiovascular system, the pulmonary system, the gastrointestinal system, or the endocrinological system.
  • Another embodiment provides a method for treating a disorder in which modulating the function of at least one histamine H3 receptor is beneficial comprising administering to a warm-blooded animal in need of such treatment a therapeutically effective amount of crystalline form of the compound (I).
  • One embodiment relates to the use of the crystalline form of a compound (I) in the manufacture of a medicament for the treatment of at least one disorder selected from schizophrenia, narcolepsy, excessive daytime sleepiness, obesity, attention deficit hyperactivity disorder, pain, neuropathic pain, Alzheimer's disease, cognition deficiency, and cognition deficiency associated with schizophrenia.
  • A further embodiment relates to a method for the therapy of at least one disorder selected from schizophrenia, narcolepsy, excessive daytime sleepiness, obesity, attention deficit hyperactivity disorder, pain, neuropathic pain, Alzheimer's disease, cognition deficiency, and cognition deficiency associated with schizophrenia, in a warm-blooded animal in need of such therapy, wherein the method comprises administering to the animal a therapeutically effective amount of a crystalline form of the compound (I).
  • A crystalline form of the compound (I) may be useful to treat at least one autoimmune disorder. Exemplary autoimmune disorders include, but are not limited to, for example, arthritis, skin grafts, organ transplants and similar surgical needs, collagen diseases, various allergies, tumors and viruses.
  • A crystalline form of the compound (I) may be useful to treat at least one psychiatric disorder.
  • Exemplary psychiatric disorders include, but are not limited to, for example, Psychotic Disorder(s) and Schizophrenia Disorder(s), such as, for example, Schizoaffective Disorder(s), Delusional Disorder(s), Brief Psychotic Disorder(s), Shared Psychotic Disorder(s), and Psychotic Disorder(s) Due to a General Medical Condition; Dementia and other Cognitive Disorder(s); Anxiety Disorder(s), such as, for example, Panic Disorder(s) Without Agoraphobia, Panic Disorder(s) With Agoraphobia, Agoraphobia Without History of Panic Disorder(s), Specific Phobia, Social Phobia, Obsessive-Compulsive Disorder(s), Stress related Disorder(s), Posttraumatic Stress Disorder(s), Acute Stress Disorder(s), Generalized Anxiety Disorder(s) and Generalized Anxiety Disorder(s) Due to a General Medical Condition; Mood Disorder(s), such as, for example, a) Depressive Disorder(s) (including but not limited to, for example, Major Depressive Disorder(s) including depression, major depression, mood stabilization and/or apathy, and Dysthymic Disorder(s)), b) Bipolar Depression and/or Bipolar mania, such as, for example, Bipolar I (which includes, but is not limited to those with manic, depressive or mixed episodes), Bipolar II, and Bipolar Maintenance, c) Cyclothymiac's Disorder(s), and d) Mood Disorder(s) Due to a General Medical Condition; Sleep Disorder(s), such as, for example, excessive daytime sleepiness, narcolepsy, hypersomina, and sleep apnea; Disorder(s) Usually First Diagnosed in Infancy, Childhood, or Adolescence including, but not limited to, for example, Mental Retardation, Down's Syndrome, Learning Disorder(s), Motor Skills Disorder(s), Communication Disorders(s), Pervasive Developmental Disorder(s), is Attention-Deficit and Disruptive Behavior Disorder(s), Feeding and Eating Disorder(s) of Infancy or Early Childhood, Tic Disorder(s), and Elimination Disorder(s); Substance-Related Disorder(s) including, but not limited to, for example, Substance Dependence, Substance Abuse, Substance Intoxication, Substance Withdrawal, Alcohol-Related Disorder(s), Amphetamines (or Amphetamine-Like)-Related Disorder(s), Caffeine-Related Disorder(s), Cannabis-Related Disorder(s), Cocaine-Related Disorder(s), Hallucinogen-Related Disorder(s), Inhalant-Related Disorder(s), Nicotine-Related Disorder(s), Opiod-Related Disorder(s), Phencyclidine (or Phencyclidine-Like)-Related Disorder(s), and Sedative-, Hypnotic- or Anxiolytic-Related Disorder(s); Attention-Deficit and Disruptive Behavior Disorder(s); Eating Disorder(s), such as, for example, obesity; Personality Disorder(s) including, but not limited to, for example, Obsessive-Compulsive Personality Disorder(s); Impulse-Control Disorder(s); Tic Disorders including, but not limited to, for example Tourette's Disorder, Chronical Tics Syndrome, Chronic motor or vocal tic disorder; and Transient Tic Disorder. At least one of the above psychiatric disorders is defined, for example, in the American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision, Washington, D.C., American Psychiatric Association, 2000.
  • A crystalline form of the compound (I) may be useful: i) to treat obesity or being overweight (e.g., promotion of weight loss and maintenance of weight loss), eating disorders (e.g., binge eating, anorexia, bulimia and compulsive), and/or cravings (for drugs, tobacco, alcohol, any appetizing macronutrients or non-essential food items); ii) to prevent weight gain (e.g., medication-induced or subsequent to cessation of smoking); and/or iii) to modulate appetite and/or satiety. At least one solid form described herein may be suitable for treating obesity by reducing appetite and body weight and/or maintaining weight reduction and preventing rebound. At least one solid form described herein may be used to prevent or reverse medication-induced weight gain, e.g., weight gain caused by antipsychotic (neuroleptic) treatment(s); and/or weight gain associated with smoking cessation.
  • A crystalline form of the compound (I) may be useful to treat at least one Neurodegenerative Disorder.
  • Exemplary Neurodegenerative Disorders include, but are not limited to, for example, conditions associated with cognitive disorder(s) or indications with deficit(s) in cognition is such as: dementia; incl. pre-senile dementia (early onset Alzheimer's Disease); senile dementia (dementia of the Alzheimer's type); Alzheimer's Disease (AD); Familial Alzheimer's disease; Early Alzheimer's disease; mild to moderate dementia of the Alzheimer's type; delay of disease progression of Alzheimer's Disease; neurodegeneration associated with Alzheimer's disease, Mild Cognitive Impairment (MCI); Amnestic Mild Cognitive Impairment (aMCI); Age-associated Memory Impairment (AAMI); Lewy body dementia; vascular dementia (VD); HIV-dementia; AIDS dementia complex; AIDS—Neurological Complications; Frontotemporal dementia (FTD); Frontotemporal dementia Parkinson's Type (FTDP); dementia pugilistica; dementia due to infectious agents or metabolic disturbances; dementia of degenerative origin; dementia-Multi-Infarct; memory loss; cognition in Parkinson's Disease; cognition in multiple sclerosis; cognition deficits associated with chemotherapy; Cognitive Deficit in Schizophrenia (CDS); Schizoaffective disorders including schizophrenia; Age-Related Cognitive Decline (ARCD); Cognitive Impairment No Dementia (CIND); Cognitive Deficit arising from stroke or brain ischemia; Congenital and/or development disorders; progressive supranuclear palsy (PSP); amyotrophic lateral sclerosis (ALS); corticobasal degeneration (CBD); traumatic brain injury (TBI); postencephelatic parkinsonism; Pick's Disease; Niemann-Pick's Disease; Down's syndrome; Huntington's Disease; Creurtfeld-Jacob's disease; prion diseases; multiple sclerosis (MS); motor neuron diseases (MND); Parkinson's Disease (PD); β-amyloid angiopathy; cerebral amyloid angiopathy; Trinucleotide Repeat Disorders; Spinal Muscular Atrophy; Ataxia; Friedreich's Ataxia; Ataxias and Cerebellar or Spinocerebellar Degerneration; Neuromyelitis Optica; Multiple System Atrophy; Transmissible Spongiform Encephalopathies; Attention Deficit Disorder (ADD); Attention Deficit Hyperactivity Disorder (ADHD); Bipolar Disorder (BD) including acute mania, bipolar depression, bipolar maintenance; Major Depressive Disorders (MDD) including depression, major depression, mood disorder (stabilization), dysthymia and apathy; Guillain-Barré Syndrome (GBS); and Chronic Inflammatory Demyelinating Polyneuropathy (CIDP).
  • A crystalline form of the compound (I) may be useful to treat at least one Neuroinflammatory Disorder including, but not limited to, for example, Multiple Sclerosis (MS), which includes, but is not limited to, for example, Relapse Remitting Multiple Sclerosis (RRMS), Secondary Progressive Multiple Sclerosis (SPMS), and Primary Progressive Multiple Sclerosis (PPMS); Parkinson's disease; Multiple System Atrophy is (MSA); Corticobasal Degeneration; Progressive Supranuclear Paresis; Guillain-Barré Syndrome (GBS); and chronic inflammatory demyelinating polyneuropathy (CIDP).
  • A crystalline form of the compound (I) may be useful to treat at least one Attention-Deficit and Disruptive Behavior Disorder.
  • Exemplary Attention-Deficit and Disruptive Behavior Disorders include, but are not limited to, for example, attention deficit disorder (ADD), attention deficit hyperactivity disorder (ADHD), and affective disorders.
  • A crystalline form of the compound (I) may be useful to treat pain, including acute or chronic pain disorders including but not limited to, for example, Widespread pain, Localized pain, Nociceptive pain, Inflammatory pain, Central pain, Central and peripheral neuropathic pain, Diabetic neuropathic pain, Central and peripheral neurogenic pain, Central and peripheral neuralgia, Low back pain, Postoperative pain, Visceral pain, and Pelvic pain; Allodynia; Anesthesia dolorosa; Causalgia; Dysesthesia; Fibromyalgia; Hyperalgesia; Hyperesthesia; Hyperpathia; Ischemic pain; Sciatic pain; Burn-induced pain; Pain associated with cystitis including, but not limited to, interstitial cystitis; Pain associated with multiple sclerosis; Pain associated with arthritis; Pain associated with osteoarthritis; Pain associated with rheumatoid arthritis; Pain associated with pancreatitis; Pain associated with psoriasis; Pain associated with fibromyalgia; Pain associated with IBS; Pain associated with cancer; and Restless Legs Syndrome.
  • A crystalline form of the compound (I) may be useful to treat at least one of the following disorders Autism, Dyslexia, Jetlag, Hyperkinesias, Dystonias, Rage outbursts, Muscular Dystrophy, Neurofibromatosis, Spinal Cord Injury, Cerebral Palsy, Neurological Sequelae of Lupus and Post-Polio Syndrome.
  • A crystalline form of the compound (I) may be used for the manufacture of a medicament for the treatment of at least one autoimmune disorder, psychiatric disorder, obesity disorder, eating disorder, craving disorder, neurodegenerative disorder, neuroinflammatory disorder, Attention-Deficit and Disruptive Behaviour Disorder, and/or pain disorder described hereinabove.
  • A crystalline form of the compound (I) may be used for the treatment of at least one disorder selected from cognitive deficits in schizophrenia and Alzheimer's disease. One embodiment of the invention relates to the prevention and/or treatment of Alzheimer's Disease, especially the use in symptomatic treatment of mild to moderate Alzheimer's Disease or in the treatment of mild to moderate dementia of Alzheimer type.
  • Other embodiments of the invention relate to the prevention and/or treatment of disorders selected from the group consisting of attention deficit disorder (ADD), attention deficit hyperactivity disorder (ADHD) and affective disorders, wherein the affective disorders are Bipolar Disorder including acute mania, bipolar depression, bipolar maintenance, major depressive disorders (MDD) including depression, major depression, mood stabilization, schizoaffective disorders including schizophrenia, and dysthymia.
  • Another aspect provides a method for treating at least one autoimmune disorder, psychiatric disorder, obesity disorder, eating disorder, craving disorder, neurodegenerative disorder, neuroinflammatory disorder, attention-deficit and disruptive behaviour disorder, and/or pain disorder in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating at least one disorder selected from cognitive deficits in schizophrenia, narcolepsy, excessive daytime sleepiness, obesity, attention deficit hyperactivity disorder, pain, neuropathic pain, and Alzheimer's disease in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating cognitive deficits in schizophrenia in a warm-blooded animal, comprising administering to said animal in need of such treatment a to therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating obesity in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating narcolepsy in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet another aspect provides a method for treating excessive daytime sleepiness in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • One embodiment of the invention relates to the prevention and/or treatment of Alzheimer's Disease, especially the use in the delay of the disease progression of Alzheimer's Disease.
  • Other embodiments of the invention relate to the prevention and/or treatment of disorders selected from the group consisting of attention deficit disorder (ADD), attention deficit hyperactivity disorder (ADHD) and affective disorders, wherein the affective disorders are Bipolar Disorder including acute mania, bipolar depression, bipolar maintenance, major depressive disorders (MDD) including depression, major depression, mood stabilization, schizoaffective disorders including schizophrenia, and dysthymia.
  • Still another aspect provides a method for treating Alzheimer's disease in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Still yet another aspect provides a method for treating attention deficit hyperactivity disorder in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet still another aspect provides a method for treating a pain disorder in a warm-blooded animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • Yet still another aspect provides a method for treating neuropathic pain in a warm-blooded is animal, comprising administering to said animal in need of such treatment a therapeutically effective amount of a crystalline form of the compound (I).
  • In one embodiment, the warm-blooded animal is a mammalian species including, but not limited to, for example, humans and domestic animals, such as, for example, dogs, cats, and horses. In one embodiment, the warm-blooded animal is a human.
  • Another aspect provides the use of a crystalline form of the compound (I) in therapy.
  • Another embodiment provides the use of a crystalline form of the compound (I) in the manufacture of a medicament for use in therapy.
  • Another aspect of the invention is wherein a compound of formula (I) as defined herein, or a pharmaceutical composition or formulation comprising a combination comprising such a compound of formula (I) is administered, concurrently, simultaneously, sequentially, separately or adjunct with another pharmaceutically active compound or compounds selected from the following:
