EP2632933A1 - Glycyrrhetinic acid amine analogues for use in the treatment of inflammation, infectious diseases, cancer, autoimmune diseases, skin diseases, bone diseases and metabolic diseases - Google Patents

Glycyrrhetinic acid amine analogues for use in the treatment of inflammation, infectious diseases, cancer, autoimmune diseases, skin diseases, bone diseases and metabolic diseases

Info

Publication number
EP2632933A1
EP2632933A1 EP11740948.2A EP11740948A EP2632933A1 EP 2632933 A1 EP2632933 A1 EP 2632933A1 EP 11740948 A EP11740948 A EP 11740948A EP 2632933 A1 EP2632933 A1 EP 2632933A1
Authority
EP
European Patent Office
Prior art keywords
norolean
urea
oxo
hydroxy
methyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP11740948.2A
Other languages
German (de)
French (fr)
Inventor
Dirk Classen-Houben
Bernhard Kueenburg
Paul Kosma
Ulrich Jordis
Laszlo Czollner
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Onepharm Research and Development GmbH
Original Assignee
Onepharm Research and Development GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Onepharm Research and Development GmbH filed Critical Onepharm Research and Development GmbH
Priority to EP11740948.2A priority Critical patent/EP2632933A1/en
Publication of EP2632933A1 publication Critical patent/EP2632933A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J63/00Steroids in which the cyclopenta(a)hydrophenanthrene skeleton has been modified by expansion of only one ring by one or two atoms
    • C07J63/008Expansion of ring D by one atom, e.g. D homo steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Definitions

