1-(4-(2-((1-(3,4-DIFLUOROPHENYL)-1H-PYRAZOL-3-YL)METHOXY)ETHYL)PIPERAZIN-1-YL)ETHANONE SALTS

Information

  • Patent Application
  • 20180251430
  • Publication Number
    20180251430
  • Date Filed
    September 01, 2016
    7 years ago
  • Date Published
    September 06, 2018
    5 years ago
Abstract
The present invention relates to 1-(4-(2-((1-(3,4-difluorophenyl)-1 H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone salts, specifically to the hydrochloride and to the maleate, to pharmaceutical compositions comprising them, and to their use in therapy and/or prophylaxis of sigma receptor associated diseases.
Description
FIELD OF THE INVENTION

The present invention relates to 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl) methoxy)ethyl)piperazin-1-yl)ethanone salts, specifically to the hydrogen halides salts and simple carboxylic diacid salts, to pharmaceutical compositions comprising them, and to their use in therapy and/or prophylaxis of sigma receptor associated diseases.


BACKGROUND

The search for new therapeutic agents has been greatly aided in recent years by better understanding of the structure of proteins and other biomolecules associated with target diseases. One important class of these proteins is the sigma (σ) receptor, a cell surface receptor of the central nervous system (CNS) which may be related to the dysphoric, hallucinogenic and cardiac stimulant effects of opioids. From studies of the biology and function of sigma receptors, evidence has been presented that sigma receptor ligands may be useful in the treatment of psychosis and movement disorders such as dystonia and tardive dyskinesia, and motor disturbances associated with Huntington's chorea or Tourette's syndrome and in Parkinson's disease (Walker, J. M. et al, Pharmacological Reviews, 1990, 42, 355). It has been reported that the known sigma receptor ligand rimcazole clinically shows effects in the treatment of psychosis (Snyder, S. H., Largent, B. L. J. Neuropsychiatry 1989, 1, 7). The sigma binding sites have preferential affinity for the dextrorotatory isomers of certain opiate benzomorphans, such as (+)-SKF 10047, (+)-cyclazocine, and (+)-pentazocine and also for some narcoleptics such as haloperidol.


The sigma receptor has at least two subtypes, which may be discriminated by stereoselective isomers of these pharmacoactive drugs. SKF 10047 has nanomolar affinity for the sigma-1 (σ1) receptor and has micromolar affinity for the sigma-2 (σ2) isoform. Haloperidol has similar affinities for both subtypes. Endogenous sigma ligands are not known, although progesterone has been suggested to be one of them. Possible sigma-site-mediated drug effects include modulation of glutamate receptor function, neurotransmitter response, neuroprotection, behavior, and cognition (Quirion, R. et al. Trends Pharmacol. Sci., 1992, 13:85-86). Most studies have implied that sigma binding sites (receptors) are plasmalemmal elements of the signal transduction cascade. Drugs reported to be selective sigma ligands have been evaluated as antipsychotics (Hanner, M. et al. Proc. Natl. Acad. Sci., 1996, 93:8072-8077). The existence of sigma receptors in the CNS, immune and endocrine systems have suggested a likelihood that it may serve as link between the three systems.


In view of the potential therapeutic applications of agonists or antagonists of the sigma receptor, a great effort has been directed to find selective ligands. Thus, the prior art discloses different sigma receptor ligands. 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone is one of such promising sigma receptor ligands. The compound and its synthesis are disclosed and claimed in WO 2011/147910.


1-(4-(2-((1-(3,4-Difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone is a highly selective sigma-1 (σ1) receptor antagonist. It displays strong analgesic activity in the treatment and prevention of chronic and acute pain, and particularly, neuropathic pain. The compound has a molecular weight of 364.39 Da and a pKa of 6.37. The structural formula of the compound is:




embedded image


To carry out its pharmaceutical development and realize its potential, there is a need in the art for additional forms of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone that will facilitate the preparation of better formulations of this active pharmaceutical ingredient.


In this regard, alternative forms of the compound may have widely different properties such as, for example, enhanced thermodynamic stability, higher purity or improved bioavailability (e.g. better absorption, dissolution patterns). Specific compound forms could also facilitate the manufacturing (e.g. enhanced flowability), handling and storage (e.g. non-hygroscopic, long shelf life) of the compound formulations or allow the use of a lower dose of the therapeutic agent, thus decreasing its potential side effects. Thus, it is important to provide such forms, having improved properties for pharmaceutical use.


BRIEF DESCRIPTION OF THE INVENTION

In the present invention, after an extensive research on different forms of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone, it is surprisingly found and demonstrated that some of its crystalline salts and specifically the hydrogen halides salts and simple carboxylic diacid salts provides advantageous production, handling, storage and/or therapeutic properties.


Thus, in a first aspect the present invention relates to a 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of inorganic acids, sulphonic acids and organic acids.


In a preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt is selected from the group consisting of hydrochloric acid and hydrobromic acid.


In another preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt is selected from the group consisting of maleic acid, fumaric acid, oxalic acid, malonic acid and succinic acid.


In a more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt is selected from the group consisting of hydrochloride, maleate, fumarate, malonate, succinate, oxalate and/or hydrobromide.


In a still more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt is selected from hydrochloride and/or maleate.


A further aspect of the present invention includes pharmaceutical compositions comprising a 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt and at least a pharmaceutically acceptable carrier, adjuvant or vehicle.


In a further aspect the invention is directed to 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt for use as medicament, preferably as sigma ligand, i.e., for use in the treatment and/or prophylaxis of a sigma receptor mediated disease or condition.


Another aspect of this invention relates to a method of treating and/or preventing a sigma receptor mediated disease which method comprises administering to a patient in need of such a treatment a therapeutically effective amount of a compound as above defined or a pharmaceutical composition thereof.


These aspects and preferred embodiments thereof are additionally also defined in the claims.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1: X-Ray powder diffraction of example 0.



FIG. 2: 1H nuclear magnetic resonance of example 0.



FIG. 3: X-Ray powder diffraction of example 1.



FIG. 4: 1H nuclear magnetic resonance of example 1.



FIG. 5: Differential Scanning calorimetry (DSC) of example 1.



FIG. 6: 1H nuclear magnetic resonance of example 2.



FIG. 7: X-Ray powder diffraction of example 2.



FIG. 8: Differential Scanning calorimetry (DSC) of example 2.



FIG. 9: 1H nuclear magnetic resonance of example 3.



FIG. 10: X-Ray powder diffraction of example 3.



FIG. 11: Differential Scanning calorimetry (DSC) of example 3.



FIG. 12: 1H nuclear magnetic resonance of example 4.



FIG. 13: X-Ray powder diffraction of example 4.



FIG. 14: Differential Scanning calorimetry (DSC) of example 4.



FIG. 15: 1H nuclear magnetic resonance of example 5.



FIG. 16: X-Ray powder diffraction of example 5.



FIG. 17: Differential Scanning calorimetry (DSC) of example 5.



FIG. 18: 1H nuclear magnetic resonance of example 6.



FIG. 19: X-Ray powder diffraction of example 6.



FIG. 20: Differential Scanning calorimetry (DSC) of example 6.



FIG. 21: 1H nuclear magnetic resonance of example 7.



FIG. 22: X-Ray powder diffraction of example 7.



FIG. 23: Differential Scanning calorimetry (DSC) of example 7.



FIG. 24: Plasma concentration after oral administration of example 1.



FIG. 25: Plasma concentration after oral administration of example 2.





DETAILED DESCRIPTION OF THE INVENTION

Compound 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone is difficult to crystallize. On trying in different conditions, an oil was obtained in all the cases, either using precipitation or evaporation from several solvents. This oil crystallizes with seeding very slowly, and only after several days it gives a crystalline form. The difficulty to crystallize comes from its low melting point (46° C.). Thus there is a need for alternative forms of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone with a melting point higher than 46° C. which have advantages, among other, of simplifying the procedures of isolation, purification and handling.


Indeed, after an extensive screening of salts, it has been observed that a large number of acids (e.g. sulphuric acid, benzenesulphonic acid, acetic acid or L-tartaric acid) did not afford a solid when mixing with 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone, but instead an oil was always obtained.


Further, among the acids suitable for obtaining a salt in solid form, it has been surprisingly found that the strong inorganic monoacids and the organic diacids were the ones that provided better results in terms of easiness of preparation, physical stability, scaling-up, solubility, etc. This is particularly true for hydrochloric acid and maleic acid. These results are shown through the increment achieved regarding the melting point and the values for some specific properties as thermodynamic solubility or pharmacokinetic parameters as Cmax or AUC in order to find new alternative forms having desirable properties for pharmaceutical use.


Thus, in one preferred aspect, the present invention is directed to a 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt.


In another preferred aspect, the present invention is directed to 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of inorganic acids, sulphonic acids and organic acids.


