WO2015151068A1 - Synthetic analogues of 3-iodothyronamine (t1am) and uses thereof - Google Patents

Synthetic analogues of 3-iodothyronamine (t1am) and uses thereof Download PDF

Info

Publication number
WO2015151068A1
WO2015151068A1 PCT/IB2015/052458 IB2015052458W WO2015151068A1 WO 2015151068 A1 WO2015151068 A1 WO 2015151068A1 IB 2015052458 W IB2015052458 W IB 2015052458W WO 2015151068 A1 WO2015151068 A1 WO 2015151068A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
cooh
compound according
mmoles
nmr
Prior art date
Application number
PCT/IB2015/052458
Other languages
French (fr)
Inventor
Grazia Chiellini
Simona Rapposelli
Riccardo ZUCCHI
Original Assignee
Universita' Di Pisa
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universita' Di Pisa filed Critical Universita' Di Pisa
Publication of WO2015151068A1 publication Critical patent/WO2015151068A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C217/00Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton
    • C07C217/78Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton
    • C07C217/80Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the same carbon skeleton having amino groups and etherified hydroxy groups bound to carbon atoms of non-condensed six-membered aromatic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/27Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by etherified hydroxy groups
    • C07C205/35Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by etherified hydroxy groups having nitro groups and etherified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/49Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by carboxyl groups
    • C07C205/56Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by carboxyl groups having nitro groups bound to carbon atoms of six-membered aromatic rings and carboxyl groups bound to acyclic carbon atoms of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C211/00Compounds containing amino groups bound to a carbon skeleton
    • C07C211/43Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • C07C211/44Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton having amino groups bound to only one six-membered aromatic ring
    • C07C211/49Compounds containing amino groups bound to a carbon skeleton having amino groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton having amino groups bound to only one six-membered aromatic ring having at least two amino groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C217/00Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton
    • C07C217/02Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C217/04Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C217/06Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one etherified hydroxy group and one amino group bound to the carbon skeleton, which is not further substituted
    • C07C217/14Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one etherified hydroxy group and one amino group bound to the carbon skeleton, which is not further substituted the oxygen atom of the etherified hydroxy group being further bound to a carbon atom of a six-membered aromatic ring
    • C07C217/18Compounds containing amino and etherified hydroxy groups bound to the same carbon skeleton having etherified hydroxy groups and amino groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated having only one etherified hydroxy group and one amino group bound to the carbon skeleton, which is not further substituted the oxygen atom of the etherified hydroxy group being further bound to a carbon atom of a six-membered aromatic ring the six-membered aromatic ring or condensed ring system containing that ring being further substituted
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/40Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino groups bound to carbon atoms of at least one six-membered aromatic ring and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton

