MXPA06012870A - Substantially pure 2-{[2-(2- methylamino -pyrimidin-4- yl)-1h- indole-5- carbonyl] -amino}-3 -(phenylpyridin -2-yl-amino)- propionic acid as an ikb kinase inhibitor. - Google Patents

Substantially pure 2-{[2-(2- methylamino -pyrimidin-4- yl)-1h- indole-5- carbonyl] -amino}-3 -(phenylpyridin -2-yl-amino)- propionic acid as an ikb kinase inhibitor.

Info

Publication number
MXPA06012870A
MXPA06012870A MXPA06012870A MXPA06012870A MXPA06012870A MX PA06012870 A MXPA06012870 A MX PA06012870A MX PA06012870 A MXPA06012870 A MX PA06012870A MX PA06012870 A MXPA06012870 A MX PA06012870A MX PA06012870 A MXPA06012870 A MX PA06012870A
Authority
MX
Mexico
Prior art keywords
compound
effective amount
ikk
inhibitors
patient
Prior art date
Application number
MXPA06012870A
Other languages
Spanish (es)
Inventor
David J Aldous
Paul Joseph Cox
Olaf Ritzeler
El-Bdaoui Haddad
Original Assignee
Aventis Pharma Inc
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 Aventis Pharma Inc filed Critical Aventis Pharma Inc
Publication of MXPA06012870A publication Critical patent/MXPA06012870A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pulmonology (AREA)
  • Virology (AREA)
  • AIDS & HIV (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Otolaryngology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention is directed to the substantially pure compound of formula (A), or pharmaceutically acceptable salt, or solvate of said compound; to a pharmaceutical composition comprising a pharmaceutically effective amount of the compound of formula (A), and a pharmaceutically acceptable carrier; and the use of a compound of formula (A) having activity as an inhibitor, preferably a selective inhibitor, of IkB (IKK), particularly IKK-2, and methods related thereto.