In another preferred aspect, the present invention is directed to 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloric acid and hydrobromic acid.


In another preferred aspect, the present invention is directed to 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of maleic acid, fumaric acid, oxalic acid, malonic acid and succinic acid.


In another preferred aspect, the present invention is directed to 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloride, maleate, fumarate, malonate, succinate, oxalate and/or hydrobromide.


In a still more preferred aspect, the present invention is directed to 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone hydrochloride and 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl) ethanone maleate.


As noted previously, it has been reported that 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone is a highly selective sigma-1 (σ1) receptor antagonist, displaying strong analgesic activity in the treatment and prevention of chronic and acute pain, and particularly, neuropathic pain (see WO 2011/147910).


It has now been found that 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salts are particularly suitable for use as medicament.


It has also been found that 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of inorganic acids, sulphonic acids and organic acids is particularly suitable for use as medicament.


It has also been found that 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloric acid and hydrobromic acid is particularly suitable for use as medicament.


It has also been found that 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of maleic acid, fumaric acid, oxalic acid, malonic acid and succinic acid is particularly suitable for use as medicament.


It has also been found that 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloride, maleate, fumarate, malonate, succinate, oxalate and/or hydrobromide is particularly suitable for use as medicament.


It has now been found that the hydrochloride salt of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone or the maleate salt of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone are particularly suitable for use as medicament.


The present invention therefore further provides medicaments or pharmaceutical compositions comprising a 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy) ethyl)piperazin-1-yl)ethanone crystalline salt together with at least a pharmaceutically acceptable carrier, adjuvant, or vehicle, for administration to a patient.


The present invention therefore further provides medicaments or pharmaceutical compositions comprising a 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy) ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of inorganic acids, sulphonic acids and organic acids together with at least a pharmaceutically acceptable carrier, adjuvant, or vehicle, for administration to a patient.


The present invention therefore further provides medicaments or pharmaceutical compositions comprising a 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy) ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloric acid and hydrobromic acid together with at least a pharmaceutically acceptable carrier, adjuvant, or vehicle, for administration to a patient.


The present invention therefore further provides medicaments or pharmaceutical compositions comprising a 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy) ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of maleic acid, fumaric acid, oxalic acid, malonic acid and succinic acid together with at least a pharmaceutically acceptable carrier, adjuvant, or vehicle, for administration to a patient.


The present invention therefore further provides medicaments or pharmaceutical compositions comprising a 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloride, maleate, fumarate, malonate, succinate, oxalate and/or hydrobromide together with at least a pharmaceutically acceptable carrier, adjuvant, or vehicle, for administration to a patient.


The present invention therefore further provides medicaments or pharmaceutical compositions comprising 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy) ethyl)piperazin-1-yl)ethanone hydrochloride or 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone maleate together with at least a pharmaceutically acceptable carrier, adjuvant, or vehicle, for administration to a patient.


Examples of pharmaceutical compositions include any solid (tablets, pills, capsules, granules etc.) or liquid (solutions, suspensions or emulsions) composition for oral, topical or parenteral administration.


In a preferred embodiment the pharmaceutical compositions are in oral form, either solid or liquid. Suitable dose forms for oral administration may be tablets, capsules, syrops or solutions and may contain conventional excipients known in the art such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, for example lactose, sugar, maize starch, calcium phosphate, sorbitol or glycine; tabletting lubricants, for example magnesium stearate; disintegrants, for example starch, polyvinylpyrrolidone, sodium starch glycollate or microcrystalline cellulose; or pharmaceutically acceptable wetting agents such as sodium lauryl sulfate.


The solid oral compositions may be prepared by conventional methods of blending, filling or tabletting. Repeated blending operations may be used to distribute the active agent throughout those compositions employing large quantities of fillers. Such operations are conventional in the art. The tablets may for example be prepared by wet or dry granulation and optionally coated according to methods well known in normal pharmaceutical practice, in particular with an enteric coating.


The pharmaceutical compositions may also be adapted for parenteral administration, such as sterile solutions, suspensions or lyophilized products in the appropriate unit dosage form. Adequate excipients can be used, such as bulking agents, buffering agents or surfactants.


Administration of the compounds or compositions of the present invention may be by any suitable method, such as intravenous infusion, oral preparations, and intraperitoneal and intravenous administration. Oral administration is preferred because of the convenience for the patient and the chronic character of the diseases to be treated.


The compounds and compositions of this invention may be used with other drugs to provide a combination therapy. The other drugs may form part of the same composition, or be provided as a separate composition for administration at the same time or at different time.


The auxiliary materials or additives of a pharmaceutical composition according to the present invention can be selected among carriers, excipients, support materials, lubricants, fillers, solvents, diluents, colorants, flavour conditioners such as sugars, antioxidants, binders, adhesives, disintegrants, anti-adherents, glidants and/or agglutinants. In the case of suppositories, this may imply waxes or fatty acid esters or preservatives, emulsifiers and/or carriers for parenteral application. The selection of these auxiliary materials and/or additives and the amounts to be used will depend on the form of application of the pharmaceutical composition.


The medicament or pharmaceutical composition according to the present invention may be in any form suitable for the application to humans and/or animals, preferably humans including infants, children and adults and can be produced by standard procedures known to those skilled in the art. Therefore, the formulation in accordance with the invention may be adapted for topical or systemic application, particularly for dermal, transdermal, subcutaneous, intramuscular, intra-articular, intraperito neal, intravenous, intra-arterial, intravesical, intraosseous, intracavernosal, pulmonary, buccal, sublingual, ocular, intravitreal, intranasal, percutaneous, rectal, vaginal, oral, epidural, intrathecal, intraventricular, intracerebral, intracerebroventricular, intra cisternal, intraspinal, perispinal, intracranial, delivery via needles or catheters with or without pump devices, or other application routes.


The mentioned formulations will be prepared using standard methods such as those described or referred to in the Spanish and US Pharmacopoeias and similar reference texts.


In one embodiment of the invention the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt is used in therapeutically effective amounts.


In another embodiment of the invention the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of inorganic acids, sulphonic acids and organic acids is used in therapeutically effective amounts.


In another embodiment of the invention the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloric acid and hydrobromic acid is used in therapeutically effective amounts.


In another embodiment of the invention the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of maleic acid, fumaric acid, oxalic acid, malonic acid and succinic acid is used in therapeutically effective amounts.


In a preferred embodiment of the invention the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloride, maleate, fumarate, malonate, succinate, oxalate and/or hydrobromide is used in therapeutically effective amounts.


In still more preferred embodiment of the invention the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone hydrochloride or the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone maleate are used in therapeutically effective amounts.


Generally an effective administered amount of a compound of the invention will depend on the relative efficacy of the compound chosen, the severity of the disorder being treated and the weight of the sufferer. The physician will determine the dosage of the present therapeutic agents which will be most suitable and it will vary with the form of administration and the particular compound chosen, and furthermore, it will vary with the patient under treatment, the age of the patient, the type of disease or condition being treated. When the composition is administered orally, larger quantities of the active agent will be required to produce the same effect as a smaller quantity given parenterally. The active compound will typically be administered once or more times a day for example 1, 2, 3 or 4 times daily, with typical total daily doses in the range of from 0.1 to 1000 mg/kg/day.


Particularly, 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salts are useful for the treatment and/or prophylaxis of a sigma receptor mediated disease or condition.


In a preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salts are used in the manufacture of a medicament for the treatment and/or prophylaxis of a disease selected from the group consisting of diarrhoea; lipoprotein disorders; migraine; obesity; arthritis; hypertension; arrhythmia; ulcer; learning, memory and attention deficits; cognition disorders; neurodegenerative diseases; demyelinating diseases; addiction to drugs and chemical substances including cocaine, amphetamine, ethanol and nicotine; tardive diskinesia; ischemic stroke; epilepsy; stroke; stress; cancer; psychotic conditions, in particular depression, anxiety or schizophrenia; inflammation; or autoimmune diseases.


In a still more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethylpiperazin-1-yl)ethanone crystalline salts are used in the manufacture of a medicament for the treatment and/or prophylaxis of pain, preferably neuropathic pain, inflammatory pain or other pain conditions involving allodynia and/or hyperalgesia.


Particularly, 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of inorganic acids, sulphonic acids and organic acids is useful for the treatment and/or prophylaxis of a sigma receptor mediated disease or condition.


In a more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of inorganic acids, sulphonic acids and organic acids is used in the manufacture of a medicament for the treatment and/or prophylaxis of a disease selected from the group consisting of diarrhoea; lipoprotein disorders; migraine; obesity; arthritis; hypertension; arrhythmia; ulcer; learning, memory and attention deficits; cognition disorders; neurodegenerative diseases; demyelinating diseases; addiction to drugs and chemical substances including cocaine, amphetamine, ethanol and nicotine; tardive diskinesia; ischemic stroke; epilepsy; stroke; stress; cancer; psychotic conditions, in particular depression, anxiety or schizophrenia; inflammation; or autoimmune diseases.