  • This invention relates generally to the field of medicinal chemistry. More specifically, it relates to novel triterpene derivatives, preferably glycyrrhetinic acid derivatives that have pharmacologic activities, formulations containing such and their use to diagnose, cure or prevent certain diseases.
  • Licorice root is one of the most ancient medical plants being used in the traditional Chinese, Vietnamese, Indian and Arabian medicine.
  • the most important, and well-known bioactive component of licorice root is glycyrrhizin (GL), a natural product of the class of triterpene glycosides, also called saponins.
  • Glycyrrhetinic acid (GA) is the aglycone of GL and thus consists only of the triterpene part without the attached sugar molecules (see Figure 1 ).
  • a variety of pharmacological activities for GL and GA have been reported over the last decades comprising in vitro and in vivo studies. A good number of publications can be found in the field of steroid metabolism
  • GA has been widely reported as a potent inhibitor of intercellular gap-junctional communication most likely involving connexin43.
  • FIG. 1 Structure of Glycyrrhizin (GL) and Glycyrrhetinic acid (GA) Impact on Apoptosis and Oxidative Stress
  • GA is a potent inducer of mitochondrial permeability transition and can trigger the pro-apoptotic pathway.
  • GA is a potent inhibitor of bile acid-induced apoptosis and necrosis in a manner consistent with its antioxidative effect, significantly decreases neutrophil-generated oxygen species and inhibits the generation of inflammatory mediators. Below a certain concentration, GA prevents oxidative stress and
  • GA mitochondrial permeability transition but at higher concentrations GA induces oxidative stress in certain tissues. GA reveals also an effect on the protein expression of markers of oxidative stress (PAI-1 and p22 phox ) and scavenges oxygen free radicals in polymorphonuclear leukocytes (PMN). In addition GL stimulates DNA synthesis, proliferation in hepatocytes, and tyrosine phosphorylation of the EGF receptor and p42 MAP kinase.
  • PAI-1 and p22 phox markers of oxidative stress
  • PMN polymorphonuclear leukocytes
  • GL stimulates DNA synthesis, proliferation in hepatocytes, and tyrosine phosphorylation of the EGF receptor and p42 MAP kinase.
  • GL, GA and derivatives showed inhibition of replication, growth, proliferation or specific proteins of various viral and bacterial pathogens in vitro and in vivo.
  • examples include SARS-coronavirus replication, influenza A virus (H1 N1 , H2N2, H3N2), herpes simplex virus (HSV), hepatitis C virus, hepatitis A, HIV-1 -induced cytopathogenicity, hepatitis B virus (HBV), hyaluronate lyase from Streptococcus agalactiae, diverse species of periodontopathogenic and capnophilic bacteria, clarithromycin- and metronidazole-resistant strains of Heliobacter pylori, plaque formation in Japan encephalitis virus (JEV), vaccinia virus, Epstein-Barr virus (EBV), and Leishmania donovani.
  • GA and GL result in reduced levels of IL-10 and IL-4, but increased levels of IL-12, IFN-gamma, T
  • GA and GL treatment significantly reduces the increase of serum transaminases induced by D-galactosamine (GaIN), CCI4, or retrorsine.
  • GA inhibits the proliferation and collagen production of hepatic stellate cells (HSCs), down-regulates the mRNA expression of type III and I procollagen, and reduces the deposition of type III and I collagen in fibrotic liver.
  • HSCs hepatic stellate cells
  • GA also prevents the depletion of glutathione in the livers of CCI4-intoxicated mice and protects gel entrapped hepatocytes from tacrine toxicity.
  • GA treatment attenuates bile duct and hepatocyte damages in acute vanishing bile duct syndrome (AVBDS) rat model induced by a-naphthylisothiocyanate (ANIT).
  • AZADS acute vanishing bile duct syndrome
  • ANIT a-naphthylisothiocyanate
  • GA and GL inhibit secretory type IIA phospholipase A2 purified from the synovial fluids of patients with rheumatoid arthritis. GA inhibits the classical
  • complement C3 is a GL-binding protein and GA induces conformational changes in C3.
  • GA two trypsin-resistent fragments of C3a were immuno-precipitated with anti-C3a which could be selectively purified from the synovial fluids of patients with rheumatoid arthritis.
  • phosphorylation of C3a by CK-2 was completely inhibited by 30 ⁇ GA.
  • GL 100 ⁇
  • GA significantly improved bleeding on probing and gingival inflammation in a clinical study evaluating the local application of a paste containing GA.
  • the anti-inflammatory activity of GA is similar to hydrocortisone on formalin- induced arthritis in albino rats. Repeated treatment with GA significantly inhibits paw edema of rats with adjuvant arthritis (AA) and croton oil-induced mouse-ear-edema, decreases T-lymphocyte ratio, reduces proliferation of synovial cells and pannus formation, and eliminates the destruction of articular cartilage in inflamed joints of AA rat.
  • AA adjuvant arthritis
  • croton oil-induced mouse-ear-edema decreases T-lymphocyte ratio
  • reduces proliferation of synovial cells and pannus formation reduces the destruction of articular cartilage in inflamed joints of AA rat.
  • GA suppresses TNFa-induced IL-8 production through blockade in the phosphorylation of MAPKs, following ⁇ degradation and NFKB activation.
  • GL enhances interleukin-2 (IL-2) secretion and IL-2 receptor (IL-2R) expression.
  • IL-2R interleukin-2 receptor
  • GL promotes tyrosine phosphorylation of p56 induced by anti-CD3.
  • GL augments lipopolysaccharide (LPS)-induced IL-12 p40 mRNA expression, transcription of IL-12 mRNAs and IL-12-protein production.
  • LPS lipopolysaccharide
  • GL increases production of IL-10 in vitro and in mice with Con A-induced hepatitis.
  • GL inhibits prostaglandin E2 production and release of [3H]arachidonic acid.
  • GA lowers inflammatory capillary permeability, inhibits neutrophil emigration and prostaglandin E2 synthesis, and scavenges free radicals in a rat model of histamine, carrageenan, or ararachidonic acid-induced peritonitis.
  • GA dose-dependently increases NO production and iNOS mRNA through activation of protein/DNA binding of NF- ⁇ to its cognate site, enhances the production of nitric oxide from IFN- ⁇ activated cells and tumor cell killing by macrophages activated with IFN- ⁇ . This tumor cell killing is mainly by nitric oxide.
  • SDR Short Chain Dehydrogenase Reductases
  • GA is a potent non-competitive inhibitor of different hydroxysteroid
  • HSD dehydrogenases
  • GL and GA can bind to mineralocorticoid and glucocorticoid receptors with low but sufficient affinity in order to explain the mineralocorticoid-like side effects.
  • GA potentiates the action of aldosterone and facilitates the active transport of sodium in frog skin epithelium. GA stimulates an increase in steroid production in adrenal cells lacking intact cell junctions.
  • Examples include the reversible, gradual, constant and significant increase in systolic blood pressure, reduction in diuresis and increase in renal sodium retention, the reduction of thigh circumference and thickness of the subcutaneous fat layer in human volunteers after topical application, the reduction of metabolic detoxification of the cigarette smoke carcinogen nitrosamine 4-methylnitrosamino-1-(3-pyridyl)-1 -butanone (NNK), the involution of the thymus and thymocyte apoptosis, the potentiation of corticosteroid effects in cultured primary human bronchial epithelial cells (PBECs), ear swelling in dinitrofluorobenzene challenged mice, human volunteer skin
  • vasoconstrictor assay and lung tissue the retardation of the development of autoimmune disease, as well as the increased glucose use in subregions of the hypothalamus, hippocampus, neocortex and subthalamus.
  • ⁇ ⁇ ⁇ -HSD mRNA is expressed in neurones of the hypothalamic paraventricular nucleus (PVN) where corticotrophin-releasing factor-41 (CRF-41 ) is synthesized and GA decreases the release of CRF-41 into hypophysial portal blood in rats, suggesting that 1 1 ⁇ -HSD regulates the effective corticosterone feedback signal to CRF-41 neurons.
  • PVN hypothalamic paraventricular nucleus
  • GA inhibits oxidative stress DMBA TPA-induced skin tumor formation, inhibits ear edema and ornithine decarboxylase activity induced by croton oil in mice, protects against rapid DNA damage and decreases unscheduled DNA synthesis induced by benzo[a]pyrene, increases the antiproliferative effect of glucocorticoids In MCF-7 and ZR-75-1 breast cancer cells, reduces the tumor weight in rats transplanted with 'Oberling-Guerin' myeloma, inhibits proliferation of HepG2 human hepatoma cell line, inhibits the mutagenicity of benzo[a]pyrene, 2-aminofluorene and aflatoxin B1 , and protects against tumor initiation as well as tumor promotion by 7,12- dimethylbenz[a]anthracene (DMBA) and 12-0-tetradecanoylphorbol-13-acetate.
  • DMBA 7,12- dimethylbenz[a]anthracene
  • GA also increases the accumulation of calcein, a fluorescent substrate of multidrug resistance protein 1 (MRP1 ) and of daunorubicin, a fluorescent substrate of P-glycoprotein, resulting in sensitivity to anticancer drugs, showing that GA reverses multidrug resistance.
  • MRP1 multidrug resistance protein 1
  • daunorubicin a fluorescent substrate of P-glycoprotein
  • GA inhibits intercellular gap-junctional communication in human fibroblasts and cultured rat neonatal cardiomyocytes, as well as type 1 or type 2A protein
  • GA inhibits fluorescence replacement after photobleaching (FRAP) in primary chick osteocyte cultures, also indicating gap junction blockade.
  • GA increases the apparent cell input resistance and completely blocks membrane chloride conductance blocked while Na + and K + conductance are virtually unchanged.
  • GA in a concentration-dependent fashion attenuates EDHF-type relaxations to acetylcholine (ACh), observed in the presence of NG-nitro-L-arginine methyl ester (L-NAME) and indomethacin, modulates contractions produced by nor-adrenalin or high-K solutions and significantly reduces ACh-induced hyperpolarizations in both, endothelial and smooth muscle cells of guinea pig coronary and rat mesenteric arteries. Inhibition of the EDHF-hyperpolarization and relaxation in the smooth muscle may stem from the inhibition of endothelial cell hyperpolarization. GA quickly blocked electrical communication between smooth muscle and endothelial cells in guinea-pig mesenteric arterioles.
  • GA inhibits pressure-induced myogenic tone of rat middle cerebral arteries and vasopressin-induced vasoconstriction, increases input resistance in rat isolated mesenteric small arteries, desynchronised isolated smooth muscle cells, and had nonjunctional effects on membrane currents. GA significantly increases the frequency of phrenic bursts decreases the peak amplitude of integrated phrenic nerve discharge in an arterially perfused rat preparation.
  • GA inhibits the spike component of the action potential (AP), reduces
  • GA inhibits frequencies of paced contractions, likely owing to inhibition of l-type Ca 2+ channels, reduces the amplitudes of spontaneous and nerve-induced
  • GA also inhibits the spread of Lucifer yellow, increases input resistance, decreases cell capacitance in interstitial cells of Cajal networks and decreased L-type Ca 2+ current without affecting the voltage dependence of this current.
  • GA decreased the postsynaptic light response in murine retinal ganglion cells to 30 % of control.
  • GA reduces the bon resorption in rheumatoid arthritis and periodontits.
  • GA reduces coughing in guinea-pigs by 50 % compared to saline.
  • GA increases cytoplasmic free Ca 2+ and inhibits Ca 2+ increases induced by antigen, ATP, phenyephrine and thrombin. GA inhibits dexamethasone-induced increases in the histamine synthesis and histamine release. GA inhibits histidine decarboxylase and maturation of mast cells, lowers expression of PKC delta mRNA suggesting that the inhibition of histamine synthesis by GA is regulated by nPKC delta. GA significantly inhibits the degranulation of RBL-2H3 cells induced by IgE with the antigen (DNP-HSA) and rat peritoneal mast cells induced by compound 48/80. GA inhibits the passive cutaneous anaphylactic reaction as well as the scratching behaviour in mice induced by compound 48/80 and the production of IgE in ovalbumin- induced asthma mice.
  • GA sodium salt strongly counteracts arrhythmia induced by chloroform, lengthens the appearance time of arrhythmia induced by CaC ⁇ , slightly retards the heart rate of rats and rabbits, and partly antagonizes the acceleration effect of isoproterenol on rabbit hearts.
  • GA competitively inhibits the Na + /K + -ATPase of canine kidney basolateral membranes.
  • GA significantly increases insulin-stimulated glucose uptake in 3T3-L1 adipocytes, glucose-stimulated insulin secretion in islets isolated from mice and induces mRNA levels of insulin receptor substrate-2, pancreas duodenum homeobox-1 and glucokinase in islets.
  • New triterpene derivatives preferably glycyrrhetinic acid derivatives have been prepared and show desirable biological and pharmacologic activities relevant for the diagnosis, prevention and therapy of certain diseases.
  • one aspect of the present invention includes compounds having the following general structural formula I:
  • R 3a and R 3b are independently from one another selected from hydrogen
  • the configuration at position 18 is R or S;
  • R 30 is selected from hydrogen, -R a , or -O-R a ;
  • each R b independently of one another is selected from hydroxy or from optionally substituted, Ci- 6 alkyl, C 2-6 alkenyl, C 2-8 alkynyl,, -(CH 2 ) n -C 3 - 8 cycloalkyl,,-CF 3 ,
  • each n independently of one another denotes 0, 1 or 2;
  • R 30 is selected from hydrogen, Ci- 6 alkyl, C 2-6 alkenyl, C 2-8 alkynyl, -CF 3 , -(CH 2 ) n -C6-i 4 aryl, -(CH 2 ) n -C 5- i 4 heteroaryl, and -(CH 2 ) n -C 3 - 8 cycloalkyl.
  • Another aspect of the invention relates to compounds of general formula (I), wherein R 30 is hydrogen.
  • Another aspect of the invention relates to compounds of general formula (I), wherein R 3a is hydrogen and
  • Another aspect of the invention relates to compounds of general formula (I), wherein R 3b is selected from -OH, -O-acetyl, -O-succinyl, -NH 2 , -NH-acetyl, -NH- succinyl, -NH-S(O) 2 CF 3 , -NH-S(O) 2 CH 3 and -NH-S(O) 2 CH 2 CH 2 COOH.
  • Another aspect of the invention relates to compounds of general formula (I), wherein R 3b is hydrogen and
  • R 3a is selected from -OH, -O-acetyl, -O-succinyl, -NH 2 , -NH-acetyl, -NH- succinyl, -NH-S(O) 2 CF 3 , -NH-S(O) 2 CH 3 and -NH-S(O) 2 CH 2 CH 2 COOH.
  • Another aspect of the invention relates to a compound selected from the group consisting of
  • Another aspect of the invention relates to compounds of general formula (I), or the pharmacologically effective salts thereof, as medicaments.
  • the present invention includes a pharmaceutical preparation, containing as active substance one or more compounds of general Formula I, or the pharmacologically effective salts thereof, optionally in combination with conventional excipients and/or carriers.
  • the present invention includes the use of compounds of general Formula I for preparing a medicament for the treatment and/or prevention of chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer.
  • the present invention includes a use of a compound or a combination of compounds of the invention as a medicament or as a diagnostic.
  • the present invention includes use of a compound or a combination of compounds of the invention to treat a condition or disease that benefits from the inhibition of ⁇ ⁇ ⁇ -HSD isozymes.
  • the condition or disease that benefit from ⁇ ⁇ ⁇ -HSD inhibition are chronic inflammatory diseases.
  • a method of treating chronic inflammatory diseases comprising administering an effective amount of a compound of the invention to a subject in need thereof.
  • the invention includes a use of a compound of the invention to treat chronic inflammatory diseases, as well as a use of a compound of the invention to prepare a medicament to treat chronic inflammatory diseases.
  • the present invention includes a method of treating autoimmune diseases, comprising administering an effective amount of a compound of the invention to a subject in need thereof. Further the invention includes a use of a compound of the invention to treat autoimmune diseases, as well as a use of a compound of the invention to prepare a medicament to treat autoimmune diseases.
  • Also included within the scope of the present invention is a method of treating skin diseases, comprising administering an effective amount of a compound of the invention to a subject in need thereof. Further the invention includes a use of a compound of the invention to treat skin diseases, as well as a use of a compound of the invention to prepare a medicament to treat skin diseases.
  • the present invention also includes a method of treating metabolic diseases, comprising administering an effective amount of a compound of the invention to a subject in need thereof.
  • the invention also includes a use of an ⁇ ⁇ ⁇ -HSD inhibiting compound of the invention to treat metabolic diseases, as well as a use of an ⁇ ⁇ ⁇ -HSD inhibiting compound of the invention to prepare a medicament to treat and metabolic diseases.
  • An additional aspect of the present invention is a method of treating infectious diseases comprising administering an effective amount of a compound of the invention to a subject in need thereof. Also included in the present invention is a use of a compound of the invention to treat infectious diseases as well as a use of a compound of the invention to prepare a medicament to treat infectious diseases.
  • a further aspect of the present invention is a method of treating cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof. Also included in the present invention is a use of a compound of the invention to treat cancer as well as a use of a compound of the invention to prepare a medicament to treat cancer.
  • the compounds of the invention include compounds of Formula I as
  • alkyl when used alone or in combination with other groups or atoms, refers to a saturated straight or branched chain consisting solely of 1 to 6 hydrogen-substituted carbon atoms, and includes methyl, ethyl, propyl, isopropyl, n-butyl, 1 -methylpropyl, isobutyl, t-butyl, 2,2-dimethylbutyl, n-pentyl, 2- methylpentyl, 3-methylpentyl, 4-methylpentyl, n-hexyl and the like.
  • alkenyl refers to a partially unsaturated straight or branched chain consisting solely of 2 to 6 hydrogen-substituted carbon atoms that contains at least one double bond, and includes vinyl, allyl, 2-methylprop-1 - enyl, but-1 -enyl, but-2-enyl, but-3-enyl, buta-1 ,3-dienyl, penta-1 ,3-dienyl, penta-2,4- dienyl, 2-methylbut-1 -enyl, 2-methylpent-1-enyl, 4-methylpent-1 -enyl, 4-methylpent-2- enyl, 2-methylpent-2-enyl, 4-methylpenta-1 ,3-dienyl, hexen-1 -yl and the like.
  • alkynyl refers to a partially unsaturated straight or branched chain consisting solely of 2 to 8 hydrogen-substituted carbon atoms that contains at least one triple bond, and includes ethynyl, 1-propynyl, 2- propynyl, 2-methylprop-1 -ynyl, 1 -butynyl, 2-butynyl, 3-butynyl, 1 ,3-butadiynyl, 3- methylbut-1 -ynyl, 4-methylbut-ynyl, 4-methylbut-2-ynyl, 2-methylbut-1 -ynyl, 1 -pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1 ,3-pentadiynyl, 1 ,4-pentadiynyl, 3-methylpent-1 - ynyl, 4-methylpent-2-ynyl, 1-propyny
  • hydroxyalkyl refers to an aliphatic alkyl-, alkenyl-, or alkynyl-groups substituted with one or more hydroxyl groups, and includes 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4- hydroxybutyl, 2-hydroxy-1 -methyl-ethyl, 2-hydroxy-1 -ethyl-ethyl, 2,3-bihydroxypropyl, 2,3,4-trihydroxybutyl, 2-hydroxy-1 -hydroxymethyl-ethyl, 3-hydroxy-2-hydroxymethyl- propyl, 3-hydroxy-1 -(2-hydroxyethyl)-propyl, and the like.
  • carboxylic acid refers to an aliphatic mono- or dicarboxylic acid group containing from 2 to 18 carbon atoms that may optionally be substituted with one or more, identical or different substituents, independently selected from Ci- 4 alkyl, fluoro-substituted Ci -4 alkyl, halo, OCi -4 alkyl, fluoro-substituted OCi- 4 alkyl, aryl, heteroaryl, NH 2 , NH(alkyl), N(alkyl) 2 , C0 2 H, C0 2 (alkyl), N0 2 and CN.
  • carboxylic acids examples include formic acid, acetic acid, propionic acid, butyric acid, valeric acid, caproic acid, enanthic acid, caprylic acid, acrylic acid, pyruvic acid, acetoacetic acid, oxalic acid, malonic acid, malic acid, fumaric acid, succinic acid, glutaric acid, adipic acid, lactic acid, and the like.
  • carboxylic acid ester refers to alkylesters, arylesters, alkenylesters, alkynylester, cycloalkylesters, hydroxyalkylesters, and the like of above defined carboxylic acids.
  • carboxylic acid esters include methylesters, ethylesters, isopropylesters, t-butylesters, benzylesters,
  • cycloalkyl when used alone or in combination with other groups or atoms, refers to a saturated or unsaturated ring consisting solely of 3 to 8 carbon atoms, that may optionally be substituted with one or more, identical or different substituents, suitably one to three substituents,
  • Ci -4 alkyl independently selected from Ci -4 alkyl, fluoro-substituted Ci -4 alkyl, halo, OCi -4 alkyl, fluoro-substituted OCi -4 alkyl, NH 2 , NH(alkyl), N(alkyl) 2 , C0 2 H, C0 2 (alkyl), N0 2 and CN.
  • cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl and the like.
  • aryl refers to an aromatic mono- or bicyclic group containing from 6 to 14 carbon atoms that may be optionally fused with a fully or partially saturated or unsaturated carbocyclic ring and may optionally be substituted with one or more, identical or different substituents, suitably one to three substituents, independently selected from Ci -4 alkyl, fluoro-substituted Ci -4 alkyl, halo, OCi -4 alkyl, fluoro-substituted OCi -4 alkyl, NH 2 , NH(alkyl), N(alkyl) 2 , C0 2 H, C0 2 (alkyl), N0 2 and CN.
  • substituents suitably one to three substituents, independently selected from Ci -4 alkyl, fluoro-substituted Ci -4 alkyl, halo, OCi -4 alkyl, fluoro-substituted OCi -4 alkyl, NH 2 , NH(
  • heteroaryl refers to an aromatic mono- or bicyclic group containing from 5 to 14 carbon atoms, of which one to five is replaced with a heteroatom selected from N, S and O, that may optionally be reduced to a non- aromatic heterocycle and may optionally be substituted with one or more, identical or different substituents, suitably one to three substituents, independently selected from Ci -4 alkyl, fluoro-substituted Ci -4 alkyl, halo, OCi -4 alkyl, fluoro-substituted OCi -4 alkyl, NH 2 , NH(alkyl), N(alkyl) 2 , C0 2 H, C0 2 (alkyl), N0 2 and CN.
  • heteroaryl groups include pyrrolyl, dihydropyrrolyl, pyrrolidinyl, indolyl, isoindolyl, indolizinyl, imidazolyl, pyrazolyl, benzimidazolyl, imidazo(1 ,2-a)pyridinyl, indazolyl, purinyl, pyrrolo(2,3-c)pyridinyl, pyrrolo(3,2-c)pyridinyl, pyrrolo(2,3-b)pyridinyl, pyrazolo(1 ,5- a)pyridinyl, 1 ,2,3-triazolyl, 1 ,2,4-triazolyl, tetrazolyl, oxazolyl, isoxazolyl, 1 ,3,4- oxadiazolyl, 1 ,2,5-oxadiazolyl, 1 ,2,4-oxadiazolyl, 1 ,2,3-o
  • sugar when used alone or in combination with other groups or atoms, refers to naturally occurring or chemically modified carbohydrates that might be connected via glycosidic bonds to a free hydroxyl group of additional sugars and may optionally be substituted with one or more, identical or different substituents, suitably one to three substituents.
  • sugars include but are not limited to ⁇ -D-glucospyranose, a-L-rhamnopyranose, ⁇ -D-galactopyranose, ⁇ -D-xylopyranose, a-L-arabinofuranose, ⁇ -D-quinovopyranose, ⁇ -D-apiofuranose, gentibiose, chinovopyranose, fucopyranosyl, ⁇ -D-glucopyranosiduronic acid, ⁇ -D- galactopyranosiduronic acid, ⁇ -D-ribopyranose, ⁇ -D-xylofuranose, ⁇ -D-allopyranose, ⁇ -D-arabinopyranose, ⁇ -D-apiopyranose and the like.
  • fluoro-substituted means that, in the group being described, one or more, including all, of the hydrogen atoms has been replaced by F.
  • a fluoro-substituted alkyl includes
  • halogen and halo include F, CI, Br, and I.
  • point of attachment of the designated side chain is described first followed by the adjacent functionality toward the terminal portion.
  • a substituent's point of attachment may also be indicated by a dashed line to indicate the point(s) of attachment, followed by the adjacent functionality and ending with the terminal functionality.
  • R', R" and R'" is each independently selected from the group consisting of hydrogen, or optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl.
  • radicals can be selected independently from each other, the term independently means the radicals may be the same or may be different.
  • pharmaceutically acceptable means compatible with the treatment of animals, in particular, humans.
  • pharmaceutically acceptable salt includes both pharmaceutically acceptable acid addition salts and pharmaceutically acceptable basic addition salts.
  • pharmaceutically acceptable acid addition salt means any non-toxic organic or inorganic salt of any base compound of the disclosure, or any of its intermediates.
  • Basic compounds of the disclosure that may form an acid addition salt include, for example, compounds that contain a basic nitrogen atom.
  • Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric and phosphoric acids, as well as metal salts such as sodium monohydrogen
  • organic acids that form suitable salts include mono-, di-, and tricarboxylic acids such as glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, benzoic, phenylacetic, cinnamic and salicylic acids, as well as sulfonic acids such as p- toluene sulfonic and methanesulfonic acids.
  • Either the mono-, di- or the tri-acid salts can be formed, and such salts may exist in either a hydrated, solvated or substantially anhydrous form.
  • the acid addition salts of the compounds of the disclosure are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms.
  • the selection of the appropriate salt will be known to one skilled in the art.
  • Other non- pharmaceutically acceptable acid addition salts e.g. oxalates, may be used, for example, in the isolation of the compounds of the disclosure, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • pharmaceutically acceptable basic salt means any non-toxic organic or inorganic basic addition salt of any acid compound of the invention, or any of its intermediates, which are suitable for or compatible with the treatment of animals, in particular humans.
  • Acidic compounds of the invention that may form a basic addition salt include, for example compounds that contain carboxylic acid, sulfonic acid, sulfinic acid, sulfonamide, N-unsubstituted tetrazole, phosphoric acid ester, or sulfuric acid ester.
  • Illustrative inorganic bases which form suitable salts include lithium, sodium, potassium, calcium, magnesium, or barium hydroxide.
  • Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as methylamine, trimethylamine and picoline or ammonia.
  • the selection of the appropriate salt will be known to a person skilled in the art.
  • Other non-pharmaceutically acceptable basic addition salts may be used, for example, in the isolation of the compounds of the invention, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
  • the formation of a desired compound salt is achieved using standard techniques. For example, the neutral compound is treated with a base in a suitable solvent and the formed salt is isolated by filtration, extraction or any other suitable method.
  • subject or patient or synonym thereto includes all members of the animal kingdom, especially mammals, including human.