      • (i) antidepressants including for example agomelatine, amitriptyline, amoxapine, bupropion, citalopram, clomipramine, desipramine, doxepin duloxetine, elzasonan, escitalopram, fluvoxamine, fluoxetine, gepirone, imipramine, ipsapirone, maprotiline, nortriptyline, nefazodone, paroxetine, phenelzine, protriptyline, ramelteon, reboxetine, robalzotan, sertraline, sibutramine, thionisoxetine, tranylcypromaine, trazodone, trimipramine, venlafaxine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (ii) atypical antipsychotics including for example quetiapine and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (iii) antipsychotics including for example amisulpride, aripiprazole, asenapine, benzisoxidil, bifeprunox, carbamazepine, clozapine, chlorpromazine, debenzapine, divalproex, duloxetine, eszopiclone, haloperidol, iloperidone, lamotrigine, loxapine, mesoridazine, olanzapine, paliperidone, perlapine, perphenazine, phenothiazine, phenylbutylpiperidine, pimozide, prochlorperazine, risperidone, sertindole, sulpiride, suproclone, suriclone, thioridazine, trifluoperazine, trimetozine, valproate, valproic acid, zopiclone, zotepine, ziprasidone and equivalents and pharmaceutically active isomer(s) and is metabolite(s) thereof;
      • (iv) anxiolytics including for example alnespirone, azapirones, benzodiazepines, barbiturates such as adinazolam, alprazolam, balezepam, bentazepam, bromazepam, brotizolam, buspirone, clonazepam, clorazepate, chlordiazepoxide, cyprazepam, diazepam, diphenhydramine, estazolam, fenobam, flunitrazepam, flurazepam, fosazepam, lorazepam, lormetazepam, meprobamate, midazolam, nitrazepam, oxazepam, prazepam, quazepam, reclazepam, tracazolate, trepipam, temazepam, triazolam, uldazepam, zolazepam and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (v) anticonvulsants including for example carbamazepine, clonazepam, ethosuximide, felbamate, fosphenyloin, gabapentin, lacosamide, lamotrogine, levetiracetam, oxcarbazepine, phenobarbital, phenyloin, pregabaline, rufinamide, topiramate, valproate, vigabatrine, zonisamide, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (vi) Alzheimer's therapies including for example donepezil, rivastigmine, galantamine, memantine, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (vii) Parkinson's therapies including for example levodopa, dopamine agonists such as apomorphine, bromocriptine, cabergoline, pramipexol, ropinirole, and rotigotine, MAO-B inhibitors such as selegeline and rasagiline, and other dopaminergics such as tolcapone and entacapone, A-2 inhibitors, dopamine reuptake inhibitors, NMDA antagonists, Nicotine agonists, and inhibitors of neuronal nitric oxide synthase and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (viii) migraine therapies including for example almotriptan, amantadine, bromocriptine, butalbital, cabergoline, dichloralphenazone, dihydroergotamine, eletriptan, frovatriptan, lisuride, naratriptan, pergolide, pizotiphen, pramipexole, rizatriptan, ropinirole, sumatriptan, zolmitriptan, zomitriptan, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (ix) stroke therapies including for example thrombolytic therapy with eg activase and desmoteplase, abciximab, citicoline, clopidogrel, eptifibatide, minocycline, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (x) urinary incontinence therapies including for example darafenacin, falvoxate, oxybutynin, propiverine, robalzotan, solifenacin, tolterodine and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (xi) neuropathic pain therapies including lidocain, capsaicin, and anticonvulsants is such as gabapentin, pregabalin, and antidepressants such as duloxetine, venlafaxine, amitriptyline, klomipramine, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (xii) nociceptive pain therapies including paracetamol, NSAIDS and coxibs, such as celecoxib, etoricoxib, lumiracoxib, valdecoxib, parecoxib, diclofenac, loxoprofen, naproxen, ketoprofen, ibuprofen, nabumeton, meloxicam, piroxicam and opioids such as morphine, oxycodone, buprenorfin, tramadol and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (xiii) insomnia therapies including for example agomelatine, allobarbital, alonimid, amobarbital, benzoctamine, butabarbital, capuride, chloral, cloperidone, clorethate, dexclamol, ethchlorvynol, etomidate, glutethimide, halazepam, hydroxyzine, mecloqualone, melatonin, mephobarbital, methaqualone, midaflur, nisobamate, pentobarbital, phenobarbital, propofol, ramelteon, roletamide, triclofos, secobarbital, zaleplon, zolpidem and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (xiv) mood stabilizers including for example carbamazepine, divalproex, gabapentin, lamotrigine, lithium, olanzapine, quetiapine, valproate, valproic acid, verapamil, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof;
      • (xv) obesity therapies, such as, for example, anti-obesity drugs that affect energy expenditure, glycolysis, gluconeogenesis, glucogenolysis, lipolysis, lipogenesis, fat absorption, fat storage, fat excretion, hunger and/or satiety and/or craving mechanisms, appetite/motivation, food intake, and G-I motility; very low calorie diets (VLCD); and low-calorie diets (LCD);
      • (xvi) therapeutic agents useful in treating obesity associated disorders, such as, for example, biguanide drugs, insulin (synthetic insulin analogues) and oral antihyperglycemics (these are divided into prandial glucose regulators and alpha-glucosidase inhibitors), PPAR modulating agents, such as, for example, PPAR alpha and/or gamma agonists; sulfonylureas; cholesterol-lowering agents, such as, for example, inhibitors of HMG-CoA reductase (3-hydroxy-3-methylglutaryl coenzyme A reductase); an inhibitor of the ileal bile acid transport system (IBAT inhibitor); a bile acid binding resin; bile acid sequestering agent, such as, for example, colestipol, cholestyramine, or cholestagel; a CETP (cholesterol ester transfer protein) inhibitor; a cholesterol absorption is antagonist; a MTP (microsomal transfer protein) inhibitor; a nicotinic acid derivative, including slow release and combination products; a phytosterol compound; probucol; an anti-coagulant; an omega-3 fatty acid; an anti-obesity therapy, such as, for example, sibutramine, phentermine, orlistat, bupropion, ephedrine, and thyroxine; an antihypertensive, such as, for example, an angiotensin converting enzyme (ACE) inhibitor, an angiotensin II receptor antagonist, an adrenergic blocker, an alpha adrenergic blocker, a beta adrenergic blocker, a mixed alpha/beta adrenergic blocker, an adrenergic stimulant, calcium channel blocker, an AT-1 blocker, a saluretic, a diuretic, and a vasodilator; a melanin concentrating hormone (MCH) modulator; an NPY receptor modulator; an orexin receptor modulator; a phosphoinositide-dependent protein kinase (PDK) modulator; modulators of nuclear receptors, such as, for example, LXR, FXR, RXR, GR, ERRα, β, PPARα, β, γ and RORalpha; a monoamine transmission-modulating agent, such as, for example, a selective serotonin reuptake inhibitor (SSRI), a noradrenaline reuptake inhibitor (NARI), a noradrenaline-serotonin reuptake inhibitor (SNRI), a monoamine oxidase inhibitor (MAOI), a tricyclic antidepressive agent (TCA), a noradrenergic and specific serotonergic antidepressant (NaS SA); a serotonin receptor modulator; a leptin/leptin receptor modulator; a ghrelin/ghrelin receptor modulator; a DPP-IV inhibitor; and equivalents and pharmaceutically active isomer(s), metabolite(s), and pharamaceutically acceptable salts, solvates, and prodrugs thereof;
      • (xvii) agents for treating ADHD, such as, for example, amphetamine, methamphetamine, dextroamphetamine, atomoxetine, methylphenidate, dexmethylphenidate, modafinil, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof; and
      • (xviii) agents used to treat substance abuse disorders, dependence, and withdrawal, such as, for example, nicotine replacement therapies (i.e., gum, patches, and nasal spray); nicotinergic receptor agonists, partial agonists, and antagonists, (e.g., varenicline); acomprosate, bupropion, clonidine, disulfuram, methadone, naloxone, naltrexone, and equivalents and pharmaceutically active isomer(s) and metabolite(s) thereof.
  • The dose required for the therapeutic or preventive treatment of a particular disease will necessarily be varied depending on the host treated, the route of administration and the severity of the illness being treated.
  • For veterinary use the amounts of different components, the dosage form and the dose of the medicament may vary and will depend on various factors such as, for example the individual requirement of the animal treated.
  • When employed in combination with at least one solid form described herein, the above other pharmaceutically active compound may be used, for example, in the amounts indicated in the Physicians' Desk Reference (PDR; e.g., 64th ed. 2010) or approved dosage ranges and/or dosage described in published references or as otherwise determined by one of ordinary skill in the art.
  • Dosages can be readily ascertained by those skilled in the art based on this disclosure and the knowledge in the art. Thus, the skilled person can readily determine the amount of crystalline form and optional additives, vehicles, and/or carriers in compositions and to be administered in methods provided herein. The specific dose level and frequency of dosage for any particular subject, however, may vary and generally depends on a variety of factors, including, but not limited to, for example, the dissolution and/or bioavailability of the solid form(s) described herein; species, age, body weight, general health, sex, and diet of the subject; mode and time of administration; rate of excretion; drug combination; and severity of the particular condition.
  • In the context of the present specification, the term “therapy” also includes “prevention” unless there are specific indications to the contrary. The terms “therapeutic” and “therapeutically” should be construed accordingly.
  • In the context of the present specification, the term “disorder” also includes “condition” unless there are specific indications to the contrary.
  • In one embodiment of the invention the combination comprises the group of compounds
  • (a) and (b) as defined below:
  • (a) a first therapeutic agent, which is a H3 inhibitor and (b) a second therapeutic agent, which is a NMDA-receptor antagonist selected from:
  • (a) a first therapeutic agent, which is a crystalline form of compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, such as for example Form I, and (b) a second therapeutic agent, which is memantine;
  • (a) a first therapeutic agent, which is a H3 antagonist or inverse agonist and (b) a second therapeutic agent, which is a acetyl choline esteras inhibitor.
  • (a) a first therapeutic agent, which is a crystalline form of compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, such as for example Form I, and (b) a second therapeutic agent, which is a donepezil.
  • (a) a first therapeutic agent, which is a crystalline form of compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, such as for example Form I, and (b) a second therapeutic agent, which is a rivastigmine.
  • (a) a first therapeutic agent, which is a crystalline form of compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, such as for example Form I, and (b) a second therapeutic agent, which is a galantamine.
  • (a) a first therapeutic agent, which is a H3 inhibitor and (b) a second therapeutic agent, which is a voltage-gated calcium channel inhibitor selected from:
  • (a) a first therapeutic agent, which is a crystalline form of compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, such as for example Form I, and (b) a second therapeutic agent, which is pregabalin.
  • (a) a first therapeutic agent, which is a crystalline form of compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, such as for example Form I, and (b) a second therapeutic agent, which is gabapentin.
  • Such combination products employ the compound of this invention within the dosage range described herein and the other pharmaceutically active compound or compounds within approved dosage ranges and/or the dosage described in the publication reference.
  • Methods of Preparation
  • A process for the preparation of a crystalline form of a compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, Form I, are described in comprising the following steps:
      • a) dissolving (1s, 25)-2-(4-Carbamoyl-phenyl)-cyclopropanecarboxylic acid and cyclobutylpiperazine or a suitable salt thereof (for example the dihydrochloride) in is a suitable solvent such as DMSO in the presence of a base such as N-methylmorpholine;
      • b) adding an activating agent such as a mixture of 1-hydroxybenzotriazole/N-methylmorpholine and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride in a suitable solvent such as DMSO; followed by
      • c) heating the solution to 60° C. and adjusting pH to about 8 with a base such as an trialkylamine for example triethylamine; followed by
      • d) cooling to 20° C., adding water and let to stirring for 16 hr.; followed by
      • e) filtering off the product; followed by
      • f) slurry washing with cold water; followed by
      • g) drying the obtained solid under vacuum at 40° C. to obtain a crystalline Form I of compound (I).
  • Alternatively, the compound of formula (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, dissolved or as a slurry of amorphous material, may be crystallised in a suitable solvent, such as, for example dimethylsulfoxide (DMSO), water or mixtures thereof. Other suitable solvents for crystallisation are lower alcohols, such as, for example, methanol, ethanol, 1-propanol, 2-propanol. 1-butanol and water or mixtures thereof.
  • Crystallization of the compound of formula (I) from an appropriate solvent system, containing at least one solvent, may be achieved by attaining supersaturation in a solvent system by solvent evaporation, by temperature decrease, and/or via the addition of antisolvent (i.e. a solvent in which the compounds of the invention are poorly soluble) and/or by a chemical reaction. An example of a suitable solvent is DMSO and an example of a suitable antisolvent is water.
  • Crystallisation temperatures and times depend upon the concentration in the solution and the solvent system used.
  • Crystallisation may also be initiated and/or effected by way of standard techniques, for example with or without seeding with crystals of the appropriate crystalline compound of the invention.
  • The crystalline form of a compounds of formula (I) may be isolated using techniques, which are well known to those skilled in the art, for example decanting, filtering or centrifuging.
  • The crystalline form of a compounds of formula (I) may be dried using standard is techniques, which are well known to those skilled in the art.