In a still more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethylpiperazin-1-yl)ethanone crystalline salt selected from the group consisting of inorganic acids, sulphonic acids and organic acids is used in the manufacture of a medicament for the treatment and/or prophylaxis of pain, preferably neuropathic pain, inflammatory pain or other pain conditions involving allodynia and/or hyperalgesia.


Particularly, 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of is selected from the group consisting of hydrochloric acid and hydrobromic acid is useful for the treatment and/or prophylaxis of a sigma receptor mediated disease or condition.


In a more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt is selected from the group consisting of hydrochloric acid and hydrobromic acid is used in the manufacture of a medicament for the treatment and/or prophylaxis of a disease selected from the group consisting of diarrhoea; lipoprotein disorders; migraine; obesity; arthritis; hypertension; arrhythmia; ulcer; learning, memory and attention deficits; cognition disorders; neurodegenerative diseases; demyelinating diseases; addiction to drugs and chemical substances including cocaine, amphetamine, ethanol and nicotine; tardive diskinesia; ischemic stroke; epilepsy; stroke; stress; cancer; psychotic conditions, in particular depression, anxiety or schizophrenia; inflammation; or autoimmune diseases.


In a still more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt is selected from the group consisting of hydrochloric acid and hydrobromic acid is used in the manufacture of a medicament for the treatment and/or prophylaxis of pain, preferably neuropathic pain, inflammatory pain or other pain conditions involving allodynia and/or hyperalgesia.


Particularly, 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of selected from the group consisting of maleic acid, fumaric acid, oxalic acid, malonic acid and succinic acid is useful for the treatment and/or prophylaxis of a sigma receptor mediated disease or condition.


In a more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of maleic acid, fumaric acid, oxalic acid, malonic acid and succinic acid is used in the manufacture of a medicament for the treatment and/or prophylaxis of a disease selected from the group consisting of diarrhoea; lipoprotein disorders; migraine; obesity; arthritis; hypertension; arrhythmia; ulcer; learning, memory and attention deficits; cognition disorders; neurodegenerative diseases; demyelinating diseases; addiction to drugs and chemical substances including cocaine, amphetamine, ethanol and nicotine; tardive diskinesia; ischemic stroke; epilepsy; stroke; stress; cancer; psychotic conditions, in particular depression, anxiety or schizophrenia; inflammation; or autoimmune diseases.


In a still more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of maleic acid, fumaric acid, oxalic acid, malonic acid and succinic acid is used in the manufacture of a medicament for the treatment and/or prophylaxis of pain, preferably neuropathic pain, inflammatory pain or other pain conditions involving allodynia and/or hyperalgesia.


Particularly, 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloride, maleate, fumarate, malonate, succinate, oxalate and/or hydrobromide is useful for the treatment and/or prophylaxis of a sigma receptor mediated disease or condition.


In a more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloride, maleate, fumarate, malonate, succinate, oxalate and/or hydrobromide is used in the manufacture of a medicament for the treatment and/or prophylaxis of a disease selected from the group consisting of diarrhoea; lipoprotein disorders; migraine; obesity; arthritis; hypertension; arrhythmia; ulcer; learning, memory and attention deficits; cognition disorders; neurodegenerative diseases; demyelinating diseases; addiction to drugs and chemical substances including cocaine, amphetamine, ethanol and nicotine; tardive diskinesia; ischemic stroke; epilepsy; stroke; stress; cancer; psychotic conditions, in particular depression, anxiety or schizophrenia; inflammation; or autoimmune diseases.


In a still more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone crystalline salt selected from the group consisting of hydrochloride, maleate, fumarate, malonate, succinate, oxalate and/or hydrobromide is used in the manufacture of a medicament for the treatment and/or prophylaxis of pain, preferably neuropathic pain, inflammatory pain or other pain conditions involving allodynia and/or hyperalgesia.


More particularly, 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone hydrochloride or 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone maleate are useful for the treatment and/or prophylaxis of a sigma receptor mediated disease or condition.


In a more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone hydrochloride or the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone maleate are used in the manufacture of a medicament for the treatment and/or prophylaxis of a disease selected from the group consisting of diarrhoea; lipoprotein disorders; migraine; obesity; arthritis; hypertension; arrhythmia; ulcer; learning, memory and attention deficits; cognition disorders; neurodegenerative diseases; demyelinating diseases; addiction to drugs and chemical substances including cocaine, amphetamine, ethanol and nicotine; tardive diskinesia; ischemic stroke; epilepsy; stroke; stress; cancer; psychotic conditions, in particular depression, anxiety or schizophrenia; inflammation; or autoimmune diseases.


In a still more preferred embodiment the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone hydrochloride or the 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone maleate are used in the manufacture of a medicament for the treatment and/or prophylaxis of pain, preferably neuropathic pain, inflammatory pain or other pain conditions involving allodynia and/or hyperalgesia.


The following examples are merely illustrative of certain embodiments of the invention and cannot be considered as restricting it in any way.


EXAMPLES

Analytical Techniques


The following techniques have been used in this invention for identifying either 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl) ethanone or its different salts obtained:

    • Proton Nuclear Magnetic Resonance (1H-NMR)
    • Proton nuclear magnetic resonance analyses were recorded in deuterated methanol (CD3OD) in a Varian Mercury 400 spectrometer, equipped with a broadband probe ATB 1H/19F/X of 5 mm. Spectra were acquired dissolving 5-10 mg of sample in 0.7 mL of deuterated solvent.
    • X-Ray Powder Diffraction (XRPD) characterization
    • XRPD analysis was performed using a Philips X'Pert diffractometer with Cu Kα radiation in Bragg-Brentano geometry. The system is equipped with a monodimensional, real time multiple strip detector. Diffractograms were recorded from 3° to 40° (2θ) at a scan rate of 17.6° per minute.


Differential Scanning calorimetry analysis (DSC)


DSC analyses were recorded in a Mettler Toledo DSC822e. Samples of 1-2 mg were weighted into 40 μl aluminum crucibles with a pinhole lid, and were heated, under nitrogen (50 mL/min), from 30 to 300° C. at a heating rate of 10° C./min. Data collection and evaluation were done with software STARe.


Example 0
Characterization of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl) methoxy)ethyl)piperazin-1-yl)ethanone

Example 0 can be prepared as disclosed in the previous patent application WO 2011/147910 and was characterized by X-Ray powder diffraction (FIG. 1) and by 1H nuclear magnetic resonance (FIG. 2).


Alternatively, example 0 can be obtained as follows:




embedded image


To a suspension of 1-(4-(2-hydroxyethyl)piperazin-1-yl)ethanone (1) (16.98 g, 81.3 mmol) in tetrahydrofuran (150 mL) at 0° C., NaH (60% mineral oil, 8.13 g, 203.4 mmol) was added turing 15 min. The mixture was stirred for 10 min and (1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methyl 4-methylbenzenesulfonate (33) (24.70 g, 67.8 mmol) in tetrahydrofuran (200 mL) was added during 25 min. The suspension was allowed to reach room temperature and stirred for 19 h. The mixture was cooled to 0° C. and H2O (15 mL) was slowly added. The suspension was allowed to reach room temperature and stirred for 10 min. Ethyl acetate (350 mL) and saturated aqueous NH4Cl solution (300 mL), were added to the mixture and the phases separated. The aqueous phase was extracted with ethyl acetate (1×250 mL), and the combined organic phases were washed with saturated aqueous NaHCO3 solution (2×400 mL), dried over anhydrous Na2SO4 and filtered. The solvent was removed and the crude orange oil thus obtained was purified by chromatography over silica-gel (CH2Cl2/MeOH/NH4OH 98:2:1-95:5:1), to give the title compound (LB19) as an orange solid (20.50 g, 83% yield).


General Method for Obtaining Salts of Example 0


Previously a solubility study was performed with compound Example 0. The results are shown in Table 1 wherein number of volumes needed to dissolve the solid in the corresponding solvent at room temperature is shown. If at 50 volumes, dissolution was not observed at room temperature, mixture was heated to reflux temperature.









TABLE 1







Example 0 compound solubility










Product
Example 0







H2O
 50[1]



CH3CN
15 



EtOH
4



MIBK
17 



THF
4



CH2Cl2
3








[1]No dissolution was observed at room temperature and included when mixture was heated to reflux temperature.