  • the subject or patient is suitably a human.
  • treatment is an approach for obtaining beneficial or desired results, including clinical results.
  • beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e. not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Palliating a disease or disorder means that the extent and/or undesirable clinical manifestations of a disorder or a disease state are lessened and/or time course of the progression is slowed or lengthened, as compared to not treating the disorder.
  • prevention or prophylaxis, or synonym thereto, as used herein refers to a reduction in the risk or probability of a patient becoming afflicted with the disease or manifesting a symptom associated with the disease.
  • therapeutically effective amount, effective amount or sufficient amount of a compound of the present invention is a quantity sufficient to, when administered to the subject, including a mammal, for example a human, effect beneficial or desired results, including clinical results, and, as such, an effective amount or synonym thereof depends upon the context in which it is being applied. For example, in the context of 1 1 ⁇ -HSD inhibition, it is an amount of the compound sufficient to achieve an inhibition of 1 1 ⁇ -HSD activity compared to the response obtained without administration of the compound.
  • therapeutically effective amounts of the compounds of the present invention are used to treat, modulate, attenuate, reverse, or affect a disease or conditions that benefits from an inhibition of 1 1 ⁇ -HSD, for example, chronic inflammatory diseases.
  • an effective amount is intended to mean that amount of a compound that is sufficient to treat, prevent or inhibit such diseases or conditions.
  • the amount of a given compound of the present invention that will correspond to such an amount will vary depending upon various factors, such as the given drug or compound, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art.
  • a therapeutically effective amount of a compound of the present invention is an amount which prevents, inhibits, suppresses or reduces a disease or conditions that benefits from an inhibition of 1 1 ⁇ -HSD, for example, chronic inflammatory diseases as determined by clinical symptoms in a subject as compared to a control.
  • a therapeutically effective amount of a compound of the present invention may be readily determined by one of ordinary skill by routine methods known in the art.
  • a therapeutically effective amount of a compound of the present invention ranges from about 0.01 to about 100 mg/kg body weight, suitably about 0.02 to about 50 mg/kg body weight, and more suitably, from about 0.05 to about 20 mg/kg body weight.
  • the skilled artisan will appreciate that certain factors may influence the dosage required to effectively treat a subject, or prevent a subject, suffering from a disease or conditions that benefits from an inhibition of 1 1 ⁇ -HSD activity, for example chronic inflammatory diseases, and these factors include, but are not limited to, the severity of the disease or disorder, previous treatments, the general health and/or age of the subject and other diseases present.
  • a treatment or prevention regime of a subject with a therapeutically effective amount of the compound of the present invention may consist of a single administration, or alternatively comprise a series of applications.
  • the compound of the present invention may be administered at least once a week.
  • the compound may be administered to the subject from about one time per week to about three times daily for a given treatment.
  • the length of the treatment period depends on a variety of factors, such as the severity of the disease, the age of the patient, the concentration and the activity of the compounds of the present invention, or a combination thereof.
  • the effective dosage of the compound used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required.
  • the term administered contemporaneously means that two substances are administered to a subject in such a way that they are both biologically active in the subject at the same time.
  • the exact details of the administration will depend on the pharmacokinetics of the two substances in the presence of each other, and can include administering one substance within 24 hours of administration of the other, if the pharmacokinetics is suitable. Designs of suitable dosing regimens are routine for one skilled in the art.
  • two substances will be administered substantially simultaneously, i.e. within minutes of each other, or in a single composition that comprises both substances.
  • 1 1 ⁇ -HSD activity is to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another conditions.
  • the term comprising and its derivatives are intended to be open ended terms that specify the presence of the stated features, elements, components, groups, integers, and/or steps, but do not exclude the presence of other unstated features, elements, components, groups, integers and/or steps.
  • the foregoing also applies to words having similar meanings such as the terms, including, having and their derivatives.
  • terms of degree such as substantially, about and approximately as used herein mean a reasonable amount of deviation of the modified term such that the end result is not significantly changed. These terms of degree should be construed as including a deviation of at least ⁇ 5% of the modified term if this deviation would not negate the meaning of the word it modifies.
  • a new class of compounds derived from glycyrrhetinic acid has been identified as drugs for the treatment of chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer.
  • the compounds according to the invention may be readily prepared by a person skilled in the art on the basis of his general knowledge. Results show, that glycyrrhetinic acid derivatives, namely compound 1 , inhibit both, ⁇ ⁇ ⁇ -HSDs enzyme activity and TNFa expression.
  • compound 1 and related compounds are a novel class of mechanism-based drugs against chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer that act as inhibitors of ⁇ ⁇ ⁇ -HSDs enzyme activity and TNFa expression.
  • the present invention includes a compound selected from a compound of Formula I:
  • R 3a and R 3b are independently from one another selected from hydrogen
  • the configuration at position 18 is R or S;
  • R 30 is selected from hydrogen, -R a , or -O-R a ;
  • each R b independently of one another is selected from hydroxy or from optionally substituted, Ci- 6 alkyl, C 2-6 alkenyl, C 2-8 alkynyl,, -(CH 2 ) n -C3- 8 cycloalkyl,,-CF3,
  • each n independently of one another denotes 0, 1 or 2;
  • the compounds of the invention may exist in a continuum of solid states ranging from fully amorphous to fully crystalline.
  • amorphous refers to a state in which the material lacks long range order at the molecular level and, depending upon temperature, may exhibit the physical properties of a solid or a liquid. Typically such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid.
  • crystalline refers to a solid phase in which the material has a regular ordered internal structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterized by a phase change, typically first order (melting point).
  • the compounds of the invention may also exist in unsolvated and solvated forms.
  • solvate is used herein to describe a molecular complex comprising the compound of the invention and one or more pharmaceutically acceptable solvent molecules, for example, ethanol.
  • solvent molecules for example, ethanol.
  • hydrate is employed when said solvent is water.
  • the complex When the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity. When, however, the solvent or water is weakly bound the water/solvent content will be dependent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm.
  • references to compounds of Formula I include references to salts, solvates, prodrugs and multi-component complexes thereof.
  • the compounds of Formula I can be prepared using methods known in the art, for example, 18a- and ⁇ ⁇ -glycyrrhetinic acid and their methyl esters may be converted into the corresponding dienones by reaction with 2-iodoxybenzoic acid as per a reported method [8].
  • the present invention includes radiolabeled forms of the compounds of the invention, for example, compounds of the invention labeled by incorporation within the structure 3 H, 11 C or 14 C or a radioactive halogen such as 125 l and 18 F.
  • a radiolabeled compound of the invention may be prepared using standard methods known in the art.
  • tritium may be incorporated into a compound of the invention using standard techniques, for example by hydrogenation of a suitable precursor to a compound of the invention using tritium gas and a catalyst.
  • a compound of the invention containing radioactive iodine may be prepared from the corresponding trialkyltin (suitably trimethyltin) derivative using standard iodination conditions, such as [ 125 l] sodium iodide in the presence of chloramine-T in a suitable solvent, such as dimethylformamide.
  • the trialkyltin compound may be prepared from the corresponding non-radioactive halo, suitably iodo, compound using standard palladium-catalyzed stannylation conditions, for example hexamethylditin in the presence of tetrakis
  • a compound of the invention containing a radioactive fluorine may be prepared, for example, by reaction of K[ 18 F]/K222 with a suitable precursor compound, such as a compound of Formula I comprising a suitable leaving group, for example a tosyl group, that may be displaced with the 18 F anion.
  • a suitable precursor compound such as a compound of Formula I comprising a suitable leaving group, for example a tosyl group, that may be displaced with the 18 F anion.
  • the present invention relates to novel compounds of Formula I, accordingly the present invention includes all uses of these compounds including, for example, in therapeutic and diagnostic applications.
  • the present invention accordingly includes the use of a compound or a combination of compounds of the invention as a medicament or as a diagnostic.
  • certain compounds of the invention are useful for treating any condition or disease that benefits from an inhibition of 1 1 ⁇ -HSD.
  • the conditions or diseases that benefit from an inhibition of 1 1 ⁇ -HSD are chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer.
  • the present invention includes a method of treating chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof.
  • the invention also includes a use of a compound of the invention to treat chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof.
  • the invention also includes a use of a compound of the invention to treat chronic
  • inflammatory diseases autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer and a use of a compound of the invention to prepare a medicament to treat chronic inflammatory diseases,
  • the chronic inflammatory disease is selected from stomatitis, gingivitis, periodontitis, peri-implantitis and osteoarthritis.
  • the autoimmune disease is selected from rheumatoid arthritis, systemic lupus erythematosus (SLE), Sjogren's syndrome, scleroderma, dermatomyositis, and polymyositis, inflammatory bowel diseases like Crohn's disease and ulcerative colitis.
  • the skin disease is selected from dermatitis, contact dermatitis, allergic dermatitis, atopic dermatitis, psoriasis, eczema, prurigo simplex acuta, prurigo simplex subacuta, prurigo nodularis, alopecia areata, Idiopathic thrombocytopenic purpura, pemphigus vulgaris, actinic keratosis.
  • the bone disease is selected from inflammation and/or immune mediated bone loss, osteoporosis, postmenopausal osteoporosis, Paget's disease, lytic bone metastases, arthritis, juvenile chronic arthritis, adjuvant arthritis, infectious diseases, bone loss by cancer, bone loss by HIV, tooth loss, bone marrow inflammation, synovial inflammation, cartilage and/or bone erosion and/or proteoglycan damage, osteopenie, osteosclerose, osteonecrosis.
  • the metabolic disease or disorder is selected from fasting hyperglycemia, diabetes mellitus, in particular insulin dependent type II diabetes, impaired fasting glucose, impaired glucose tolerance, insulin resistance, high blood pressure, central obesity (also known as visceral adiposity), decreased HDL cholesterol and elevated triglycerides.
  • the infectious disease is selected from viral, bacterial or fungal infections.
  • the cancer is selected from bladder cancer, breast cancer, colorectal cancer, cutaneous melanoma, skin cancer, squamous cell carcinoma of the skin, endometrial cancer, leukemia, lung cancer, non-Hodgkin lymphoma, ovarian cancer, pancreatic cancer, and prostate cancer.
  • the present invention also includes a method of treating chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof. Further the invention includes a use of a compound of the invention to treat chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer, as well as a use of a compound of the invention to prepare a medicament to treat these diseases.
  • the ⁇ ⁇ ⁇ -HSD inhibiting compound is compound 1.
  • a person skilled in the art would be able to identify ⁇ ⁇ ⁇ -HSD inhibiting compounds of the invention using, for example enzyme assays with mammalian 1 1 ⁇ - HSD isolated from specific tissue or expressed in transfected cell lines as described in the examples below and in literature [9-12].
  • the compounds of the invention are useful for treating any condition or disease that benefits from a downregulation in the expression or activity of TNFa.
  • the conditions or diseases that that benefit from a downregulation in the expression or activity of TNFa are inflammatory diseases, in particular rheumatoid arthritis, psoriasis, periodontitis, systemic lupus erythematosus (SLE), Sjogren's syndrome, scleroderma, dermatomyositis, and polymyositis, Crohn's disease and ulcerative colitis, asthma bronchiale.
  • a method of treating inflammatory diseases comprising administering a TNFa - downregulating effective amount of a compound of the invention to a subject in need thereof.
  • a person skilled in the art would be able to identify TNFa-downregulating compounds of the invention by contacting one or more cells with a compound of the invention and assaying for the presence of one TNFa and comparing the levels of TNFa in the one or more cells with that of controls. Such methods are known in the art [13,14] and are described in the examples herein below.
  • the compounds of the invention are suitably formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo.
  • the present invention includes a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable carrier or diluent.
  • compositions containing the compounds of the invention can be prepared by known methods for the preparation of pharmaceutically acceptable compositions which can be administered to subjects, such that an effective quantity of the active substance is combined in a mixture with a pharmaceutically acceptable vehicle.
  • compositions include, albeit not exclusively, solutions of the substances in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered solutions with a suitable pH and iso-osmotic with the physiological fluids.
  • the described compounds, salts or solvates thereof may be administered to a patient in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art.
  • the compositions of the invention may be administered, for example, by peroral, parenteral, buccal, sublingual, nasal, rectal, patch, pump or transdermal (topical) administration and the pharmaceutical compositions formulated accordingly.
  • Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.
  • a compound of the invention may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet.
  • the compound of the invention may be incorporated with excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • a compound of the invention may also be administered parenterally. Solutions of a compound of the invention can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, propylene glycol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. A person skilled in the art would know how to prepare suitable formulations.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersion and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. Ampoules are convenient unit dosages.
  • a pharmaceutical form suitable for injectable use includes sterile powders for the direct needle-free injection of the substance into the outer layer of skin in a simple-to-use-device.
  • compositions for nasal and pulmonary administration may conveniently be formulated as aerosols, drops, gels and powders.
  • Aerosol formulations typically comprise a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device.
  • the sealed container may be a unitary dispensing device such as a single dose inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal after use.
  • the dosage form comprises an aerosol dispenser, it will contain a propellant which can be a compressed gas such as compressed air or an organic propellant such as hydrofluoroalkanes.
  • the aerosol dosage forms can also take the form of a pump-atomizer. Where the dosage form comprises a dry powder inhaler, it can contain a propellant or rely on the force of patient inhalation to entrain powder from the device and subsequently break-up the powder into small aerosol particles
  • compositions suitable for buccal or sublingual administration include tablets, lozenges, pastilles, and patches wherein the active ingredient is formulated with a carrier such as sugar, acacia, tragacanth, or gelatin and glycerine.
  • Compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base such as cocoa butter.
  • Compositions for topical administration may include, for example, propylene glycol, isopropyl alcohol, mineral oil and glycerin.
  • Preparations suitable for topical administration include liquid or semi-liquid preparations such as liniments, lotions, applicants, and oil-in-water or water-in-oil emulsions such as creams, ointments or pastes; or solutions or suspensions such as drops.
  • suitable preparations for topical administration include patches wherein the ingredient is formulated with carriers such as adhesives, solvents, or polymers.
  • the topical preparations may include one or more additional ingredients such as diluents, buffers, flavoring agents, binders, surface active agents, thickeners, lubricants, preservatives, e.g. methyl hydroxybenzoate (including anti-oxidants), emulsifying agents and the like.
  • additional ingredients such as diluents, buffers, flavoring agents, binders, surface active agents, thickeners, lubricants, preservatives, e.g. methyl hydroxybenzoate (including anti-oxidants), emulsifying agents and the like.
  • Sustained or direct release compositions can be formulated, e.g. liposomes or those wherein the active compound is protected with differentially degradable coatings, such as by microencapsulation, multiple coatings, etc or those wherein the compound is imbedded into a polymer matrix such as polylactid or the like. It is also possible to freeze-dry the compounds of the invention and use the lyophilisates obtained, for example, for the preparation of products for injection.
  • the dosage of the compounds of Formula I and/or compositions of the invention can vary depending on many factors such as the pharmacodynamic properties of the compound, the mode of administration, the age, health and weight of the recipient, the nature and extent of the symptoms, the frequency of the treatment and the type of concurrent treatment, if any, and the clearance rate of the compound in the human or animal to be treated.
  • One of skill in the art can determine the appropriate dosage based on the above factors.
  • the compounds of Formula I may be administered initially in a suitable dosage that may be adjusted as required, depending on the clinical response.
  • the compounds of the invention may be administered to a subject alone or in combination with pharmaceutically acceptable carriers, as noted above, and/or with other pharmaceutically active agents for the treatment of chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer, the proportion of which is determined by the solubility and chemical nature of the compounds, chosen route of administration and standard pharmaceutical practice.
  • the compounds of Formula I, or salts or solvates thereof can be used alone or in combination with other agents or therapies that treat inflammatory and autoimmune diseases, for example, but not limited to, non-steroidal anti-inflammatory drugs
  • NAIDS neurodegenerative disease modifying anti-rheumatic drugs
  • DMARDs disease modifying anti-rheumatic drugs
  • TNFa blockers TNFa blockers
  • IL-1 interleukin 1 blockers
  • monoclonal antibodies against B cells T cell activation blocker.
  • the compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies that treat skin diseases, for example, but not limited to, corticosteroids, immunomodulating agents, retinoids, urea, zinc oxide, panthenol, antibiotics, and antimycotics.
  • agents or therapies that treat skin diseases, for example, but not limited to, corticosteroids, immunomodulating agents, retinoids, urea, zinc oxide, panthenol, antibiotics, and antimycotics.
  • the compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies that treat metabolic diseases, for example, but not limited to, sulfonylureas, meglitinides, biguanides, thiazolidinediones (TZDs), and a-glucosidase inhibitors.
  • metabolic diseases for example, but not limited to, sulfonylureas, meglitinides, biguanides, thiazolidinediones (TZDs), and a-glucosidase inhibitors.
  • the compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies that treat infectious diseases, for example, but not limited to, antibiotics like aminoglycosides, ansamycins,
  • carbacephem carbapenems, cephalosporins, glycopeptides, macrolides,
  • monobactams penicillins, polypeptides, quinolones, sulfonamides, and tetracyclines, antimycotics like polyenes, imidazoles, triazoles, allylamines, and echinocandins, and antivirals like cell entry blockers, nucleoside analogues, reverse transcriptase inhibitors, protease inhibitors, and neuraminidase inhibitors.
  • the compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies that treat cancer, for example, but not limited to, cytotoxic drugs, kinase inhibitors, antibodies and immunotherapy, selective receptor modulators, non-steroidal anti-inflammatory drugs (NSAIDS), corticosteroids, and enzyme modulators.
  • cytotoxic drugs for example, but not limited to, cytotoxic drugs, kinase inhibitors, antibodies and immunotherapy, selective receptor modulators, non-steroidal anti-inflammatory drugs (NSAIDS), corticosteroids, and enzyme modulators.
  • NSAIDS non-steroidal anti-inflammatory drugs
  • corticosteroids corticosteroids
  • NMR spectra were recorded at 297 K in CDCI3, pyridine-ds, DMSO-de or MeOD with a Bruker AC 200 spectrometer ( 1 H at 200.13 MHz, 13 C at 50.31 MHz), a Bruker DPX 300 spectrometer ( 1 H at 300.13 MHz, 13 C at 75.47 MHz), a Bruker AC 400 spectrometer ( 1 H at 400.13 MHz, 13 C at 100.61 MHz) and with a Bruker DPX 400 spectrometer ( 1 H at 400.13 MHz, 13 C at 100.61 MHz) using standard Bruker NMR software.
  • HPLC was performed using a Waters 2695 instrument with Merck Chromolith RP18 columns and a gradient of 3 % to 60 % acetonitrile and water containing 0.1 % formic acid at a flow of 1 .0 to 3.0 mL/min.
  • Preparative LC was performed using a Waters instrument with Merck Geminy RP18 columns and a gradient of 3 % to 60 % acetonitrile and water containing 0.1 % formic acid at a flow of 25 mL/min.
  • the HPLC reported purity is the number generated for the peak area as calculated using the Waters Millennium software with the Maxplot option for the UV maximum of the corresponding peak. Mass spectra were measure in Cl-mode with ammonia as a reagent gas on a thermo scientific.
  • the steroids were extracted with ethylacetate, separated by thin layer chromatography (TLC) (SIL G-25 UV254, Macherey-Nagel, Oensingen, Switzerland) using a solvent system of 9:1 (v/v) chloroform:methanol and the band corresponding to cortisone was excised. The product was run for a second
  • HEK-293 cells do not show ⁇ ⁇ ⁇ -HSDI activity. 1 i p-HSD2 mRNA was detectable by RT-PCR, but activity was not detectable upon incubation of radiolabeled Cortisol with cell lysate for 8 h.
  • HEK-293 cells were transfected with the plasmid for expression of carboxy-terminally FLAG-epitope tagged 1 1 ⁇ -HSDI or 1 1 ⁇ - HSD2, respectively, as described previously [18].
  • Transfected cells were selected by cultivation in presence of 800 ⁇ g/mL of G-418. Non-resistant cells were removed by replacing the cell culture medium every third day for 3 weeks. From these cells, eight clones each were then selected and tested for protein expression, by
  • HEK-293 cells stably transfected with either 1 1 ⁇ -HSDI or 1 i p-HSD2 were grown in 10 cm dishes to 90 % confluence. Cells were rinsed once with phosphate- buffered saline and resuspended in 2 mL of ice-cold buffer TS2 containing 100 mM NaCI, 1 mM EGTA, 1 mM EDTA, 1 mM MgCI 2 , 250 mM sucrose, 20 mM Tris-HCI, pH 7.4.
  • oxidative activity of 1 1 ⁇ -HSD enzymes cells were lysed by sonication and the cell lysate diluted 1 :12 in buffer TS2 (at 4 °C). Reactions were carried out in 96-well optical PCR reaction plates (Applied Biosystems, Foster City, CA) and tubes were capped during the reaction to avoid evaporation.
  • Reactions were started by simultaneously adding 10 ⁇ _ of cell lysate and 10 ⁇ _ of TS2 buffer containing the appropriate concentration of the compound to be tested to 10 ⁇ _ of TS2 buffer containing NAD+, 30 nCi of [1 ,2,6, 7-3H]-cortisol and unlabeled Cortisol to give a final concentration of 400 ⁇ NAD+ and 10 nM Cortisol.
  • Stock solutions of the compounds in methanol or in DMSO were diluted in TS2 buffer to yield the appropriate concentrations, whereby the concentration of methanol or DMSO in the reactions was below 0.1 %. Control reactions with or without 0.1 % of the solvent showed the same activity.
  • reductase activity was measured in a reaction containing NADPH, 30 nCi of [1 ,2,6, 7-3H]-cortisone and unlabeled cortisone, whereby final concentrations were 400 ⁇ NADPH and 10 nM cortisone.
  • No loss of 1 1 P-HSD2 activity was observed upon freezing of cell lysates for up to 1 month.
  • ⁇ ⁇ ⁇ -HSDI activity declined after cell disruption, with a concomitant loss of affinity for its substrate but without any significant loss of apparent Vmax. Activities were determined measuring the conversion of either radiolabeled cortisone or Cortisol for 5-20 min using substrate concentrations in the range between 10 nM and 10 ⁇ .
  • ⁇ ⁇ ⁇ -HSDI activities were measured immediately after cell disruption. All measurements included a negative control in absence of environmental compound and a positive control containing glycyrrhetinic acid at a final concentration of 10 ⁇ . Results are expressed as mean ⁇ S.D. and consist of at least three independent measurements.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Pain & Pain Management (AREA)
  • Transplantation (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Dermatology (AREA)
  • Steroid Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The present invention encompasses compounds of general formula (I) wherein R3a to R31 and X are defined as in claim 1, which are suitable for the treatment of and/or prevention of chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer.