  • Alternatively, a crystalline form of compound of formula (I) may be further purified by column chromatography on silica eluting with a suitable organic solvent or a mixture of solvents, such as for example mixtures of dichloromethane and methanol optionally containing ammonia in methanol.
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 shows an X-ray powder diffractogram (XRDP) pattern for Form I of Compound (I).
  • FIG. 2 shows a differential scanning calorimetry (DSC) thermogram for Form I of Compound (I).
  • WORKING EXAMPLES
  • The invention is further defined in the following Examples. It should be understood that the Examples are given by way of illustration only. From the above discussion and the Examples, one skilled in the art can ascertain the essential characteristics of the invention, and without departing from the spirit and scope thereof, can make various changes and modifications to adapt the invention to various uses and conditions. As a result, the invention is not limited by the illustrative examples set forth herein below, but rather defined by the claims appended hereto.
  • All temperatures are in degrees Celsius (° C.) and are uncorrected. Unless otherwise noted, commercial reagents used in preparing the example compounds were used as received without additional purification.
  • Unless otherwise noted, the solvents used in preparing the example compounds were commercial anhydrous grades and were used without further drying or purification. Compounds used as starting materials in the Example and Methods are commercially available otherwise a process for preparing them are described in the Intermediates A-F herein.
  • Example 1 4-{(1S,2S)-2-[(4-Cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide, Form I
  • Figure US20110201623A1-20110818-C00003
  • First Method
  • Intermediate E (5.52 g, 26.7 mmoles, 99.1% w/w) and Intermediate F (6.07 g, 28.0 mmoles, 98.40% w/w) were mixed in DMSO (82 mL) at tjacket=22° C. N-Methylmorpholine (2.94 mL, 27.2 mmoles) was added over 5 min. The charging vessel was rinsed with DMSO (2.8 mL). HOBt/NMM solution (1.80 g, 2.66 mmoles, 20% w/w) was added in one portion. The charging vessel was rinsed with DMSO (2.8 mL). EDCI×HCl (7.16 g, 38.0 mmoles) was added over 10 min. at tjacket=22° C. The reaction was complete after 2 h. The reaction solution was then heated to 60° C. and pH adjusted with TEA (5.18 g g, 51.2 mmol) to pH-8. The solid mixture was cooled to 20° C. after which H2O (69.8 mL) was added and left to stir for 16 h. The product was filtered off, and slurry washed with cold H2O (2×33 mL). Drying under vacuum at 40° C. gave 7.53 g (22.8 mmoles, 99.0% w/w), 85% yield. 1H-NMR (DMSO-d6): δ 7.91 (br s, 1H), 7.78 (d, J=8.0 Hz, 2H), 7.29 (br s, 1H), 7.24 (d, J=8.0 Hz, 2H), 3.68-3.39 (m, 4H), 2.72-2.62 (m, 1H), 2.40-2.29 (m, 2H), 2.26-2.12 (m, 4H), 1.99-1.88 (m, 2H), 1.83-1.70 (m, 2H), 1.67-1.56 (m, 2H), 1.47-1.39 (m, 1H), 1.28-1.20 (m, 1H); LC-MS (ESI): m/z 328 (M+1). The chiral purity of the product was analyzed on a chiral column with UV-detection (250 nm) using isocratic method (mobile phase: Heptane/EtOH (80/20)+0.1% Diethylamine) on Chiralpak AD-H, 4.6×150 mm, giving an enantiomeric purity of >99% ee.
  • Second Method
  • 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide (10 gram, 30.54 mmoles) was dissolved in DMSO (83 ml) at 70° C. and screened filtered into a reactor. The filter was rinsed with DMSO (17 ml) into the reactor. The temperature was decreased to 55° C. and seed crystals of 4-{(1S,25)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide were added (0.2 gram, 0.61 moles). Then after 30 is minutes the slurry was cooled to 20° C. over 3.5 hrs. At 20° C., water (40 ml) was charged over 2.5 hrs. After charging the slurry remained at 20° C. for additional 12 hrs. before isolation. The solid product was washed (displacement wash) with a mixture of DMSO (28 ml) and water (12 ml) followed by three water (3×40 ml), one slurry wash and two displacement washes. Then the solid product was dried for 18 hrs. at 60° C. to obtain the title compound in 9.32 gram (28.5 mmoles), 93% yield as crystals.
  • Third Method
  • 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide (3.0 gram, 9.16 mmoles) was dissolved in methanol (34.5 ml) and water (6.0 ml) at 65° C. and screened filtered into a reactor. Then the temperature was decreased to 55° C. and followed by addition of seed crystals of 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide (0.06 gram, 0.18 mmoles). After 5 minutes the slurry was cooled to 20° C. over 3.5 hrs. At 20° C., water (31.5 ml) was charged over 3.5 hrs. Then after additional 2 hrs, the solid form was isolated and washed twice with a mixture of methanol (9 ml) and water (3 ml). Finally, the solid product was dried at 60° C. for 15 hrs to obtain the title compound in 2.49 gram. (7.60 mmoles), 81% yield as crystals.
  • The solid product obtained in the first method was analysed by XRPD. A representative XRPD pattern is shown in FIG. 1. Selected peaks are provided in Table 1. The XRPD pattern confirmed the solid material to be Crystalline Form I.
  • TABLE 1
    Selected XRPD peaks of Crystalline Form I
    Measured
    Angle Relative
    Peak [°2Th.] intensity
    1 4.9 s
    2 5.3 m
    3 9.0 w
    4 12.6 w
    5 16.4 m
    6 16.6 m
    7 18.3 vs
    8 19.6 s
    9 20.4 s
    10 21.2 m
    11 23.2 m
    12 24.6 w
  • The solid product obtained in the second and third method, respectively, were analysed by XRPD and a representative XRPD pattern as shown in FIG. 1 and with selected peaks as are provided in Table 1 were obtained. The XRPD pattern confirmed the solid material to be Crystalline Form I.
  • to Solid product obtained according to method 1 of the Example 1 was analyzed by thermal techniques. DSC analysis indicated that Form I of Compound (I) is a high melting solid. The DSC-trace shows a small endothermic event, onset at 225° C., followed by a distinct endothermic event, onset at 235° C. A representative DSC thermogram is shown in FIG. 2.
  • X-Ray Powder Diffraction Analysis
  • An XRDP pattern for Crystalline Form I was collected using an XRPD instrumentation as described below.
  • An X-Ray Powder Diffraction (XRPD) pattern was collected under ambient conditions on a PANalytical X'Pert PRO MPD theta-theta system using long-fine-focus Cu Kα-radiation, wavelength of X-rays 1.5418 Å, at 45 kV and 40 mA. A programmable divergence slit and a programmable anti-scatter slit giving an irradiated length of 10 mm were used. 0.02 radian Soller slits were used on the incident and on the diffracted beam path. A 20 mm fixed mask was used on the incident beam path and a Nickel-filter was placed in front of a PIXcel-detector using 255 active channels. A thin flat sample was prepared on a flat zero background plate made of silicon using a spatula. The plate was mounted in a sample holder and rotated in a horizontal position during measurement. A diffraction pattern was collected between 2°2theta and 40°2theta in a continuous scan mode. Total time for the scan was approximately 10 minutes.
  • It is known in the art that an X-ray powder diffraction pattern may be obtained which has one or more measurement errors depending on measurement conditions (such as is equipment, sample preparation or machine used). In particular, it is generally known that intensities in an X-ray powder diffraction pattern may fluctuate depending on measurement conditions and sample preparation. For example, persons skilled in the art of X-ray powder diffraction will realize that the relative intensities of peaks may vary according to the orientation of the sample under test and on the type and setting of the instrument used. The skilled person will also realize that the position of reflections can be affected by the precise height at which the sample sits in the diffractometer and the zero calibration of the diffractometer. The surface planarity of the sample may also have a small effect. Hence a person skilled in the art will appreciate that the diffraction pattern data presented herein is not to be construed as absolute and any crystalline form that provides a power diffraction pattern substantially identical to those disclosed herein fall within the scope of the present disclosure (for further information see Jenkins, R & Snyder, R. L. ‘Introduction to X-Ray Powder Diffractometry’ John Wiley & Sons, 1996).
  • When herein reference is made to a compound according to the invention being crystalline, suitably the degree of crystallinity as determined by X-ray powder diffraction data, is for example greater than about 10%, is for example greater than about 20%, is for example greater than about 30%, is for example greater than about 40%, is for example greater than about 50%, is for example greater than about 60%, such as greater than about 80%, particularly greater than about 90%, more particularly greater than about 95%. In embodiments of the invention, the degree of crystallinity as determined by X-ray powder diffraction data is greater than about 98%, wherein the % crystallinity refers to the % by weight of the total sample mass which is crystalline.
  • Peak search on XRDP data of Crystalline Form I.
  • A manual peak search was done preceded by an angle correction against NIST SRM 676 alumina (α-Al2O3) standard.
  • The measured relative intensities vs. the strongest peak are given as very strong (vs) above 50%, as strong (s) between 25 and 50%, as medium (m) between 10 and 25% and as weak (w) between 5 and 10% relative peak height.
  • Differential Scanning Calorimetry (DSC) Analysis
  • DSC from 25° C. to 350° C. was performed under nitrogen in an aluminum sample cup with a perforated lid using a NETZSCH DSC 204 instrument. The scan rate was 10° C. per is minute. The sample size was less than 1 mg. It is well known that the DSC onset and peak temperatures as well as energy values may vary due to, for example, the purity of the sample and sample size and due to instrumental parameters, especially the temperature scan rate. Hence the DSC data presented are not to be taken as absolute values. A person skilled in the art can set up instrumental parameters for a Differential scanning calorimeter so that data comparable to the data presented here can be collected according to standard methods, for example those described in Höhne, G. W. H. et al (1996), Differential Scanning calorimetry, Springer, Berlin.
  • Synthesis of Intermediates Intermediate A (R)-1-(4-Bromo-phenyl)-2-chloro-ethanol
  • Figure US20110201623A1-20110818-C00004
  • Borane dimethylsulfide (2.0 kg, 24.8 moles, 94% w/w) was mixed in toluene (8 L) at tjacket=20° C. (R)-(+)-Methyl-CBS-oxazaborolidine (2.6 kg, 2.74 moles, 1M) as a toluene solution was added. The charging vessel was rinsed with toluene (0.5 L) and tjacket was set to 45° C. 1-(4-Bromo-phenyl)-2-chloro-ethanone (7.84 kg, 33.6 moles), which is commercially available from Jiangyan Keyan Fine Chemical Co. Ltd, was dissolved in 2-MeTHF (75 L) in a separate vessel and when tinner was above 40° C. in the first vessel, the 2-MeTHF solution was added during 3 h. The latter vessel was rinsed with 2-MeTHF (2 L) and added to the reaction mixture, which was left stirring at tjacket=45° C. for 1 h. At full conversion, the reaction mixture was cooled to tjacket=10° C. before slow quench with MeOH (36 L). The first litre of MeOH was added during 30 min. and the rest during additional 30 min. MeOH was distilled off under vacuum at tjacket=50° C. The organic solution left was cooled to tjacket=20° C., washed with 1M HCl in H2O (7 L conc HCl+73 L H2O) and concentrated under vacuum at tjacket=50° C. to approximately 40 L. Intermediate A obtained in a 2-MeTHF solution can be stored at 10° C. for 20 h or used directly in the next synthetic step.
  • Intermediate B (R)-2-(4-Bromo-phenyl)-oxirane
  • Figure US20110201623A1-20110818-C00005
  • Aliquat® 175 (methyl tributyl ammonium chloride) (1.12 kg, 4.75 moles) was added to Intermediate A as a 2-MeTHF solution (33.6 moles, 40 L) at tjacket=20° C. NaOH (5.1 kg, 57.4 moles, 45% w/w) diluted in H2O (2 L) was added during 20 min. The reaction mixture was left stirring at tjacket=20° C. for 2 h. At full conversion the aq. phase was separated off and the organic phase washed with H2O (2×25 L). 2-MeTHF (25 L) was added and the organic phase concentrated under vacuum at tjacket=50° C. to approximately 30 L. Intermediate B obtained in a 2-MeTHF solution, can be stored at 5° C. for 140 h or used directly in the next synthetic step.
  • Intermediate C (1S,2S)-2-(4-Bromo-phenyl)-cyclopropanecarboxylic acid
  • Figure US20110201623A1-20110818-C00006
  • Triethyl phosphonoacetate (10.5 L, 51.9 moles, 98% w/w) was dissolved in 2-MeTHF (14 L) at tjacket=−20° C. Hexyl lithium in hexane (21 L, 48.3 moles, 2.3 M) was added at a rate to maintain tinner below 0° C. The charging vessel was rinsed with 2-MeTHF (3 L) and the reaction solution was left stirring at tjacket=10° C. Intermediate B as a 2-MeTHF solution (33.6 moles, 30 L) was added during 20 min. The charging vessel was rinsed with 2-s MeTHF (2 L) and the reaction solution was left stirring at tjacket=65° C. for at least 16 h with the last 3 h at tjacket=75° C. At full conversion the reaction solution was cooled to tjacket=20° C. NaOH (7.6 kg, 85.5 moles, 45% w/w) diluted in H2O (12 L) was added over 20 min. The reaction solution obtained was left stirring at tjacket=60° C. for at least 2 h. At full conversion the reaction solution was cooled to tjacket =20° C., the aq. phase was separated off and the organic phase was extracted with H2O (37 L). The combined aq. phases were acidified to pH<3.5 with H3PO4 (9 L, 131 moles, 85% w/w) diluted in H2O (12.5 L). Only 17 L of the diluted H3PO4(aq) was used to achieve the pH<3.5. The acidic aq. phase was extracted with 2-MeTHF (2×15 L). The combined organic phases including rinsing with 2-MeTHF (2 L) were concentrated under vacuum at tjacket=50° C. to approximately 11 L. is The 2-MeTHF solution was diluted with EtOH (14.5 L) at tjacket=35° C. and H2O (16 L) was added over 20 min. The reaction solution was cooled to tjacket=28° C. Seed (16 g, 0.066 moles) was added and the solution was stirred for 2 h at tjacket=28° C. The reaction mixture was cooled to tjacket =0° C. over 6 h and left stirring for at least 1 h. Additional H2O (8 L) was added during 40 min. and the product was filtered off and washed with cold H2O (10 L). Drying under vacuum at 40° C. gave 6.18 kg Intermediate C (21.5 moles, 84% w/w), 64% yield over four steps from 7.84 kg 1-(4-bromo-phenyl)-2-chloro-ethanone (33.6 moles).