The acids used to investigate the crystalline salts of example 0 were selected according to the following criteria:

    • Acids with enough acidity to protonate the example 0
    • Acids that are pharmaceutically acceptable compounds


The selection of the acids was then carried out starting from the list in P. H. Stahl, C. G. Wemuth, Handbook of Pharmaceutical Salts: Properties, Selection and Use 2002. Acids having a pKa low enough to form a salt with example 0 (pKa(base)−pKa(acid)>3) were chosen and are shown in table 2













TABLE 2





Acid
Purity (%)
PKa1
pKa2
pKa3



















Phosphoric acid

2.15
7.20
12.35 


Maleic acid
  99.3
1.92
6.23



Benzensulphonic acid

0.7




Sulfuric acid

Strong
1.99



Acetic acid

4.75




Propanoic acid

4.87




Methanesulfonic acid

−1.2




Ethanesulfonic acid

2.05




Benzoic acid

4.2




Cinnamic acid

4.44




Nicotinic acid

4.85




Salicylic acid

2.97




Capric acid

4.9




Caproic acid

4.88




Caprylic acid

4.89




Citric acid

3.13
4.76
6.40


Fumaric acid
100
3.03
4.38



Malonic acid
100
2.83
5.70



Oxalic acid
100
1.25
4.27



Succinic acid
100
4.21
5.64



L-(+)-Tartaric acid

3.02
4.36



Hydrobromic acid
100
Strong




Nitric acid

Strong




Hydrochloric acid
  99.4
Strong











Although several of the acids selected have two or even three (citric acid) acidic positions, in principle, only sulfuric acid has a second proton acidic enough to form the disalt with example 0. In total there are twenty four different salts that could be formed.


Experimental Part:


General Experimental Conditions:


Wet Grinding Experiments:


General procedure: In a microtube of 2 mL, Example 0 and 1 eq. of the corresponding acid were added. One drop of solvent and two steel balls were added to each tube and the resulting mixture was grinded in a ball mill (15 min, 30 Hz, three times) and dried. In case of liquid acids, an ethyl acetate solution was previously prepared and the necessary volume for 1 eq of acid was added to the microtube.


Results obtained are shown in Table 3












TABLE 3





Acid
Solvent
Observation
XRPD Result







Benzenesulfonic acid
H2O
Yellow oil
Amorphous


Benzenesulfonic acid
CH3CN
Yellow oil
Amorphous


Benzenesulfonic acid
THF
Yellow oil
Amorphous


Benzenesulfonic acid
EtOH
Yellow oil
Amorphous


Benzenesulfonic acid
Heptane
Pastous solid
Amorphous


Benzenesulfonic acid
Et2O
Pastous solid
Amorphous


Benzenesulfonic acid
Cyclohexane
Pastous solid
Amorphous


Benzensulphonic acid
EtOH—Et2O
Pastous solid
Amorphous


Benzensulphonic acid
CH2Cl2
Pastous solid
Amorphous


Benzensulphonic acid
MIBK
Pastous solid
Amorphous


Benzensulphonic acid
Heptane
Pastous solid
Amorphous


Benzensulphonic acid
Toluene
Pastous solid
Amorphous


Maleic acid
MIBK
Yellow oil
Amorphous


Maleic acid
CH2Cl2
Yellow oil
Amorphous


Maleic acid
EtOH
Off White pastous solid
Example 2


Maleic acid
Cyclohexane
White solid
Example 2


Maleic acid
Heptane
White solid
Example 2


Maleic acid
Et2O
White solid
Example 2


Maleic acid
MTBE
White solid
Example 2


Maleic acid
MTBE
White solid
Example 2


Maleic acid
MTBE
White solid
Example 2


Maleic acid
H2O
no crystallisation



Maleic acid
CH3CN—Et2O
Pastous solid
Amorphous


Maleic acid
THF
White solid
Example 2


Fumaric acid
THF
White solid
Example 3


Fumaric acid
IPA
White solid
Example 3


Fumaric acid
EtOH
Yellow oil



Fumaric acid
IPA
White solid
Example 3


Fumaric acid
IPA
Off white solid
Example 3


Malonic acid
MIBK
White solid
Example 4


Malonic acid
MTBE
White solid
Example 4 (low





crystallinity)


Malonic acid
Et2O
White solid
Example 4 (low





crystallinity)


Malonic acid
Ether
White solid
Example 4


Malonic acid
MIBK
White solid
Example 4


Malonic acid
EtOH
Yellow oil



Malonic acid
CHCl3
Yellow paste



Capric acid
H2O
Yellowish oil



Capric acid
CH3CN
Yellowish oil



Capric acid
CHCl3
Yellowish oil



Capric acid
AcOEt
Yellowish oil



Capric acid
MTBE
brown paste



Capric acid
Toluene
brown oil



Capric acid
CH3CN
Off white pastous solid
Amorphous


Capric acid
EtOH
Yellow oil



Capric acid
H2O/AcOiBu
Yellow oil



Capric acid
CH3CN/MIBK
Yellow oil



Capric acid
CH2Cl2
Yellow oil



Sulphuric acid
H2O
oil



Sulphuric acid
CH3CN
oil



Sulphuric acid
IPA
oil



Sulphuric acid
H2O
oil



Sulphuric acid
Heptane
oil
Amorphous


Sulphuric acid
Acetone
oil
Amorphous


Succinic acid
MIBK
White solid
Example 6


Succinic acid
MIBK
White solid
Example 6


Succinic acid
MTBE
Off white solid
Example 6


Succinic acid
MIBK
White solid
Example 6


Succinic acid
H2O
Paste
Example 6 (low





crystallinity)