Description

GLYCYRRHETINIC ACID AMINE ANALOGUES FOR USE IN THE TREATMENT OF INFLAMMATION, INFECTIOUS DISEASES, CANCER, AUTOIMMUNE DISEASES,
SKIN DISEASES, BONE DISEASES AND METABOLIC DISEASES
Field of the Invention
This invention relates generally to the field of medicinal chemistry. More specifically, it relates to novel triterpene derivatives, preferably glycyrrhetinic acid derivatives that have pharmacologic activities, formulations containing such and their use to diagnose, cure or prevent certain diseases.
Background of the Invention
Licorice root is one of the most ancient medical plants being used in the traditional Chinese, Tibetan, Indian and Arabian medicine. The most important, and well-known bioactive component of licorice root is glycyrrhizin (GL), a natural product of the class of triterpene glycosides, also called saponins. Glycyrrhetinic acid (GA) is the aglycone of GL and thus consists only of the triterpene part without the attached sugar molecules (see Figure 1 ). A variety of pharmacological activities for GL and GA have been reported over the last decades comprising in vitro and in vivo studies. A good number of publications can be found in the field of steroid metabolism
predominantly describing the inhibitory activity for 1 1 β-HSDs with various
pharmacological effects. GA has been widely reported as a potent inhibitor of intercellular gap-junctional communication most likely involving connexin43.
Furthermore, anti-inflammatory/immunemodulatory effects were reported suggesting several targets involved in the inflammatory process. Several papers report liver protective and anti-cancer properties whereas the impact on apoptosis/oxidative stress has been discussed controversially. Finally, antibiotic and antiviral effects have been reported comprising antibacterial effects on periodontopathogenic bacteria.
The pharmacology and toxicology of GL and GA has been comprehensively reviewed [1 -7].
Figure 1 : Structure of Glycyrrhizin (GL) and Glycyrrhetinic acid (GA) Impact on Apoptosis and Oxidative Stress
GA is a potent inducer of mitochondrial permeability transition and can trigger the pro-apoptotic pathway. GA is a potent inhibitor of bile acid-induced apoptosis and necrosis in a manner consistent with its antioxidative effect, significantly decreases neutrophil-generated oxygen species and inhibits the generation of inflammatory mediators. Below a certain concentration, GA prevents oxidative stress and
mitochondrial permeability transition but at higher concentrations GA induces oxidative stress in certain tissues. GA reveals also an effect on the protein expression of markers of oxidative stress (PAI-1 and p22phox) and scavenges oxygen free radicals in polymorphonuclear leukocytes (PMN). In addition GL stimulates DNA synthesis, proliferation in hepatocytes, and tyrosine phosphorylation of the EGF receptor and p42 MAP kinase.
Antibiotic and Antiviral Effects
GL, GA and derivatives showed inhibition of replication, growth, proliferation or specific proteins of various viral and bacterial pathogens in vitro and in vivo. Examples include SARS-coronavirus replication, influenza A virus (H1 N1 , H2N2, H3N2), herpes simplex virus (HSV), hepatitis C virus, hepatitis A, HIV-1 -induced cytopathogenicity, hepatitis B virus (HBV), hyaluronate lyase from Streptococcus agalactiae, diverse species of periodontopathogenic and capnophilic bacteria, clarithromycin- and metronidazole-resistant strains of Heliobacter pylori, plaque formation in Japan encephalitis virus (JEV), vaccinia virus, Epstein-Barr virus (EBV), and Leishmania donovani. In addition GA and GL result in reduced levels of IL-10 and IL-4, but increased levels of IL-12, IFN-gamma, TNF-alpha, and inducible NO synthase.
Liver Protective Effects
GA and GL treatment significantly reduces the increase of serum transaminases induced by D-galactosamine (GaIN), CCI4, or retrorsine. GA inhibits the proliferation and collagen production of hepatic stellate cells (HSCs), down-regulates the mRNA expression of type III and I procollagen, and reduces the deposition of type III and I collagen in fibrotic liver. GA also prevents the depletion of glutathione in the livers of CCI4-intoxicated mice and protects gel entrapped hepatocytes from tacrine toxicity.
GA treatment attenuates bile duct and hepatocyte damages in acute vanishing bile duct syndrome (AVBDS) rat model induced by a-naphthylisothiocyanate (ANIT).
Anti-Inflammatory and Immunmodulatory Effects
GA and GL inhibit secretory type IIA phospholipase A2 purified from the synovial fluids of patients with rheumatoid arthritis. GA inhibits the classical
complement pathway at the level of C2, complement C3 is a GL-binding protein and GA induces conformational changes in C3. In the presence of GA, two trypsin-resistent fragments of C3a were immuno-precipitated with anti-C3a which could be selectively purified from the synovial fluids of patients with rheumatoid arthritis. In addition, phosphorylation of C3a by CK-2 was completely inhibited by 30 μΜ GA. GL (100 μΜ) induces conformational changes in high mobility group box (HMGB)1 and 2 and completely inhibits the phosphorylation of HMGB1/2 by PKC and CK-I.
GA significantly improved bleeding on probing and gingival inflammation in a clinical study evaluating the local application of a paste containing GA.
The anti-inflammatory activity of GA is similar to hydrocortisone on formalin- induced arthritis in albino rats. Repeated treatment with GA significantly inhibits paw edema of rats with adjuvant arthritis (AA) and croton oil-induced mouse-ear-edema, decreases T-lymphocyte ratio, reduces proliferation of synovial cells and pannus formation, and eliminates the destruction of articular cartilage in inflamed joints of AA rat.
GA suppresses TNFa-induced IL-8 production through blockade in the phosphorylation of MAPKs, following ΙκΒα degradation and NFKB activation. GL enhances interleukin-2 (IL-2) secretion and IL-2 receptor (IL-2R) expression. In addition GL promotes tyrosine phosphorylation of p56 induced by anti-CD3. GL augments lipopolysaccharide (LPS)-induced IL-12 p40 mRNA expression, transcription of IL-12 mRNAs and IL-12-protein production. GL increases production of IL-10 in vitro and in mice with Con A-induced hepatitis. GL inhibits prostaglandin E2 production and release of [3H]arachidonic acid. GA lowers inflammatory capillary permeability, inhibits neutrophil emigration and prostaglandin E2 synthesis, and scavenges free radicals in a rat model of histamine, carrageenan, or ararachidonic acid-induced peritonitis. GA dose-dependently increases NO production and iNOS mRNA through activation of protein/DNA binding of NF-κΒ to its cognate site, enhances the production of nitric oxide from IFN-γ activated cells and tumor cell killing by macrophages activated with IFN-γ. This tumor cell killing is mainly by nitric oxide.
Anti-inflammatory activities of natural triterpenoids including GA have been reviewed recently.
Short Chain Dehydrogenase Reductases (SDR) and Corticoid Metabolism
GA is a potent non-competitive inhibitor of different hydroxysteroid
dehydrogenases (HSD). GA inhibits 1 1 β-HSD 1 and 11 β-HSD 2 involved in the metabolism of corticosteroids, 3a-HSD involved in inflammatory processes, 3α/β,20β- HSD involved in the metabolism of androgens and progestins, δβ-HSD involved in the metabolism of Cortisol, aldosterone and testosterone, and 3β-Η5ϋ involved in the metabolism of aldosterone and other steroids.
GL and GA can bind to mineralocorticoid and glucocorticoid receptors with low but sufficient affinity in order to explain the mineralocorticoid-like side effects. GA potentiates the action of aldosterone and facilitates the active transport of sodium in frog skin epithelium. GA stimulates an increase in steroid production in adrenal cells lacking intact cell junctions.
Especially the modification of corticosteroid levels by inhibition of 1 1 β-HSD 1 and 2 by GA has been connected to numerous biological states and diseases.
Examples include the reversible, gradual, constant and significant increase in systolic blood pressure, reduction in diuresis and increase in renal sodium retention, the reduction of thigh circumference and thickness of the subcutaneous fat layer in human volunteers after topical application, the reduction of metabolic detoxification of the cigarette smoke carcinogen nitrosamine 4-methylnitrosamino-1-(3-pyridyl)-1 -butanone (NNK), the involution of the thymus and thymocyte apoptosis, the potentiation of corticosteroid effects in cultured primary human bronchial epithelial cells (PBECs), ear swelling in dinitrofluorobenzene challenged mice, human volunteer skin
vasoconstrictor assay and lung tissue, the retardation of the development of autoimmune disease, as well as the increased glucose use in subregions of the hypothalamus, hippocampus, neocortex and subthalamus.
Ι ΐ β-HSD mRNA is expressed in neurones of the hypothalamic paraventricular nucleus (PVN) where corticotrophin-releasing factor-41 (CRF-41 ) is synthesized and GA decreases the release of CRF-41 into hypophysial portal blood in rats, suggesting that 1 1 β-HSD regulates the effective corticosterone feedback signal to CRF-41 neurons.
Anticancer Effects
GA inhibits oxidative stress DMBA TPA-induced skin tumor formation, inhibits ear edema and ornithine decarboxylase activity induced by croton oil in mice, protects against rapid DNA damage and decreases unscheduled DNA synthesis induced by benzo[a]pyrene, increases the antiproliferative effect of glucocorticoids In MCF-7 and ZR-75-1 breast cancer cells, reduces the tumor weight in rats transplanted with 'Oberling-Guerin' myeloma, inhibits proliferation of HepG2 human hepatoma cell line, inhibits the mutagenicity of benzo[a]pyrene, 2-aminofluorene and aflatoxin B1 , and protects against tumor initiation as well as tumor promotion by 7,12- dimethylbenz[a]anthracene (DMBA) and 12-0-tetradecanoylphorbol-13-acetate.
GA also increases the accumulation of calcein, a fluorescent substrate of multidrug resistance protein 1 (MRP1 ) and of daunorubicin, a fluorescent substrate of P-glycoprotein, resulting in sensitivity to anticancer drugs, showing that GA reverses multidrug resistance.
Gap Junction Blockade and Endothelial Relaxation
GA inhibits intercellular gap-junctional communication in human fibroblasts and cultured rat neonatal cardiomyocytes, as well as type 1 or type 2A protein
phosphatase-mediated Connexin43 dephosphorylation in WB-F344 rat liver epithelial cells. GA inhibits fluorescence replacement after photobleaching (FRAP) in primary chick osteocyte cultures, also indicating gap junction blockade.
GA increases the apparent cell input resistance and completely blocks membrane chloride conductance blocked while Na+ and K+ conductance are virtually unchanged.
GA in a concentration-dependent fashion attenuates EDHF-type relaxations to acetylcholine (ACh), observed in the presence of NG-nitro-L-arginine methyl ester (L-NAME) and indomethacin, modulates contractions produced by nor-adrenalin or high-K solutions and significantly reduces ACh-induced hyperpolarizations in both, endothelial and smooth muscle cells of guinea pig coronary and rat mesenteric arteries. Inhibition of the EDHF-hyperpolarization and relaxation in the smooth muscle may stem from the inhibition of endothelial cell hyperpolarization. GA quickly blocked electrical communication between smooth muscle and endothelial cells in guinea-pig mesenteric arterioles.
GA inhibits pressure-induced myogenic tone of rat middle cerebral arteries and vasopressin-induced vasoconstriction, increases input resistance in rat isolated mesenteric small arteries, desynchronised isolated smooth muscle cells, and had nonjunctional effects on membrane currents. GA significantly increases the frequency of phrenic bursts decreases the peak amplitude of integrated phrenic nerve discharge in an arterially perfused rat preparation.
GA inhibits the spike component of the action potential (AP), reduces
contraction evoked by electrical stimulation, inhibits slow depolarization with
superimposed APs and phasic contractions of the ureter induced by neurokinin A, and inhibits the KCI-evoked APs and phasic contractions without affecting the sustained responses in the guinea pig ureter.
GA inhibits frequencies of paced contractions, likely owing to inhibition of l-type Ca2+ channels, reduces the amplitudes of spontaneous and nerve-induced
contractions, decreases phasic contractions and depolarizes resting membrane potential in murine small intestinal muscles. GA also inhibits the spread of Lucifer yellow, increases input resistance, decreases cell capacitance in interstitial cells of Cajal networks and decreased L-type Ca2+ current without affecting the voltage dependence of this current.
GA decreased the postsynaptic light response in murine retinal ganglion cells to 30 % of control.
Other Effects
GA reduces the bon resorption in rheumatoid arthritis and periodontits.
GA reduces coughing in guinea-pigs by 50 % compared to saline.
GA increases cytoplasmic free Ca2+ and inhibits Ca2+ increases induced by antigen, ATP, phenyephrine and thrombin. GA inhibits dexamethasone-induced increases in the histamine synthesis and histamine release. GA inhibits histidine decarboxylase and maturation of mast cells, lowers expression of PKC delta mRNA suggesting that the inhibition of histamine synthesis by GA is regulated by nPKC delta. GA significantly inhibits the degranulation of RBL-2H3 cells induced by IgE with the antigen (DNP-HSA) and rat peritoneal mast cells induced by compound 48/80. GA inhibits the passive cutaneous anaphylactic reaction as well as the scratching behaviour in mice induced by compound 48/80 and the production of IgE in ovalbumin- induced asthma mice.
GA sodium salt strongly counteracts arrhythmia induced by chloroform, lengthens the appearance time of arrhythmia induced by CaC^, slightly retards the heart rate of rats and rabbits, and partly antagonizes the acceleration effect of isoproterenol on rabbit hearts.
GA competitively inhibits the Na+/K+-ATPase of canine kidney basolateral membranes.
GA significantly increases insulin-stimulated glucose uptake in 3T3-L1 adipocytes, glucose-stimulated insulin secretion in islets isolated from mice and induces mRNA levels of insulin receptor substrate-2, pancreas duodenum homeobox-1 and glucokinase in islets.
Summary of the Invention
New triterpene derivatives, preferably glycyrrhetinic acid derivatives have been prepared and show desirable biological and pharmacologic activities relevant for the diagnosis, prevention and therapy of certain diseases.
Accordingly, one aspect of the present invention includes compounds having the following general structural formula I:
wherein
R3a and R3b together are selected from =0, =NRa, =N-0-Ra, or
R3a and R3b are independently from one another selected from hydrogen,
-0-Ra, -0-C(=0)-Ra, -N(Ra)2, -NH-C(=0)-Ra and -NH-S(=0)2Ra; and
R11a and R11b together are selected from =0, =NRa, =NH, =N-0-Ra, =N-OH; R11a and R11b are independently from one another selected from hydrogen, -OH, -O-Ra, -O-C(=O)-Ra, -N(Ra)2, Ci-6alkyl, C2-8alkynyl, fluorine, chlorine, and bromine; and
a single or a double bond is present at position 12-13; and
the configuration at position 18 is R or S; and
X is selected from -C(=O)-1, -S(=O)2- and -C(=S)-; and
R30 is selected from hydrogen, -Ra, or -O-Ra; and
R31 is selected from -Rb, -NH2, -NHRb, -N(Rb)2, -NH-S(=O)2-Rb,
-NH-NH-C(=O)-NH2, -NH-NH-C(=O)-NHRb, -NH-NH-C(=O)-N(Rb)2; and each Ra independently of one another is selected from hydrogen or from optionally substituted hydroxyalkyl, Ci-6alkyl, C2-6alkenyl, C2-8alkynyl, -CF3, carboxylic acid, - (CH2)n-C6-i4aryl, -CH=CH-C6-i4aryl, -C≡C-C6-i4aryl, -(CH2)n-C5-i4heteroaryl, -CH=CH-C5-i4heteroaryl, -C≡C-C5-i4heteroaryl, -(CH2)n-C3-8cycloalkyl,
-CH=CH-C3-8cycloalkyl and -C≡C-C3-8cycloalkyl; and
each Rb independently of one another is selected from hydroxy or from optionally substituted, Ci-6alkyl, C2-6alkenyl, C2-8alkynyl,, -(CH2)n-C3-8cycloalkyl,,-CF3,
-(CH2)n-C6-i4aryl; and
each n independently of one another denotes 0, 1 or 2;
optionally in the form of their pharmaceutically effective salts, solvates, prodrugs, tautomers, racemates, enantiomers, diastereomers or mixtures thereof.
One aspect of the invention relates to compounds of general formula (I), wherein R11a and R11b together denote =O; and
a double bond is present at position 12-13.
Another aspect of the invention relates to compounds of general formula (I), wherein R30 is selected from hydrogen, Ci-6alkyl, C2-6alkenyl, C2-8alkynyl, -CF3, -(CH2)n-C6-i4aryl, -(CH2)n-C5-i4heteroaryl, and -(CH2)n-C3-8cycloalkyl.
Another aspect of the invention relates to compounds of general formula (I), wherein R30 is hydrogen.
Another aspect of the invention relates to compounds of general formula (I), wherein R3a is hydrogen and
R3b is selected from-O-Ra, -O-C(=O)-Ra, -N(Ra)2, -NH-C(=O)-Ra, and -NH- S(=O)2Ra. Another aspect of the invention relates to compounds of general formula (I), wherein R3b is selected from -OH, -O-acetyl, -O-succinyl, -NH2, -NH-acetyl, -NH- succinyl, -NH-S(O)2CF3, -NH-S(O)2CH3 and -NH-S(O)2CH2CH2COOH.
Another aspect of the invention relates to compounds of general formula (I), wherein R3b is hydrogen and
R3a is selected from -O-Ra, -O-C(=O)-Ra, -N(Ra)2, -NH-C(=O)-Ra, and -NH- S(=O)2Ra.
Another aspect of the invention relates to compounds of general formula (I), wherein R3a is selected from -OH, -O-acetyl, -O-succinyl, -NH2, -NH-acetyl, -NH- succinyl, -NH-S(O)2CF3, -NH-S(O)2CH3 and -NH-S(O)2CH2CH2COOH.
Another aspect of the invention relates to compounds of general formula (I), wherein R3a and R3b together are selected from =0, =N-methyl, =NH, =N-O-methyl, and =N-OH.
Another aspect of the invention relates to a compound selected from the group consisting of
Λ/-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-[(3p,18p,20P)-3-amino-1 1-oxo-30-norolean-12-en-20-yl] urea,
Λ/-[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, Λ/-[(3β, 18p,20P)-3-(methylsulfonamino)-11 -oxo-30-norolean-12-en-20-yl]urea,
/V-[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]urea,
/V-[(18p,20P)-3-(hydroxyimino)-1 1-oxo-30-norolean-12-en-20-yl] urea,
/V-hydroxy-/V-methyl-/V '-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/V '-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl] urea,
/V-hydroxy-/V-methyl-/V '-[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/V -[(3ββ, 18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V-methyl-/V '-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20- yl]urea, /V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/V '-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30- norolean-12-en-20-yl] urea
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(methylsulfonamino)-1 1 -oxo-30-norolean-12- en-20-yl]urea,
/V-hydroxy-/V-methyl-/ -[(18β,20β)-3, 1 1 -dioxo-30-norolean-12-en-20-yl]urea,
/V-hydroxy-/V-methyl-/ -[(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V -[(3p,18p,20P)-3-(acetyloxy)-1 1-oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V '-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea,
N-hydroxy-N -[(3p,18p,20P)-3-(acetamino)-1 1-oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V '-[(3β, 18p,20P)-3-amino -1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-hydroxy-/V -[(3β, 18p,20P)-3-(succinyloxy)-11 -oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V -[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V -[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-11 -oxo-30-norolean-12-en- 20-yl]urea,
/V-hydroxy-N'-[(3p, 18p,20P)-3-(methylsulfonamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V -[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]urea,
N-hydroxy-N -[(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/ -[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-methyl-/ -[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-methyl-/ -[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, N-methyl-/V -[(3p,18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]urea,
N-methyl-N -[(3p,18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/V '-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/V -[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-11 -oxo-30-norolean-12-en- 20-yl]urea,
N-methyl-N -[(3p,18p,20P)-3-(methylsulfonamino)-1 1-oxo-30-norolean-12-en-20- yl]urea,
/V-methyl-/V -[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]urea,
N-methyl-N -[(18p,20P)-3-(hydroxyimino)-11 -oxo-30-norolean-12-en-20-yl]urea, /V-methylsulfonyl-/ -[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea,
N-methylsulfonyl-/V -[(3p,18p,20P)-3-(acetamino)-1 1-oxo-30-norolean-12-en-20- yl]urea,
/V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-methylsulfonyl-N '-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30-norolean- 12-en-20-yl]urea,
N-methylsulfonyl-N -[(3p,18p,20P)-3-(methylsulfonamino)-11 -oxo-30-norolean-12-en- 20-yl]urea,
/V-methylsulfonyl-/ -[(18β,20β)-3, 1 1 -dioxo-30-norolean-12-en-20-yl]urea,
N-methylsulfonyl-N -[(18p,20P)-3-(hydroxyimino)-1 1-oxo-30-norolean-12-en-20-yl]urea, Λ/-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl )sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl )sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl )sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(methylsulfonamino)-11 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl)sulfonyl]urea,
/V-[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]-/V -[(trifluoromethyl)sulfonyl]urea,
/V-[(18p,20P)-3-(hydroxyimino)-1 1-oxo-30-norolean-12-en-20-yl]-/V -
[(trifluoromethyl)sulfonyl]urea,
(3β, 18p,20P)-3-hydroxy-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-(acetyloxy)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-amino-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-(acetamino)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-(succinyloxy)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-11 -one, (3β, 18p,20P)-3-(succinylamino)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 - one,
(3β, 18p,20P)-3-(trifluoromethylsulfonam
12-en-1 1 -one,
(3β, 18p,20P)-3-(methylsulfonamino)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en- 1 1 -one,
(18β,20β)-3, 1 1 -dioxo-30-norolean-12-en-20-yl]urea,
(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-/ -(phenylmethyl) urea, and
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-1 ,2-hydrazine- dicarboxamide.
Another aspect of the invention relates to compounds of general formula (I), or the pharmacologically effective salts thereof, as medicaments.
In a further aspect, the present invention includes a pharmaceutical preparation, containing as active substance one or more compounds of general Formula I, or the pharmacologically effective salts thereof, optionally in combination with conventional excipients and/or carriers.
In a further aspect, the present invention includes the use of compounds of general Formula I for preparing a medicament for the treatment and/or prevention of chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer.