  • Recrystallization of Intermediate C: Two batches of Intermediate C (6.18+7.04 kg) were mixed in EtOH (52 L) and heated at tjacket=70° C. H2O (52 L) was added. The reaction solution was cooled to tjacket=30° C. over 2.5 h. H2O (16 L) was added during 20 min. and the crystallization was cooled to tjacket=20° C. during 3 h. The product was filtered off and washed with a mixture of H2O (8 L) and EtOH (2 L). Drying under vacuum at 40° C. gave 10.0 kg Intermediate (41.5 moles, 88% w/w), which was redissolved in toluene (39 L) and isooctane (57 L) at tjacket=60° C. A clear solution was obtained. The reaction solution was cooled to tjacket=45° C. and left stirring for 1 h, then cooled to tjacket=20° C. over 2 h. The product was filtered off and washed with a mixture of toluene (4 L) and isooctane (36 L) in two portions. Drying under vacuum at 40° C. gave 7.4 kg Intermediate C (29.8 moles, 97% w/w), 44% yield over four steps from 7.84+7.93 kg 1-(4-bromo-phenyl)-2-chloro-ethanone (67.5 moles). 1H-NMR (DMSO-d6): δ 12.36 (s, 1H), 7.44 (d, 2H, J=8 Hz), 7.13 (d, 2H, J=8 Hz), 2.39 (m, 1H), 1.81 (m, 1H), 1.43 (m, 1H), 1.33 (m, 1H); 13C-NMR (DMSO-d6): δ 173.76, 139.88, 131.20, 128.24, 119.14, 24.73, 24.31, 16.78; LC-MS (ESI): m/z 239 (M−1 (Br79)) and 241 (M−1 (Br81)).
  • Intermediate D (1S,2S)-2-(4-Cyano-phenyl)-cyclopropanecarboxylic acid
  • Figure US20110201623A1-20110818-C00007
  • Intermediate C (3.7 kg, 14.9 moles, 97% w/w) and zinc-dust (98%+, <10 μm) (99 g, 1.51 moles) were mixed with DMF (13.5 L) and the slurry was stirred at tjacket=20° C. The mixture was inerted and left with N2 pressure of 0.1-0.2 bar. Bis(tri-t-butylphosphine)palladium (0) (27.5 g, 0.054 moles) was added to the slurry, and the vessel was inerted and left with N2 pressure of 0.1-0.2 bar. The mixture was heated to tjacket=45° C., Zn (CN)2 (1.0 kg, 8.52 moles) was added to the suspension in one portion, and the system was inerted and left with N2 pressure of 0.1-0.2 bar (N. B. Cyanide salts are highly toxic). The resulting mixture was heated to tjacket=75° C. and stirred for at least 2 h. At full is conversion the reaction mixture was cooled to tjacket=20° C. Thiol-functionalized silica (Silicycle, SiliaBond Thiol) (1.07 kg, 28% w/w) was added and the vessel was inerted. The reaction mixture was stirred for at least 36 h at tjacket=20° C. The scavenger was filtered off via a filter with activated charcoal or equivalent (pall-filter). The vessel and the filter system were washed with 2-MeTHF (53 L). The filtrate and washings were combined and stirred at tjacket=5° C. A pale yellow liquid resulted. NaCl (3.5 kg) in H2O (16.4 L) was added during 15 min. at such a rate so the inner temperature remained below 15° C. The resulting reaction mixture was heated to tjacket=45° C. and the aq. phase was separated off. The organic phase was washed with NaHSO4×H2O in H2O (2×(2.87 kg+16.4 L)) and NaCl in H2O (3.5 kg+16.4 L). The organic phase was cooled to tjacket=10° C. and NaOH (1.54 kg, 19.3 moles, 50% w/w) diluted in H2O (41 L) was added during 45 min. The resulting reaction mixture was heated to tjacket=30° C. and the organic phase separated off. The aq. phase was stirred at tjacket=20° C. and pH adjusted to 6.5 with H3PO4 (0.90 kg, 7.81 moles, 85% w/w) diluted in H2O (5.3 L) at a rate that maintained the inner temperature below 25° C. 2-MeTHF and H2O were distilled off under vacuum until a volume 85-90% of the volume prior to distillation, approximately 8 L. The reaction mixture was cooled to tjacket=0° C. and continued charging off H3PO4 (1.17 kg, 10.1 moles, 85% w/w) diluted in H2O (8.2 L) until pH=4. The slurry was left stirring overnight at tjacket=10° C. The product was filtered off, washed with H2O (2×4 L). Drying under vacuum at 40° C. gave Intermediate D (2.24 kg, 11.2 moles, 93.2% w/w), 75% yield. 1H-NMR (DMSO-d6): δ 12.45 (s, 1H), 7.72 (d, 2H, J=8 Hz), 7.37 (d, 2H, J=8 Hz), 2.50 (m, 1H), 1.94 (m, 1H), 1.50 (m, 1H), 1.42 (m, 1H); 13C-NMR (DMSO-d6): δ 173.51, 146.68, 132.27, 126.93, 118.97, 108.85, 25.16, 25.04, 17.44; LC-MS (ESI): m/z 186 (M−1).
  • Intermediate E (1S,2S)-2-(4-Carbamoyl-phenyl)-cyclopropanecarboxylic acid
  • Figure US20110201623A1-20110818-C00008
  • Intermediate D (4.46 kg, 22.0 moles, 92.5% w/w) was mixed in H2O (40 L) at tjacket=30° C. NaOH (2.25 kg, 28.1 moles, 50% w/w) diluted in H2O (6 L) was added at such a rate so tinner remained below 35° C. The charging vessel was rinsed with H2O (1 L). If the pH was not >12, more NaOH was charged in the same concentration as previously. Hydrogen peroxide (4.89 kg, 50.3 moles, 35% w/w) was added at a rate to maintain tinner below 35° C. The charging vessel was rinsed with H2O (1 L) and the reaction slurry was left stirring for 0.5-1.0 h. At full conversion the reaction mixture was cooled to tjacket=0° C. and left stirring for at least 0.5 h when the temperature was reached. The sodium salt of Intermediate E was filtered off and washed with cold H2O (2×7 L). The solid was slurry washed on the filter with NaHSO4×H2O (2.76 kg, 20.0 moles) diluted in H2O (35 L). The slurry was kept stirring at tjacket=0° C. for 1 h. If the pH was not <3.7, it was adjusted with NaHSO4×H2O in H2O. The product was filtered off, washed with cold H2O (3×14 L). Drying under vacuum at 40° C. gave Intermediate E (4.0 kg, 18.2 moles, 93.4% w/w), 83% yield. 1H-NMR (DMSO-d6): δ 12.40 (s, 1H), 7.94 (s, 1H), 7.79 (d, 2H, J=8 Hz), 7.32 (s, 1H), 7.23 (d, 2H, J=8 Hz), 2.44 (m, 1H), 1.88 (m, 1H), 1.47 (m, 1H), 1.39 (m, 1H); 13C-NMR (DMSO-d6): δ 173.83, 167.67, 143.94, 132.17, 127.68, 125.73, 25.21, 24.67, 17.11; LC-MS (ESI): m/z 206 (M+1). The product was analyzed on a chiral column with UV-detection using isocratic method (mobile phase: EtOH/Isohexane/TFA (15/85/0.1 v/v/v)) on Kromosil 3-Amycoat, 150×4.6 mm, 3 μm particle size, giving an enantiomeric purity of >99% ee, Rt=13.40 min (isomer 1) and 22.22 min (isomer 2).
  • Intermediate F 1-Cyclobutylpiperazine×2HCl
  • Figure US20110201623A1-20110818-C00009
  • N-Boc-piperazine (46 g, 0.25 moles), which is commercially available from SAFC, was dissolved in EtOH (415 mL) at tjackei=20° C. Acetic acid (140 mL) was added in one portion followed by the addition of cyclobutanone (26.5 g, 0.37 moles). The charging vessel was rinsed with EtOH (25 mL) and the light yellow solution was left stirring at tjacket=20° C. for 1 h. NaBH(OAc)3 (80 g, 0.36 moles, 95% w/w) was added in 20 portions is over 2 h. EtOH (25 mL) was used for rinsing. The reaction mixture was left stirring for 2 h. At full conversion NAOH (296 g, 3.70 moles, 50% w/w) diluted in H2O (230 mL) was added at such a rate so tinner remained below 35° C.
  • EtOH was distilled off under vacuum at tjacket=45° C. to approximately 650 mL. The water phase was extracted with toluene (550 mL) at tjacket=45° C. and the obtained organic phase was concentrated under vacuum at tjacket=45° C. to approximately 250 mL. The toluene solution was diluted with 2-propanol (140 mL) at tjacket=20° C. and H2O (2.2 mL, 0.12 moles) was added. HCl in 2-propanol (82 mL, 0.49 moles, 6M) diluted in 2-propanol (140 mL) was added over 30 min at tjacket=20° C. The reaction solution was heated to tjacket=48° C. HCl in 2-propanol (164 mL, 0.99 moles, 6M) diluted in 2-propanol (276 mL) was added over 2 h at tjacket=46° C. The reaction solution was kept at tjacket=48° C. for an additional 4 h before cooling to tjacket=10° C. over 1 h. The product was filtered off and washed with cold 2-propanol (2×230 mL). Drying under vacuum at 40° C. gave 44 g Intermediate F (0.20 moles, 95.9% w/w), 80% yield. 1H-NMR (DMSO-d6): δ 12.46 (s, 1H), 10.07 (s, 2H), 3.73 (m, 1H), 3.05-3.61 (m, 8H), 2.37 (m, 2H), 2.14 (m, 2H), 1.70 (m, 2H); 13C-NMR (DMSO-d6): δ 58.05, 44.67, 39.59, 24.38, 13.18.
  • General Methods
  • 1H NMR spectra were recorded in the indicated deuterated solvent at 400 MHz or 500 MHz. The 400 MHz spectra were obtained using a Bruker av400 NMR spectrometer equipped with a 3 mm flow injection SEI 1H/D-13C probe head with Z-gradients, using a BEST 215 liquid handler for sample injection, or using a Bruker DPX400 NMRor Bruker 500 MHz ultrashield spectrometer equipped with a 4-nucleus probehead with Z-gradients. Chemical shifts are given in ppm down- and upfield from TMS. Resonance multiplicities are denoted s, d, t, q, m and br for singlet, doublet, triplet, quartet, multiplet, and broad respectively.
  • Mass spectra (MS) were run using an automated system with electrospray (+ESI) ionization. Generally, only spectra where parent masses are observed are reported. The lowest mass major ion is reported for molecules where isotope splitting results in multiple mass spectral peaks (for example when chlorine or bromine is present).
  • LC-MS analyses were recorded on a Waters LCMS equipped with a Waters X-Terra MS, C8-column, (3.5 μm, 100 mm×3.0 mm i.d.). The mobile phase system consisted of A: 10 mM ammonium acetate in water/acetonitrile (95:5) and B: acetonitrile. A linear gradient was applied running from 0% to 100% B in 4-5 minutes with a flow rate of 1.0 mL/min. The mass spectrometer was equipped with an electrospray ion source (ESI) operated in a positive or negative ion mode. The capillary voltage was 3 kV and the mass spectrometer was typically scanned between m/z 100-700. Alternative, LC-MS HPLC conditions were as follows: Column: Agilent Zorbax SB-C8 (5 μm, 50 mm×2 mm i.d) Flow: 1.0 mL/minGradient: 95% A to 100% Bin 5 min. A=5% acetonitrile in water with 0.1% formic acid and B=acetonitrile with 0.1% formic acid, UV-DAD 210-400 nm. Alternative, LC-MS analyses were recorded on a Waters 2790 LCMS equipped with a Phenomenex Luna C18 (5 μm, 50×4.6 mm i.d.) The mobile phase system consisted of A: 10 mM ammonium formate (pH 4) in water and B: acetonitrile. A linear gradient was applied running from 95% to 5% B in 5 minutes with a flow rate of 2.0 mL/min. The mass spectrometer was equipped with an electrospray ion source (ESI) operated in a positive or negative ion mode. The capillary voltage was 3 kV and the mass spectrometer was typically scanned between m/z 100-700. Alternative, LC-MS analyses were recorded on a Agilent 1200 LCMS equipped with a Zorbax SB C8 (3.5 μm, 150×4.6 mm i.d.) The mobile phase system consisted of A: 0.05% TFA in water and B: acetonitrile. A linear gradient was applied running from 10% to 90% B in 8 minutes with a flow rate of 1.0 mL/min. The mass spectrometer was equipped with an electrospray ion source (ESI) operated in a positive or negative ion mode. The capillary voltage was 3 kV and the mass spectrometer was typically scanned between m/z 100-700.
  • The compounds have been named using CambridgeSoft MedChem ELN v2.1, ACD/Name, version 8.08, software from Advanced Chemistry Development, Inc. (ACD/to
  • ABBREVIATION LIST
  • ACN: acetonitrile; aq: aqueous; br: broad; Bu: butyl; calcd: calculated; Celite®: brand of diatomaceous earth filtering agent, registered trader of Celite Corporation; d: doublet; dd: doublet of doublet; ddd: doublet of doublet of doublet; dddd: doublet of doublet of doublet of doublet; DMF: N,N-dimethyl formamide; DMSO: dimethyl sulfoxide; dq: doublet of is quartet; DSC: differential scanning calorimetry; dt: doublet of triplet; DVS: dynamic vapour sorption; EDC: 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride; ESI: electrospray ionization; EtOAc: ethyl acetate; EtOH: ethanol; Et: ethyl; FT-IR: Fourier-transform infrared; FT-Raman: Fourier transform Raman; g: gram; h: hour(s); 1H NMR: proton nuclear magnetic resonance; HOBT: N-Hydroxybenzotriazole; HPLC: high pressure liquid chromatography; iPrOH: iso-propanol; L: liter; m: multiplet; M: molar; mL: milliliter; Me: methyl; MeOH: methanol; mg: milligram; 2-MeTHF: 2-methyl tetrahydrofuran; MHz: megahertz; min: minute(s); mmol: millimole; mol: mole; MS: mass spectrometry; MTBE: methyl tert-butyl ether; NaHCO3: sodium bicarbonate; Pd/C: palladium on carbon; ppm: parts per million; q: quartet; quin: quintet; rt: room temperature; s: singlet; sat: saturated; t: triplet; TEA: triethylamine; tBuOH: tert-butanol; td: triplet of doublet; TFA: trifluoroacetic acid; TGA=thermalgravimetric analysis; THF: tetrahydrofuran; UV=ultraviolet; XRPD=X-ray powder diffraction; and the prefixes n-, s-, i-, t- and tert- have their usual meanings normal, secondary, iso, and tertiary.
  • Pharmacology
  • Human Histamine H3 Binding Assay with the Agonist Radioligand [3H]-N-a-Methylhistamine
  • The H3 binding assay was used to evaluate the ability of a compound of formula (I) to inhibit [3H]-N-a-methylhistamine under the conditions as described below: All binding assays were performed in a buffer consisting of 20 mmol/L Hepes, 100 mmol/L NaCl. The pH was set at 7.4 at room temperature. Liquid handling robots were used to prepare, in drug plates, 10 points dose response curves of the compound of formula (I) (3-fold serial dilutions, starting at 10 μmol/L). The assay, performed in 96-well plates, consisted of 100 μL, containing 20 μL of buffer alone for total binding (column #1), 20 μL of imetit (5×) for the non specific binding (column #2), 20 μL of the compound of formula (I) (5×) at varying concentrations (column #3 to #12), plus 20 μL of [3H]-N-a-Methylhistamine (25 000 dpm/well, 1.5 nmol/L) in all wells and finally 60 μL of membranes (20 μg of protein/well) mix is added to start the reaction. Membranes and SPA beads (1000 μg/well) were mixed together and incubated for 30 minutes at room is temperature prior to the start of the assay. Eight wells (column #1) were used to define total binding and 1 μmol/L imetit (eight wells, column #2) define the non-specific binding. Plates were then mixed on an orbital mixer, incubated 90 minutes at room temperature, and then read on a Trilux 384™ counter.
  • Guanosine 5′-O-(3-[35S]thio)triphosphate [GTPgS] Binding Assay
  • [35S]GTPγS binding assay was performed in a buffer consisting of 20 mmol/L Hepes, 100 mmol/L NaCl, 10 mmol/L MgCl2, 3 μg/mL saponine and 10 μmol/L GDP. The pH was set at 7.4 at room temperature. Liquid handling robots were used to prepare, in drug plates, 10 points dose response curves of the compound of formula (I) (3-fold serial dilutions, starting at 1 μmol/L for hH3 receptors and 10 μmol/L for rH3 receptors). The assay, performed in 96-well plates, consisted of 120 μL, containing 20 μL of RαMH (EC80, antagonist mode), 20 μL the compound of formula (I) at varying concentrations, 20 μL of the tracer [35S]GTPγS (60000 dpm/well, 0.2 nmol/L) and finally 60 μL of membranes (10 μg of protein/well for hH3) mix is added to start the reaction. Membranes, SPA beads (125 μg/well) and GDP were mixed together and incubated for 30 minutes at room temperature prior to the start of the assay. Eight wells were used to define baseline and RαMH EC80 (30 nmol/L for hH3 receptors) define the positive control. Plates were then mixed (2 minutes) on an orbital mixer, incubated 60 minutes at room temperature, and then read on a Trilux 384™ counter.
  • Results
  • A compound formula (I) showing pIC50 values that were generated in accordance with the assays described above, the inhibit specific binding of [3H]-N-α-Methyl Histamine to the human H3 receptor (445 aa)) as 7.9±0.060 (13 nmol/L) and the inhibition of H3 agonist R-α-Methyl-Histamine stimulated-[35S]GTPγS binding as 8.5±0.14 (3.0 nmol/L).