Description

ACID 2-. { [2- (2-METHYLAMINOPYRIMIDIN-4-IL) -lH-INDOL-5-C ARBONIL] AMINO} -3- (FENILPIRIDIN-2-INO) PROPIONIC SUBSTANTIALLY PURE AS INHIBITOR OF THE KINASE I? B FIELD OF THE INVENTION This invention is directed to an indole derivative, to its preparation, to a pharmaceutical composition comprising the compound, to its use and to its intermediates.
BACKGROUND OF THE INVENTION NF-? B is a heterodimeric transcription factor that regulates the expression of multiple inflammatory genes. The expression of more than 70 known proteins is transcriptionally regulated by the binding of NF-? B to specific sequence elements in the promoter region of these genes (Baeuerle and Baichwal, Advances in Immunology, 65: 111-137, 1997). NF-? B has been implicated in many pathophysiological processes, including angiogenesis (Koch et al., Nature, '376: 517-519, 1995), atherosclerosis (Brand et al, J Clin Juv, 97: 1715-1722, 1996), endotoxic shock and sepsis (Bohrer et al, J. Clin, tnv, 100: 972-985, 1997), inflammatory bowel disease (Panes et al, Am. J. Physiol, 269: H1955-H1964, 1995) , damage by ischemia / reperfusion (Zwacka et al, Nature Medicine, 4: 698-704, 1998), and allergic lung inflammation (Gosset et al, Int. Arch. Allergy Immunol, 106: 69-77, 1995). Therefore, the inhibition of NF-? B by acting on the regulatory proteins in the NF-? B activation pathway represents an attractive strategy to generate anti-inflammatory therapeutic compounds due to the central role of NF-? B in inflammatory disorders.
The I? B kinases (IKK) are key regulatory signaling molecules that coordinate the activation of NF-? B. Two IKK, IKK-1 (IKK-a) and IKK-2 (D K-ß), are structurally exclusive kinases containing an N-terminal kinase domain with a dual serine activation loop, a leucine zipper domain, and a helix-loop-helix domain and a C-terminal serine cluster. IKK enzymes show relatively little sequence homology to other kinases, and early profiles with known kinase inhibitors have not identified compounds with surprising potency. Kinetic analyzes show that IKK-2 binds and phosphorylates I I Ba and I I Be with high and relatively equal affinities (Heilker et al, 1999). Recombinant IKK-2 phosphorylates I-Ba 26-42 peptide with almost the same affinity as full-length I? Ba, however, the complex of the native IKK enzyme phosphorylates the full length I? Ba in a manner 25,000 times more efficient, which suggests that there are important regulatory sequences in the C-thermal region of I? Ba, or other regulatory proteins in the complex of the IKK enzyme that accelerate the rate of catalysis (Burke et al, Journal of Biological Chemistry, 274: 36146-36152, 1999). The phosphorylation of I? Ba occurs through a random sequential kinetic mechanism, which means that ATP or I? Ba can bind ppmero to IKK-2, and that both must be bound before phosphorylation of I can occur. Ba (Peet and Li, Journal of Biological Chemistry, 274: 32655-32661, 1999) IKK-2 binds ATP with an exclusively high affinity (K? = 130 nM), compared to other septen threonine-kinase, such as p38 and JNK, which indicates perhaps a Exclusive ATP binding pocket that reflects the relatively low activity against muhco inhibitors of broad specificity when tested against IKK-2. To date, no crystal structure of IKK-2 has been indicated. However, homology modeling has identified 3 structural domains, which include a N-thermal cmase domain with an activation loop, a leucma zipper domain that probably mediates the formation of homo / heterodimers of IKK-1 and IKK-2, and a C-terminal hehce-loop-hehce with a pca tail in sepna. The activation of IKK-2 depends on the phosphorylation of serine 177 and 181 in the activation loop or T. Mutations of the alamna suppy the activity, whereas the glutamate mutations produce a constitutively active enzyme (Mercuno et al, Science , 278: 860-866, 1997; Delhase et al, Science, 284: 30-313, 1999) IKK-1 and IKK-2 both appear as heterodimers and as homodimers of IKK-2, and are associated with a cytoplasmic enzyme complex of 700-900 kDa called the "IKK signaling" (Mercurio et al, Science, 278: 860-866, 1997). Another component, IKKAP-1 or NEMO / DK ?, has no apparent catalytic function, but is directly associated with IKK-2 and is necessary for the complete activation of NF-? B (Mercurio et al., Mol Cell Biol, 19: 1526-1538, 1999). It has been shown that many immunological mediators and inflmatenes, including TNFa, hpoposaccharide (LPS), JJL-1/3, CD3 / CD28 (presentation to the antigen), CD40L, FasL, viral infection, and oxidative stress, lead to the activation of NF -? B. Although the receptor complexes that transduce these diverse stimuli are very different in their protein components, it is understood that each of these stimulation events leads to the activation of the IKK and NF-? B.
The IKK complex appears to be the central integrator of various mflamatory signals leading to the phosphorylation of IκB. IKKs are activated in serine residues by upstream kinases, including the kinase inducing NF-? B, NIK (Malinin et al, Nature, 385: 540-544, 1997), and MEKK-1 (Yujiri et al, Science, 282: 1911-1914, 1998). The differential activities of NIK and MEKK-1 remains unclear, although initial data indicate that these kinases can preferentially activate IKK-1 and IKK-2, respectively. Activated IKK phosphorylates a cytoplasmic inhibitory protein, I? B, which binds to NF-? B, thereby masking a nuclear localization signal present in the ReI proteins (Cramer et al, Structure, 7: R1-R6, 1999). IKK phosphorylation of I? B in serines 32 and 36 forms a structural motif recognized by the E3 ligase, / 3TRcP (Yaron et al, Nature, 396: 590-594, 1998). The 3TRcP anchor produces the formation of a ligase complex that polyubiquitin I? B, thereby marking it for degradation by the 26S proteosome. Then free NF-? B is identified by nuclear transport proteins, which translocate it to the nucleus where it can be associated with sequence-specific regulatory elements on gene promoters.