In a further aspect, the present invention includes a use of a compound or a combination of compounds of the invention as a medicament or as a diagnostic.
In a further aspect, the present invention includes use of a compound or a combination of compounds of the invention to treat a condition or disease that benefits from the inhibition of Ι ΐ β-HSD isozymes. In particular embodiments the condition or disease that benefit from Ι ΐ β-HSD inhibition are chronic inflammatory diseases.
Accordingly, also included within the scope of the present invention is a method of treating chronic inflammatory diseases, comprising administering an effective amount of a compound of the invention to a subject in need thereof. Further the invention includes a use of a compound of the invention to treat chronic inflammatory diseases, as well as a use of a compound of the invention to prepare a medicament to treat chronic inflammatory diseases.
In a further aspect, the present invention includes a method of treating autoimmune diseases, comprising administering an effective amount of a compound of the invention to a subject in need thereof. Further the invention includes a use of a compound of the invention to treat autoimmune diseases, as well as a use of a compound of the invention to prepare a medicament to treat autoimmune diseases.
Also included within the scope of the present invention is a method of treating skin diseases, comprising administering an effective amount of a compound of the invention to a subject in need thereof. Further the invention includes a use of a compound of the invention to treat skin diseases, as well as a use of a compound of the invention to prepare a medicament to treat skin diseases.
The present invention also includes a method of treating metabolic diseases, comprising administering an effective amount of a compound of the invention to a subject in need thereof. The invention also includes a use of an Ι ΐ β-HSD inhibiting compound of the invention to treat metabolic diseases, as well as a use of an Ι ΐ β-HSD inhibiting compound of the invention to prepare a medicament to treat and metabolic diseases.
An additional aspect of the present invention is a method of treating infectious diseases comprising administering an effective amount of a compound of the invention to a subject in need thereof. Also included in the present invention is a use of a compound of the invention to treat infectious diseases as well as a use of a compound of the invention to prepare a medicament to treat infectious diseases.
A further aspect of the present invention is a method of treating cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof. Also included in the present invention is a use of a compound of the invention to treat cancer as well as a use of a compound of the invention to prepare a medicament to treat cancer.
Other features and advantages of the present invention will become apparent from the detailed description. It should be understood, however, that the detailed description and the specific examples while indicating preferred embodiments of the invention are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
Detailed Description of the Invention
The compounds of the invention include compounds of Formula I as
hereinbefore defined, including all polymorphs and crystal habits thereof, salts, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labeled compounds of Formula I.
Unless specified otherwise, the term alkyl, when used alone or in combination with other groups or atoms, refers to a saturated straight or branched chain consisting solely of 1 to 6 hydrogen-substituted carbon atoms, and includes methyl, ethyl, propyl, isopropyl, n-butyl, 1 -methylpropyl, isobutyl, t-butyl, 2,2-dimethylbutyl, n-pentyl, 2- methylpentyl, 3-methylpentyl, 4-methylpentyl, n-hexyl and the like.
Unless specified otherwise, the term alkenyl refers to a partially unsaturated straight or branched chain consisting solely of 2 to 6 hydrogen-substituted carbon atoms that contains at least one double bond, and includes vinyl, allyl, 2-methylprop-1 - enyl, but-1 -enyl, but-2-enyl, but-3-enyl, buta-1 ,3-dienyl, penta-1 ,3-dienyl, penta-2,4- dienyl, 2-methylbut-1 -enyl, 2-methylpent-1-enyl, 4-methylpent-1 -enyl, 4-methylpent-2- enyl, 2-methylpent-2-enyl, 4-methylpenta-1 ,3-dienyl, hexen-1 -yl and the like.
Unless specified otherwise, the term alkynyl refers to a partially unsaturated straight or branched chain consisting solely of 2 to 8 hydrogen-substituted carbon atoms that contains at least one triple bond, and includes ethynyl, 1-propynyl, 2- propynyl, 2-methylprop-1 -ynyl, 1 -butynyl, 2-butynyl, 3-butynyl, 1 ,3-butadiynyl, 3- methylbut-1 -ynyl, 4-methylbut-ynyl, 4-methylbut-2-ynyl, 2-methylbut-1 -ynyl, 1 -pentynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1 ,3-pentadiynyl, 1 ,4-pentadiynyl, 3-methylpent-1 - ynyl, 4-methylpent-2-ynyl, 4-methylpent-2-ynyl, 1 -hexynyl, and the like.
Unless specified otherwise, the term hydroxyalkyl refers to an aliphatic alkyl-, alkenyl-, or alkynyl-groups substituted with one or more hydroxyl groups, and includes 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4- hydroxybutyl, 2-hydroxy-1 -methyl-ethyl, 2-hydroxy-1 -ethyl-ethyl, 2,3-bihydroxypropyl, 2,3,4-trihydroxybutyl, 2-hydroxy-1 -hydroxymethyl-ethyl, 3-hydroxy-2-hydroxymethyl- propyl, 3-hydroxy-1 -(2-hydroxyethyl)-propyl, and the like.
Unless specified otherwise, the term carboxylic acid refers to an aliphatic mono- or dicarboxylic acid group containing from 2 to 18 carbon atoms that may optionally be substituted with one or more, identical or different substituents, independently selected from Ci-4alkyl, fluoro-substituted Ci-4alkyl, halo, OCi-4alkyl, fluoro-substituted OCi- 4alkyl, aryl, heteroaryl, NH2, NH(alkyl), N(alkyl)2, C02H, C02(alkyl), N02 and CN.
Examples of carboxylic acids include formic acid, acetic acid, propionic acid, butyric acid, valeric acid, caproic acid, enanthic acid, caprylic acid, acrylic acid, pyruvic acid, acetoacetic acid, oxalic acid, malonic acid, malic acid, fumaric acid, succinic acid, glutaric acid, adipic acid, lactic acid, and the like.
Unless specified otherwise, the term carboxylic acid ester refers to alkylesters, arylesters, alkenylesters, alkynylester, cycloalkylesters, hydroxyalkylesters, and the like of above defined carboxylic acids. Examples of carboxylic acid esters include methylesters, ethylesters, isopropylesters, t-butylesters, benzylesters,
benzhydrylesters, allylesters, and the like.
Unless specified otherwise, the term cycloalkyl, when used alone or in combination with other groups or atoms, refers to a saturated or unsaturated ring consisting solely of 3 to 8 carbon atoms, that may optionally be substituted with one or more, identical or different substituents, suitably one to three substituents,
independently selected from Ci-4alkyl, fluoro-substituted Ci-4alkyl, halo, OCi-4alkyl, fluoro-substituted OCi-4alkyl, NH2, NH(alkyl), N(alkyl)2, C02H, C02(alkyl), N02 and CN. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl and the like.
Unless specified otherwise, the term aryl refers to an aromatic mono- or bicyclic group containing from 6 to 14 carbon atoms that may be optionally fused with a fully or partially saturated or unsaturated carbocyclic ring and may optionally be substituted with one or more, identical or different substituents, suitably one to three substituents, independently selected from Ci-4alkyl, fluoro-substituted Ci-4alkyl, halo, OCi-4alkyl, fluoro-substituted OCi-4alkyl, NH2, NH(alkyl), N(alkyl)2, C02H, C02(alkyl), N02 and CN. Examples of aryl groups include phenyl, naphthyl, indanyl, and the like.
Unless specified otherwise, the term heteroaryl refers to an aromatic mono- or bicyclic group containing from 5 to 14 carbon atoms, of which one to five is replaced with a heteroatom selected from N, S and O, that may optionally be reduced to a non- aromatic heterocycle and may optionally be substituted with one or more, identical or different substituents, suitably one to three substituents, independently selected from Ci-4alkyl, fluoro-substituted Ci-4alkyl, halo, OCi-4alkyl, fluoro-substituted OCi-4alkyl, NH2, NH(alkyl), N(alkyl)2, C02H, C02(alkyl), N02 and CN. Examples of heteroaryl groups include pyrrolyl, dihydropyrrolyl, pyrrolidinyl, indolyl, isoindolyl, indolizinyl, imidazolyl, pyrazolyl, benzimidazolyl, imidazo(1 ,2-a)pyridinyl, indazolyl, purinyl, pyrrolo(2,3-c)pyridinyl, pyrrolo(3,2-c)pyridinyl, pyrrolo(2,3-b)pyridinyl, pyrazolo(1 ,5- a)pyridinyl, 1 ,2,3-triazolyl, 1 ,2,4-triazolyl, tetrazolyl, oxazolyl, isoxazolyl, 1 ,3,4- oxadiazolyl, 1 ,2,5-oxadiazolyl, 1 ,2,4-oxadiazolyl, 1 ,2,3-oxadiazolyl, thiazolyl, isothiazolyl, 1 ,3,4-thiadiazolyl, 1 ,2,5-thiadiazolyl, 1 ,2,4-thiadiazolyl, 1 ,2,3-thiadiazolyl, furanyl, dihydrofuranyl, tetrahydrofuranyl, benzofuranyl, isobenzofuranyl, thiophenyl, dihydrothiophenyl, tetrahydrothiophenyl, benzothiophenyl, benzoisothiophenyl, pyridyl, piperidinyl, quinolinyl, isoquinolinyl, quinolizinyl, pyrazinyl, pyridazinyl, pyrimidinyl, pyranyl, tetrahydropyranyl, 1 ,2,3-triazinyl, 1 ,2,4-triazinyl, 1 ,3,5-triazinyl, chromenyl, morpholinyl, diazepinyl, benzodiazepinyl, and the like.
Unless specified otherwise, the term sugar, when used alone or in combination with other groups or atoms, refers to naturally occurring or chemically modified carbohydrates that might be connected via glycosidic bonds to a free hydroxyl group of additional sugars and may optionally be substituted with one or more, identical or different substituents, suitably one to three substituents. Examples of sugars include but are not limited to β-D-glucospyranose, a-L-rhamnopyranose, β-D-galactopyranose, β-D-xylopyranose, a-L-arabinofuranose, β-D-quinovopyranose, β-D-apiofuranose, gentibiose, chinovopyranose, fucopyranosyl, β-D-glucopyranosiduronic acid, β-D- galactopyranosiduronic acid, β-D-ribopyranose, β-D-xylofuranose, β-D-allopyranose, β-D-arabinopyranose, β-D-apiopyranose and the like.
Unless specified otherwise, the term fluoro-substituted as used herein means that, in the group being described, one or more, including all, of the hydrogen atoms has been replaced by F. For example, a fluoro-substituted alkyl includes
trifluoromethyl, trifluoroethyl, pentafluoroethyl and the like.
Unless specified otherwise, as used herein, the terms halogen and halo include F, CI, Br, and I. Under standard nomenclature rules used throughout this disclosure, the point of attachment of the designated side chain is described first followed by the adjacent functionality toward the terminal portion. A substituent's point of attachment may also be indicated by a dashed line to indicate the point(s) of attachment, followed by the adjacent functionality and ending with the terminal functionality.
The term optionally substituted in each instance if not further specified refers to between 1 and 10 substituents, e.g. 1 ,2,3,4,5,6,7,8,9, or 10 substituents which are in each instance independently selected form the group consisting of halogen, in particular F, CI, Br, or I; N02, CN, OR', NR'R", C(=O)OR', C(=O)NR'R", OC(=O)R', NR'C(=O)R", OC(=O)OR', NR'C(=O)NR"R"', NR'S(=O)2R", C(=O)R', S(=O)2NR'R", CR'R'OH, and R';
R', R" and R'" is each independently selected from the group consisting of hydrogen, or optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl.
If two or more radicals can be selected independently from each other, the term independently means the radicals may be the same or may be different.
It is intended that the definition of any substituent or variable at a particular location in a molecule be independent of its definitions elsewhere in that molecule. It is understood that substituents and substitution patterns on the compounds of this invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art as well as those methods set forth herein.
The term pharmaceutically acceptable means compatible with the treatment of animals, in particular, humans. The term pharmaceutically acceptable salt includes both pharmaceutically acceptable acid addition salts and pharmaceutically acceptable basic addition salts.
The term pharmaceutically acceptable acid addition salt as used herein means any non-toxic organic or inorganic salt of any base compound of the disclosure, or any of its intermediates. Basic compounds of the disclosure that may form an acid addition salt include, for example, compounds that contain a basic nitrogen atom. Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric and phosphoric acids, as well as metal salts such as sodium monohydrogen
orthophosphate and potassium hydrogen sulfate. Illustrative organic acids that form suitable salts include mono-, di-, and tricarboxylic acids such as glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, benzoic, phenylacetic, cinnamic and salicylic acids, as well as sulfonic acids such as p- toluene sulfonic and methanesulfonic acids. Either the mono-, di- or the tri-acid salts can be formed, and such salts may exist in either a hydrated, solvated or substantially anhydrous form. In general, the acid addition salts of the compounds of the disclosure are more soluble in water and various hydrophilic organic solvents, and generally demonstrate higher melting points in comparison to their free base forms. The selection of the appropriate salt will be known to one skilled in the art. Other non- pharmaceutically acceptable acid addition salts, e.g. oxalates, may be used, for example, in the isolation of the compounds of the disclosure, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt.
The term pharmaceutically acceptable basic salt as used herein means any non-toxic organic or inorganic basic addition salt of any acid compound of the invention, or any of its intermediates, which are suitable for or compatible with the treatment of animals, in particular humans. Acidic compounds of the invention that may form a basic addition salt include, for example compounds that contain carboxylic acid, sulfonic acid, sulfinic acid, sulfonamide, N-unsubstituted tetrazole, phosphoric acid ester, or sulfuric acid ester. Illustrative inorganic bases which form suitable salts include lithium, sodium, potassium, calcium, magnesium, or barium hydroxide.
Illustrative organic bases which form suitable salts include aliphatic, alicyclic or aromatic organic amines such as methylamine, trimethylamine and picoline or ammonia. The selection of the appropriate salt will be known to a person skilled in the art. Other non-pharmaceutically acceptable basic addition salts, may be used, for example, in the isolation of the compounds of the invention, for laboratory use, or for subsequent conversion to a pharmaceutically acceptable acid addition salt. The formation of a desired compound salt is achieved using standard techniques. For example, the neutral compound is treated with a base in a suitable solvent and the formed salt is isolated by filtration, extraction or any other suitable method.
The term subject or patient or synonym thereto, as used herein includes all members of the animal kingdom, especially mammals, including human. The subject or patient is suitably a human.
As used herein, and as well understood in the art, treatment is an approach for obtaining beneficial or desired results, including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e. not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
Palliating a disease or disorder means that the extent and/or undesirable clinical manifestations of a disorder or a disease state are lessened and/or time course of the progression is slowed or lengthened, as compared to not treating the disorder. The term prevention or prophylaxis, or synonym thereto, as used herein refers to a reduction in the risk or probability of a patient becoming afflicted with the disease or manifesting a symptom associated with the disease.
The term therapeutically effective amount, effective amount or sufficient amount of a compound of the present invention is a quantity sufficient to, when administered to the subject, including a mammal, for example a human, effect beneficial or desired results, including clinical results, and, as such, an effective amount or synonym thereof depends upon the context in which it is being applied. For example, in the context of 1 1 β-HSD inhibition, it is an amount of the compound sufficient to achieve an inhibition of 1 1 β-HSD activity compared to the response obtained without administration of the compound. In the context of disease, therapeutically effective amounts of the compounds of the present invention are used to treat, modulate, attenuate, reverse, or affect a disease or conditions that benefits from an inhibition of 1 1 β-HSD, for example, chronic inflammatory diseases. An effective amount is intended to mean that amount of a compound that is sufficient to treat, prevent or inhibit such diseases or conditions. The amount of a given compound of the present invention that will correspond to such an amount will vary depending upon various factors, such as the given drug or compound, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art. Also, as used herein, a therapeutically effective amount of a compound of the present invention is an amount which prevents, inhibits, suppresses or reduces a disease or conditions that benefits from an inhibition of 1 1 β-HSD, for example, chronic inflammatory diseases as determined by clinical symptoms in a subject as compared to a control. As defined herein, a therapeutically effective amount of a compound of the present invention may be readily determined by one of ordinary skill by routine methods known in the art.
In an embodiment, a therapeutically effective amount of a compound of the present invention ranges from about 0.01 to about 100 mg/kg body weight, suitably about 0.02 to about 50 mg/kg body weight, and more suitably, from about 0.05 to about 20 mg/kg body weight. The skilled artisan will appreciate that certain factors may influence the dosage required to effectively treat a subject, or prevent a subject, suffering from a disease or conditions that benefits from an inhibition of 1 1 β-HSD activity, for example chronic inflammatory diseases, and these factors include, but are not limited to, the severity of the disease or disorder, previous treatments, the general health and/or age of the subject and other diseases present.
Moreover, a treatment or prevention regime of a subject with a therapeutically effective amount of the compound of the present invention may consist of a single administration, or alternatively comprise a series of applications. For example, the compound of the present invention may be administered at least once a week.
However, in another embodiment, the compound may be administered to the subject from about one time per week to about three times daily for a given treatment. The length of the treatment period depends on a variety of factors, such as the severity of the disease, the age of the patient, the concentration and the activity of the compounds of the present invention, or a combination thereof. It will also be appreciated that the effective dosage of the compound used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required.
As used herein the term administered contemporaneously means that two substances are administered to a subject in such a way that they are both biologically active in the subject at the same time. The exact details of the administration will depend on the pharmacokinetics of the two substances in the presence of each other, and can include administering one substance within 24 hours of administration of the other, if the pharmacokinetics is suitable. Designs of suitable dosing regimens are routine for one skilled in the art. In particular embodiments, two substances will be administered substantially simultaneously, i.e. within minutes of each other, or in a single composition that comprises both substances.
To inhibit or suppress or reduce or downregulate a function or activity, such
1 1 β-HSD activity, is to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another conditions.
In understanding the scope of the present disclosure, the term comprising and its derivatives, as used herein, are intended to be open ended terms that specify the presence of the stated features, elements, components, groups, integers, and/or steps, but do not exclude the presence of other unstated features, elements, components, groups, integers and/or steps. The foregoing also applies to words having similar meanings such as the terms, including, having and their derivatives. Finally, terms of degree such as substantially, about and approximately as used herein mean a reasonable amount of deviation of the modified term such that the end result is not significantly changed. These terms of degree should be construed as including a deviation of at least ±5% of the modified term if this deviation would not negate the meaning of the word it modifies.
Unless otherwise indicated, the terms a, an, and the as used herein mean one or more that one.
Compounds of the Invention
A new class of compounds derived from glycyrrhetinic acid has been identified as drugs for the treatment of chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer. The compounds according to the invention may be readily prepared by a person skilled in the art on the basis of his general knowledge. Results show, that glycyrrhetinic acid derivatives, namely compound 1 , inhibit both, Ι Ι β-HSDs enzyme activity and TNFa expression. Accordingly, compound 1 and related compounds are a novel class of mechanism-based drugs against chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer that act as inhibitors of Ι Ι β-HSDs enzyme activity and TNFa expression.
Accordingly, in one its aspects, the present invention includes a compound selected from a compound of Formula I:
wherein
R3a and R3b together are selected from =0, =NRa, =N-0-Ra, or
R3a and R3b are independently from one another selected from hydrogen,
-0-Ra, -0-C(=0)-Ra, -N(Ra)2, -NH-C(=0)-Ra and -NH-S(=0)2Ra; and
R11a and R11b together are selected from =0, =NRa, =NH, =N-0-Ra, =N-OH; R11a and R11b are independently from one another selected from hydrogen, -OH, -O-Ra, -O-C(=O)-Ra, -N(Ra)2, Ci-6alkyl, C2-8alkynyl, fluorine, chlorine, and bromine; and
a single or a double bond is present at position 12-13; and
the configuration at position 18 is R or S; and
X is selected from -C(=O)-1 , -S(=O)2- and -C(=S)-; and
R30 is selected from hydrogen, -Ra, or -O-Ra; and
R31 is selected from -Rb, -NH2, -NHRb, -N(Rb)2, -NH-S(=O)2-Rb,
-NH-NH-C(=O)-NH2, -NH-NH-C(=O)-NHRb, -NH-NH-C(=O)-N(Rb)2; and each Ra independently of one another is selected from hydrogen or from optionally substituted hydroxyalkyl, Ci-6alkyl, C2-6alkenyl, C2-8alkynyl, -CF3, carboxylic acid, - (CH2)n-C6-i4aryl, -CH=CH-C6-i4aryl, -C≡C-C6-i4aryl, -(CH2)n-C5-i4heteroaryl, -CH=CH-C5-i4heteroaryl, -C≡C-C5-i4heteroaryl, -(CH2)n-C3-8cycloalkyl,