Definitions

  • the present invention generally refers to synthetic analogues of thyroid hormones. More specifically, the invention relates to synthetic analogues of 3-iodothyronamine.
  • pharmaceutical compositions comprising at least one of the said synthetic analogues of 3- iodothyronamine as the active ingredient in combination with a pharmaceutically acceptable carrier and/or excipient, as well as medical uses of the said synthetic 3- iodothyronamine analogues.
  • 3-Iodothyronamine (TI AM) is an endogenous substance first identified in 2004 (Scanlan et al, Nat Med. 2004 Jun; 10(6):638-42. Epub 2004 May 16).
  • thyroid hormones It is a derivative of thyroid hormones, which with full rights may be regarded as a distinct hormone, as it is an endogenous compound that so far has been virtually identified in all tissues. It is also a potent agonist of membrane receptors known as "trace amine-associated receptors " (TAAR), which are expressed in many organs and until a few years ago were thought to be orphan receptors. TAAR was further suggested also to interact with other molecular targets, such as monoamine carriers, mitochondrial proteins and. at least in some tissues, with alpha-2-adrenergic receptors. But TI AM was not shown to be a ligand for nuclear receptors of thyroid hormones (TR). Finally, TI AM produces significant functional effects.
  • TR thyroid hormones
  • the first effects to be observed consisted of a decrease in body temperature and heart contractility. Now we know that such effects occur at concentrations several orders of magnitude higher than the physiological ones. However, effects have been discovered which are detected at concentrations similar to the physiological ones, which are related to regulation of the metabolism and function of the central nervous system. Particularly, the literature shows that TI AM has an anti-insulin action, it favours the lipid catabolism over glucose metabolism and facilitates learning.
  • TI AM is in the treatment of obesity.
  • chronic administration ( 1 week) of TI AM at a low dosage in experimental animals resulted in a significant decrease in body weight, without change in food intake, and glucose tolerance,
  • the effect proved to be persistent at 2 weeks after treatment discontinuation, and is most likely linked to a complex expression modulation of proteins regulating homeostasis of glucose and lipids, such as sirtuins.
  • T I AM is rapidly metabolized by several enzymatic systems, such as: amine oxidases (MAO, SSAO), with formation of 3- iodothyroacetic acid; deiodinases (DI03), with formation of thyronamine; sulfotransferases (SULT 1 A 1 and SULT 1 A3); N-acetyltransferases; glucuronidases.
  • MAO amine oxidases
  • DI03 deiodinases
  • DI03 sulfotransferases
  • SULT 1 A 1 and SULT 1 A3 N-acetyltransferases
  • glucuronidases glucuronidases.
  • T I AM 3-iodothyronamine
  • the synthetic analogues of 3-iodothyronamine which are the object of the present invention, are pharmaceutically effective active ingredients suitable to be used in treating diseases such as, for example, obesity, dyslipidemias, hypothyroidism (optionally in association with the thyroid hormone), neuropsychiatric disorders, behaviour disorders, cardiovascular disorders, hormone disorders such as, for example, Polycystic ovary syndrome (PCOS), ageing-related disorders.
  • diseases such as, for example, obesity, dyslipidemias, hypothyroidism (optionally in association with the thyroid hormone), neuropsychiatric disorders, behaviour disorders, cardiovascular disorders, hormone disorders such as, for example, Polycystic ovary syndrome (PCOS), ageing-related disorders.
  • PCOS Polycystic ovary syndrome
  • T I AM 3-iodothyronamine
  • X is selected from O (oxygen) and CH 2 ;
  • n is an integer comprised between 0 (zero) and 3;
  • Z is selected from NH2, N0 2 and OR 3 ;
  • R 2 is selected from the group consisting of CH 2 NH 2 , CH 2 NR R 5 , COOH, CN and COOR 4 ; wherein R, R 1 , R 3 , R 4 and R 5 are independently selected from H and lower alkyl.
  • lower alkyl is to be understood as a linear or branched alkyl group having from 1 to 4 carbon atoms, particularly methyl, ethyl, iso- propyl, n-propyl, iso-butyl, n-butyl, sec-butyl or tert-butyl.
  • Pharmaceutically acceptable salts of the compounds of formula (I) are also included in the scope of the present invention.
  • Inorganic acids that can be used for obtaining pharmaceutically acceptable salts are, for instance, hydrochloric acid, phosphoric acid and sulfuric acid. Oxalates, tartrates, maleates, citrates and succinates are included among salts derived from the inorganic acids.
  • R and R 1 are both H.
  • R and R 1 are both methyl. In still another preferred embodiment, R is methyl and R 1 is H. In still a further preferred embodiment, n is zero or 1 .
  • R 2 is CH 2 NH 2 or COOH.
  • Preferred compounds that fall within the scope of the present invention are listed hereinafter and are defined with reference to formula (I):
  • Example 1 synthesis of T1AM analogues according to the invention
  • the structure of the compounds was assessed by ⁇ -NMR and mass spectrophotometry. The most significant elements of the ⁇ -NMR and MS spectra were reported. All the synthesized compounds exhibit spectrum data in agreement with the assigned structures.
  • the nuclear magnetic resonance spectra were performed with a Varian Gemini 200 MHz or Bruker TopSpin 3.2 400 MHz spectrometer; the solutions are approximately at 5% in CDC1 , CD 3 OD or DMSO-d 6 . The chemical shifts were expressed in ppms ( ⁇ scale). The melting points were determined under a Kolfler microscope and are uncorrected.
  • the elemental analyses were carried out at the laboratory of Analytical Chemistry of the present inventors; the differences between the theoretical values and those obtained were comprised within a range of ⁇ 0.4%.
  • the evaporations were performed in a rotary evaporator and the dehydration of the organic phases was carried out by using Na 2 SC>4.
  • the analytical TLCs were performed by using MERCK silica gel (G60) plates with a 20 x 20.2 mm fluorescence indicator. The spots were detected by a UV lamp (256nm). For column chromatography, 70-230 mesh silica gel was used. For filtration through celite, celite * 521 was used.
  • Reagents and conditions a: 4-Nitrobenzyl bromide, K2CO3, PdCl 2 , Acetone/H 2 0, r.t., 72 hrs; b: BBr 3 , CH 2 C1 2 , 0°C. 1 hr; c: BrCH 2 CN, DMF, Cs 2 C0 3 , r.t., 30 min; d: L1AIH4, AICI3, THF, reflux, 12 hrs.
  • Derivatives SG I and SG_2 were obtained by following the synthesis procedure shown in SCHEME 1.
  • the subsequent demethylation reaction in the presence of BBr 3 at 0°C resulted in the corresponding phenol derivatives 2a,b, which were subjected to an alkylation reaction with bromoacetonitrile to give the cyano derivatives 3a, b.
  • the compounds were finally reduced with LiAlHj/AlCb into the final products SG_1 and SG_2
  • the derivative la,b is solubilized in the minimum amount of DCM, placed under N 2 at - 78°C.
  • BBr 3 (3.94 mL, 1 .24 mmoles) is added dropwise to this solution.
  • the reaction is stirred for 5 min at -78°C, and then kept at 0°C for 1 .5 hrs in an ice bath. Thereafter, the reaction is diluted with water and extracted with DCM. The organic layer is dried, filtered arid evaporated, to give compounds 2a,b. 4-(4-nitroben ⁇ yl)phenol (2a). White oil. (95% yield).
  • the phenol derivatives 2a, b (100 mg, 0.44 mmoles) were solubilized in the minimum amount of DMF, Cs 2 C0 3 (725 mg, 2.22 mmoles) was added till formation of the phenate. Then, BrCH 2 CN (0.03 mL, 0.44 mmoles) was added to the reaction mixture. The reaction was stirred at r.t. for 30 min. Thereafter, a IN HC1 solution was added to the mixture, which was extracted with AcOEt. The combined organic layers were successively washed with a saturated NaCl solution and ice. The organic layer was dried, filtered and evaporated.
  • Reagents and conditions a: BrCH 2 COOEt, DMF, Cs 2 C0 3 , r.t, 30 min; b: NaOH 10%, MeOH, reflux, lhr; c: Hydrazine hydrate, Charcoal, FeCl 3 , MeOH, reflux, 12 hrs.
  • Reagents and conditions a: Pd/C, AcOH. EtOH, r.t., 48 hrs; b: NaN0 2 , H 2 S0 4 , H 2 0, 100°C, 1 hr; c: L1AIH4, A1C1 3 , THF, reflux. 12 hrs.
  • SG 10 Reagents and conditions: a: KHF 2 , MeOH/H 2 0, r.t., 30 min; b: 4-Nitrobenzyl bromide, PdCl 2 dppf, Cs 2 C0 3 , H 2 0/Dioxane, 95°C, 24 hrs; c: SOCl 2 , CHCI3, r.t., 2hrs; d: NaCN, H 2 0/CH 3 CN, 100°C, 150 W, 8 bars, 4 x 20 min cycles; e: LiAlH , AICI3, THF, reflux, 12 hrs; f: H 2 S0 4 50%. reflux, 30 min; g: Hydrazine hydrate, Charcoal, FeCl 3 , MeOH, reflux, 12 hrs.
  • the experimental model used is the isolated perfused rat heart with the working heart technique (Zucchi R, Ronca-Testoni S, Yu G, Galbani P, Ronca G, Mariani M. Effect of ischemia and reperfusion on cardiac ryanodine receptors - sarcoplasmic reticulum Ca 2+ channels. Circ Res 1994: 74:271 -280), which allows for monitoring the main haemodynamic variables, such as: aortic flow, coronary flow, cardiac output, heart rate, aortic pressure.
  • the hearts of Sprague-Dawley rats (275-300 g of weight) were removed and perfused by using the working heart apparatus, in which the preload (height of the atrial chamber) and the afterload (height of the aortic chamber) were set at heights of 20 and 70 cm. respectively.
  • the aortic and coronary flows were measured by collecting the perfusion liquid from the aortic and atrial chambers, respectively, into a graduated cylinder.
  • the cardiac output was determined as the sum of the aortic and coronary flows.
  • the perfusion medium was the rebs-Henseleit buffer solution (pH 7.4), which has the following composition (mmol/L): NaCl 1 1 8, NaHC0 3 25, C1 4.5, H 2 P0 4 1 .2, MgS0 4 1 .2, CaCl 2 2.5, and glucose 1 ).
  • the perfusion medium was kept in recirculation by a mechanical pump and was equilibrated with an 0 2 (95%)/C0 2 (5%) mixture. The temperature was maintained constant throughout the experiment at 37-37.4°C. After a 10 min equilibration time, the compounds (SG I and SG 2) were added to the perfusion medium, at concentrations within the range of from 100 nM to 100 microM, and the haemodynamic variables were monitored for 60 min.
  • T0AM thyronamine
  • T1 AM 3-iodothyronamine
  • Example 3 metabolic effects In male mice (CD 1 , weighing 25.2 +/- 3.2 g; Harlan-Nossan (Italy) fasted for 4 hrs before being subjected to an i.c.v injection (Manni et al, Br J Pharmacol 2013; 168(2):354-362) with saline (controls) or with one of the test compounds (T l A at doses of 1 .32 microgrKg- 1 and 4 microgrKg- 1 ; T0AM, T1 AM and SG I at 1 .32 microgrKg- 1 ), the glucose levels were measured in blood collected from the venous vessels of the tail. As shown in Figure 2, SG- 1 administered i.e. v.
  • FIG. 1 shows the effects of SG I on glycaemia.
  • Example 4 agonistic TAAR 1 activity
  • HEK-293 cells transfected with the murine TAAR 1 receptor (mTAAR l ) were examined for the induction of cAMP production by the BRET (Bioluminescence Resonance Energy Transfer) technique according to the method described in literature (Espinoza S, Salahpour A, Masri B, Sotnikova TD, Messa M, Barak LS, Caron MG, Gainetdinov RR. Functional interaction between trace amine-associated receptor 1 and dopamine D2 receptor. Mol Pharmacol. 201 1 ; 80(3):416-25).
  • BRET Bioluminescence Resonance Energy Transfer
  • Figure 3 shows the dose-response curves for activation of mTAAR l . Particularly, it shows the differential cAMP levels by using the cAMP BRET biosensor in HE -293T cells that express mTAARl .
  • the values on the Y-axis in each panel denote the Rluc/YFP ratio, which is known as the BRET ratio.
  • the table that follows shows the preferred compounds of the present invention.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Novel synthetic analogues of 3-iodothyronamine of formula (I), pharmaceutical compositions containing the same, and medical uses thereof are described.