Claims (22)

1. A crystalline form of compound (I), 4-{(1S,2S)-2-[(4-cyclobutylpiperazin-1-yl)carbonyl]-cyclopropyl}-benzamide,
Figure US20110201623A1-20110818-C00010
2. The crystalline form of compound (I) of claim 1, wherein said form has an XRPD pattern (Cu Kα) with at least one peak at about 18.3 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
3. The crystalline form of compound (I) of claim 1, wherein in that said form has an XRPD pattern (Cu Kα) with at least two peaks at about 4.9 and about 18.3 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
4. The crystalline form of compound (I) of claim 1, wherein said form has an XRPD pattern (Cu Kα) with at least three peaks at about 4.9, about 18.3 and about 20.4 °2Theta when measured using radiation with a wavelength of about 1.54 angstroms.
5. The crystalline form of compound (I) of claim 1, wherein said form has an XRPD pattern (Cu Kα) with at least four peaks at about 4.9, about 18.3 about 19.6 and 20.4 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
6. The crystalline form of compound (I) of claim 3, wherein the XRPD pattern (Cu Kα) further comprises peaks at about 16, 4 and about 16.6, about 18.3 and about 19.6 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
7. The crystalline form of compound (I) of claim 3, wherein the XRPD pattern (Cu Kα) further comprises a peak at about 5.3 °2Theta, when measured using radiation with a wavelength of about 1.54 angstroms.
8. The crystalline form of compound (I) of claim 1, wherein said form has an XRPD pattern (Cu Kα) with peaks at about 4.9, about 5.3, about 9.0, about 12.6, about 16.4, about 16.6, about 18.3, about 19.6, about 20.4, about 21.2, about 23.2 and about 24.6 °2Theta when measured using radiation with a wavelength of about 1.54 angstroms.
9. The crystalline form of compound (I) of claim 1, wherein the XRPD pattern is essentially as shown in FIG. 1.
10. Thecrystalline form of compound (I) of claim 3, wherein the DSC thermogram comprises an endotherm at about 235° C.
11. The crystalline form of compound (I) of claim 10, wherein the DSC thermogram comprises an additional small endotherm at about 225° C.
12. The crystalline form of compound (I) of claim 3, wherein the DSC thermogram is essentially as shown in FIG. 2.
13. The crystalline form of compound (I) of claim 1, which is substantially pure.
14. A pharmaceutical formulation comprising a crystalline form of compound (I) as claimed in claim 1 in admixture with a pharmaceutically acceptable adjuvant, diluent and/or carrier.
15. (canceled)
16. (canceled)
17. (canceled)
18. A method for the treatment of a disorder selected from schizophrenia, narcolepsy, excessive daytime sleepiness, obesity, attention deficit hyperactivity disorder, pain, neuropathic pain, Alzheimer's disease, cognition deficiency, and cognition deficiency associated with schizophrenia, in a warm-blooded animal, comprising administering to said animal in need of such therapy a therapeutically effective amount of a crystalline form of compound (I) according to claim 1.
19. The crystalline form of compound (I) of claim 8, wherein the DSC thermogram comprises an endotherm at about 235° C.
20. The crystalline form of compound (I) of claim 9, wherein the DSC thermogram comprises an endotherm at about 235° C.
21. The crystalline form of compound (I) of claim 8, wherein the DSC thermogram is essentially as shown in FIG. 2.
22. The crystalline form of compound (I) of claim 9, wherein the DSC thermogram is essentially as shown in FIG. 2.
US13/029,362 2010-02-18 2011-02-17 Crystalline Form Of A Cyclopropyl Benzamide Derivative Abandoned US20110201623A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/029,362 US20110201623A1 (en) 2010-02-18 2011-02-17 Crystalline Form Of A Cyclopropyl Benzamide Derivative