-CH=CH-C3-8cycloalkyl and -C≡C-C3-8cycloalkyl; and
each Rb independently of one another is selected from hydroxy or from optionally substituted, Ci-6alkyl, C2-6alkenyl, C2-8alkynyl,, -(CH2)n-C3-8cycloalkyl,,-CF3,
-(CH2)n-C6-i4aryl; and
each n independently of one another denotes 0, 1 or 2;
optionally in the form of their pharmaceutically effective salts, solvates, prodrugs, tautomers, racemates, enantiomers, diastereomers or mixtures thereof.
The compounds of the invention may exist in a continuum of solid states ranging from fully amorphous to fully crystalline. The term amorphous refers to a state in which the material lacks long range order at the molecular level and, depending upon temperature, may exhibit the physical properties of a solid or a liquid. Typically such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the properties of a solid, are more formally described as a liquid. Upon heating, a change from solid to liquid properties occurs which is characterized by a change of state, typically second order (glass transition). The term crystalline refers to a solid phase in which the material has a regular ordered internal structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficiently will also exhibit the properties of a liquid, but the change from solid to liquid is characterized by a phase change, typically first order (melting point).
The compounds of the invention may also exist in unsolvated and solvated forms. The term solvate is used herein to describe a molecular complex comprising the compound of the invention and one or more pharmaceutically acceptable solvent molecules, for example, ethanol. The term hydrate is employed when said solvent is water. When the solvent or water is tightly bound, the complex will have a well-defined stoichiometry independent of humidity. When, however, the solvent or water is weakly bound the water/solvent content will be dependent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm.
Herein all references to compounds of Formula I include references to salts, solvates, prodrugs and multi-component complexes thereof.
The compounds of Formula I can be prepared using methods known in the art, for example, 18a- and Ι δβ-glycyrrhetinic acid and their methyl esters may be converted into the corresponding dienones by reaction with 2-iodoxybenzoic acid as per a reported method [8].
The present invention includes radiolabeled forms of the compounds of the invention, for example, compounds of the invention labeled by incorporation within the structure 3H, 11C or 14C or a radioactive halogen such as 125l and 18F. A radiolabeled compound of the invention may be prepared using standard methods known in the art. For example, tritium may be incorporated into a compound of the invention using standard techniques, for example by hydrogenation of a suitable precursor to a compound of the invention using tritium gas and a catalyst. Alternatively, a compound of the invention containing radioactive iodine may be prepared from the corresponding trialkyltin (suitably trimethyltin) derivative using standard iodination conditions, such as [125l] sodium iodide in the presence of chloramine-T in a suitable solvent, such as dimethylformamide. The trialkyltin compound may be prepared from the corresponding non-radioactive halo, suitably iodo, compound using standard palladium-catalyzed stannylation conditions, for example hexamethylditin in the presence of tetrakis
(triphenylphosphine) palladium (0) in an inert solvent, such as dioxane, and at elevated temperatures. Further, a compound of the invention containing a radioactive fluorine may be prepared, for example, by reaction of K[18F]/K222 with a suitable precursor compound, such as a compound of Formula I comprising a suitable leaving group, for example a tosyl group, that may be displaced with the 18F anion.
Methods and Compositions
The present invention relates to novel compounds of Formula I, accordingly the present invention includes all uses of these compounds including, for example, in therapeutic and diagnostic applications. The present invention accordingly includes the use of a compound or a combination of compounds of the invention as a medicament or as a diagnostic.
In their ability to inhibit the enzymatic activity of Ι ΐ β-HSD, certain compounds of the invention are useful for treating any condition or disease that benefits from an inhibition of 1 1 β-HSD. In an embodiment of the invention, the conditions or diseases that benefit from an inhibition of 1 1 β-HSD are chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer.
Accordingly, the present invention includes a method of treating chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof. The invention also includes a use of a compound of the invention to treat chronic
inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer and a use of a compound of the invention to prepare a medicament to treat chronic inflammatory diseases,
autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer. In embodiments of the invention the chronic inflammatory disease is selected from stomatitis, gingivitis, periodontitis, peri-implantitis and osteoarthritis. In embodiments of the invention the autoimmune disease is selected from rheumatoid arthritis, systemic lupus erythematosus (SLE), Sjogren's syndrome, scleroderma, dermatomyositis, and polymyositis, inflammatory bowel diseases like Crohn's disease and ulcerative colitis. In embodiments of the invention the skin disease is selected from dermatitis, contact dermatitis, allergic dermatitis, atopic dermatitis, psoriasis, eczema, prurigo simplex acuta, prurigo simplex subacuta, prurigo nodularis, alopecia areata, Idiopathic thrombocytopenic purpura, pemphigus vulgaris, actinic keratosis. In embodiments of the invention the bone disease is selected from inflammation and/or immune mediated bone loss, osteoporosis, postmenopausal osteoporosis, Paget's disease, lytic bone metastases, arthritis, juvenile chronic arthritis, adjuvant arthritis, infectious diseases, bone loss by cancer, bone loss by HIV, tooth loss, bone marrow inflammation, synovial inflammation, cartilage and/or bone erosion and/or proteoglycan damage, osteopenie, osteosclerose, osteonecrosis. In embodiments of the invention the metabolic disease or disorder is selected from fasting hyperglycemia, diabetes mellitus, in particular insulin dependent type II diabetes, impaired fasting glucose, impaired glucose tolerance, insulin resistance, high blood pressure, central obesity (also known as visceral adiposity), decreased HDL cholesterol and elevated triglycerides. In embodiments of the invention the infectious disease is selected from viral, bacterial or fungal infections. In embodiments of the invention the cancer is selected from bladder cancer, breast cancer, colorectal cancer, cutaneous melanoma, skin cancer, squamous cell carcinoma of the skin, endometrial cancer, leukemia, lung cancer, non-Hodgkin lymphoma, ovarian cancer, pancreatic cancer, and prostate cancer.
The present invention also includes a method of treating chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer comprising administering an effective amount of a compound of the invention to a subject in need thereof. Further the invention includes a use of a compound of the invention to treat chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer, as well as a use of a compound of the invention to prepare a medicament to treat these diseases.
In an embodiment of the invention the Ι ΐ β-HSD inhibiting compound is compound 1. A person skilled in the art would be able to identify Ι ΐ β-HSD inhibiting compounds of the invention using, for example enzyme assays with mammalian 1 1 β- HSD isolated from specific tissue or expressed in transfected cell lines as described in the examples below and in literature [9-12].
In their ability to downregulate the expression or activity of TNFa, the
compounds of the invention are useful for treating any condition or disease that benefits from a downregulation in the expression or activity of TNFa. In an embodiment of the invention, the conditions or diseases that that benefit from a downregulation in the expression or activity of TNFa, are inflammatory diseases, in particular rheumatoid arthritis, psoriasis, periodontitis, systemic lupus erythematosus (SLE), Sjogren's syndrome, scleroderma, dermatomyositis, and polymyositis, Crohn's disease and ulcerative colitis, asthma bronchiale.
Accordingly, in a further embodiment of the present invention, there is included a method of treating inflammatory diseases comprising administering a TNFa - downregulating effective amount of a compound of the invention to a subject in need thereof. A person skilled in the art would be able to identify TNFa-downregulating compounds of the invention by contacting one or more cells with a compound of the invention and assaying for the presence of one TNFa and comparing the levels of TNFa in the one or more cells with that of controls. Such methods are known in the art [13,14] and are described in the examples herein below.
The compounds of the invention are suitably formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo. Accordingly, in another aspect, the present invention includes a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable carrier or diluent.
The compositions containing the compounds of the invention can be prepared by known methods for the preparation of pharmaceutically acceptable compositions which can be administered to subjects, such that an effective quantity of the active substance is combined in a mixture with a pharmaceutically acceptable vehicle.
Suitable vehicles are described in literature [15-17]. On this basis, the compositions include, albeit not exclusively, solutions of the substances in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered solutions with a suitable pH and iso-osmotic with the physiological fluids.
In accordance with the methods of the invention, the described compounds, salts or solvates thereof may be administered to a patient in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art. The compositions of the invention may be administered, for example, by peroral, parenteral, buccal, sublingual, nasal, rectal, patch, pump or transdermal (topical) administration and the pharmaceutical compositions formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.
A compound of the invention may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet. For oral therapeutic administration, the compound of the invention may be incorporated with excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. A compound of the invention may also be administered parenterally. Solutions of a compound of the invention can be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, propylene glycol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. A person skilled in the art would know how to prepare suitable formulations.
The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersion and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. Ampoules are convenient unit dosages. In addition, a pharmaceutical form suitable for injectable use includes sterile powders for the direct needle-free injection of the substance into the outer layer of skin in a simple-to-use-device.
Compositions for nasal and pulmonary administration may conveniently be formulated as aerosols, drops, gels and powders. Aerosol formulations typically comprise a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device.
Alternatively, the sealed container may be a unitary dispensing device such as a single dose inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal after use. Where the dosage form comprises an aerosol dispenser, it will contain a propellant which can be a compressed gas such as compressed air or an organic propellant such as hydrofluoroalkanes. The aerosol dosage forms can also take the form of a pump-atomizer. Where the dosage form comprises a dry powder inhaler, it can contain a propellant or rely on the force of patient inhalation to entrain powder from the device and subsequently break-up the powder into small aerosol particles
Compositions suitable for buccal or sublingual administration include tablets, lozenges, pastilles, and patches wherein the active ingredient is formulated with a carrier such as sugar, acacia, tragacanth, or gelatin and glycerine. Compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base such as cocoa butter. Compositions for topical administration may include, for example, propylene glycol, isopropyl alcohol, mineral oil and glycerin. Preparations suitable for topical administration include liquid or semi-liquid preparations such as liniments, lotions, applicants, and oil-in-water or water-in-oil emulsions such as creams, ointments or pastes; or solutions or suspensions such as drops. Moreover, suitable preparations for topical administration include patches wherein the ingredient is formulated with carriers such as adhesives, solvents, or polymers. In addition to the aforementioned
ingredients, the topical preparations may include one or more additional ingredients such as diluents, buffers, flavoring agents, binders, surface active agents, thickeners, lubricants, preservatives, e.g. methyl hydroxybenzoate (including anti-oxidants), emulsifying agents and the like.
Sustained or direct release compositions can be formulated, e.g. liposomes or those wherein the active compound is protected with differentially degradable coatings, such as by microencapsulation, multiple coatings, etc or those wherein the compound is imbedded into a polymer matrix such as polylactid or the like. It is also possible to freeze-dry the compounds of the invention and use the lyophilisates obtained, for example, for the preparation of products for injection.
The dosage of the compounds of Formula I and/or compositions of the invention can vary depending on many factors such as the pharmacodynamic properties of the compound, the mode of administration, the age, health and weight of the recipient, the nature and extent of the symptoms, the frequency of the treatment and the type of concurrent treatment, if any, and the clearance rate of the compound in the human or animal to be treated. One of skill in the art can determine the appropriate dosage based on the above factors. The compounds of Formula I may be administered initially in a suitable dosage that may be adjusted as required, depending on the clinical response.
The compounds of the invention may be administered to a subject alone or in combination with pharmaceutically acceptable carriers, as noted above, and/or with other pharmaceutically active agents for the treatment of chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer, the proportion of which is determined by the solubility and chemical nature of the compounds, chosen route of administration and standard pharmaceutical practice. The compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies that treat inflammatory and autoimmune diseases, for example, but not limited to, non-steroidal anti-inflammatory drugs
(NSAIDS), corticosteroids, disease modifying anti-rheumatic drugs (DMARDs), TNFa blockers, interleukin 1 (IL-1 ) blockers, monoclonal antibodies against B cells, and T cell activation blocker.
The compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies that treat skin diseases, for example, but not limited to, corticosteroids, immunomodulating agents, retinoids, urea, zinc oxide, panthenol, antibiotics, and antimycotics.
The compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies that treat metabolic diseases, for example, but not limited to, sulfonylureas, meglitinides, biguanides, thiazolidinediones (TZDs), and a-glucosidase inhibitors.
The compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies that treat infectious diseases, for example, but not limited to, antibiotics like aminoglycosides, ansamycins,
carbacephem, carbapenems, cephalosporins, glycopeptides, macrolides,
monobactams, penicillins, polypeptides, quinolones, sulfonamides, and tetracyclines, antimycotics like polyenes, imidazoles, triazoles, allylamines, and echinocandins, and antivirals like cell entry blockers, nucleoside analogues, reverse transcriptase inhibitors, protease inhibitors, and neuraminidase inhibitors.
The compounds of Formula I, or salts or solvates thereof, can be used alone or in combination with other agents or therapies that treat cancer, for example, but not limited to, cytotoxic drugs, kinase inhibitors, antibodies and immunotherapy, selective receptor modulators, non-steroidal anti-inflammatory drugs (NSAIDS), corticosteroids, and enzyme modulators.
While the following Examples illustrate the invention in further detail, it will be appreciated that the invention is not limited to the specific examples.
Examples
All solvents were purified and dried by standard procedures. Melting points were measured on a Buchi B-545 melting point apparatus or a Kofler hot stage microscope and are uncorrected. Column chromatography was performed on silica gel 60 (230- 400 mesh, Merck). Reactions were monitored by TLC on silica gel 60 F254 pre-coated glass plates (Merck) or on silica gel 60 F254 HPTLC pre-coated glass plates with 2.5 cm concentration zone (Merck); spots were detected by UV light examination or visualized by spraying with anisaldehyde sulfuric acid, phosphorous molybdic acid, with a mixture of cerium (IV) sulfate and ammonium molybdate or ninhydrine and heating. Concentration of solutions was performed at reduced pressure at
temperatures below 50 °C. NMR spectra were recorded at 297 K in CDCI3, pyridine-ds, DMSO-de or MeOD with a Bruker AC 200 spectrometer (1H at 200.13 MHz, 13C at 50.31 MHz), a Bruker DPX 300 spectrometer (1H at 300.13 MHz, 13C at 75.47 MHz), a Bruker AC 400 spectrometer (1H at 400.13 MHz, 13C at 100.61 MHz) and with a Bruker DPX 400 spectrometer (1H at 400.13 MHz, 13C at 100.61 MHz) using standard Bruker NMR software. 1H NMR spectra were referenced to tetramethylsilane. 13C NMR spectra were referenced to chloroform (δ 77.00). Infrared spectra were recorded on a BIORAD ATR-FT-IR spectrometer as solutions in DCM or methanol. Elemental analyses were measured with an EA 1108 CHNS-0 from Carlo Erba. Compounds were purified by MPLC (medium pressure liquid chromatography) using preparative silica gel (40-63 mm) columns. HPLC was performed using a Waters 2695 instrument with Merck Chromolith RP18 columns and a gradient of 3 % to 60 % acetonitrile and water containing 0.1 % formic acid at a flow of 1 .0 to 3.0 mL/min. Preparative LC was performed using a Waters instrument with Merck Geminy RP18 columns and a gradient of 3 % to 60 % acetonitrile and water containing 0.1 % formic acid at a flow of 25 mL/min. The HPLC reported purity is the number generated for the peak area as calculated using the Waters Millennium software with the Maxplot option for the UV maximum of the corresponding peak. Mass spectra were measure in Cl-mode with ammonia as a reagent gas on a thermo scientific.
Example 1 : Precursor
(3β,18β,20β)-3-(ΑοβΙγΙοχγ)-20-Ϊ5θογ3Π3ΐο-30-ηοΓθΙβ3η-12-βη-11 -οηβ A stirred suspension of glycyrrhetinic acid (9.4 g, 20 mmol) in acetyl chloride was heated up to 50°C for 1 h. After 1 h acetylchloride from cleared solution was removed under vacuum to get the residue. Diethylether (50 mL) was added to the residue and filtered. After drying overnight at room temperature under vacuum yielded 9.1 g (86 %) of (3p,18p,20P)-3-(acetyloxy)-1 1 -oxo-olean-12-en-29-oyl chloride as white powder.
The powder (5 g, 9.41 mmol) was suspended in acetone (300 mL). To the stirred suspension was added sodiumazide (1 .5 g, 23 mmol). After 30 min ice water (500 mL) was added to the reaction mixture. Solids were filtered suck dried and further dissolved in chloroform (500 mL) and refluxed overnight. Solvent was removed from reaction mixture to get crude product. Purification of the crude product using 50 mg silica with a mixture of 0-10 % diethylether in DCM yielded 4.3 g, (89.6 %) of
(3p,18p,20P)-3-(acetyloxy)-20-isocyanato-30-norolean-12-en-11 -one as white powder.
1H NMR (200 MHz, CDCI3) δ 5.63 (s, 1 H), 2.77 (m, 1 H), 2.25 (m, 2H), 2.03 (s, 3H), 1 , 42-1 .95 (m, 19H), 1 .35 (s, 3H), 1 .30 (s, 3H), 1 .14 (s, 3H), 1 .12 (s. 3H), 0.88 (s, 3H), 0.86 (s, 6H)
13C NMR (200 MHz, CDCI3) δ 199.76, 170.94, 168.17, 128.56, 122.1 1 , 80.53, 61 .70, 58.59 54.97, 47.21 , 45.34, 44.44, 43.17, 38.75, 38.00, 36.90, 36.17, 34.77, 32.64, 31 .84, 31 .70, 28.14, 28.00, 26.28, 26.16, 23.51 , 23.29, 21 .27, 18.65, 17.33, 16.65, 16.37
Example 2: Compound 2
Λ/-[(3β,18β,20β)-3-(ΑοθΙγΙοχγ)-11 -οχο-30-ηοΓθΙθ3η-12-θη-20-γΙ] urea
To a stirred solution of (3p,18p,20P)-3-(acetyloxy)-20-isocyanato-30-norolean- 12-en-1 1 -one (2 g, 3.9 mmol) in chloroform (200 mL) was purged with ammonia gas for 15 min at 0-5°C. Excess of ammonia was removed by purging compressed air. Solvent was removed under vacuum to get residue. Purification of the residue on silica with a mixture of 0-5 % diethyl ether in DCM yielded 1.5 g (73 %) of the desired product as white powder.
1H NMR (200 MHz, CDCI3) δ 5.68 (s, 1 H), 4.49 (m, 1 H), 2.70 (m, 1 H), 2.15-2.31 (m, 4H), 2.03 (s, 3H), 1 .40-1 .97 (m, 19H), 1 .32 (s, 3H), 1 .30 (s, 3H), 1 .13 (s, 3H), 1 .10 (s, 3H), 0.89 (s, 6H), 0.79 (s, 3H).
13C NMR (200 MHz, CDCI3) δ 200.41 , 171 .03, 170.10, 158.86, 128.06, 80.58, 61 .71 , 54.95, 52.28, 52.18, 46.59, 45.45, 43.33, 43.09, 38.89, 38.01 , 36.87, 35.78, 32.67, 31 .88, 31 .28, 28.54, 28.39, 28.01 , 26.20, 23.53, 23.16, 21 .30, 18.64, 17.32, 16.66, 16.37.
Example 3: Compound 3
Λ/-[(3β,18β,20β)-3-ΗγςΐΓθχγ-11 -οχο-30-ηοΓθΙθ3η-12-θη-20-γΙ] urea
To a stirred solution of /V-[(3p,18p,20P)-3-(acetyloxy)-1 1-oxo-30-norolean-12- en-20-yl] urea (200 mg, 0.38 mmol) in 20 ml_ of methanol was added 4.2 g (75 mmol) of KOH. The reaction mixture was stirred at room temperature. After 16 h the solvent was removed under vacuum, the residue was diluted with water (20 ml_) and the pH was adjusted to 2-3 using 2N HCI solution. The reaction mixture was extracted with 2x30 ml_ of DCM. The combined organic layer was washed with 20 ml_ of water and brine and finally dried over sodium sulphate. The separated organic solvent was removed under vacuum. Crystallisation of the crude product from acetone (2 ml_) yielded the final product (95.5 mg, 54 %) as white powder.
1H NMR (200 MHz, Pyridine-d5) δ 6.62 (s, 1 H), 5.80 (s, 1 H), 3.45 (m, 1 H), 3.12 (m, 1 H), 2.56-1 .36 (m, 22H), 1 .28 (s, 6H), 1 .23 (s, 3H), 1 .06 (s, 3H), 1 .04 (s, 3H), 0.80 (s, 3H) 13C NMR (200 MHz, Pyridine, d5) δ 200.35, 170.49, 160.51 , 128.90, 78.35, 62.58, 55.75, 52.83, 47.21 , 46.00, 43.98, 43.56, 40.24, 38.05, 36.76, 33.43, 33.53 32.65, 32.54, 29.43, 29.20, 29.05, 28.55, 27.17, 27.01 , 23.80, 19.27, 18.37, 17.27, 17.06
Example 4: Compound 4
Λ/-[(18β,20β)-3,11-Οϊοχο-30-ηοΓθΙθ3η-12-θη-20-γΙ] urea