Succinic acid
CH3CN
White solid
Example 6


Succinic acid
MTBE
White solid
Example 6


Succinic acid
AcOEt
White solid
Example 6


Succinic acid
EtOH
Off white pastous solid
Amorphous


Succinic acid
EtOH
Yellow oil



Succinic acid
MIBK
White solid
Example 6


Caprylic acid
CH2Cl2
brown oil



Caprylic acid
Et2O
brown oil



Caprylic acid
AcOEt
Yellow oil



Caprylic acid
Heptane
Colorless solution



Caprylic acid
AcOEt
Yellow oil



Caproic acid
AcOEt
Yellow oil



Caproic acid
Toluene
brown oil



Caproic acid
/Heptane
Colorless solution



Caproic acid
AcOEt
Yellow oil



Caproic acid
Et2O
brown oil



Caproic acid
CH2Cl2
brown oil



Propionic acid
CH2Cl2
brown oil



Propionic acid
Et2O
brown oil



Propionic acid
AcOEt
Yellow oil



Propionic acid
Toluene
brown oil



Propionic acid
AcOEt
Yellow oil



Propionic acid
Heptane
Brown oil



Phosphoric acid
Dioxane
oil
Amorphous


Phosphoric acid
EtOH
oil
Amorphous


Methanesulfonic acid
AcOEt
Yellow oil



Methanesulfonic acid
CH2Cl2
brown oil



Methanesulfonic acid
Toluene
Greenish paste
Amorphous


Methanesulfonic acid
Et2O
brown oil



Methanesulfonic acid
AcOEt
Yellow oil



Methanesulfonic acid
Toluene
yellow oil



Methanesulfonic acid
Heptane
Brown oil



Methanesulfonic acid
Cyclohexane
Yellow oil



Ethanesulfonic acid
Cyclohexane
Yellow oil



Ethanesulfonic acid
Heptane
Brown oil



Ethanesulfonic acid
Et2O
brown oil



Ethanesulfonic acid
CH2Cl2
brown oil



Ethanesulfonic acid
Toluene
Greenish paste
Amorphous


Ethanesulfonic acid
AcOEt
Yellow oil



Ethanesulfonic acid
Toluene
yellow oil



Ethanesulfonic acid
AcOEt
Yellow oil



Salicylic acid
H2O
Yellowish oil



Salicylic acid
AcOEt
Yellowish oil



Salicylic acid
CH2Cl2
Yellowish oil



Salicylic acid
CH2Cl2
Yellow oil



Salicylic acid
Toluene
Reddish oil



Salicylic acid
CH3CN
Yellow oil



Salicylic acid
MTBE
Off white solid
Amorphous


Salicylic acid
AcOEt
Yellow oil



Salicylic acid
CH2Cl2
Reddish oil



Salicylic acid
Toluene
Brown oil



Salicylic acid
EtOH
Yellow oil



Salicylic acid
toluene/cyclohexane
Yellow paste
Amorphous


Salicylic acid
MTBE/cyclohexane
Yellow paste
Salicylic acid


Salicylic acid
Cyclohexane
Yellowish paste
Amorphous


Salicylic acid
Cyclohexane
Off white pastous solid
Amorphous


Salicylic acid
CH3CN
Yellowish oil



Salicylic acid
MTBE
Yellowish oil



Nicotinic acid
H2O
White paste
Nicotinic acid


Nicotinic acid
CH3CN
Yellowish oil



Nicotinic acid
MTBE
Yellowish oil



Nicotinic acid
AcOEt
Yellowish oil



Nicotinic acid
CH2Cl2
Yellowish oil



Nicotinic acid
MTBE
Off white pastous solid
Nicotinic acid


Nicotinic acid
CH2Cl2
White solid
Nicotinic acid


Nicotinic acid
IPA
Off white solid
Nicotinic acid


Nicotinic acid
CH3CN
Off white solid
Nicotinic acid


Nicotinic acid
MTBE
Off white solid
Nicotinic acid


Nicotinic acid
AcOEt
Off white solid
Nicotinic acid


Nicotinic acid
CH2Cl2
Off white solid
Nicotinic acid


Nicotinic acid
MTBE
White solid
Nicotinic acid


Nicotinic acid
EtOH
Yellow solid
Amorphous +





nicotinic acid


Nicotinic acid
CH3CN
White solid
Nicotinic acid


Nicotinic acid
IPA
White solid
Nicotinic acid


Citric acid
Toluene
Yellow oil



Citric acid
CH3CN
Yellow oil



Citric acid
MTBE
Off white solid
Amorphous


Citric acid
AcOEt
Yellow oil



Citric acid
CH2Cl2
Yellow oil



Citric acid
CH3CN
Brown oil



Citric acid
MIBK
Off white solid
Citric acid


Citric acid
EtOH
Yellow oil



Citric acid
Et2O
Yellowish paste
Amorphous


Citric acid
AcOEt
Yellow solidl
Citric acid


Citric acid
CH3CN/cyclohexane
Off white paste
Amorphous


Citric acid
H2O
Yellowish oil



Citric acid
CH3CN
Yellowish oil



Citric acid
MTBE
Yellowish oil



Citric acid
AcOEt
Yellowish oil



Citric acid
CH2Cl2
Yellowish oil



Citric acid
CH2Cl2
Yellow oil



Citric acid
Et2O
Off white pastous solid
Amorphous


Benzoic acid
MTBE
Off white pastous solid
Amorphous


Benzoic acid
H2O/AcOiBu
Yellow oil



Benzoic acid
CH3CN/MIBK
Yellow oil



Benzoic acid
CHCl3/Toluene
Off white solid
Example 1


Benzoic acid
AcOEt/Heptane
Yellow paste/oil
Amorphous


Benzoic acid
Et2O
Yellow paste/oil



Benzoic acid
CH2Cl2
Yellow oil



Benzoic acid
EtOH
Brown oil



Benzoic acid
MTBE
brown oil



Benzoic acid
H2O
Yellowish oil



Benzoic acid
CH3CN
Yellowish oil



Benzoic acid
CHCl3
Yellowish oil



Benzoic acid
AcOEt
Yellowish oil



Cinnamic acid
MTBE
Off white pastous solid
Amorphous


Cinnamic acid
MTBE
brown oil



Cinnamic acid
CH2Cl2
Brown oil



Cinnamic acid
H2O
Yellowish oil



Cinnamic acid
CH3CN
Yellowish oil



Cinnamic acid
CHCl3
Yellowish oil



Cinnamic acid
AcOEt
Yellowish oil



Cinnamic acid
H2O/AcOiBu
Yellow oil



Cinnamic acid
CH3CN/MIBK
Brown oil



Cinnamic acid
AcOEt/Heptane
Yellow paste/oil
Amorphous


Cinnamic acid
Et2O
Yellow paste/oil



Cinnamic acid
EtOH
Yellow oil



Oxalic acid
MTBE
White solid
Example 5


Oxalic acid
AcOiBu
White solid
Example 5


Oxalic acid
EtOH
White solid
Example 5


Oxalic acid
IPA
White solid
Example 5


Oxalic acid
MIBK
White solid
Example 5


Oxalic acid
AcOEt
White solid
Example 5


Oxalic acid
AcOEt
White solid
Example 5


Oxalic acid
AcOEt
White solid
Example 5


Oxalic acid
MIBK
Off white solid
Example 5


Oxalic acid
MIBK
Off white solid
Example 5


Oxalic acid
MIBK
Off white solid
Example 5


Hydrobromic acid
Cyclohexane
Yellow oil



Hydrobromic acid
IPA
Brown paste
Amorphous


Hydrobromic acid
CH3CN
Brown paste
Amorphous


Hydrobromic acid
Acetone
Brown paste
Amorphous


Hydrobromic acid
IPA
Brown paste
Amorphous


Hydrobromic acid
CH3CN
Brown paste
Amorphous


Hydrobromic acid
Acetone
Brown paste
Amorphous


Hydrobromic acid
Toluene
No evolution



Hydrobromic acid
MIBK/IPA
White solid
Example 7


Hydrobromic acid
THF/IPA
White solid
Example 7


Hydrobromic acid
MTBE/IPA
White solid
Example 7


Hydrobromic acid
Toluene/IPA
White solid
Example 7


Hydrobromic acid
MIBK/IPA
White solid
Example 7


Hydrobromic acid
IPA
Yellow liquid



Hydrobromic acid
CH3CN
Yellow oil



Hydrobromic acid
Acetone
Yellow oil



Hydrobromic acid
THF
brown oil



Tartaric acid
AcOiBu
Off white solid
Tartaric acid


Tartaric acid
MIBK
Off white solid
Tartaric acid


Tartaric acid
Et2O
Off white solid
Tartaric acid


Tartaric acid
Toluene
Off white solid
Tartaric acid


Tartaric acid
MIBK
Off white solid
Tartaric acid


Tartaric acid
CHCl3
Yellow oil



Tartaric acid
CH3CN
Yellow oil



Tartaric acid
THF
Yellow oil



Tartaric acid
Dioxane
Off white solid
Amorphous


Tartaric acid
H2O
yellow oil



Tartaric acid
Acetone
Yellow oil



Tartaric acid
CH3CN
Oil + solvent



Tartaric acid
EtOH
Yellow oil



Tartaric acid
H2O
Off white pastous solid
Amorphous


Tartaric acid
CH3CN
Off white pastous solid
Amorphous


Tartaric acid
THF
Off white pastous solid
Amorphous


Tartaric acid
MTBE
Off white pastous solid
Amorphous


Tartaric acid
EtOH
Off white pastous solid
Amorphous


Tartaric acid
MIBK
Yellowish paste
Amorphous


Nitric acid
Toluene
No evolution



Nitric acid
MTBE
No evolution



Nitric acid
AcOEt
No evolution



Nitric acid
Cyclohexane
Yellow oil



Nitric acid
IPA
Yellow liquid



Nitric acid
CH3CN
Yellow oil



Nitric acid
Acetone
Yellow oil



Nitric acid
THF
yellow oil



Acetic acid
AcOEt
Yellow oil



Acetic acid
Et2O
brown oil



Acetic acid
AcOEt
Yellow oil



Acetic acid
CH2Cl2
brown oil



Acetic acid
Toluene
brown oil



Acetic acid
MTBE
No evolution






Where MIBK stands for methyl isobutyl ketone, MTBE stands for methyl tert-butylether, IPA stands for isopropanol and THF stands for tetrahydrofuran.






From the above experiments and the corresponding DSC data, as shown in Table 4, it can be concluded that increasing melting point regarding 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone (example 0) is achieved in order to find new alternative forms having desirable properties for pharmaceutical use.












TABLE 4





Example


MP (° C.)


no
Acid Structure
Acid Name
DSC







1
HCl
Hydrochloric
155-157




acid


2
Cis-HOOC—CH═CH—COOH
Maleic acid
160-162


3
Trans-HOOC—CH═CH—COOH
Fumaric acid
132-133


4
HOOC—CH2—COOH
Malonic acid
101-103


5
HOOC—COOH
Oxalic acid
160-162


6
HOOC—CH2—CH2—COOH
Succinic acid
102-104


7
HBr
Hydrobromic
170-171




acid





Example 0 MP: 46° C.






The above mentioned examples 1 to 7 can be specifically obtained according to the following procedures:


Example 1
Synthesis of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}methyl)piperazine hydrochloride



embedded image


To a solution of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyhpiperazine (57.41 g, 157.55 mmol) in ethyl acetate (900 mL), HCI.Et2O (2.0 M, 86.7 mL, 173.30 mmol) was added and the mixture was stirred at room temperature for 2 h. The mixture was evaporated to dryness, ethyl ether (300 mL) was added and evaporated again. This process was repeated two times with CH2Cl2 and ethyl ether. The solid thus obtained was triturated with hexane (400 mL) and filtered, washed with hexane (200 mL) and with ethyl ether/hexane (1:1, 100 mL). The solid was dried to give the title compound (61.2 g, 97% yield).


RMN-1H (CD3OD, 400 MHz, □): 8.24 (d, J=2.7 Hz, 1H, ArH); 7.76 (ddd, J=11.7, 7.0, 2.7 Hz, 1H, ArH); 7.61-7.55 (m, 1H, ArH); 7.47-7.37(m, 1H, ArH); 6.58 (d, J=2.5 Hz, 1H, ArH); 4.71 (s, 2H, CH2); 4.59 (sa, 1H, CH2); 4.20-4.05 (m, 1H, CH2); 3.96-3.85 (m, 2H, CH2); 3.69-3.39 (m, 4H, CH2); 3.24-2.99 (m, 2H, CH2); 2.14 (s, 3H, CH3). (FIG. 4)


EM-ESI+m/z: 365 (M+1-HCl).