Description

Synthetic analogues of 3-iodothyronamine (TI AM) and uses thereof
The present invention generally refers to synthetic analogues of thyroid hormones. More specifically, the invention relates to synthetic analogues of 3-iodothyronamine. pharmaceutical compositions comprising at least one of the said synthetic analogues of 3- iodothyronamine as the active ingredient in combination with a pharmaceutically acceptable carrier and/or excipient, as well as medical uses of the said synthetic 3- iodothyronamine analogues. 3-Iodothyronamine (TI AM) is an endogenous substance first identified in 2004 (Scanlan et al, Nat Med. 2004 Jun; 10(6):638-42. Epub 2004 May 16). It is a derivative of thyroid hormones, which with full rights may be regarded as a distinct hormone, as it is an endogenous compound that so far has been virtually identified in all tissues. It is also a potent agonist of membrane receptors known as "trace amine-associated receptors " (TAAR), which are expressed in many organs and until a few years ago were thought to be orphan receptors. TI AM was further suggested also to interact with other molecular targets, such as monoamine carriers, mitochondrial proteins and. at least in some tissues, with alpha-2-adrenergic receptors. But TI AM was not shown to be a ligand for nuclear receptors of thyroid hormones (TR). Finally, TI AM produces significant functional effects. The first effects to be observed consisted of a decrease in body temperature and heart contractility. Now we know that such effects occur at concentrations several orders of magnitude higher than the physiological ones. However, effects have been discovered which are detected at concentrations similar to the physiological ones, which are related to regulation of the metabolism and function of the central nervous system. Particularly, the literature shows that TI AM has an anti-insulin action, it favours the lipid catabolism over glucose metabolism and facilitates learning.
However, the most promising therapeutic application of TI AM is in the treatment of obesity. In fact, a recent experimental study described that chronic administration ( 1 week) of TI AM at a low dosage in experimental animals resulted in a significant decrease in body weight, without change in food intake, and glucose tolerance, The effect proved to be persistent at 2 weeks after treatment discontinuation, and is most likely linked to a complex expression modulation of proteins regulating homeostasis of glucose and lipids, such as sirtuins.
As extensively described in the literature, T I AM is rapidly metabolized by several enzymatic systems, such as: amine oxidases (MAO, SSAO), with formation of 3- iodothyroacetic acid; deiodinases (DI03), with formation of thyronamine; sulfotransferases (SULT 1 A 1 and SULT 1 A3); N-acetyltransferases; glucuronidases.
This obviously limits the possibilities of therapeutic uses of T I AM.
Accordingly, the development of synthetic T I AM analogues preferably provided with an enhanced biopharmacological profile and/or improved pharmacokinetic properties appears to be of fundamental importance for the purpose of an effective therapeutic application. In order to meet such a requirement, the present inventors synthesized a set of synthetic analogues of 3-iodothyronamine (T I AM) which, through a preliminary screening in experimental models available at the laboratories of these same inventors and previously used for studying the functional (isolated working rat heart) and metabolic (icv administration to mice) effects of T I AM, demonstrated an optimal mimicking of the effects induced by administration of TI AM or its endogenous derivative thyronamine (TOAM). These experiments are showed in detail in the section concerning the Examples. The results obtained show that the synthetic analogues of 3-iodothyronamine, which are the object of the present invention, are pharmaceutically effective active ingredients suitable to be used in treating diseases such as, for example, obesity, dyslipidemias, hypothyroidism (optionally in association with the thyroid hormone), neuropsychiatric disorders, behaviour disorders, cardiovascular disorders, hormone disorders such as, for example, Polycystic ovary syndrome (PCOS), ageing-related disorders.
The synthetic analogues of 3-iodothyronamine (T I AM) that form the subject of the present invention are represented by the following general formula (I):
Figure imgf000004_0001
wherein X is selected from O (oxygen) and CH2;
n is an integer comprised between 0 (zero) and 3;
Z is selected from NH2, N02 and OR3; and
R2 is selected from the group consisting of CH2NH2, CH2NR R5, COOH, CN and COOR4; wherein R, R1 , R3, R4 and R5 are independently selected from H and lower alkyl.
In the present description, the term "lower alkyl" is to be understood as a linear or branched alkyl group having from 1 to 4 carbon atoms, particularly methyl, ethyl, iso- propyl, n-propyl, iso-butyl, n-butyl, sec-butyl or tert-butyl.
Pharmaceutically acceptable salts of the compounds of formula (I) are also included in the scope of the present invention. Inorganic acids that can be used for obtaining pharmaceutically acceptable salts are, for instance, hydrochloric acid, phosphoric acid and sulfuric acid. Oxalates, tartrates, maleates, citrates and succinates are included among salts derived from the inorganic acids.
In a preferred embodiment of the invention, R and R1 are both H.
In another preferred embodiment, R and R1 are both methyl. In still another preferred embodiment, R is methyl and R1 is H. In still a further preferred embodiment, n is zero or 1 .
In still another preferred embodiment, R2 is CH2NH2 or COOH. Preferred compounds that fall within the scope of the present invention are listed hereinafter and are defined with reference to formula (I):
compound SG_1 , wherein R = R.' = H; X = 0; n = l ; Z = NH2; R2 = CH2NH2;
compound SG_2, wherein R = CH3; R1 = H; X = O; n = 1 ; Z = NH2 ; R2 = CH2NH2;
compound SG_3, wherein R = R' = H; X = 0; n = l ; Z = N02; R2 = COOH;
compound SG_4, wherein R = CH3; R1 = H; X = O; n = 1 ; Z = N02; R2 = COOH;
compound SG_5, wherein R = R' = H; X = 0; n = l ; Z = NH2; R2 - COOH;
compound SG_6, wherein R = methyl; R1 = H; X = O; n = 1 ; Z = NH2; R2 = COOH;
compound SG_7, wherein R - R1 - H; X = O; n = 1 ; Z = OH; R2 = CH2NH2;
compound SG_8. wherein R = R1 = H; X = CH2; n = 0; Z = NH2; R2 = CH2NH2;
compound SG_9, wherein R = R1 = H; X = CH2; n = 0; Z = N02; R2 = COOH; and compound SGJ 0, wherein R = R1 = H; X = CH2; n = 0; Z = NH2; R2 - COOH.
The synthesis of the said compounds is illustrated hereunder in the Examples section and more particularly in the reaction schemes 1 -4. Such reaction schemes illustrate general synthesis procedures that can be used for the synthesis of all compounds falling within the scope of the above-shown general formula (I).
The examples that follow are provided for illustrative purposes only and are not intended as limiting the scope of the invention, as defined in the appended claims.
EXAMPLES
Example 1 : synthesis of T1AM analogues according to the invention
The structure of the compounds was assessed by Ή-NMR and mass spectrophotometry. The most significant elements of the Ή-NMR and MS spectra were reported. All the synthesized compounds exhibit spectrum data in agreement with the assigned structures. The nuclear magnetic resonance spectra were performed with a Varian Gemini 200 MHz or Bruker TopSpin 3.2 400 MHz spectrometer; the solutions are approximately at 5% in CDC1 , CD3OD or DMSO-d6. The chemical shifts were expressed in ppms (δ scale). The melting points were determined under a Kolfler microscope and are uncorrected. The elemental analyses were carried out at the laboratory of Analytical Chemistry of the present inventors; the differences between the theoretical values and those obtained were comprised within a range of ± 0.4%. The evaporations were performed in a rotary evaporator and the dehydration of the organic phases was carried out by using Na2SC>4. The analytical TLCs were performed by using MERCK silica gel (G60) plates with a 20 x 20.2 mm fluorescence indicator. The spots were detected by a UV lamp (256nm). For column chromatography, 70-230 mesh silica gel was used. For filtration through celite, celite * 521 was used.
SCHEME 1
Figure imgf000006_0001
Reagents and conditions: a: 4-Nitrobenzyl bromide, K2CO3, PdCl2, Acetone/H20, r.t., 72 hrs; b: BBr3, CH2C12, 0°C. 1 hr; c: BrCH2CN, DMF, Cs2C03, r.t., 30 min; d: L1AIH4, AICI3, THF, reflux, 12 hrs.