To a stirred solution of /V-[(3p,18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en- 20-yl] urea (100 mg, 0.21 mmol) in 30 ml_ of acetone was added 0.20 ml_ of Jones reagent over 20 min at 0 °C until the brown colour persisted. The reaction mixture was stirred for 30 min at 0 °C, diluted drop by drop with isopropanol until the brown colour disappeared. The reaction mixture was filtered and the residue was washed with 20 ml_ of DCM. Evaporation of the clear filtrate yielded the crude product which was purified on silica with a mixture of 0-5 % methanol in DCM to yield the final product (70 mg, 70.2 %) as white powder.
13C NMR (200 MHz, Pyridine-d5) δ 216.32, 199.28, 170.55, 159.98, 128.65, 61 .62, 55.52, 52.79, 48.21 , 47.49, 45.73, 44.08, 45.73, 44.08, 43.42, 40.40, 37.45, 36.69, 34.89, 32.73, 32.57, 32.52, 29.40, 29.08, 27.00, 23.65, 21 .99, 19.44, 18.95, Example 5: Compound 5
Λ/-[(18β,20β)-3-(Hydroxyimino)-11-oxo-30-norolean-12-en-20-yl] urea
To a solution of Λ/-[(18β,20β)-3,1 1 -dioxo-30-norolean-12-en-20-yl] urea
(200 mg, 0.41 mmol) in 2.5 mL pyridine were added 150 mg (2.16 mmol) of
hydroxylamine hydrochloride. The reaction mixture was stirred at 50 °C. After 2 h the solvent was removed under vacuum and the residue crystallised from water (20 mL) to yield the final product (120 mg, 58.3 %) as white solid material.
CI-MS, m/z (Irei. (%)): 480.99 (100.00); 436.79 (81 .81 ); 439.08 (79.57); 465.07 (67.82); 437.95 (66.60); 367.1 1 (46.1 1 ); 454.98 (45.32); 423.02 (32.45); 482.04 (31.99); 466.06 (31 .85).
1H NMR (200 MHz, Pyridine-d5) δ 6.37 (s, 1 H), 5.61 (s, 1 H), 3.42, (m, 1 H), 3.13 (m, 1 H), 1.44-2.62 (m, 22H), 1 .34 (s, 6H), 1 .25 (s, 6H), 1 .14 (s, 3H), 1 .06 (s, 3H), 0.76 (s, 3H)
13C NMR (200 MHz, Pyridine-d5) δ 199.64, 170.25, 164.41 , 160.15, 128.76,
62.00, 56.15, 52.82, 48.21 , 47.37, 45.90, 44.02, 43.52, 40.82, 39.86, 37.94, 36.72, 32.96, 32.52, 29.40, 29.07, 28.35, 27.1 1 , 26.95, 24.28, 23.70, 19.14, 18.96, 17.95, 16.36 Example 6: Compound 6
Λ/-Hydroxy-Λ/-methyl-Λ/'-[(3β,18β,20β)-3-(acetyloxy)-11 -oxo-30-norolean-12- en-20-yl] urea
To a suspension of NaHC03 (30 mg, 0.36 mmol) in 30 ml_ acetone was added
/V-methyl-hydroxylamine hydrochloride (25 mg, 0.30 mmol). The reaction mixture was stirred at room temperature and a solution of (3p,18p,20P)-3-(acetyloxy)-20- isocyanato-30-norolean-12-en-1 1 -one (100 mg, 0.20 mmol) in 30 ml_ of DCM was added drop by drop. The reaction mixture was stirred for 2 h at room temperature, diluted with water and extracted three times with 20 ml_ of DCM. The combined organic layer was washed with je 20 ml_ of 2N HCI solution and water. The separated organic layer was dried over sodium sulphate and concentrated in vacuum. Purification of the residue on silica with a mixture of 0-5 % methanol in DCM yielded 70 mg (64 %) of the product as white powder.
CI-MS, m/z (lrei. (%)): 509.97 (100.00); 467.07 (50.13); 484.00 (43.95); 540.71
(40.01 ); 51 1 .01 (33.93); 539.93 (19.47); 468.09 (15.48); 541 .76 (14.45); 485.01 (13.36); 483.25 (1 1 .47).
1H NMR (200 MHz, CDCI3) δ 5.92 (s, 1 H), 5.61 (s, 1 H), 4.50, (m, 1 H), 3.09 (s, 3H), 2.74 (m, 1 H), 2.16-121 (m, 30H), 1 .14 (s, 3H), 1 .10 (s, 3H), 0.80 (s, 6H), 0.81 (s, 3H)
13C NMR (200 MHz, CDCI3) δ 200.87, 171 .03, 170.36, 160.98, 128.12, 80.51 , 61 .71 , 54.96, 52.33, 47.08, 45.54, 43.41 , 42.85, 39.86, 38.77, 38.02, 36.93, 35.69, 32.68, 31 .87, 31 ,38, 28.44, 28.27, 28.00, 26.34, 26.20, 23.52, 23.10, 21 .28, 18.66, 17.30, 16.66, 16.39 Example 7: Compound 7
Λ/-ΗγάΓθχγ-Λ/'-[(3β,18β,20β)-3-(3θβΙγΙοχγ)-11 -οχο-30-ηοΓθΙβ3η-12-βη-20-γΙ] urea
To a suspension of NaHC03 (48 mg, 0.58 mmol) in 30.0 ml_ of THF was added
41 mg (0.59 mmol) of hydroxylamine hydrochloride. The reaction mixture was stirred at room temperature and (3p,18p,20P)-3-(acetyloxy)-20-isocyanato-30-norolean-12-en- 1 1 -one (200 mg, 0.39 mmol.) was added. After 2 h the reaction mixture was filtered and concentrated to get the crude compound. Purification of the residue on silica with a mixture of 0-5 % methanol in DCM yielded 120 mg (56.6 %) of the product as white powder.
CI-MS, m/z (Irei. (%)): 509.97 (100.00); 483.92 (52.60); 467.03 (49.74); 51 1.00 (35.14); 258.95 (22.52); 525.47 (20.04); 135.01 (19.74); 524.42 (19.64); 468.05
(18.82); 526.41 (18.12).
1H NMR (200 MHz, CDCI3) δ 5.81 (s, 1 H), 5.70 (s, 1 H), 4.50 (m, 1 H), 2.70 (m,
1 H), 2.35-1 .44 (m, 25H), 1 .33 (s, 6H), 1 .13, (s, 3H), 1 .09 (s, 3H), 0.86 (s, 6H), 0.81 (s, 3H)
13C NMR (200 MHz, CDCI3) δ 201 .04, 171 .01 , 170.61 , 161.23, 128.12, 80.52, 61 .67, 54.94, 52.58, 47.04, 45.46, 43.34, 41 .27, 38.74, 38.02, 36.96, 35.54, 32.89, 32.69, 31 .82, 28.47, 28.30, 28.01 , 26.41 , 26.31 ,23.52, 23.02, 21 .29, 18.68, 17.29, 16.66, 16.40 Example 8: Compound 8
Λ/-ΜθΙήγΐ5υΙίοηγΙ-Λ/'-[(3β,18β,20β)-3-(3θθΙγΙοχγ)-11 -οχο-30-ηοΓθΙθ3η-12-θη- 20-yl] urea
To a stirred solution of (3p,18p,20P)-3-(acetyloxy)-20-isocyanato-30-norolean-
12-en-1 1 -one (200 mg, 0.39 mmol) in 50 ml_ DCM was added 56 mg (0.59 mmol) of methane sulphonamide. The reaction mixture was stirred at room temperature. After 3 h the reaction mixture was quenched with water, the organic layer was separated and the aqueous layer was extracted with DCM. The combined organic layer was washed with water, dried over sodium sulphate and the solvent removed under vacuum.
Purification of the residue on silica with a mixture of 0-5 % methanol in DCM yielded 120 mg (50.6 %) of the product as white powder.
CI-MS, m/z (Irei. (%)): 483.87 (100.00); 467.02 (73.12); 338.05 (41.80); 509.99 (36.93); 189.00 (32.56); 484.88 (30.73); 468.05 (27.71 ); 256.95 (15.10); 135.05 (13.72); 121 .93 (12.99).
1H NMR (200 MHz, CDCI3) δ 6.10 (s, 1 H), 5.62 (s, 1 H), 4.50 (m, 1 H), 3.24 (s, 3H), 2.68 (m, 1 H), 2.33 (s, 1 H), 2.17-1 .40 (m, 23H), 1 .36 (s, 3H), 1 .33 (s, 3H), 1 .14 (s, 3H), 1 .1 1 (s, 3H), 0.87 (s, 6H), 0.84 (s, 3H)
13C NMR (200 MHz, CDCI3) δ 200.38, 171 .13, 169.26, 150.74, 128.26, 80.58, 61 .73, 54.93, 53.71 , 46.73, 45.48, 43.30, 42.10, 42.02, 38.75, 38.01 , 36.88, 35.69, 32.65, 31 .86, 31 .32, 28.40, 28.01 , 27.86, 26.31 , 26.10, 23.51 , 23.20, 21 .31 , 18.63, 17.30, 16.66, 16.37 Example 9: Compound 9
Λ/-[(3β,18β,20β)-3-(ΑοθΙγΙοχγ)-11 -οχο-30-ηοΓθΙθ3η-12-θη-20-γΙ]-Λ/'- (phenylmethyl)urea
To a solution of (3p,18p,20P)-3-(acetyloxy)-20-isocyanato-30-norolean-12-en- 1 1 -one (200 mg, 0.3924 mmol) in 50 mL DCM was added 63 mg (0.59 mmol) of benzyl amine. The reaction mixture was stirred at room temperature. After 3 h the solvent was removed under vacuum and the residue purified on silica with a mixture of 0-10 % methanol in DCM to yield 160 mg (66 %) of the product as white powder.
1H NMR (200 MHz, CDCI3) δ 7.26 (m, 5H), 5.56 (s, 1 H), 4.48 (m, 1 H), 4.28 (m, 2H), 2.72 (m, 1 H), 2.29-1 .40 (m, 25H), 1 .29 (s, 6H), 1 .13 (s, 3H), 1 .09 (s, 6H), 0.86 (s, 6H), 0.71 (s, 3H)
13C NMR (200 MHz, CDCI3) δ 200.08, 171 .07, 169.62, 157.95, 139.55, 128.60, 128,45, 128.13, 127.14, 80.60, 61 .68, 54.95, 52.40, 46.54, 45.41 , 44.24, 43.41 , 43.28, 38.80, 38.01 , 36.83, 35.74, 32.66, 31 .79, 31 .12, 28.71 , 28.28, 28.02, 26.32, 26.14, 23.50, 23.20, 21.30, 18.62, 17.34, 16.67, 16.37
Example 10: Compound 10
Λ/-[(3β,18β,20β)-3-(ΑοθΙγΙοχγ)-11 -οχο-30-ηοΓθΙθ3η-12-θη-20-γΙ]-1 ,2- hydrazinedicarboxamide
To a stirred suspension of semicarbazide hydrochloride (66 mg, 0.59 mmol) in 20 mL of THF were added sodium bicarbonate (50 mg, 0.60 mmol) and (3β,18β,20β)- 3-(acetyloxy)-20-isocyanato-30-norolean-12-en-1 1 -one (200 mg, 0.39 mmol) The reaction mixture was stirred for 2 h at room temperature, filtered and the solvent removed in vacuum. The residue was dissolved in DCM, washed with water and the separated organic layer was dried over sodium sulphate. After filtration the solvent was removed under vacuum and the residue purified on silica with a mixture of 0-8 % methanol in DCM to yield 130 mg (56.7 %) of the product as white powder.
CI-MS, m/z (Irei. (%)): 510.00 (100.00); 75.98 (70.54); 510.95 (42.86); 466.99 (38.54); 483.97 (35.09); 134.99 (30.35); 338.08 (28.77); 299.83 (28.68); 135.88 (28.41 ); 258.97 (25.85)
1H NMR (200 MHz, Pyridine-d5) δ 6.15 (s, 1 H), 5.78 (s, 1 H), 4.72 (m, 1 H), 3.05 (m, 1 H), 2.36 (m, 3H), 2.03-1 .34 (m, 22H), 1 .27 (s, 3H), 1 .22 (s, 3H), 1 .00 (s, 3H), 0.89 (s, 6H), 077 (s, 3H)
13C NMR (200 MHz, Pyridine-d5) δ 199.74, 171.03, 169.98, 162.08, 159.43, 128.92, 80.89, 62.21 , 55.43, 52.99, 47.42, 45.89, 43.93, 42.17, 39.48, 38.70, 37.78, 36.57, 33.36, 33.16, 32.44, 29.16, 28.91 , 28.55, 27.10, 26.92, 24.42, 23.76, 21.57, 19.15, 18.05, 17.45, 17.13
Example 11 : Compound 11
Λ/-[(3β,18β,20β)-3-(ΑοθΙγΙοχγ)-11 -οχο-30-ηοΓθΙθ3η-12-θη-20-γΙ]-Λ/'- [(trifluoromethyl)sulfonyl]urea
A mixture of trifluoromethane sulphonamide (87 mg, 0.58 mmol) and
(3p,18p,20P)-3-(acetyloxy)-20-isocyanato-30-norolean-12-en-11 -one (200 mg, 0.39 mmol) in 50 mL DCM was stirred for 3 h at room temperature. The reaction mixture was quenched with water and extracted with DCM. The separated organic layer was washed with water and dried over sodium sulphate. After filtration the solvent was removed in vacuum and the residue purified on silica with a mixture of 0-10 % methanol in DCM to yield 110 mg (42.6 %) of the product as white powder.
CI-MS, m/z (Irei. (%)): 525.80 (100.00); 509.91 (75.93); 524.82 (57.79); 523.98 (44.49); 523.05 (38.02); 526.61 (35.82); 510.91 (30.23); 521 .06 (26.76); 527.32 (24.65); 522.04 (18.94).
1H NMR (200 MHz, Pyridine-d5) δ 5.76 (s, 1 H), 5.65 (s, 1 H), 4.72 (m, 1 H), 3.10 (m, 1 H), 2.44-1 .51 (m, 22H), 1 .43 (s, 3H), 1 .36 (s, 3H), 1 .23 (s, 3H), 1 .05 (s, 3H), 0.90 (s, 3H), 0.89 (s, 3H), 0.77 (s, 3H)
13C NMR (200 MHz, Pyridine-d5) δ 199.96, 171.08, 170.15, 162.28, 128.84, 80.93, 62.31 , 55.50, 53.57, 47.48, 45.92, 44.02, 42.03, 39.61 , 38.71 , 37,82, 36.43, 33.23, 33.03, 32.43, 28.96, 28.60, 28.55, 27.12, 27.05, 24.44, 23.79, 21 .58, 19.18, 18.08, 17.44, 17.13
Example 12: Compound 12
(3β,18 β,20β)-3-(Acetyloxy)-20-(Λ/-methylsulfonyl-amino)-30-norolean-12- en-11 -one
To a stirred suspension of (3p,18p,20P)-3-(acetyloxy)-20-isocyanato-30- norolean-12-en-11 -one (200 mg, 0.40 mmol) in 20 mL dioxane was added NaOH (109 mg, 1 .95 mmol). After 3 h the reaction mixture was filtered and the solvent removed under vacuum. Purification of crude product using 50 mg silica with a mixture of 0-10 % methanol in DCM yielded 100 mg (52 %) of 3-hydroxy-20-amino-30-norolean-12-en- 1 1 -one as white powder. 1H NMR (200 MHz, CDCI3) δ 5.60 (s, 1 H), 4.45 (m, 1 H), 2.72 (m, 1 H), 1.39-2.30 (m, 25H), 1 .31 (s, 6H), 1.13 (s, 3H), 1 .09 (s, 3H), 0.85 (s, 6H), 0.80 (s, 3H).
13C NMR (200 MHz, CDCI3) δ 200.07, 171 .03, 169.73, 128.12, 80.62, 61 .65, 54.92, 52.38, 46.55, 45.39, 43.30, 42.17, 38.73, 37.99, 36.83, 35.96, 32.66, 31.91 , 31 .58, 28.90, 28.51 , 28.01 , 26.36, 26.18, 23.52, 23.16, 21 .31 , 18.62, 17.33, 16.66, 16.37.
To a stirred solution of 3-hydroxy-20-amino-30-norolean-12-en-1 1-one (200 mg, 0.4152 mmol) in 25 ml_ dry DCM were added TEA (84 mg, 0.83 mmol) and freshly distilled methanesulphonylchloride (75 mg, 0.65 mmol). The reaction mixture was stirred at 0 °C for 3 h and afterwards quenched with water. The organic layer was separated and the aqueous layer extracted twice with 20 ml_ of DCM. The combined organic layer was dried over sodium sulfate, filtered and the solvent was evaporated. Purification of the residue on silica with a mixture of 0-10 % methanol in DCM yielded 1 10 mg (47 %) of the final product as white powder.
CI-MS, m/z (lrei. (%)): 578.53 (100.00); 577.04 (28.47); 338.03 (27.53); 577.76
(25.17); 579.57 (25.04); 575.93 (11 .64); 338.99 (8.67); 580.58 (7.89); 467.00 (5.09); 134.98 (5.04).
1H NMR (200 MHz, CDCI3) δ 5.65 (s, 1 H), 5.30 (s, 1 H), 4.52 (m, 1 H), 3.04 (s, 3H), 2.81 (m, 1 H), 2.33-1 .54 (m, 23H), 1 .44 (s, 3H), 1 .33 (s, 3H), 1 .15 (s, 3H), 1 .12 (s, 3H), 0.89 (s, 3H), 0.87 (s, 6H)
13C NMR (200 MHz, CDCI3) δ 199.71 , 171 .03, 168.10, 128.55, 80.57, 61.69, 56.43, 54.96, 46.15, 45.38, 44.66, 43.20, 43.09, 38.74, 38.01 , 36.88, 35.27, 33.37, 32.65, 31 .83, 29.31 , 28.31 , 28.02, 26.23, 23.52, 23.37, 21 .30, 18.66, 17.35, 16.66, 16.37
The following example compounds are synthesized analogously to the procedures as described above.
Λ/-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-[(3p,18p,20P)-3-amino-1 1-oxo-30-norolean-12-en-20-yl] urea,
Λ/-[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, Λ/-[(3β, 18p,20P)-3-(methylsulfonamino)-11 -oxo-30-norolean-12-en-20-yl]urea, /V-[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]urea,
/V-[(18p,20P)-3-(hydroxyimino)-1 1-oxo-30-norolean-12-en-20-yl] urea,
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/V '-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl] urea,
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/ -[(3ββ, 18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V-methyl-/V '-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30- norolean-12-en-20-yl] urea
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(methylsulfonamino)-1 1 -oxo-30-norolean-12- en-20-yl]urea,
/V-hydroxy-/V-methyl-/V -[(18β,20β)-3, 1 1 -dioxo-30-norolean-12-en-20-yl]urea,
/V-hydroxy-/V-methyl-/V -[(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V -[(3p,18p,20P)-3-(acetyloxy)-1 1-oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/ -[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea,
N-hydroxy-N -[(3p,18p,20P)-3-(acetamino)-1 1-oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/ -[(3β, 18p,20P)-3-amino -1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-hydroxy-/ -[(3β, 18p,20P)-3-(succinyloxy)-11 -oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/ -[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V -[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-11 -oxo-30-norolean-12-en- 20-yl]urea,
/V-hydroxy-N'-[(3p, 18p,20P)-3-(methylsulfonamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V -[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]urea,
N-hydroxy-N -[(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/ -[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-methyl-/ -[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/V '-[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, N-methyl-/V -[(3p,18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]urea,
N-methyl-/V -[(3p,18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/V '-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, N-methyl-N -[(3p,18p,20P)-3-(trifluoromethylsulfonamino)-11 -oxo-30-n
20-yl]urea,
N-methyl-N -[(3p,18p,20P)-3-(methylsulfonamino)-1 1-oxo-30-norolean-12-en-20- yl]urea,
/V-methyl-/V -[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]urea,
N-methyl-N -[(18p,20P)-3-(hydroxyimino)-11 -oxo-30-norolean-12-en-20-yl]urea,
/V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea, N-methylsulfonyl-N -[(3p,18p,20P)-3-(acetamino)-1 1-oxo-30-norolean-12-en-20- yl]urea,
/V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30-norolean- 12-en-20-yl]urea,
N-methylsulfonyl-N -[(3p,18p,20P)-3-(methylsulfonamino)-11 -oxo-30-norolean-12-^ 20-yl]urea,
/V-methylsulfonyl-/ -[(18β,20β)-3, 1 1 -dioxo-30-norolean-12-en-20-yl]urea,
N-methylsulfonyl-N -[(18p,20P)-3-(hydroxyimino)-1 1-oxo-30-norolean-12-en-20-yl]^ Λ/-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-/V - [(trifluoromethyl)sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl)sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl)sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(methylsulfonamino)-11 -oxo-30-norolean-12-en-20-yl]-/V - [(trifluoromethyl)sulfonyl]urea,
N-[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]-N -[(trifluoromethyl)sulfon^^^
/V-[(18p,20P)-3-(hydroxyimino)-1 1-oxo-30-norolean-12-en-20-yl]-/V - [(trifluoromethyl)sulfonyl]urea,
(3β, 18p,20P)-3-hydroxy-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-(acetyloxy)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-amino-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-(acetamino)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-(succinyloxy)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-11 -one, (3β, 18p,20P)-3-(succinylamino)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 - one,
(3β, 18p,20P)-3-(trifluoromethylsulfona
12-en-1 1 -one,
(3β, 18p,20P)-3-(methylsulfonamino)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en- 1 1 -one,
(18β,20β)-3, 1 1 -dioxo-30-norolean-12-en-20-yl]urea,
(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-/ -(phenylmethyl) urea, and
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-1 ,2-hydrazine- dicarboxamide.
Example 13: Effects of Compounds on the Activity of Ι ΐ β-HSD
Cell culture media were purchased from Invitrogen, Basel, Switzerland, [1 ,2,6,7- 3H]-cortisol from Amersham Pharmacia, Dubendorf, Switzerland. [1 ,2,6,7-3H]- cortisone was produced by incubating 1 mCi of [1 ,2,6,7-3H]-cortisol with 1 mg of lysate of HEK-293 cells expressing 1 i p-HSD2 in the presence of 1 mM NAD+. Incubation lasted 16 h at 37°C. The steroids were extracted with ethylacetate, separated by thin layer chromatography (TLC) (SIL G-25 UV254, Macherey-Nagel, Oensingen, Switzerland) using a solvent system of 9:1 (v/v) chloroform:methanol and the band corresponding to cortisone was excised. The product was run for a second
chromatographic purification step on the same TLC system. A total of 250 μθί of
[1 ,2,6,7-3H]-cortisone was recovered. G-418 sulfate was purchased from Promega, Wallisellen, Switzerland. All other chemicals were purchased from Fluka AG, Buchs, Switzerland and of the highest grade available.
Untransfected HEK-293 cells do not show Ι ΐ β-HSDI activity. 1 i p-HSD2 mRNA was detectable by RT-PCR, but activity was not detectable upon incubation of radiolabeled Cortisol with cell lysate for 8 h. HEK-293 cells were transfected with the plasmid for expression of carboxy-terminally FLAG-epitope tagged 1 1 β-HSDI or 1 1 β- HSD2, respectively, as described previously [18]. Transfected cells were selected by cultivation in presence of 800 μg/mL of G-418. Non-resistant cells were removed by replacing the cell culture medium every third day for 3 weeks. From these cells, eight clones each were then selected and tested for protein expression, by
immunofluorescence analysis using mouse monoclonal anti-FLAG antibody M2
(Sigma) and goat anti-mouse antibody ALEXA 488, and 1 1 β-HSD activity. All of the eight clones of either Ι ΐ β-HSDI or 1 i p-HSD2 transfected cells showed similar expression and activity of the corresponding FLAG-epitope tagged Ι ΐ β-HSD construct.
HEK-293 cells stably transfected with either 1 1 β-HSDI or 1 i p-HSD2 were grown in 10 cm dishes to 90 % confluence. Cells were rinsed once with phosphate- buffered saline and resuspended in 2 mL of ice-cold buffer TS2 containing 100 mM NaCI, 1 mM EGTA, 1 mM EDTA, 1 mM MgCI2, 250 mM sucrose, 20 mM Tris-HCI, pH 7.4. For determination of oxidative activity of 1 1 β-HSD enzymes, cells were lysed by sonication and the cell lysate diluted 1 :12 in buffer TS2 (at 4 °C). Reactions were carried out in 96-well optical PCR reaction plates (Applied Biosystems, Foster City, CA) and tubes were capped during the reaction to avoid evaporation. Reactions were started by simultaneously adding 10 μΙ_ of cell lysate and 10 μΙ_ of TS2 buffer containing the appropriate concentration of the compound to be tested to 10 μΙ_ of TS2 buffer containing NAD+, 30 nCi of [1 ,2,6, 7-3H]-cortisol and unlabeled Cortisol to give a final concentration of 400 μΜ NAD+ and 10 nM Cortisol. Stock solutions of the compounds in methanol or in DMSO were diluted in TS2 buffer to yield the appropriate concentrations, whereby the concentration of methanol or DMSO in the reactions was below 0.1 %. Control reactions with or without 0.1 % of the solvent showed the same activity. After incubation at 37 °C for 10 min with shaking, 10 μΙ_ of stop solution containing 2 mMol unlabeled Cortisol and cortisone dissolved in methanol were added. Conversion of radiolabeled Cortisol was determined by separation of Cortisol and cortisone using TLC and a solvent system of 9:1 (v/v) chloroform: methanol, followed by scintillation counting [19]. In absence of inhibitors approximately 40 % of Cortisol was converted to cortisone.
Similarly, reductase activity was measured in a reaction containing NADPH, 30 nCi of [1 ,2,6, 7-3H]-cortisone and unlabeled cortisone, whereby final concentrations were 400 μΜ NADPH and 10 nM cortisone. No loss of 1 1 P-HSD2 activity was observed upon freezing of cell lysates for up to 1 month. In contrast, Ι ΐ β-HSDI activity declined after cell disruption, with a concomitant loss of affinity for its substrate but without any significant loss of apparent Vmax. Activities were determined measuring the conversion of either radiolabeled cortisone or Cortisol for 5-20 min using substrate concentrations in the range between 10 nM and 10 μΜ. Ι ΐ β-HSDI activities were measured immediately after cell disruption. All measurements included a negative control in absence of environmental compound and a positive control containing glycyrrhetinic acid at a final concentration of 10 μΜ. Results are expressed as mean ± S.D. and consist of at least three independent measurements.
Reduction οΠ Ιβ-HSDI Reduction of 11fi-HSD2
Concentration
Compound Activity (%) (average ± Activity (%) (average ±
(nmol/l)
standard deviation) standard deviation)
GA 200 3,7 ± 12,7 31 ,6 ± 15,3
2 200 12 ± 4,7 78,2 ± 2,2
3 200 16,2 ± 7,8 69,1 ± 0,9
4 200 0,9 ± 1 1 ,5 67,6 ± 2
5 200 7,5 ± 12,5 89,8 ± 0,2
7 200 7,7 ± 3,6 65,7 ± 10,2
8 200 10,4 ± 5,4 80,4 ± 1 1 ,7
1 1 200 9 ± 7,2 91 ,6 ± 8,8
12 1000 83,9 ± 1 ,7 93,6 ± 1 ,5
Reference List:
[1] R.A.Isbrucker, G.A.Burdock, Regul Toxicol Pharmacol 46 (2006) 167-192
[2] N.Nassiri Asl, H.Hosseinzadeh, Journal of Medicinal Plants 6 (2007) 1 -12 [3] M.Nassiri Asl, H.Hosseinzadeh, Phytotherapy Research 22 (2008) 709-724.
[4] F.Andersen, International Journal of Toxicology 26 (2007) 79-1 12.
[5] J.Rios et al., Studies in Natural Product Chemistry 22 (2000) 93-143.
[6] Y.H.Zhang et al., Immunology. 79 (1993) 528-534.
[7] Y.H.Zhang et al., Cell. Immunol. 162 (1995) 97-104.
[8] K.C.Nicolaou et al., JACS 124 (2002) 2245-2258.
[9] R.A.Schweizer et al., Mol. Cell Endocrinol. 212 (2003) 41-49.
[10] X.Su et al., J. Steroid Biochem. Mol. Biol. 104 (2007) 312-320.
[1 1] S.Diederich et al., Eur. J. Endocrinol. 142 (2000) 200-207.
[12] S.A.Latif et al., Mol. Cell Endocrinol. 243 (2005) 43-50.
[13] A.Ukil et al., J Immunol. 175 (2005) 1 161 -1 169.
[14] N.Abe, T.Ebina, N.lshida, Microbiol. Immunol. 26 (1982) 535-539.
[15] R.C.Rowe, P.J.Sheskey, S.C.Owen, Handbook of Pharmaceutical Excipients, 5 ed., Pharmaceutical Press, London, Chicago, 2006.
[16] United States Pharmacopeia, USP NF 2008 (United States
Pharmacopeia/National Formulary), 2008.
[17] European Directorate for the Quality of Medicines & HealthCare (EDQM),
European Pharmacopoeia, 2008.
[18] A.Odermatt et al., J. Biol. Chem. 274 (1999) 28762-28770.
[19] G.Escher et al., J. Exp. Med. 186 (1997) 189-198.
[20] D. Schuster et al., J. Med. Chem. 49 (2006) 3454-3466.
[21] X. Su et al., Bioorg. & Med. Chem. 12 (2004) 4439-4457.
[22] C. Brieskorn et al., Archiv der 303 (1970) 901 -4.
[23] A. Ech-Chahad et al. Tetrahedron Letters 46 (2005) 51 13-51 15.
[24] W. J. Boyle et al., Nature 423 (2003) 337-342.
[25] T. Suda et al., Bone 17 (1995)