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US30558310P 2010-02-18 2010-02-18
US13/029,362 US20110201623A1 (en) 2010-02-18 2011-02-17 Crystalline Form Of A Cyclopropyl Benzamide Derivative

Publications (1)

Publication Number Publication Date
US20110201623A1 true US20110201623A1 (en) 2011-08-18

Family

ID=44370080

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/579,605 Expired - Fee Related US9012452B2 (en) 2010-02-18 2011-02-17 Processes for making cyclopropyl amide derivatives and intermediates associated therewith
US13/029,362 Abandoned US20110201623A1 (en) 2010-02-18 2011-02-17 Crystalline Form Of A Cyclopropyl Benzamide Derivative

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/579,605 Expired - Fee Related US9012452B2 (en) 2010-02-18 2011-02-17 Processes for making cyclopropyl amide derivatives and intermediates associated therewith

Country Status (20)

Country Link
US (2) US9012452B2 (en)
EP (2) EP2536702A4 (en)
JP (3) JP2013520413A (en)
KR (2) KR20130004296A (en)
CN (2) CN103168027B (en)
AR (2) AR080204A1 (en)
AU (2) AU2011218492B2 (en)
BR (2) BR112012020629A2 (en)
CA (2) CA2790040A1 (en)
CL (2) CL2012002259A1 (en)
CO (1) CO6592109A2 (en)
ES (1) ES2541857T3 (en)
HK (1) HK1178142A1 (en)
IL (1) IL221431A (en)
MX (2) MX336333B (en)
NZ (2) NZ601920A (en)
RU (2) RU2012136148A (en)
SG (3) SG182732A1 (en)
TW (2) TWI494301B (en)
WO (2) WO2011102795A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100216812A1 (en) * 2009-02-20 2010-08-26 Astrazeneca Ab Cyclopropyl Amide Derivatives
US20110201622A1 (en) * 2010-02-18 2011-08-18 Collins Craig D Solid Forms Comprising A Cyclopropyl Amide Derivative
WO2014147353A1 (en) * 2013-03-22 2014-09-25 Les Laboratoires Servier Use of 4-{3-[cis-hexahydrocyclopenta[c]pyrrole-2(1h)-yl]propoxy}benzamide for treating neuropathic pain
US9012452B2 (en) 2010-02-18 2015-04-21 Astrazeneca Ab Processes for making cyclopropyl amide derivatives and intermediates associated therewith
US9029381B2 (en) 2007-08-22 2015-05-12 Astrazeneca Ab Cyclopropyl amide derivatives
CN111718281A (en) * 2013-03-22 2020-09-29 默克专利有限公司 Synthetic structural unit for preparing material for organic electroluminescent device

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103342655A (en) * 2013-07-02 2013-10-09 扬州大学 New method for synthesizing substituted amide by using substituted ethanedione dianiline Schiff base

Citations (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3331830A (en) * 1963-04-04 1967-07-18 American Cyanamid Co 1-substituted-4-substituted aminoalkylene piperazines
US3449427A (en) * 1965-06-03 1969-06-10 Smithkline Corp Aminocyclopropane derivatives of 5h-dibenzo(a,d)cycloheptenes
US3686335A (en) * 1970-12-21 1972-08-22 Smith Kline French Lab 5-vinyl-5h-di benzo(a,d)cycloheptenes
US3697506A (en) * 1970-08-07 1972-10-10 Pfizer Crystalline alkali metal salts of {60 -carboxybenzylpenicillin and process therefor
US4432987A (en) * 1982-04-23 1984-02-21 Pfizer Inc. Crystalline benzenesulfonate salts of sultamicillin
US4547505A (en) * 1983-03-25 1985-10-15 Degussa Aktiengesellschaft N-Phenyl-N-'-cycloalkylalkanoylpiperazine useful as analgetics and process for its production
US5112818A (en) * 1989-12-29 1992-05-12 Banyu Pharmaceutical Co., Ltd. 2-(2-cyclopropylpyrrolidin-4-ylthio)carbapenhem derivatives
US5434303A (en) * 1991-09-19 1995-07-18 Bayer Aktiengesellschaft Process for the preparation of 1-fluoro-cyclopropane-1-carboxylic acid
US5721359A (en) * 1993-03-12 1998-02-24 Pharmacia & Upjohn Company Crystalline ceftiofur free acid
US6284761B1 (en) * 1999-01-08 2001-09-04 Neurogen Corporation 1-phenyl-4-(1-[2-aryl]cyclopropyl)methylpiperazines: dopamine receptor ligands
US6383520B1 (en) * 1998-06-26 2002-05-07 Chugai Seiyaku Kabushiki Kaisha Fine powder of L-α-aminoadipic acid derivative, oral solid preparations containing the same, and method for treatment of bulk powders
US6521619B2 (en) * 2000-06-29 2003-02-18 Icos Corporation Aryl phenylcyclopropyl sulfide derivatives and their use as cell adhesion inhibiting anti-inflammatory and immune suppressive agents
US20040077618A1 (en) * 2002-10-22 2004-04-22 Bennani Youssef L. Cycloalkylamides and their therapeutic applications
US20040209858A1 (en) * 2002-10-22 2004-10-21 Bennani Youssef L. Cycloalkylamides and their therapeutic applications
US6861432B2 (en) * 2001-11-23 2005-03-01 Schering Aktiengesellschaft Piperazine derivatives that destabilize androgen receptors
US20050113309A1 (en) * 2002-03-22 2005-05-26 Aeri Kim Crystalline forms(2s)-n-5[amino(imino)methyl]-2-thienylmethyl-1-(2r)-2[(carboxymethyl) amino]-3,3-diphenylpropanoyl-2-pyrrolidinecarboxamide nh2o
US20050137201A1 (en) * 2003-09-04 2005-06-23 Alex Aronov Compositions useful as inhibitors of protein kinases
US20050143372A1 (en) * 2001-10-30 2005-06-30 Shomir Ghosh Compounds, pharmaceutical compositions and methods of use therefor
US7053089B2 (en) * 2001-02-23 2006-05-30 Merck & Co., Inc. N-substituted nonaryl-heterocyclic NMDA/NR2B antagonists
US7145002B2 (en) * 2001-09-26 2006-12-05 Merck & Co. Inc. Crystalline forms of carbapenem antibiotics and methods of preparation
US20070054917A1 (en) * 2003-04-23 2007-03-08 Gordon Bruton Piperazine derivates and their use for the treatment of neurological and psychiatric diseases
US20070066821A1 (en) * 2005-09-16 2007-03-22 Allison Brett D Cyclopropyl amines as modulators of the histamine h3 receptor
US20070167436A1 (en) * 2006-01-13 2007-07-19 Matthias Nettekoven Cyclohexyl piperazinyl methanone derivatives
US20080021081A1 (en) * 2006-06-23 2008-01-24 Huaqing Liu Cyclopropyl amine derivatives
US20080242653A1 (en) * 2006-06-23 2008-10-02 Huaqing Liu Cyclopropyl amine derivatives
US20090076020A1 (en) * 2007-08-22 2009-03-19 Astrazeneca Ab Cyclopropyl Amide Derivatives 978
US20090093525A1 (en) * 2007-10-04 2009-04-09 Roche Palo Alto Llc Cyclopropyl aryl amide derivatives and uses thereof
US20090154067A1 (en) * 2006-09-21 2009-06-18 Junichi Kurita Chip-type filter
US20090181981A1 (en) * 2008-01-15 2009-07-16 Jeanette Tower Dunlap Crystalline (r)-2-(4-cyclopropanesulphonyl-phenyl)-n-pyrazin-2-yl-3-(tetrahydropyran-4-yl)-propionamide
US20100216812A1 (en) * 2009-02-20 2010-08-26 Astrazeneca Ab Cyclopropyl Amide Derivatives
US20110201622A1 (en) * 2010-02-18 2011-08-18 Collins Craig D Solid Forms Comprising A Cyclopropyl Amide Derivative
US20130172560A1 (en) * 2010-02-18 2013-07-04 Astrazeneca Ab Processes for making cyclopropyl amide derivatives and intermediates associated therewith