Derivatives SG I and SG_2 were obtained by following the synthesis procedure shown in SCHEME 1. The cross-coupling reaction between p-nitrobenzyl bromide and the appropriate 4-methoxybenzeneboronic acid, using PdCl2 as the catalyst and K2C03 as the base, provided compounds la,b. The subsequent demethylation reaction in the presence of BBr3 at 0°C resulted in the corresponding phenol derivatives 2a,b, which were subjected to an alkylation reaction with bromoacetonitrile to give the cyano derivatives 3a, b. The compounds were finally reduced with LiAlHj/AlCb into the final products SG_1 and SG_2
General procedure for the synthesis of derivatives la,b p-Nitrobenzyl bromide (569 mg, 2.63 mmoles) was added to a solution of the appropriate arylboronic acid (2.63 mmoles) in acetone/F O in a 1 : 1 ratio (4 ml), placed under N2, and the resulting solution was stirred for 10 minutes. Thereafter, K2C03 ( 1 .24 g, 6.58 mmoles) and a catalytic amount of PdCl2 were added to this mixture. The mixture thus obtained was kept under stirring for 62 hours. After which, the solvent was evaporated and the aqueous layer was extracted with Et20. The organic layer was dried, filtered and evaporated. l-methoxy-4-(4-nitroben∑yl)benzene (la). The crude compound was purified by column chromatography, using an n-Hexane/AcOEt (97:3) mixture as the eluent. Yellow oil. (47% yield). Ή NMR (CDC13): δ 3.79 (s, 3H, OCH3); 4.02 (s, 2H, CH2); 6.86 (d, 2H, J = 8.3 Hz, Ar); 7.10 (d, 2H, J = 8.3, Ar); 7.32 (d, 2H, j=8.3 Hz, Ar); 8.13 (d, 2H, j=8.3 Hz, Ar) ppm. Anal. (C |4H|3N03) C, H, N. % Calc. 69.12 (C); 5.39 (H); 5.76 (N). % Obs. 69.12 (C); 5.39 (H); 5.76 (N).
4-methoxy-2-methyl- 1 -(4-nitrobenzyl)benzene (lb). The crude compound was purified by column chromatography, using an n-Hexane/AcOEt (97:3) mixture as the eluent. Yellow oil. (49% yield). Ή NMR (CDC13): δ 2.15 (s, 3H, CH3); 3.78 (s, 3H, OCH3); 4.01 (s, 2H, CH2); 6.69-6.80 (m, 2H. Ar); 7.01 (s, 1 H, Ar); 7.23 (d, 2H. J =8.4 Hz, Ar); 8.10 (d. 2H, J = 8.4 Hz, Ar) ppm. Anal. (C 15H, 5N03) C, H, N. % Calc. 70.02 (C); 5.88 (H); 5.44 (N). % Obs. 70.22 (C); 5.93 (H); 5.62 (N).
General procedure for the synthesis of derivatives 2a, b
The derivative la,b is solubilized in the minimum amount of DCM, placed under N2 at - 78°C. BBr3 (3.94 mL, 1 .24 mmoles) is added dropwise to this solution. The reaction is stirred for 5 min at -78°C, and then kept at 0°C for 1 .5 hrs in an ice bath. Thereafter, the reaction is diluted with water and extracted with DCM. The organic layer is dried, filtered arid evaporated, to give compounds 2a,b. 4-(4-nitroben∑yl)phenol (2a). White oil. (95% yield). Ή NMR (CDC13): δ 4.00 (s, 2H, CH2); 6.78 (d, 2H, J =7.5 Hz, Ar); 7.04 (d, 2H, J = 7.5 Hz, Ar) 7.32 (d, 2H, J=7.9 Hz, Ar); 8.14 (d, 2H, J=7.9 Hz, Ar) ppm. Anal. (C |3HnN03) C, H, N. % Calc. 68.1 1 (C); 4.84 (H); 6.1 1 (N). % Obs. 68.29 (C); 4.96 (H); 6.29 (N).
3-methyl-4-(4-nitroben∑yl)phenol (2b). Light yellow oil. (64% yield). Ή NMR (CDC13): δ 2.13 (s, 3H, CH3); 4.00 (s. 2H, CH2): 6.65-6.59 (m, 2H. Ar); 6.96 (s, 1 H, Ar); 7.24 (d, 2H, J=7.9 Hz. Ar); 8.1 1 (d, 2H, J=7.9 Hz, Ar) ppm. Anal. (C uH,3N03) C, H, N. % Calc. 69.12 (C); 5.39 (H); 5.76 (N). % Obs. 69.33 (C); 5.51 (H); 5.82 (N).
General procedure for the synthesis of derivatives 3a, b
The phenol derivatives 2a, b (100 mg, 0.44 mmoles) were solubilized in the minimum amount of DMF, Cs2C03 (725 mg, 2.22 mmoles) was added till formation of the phenate. Then, BrCH2CN (0.03 mL, 0.44 mmoles) was added to the reaction mixture. The reaction was stirred at r.t. for 30 min. Thereafter, a IN HC1 solution was added to the mixture, which was extracted with AcOEt. The combined organic layers were successively washed with a saturated NaCl solution and ice. The organic layer was dried, filtered and evaporated.
[4-(4-mtrobenzyl)phenoxy]acetonitrile (3a). The crude compound was purified by column chromatography, using an n-Hexane/ AcOEt (70:30) mixture as the eluent. White oil. (96% yield). Ή NMR (CDC13): δ 4.04 (s, 2H, CH2); 4.76 (s, 2H, CH2): 6.94 (d, 2H, J = 8.6 Hz, Ar); 7.15 (d. 2H, J = 8.6, Ar) 7.31 (d, 2H, J=8.6 Hz. Ar); 8.14 (d, 2H, J=8.6 Hz, Ar) ppm. Anal. (C ,5H|2N203) C, H. N. % Calc. 67.16 (C); 4.51 (H); 10.44 (N). % Obs. 67.24 (C); 4.72 (H); 10.62 (N).
3-[methyl-4-(4-nitroben∑yl)phenoxy]acetonitrile (3b). The crude compound was purified by column chromatography, using an n-Hexane/AcOEt (70:30) mixture as the eluent. White oil. (98% yield). Ή NMR (CDCI ): δ 2.19 (s, 3H, CH ); 4.04 (s, 2H, CH2); 4.76 (s, 2H. CH2); 6.81 -6.77(m, 2H, Ar); 7.08 (s. 1 H, Ar); 7.25 (d, 2H, J = 8.6 Hz, Ar); 8.13 (d, 2H, J - 8.6 Hz, Ar) ppm. Anal. (C 16H 14N2O3) C, H, N. % Calc. 68.07 (C); 5.00 (H); 9.92 (N). % Obs. 68.21 (C); 5.1 1 (H); 9.86 (N).
General procedure for the synthesis of derivatives SG I and SG 2.
A solution of 3a,b (0.34 mmoles) in THF was added to a solution of L1AIH4 (3.04 mmoles) and AICI3 (405 mg; 3.04 mmoles) in THF, placed under nitrogen, stirred at r.t. for 5 minutes. The reaction was stirred under reflux at 66°C for 12 hours. After this time, the mixture was cooled to 0°C, H20 was added, and the resulting mixture was acidified with a 10% HC1 solution. The mixture was then washed with Et20, the aqueous layer was alkalized with a 2N NaOH aqueous solution, and CHCI3 was added thereto. The resulting emulsion was filtered through a celite pad and the organic layer was finally washed with saturated NaCl, dried, filtered and evaporated under reduced pressure. 4-(4-(2-aminoethoxy)benzyl)aniline (SG_1). The crude compound was purified by formation of the hydrochloride. White solid. M.p.: 137- 135°C. (70% yield). Ή NMR (CD3OD): δ 3.35 (t, 2H, J = 5.0 Hz; CH2); 3.98 (s, 2H, CH2); 4.20 (t, 2H. J = 5.0 Hz; CH2NH2); 6.95 (d, 2H, J = 8,5 Hz, Ar); 7.16 (d, 2H. J = 8.5 Hz, Ar); 7.30 (d, 2H, J = 8.8 Hz, Ar); 7.37 (d, 2H, J = 8.8 Hz Ar) ppm. I 3C NMR (CD3OD): δ 142.73, 139.53, 134.80, 130.20. 128.99, 128.70, 128.52. 122.73, 41 .04, 40.90, 32.70 ppm. Anal. (C ,?H|8N20) C, H. N. % Calc. 74.35 (C); 7.49 (H); 1 1 .56 (N). % Obs. 74.61 (C); 7.52 (H); 1 1 .73 (N).
4-(4-(2-aminoethoxy)-2-methylben∑yl)aniline (SG_2). The crude compound was purified by formation of the hydrochloride. White solid. M.p.: 156- 1 58°C. (75% yield). Ή NMR (CD3OD): δ 3.35 (t, 2H, J = 4.9 Hz; CH3); 4.00 (s. 2H, CH2); 4.21 (t, 2H, J = 4.9 Hz; CH2NH2); 6.79-6.88 (m. 2H, Ar); 7.09 (d. 1 H, J = 8.0 Hz, Ar); 7.23-7.34 (m, 4H, Ar) ppm. I 3C NMR (CD3OD): δ 156.92. 142.50, 137.92, 13 1 .37, 130.81. 129.87, 128.26, 122.61. 1 16.44, 1 1 1.64, 63.82, 39.00. 37.49, 18.52 ppm. Anal. (C |6H2oN20) C, H, N. % Calc. 74.97 (C); 7.86 (H); 10.93 (N). % Obs. 75.00 (C); 7.91 (H); 10.89 (N).
SCHEME 2
Figure imgf000010_0001
Figure imgf000010_0002
Reagents and conditions: a: BrCH2COOEt, DMF, Cs2C03, r.t, 30 min; b: NaOH 10%, MeOH, reflux, lhr; c: Hydrazine hydrate, Charcoal, FeCl3, MeOH, reflux, 12 hrs.
Compounds SG 3 - SG_6 were synthesized by the procedure described in scheme 2. The alkylation reaction between the appropriate 2a,b phenol and ethyl -bromoacetate provided the corresponding ethyl esters 4a,b. The subsequent hydrolysis with 10% NaOH allowed for obtaining the SG_3 and SG_4 acids. The amine analogues SG_5 and SG_6 were obtained by reduction of the nitro group with hydrazine hydrate in the presence of FeCl3 and charcoal.
General procedure for the synthesis of derivatives 4a, b The 2a,b phenol ( l OOmg, 0.44 mmoles), solubilized in the minimum amount of DMF, is reacted with BrCH2COOEt (73.5mg, 0.44 mmoles). The synthetic procedure and the treatment of the reaction are analogous to those previously described for derivatives 3a, b.
Ethyl-2-(4-(4-nitroben∑yl)phenoxy)acetate (4a). Yellow oil. (83% yield). Ή NMR (CDCI3): δ 1 .27 (t, 3H, J =7.1 Hz, CH3); 4.02 (s, 2H, CH2); 4.27 (q, 2H, J =7.1 Hz, CH2); 4.60 (s, 2H, CH2); 6.86 (d, 2H, J = 8.4 Hz); 7.09 (d, 2H, J = 8.8 Hz); 7.31 (d, 2H, J = 8.8 Hz); 8. 13 (d, 2H, J = 8.4 Hz) ppm. Anal. (C 7H1 7NO5) C. H, N. % Calc. 64.75 (C); 5.43 (H); 4.44 (N). % Obs. 64.80 (C); 5.71 (H); 4.69 (N). Ethyl-2-(S- ethyl-4-(4-m(robenzyl)phenoxy)acetate (4b). Yellow oil. (60% yield). Ή NMR (CDC13): δ 1 .26 (t, 3H, J =7.0 Hz, CH3); 2.12 (s, 3H, CH3); 3.98 (s, 2H, CH2); 4.23 (q, 2H, J =7.0 Hz, CH2); 4.57 (s, 2H, CH2); 6.32-6.79 (m, 2H, Ar); 6.97 (d, 1 H, J = 8.2, Ar) 7.21 (d, 2H, J =8.5 Hz, Ar); 8.08 (d, 2H, J = 8.5 Hz, Ar) ppm. Anal.
Figure imgf000011_0001
C, H, N. % Calc. 65.64 (C); 5.81 (H); 4.25 (N). % Obs. 65.91 (C); 5.93 (H); 4.39 (N).
General procedure for the synthesis of derivatives SG 3 and SG 4
A 10% NaOH solution (0.2 ml) was added to a solution of the ester derivative 4a,b (0.57 mmoles) in MeOH; the resulting solution was stirred under reflux for 30 minutes. Thereafter, the reaction mixture was set at 0°C, and H20 was added thereto; the precipitated solid was collected by filtration and washed with H20 giving the desired products. 2-(4-(4-Nitrobenzyl)phenoxy)acetic acid (SG_3). White solid. M.p.: 162-164°C. (87% yield). Ή NMR (CDC13): δ 4.02 (s, 2H, CH2); 4.66 (s, 2H, CH2); 6.88 (d, 2H, J = 8.4 Hz, Ar); 7.1 1 (d, 2H, J = 8.4 Hz, Ar); 7.31 (d, 2H, J = 8.4 Hz, Ar); 8.14 (d, 2H, J = 8.4 Hz, Ar) ppm. Anal. (0 ,5Η,3ΝΟ5) C, H, N. % Calc. 62.72 (C); 4.56 (H); 4.88 (N). % Obs. 62.91 (C); 4.23 (H); 4.59 (N).