Claims

Compounds of general Formula I,
wherein
R3a and R3b together are selected from =0, =NRa, =N-0-Ra, or
R3a and R3b are independently from one another selected from hydrogen,
-0-Ra, -0-C(=0)-Ra, -N(Ra)2, -NH-C(=0)-Ra and -NH-S(=0)2Ra; and
R11a and R11b together are selected from =0, =NRa, =NH, =N-0-Ra, =N-OH; or
R11a and R11b are independently from one another selected from hydrogen, -OH, -O-Ra, -O-C(=O)-Ra, -N(Ra)2, Ci-6alkyl, C2-8alkynyl, fluorine, chlorine, and bromine; and
a single or a double bond is present at position 12-13; and
the configuration at position 18 is R or S; and
X is selected from -C(=O)-1, -S(=O)2- and -C(=S)-; and
R30 is selected from hydrogen, -Ra, or -O-Ra; and
R31 is selected from -Rb, -NH2, -NHRb, -N(Rb)2, -NH-S(=O)2-Rb,
-NH-NH-C(=O)-NH2, -NH-NH-C(=O)-NHRb, -NH-NH-C(=O)-N(Rb)2; and each Ra independently of one another is selected from hydrogen or from optionally substituted hydroxyalkyl, Ci-6alkyl, C2-6alkenyl, C2-8alkynyl, -CF3, carboxylic acid, - (CH2)n-C6-i4aryl, -CH=CH-C6-i4aryl, -C≡C-C6-i4aryl, -(CH2)n-C5-i4heteroaryl, -CH=CH-C5-i4heteroaryl, -C≡C-C5-i4heteroaryl, -(CH2)n-C3-8cycloalkyl,
-CH=CH-C3-8cycloalkyl and -C≡C-C3-8cycloalkyl; and
each Rb independently of one another is selected from hydroxy or from optionally substituted, Ci^alkyl, C2-6alkenyl, C2-8alkynyl,, -(CH2)n-C3-8cycloalkyl,,-CF3,
-(CH2)n-C6-i4aryl; and
each n independently of one another denotes 0, 1 or 2; optionally in the form of their pharmaceutically effective salts, solvates, prodrugs, tautomers, racemates, enantiomers, diastereomers or mixtures thereof.
2. Compounds according to claim 1 , wherein
R11a and R11b together denote =0; and
a double bond is present at position 12-13.
3. Compounds according to one of claimsl to 2, wherein
R30 is selected from hydrogen, Ci-6alkyl, C2-6alkenyl, C2-8alkynyl, -CF3,
-(CH2)n-C6-i4aryl, -(CH2)n-C5-i4heteroaryl, and -(CH2)n-C3-8cycloalkyl.
4. Compounds according to claim 3, wherein
R30 is hydrogen.
5. Compounds according to one of claims 1 to 4, wherein
R3a is hydrogen and
R3b is selected from-0-Ra, -0-C(=0)-Ra, -N(Ra)2, -NH-C(=0)-Ra, and -NH- S(=0)2Ra.
6. Compounds according to claim 5, wherein
R3b is selected from -OH, -O-acetyl, -O-succinyl, -NH2, -NH-acetyl, -NH-succinyl, -NH-S(O)2CF3, -NH-S(O)2CH3 and -NH-S(O)2CH2CH2COOH.
7. Compounds according to one of claims 1 to 4, wherein
R3b is hydrogen and
R3a is selected from -O-Ra, -O-C(=O)-Ra, -N(Ra)2, -NH-C(=O)-Ra, and -NH- S(=O)2Ra.
8. Compounds according to claim 7, wherein
R3a is selected from -OH, -O-acetyl, -O-succinyl, -NH2, -NH-acetyl, -NH-succinyl, -NH-S(O)2CF3, -NH-S(O)2CH3 and -NH-S(O)2CH2CH2COOH.
9. Compounds according to one of claims 1 to 4, wherein
R3a and R3b together are selected from =0, =N-methyl, =NH, =N-O-methyl, and =N-OH.
10. A compound selected from the group consisting of
Λ/-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-[(3p,18p,20P)-3-amino-1 1-oxo-30-norolean-12-en-20-yl] urea,
Λ/-[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, Λ/-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, Λ/-[(3β, 18p,20P)-3-(methylsulfonamino)-11 -oxo-30-norolean-12-en-20-yl]urea,
/V-[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]urea,
/V-[(18p,20P)-3-(hydroxyimino)-1 1-oxo-30-norolean-12-en-20-yl] urea,
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl] urea,
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/ -[(3ββ, 18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V-methyl-/ -[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30- norolean-12-en-20-yl] urea
/V-hydroxy-/V-methyl-/V '-[(3β, 18p,20P)-3-(methylsulfonamino)-1 1 -oxo-30-norolean-12- en-20-yl]urea,
/V-hydroxy-/V-methyl-/ -[(18β,20β)-3, 1 1 -dioxo-30-norolean-12-en-20-yl]urea,
/V-hydroxy-/V-methyl-/ -[(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V -[(3p,18p,20P)-3-(acetyloxy)-1 1-oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V '-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea,
N-hydroxy-N -[(3p,18p,20P)-3-(acetamino)-1 1-oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V -[(3β, 18p,20P)-3-amino -1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-hydroxy-/V '-[(3β, 18p,20P)-3-(succinyloxy)-11 -oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V -[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-hydroxy-/V -[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-11 -oxo-30-norolean-12-en- 20-yl]urea,
/V-hydroxy-N'-[(3p, 18p,20P)-3-(methylsulfonamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-hydroxy-/V -[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]urea,
N-hydroxy-N -[(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/V '-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/V '-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea,
/V-methyl-/V '-[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, N-methyl-/V -[(3p,18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]urea,
N-methyl-/V -[(3p,18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/V '-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methyl-/V '-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-11 -oxo-30-norolean-12-en- 20-yl]urea,
N-methyl-N -[(3p,18p,20P)-3-(methylsulfonamino)-1 1-oxo-30-norolean-12-en-20- yl]urea,
/V-methyl-/V -[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]urea,
N-methyl-N -[(18p,20P)-3-(hydroxyimino)-11 -oxo-30-norolean-12-en-20-yl]urea, /V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]urea, N-methylsulfonyl-N -[(3p,18p,20P)-3-(acetamino)-1 1-oxo-30-norolean-12-en-20- yl]urea,
/V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]urea, /V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20- yl]urea,
/V-methylsulfonyl-/V '-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30-norolean- 12-en-20-yl]urea,
N-methylsulfonyl-N -[(3p,18p,20P)-3-(methylsulfonamino)-11 -oxo-30-norolean-12-^ 20-yl]urea,
/V-methylsulfonyl-/ -[(18β,20β)-3, 1 1 -dioxo-30-norolean-12-en-20-yl]urea,
N-methylsulfonyl-N -[(18p,20P)-3-(hydroxyimino)-1 1-oxo-30-norolean-12-en-20-yl]^ Λ/-[(3β, 18p,20P)-3-hydroxy-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-amino-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonylurea,
Λ/-[(3β, 18p,20P)-3-(acetamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ -[(trifluoromethyl) sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(succinyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl)sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(succinylamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl)sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl)sulfonyl]urea,
Λ/-[(3β, 18p,20P)-3-(methylsulfonamino)-1 1 -oxo-30-norolean-12-en-20-yl]-/ - [(trifluoromethyl)sulfonyl]urea,
N-[(18p,20P)-3,1 1 -dioxo-30-norolean-12-en-20-yl]-N -[(trifluoromethyl)sulfony^
/V-[(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20-yl]-/V - [(trifluoromethyl)sulfonyl]urea,
(3β, 18p,20P)-3-hydroxy-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-(acetyloxy)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-amino-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one,
(3β, 18p,20P)-3-(acetamino)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-(succinyloxy)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 -one, (3β, 18p,20P)-3-(succinylamino)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en-1 1 - one,
(3β, 18p,20P)-3-(trifluoromethylsulfonamino)-20-(N-methylsulfonyl-amino)-30-norolean- 12-en-1 1 -one,
(3β, 18p,20P)-3-(methylsulfonamino)-20-(/V-methylsulfonyl-amino)-30-norolean-12-en- 1 1 -one,
(18β,20β)-3, 1 1 -dioxo-30-norolean-12-en-20-yl]urea,
(18p,20P)-3-(hydroxyimino)-1 1 -oxo-30-norolean-12-en-20-yl]urea,
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-/ -(phenylmethyl) urea, and
Λ/-[(3β, 18p,20P)-3-(acetyloxy)-1 1 -oxo-30-norolean-12-en-20-yl]-1 ,2-hydrazine- dicarboxamide.
1 1 . Compounds, or the pharmacologically effective salts thereof, according to one of the claims 1 to 10, as medicaments.
12. Use of compounds of general formula I according to one of the claims 1 to10 for preparing a medicament for the treatment and/or prevention of chronic inflammatory diseases, autoimmune diseases, skin diseases, bone diseases, metabolic diseases, infectious diseases and cancer.
13. Pharmaceutical preparation, containing as active substance one or more compounds of general formula I according to one of the claims 1 to 10, or the pharmacologically effective salts thereof, optionally in combination with conventional excipients and/or carriers.
EP11740948.2A 2010-08-10 2011-08-09 Glycyrrhetinic acid amine analogues for use in the treatment of inflammation, infectious diseases, cancer, autoimmune diseases, skin diseases, bone diseases and metabolic diseases Withdrawn EP2632933A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP11740948.2A EP2632933A1 (en) 2010-08-10 2011-08-09 Glycyrrhetinic acid amine analogues for use in the treatment of inflammation, infectious diseases, cancer, autoimmune diseases, skin diseases, bone diseases and metabolic diseases

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP10172411 2010-08-10
PCT/EP2011/063702 WO2012020021A1 (en) 2010-08-10 2011-08-09 Glycyrrhetinic acid amine analogues for use in the treatment of inflammation, infectious diseases, cancer, autoimmune diseases, skin diseases, bone diseases and metabolic diseases
EP11740948.2A EP2632933A1 (en) 2010-08-10 2011-08-09 Glycyrrhetinic acid amine analogues for use in the treatment of inflammation, infectious diseases, cancer, autoimmune diseases, skin diseases, bone diseases and metabolic diseases

Publications (1)

Publication Number Publication Date
EP2632933A1 true EP2632933A1 (en) 2013-09-04

Family

ID=44629815

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11740948.2A Withdrawn EP2632933A1 (en) 2010-08-10 2011-08-09 Glycyrrhetinic acid amine analogues for use in the treatment of inflammation, infectious diseases, cancer, autoimmune diseases, skin diseases, bone diseases and metabolic diseases

Country Status (2)

Country Link
EP (1) EP2632933A1 (en)
WO (1) WO2012020021A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108276468B (en) * 2018-03-13 2019-12-31 中国农业科学院兰州畜牧与兽药研究所 Glycyrrhetinic acid ester, preparation method thereof and application thereof in preparation of antiviral drugs
WO2019222800A1 (en) * 2018-05-21 2019-11-28 Hudson Institute of Medical Research Methods for the treatment or prevention of autoimmune or autoinflammatory diseases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2012020021A1 *

Also Published As

Publication number Publication date
WO2012020021A1 (en) 2012-02-16
WO2012020021A9 (en) 2012-05-10

Similar Documents

Publication Publication Date Title
US8476315B2 (en) N-hydroxy C29-amide derivatives of oleandrane
EP1278763B1 (en) 3-nitrogen-6,7-dioxygen steroids and uses related thereto
CA2869783C (en) 2.2-difluoropropionamide derivatives of bardoxolone methyl, polymorphic forms and methods of use thereof
TWI584808B (en) Antioxidant inflammation modulators: c-17 homologated oleanolic acid derivatives
EP2576585B1 (en) Modified c-3 betulinic acid derivatives as hiv maturation inhibitors
EP2814834B1 (en) C-3 cycloalkenyl triterpenoids with hiv maturation inhibitory activity
BR112015027012B1 (en) POLYMORPHIC FORMS OF A 2,2-DIFLUOROPROPIONAMIDE COMPOUND OF BARDOXOLONE METHYL, PHARMACEUTICAL COMPOSITION COMPRISING THE SAME AND THERAPEUTIC OR PROPHYLACTIC USE THEREOF
CA2902513A1 (en) C-3 alkyl and alkenyl modified betulinic acid derivatives useful in the treatment of hiv
JP6549544B2 (en) Novel compounds for treating cancer and other diseases
Salomatina et al. Synthesis of novel 2-cyano substituted glycyrrhetinic acid derivatives as inhibitors of cancer cells growth and NO production in LPS-activated J-774 cells
Khusnutdinova et al. Inhibition of alpha-glucosidase by synthetic derivatives of lupane, oleanane, ursane and dammarane triterpenoids
JP2017521459A (en) Hydroxysteroid compounds, intermediates thereof, methods for their preparation, compositions and uses
CA3013417C (en) C-3 and c-17 modified triterpenoids as hiv-1 inhibitors
EP2632933A1 (en) Glycyrrhetinic acid amine analogues for use in the treatment of inflammation, infectious diseases, cancer, autoimmune diseases, skin diseases, bone diseases and metabolic diseases
EP2258713A1 (en) Antiviral Triterpene Derivatives
Smirnova et al. Synthesis and evaluation of 29-norcycloartane triterpenoids as α-glucosidase inhibitors
WO2012020019A1 (en) Novel triterpene derivatives
WO2013091144A1 (en) Propenoate derivatives of betulin
Czollner et al. EP 2228380 Novel Triterpene derivatives
Kaledina et al. Synthesis and properties of 2-cyanoethoxy derivatives of dammarane triterpenoids
Classen-Houben et al. Kosma et a
Yang et al. Steroidal saponins with anti-inflammatory activity
WO2016077569A1 (en) C17-aryl substituted betulinic acid analogs

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130626

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20141111