Example 1 was additionally characterized by X-Ray powder diffraction (FIG. 3) and by DSC (FIG. 5).


Example 2
Synthesis of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}methyl)piperazine maleate



embedded image


To a 2 mL Eppendorf tube containing 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (19 mg, 0.052 mmol) and maleic acid (6 mg, 0.052 mmol), 1 drop of ethanol and two stainless steel grinding balls were added before milling for 45 minutes at a rate of 30 Hz (3×15 minutes) with a Retsch Ball Mill MM400. After drying under vacuum at room temperature the title compound was obtained as an off white pasty solid to which ethyl ether (0.2 mL) was added before stirring at room temperature for 2 h. The resulting mixture was isolated by centrifugation (RT, 14000 rpm, 10 min). After drying under vacuum at room temperature the title compound was obtained as a solid with a good crystallinity (17 mg, 82% yield).


RMN-1H (CD3OD, 400 MHz, □): 8.23 (d, J=2.7 Hz, 1H, ArH); 7.76 (ddd, J=11.1, 7.0, 2.7 Hz, 1H, ArH); 7.62-7.55 (m, 1H, ArH); 7.46-7.36 (m, 1H, ArH); 6.57 (d, J=2.7 Hz, 1H, ArH); 6.26 (s, 2H, CH=); 4.69 (s, 2H, CH2); 3.92-3.84 (m, 2H, CH2); 3.84-3.70 (m, 4H, CH2); 3.39-3.15 (m, 6H, CH2); 2.13 (s, 3H, CH3). (FIG. 6)


Example 2 was additionally characterized characterized by X-Ray powder diffraction (FIG. 7) and by DSC (FIG. 8).


Alternatively, the compound of Example 2 can be prepared using the following procedure:


To an assay tube equipped with magnetic stirrer containing a turbid solution of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (109.1 mg, 0.2994 mmol) in tert-butyl methyl ether (1.1 mL) at 45° C., maleic acid (35.2 mg, 0.303 mmol) was added. The resulting paste was vigorously stirred 1 h at 45° C. affording a suspension of a crystalline solid. Then the resultant suspension was cooled down to room temperature and stirred for 2 h. The solid was filtered with a sintered funnel (porosity 3) and washed with tert-butyl methyl ether (1×2 vol.). After drying under vacuum at room temperature the title compound was obtained as a crystalline solid (109 mg, 76% yield).


Example 3
Synthesis of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}methyl)piperazine fumarate



embedded image


To an assay tube equipped with magnetic stirrer containing 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (35 mg, 0.1 mmol) and fumaric acid (11.7 mg, 0.1 mmol, 1 eq.), isopropanol (0.35 mL) was added at room temperature. After 3 h of stirring at room temperature, the resultant suspension was filtered and washed with isopropanol (1×1.5 vol.). After drying under vacuum at room temperature, the title compound was obtained as a crystalline solid (33 mg, 68% yield).


RMN-1H (CD3OD, 400 MHz, □): 8.21 (d, J=2.7 Hz, 1H, ArH); 7.75 (ddd, J=11.1, 7.0, 2.7 Hz, 1H, ArH); 7.61-7.54 (m, 1H, ArH); 7.45-7.35 (m, 1H, ArH); 6.73 (s, 2 H, CH═); 6.55 (d, J=2.7 Hz, 1H, ArH); 4.64 (s, 2H, CH2); 3.81-3.74 (m, 2H, CH2); 3.73-3.61 (m, 4H, CH2); 3.01-2.94 (m, 2H, CH2); 2.94-2.87 (m, 2H, CH2); 2.87-2.79 (m, 2H, CH2); 2.11 (s, 3H, CH3). (FIG. 9)


Example 3 was additionally characterized characterized by X-Ray powder diffraction (FIG. 10) and by DSC (FIG. 11).


Example 4
Synthesis of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}methyl)piperazine malonate



embedded image


To an assay tube equipped with magnetic stirrer containing 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (35 mg, 0.1 mmol) and fumaric acid (10 mg, 0.1 mmol), methyl isobutyl ketone (0.3 mL) at room temperature was added. After 3 h the resultant suspension was filtered and washed with methyl isobutyl ketone (1×1.5 vol.). After drying under vacuum at room temperature, the title compound was obtained as a solid (35 mg, 75% yield).


RMN-1H (CDCl3, 400 MHz, □): 7.84 (d, J=2.7 Hz, 1H, ArH); 7.57 (ddd, J=11.1, 7.0, 2.7 Hz, 1H, ArH); 7.41-7.34 (m, 1H, ArH); 7.30-7.21 (m, 1H, ArH); 6.45 (d, J=2.7 Hz, 1H, ArH); 4.61 (s, 2H, CH2); 3.96-3.83 (m, 4H, CH2); 3.81-3.73 (m, 2H, CH2); 3.29-3.17 (m, 6H, CH2); 3.17-3.09 (m, 2H, CH2); 2.11 (s, 3H, CH3). (FIG. 12)


Example 4 was additionally characterized characterized by X-Ray powder diffraction (FIG. 13) and by DSC (FIG. 14).


Example 5
Synthesis of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}methyl)piperazine oxalate



embedded image


To a 2 mL Eppendorf tube containing 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (25 mg, 0.068 mmol) and oxalic acid (6.9 mg, 0.077 mmol), 1 drop of tert-butyl methyl ether and two stainless steel grinding balls were added before milling 45 minutes at a rate of 30 Hz (3×15 minutes) with a Retsch Ball Mill MM400. After drying under vacuum at room temperature, the title compound was obtained as a crystalline solid in a quantitative yield.


RMN-1H (CD3OD, 400 MHz, □): 8.22 (d, J=2.7 Hz, 1H, ArH); 7.76 (ddd, J=11.7, 7.0, 2.7 Hz, 1H, ArH); 7.61-7.55 (m, 1H, ArH); 7.45-7.36 (m, 1H, ArH); 6.57 (d, J=2.7 Hz, 1H, ArH); 4.67 (s, 2H, CH2); 3.92-3.85 (m, 2H, CH2); 3.82-3.75 (m, 4H, CH2); 3.38-3.29 (m, 4H, CH2); 3.29-3.21 (m, 2H, CH2); 2.13 (s, 3H, CH3). (FIG. 15)


Example 5 was additionally characterized characterized by X-Ray powder diffraction (FIG. 16) and by DSC (FIG. 17).


Alternatively, the compound of Example 5 can be prepared using the following procedure:


To an Eppendorf tube equipped with magnetic stirrer containing a solution of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (30 mg, 0.082 mmol) in ethyl acetate (0.3 mL), oxalic acid (7.5 mg, 0.083 mmol) was added at room temperature. After 3 h of stirring a precipitate was observed and the resultant suspension was centrifuged to isolate the solid. Ethyl acetate (0.2 mL) was added to the solid and centrifuged again. The recovered solid was dried under vacuum at room temperature to afford the title compound as a crystalline solid (37 mg, 99% yield).


Example 6
Synthesis of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}methyl)piperazine succinate



embedded image


To a 2 mL Eppendorf tube containing 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (24.6 mg, 0.067 mmol) and succinic acid (10 mg, 0.084 mmol), 1 drop of tert-butyl methyl ether and two stainless steel grinding balls were added before milling 45 minutes at a rate of 30 Hz (3×15 minutes) with a Retsch Ball Mill MM400. The resulting solid was dried under vacuum at room temperature to afford an amorphous solid (25 mg) to which isobutyl methyl ketone (0.2 mL) was added. The mixture was stirred at room temperature for 16 h and the resulting suspension was centrifuged. The solid thus obtained was dried at room temperature under vacuum to give the title compound as a solid (15 mg, 46% yield).


RMN-1H (CD3OD, 400 MHz, □): 8.20 (d, J=2.7 Hz, 1H, ArH); 7.75 (ddd, J=11.7, 7.0, 2.7 Hz, 1H, ArH); 7.60-7.54 (m, 1H, ArH); 7.44-7.35 (m, 1H, ArH); 6.55 (d, J=2.7 Hz, 1H, ArH); 4.61 (s, 2H, CH2); 3.72 (t, J=5.5, 2H, CH2); 3.67-3.53 (m, 4H, CH2); 2.84-2.73 (m, 2H, CH2); 2.73-2.65 (m, 2H, CH2); 2.65-2.58 (m, 2H, CH2); 2.56 (s, 4H); 2.0.9 (s, 3H, CH3). (FIG. 18).


Example 6 was additionally characterized characterized by X-Ray powder diffraction (FIG. 19) and by DSC (FIG. 20).