2-(4-(4-Nitrobenzyl)-3-methylphenoxy)acetic acid (SG_4). White solid. M.p.: 166-168°C. (90% yield). Ή NMR (CDC13): δ 2.17 (s, 3H, 4.02 CH3), 4.02 (s, 2H, CH2); 4.68 (s. 2H, CH2); 6.70-6.80 (m, 2H, Ar); 7.03 (d, 1 H, J = 8.2, Ar); 7.24 (d, 2H, J =7.9 Hz, Ar); 8.12 (d, 2H, J =7.9 Hz. Ar) ppm. Anal. (C|6H |5N05) C, H, N. % Calc. 63.78 (C); 5.02 (H); 4.65 (N). % Obs. 63.87 (C); 5.21 (H); 4.51 (N).
General procedure for the synthesis of derivatives SG 5 and SG_6
Charcoal (33.7 mg) and FeCl3 (7 mg) were added to a solution of the nitro derivative (0.64 mmoles) solubilized in the minimum amount of MeOH (4 ml). The reaction mixture was heated to reflux for 15 minutes, then a solution of hydrazine hydrate was added (0.33 ml, 6.71 mmoles), and the resulting mix was refluxed and agitated overnight. After this time, the mixture was filtered and the filtrate was evaporated. The residue was taken up with AcOEt and washed with H20; the organic layer was dried, filtered and evaporated.
2-(4-(4-Ammoben∑yl)phenoxy)acetic acid (SG_5). White solid. M.p. : 153- 155°C. (70% yield). Ή NMR (CDC13): δ 3.82 (s, 2H, CH2); 4.54 (s. 2H, CH:); 6.62 (d, 2H, J = 8.4 Hz, Ar); 6.81 (d, 2H, J = 8.4 Hz, Ar); 6.95 (d. 2H, J = 8.4 Hz, Ar); 7.1 1 (d, 2H, J = 8.4 Hz Ar) ppm. UC NMR (CDC13): δ 168.76, 1 55.34, 144.52, 135.92, 131 .22, 130.04. 129.65, 1 15.34, 1 14.49, 67.09, 40.14 ppm. Anal. (0 ,5Η,5ΝΟ3) C, H, N. % Calc. 70.02 (C); 5.88 (H); 5.44 (N). % Obs. 70.39 (C); 5.93 (H); 5.59 (N).
2-(4-(4-Aminobenzyl)-3-methylphenoxy)acetic acid (SG_6). White solid. M.p.: 175-177°C. (65% yield). Ή NMR (CD3OD): δ 2.16 (s, 3H, CH3); 3.77 (s, 2H, CH2); 4.36 (s. 2H. CH2); 6.57-6.80 (m, 4H. Ar); 6.81 -6.89 (m, 2H. Ar) 6.92-7.01 (m, 2H. Ar) ppm. Anal. (C|6H| 7N03) C, H, N. % Calc. 70.83 (C); 6.32 (H); 5.16 (N). % Obs. 70.90 (C); 6.37 (H); 5.29 (N).
SCHEME 3
Figure imgf000012_0001
Reagents and conditions: a: Pd/C, AcOH. EtOH, r.t., 48 hrs; b: NaN02, H2S04, H20, 100°C, 1 hr; c: L1AIH4, A1C13, THF, reflux. 12 hrs.
Compound SG_7 was synthesized by the procedure described in scheme 3. The catalytic hydrogenation of 3a by using Pd/C in concentrated AcOH provided the aniline derivative 5. which upon reaction with NaN02 in H2S04 provided the phenol 6 through formation of the diazonium salt as the intermediate. Compound 6 was finally reduced with LiAlH /AlCl3 into the final product SG_7.
Synthesis of derivative 5
2-(4-(4-Aminobenzyl)phenoxy)acetomtrile (5). A solution of the nitro derivative 3a (136 mg. 0.5 mmoles) in AcOEt was hydrogenated by using as the catalyst Pd/C (28.3 mg) and AcOH, for 48 hours at room temperature. After this time, the catalyst was filtered through celite and the solvent evaporated. The crude product 5 was used in the subsequent reaction without further purification. Yellow oil. (52% yield). Ή NM (CDCI3) δ: 3.83 (s, 2H, CH2); 4.73 (s, 2H, CH2CN); 6.63 (d, 2H, J = 8.4 Hz. Ar); 6.89 (d, 2H, J =8.4 Hz, Ar); 6.95 (d, 2H, J = 8.4 Hz, Ar); 7.14 (d. 2H, J = 8.4 Hz, Ar) ppm. Anal. (C | 5H14N20) C, H, N. % Calc. 75.61 (C); 5.92 (H); 1 1.76 (N). % Obs. 75.70 (C); 6.07 (H); 1 1 .61 (N). Synthesis of derivative 6
2-(4-(4-Hydroxybenzyl)phenoxy)acetomtrile (6). To a suspension of the amino-derivative 5 (63,0 mg, 0.26 mmoles) in H20 (0.46 ml), under stirring, cone. H2S04 (0,06 ml) was added dropwise. The mixture thus obtained was stirred at room temperature for 20 min. To this solution a NaN02 solution ( 17.9 mg, 0.26 mmoles) in H20 (0.19 ml) was added dropwise. The resulting solution was stirred at 100°C for 1 hr. After this time, the mixture was cooled, extracted with AcOEt. The organic layer was washed with NaCl, dehydrated, filtered and evaporated, to give the crude product 6. White solid. M.p.: 1 51 -1 53°C. (61 % yield). Ή NMR (CDCI3) δ: 3.87 (s, 2H, CH2); 4.74 (s. 2H, CH2CN); 6.76 (d, 2H, J - 8.4 Hz, Ar); 6.90 (d, 2H, J = 8.4 Hz, Ar); 7.02 (d. 2H, J = 8.4 Hz, Ar); 7.14 (d. 2H, J = 8.4 Hz, Ar) ppm. Anal. (C |5H,3N02) C, H, N. % Calc. 75.30 (C); 5.48 (H); 5.85 (N). % Obs. 74.92 (C); 5.48 (H); 6.13 (N).
Synthesis of derivative SG 7
4-(4-(2-Ami)ioethoxy)benzyi)phenol (SG_7). A solution of the cyano derivative 6 (32.3 mg; 0.13 mmoles) in THF was added to a LiAlH4 ( l .2 l mmoles) and AICI3 (161 mg; l .2 l mmoles) solution in THF, under nitrogen, left stirring at r.t. for 5 minutes. The reaction was stirred under reflux at 66°C for 12 hours. Thereafter, the mixture was cooled to 0°C, H20 was added, and the resulting mix was acidified with 10% HCl. The solution was then washed with Et20, the aqueous layer was alkalized with 2N NaOH, and CHC13 was added thereto. The resulting emulsion was filtered through a celite pad, and the organic layer was finally washed with saturated NaCl, dehydrated, filtered and evaporated under reduced pressure, providing the crude product SG_7 which was purified by formation of the hydrochloride. White solid. M.p.: 175- 177°C. (50% yield). Ή NMR (CD3OD): δ 3.34 (t. 2H, J = 4.8 Hz; CH2); 3.80 (s, 2H, CH2); 4.20 (t, 2H, J = 4.8 Hz; CH2NH2); 6.68 (d, 2H, J = 8.4 Hz, Ar); 6.91 (d, 2H, J = 8.8 Hz, Ar); 6.97 (d, 2H, J = 8.4 Hz, Ar); 7.1 1 (d, 2H, J = 8.8 Hz Ar) ppm. Anal. (C |5H|7N02) C, H, N. % Calc. 74.05 (C); 7.04 (H); 5.76 (N). % Obs. 74.31 (C); 7.22 (H); 5.73 (N).
SCHEME 4
Figure imgf000014_0001
7 8
Figure imgf000014_0002
d
Figure imgf000014_0003
e
9
Figure imgf000014_0004
SG 10 Reagents and conditions: a: KHF2, MeOH/H20, r.t., 30 min; b: 4-Nitrobenzyl bromide, PdCl2 dppf, Cs2C03, H20/Dioxane, 95°C, 24 hrs; c: SOCl2, CHCI3, r.t., 2hrs; d: NaCN, H20/CH3CN, 100°C, 150 W, 8 bars, 4 x 20 min cycles; e: LiAlH , AICI3, THF, reflux, 12 hrs; f: H2S04 50%. reflux, 30 min; g: Hydrazine hydrate, Charcoal, FeCl3, MeOH, reflux, 12 hrs.
Compounds SG_8, SG_9, SG_10 were synthesized by the procedure described in scheme 4. Through salification of the commercially available 4-hydroxymethylboronic acid with HF2, the derivative 7 was obtained, which was cross-coupled with 4-nitrobenzyl bromide to give compound 8. The subsequent chlorination reaction in the presence of SOCl2 provided the corresponding chloro-derivative 9, which by nucleophilic substitution with NaCN, gave the cyano-derivative 10. Reduction of 10 with L1AIH4 and in the presence of A1C13 provided the amino-compound SG_8, while its hydrolysis with H2S04 allowed for obtaining the acid SG_9. The subsequent reaction of derivative SG_9 with hydrazine hydrate in the presence of FeCl3 and charcoal provided the corresponding 4-(p- aminobenzyl)phenylacetic acid SG_10.
Synthesis of deriv ative 7 Potassium trifluoro[4-(hydroxymethyl)phenyl] -borate (7). KHF2 (2.7 g; 34.2 mmoles) and a few drops of H20 were added to a solution of 4-hydroxymethyl-phenylboronic acid ( 1.3 g; 8.55 mmoles) in MeOH. The mixture was stirred at room temperature for 30 minutes. After this time, the solvent was evaporated and the solid obtained was purified by crystallization from iPrOH, to give the desired derivative 5. White solid. (63% yield). Ή NMR (CD3OD): δ 4.53 (s, 2H. CH2); 7.18 (d. 2H, J = 7.2 Hz, Ar); 7.48 (d, 2H, J = 7.2 Hz, Ar) ppm. Anal. (C7H7BF3KO) C, H, N. % Calc. 39.28 (C); 3.30 (H). % Obs. 39.55 (C); 3.43 (H).
Synthesis of derivative 8
(4-(4-Nitroben∑yl)phenyl)methaiiol (8). p-Nitrobenzyl bromide (513 mg; 2.38 mmoles). cesium carbonate (2.30 g; 7.1 mmoles) and PdCl2dppf (34.8 mg; 0.05 mmoles) were added to a solution of the salt 7 (509 mg; 2.38 mmoles) in dioxane/H20 in a 9: 1 ratio (17 ml), under nitrogen. The mixture thus obtained was stirred at 95°C for 24 hours. Thereafter, the organic solvent was evaporated and the aqueous layer extracted with CH2CI2. The organic layer was dried, filtered and evaporated, giving a crude product, which was purified by column chromatography by using an n-Hexane/AcOEt (70:30) mixture as the eluent. White oil. (32% yield). Ή NMR (CDC13): δ 4.08 (s, 2H, CH2); 4.68 (s, 2H, CH2OH); 7.17 (d, 2H, J = 8.0 Hz Ar); 7.34-7.32 (m, 4H, Ar); 8.14 (d, 2H. J= 8.4 Hz, Ar) ppm. Anal. (C |4H,3N0 ) C. H, N. % Calc. 69.12 (C); 5.39 (H); 5.76 (N). % Obs. 69.03 (C); 5.63 (H); 5.58 (N).
Synthesis of derivative 9 l-(Chloromethyl)-4-(4-mtrobemyl)benzene (9). SOCl2 (0.009 ml) was added to a solution of 8 (86.4 mg; 0.35 mmoles) in CHCI3 at 0°C. The obtained mixture was stirred at room temperature for 2 hours. After this time, the solvent was evaporated and the obtained solid was taken up in H20 and alkalized with IN NaOH; the aqueous layer was extracted with CH2CI2. The organic layer was dried and evaporated to give the desired crude product 9 which was used in the subsequent reaction without further purification. Yellow oil. (83% yield). Ή NMR (CDC13): δ 4.08 (s. 2H, CH2); 4.57 (s, 2H, CH2); 7.1 7 (d, 2H. J = 8.0 Hz, Ar); 7.35-7.32 (m, 4H, Ar); 8.15 (d. 2H, J = 8.8 Hz Ar) ppm. Anal. (C 14H 12CINO2) C, H, N. % Calc. 64.25 (C); 4.62 (H); 5.35 (N). % Obs. 63.96 (C); 4.46 (H); 5.71 (N).