Family Cites Families (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL128365C (en) 1963-11-05
CS244821B2 (en) 1983-06-16 1986-08-14 Boehringer Ingelheim Ltd Production method of new substituted phenylalkyl(piperazinyl or homopiperazinyle)-prpylureas or thioureas
DE3418167A1 (en) 1984-05-16 1985-11-21 Bayer Ag, 5090 Leverkusen Use of amides for improving the crop plant compatibility of herbicidally active heteroaryloxyacetamides
DE3600288A1 (en) 1986-01-08 1987-07-16 Bayer Ag USE OF AMIDES FOR IMPROVING THE CROP PLANT TOLERABILITY OF HERBICIDE-ACTIVE SULFONYL URINE DERIVATIVES
DE3618004A1 (en) 1986-05-28 1987-12-03 Bayer Ag Use of amides for improving the crop plant tolerance of herbicidally active sulphonyliso(thio)urea derivatives
CA2071897A1 (en) 1989-12-28 1991-06-29 Richard A. Glennon Sigma receptor ligands and the use thereof
EP0531271A1 (en) 1991-08-22 1993-03-10 Monsanto Company Safening herbicidal pyrazolysulfonylureas
AU4368996A (en) 1994-11-23 1996-06-17 Neurogen Corporation Certain 4-aminomethyl-2-substituted imidazole derivatives and 2-aminomethyl-4-substituted imidazole derivatives; new classes of dopamine receptor subtype specific ligands
US5604253A (en) 1995-05-22 1997-02-18 Merck Frosst Canada, Inc. N-benzylindol-3-yl propanoic acid derivatives as cyclooxygenase inhibitors
US5859246A (en) 1997-01-30 1999-01-12 Neurogen Corporation 1-phenyl-4-benzylpiperazines: dopamine receptor subtype specific ligands
EP0968200A1 (en) 1997-02-24 2000-01-05 ZymoGenetics, Inc. Calcitonin mimetics
US6160134A (en) 1997-12-24 2000-12-12 Bristol-Myers Squibb Co. Process for preparing chiral cyclopropane carboxylic acids and acyl guanidines
ZA99607B (en) 1998-01-27 1999-07-27 Rhone Poulenc Rorer Pharma Substituted oxoazaheterocyclyl factor xa inhibitors.
US6268367B1 (en) 1998-02-23 2001-07-31 Zymogenetics, Inc. Piperazine derivatives for treating bone deficit conditions
CN1167694C (en) 1999-01-08 2004-09-22 纽罗根公司 1-phenyl-4-(1-[2-aryl]cyclopropyl) methylpiperazines: dopamine receptor ligands
GB9913083D0 (en) * 1999-06-04 1999-08-04 Novartis Ag Organic compounds
US6936602B1 (en) 1999-06-16 2005-08-30 Takeda Chemical Industries, Ltd. Benzazepine derivatives, process for the preparation of the same and uses thereof
AU7715300A (en) 1999-09-27 2001-04-30 Merck & Co., Inc. Method of preventing osteoporosis
CA2396079A1 (en) * 2000-01-07 2001-07-19 Transform Pharmaceuticals, Inc. High-throughput formation, identification, and analysis of diverse solid-forms
AU6872401A (en) 2000-06-29 2002-01-14 Abbott Lab Aryl phenycyclopropyl sulfide derivatives and their use as cell adhesion-inhibiting anti-inflammatory and immune-suppressive agents
CN1443170A (en) 2000-07-20 2003-09-17 神经原公司 Capsaicin receptor ligands
JPWO2002051781A1 (en) 2000-12-25 2004-04-22 味の素株式会社 Method for producing optically active halohydrin compound
DE60234453D1 (en) 2001-07-02 2009-12-31 High Point Pharmaceuticals Llc SUBSTITUTED PIPERAZINE AND DIAZEPANDERIVATES FOR USE AS HISTAMINE H3 RECEPTOR MODULATORS
WO2003014110A1 (en) 2001-08-08 2003-02-20 Takeda Chemical Industries, Ltd. Benzazepine derivative, process for producing the same, and use
AU2003236686A1 (en) 2002-06-07 2003-12-22 Altana Pharma Ag 4,5-dihydro-imidazo(4,5,1-j) quinolin-6-ones as parp inhibitors
AU2003284402A1 (en) 2002-11-15 2004-06-15 Yamanouchi Pharmaceutical Co., Ltd. Antagonist to melanin-concentrating hormone receptor
ATE384724T1 (en) 2002-12-13 2008-02-15 Smithkline Beecham Corp CYCLOPROPYL COMPOUNDS AS CCR5 ANTAGONISTS
RS20050789A (en) 2003-05-01 2008-04-04 Bristol-Myers Squibb Company, Aryl-substituted pyrazole-amide compounds useful as kinase inhibitors
ZA200605248B (en) 2003-12-15 2007-10-31 Japan Tobacco Inc Cyclopropane compounds and pharmaceutical use thereof
MX2007000028A (en) 2004-07-06 2007-07-20 Xenon Pharmaceuticals Inc Nicotinamide derivatives and their use as therapeutic agents.
AR050865A1 (en) 2004-09-09 2006-11-29 Sanofi Aventis DERIVATIVES OF 2- MORFOLINO-4-PIRIMIDONA
WO2006036015A2 (en) 2004-09-29 2006-04-06 Mitsubishi Pharma Corporation 6- (pyridinyl) -4-pyrimidone derivates as tau protein kinase 1 inhibitors
PT1802307E (en) 2004-10-15 2008-06-06 Glaxo Group Ltd Pyrrolidine derivatives as histamine receptors ligands
BRPI0517423A (en) 2004-12-27 2008-10-07 Astrazeneca Ab compound, pharmaceutical composition, method for treating or preventing neurological and psychiatric disorders associated with glutamate dysfunction in an animal, and, use in a compound
CA2595882A1 (en) 2005-01-26 2006-08-03 Pharmacia & Upjohn Company Llc Thieno [2,3-d] pyrimidine compounds as inhibitors of adp-mediated platelets aggregation
TW200640863A (en) * 2005-02-15 2006-12-01 Glaxo Group Ltd Compounds which potentiate glutamate receptor and uses thereof in medicine
RU2408580C2 (en) 2005-02-17 2011-01-10 Астеллас Фарма Инк. Pyridyl nonaromatic nitrogen-containing heterocyclo-1-carboxylate derivative
CA2602358A1 (en) 2005-03-25 2006-09-28 Pharmacia & Upjohn Company Llc 4-piperazinnylthieno [2,3-d] pyrimidine compounds as platelet aggregation inhibitors
JP2008534573A (en) 2005-03-28 2008-08-28 ファルマシア・アンド・アップジョン・カンパニー・エルエルシー 4-piperazinothieno [2,3-D] pyrimidine compounds as platelet aggregation inhibitors
WO2006103544A2 (en) 2005-03-28 2006-10-05 Pharmacia & Upjohn Company Llc 4-piperazinylthieno [2, 3-d] pyrimidine compounds as platelet aggregation inhibitors
WO2006103545A1 (en) 2005-03-28 2006-10-05 Pharmacia & Upjohn Company Llc 4-piperazinylthieno [2,3-d] pyrimidine compounds as platelet aggregation inhibitors
CA2615380A1 (en) 2005-07-15 2007-01-25 Schering Corporation Quinazoline derivatives useful in cancer treatment
TW200740779A (en) * 2005-07-22 2007-11-01 Mitsubishi Pharma Corp Intermediate compound for synthesizing pharmaceutical agent and production method thereof
AU2006275568A1 (en) 2005-08-02 2007-02-08 Neurogen Corporation Dipiperazinyl ketones and related analogues
US8158673B2 (en) 2005-10-27 2012-04-17 Pfizer Inc. Histamine-3 receptor antagonists
SI1951699T1 (en) 2005-10-31 2013-01-31 Janssen Pharmaceutica N.V. Processes for the preparation of cyclopropyl-amide derivatives
TW200740803A (en) 2005-12-21 2007-11-01 Vertex Pharma Heterocyclic derivatives as modulators of ion channels
WO2007076140A2 (en) 2005-12-23 2007-07-05 University Of Cincinnati Treatment methods employing histamine h3 receptor antagonists, including betahistine
CN101384581B (en) * 2006-02-17 2013-09-18 弗·哈夫曼-拉罗切有限公司 Benzoyl-piperidine derivatives as 5HT2/D3 modulators
WO2007098536A1 (en) 2006-03-01 2007-09-07 Cementech Pty Ltd Matrix for masonry elements and method of manufacture thereof
JP2009132621A (en) 2006-03-13 2009-06-18 Ajinomoto Co Inc Method for producing cyclopropylamide compound
WO2007111921A1 (en) 2006-03-23 2007-10-04 Amgen Inc. 1-phenylsulfonyl-diaza heterocyclic amide compounds and their uses as modulators of hydroxsteroid dehydrogenases
WO2007138928A1 (en) 2006-05-26 2007-12-06 Kaneka Corporation Process for production of optically active 3-amino-2 -hydroxypropionic cyclopropylamide derivatives and salts thereof
US20100016293A1 (en) 2006-07-03 2010-01-21 Rogier Adriaan Smits Quinazolines and Related Heterocyclic Compounds, and Their Therapeutic Use
GB0615620D0 (en) * 2006-08-05 2006-09-13 Astrazeneca Ab A process for the preparation of optically active intermediates
WO2008024284A2 (en) * 2006-08-21 2008-02-28 Merck & Co., Inc. Sulfonylated piperazines as cannabinoid-1 receptor modulators
DE102006056526A1 (en) 2006-11-30 2008-06-05 Archimica Gmbh Process for the stereoselective synthesis of chiral epoxides by ADH reduction of alpha-leaving group-substituted ketones and cyclization
US7737149B2 (en) * 2006-12-21 2010-06-15 Astrazeneca Ab N-[5-[2-(3,5-dimethoxyphenyl)ethyl]-2H-pyrazol-3-yl]-4-(3,5-dimethylpiperazin-1-yl)benzamide and salts thereof
WO2008147864A2 (en) 2007-05-22 2008-12-04 Xenon Pharmaceuticals Inc. Methods of using piperazine compounds in treating sodium channel-mediated diseases or conditions
EP2150115B1 (en) 2007-05-23 2013-09-18 Merck Sharp & Dohme Corp. Cyclopropyl pyrrolidine orexin receptor antagonists
DE602008002598D1 (en) 2007-06-11 2010-10-28 Hoffmann La Roche Cyclohexylderivate
DE102008049371A1 (en) 2008-03-06 2009-09-10 Mühlbauer Ag Device with an RFID transponder in an electrically conductive object and manufacturing method thereof
JP2011525478A (en) * 2008-03-20 2011-09-22 フォレスト・ラボラトリーズ・ホールディングス・リミテッド Novel piperazine derivatives as inhibitors of stearoyl-CoA desaturase
US8691804B2 (en) * 2008-05-08 2014-04-08 Evotec Ag Azetidines and cyclobutanes as histamine H3 receptor antagonists
CN105152919A (en) 2008-07-28 2015-12-16 赛丹思科大学 Compounds for the treatment of metabolic diseases
CN101462980B (en) * 2009-01-05 2013-01-09 扬州天辰精细化工有限公司 Industrial production method of 2,6-difluorobenzamide