Alternatively, the compound of Example 6 can be prepared using the following procedure:


To an Eppendorf tube equipped with magnetic stirrer containing a solution of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (30 mg, 0.082 mmol) in ethyl acetate (0.3 mL), succinic acid (10 mg, 0.084 mmol) was added at room temperature. After one night of stirring at room temperature a precipitate was observed. The resultant suspension was centrifuged (25° C., 14000 rpm, 10 min) to isolate the solid. Tert-butyl methyl ether (0.2 mL) was added and the mixture centrifuged again (25° C., 14000 rpm, 10 min). The recovered solid was dried under vacuum at room temperature to afford the title compound as a solid (36 mg, 91% yield).


Example 7
Synthesis of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}methyl)piperazine hydrobromide



embedded image


To an Eppendorf tube equipped with magnetic stirrer containing a solution of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (15 mg, 0.041 mmol) in isobutyl methyl ketone (0.3 mL), a solution of HBr in IPA (50 μl of a solution prepared from 50 μl 48% HBr aqueous and 0.5 mL IPA, 0.04 mmol) was added at room temperature. After 2 h of stirring a precipitate was observed. The resultant suspension was centrifuged and the solid thus obtained was dried under vacuum at room temperature to afford the title compound as a crystalline solid (14 mg, 79% yield).


RMN-1H (CD3OD, 400 MHz, □): 8.24 (d, J=2.7 Hz, 1H, ArH); 7.76 (ddd, J=11.7, 7.0, 2.7 Hz, 1H, ArH); 7.62-7.55 (m, 1H, ArH); 7.47-7.37(m, 1H, ArH); 6.58 (d, J=2.5 Hz, 1H, ArH); 4.70 (s, 2H, CH2); 4.59 (sa, 1H, CH2); 4.11 (sa, 1H, CH2); 3.96-3.85 (m, 2H, CH2); 3.74-3.49 (m, 2H, CH2); 3.49-3.41 (m, 2H, CH2); 3.26-2.98 (m, 2H, CH2); 2.14 (s, 3H, CH3). (FIG. 21).


Example 7 was additionally characterized characterized by X-Ray powder diffraction (FIG. 22) and by DSC (FIG. 23).


Alternatively, the compound of Example 7 can be prepared using the following procedure:


To an assay tube equipped with magnetic stirrer containing a solution of 1-acetyl-4-({[1-(3,4-difluorophenyl)-1H-pyrazol-3-yl]methoxy}ethyl)piperazine (47 mg, 0.129 mmol) in isobutyl methyl ketone (0.7 mL), a solution of HBr in isopropanol (140 μl of a solution prepared from 150 μl 48% HBr aqueous and 1.5 mL isopropanol, 0.13 mmol) was added at room temperature. After 3 h of stirring precipitation was not observed. Therefore the solution was seeded with the previous compound and a precipitate appeared. After 2 h of stirring at room temperature, the resultant suspension was filtered and washed with isobutyl methyl ketone (0.5 mL). After drying under vacuum at room temperature, the title compound was obtained as a crystalline solid (37 mg, 65% yield).


Example 8
Thermodynamic Solubility

General protocol for thermodynamic solubility at pH 7.4 and pH 2 are described.


A) Thermodynamic Solubility at pH 7.4


Buffer Solution


Phosphate buffer at pH 7.4 (25 mM) was prepared as follows:


A solution 25 mM of Na2HPO4.12H2O (for 1 L of water, weight 8.96 g) was prepared


A solution 25 mM de KH2PO4 (for 1 L of water weight 3.4 g) was prepared.


Disodium phosphate solution (812 mL) and potassium phosphate (182 mL) solution were mixed and pH checked to be 7.4.


Equipment

    • Precision analytical balance Mettler Toledo AT20.
    • Analytical balance Mettler Toledo PJ300.
    • Liquid chromatograph Waters Alliance 2695.
    • Stirrer Thermomixer Control of Eppendorf a 25° C. y 1250 rpm
    • pHmeter with combined semi-micro electrode.


Procedure


Test Substance


Around 4 mg of compound in an HPLC vial (by duplicate) was dissolved in 1 mL of buffer solution. After stirring in Thermomixer Comfort system for 24 hours at 25° C., in order to achieve thermodynamic equilibrium, solution was centrifuged at 3000 rpm for 15 minutes.


The resulting upper layer was collected with a glass pipette and transferred to the HPLC vials in order to inject them (10 μL) directly to the HPLC instrument.


Standards


Standards were made in methanol to ensure overall compound solubility. Preparation of standard calibrators is illustrated below:


Sol.A: 4 mg in 1 mL methanol (4000 μg/mL)


Sol.B: 0.5 mL Sol.A to 5 mL with methanol (400 μg/mL)


Sol.C: 1 mL Sol.B to 10 mL with methanol (40 μg/mL)


Sol.D: 5 mL Sol.0 to 50 mL with methanol (4 μg/mL)


Sol.E: 4 mL Sol.D to 10 mL with methanol (1.6 μg/mL)


Calibration curve was created through 10 μL injection of standards, beginning with the more diluted standard. Blanks were also injected, for checking the absence of contamination.


10 μl of test substance were injected, by duplicate, and the average peak area interpolated in the calibration curve (see Tables Examples below).


Chromatographic Conditions

    • Column: XBridge C18 (or similar) 2.5 μm 4.6×50 mm
    • Temperature: 35° C.
    • Mobile phase: ACN/ammonium bicarbonate 10 mM.
    • Gradient: 0-3.5 min: from 15% CAN to 95% CAN
      • 3.5-5 min: 95% ACN
      • 5-6 min: 95 a 15% ACN
      • 6-8 min: 15% ACN
    • Flow: 1.5 mL/min
    • Detection: around the maximum UV wavelength.


B) Thermodynamic Solubility at pH 2


The same previous procedure was performed with HCl 0.01N, instead of buffer solution.


8.1 Thermodinamical Solubility for Example 1


According to the described protocol Example 1 was completely dissolved so the solubility was higher than 4000 μg/mL (pH=7.4). (See Table 5 and Table 6).









TABLE 5







SAMPLES


















Sample
Condition
Vial
RT
Date Acquired
Dilution
Inj.Vol.
Detection
Area
Height





















1
Example 1 PROB pH 7.4
pH 7.4
44
2.2
21/04/2012 6:01
1
10
PDA 290.0 nm
4351307
1096492



(1)


2
Example 1 PROB pH 7.4 (1)
pH 7.4
44
2.2
21/04/2012 6:10
1
10
PDA 290.0 nm
4230788
1078515


3
Example 1 PROB pH 7.4 (2)
pH 7.4
45
2.2
21/04/2012 6:19
1
10
PDA 290.0 nm
4757240
1203806


4
Example 1 PROB pH 7.4 (2)
pH 7.4
45
2.2
21/04/2012 6:28
1
10
PDA 290.0 nm
4804788
1214183






















TABLE 6







Conc.
Units
Res Id
Cal Id
SampleWeight





















1
3928
μg/ml
13370
13347
1


2
3817.1
μg/ml
13371
13347
1


3
4301.5
μg/ml
13372
13347
1


4
4345.3
μg/ml
13373
13347
1


Mean
4097.976


% RSD
6.5









8.2 Thermodinamical Solubility for Example 2


According to the described protocol Example 2 was completely dissolved so the solubility was higher than 4000 μg/mL (pH=7.4). (See Table 7 and Table 8).









TABLE 7







SAMPLES


















Sample
Condition
Vial
RT
Date Acquired
Dilution
Inj.Vol.
Detection
Area
Height





















1
Example 1 PROB pH 7.4 (1)
pH 7.4
11
2.8
15/04/2015 13:33
1
10
PDA 290.0 nm
4533277
1173198


2
Example 1 PROB pH 7.4 (1)
pH 7.4
11
2.8
15/04/2015 13:42
1
10
PDA 290.0 nm
4355437
1154617


3
Example 1 PROB pH 7.4 (2)
pH 7.4
12
2.8
15/04/2015 13:51
1
10
PDA 290.0 nm
4288239
1131879


4
Example 1 PROB pH 7.4 (2)
pH 7.4
12
2.8
15/04/2015 14:00
1
10
PDA 290.0 nm
4475398
1174356






















TABLE 8







Conc.
Units
Res Id
Cal Id
SampleWeight





















1
4593.8
μg/ml
2800
2784
1


2
4408.7
μg/ml
2801
2784
1


3
4338.8
μg/ml
2802
2784
1


4
4533.6
μg/ml
2803
2784
1


Mean
4468.706


% RSD
2.6









Example 9
Pharmacokinetic Parameters Cmax and AUC

The pharmacokinetics of Example 1 and 2 were tested using the following protocol:


Animals


Male Wistar rats weighing 250 to 300 g (±20) supplied by Harlan were used. Water and food was available ad libitum throughout the study.