Synthesis of derivative 10 2-(4-(4-Nitrobenzyl)phenyl)acetowtrile (10). NaCN (57.0 mg; 1 .16 mmoles) in H20 (0.26 ml) was added to a solution of derivative 9 (152 mg; 0.58 mmoles) in CH3CN (0.78 ml). The mixture thus obtained was placed in a microwave oven, setting the following parameters: temperature 100°C; power 150 W; pressure 8 bars; time 20 minutes. After cooling, the reaction mixture was extracted with CH2C12 and the organic layer was dried and evaporated under reduced pressure, obtaining the desired crude product 10. Yellow oil. (86% yield). Ή NMR (CDCI3): δ 3.73 (s, 2H, CH2), 4.08 (s, 2H, CH2); 7.19 (d, 2H, J = 8.0 Hz; Ar); 7.33-7.28 (m. 4H. Ar); 8.15 (d, 2H, J = 8.8 Hz; Ar) ppm. Anal. (C |5H|2N202) C, H, N. % Calc. 71 .42 (C); 4.79 (H); 1 1 .10 (N). % Obs. 71 .56 (C); 4.64 (H); 1 1 .33 (N).
Synthesis of derivative SG_8
4-(4-(2-Aminoethyl)benzy\)aml\ne (SG_8). A solution of the cyano derivative 10 (85.0 mg; 0.34 mmoles) in THF was added to a LiAlH4 (3.04 mmoles) and A1C13 (405 mg; 3.04 mmoles) solution in THF, placed under nitrogen, left stirring at room temperature for 5 minutes. The reaction was stirred under reflux at 66°C for 12 hours. After this time, the mixture was cooled to 0°C, H20 was added, and the resulting mix was acidified with 10% HC1. The solution was then washed with Et20, the aqueous layer was alkalized with 2N NaOH, and CHCI3 was added thereto. The resulting emulsion was filtered through a celite pad and the organic layer was finally washed with saturated NaCl, dried, filtered and evaporated under reduced pressure, to give the crude product SG_8 which was purified by formation of the hydrochloride. White solid. M.p.: 165-167°C. (70% yield). Ή NMR (CD3OD): δ 2.93 (t. 2H. J = 7.8 Hz; CH2); 3.15 (t, 2H, J - 7.8 Hz; CH2); 4.02 (s, 2H, CH2); 7.25-7.20 (m, 4H, Ar); 7.33 (d, 2H,J = 8.4 Hz. Ar) 7.38 (d, 2H, J = 8.4 Hz, Ar) ppm. 13C NMR (CD3OD): 5 142.83, 139.53, 134.58, 130.20, 129.12, 128.67, 128.50, 122.71 , 40.54, 40.30. 32.75 ppm. Anal. (C ,5H|8N2) C. H, N. % Calc. 79.61 (C); 8.02 (H); 12.38 (N). % Obs. 79.45 (C); 8.18 (H); 12.77 (N).
Synthesis of derivative SG_9
2-(4-(4-Ni(roben∑y )p enyl)ace(ic acid (SG_9). A solution of 10 (51 .6 mg; 0.20 mmoles) in 50% H2SO4 (0.2 ml) was stirred under reflux for 30 minutes. After this time, the reaction mixture was set at 0°C, and H20 was added thereto; the precipitated solid was collected by filtration and washed with H20, providing the desired product SG_8. Yellow solid. M.p.: 175- 1 77°C. (79% yield). Ή NMR (CDCI3): δ 3.58 (s, 2H. CH2); 4.09 (s, 2H, CH2); 7.19 (d, 2H, J = 7.6 Hz, Ar); 7.37-7.27 (m, 4H, Ar); 8.17 (d, 2H, J = 7.6 Hz, Ar) ppm. Anal. (C 15H13NO4) C. H, N. % Calc. 66.41 (C); 4.83 (H); 5.16 (N). % Obs. 66.44 (C); 4.55 (H); 5.28 (N). Syothesis of derivative SG_10
2-(4-(4-Aminoben∑yl)phenyl)acetic acid (SG_10). Charcoal (33.7 mg) and FeClj (7 mg) were added to a solution of compound SG_8 (167 mg; 0.64 mmoles) solubilized in the minimum amount of MeOH (4 ml). The reaction mixture was heated under reflux for 15 minutes, then a hydrazine hydrate solution (0.33 ml, 6.71 mmoles) was added thereto, and the reaction was refluxed with stirring overnight. Thereafter, the mixture was filtered and the filtrate was evaporated. The residue was taken up with AcOEt and washed with H20; the organic layer was dried, filtered and evaporated, obtaining a crude product, which was purified by formation of the hydrochloride. White solid. M.p. : 154- 156°C. (72% yield). Ή NMR (CD3OD): δ 3.61 (s, 2H, CH2); 4.01 (s, 2H, CH2); 7.19 (d, 2H, J = 8.0 Hz, Ar); 7.26 (d, 2H. J = 8.0 Hz, Ar); 7.31 (d, 2H, J = 8.4 Hz, Ar); 7.37 (d, 2H, J = 8.4 Hz, Ar) ppm. Anal. (C 15H 13NO4) C. H. N. % Calc. 66.41 (C); 4.83 (H); 5.16 (N). % Obs. 66.12 (C); 5.18 (H); 5.09 (N).
Example 2: cardiac effects
The cardiac effects of thyronamine analogues falling within the scope of the present invention were assessed in acute treatment models.
The experimental model used is the isolated perfused rat heart with the working heart technique (Zucchi R, Ronca-Testoni S, Yu G, Galbani P, Ronca G, Mariani M. Effect of ischemia and reperfusion on cardiac ryanodine receptors - sarcoplasmic reticulum Ca2+ channels. Circ Res 1994: 74:271 -280), which allows for monitoring the main haemodynamic variables, such as: aortic flow, coronary flow, cardiac output, heart rate, aortic pressure.
After anaesthesia with a mixture of ether/air, the hearts of Sprague-Dawley rats (275-300 g of weight) were removed and perfused by using the working heart apparatus, in which the preload (height of the atrial chamber) and the afterload (height of the aortic chamber) were set at heights of 20 and 70 cm. respectively. The aortic and coronary flows were measured by collecting the perfusion liquid from the aortic and atrial chambers, respectively, into a graduated cylinder. The cardiac output was determined as the sum of the aortic and coronary flows. The perfusion medium was the rebs-Henseleit buffer solution (pH 7.4), which has the following composition (mmol/L): NaCl 1 1 8, NaHC03 25, C1 4.5, H2P04 1 .2, MgS04 1 .2, CaCl2 2.5, and glucose 1 ). The perfusion medium was kept in recirculation by a mechanical pump and was equilibrated with an 02 (95%)/C02 (5%) mixture. The temperature was maintained constant throughout the experiment at 37-37.4°C. After a 10 min equilibration time, the compounds (SG I and SG 2) were added to the perfusion medium, at concentrations within the range of from 100 nM to 100 microM, and the haemodynamic variables were monitored for 60 min.
As shown in Figure 1 , both compounds produced a dose-dependent negative inotropic effect (decrease in the cardiac output), with IC50 values (SG I , ^50= 0 microM; SG_2, IC50=20 microM) comparable to those obtained in previous experiments with administration of thyronamine (T0AM) and 3-iodothyronamine (T1 AM) [Scanlan et al, Nat Med 2004; 10: 638-642; Chiellini et al, FASEB J 2007; 21 , 1 597- 1608], The concentration-response curves for the effects of analogues SG_1 and SG_2 on the cardiac output are shown in Figure 1 .
Example 3: metabolic effects In male mice (CD 1 , weighing 25.2 +/- 3.2 g; Harlan-Nossan (Italy) fasted for 4 hrs before being subjected to an i.c.v injection (Manni et al, Br J Pharmacol 2013; 168(2):354-362) with saline (controls) or with one of the test compounds (T l A at doses of 1 .32 microgrKg- 1 and 4 microgrKg- 1 ; T0AM, T1 AM and SG I at 1 .32 microgrKg- 1 ), the glucose levels were measured in blood collected from the venous vessels of the tail. As shown in Figure 2, SG- 1 administered i.e. v. at 1 .32 microgr g- 1 was capable of producing a significant increase in plasma glucose, which was fully comparable to the one produced by the structurally analogous endogenous thyronamine (TOAM). Figure 2 shows the effects of SG I on glycaemia.
Example 4: agonistic TAAR 1 activity
In order to assess the effectiveness of the molecular design of the new SG set of compounds, such as novel analogues for TAAR1 receptors, HEK-293 cells transfected with the murine TAAR 1 receptor (mTAAR l ) were examined for the induction of cAMP production by the BRET (Bioluminescence Resonance Energy Transfer) technique according to the method described in literature (Espinoza S, Salahpour A, Masri B, Sotnikova TD, Messa M, Barak LS, Caron MG, Gainetdinov RR. Functional interaction between trace amine-associated receptor 1 and dopamine D2 receptor. Mol Pharmacol. 201 1 ; 80(3):416-25). After a first screening at 10 microM, the active compounds were used in dose-response experiments. As shown in Figure 3, the preliminary results demonstrated that the presence of an oxyethyl- or ethyl-amine chain at position 1 is critical for the occunence of the agonistic mTAARl activity, and compound SG 2 appears to be the most potent among the test compounds (SG I , 2, 7, 8) with an EC50 = 240 nM. Compound SG 8, although less potent than SG_2 (EC50 = 800 nM), appears to be the most effective among the tested set, with Emax = 100%, as calculated in comparison with the effect of 10 microM beta-phenylethylamine, which is the reference agonist for TAAR1 receptors. Figure 3 shows the dose-response curves for activation of mTAAR l . Particularly, it shows the differential cAMP levels by using the cAMP BRET biosensor in HE -293T cells that express mTAARl . The values on the Y-axis in each panel denote the Rluc/YFP ratio, which is known as the BRET ratio. The table that follows shows the preferred compounds of the present invention.
Figure imgf000021_0001