Patent Citations (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3331830A (en) * 1963-04-04 1967-07-18 American Cyanamid Co 1-substituted-4-substituted aminoalkylene piperazines
US3449427A (en) * 1965-06-03 1969-06-10 Smithkline Corp Aminocyclopropane derivatives of 5h-dibenzo(a,d)cycloheptenes
US3697506A (en) * 1970-08-07 1972-10-10 Pfizer Crystalline alkali metal salts of {60 -carboxybenzylpenicillin and process therefor
US3686335A (en) * 1970-12-21 1972-08-22 Smith Kline French Lab 5-vinyl-5h-di benzo(a,d)cycloheptenes
US4432987A (en) * 1982-04-23 1984-02-21 Pfizer Inc. Crystalline benzenesulfonate salts of sultamicillin
US4547505A (en) * 1983-03-25 1985-10-15 Degussa Aktiengesellschaft N-Phenyl-N-'-cycloalkylalkanoylpiperazine useful as analgetics and process for its production
US5112818A (en) * 1989-12-29 1992-05-12 Banyu Pharmaceutical Co., Ltd. 2-(2-cyclopropylpyrrolidin-4-ylthio)carbapenhem derivatives
US5434303A (en) * 1991-09-19 1995-07-18 Bayer Aktiengesellschaft Process for the preparation of 1-fluoro-cyclopropane-1-carboxylic acid
US5721359A (en) * 1993-03-12 1998-02-24 Pharmacia & Upjohn Company Crystalline ceftiofur free acid
US6383520B1 (en) * 1998-06-26 2002-05-07 Chugai Seiyaku Kabushiki Kaisha Fine powder of L-α-aminoadipic acid derivative, oral solid preparations containing the same, and method for treatment of bulk powders
US6284761B1 (en) * 1999-01-08 2001-09-04 Neurogen Corporation 1-phenyl-4-(1-[2-aryl]cyclopropyl)methylpiperazines: dopamine receptor ligands
US6544996B2 (en) * 1999-01-08 2003-04-08 Neurogen Corporation 1-phenyl-4-(1-[2-aryl]cyclopropyl) methylpiperazines: dopamine receptor ligands
US6521619B2 (en) * 2000-06-29 2003-02-18 Icos Corporation Aryl phenylcyclopropyl sulfide derivatives and their use as cell adhesion inhibiting anti-inflammatory and immune suppressive agents
US7217716B2 (en) * 2001-02-23 2007-05-15 Merck & Co., Inc. N-substituted nonaryl-heterocyclic NMDA/NR2B antagonists
US7053089B2 (en) * 2001-02-23 2006-05-30 Merck & Co., Inc. N-substituted nonaryl-heterocyclic NMDA/NR2B antagonists
US7145002B2 (en) * 2001-09-26 2006-12-05 Merck & Co. Inc. Crystalline forms of carbapenem antibiotics and methods of preparation
US20050143372A1 (en) * 2001-10-30 2005-06-30 Shomir Ghosh Compounds, pharmaceutical compositions and methods of use therefor
US6861432B2 (en) * 2001-11-23 2005-03-01 Schering Aktiengesellschaft Piperazine derivatives that destabilize androgen receptors
US20050113309A1 (en) * 2002-03-22 2005-05-26 Aeri Kim Crystalline forms(2s)-n-5[amino(imino)methyl]-2-thienylmethyl-1-(2r)-2[(carboxymethyl) amino]-3,3-diphenylpropanoyl-2-pyrrolidinecarboxamide nh2o
US20040209858A1 (en) * 2002-10-22 2004-10-21 Bennani Youssef L. Cycloalkylamides and their therapeutic applications
US20040077618A1 (en) * 2002-10-22 2004-04-22 Bennani Youssef L. Cycloalkylamides and their therapeutic applications
US20070054917A1 (en) * 2003-04-23 2007-03-08 Gordon Bruton Piperazine derivates and their use for the treatment of neurological and psychiatric diseases
US7446199B2 (en) * 2003-09-04 2008-11-04 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of protein kinases
US20050137201A1 (en) * 2003-09-04 2005-06-23 Alex Aronov Compositions useful as inhibitors of protein kinases
US20070066821A1 (en) * 2005-09-16 2007-03-22 Allison Brett D Cyclopropyl amines as modulators of the histamine h3 receptor
US20070167436A1 (en) * 2006-01-13 2007-07-19 Matthias Nettekoven Cyclohexyl piperazinyl methanone derivatives
US20080242653A1 (en) * 2006-06-23 2008-10-02 Huaqing Liu Cyclopropyl amine derivatives
US20080021081A1 (en) * 2006-06-23 2008-01-24 Huaqing Liu Cyclopropyl amine derivatives
US20090154067A1 (en) * 2006-09-21 2009-06-18 Junichi Kurita Chip-type filter
US7612987B2 (en) * 2006-09-21 2009-11-03 Panasonic Corporation Chip-type filter
US8063215B2 (en) * 2007-08-22 2011-11-22 Astrazeneca Ab Cyclopropyl amide derivatives
US20090076020A1 (en) * 2007-08-22 2009-03-19 Astrazeneca Ab Cyclopropyl Amide Derivatives 978
US20120065193A1 (en) * 2007-08-22 2012-03-15 James Arnold Cyclopropyl Amide Derivatives '978
US20090093525A1 (en) * 2007-10-04 2009-04-09 Roche Palo Alto Llc Cyclopropyl aryl amide derivatives and uses thereof
US20090181981A1 (en) * 2008-01-15 2009-07-16 Jeanette Tower Dunlap Crystalline (r)-2-(4-cyclopropanesulphonyl-phenyl)-n-pyrazin-2-yl-3-(tetrahydropyran-4-yl)-propionamide
US20100216812A1 (en) * 2009-02-20 2010-08-26 Astrazeneca Ab Cyclopropyl Amide Derivatives
US20110201622A1 (en) * 2010-02-18 2011-08-18 Collins Craig D Solid Forms Comprising A Cyclopropyl Amide Derivative
US20130172560A1 (en) * 2010-02-18 2013-07-04 Astrazeneca Ab Processes for making cyclopropyl amide derivatives and intermediates associated therewith

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Armarego et al. ("Purification of Laboratory Chemicals", 5th Ed., Chapter 1, pp. 1-30, (c) 2003, Elsevier, Cornwall, GB *
Hickey et al, JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 96, NO. 5, MAY 2007 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9029381B2 (en) 2007-08-22 2015-05-12 Astrazeneca Ab Cyclopropyl amide derivatives
US20100216812A1 (en) * 2009-02-20 2010-08-26 Astrazeneca Ab Cyclopropyl Amide Derivatives
US8993577B2 (en) 2009-02-20 2015-03-31 Astrazeneca Ab Cyclopropyl amide derivatives
US20110201622A1 (en) * 2010-02-18 2011-08-18 Collins Craig D Solid Forms Comprising A Cyclopropyl Amide Derivative
US9012452B2 (en) 2010-02-18 2015-04-21 Astrazeneca Ab Processes for making cyclopropyl amide derivatives and intermediates associated therewith
WO2014147353A1 (en) * 2013-03-22 2014-09-25 Les Laboratoires Servier Use of 4-{3-[cis-hexahydrocyclopenta[c]pyrrole-2(1h)-yl]propoxy}benzamide for treating neuropathic pain
FR3003466A1 (en) * 2013-03-22 2014-09-26 Servier Lab USE OF 4- {3- [CIS-HEXAHYDROCYCLOPENTA [C] PYRROL-2 (1H) -YL] PROPOXY} BENZAMIDE FOR THE TREATMENT OF NEUROPATHIC PAIN
CN111718281A (en) * 2013-03-22 2020-09-29 默克专利有限公司 Synthetic structural unit for preparing material for organic electroluminescent device

Also Published As

Publication number Publication date
KR20130004296A (en) 2013-01-09
WO2011102795A1 (en) 2011-08-25
EP2536702A1 (en) 2012-12-26
TW201136897A (en) 2011-11-01
US20130172560A1 (en) 2013-07-04
SG182732A1 (en) 2012-08-30
IL221431A (en) 2015-11-30
NZ601920A (en) 2014-08-29
CA2788444A1 (en) 2011-08-25
AU2011218491C1 (en) 2015-05-14
US9012452B2 (en) 2015-04-21
SG10201501226VA (en) 2015-04-29
ES2541857T3 (en) 2015-07-27
AR080204A1 (en) 2012-03-21
HK1178142A1 (en) 2013-09-06
TW201144272A (en) 2011-12-16
MX336333B (en) 2016-01-15
AU2011218491B2 (en) 2014-11-06
CO6592109A2 (en) 2013-01-02
EP2536685B1 (en) 2015-04-08
EP2536685A4 (en) 2013-07-10
BR112012020782A2 (en) 2016-05-03
RU2012139082A (en) 2014-03-27
AU2011218492B2 (en) 2014-11-13
RU2012136148A (en) 2014-03-27
EP2536685A1 (en) 2012-12-26
TWI520945B (en) 2016-02-11
MX2012009473A (en) 2012-09-12
CN103168027B (en) 2015-08-26
TWI494301B (en) 2015-08-01
CL2012002259A1 (en) 2012-11-30
BR112012020629A2 (en) 2018-06-19
CA2790040A1 (en) 2011-08-25
IL221431A0 (en) 2012-10-31
WO2011102794A1 (en) 2011-08-25
SG183274A1 (en) 2012-09-27
MX2012009480A (en) 2012-09-12
AR080205A1 (en) 2012-03-21
NZ602110A (en) 2014-09-26
CN103168027A (en) 2013-06-19
JP2016006103A (en) 2016-01-14
AU2011218491A1 (en) 2012-10-04
KR20130002316A (en) 2013-01-07
AU2011218492A1 (en) 2012-08-23
CN102869651A (en) 2013-01-09
JP2013520414A (en) 2013-06-06
CL2012002286A1 (en) 2013-01-25
EP2536702A4 (en) 2013-07-10
JP2013520413A (en) 2013-06-06

Similar Documents

Publication Publication Date Title
AU2011218492B2 (en) New crystalline form of a cyclopropyl benzamide derivative
EP2398780B1 (en) Cyclopropyl amide derivatives targeting the histamine h3 receptor
US8063215B2 (en) Cyclopropyl amide derivatives
AU2011218490B9 (en) Solid forms comprising a cyclopropyl amide derivative
EP2627329A1 (en) Trpa1 receptor antagonist
AU2013209365A1 (en) Cyclopropyl amide derivatives targeting the histamine h3 receptor

Legal Events

Date Code Title Description
AS Assignment

Owner name: ASTRAZENECA AB, SWEDEN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ASTRAZENECA PHARMACEUTICALS LP;REEL/FRAME:025899/0951

Effective date: 20110223

Owner name: ASTRAZENECA PHARMACEUTICALS LP, DELAWARE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UCZYNSKI, MICHAEL A.;REEL/FRAME:025899/0916

Effective date: 20110217

STCB Information on status: application discontinuation

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