Materials
















Supplier
Reference


















(Hydroxypropyl)methylcellulose
Sigma-Aldrich
H9262


Physiological serum Vitulia (isotonic)
ERN
999789.2


Fluorane - isoflurane
Abbot
880393H0


Ethanol
Sharlau
ET0010


Anhydrous dimethylsulphoxide (DMSO)
Carlo Erba
445131


Acetonitrile
Sigma-Aldrich
34967


Formic acid (98-100% purity)
Riedel de Haen
33015


Heparinized tubes (Microvette ®)
Sarstedt
CB300









Administration and Sample Collection


Two rats (R1 and R2) were used in the pharmacokinetic oral studies.


A single dose of tested compound was administered by oral gavage [10 mg/kg as free base (Example 0)] in 0.5% hydroxypropyl methylcellulose (1 mg/ml). From each rat, serial blood samples were collected at 15 and 30 min, 1, 2, 3, 5, 7 and 24 h.


Blood was collected from the saphenous veins into heparinized tubes. Plasma was obtained by blood centrifugation at 4° C. and 2280×g for 10 min and kept at −80° C. until analysis.


Sample Processing


Tested compound concentration in plasma samples was determined by least-squares linear regression using a ten-point calibration curve. The calibration curve was prepared in blank plasma from a working solution of 1 mg/ml in DMSO.


Samples were thawed at room temperature on the day of analysis. After plasma protein precipitation of samples and calibration standards with acetonitrile (1:4.3; v/v), the mixture was vortexed and centrifuged (4° C. and 16090×g for 10 min). Finally, an aliquot of the resultant supernatant was diluted 1/10 with water (0.1% formic acid) before analysis.


Analytical Method


Tested compound plasma concentrations were determined by high performance liquid chromatography-triple quadrupole mass spectrometry (HPLC-MS/MS) through the following method:


Column: Atlantis® T3 column (2.1×100 mm, 3 μm) (Waters).


Mobile phase: A: 0.0155% Formic water

    • B: 0.0155% Formic Acetonitrile


Autosampler wash: Solvent 1:Acetonitrile

    • Solvent 2: 5% Acetonitrile+95% Water


Pharmacokinetic Analysis


Standard pharmacokinetic parameters, such as area under the curve (AUC), peak plasma concentration (Cmax), time to peak concentration (tmax), oral bioavailability (F), total plasma clearance (Cl), volume of distribution at steady-state (Vss), mean residence time (MRT) and terminal half-life (t1/2), were determined by non-compartmental analysis of the plasma concentration-time curves (Phoenix v. 6.2.1.51, Pharsight, C A).


9.1 Pharmacokinetic Parameters for Example 1









TABLE 9







Table 9. Plasma concentration after single oral administration


of 10 mg/kg to male Wistar rat (FIG. 25)









time
Plasma concentration (ng/ml)










(h)
R1
R2












24
<lloq
<lloq


7
12.8
11.5


5
49.6
46.9


3
323.1
233.1


2
722.8
673.9


1
1066.8
1045.9


0.5
1301.9
741.6


0.25
1310.9
534.9





LLOQ: 2 ng/ml













TABLE 10







Table 10. Pharmacokinetic parameters after single


oral administration of 10 mg/kg to male Wistar rat















t1/2
Cmax
tmax
AUC
F



Animal
(h)
(ng/ml)
(h)
(ng · h/ml)
(%)a


















1
0.9
1311
0.25
2950
66



2
0.9
1046
1.00
2340
53



Mean
0.9
1178
0.63
2645
60



SD
0.0
187
0.53
432
9








aMean AUC after i.v. administration was used for F calculation







Thus it can be concluded that:

    • After oral administration to rat of 10 mg/kg, Example 1 achieves a peak plasma concentration of approximately 1000 ng/ml at 0.6 h post-administration (Table 9) and shows a good oral bioavailability (60%) (Table 10).
    • The terminal half-life is very short (<1 h). This terminal half-life is related to a high plasma clearance (70% liver blood flow).
    • Example 1 shows a volume of distribution higher than the total body water volume (1.2 vs. 0.6 l/kg). This result suggests that Example 1 is able to cross cellular membranes and/or has affinity for tissue components.


When the volume of distribution is higher than total body water it is considered the compound is widely distributed and a good therapeutic target exposure could be expected.


9.2 Pharmacokinetic Parameters for Example 2









TABLE 11







Table 11. Plasma concentration after single oral administration


of 10 mg/kg to male Wistar rat (FIG. 26)









time
Plasma concentration (ng/ml)










(h)
R1
R2












24
<lloq
<lloq


7
41
13


5
45
75


3
157
95


2
289
317


1
907
639


0.5
1251
1039


0.25
1386
1002





Lloq: 2 ng/ml













TABLE 12







Table 12. Pharmacokinetic parameters after single


oral administration of 10 mg/kg to male Wistar rat















t1/2
Cmax
tmax
AUC
F



Animal
(h)
(ng/ml)
(h)
(ng · h/ml)
(%)a


















1
1.7
1386
0.25
2227
50



2
1.2
1039
0.50
1768
40



Mean
1.5
1213
0.38
1997
45



SD
0.3
246
0.18
325
7








aMean AUC after i.v. administration was used for F calculation







Thus it can be concluded that:

    • After oral administration to rat of 10 mg/kg, Example 2 achieves a peak plasma concentration of approximately 1213 ng/ml at 0.4 h post-administration (Table 11).
    • The terminal half-life is moderate (1.5 h) (Table 12).


The peak plasma concentration is achieved at 0.63 (example 1) and 0.38 h post-administration (example 2) suggesting a fast absorption and therefore, a fast onset of action could be expected.


High exposure is preferred to assure the desired activity for the compound. After 10 mg/kg administration, the area under the curve (AUC) gives an oral exposure around 2600 (example 1) and 2000 ng·h/ml (example 2), values that could be considered high enough.


Bioavailability (F) is the fraction of the dose that reaches systemic circulation unchanged. The good value found (60% for example 1 and 45% for example 2) is considered to provoke an exposure high enough and also valid for avoiding the risk of high patient-to-patient variability of blood concentrations of a compound with low bioavailability.

Claims
  • 1-12. (canceled)
  • 13. A crystalline salt of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone.
  • 14. The crystalline salt according to claim 13 wherein the salt is selected from the group consisting of inorganic acids, sulphonic acids and organic acids.
  • 15. The crystalline salt according to claim 13 wherein said salt is selected from hydrochloride, maleate, fumarate, malonate, succinate, oxalate and/or hydrobromide.
  • 16. The crystalline salt according to claim 13 wherein the salt is the hydrochloride salt of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone.
  • 17. The crystalline salt according to claim 13 wherein the salt is the maleate salt of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone.
  • 18. A pharmaceutical composition comprising at least a crystalline salt according to claim 13.
  • 19. A method for the manufacture of a medicament comprising combining the crystalline salt as defined in claim 13 with a pharmaceutically acceptable excipient.
  • 20. A method for the treatment and/or prophylaxis of a sigma receptor mediated disease, comprising administering to a patient in need of such a treatment or prophylaxis a therapeutically effective amount of a crystalline salt as defined in claim 13.
  • 21. The method according to claim 20 wherein the disease is selected from the group consisting of diarrhoea; lipoprotein disorders; migraine; obesity; arthritis; hypertension; arrhythmia; ulcer; learning, memory and attention deficits; cognition disorders; neurodegenerative diseases; demyelinating diseases; addiction to drugs and chemical substances; tardive dyskinesia; epilepsy; stroke; stress; cancer; psychotic conditions; inflammation; or autoimmune diseases.
  • 22. The method according to claim 21, wherein the addiction to drugs and chemical substances is selected from addiction to cocaine, amphetamine, ethanol or nicotine; wherein the stroke is ischemic stroke; andwherein the psychotic conditions are selected from depression, anxiety or schizophrenia.
  • 23. The method according to claim 20 wherein the disease is pain.
  • 24. The method according to claim 23 wherein the pain is neuropathic pain, inflammatory pain or other pain conditions involving allodynia and/or hyperalgesia.
  • 25. The method according to claim 23 wherein the salt administered to the patient is the hydrochloride salt of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin1-yl)ethanone.
  • 26. The method according to claim 24 wherein the salt administered to the patient is the hydrochloride salt of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone.
  • 27. The method according to claim 23 wherein the salt administered to the patient is the maleate salt of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone.
  • 28. The method according to claim 24 wherein the salt administered to the patient is the maleate salt of 1-(4-(2-((1-(3,4-difluorophenyl)-1H-pyrazol-3-yl)methoxy)ethyl)piperazin-1-yl)ethanone.
Priority Claims (1)
Number Date Country Kind
15382436.2 Sep 2015 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2016/070604 9/1/2016 WO 00