Claims

1 . A compound of formula (I):
Figure imgf000022_0001
wherein X is selected from O (oxygen) and CH2 ;
n is an integer comprised between 0 (zero) and 3;
Z is selected from NH2, N02 and OR3;
R2 is selected from the group consisting of CH2NH2, CH2NR4R"\ COOH, CN and COOR4; wherein R, R1 , R3, R4 and R5 are independently selected from H and a linear or branched alkyl group having from 1 to 4 carbon atoms and pharmaceutically acceptable salts thereof.
2. The compound according to claim 1 , wherein said pharmaceutically acceptable salts are selected from the group consisting of hydrochlorides, phosphates, sulfates, oxalates, tartrates, maleates, citrates and succinates.
3. The compound according to claim 1 or 2, wherein R and R1 are both H.
4. The compound according to claim 1 or 2, wherein R and R1 are both methyl.
5. The compound according to claim 1 or 2, wherein R is methyl and R is H.
6. The compound according to any one of claims 1 to 5, wherein n is zero or 1 .
7. The compound according to any one of claims 1 to 6, wherein R2 is CH2NH2 or
COOH.
8. The compound according to claim 1 , which is selected from the group consisting of: compound SG_ 1 , wherein R = R' = H; X = 0; n = l ; Z = NH2; R2 = CH2NH2; compound SG_2, wherein R = CH3; R1 = H; X = O; n = 1 ; Z = NH2; R2 = CH2NH2;
compound SG_3. wherein R = R' = H; X = 0; n = l ; Z = N02; R2 = COOH;
compound SG_4, wherein R = CH3; R1 = H; X = O; n = 1 ; Z = N02; R2 = COOH;
compound SG_5, wherein R = R' = H; X = 0; n = l ; Z = NH2; R2 - COOH:
compound SG_6, wherein R = methyl; R1 = H; X = O; n = 1 ; Z = NH2; R2 = COOH;
compound SG_7, wherein R = R1 = H; X = O; n = 1 ; Z = OH; R2 = CH2NH2;
compound SG_8. wherein R = R1 = H; X = CH2; n - 0; Z = NH2; R2 = CH2NH2;
compound SG_9, wherein R = R1 = H; X = CH2; n = 0; Z = N02; R2 = COOH; and compound SGJ O, wherein R = R1 = H; X = CH2; n - 0; Z = NH2; R2 = COOH.
9. The compound according to any one of claims 1 to 8, for use as a medicament.
10. The compound according to any one of claims 1 to 8, for use in the therapeutic treatment of a disease selected from the group consisting of obesity, dyshpidemias, hypothyroidism (optionally in association with the thyroid hormone), neuropsychiatric disorders, behaviour disorders, cardiovascular disorders, hormone disorders such as, for example, Polycystic ovary Syndrome (PCOS), ageing-related disorders.
1 1 . A pharmaceutical composition comprising at least one compound according to any one of claims 1 to 8 and a pharmaceutically acceptable carrier and/or excipient.
PCT/IB2015/052458 2014-04-03 2015-04-03 Synthetic analogues of 3-iodothyronamine (t1am) and uses thereof WO2015151068A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ITTO2014A000278 2014-04-03
ITTO20140278 2014-04-03

Publications (1)

Publication Number Publication Date
WO2015151068A1 true WO2015151068A1 (en) 2015-10-08

Family

ID=50928172

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2015/052458 WO2015151068A1 (en) 2014-04-03 2015-04-03 Synthetic analogues of 3-iodothyronamine (t1am) and uses thereof

Country Status (1)

Country Link
WO (1) WO2015151068A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT201900018893A1 (en) 2019-10-15 2021-04-15 Univ Pisa Compounds for use in the therapeutic and preventive treatment of neurodegenerative diseases, in particular Alzheimer's disease

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1137733B (en) * 1954-06-05 1962-10-11 Chem Fab Promonta G M B H Process for the preparation of aryl and thienylmethylphenylalkylamines
DE3718638A1 (en) * 1987-06-04 1988-12-22 Thomae Gmbh Dr K NEW PHENYLETHANOLAMINE, MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS AND METHOD FOR THE PRODUCTION THEREOF
US5409928A (en) * 1992-10-20 1995-04-25 Otsuka Pharmaceutical Co., Ltd. Condensed pyrazole derivatives, method of manufacturing the same, and androgen inhibitor
US5830920A (en) * 1995-05-05 1998-11-03 Hoffmann-La Roche Inc. Sulfuric acid esters of sugar alcohols
US20090105347A1 (en) * 2003-04-18 2009-04-23 The Regents Of The University Of California Thyronamine derivatives and analogs and methods of use thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1137733B (en) * 1954-06-05 1962-10-11 Chem Fab Promonta G M B H Process for the preparation of aryl and thienylmethylphenylalkylamines
DE3718638A1 (en) * 1987-06-04 1988-12-22 Thomae Gmbh Dr K NEW PHENYLETHANOLAMINE, MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS AND METHOD FOR THE PRODUCTION THEREOF
US5409928A (en) * 1992-10-20 1995-04-25 Otsuka Pharmaceutical Co., Ltd. Condensed pyrazole derivatives, method of manufacturing the same, and androgen inhibitor
US5830920A (en) * 1995-05-05 1998-11-03 Hoffmann-La Roche Inc. Sulfuric acid esters of sugar alcohols
US20090105347A1 (en) * 2003-04-18 2009-04-23 The Regents Of The University Of California Thyronamine derivatives and analogs and methods of use thereof

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
CHIELLINI ET AL., FASEB J, vol. 21, 2007, pages 1597 - 1608
ESPINOZA S; SALAHPOUR A; MASRI B; SOTNIKOVA TD; MESSA M; BARAK LS; CARON MG; GAINETDINOV RR: "Functional interaction between trace amine-associated receptor 1 and dopamine D2 receptor", MOL PHARMACOL., vol. 80, no. 3, 2011, pages 416 - 25
GAUTAM PANDA ET AL: "Effect of substituents on diarylmethanes for antitubercular activity", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 42, no. 3, 1 March 2007 (2007-03-01), pages 410 - 419, XP055137166, ISSN: 0223-5234, DOI: 10.1016/j.ejmech.2006.09.020 *
KARL KINDLER ET AL: "Studien über den Mechanismus chemischer Reaktionen XVIII. Katalytisch-spezifische Kondensationen mit Dihalogeniden, I Über die Umwandlung von aliphatisch-aromatischen Dichloriden in lokalanästhesierende Amine", JUSTUS LIEBIGS ANNALEN DER CHEMIE, vol. 617, no. 1, 30 October 1958 (1958-10-30), pages 25 - 54, XP055137298, ISSN: 0075-4617, DOI: 10.1002/jlac.19586170105 *
LORNE J. BRANDES ET AL: "Correlation of the Antiproliferative Action of Diphenylmethane-Derivative Antiestrogen Binding Site Ligands with Antagonism of Histamine Binding but not of Protein Kinase C-mediated Phosphorylation", CANCER RESEARCH, vol. 48, 1988, pages 3954 - 3958, XP002732586, ISSN: 0008-5472 *
MANNI ET AL., BR J PHARMACOL, vol. 168, no. 2, 2013, pages 354 - 362
RAO P ET AL: "Novel Functionalised Troger's Bases: Synthesis of a New Class of Troger's Base Analogues Containing Dicarboxyl Functionality", TETRAHEDRON LETTERS, PERGAMON, GB, vol. 37, no. 32, 5 August 1996 (1996-08-05), pages 5791 - 5794, XP004030540, ISSN: 0040-4039, DOI: 10.1016/0040-4039(96)01227-0 *
SCANLAN ET AL., NAT MED, vol. 10, 2004, pages 638 - 642
SCANLAN ET AL., NAT MED., vol. 10, no. 6, 16 May 2004 (2004-05-16), pages 638 - 42
ZUCCHI R; RONCA-TESTONI S; YU G; GALBANI P; RONCA G; MARIANI M: "Effect of ischemia and reperfusion on cardiac ryanodine receptors - sarcoplasmic reticulum Ca2+ channels", CIRC RES, vol. 74, 1994, pages 271 - 280

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT201900018893A1 (en) 2019-10-15 2021-04-15 Univ Pisa Compounds for use in the therapeutic and preventive treatment of neurodegenerative diseases, in particular Alzheimer's disease

Similar Documents

Publication Publication Date Title
US20200331859A1 (en) Salt of omecamtiv mecarbil and process for preparing salt
JP5161764B2 (en) Process for producing optionally 2-substituted 1,6-dihydro-6-oxo-4-pyrimidinecarboxylic acid
JP6118965B2 (en) Bicyclic compound
BR112020018562A2 (en) PREPARATIVE PROCESS
KR20020067589A (en) Phenylpiperazinyl derivatives
JPWO2013147117A1 (en) Fused azole derivatives
JP2000063363A (en) New triazole derivative
CZ2015233A3 (en) Process for preparing ixazomib citrate
AU2017303898A1 (en) Production method for pyrazole-amide compound
WO2000021916A1 (en) Process for the preparation of amine derivatives
EA010392B1 (en) Imidazole derivatives, the production thereof, and the use of the same as a medicament
WO2015151068A1 (en) Synthetic analogues of 3-iodothyronamine (t1am) and uses thereof
JPH0256471A (en) Novel (hetero)aryl substituted diazole derivative, its production and adaptation thereof to treatment
JP7182562B2 (en) Preparation of 2-([1,2,3]triazol-2-yl)-benzoic acid derivatives
CN113563319B (en) Indazole heterocyclic compounds having phosphodiesterase 4B inhibitory activity
CN108017522B (en) Preparation process of 2, 6-dibromobenzene methane sulfonyl chloride
JP2017525748A (en) Synthesis of cyclocreatine and its analogues
KR20200092945A (en) Lenalidomide Crystalline Form
WO2019018718A1 (en) Benzothiazole and pyridothiazole compounds as sumo activators
CN113512032B (en) Oxadiazole thioether derivative, and preparation method and application thereof
US8815870B2 (en) 4-(2-(6-substituted-hexylidene) hydrazinyl)benzonitrile and preparation thereof
AU646708B2 (en) Imidazol-2-yl derivatives of substituted bicyclic compounds and process for their preparation
JP2021531329A (en) Maleate of benzothiophene compound, its crystalline form and its use
DK155280B (en) ANALOGY PROCEDURE FOR THE PREPARATION OF INCIDENTAL DERIVATIVES
JPWO2003055847A1 (en) Carboxylic acid derivative

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15724022

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15724022

Country of ref document: EP

Kind code of ref document: A1