Although both kinases can phosphorylate I? B in vitro, the first studies using genetic mutants indicate that IKK-2, but not DK-1, is essential for the activation of NF-? B by proinflammatory stimuli, such as 11- 1/3 and TNFo; In addition, only the catalytically inactive mutants of IKK-2 block the expression of genes regulated by NF-? B, such as the monocyte chemotactic protein (MCP-1) and the intercellular adhesion molecule (ICAM-1) (Mercurio et al, Science, 278: 860-866, 1997). Studies of knockout animals (inactivated) to detect IKK-1 and IKK-2 demonstrate these initial findings (Hu et al, Science, 284: 316-320, 1999, Li et al., Genes &Development, 13: 1322-1328 , 1999, Li et al, Science, 284: 321-324, 1999, Takeda et al, Science, 84: 313-316, 1999, Tanaka et al, Immunity, 10: 421-429, 1999). The IKK-1 animals were born alive but died after hours. The puppies showed skin abnormalities due to defective proliferation and differentiation, but did not show a large deficiency in the cytokine-induced activation of NF-? B. By contrast, IKK-2"embryos died on day 14-16 of pregnancy due to hepatic degeneration and apoptosis, which showed a striking resemblance to that observed in animals with a K A knockout (inactivated) (Beg et al, Nature, 376: 167-170, 1995). In addition, the embryonic fibroblasts of animals IKK-2"" showed a very reduced activation of NF-? B after stimulation with cytokines, whereas the rKK-1"" no.
Therefore, experiments in cells and animals indicate that D K-2 is a central regulator of the proinflammatory role of NF-? B, whereas IKK-2 is activated in response to immunological stimuli and inflmatenes and signaling pathways. Many of these immune and inflammatory mediators, including IL-1 | 3, LPS, TNFa, CD3 / CD28 (presentation to the antigen), CD40L, FasL, vinca infection, and oxidative stress, play an important role in respiratory diseases. In addition, the ubiquitous expression of NF-? B, together with its response to multiple stimuli, means that almost all cell types present in the lung are a potential target for access to anti-NF-? B / IKK-2 therapy. . This includes the alveolar epithelium, the mast cells, the fibroblasts, the vascular endoteho, and the infiltration leukocytes, including neutrophils, macrophages, lymphocytes, eosinophils, and basophils. By inhibiting the expression of genes, such as c? Cloox? Genasa-2 and 12-l? Pox? Genasa (synthesis of inflammatory mediators), peptide transporter TAP-1 (antigen processing), MHC class mvanables chains I H-2K and class II (presentation to the antigen), E-selectin and vascular cell adhesion molecule (leukocyte recruitment), microtequins-1, 2, 6, 8 (cytokines), RANTES, eotaxin, GM- CSF (chemokines), and superoxide dismutase and NADPH-quinone-oxidoreductase (reactive oxygen species), it is believed that inhibitors of D K-2 show a broad anti-inflammatory activity.
Patent application WO 94/12478, the content of which is incorporated herein by reference, discloses, inter alia, indole derivatives that inhibit the aggregation of blood platelets. Patent applications WO 01/00610 and WO 01/30774, the content of which is incorporated herein by reference, disclose indole derivatives and benzimidazole denones, which are capable of modulating NF-? B. As described above, NF-? B is a heterodimpe transcription factor that is capable of activating a large number of genes encoding, inter alia, promlammatory cytokines, such as IL-1, IL-2, TNFa or IL-6. The NF-? B is present in the cell cytosol, where it forms a complex with its inhibitor that appears in nature, I? B. The stimulation of the cells, for example by means of cytokines, leads to the I? B being phospholized and, subsequently, degrading in a proteolytic manner. This proteolytic degradation leads to the activation of NF-? B, which then migrates to the nucleus of the cell, where it activates a large number of proinflammatory genes.
In diseases such as rheumatoid arthritis, osteoartantis, asthma, chronic obstructive pulmonary disorder (COPD), nnitis, multiple sclerosis, cardiac infarction, Alzheimer's disease, type II diabetes, inflammatory bowel disease or atherosclerosis, NF-? B is activated beyond its normal degree. It is also indicated that the inhibition of NF-? B is useful for treating cancer, on its own, or in conjunction with cytostatic therapy. The inhibition of the signal chain that activates NF-? B at various points, or directly interfering in the transcnption of the gene by glucocorticoids, saccylates or gold salts, has proven to be useful in treating rheumatism.
The first step in the signal cascade mentioned above is the degradation of I? B. This phosphonylation is regulated by the specific IκB kinase. To date, it is known that inhibitors of the I? B kinase, often have the disadvantage of not being specific to inhibit only one class of kinase. For example, most inhibitors of the I? B inhibit different pools at the same time, because the structures of the catalytic domains of these cells are similar. Accordingly, the inhibitors act in an undesirable way on many enzymes, including those that possess vital function.
Chronic obstructive pulmonary disease (COPD) is a debilitating inflammatory disease of the lungs, characterized by a progressive development of a limitation of air flow that is not fully reversible (Pauwels et al, 2001). The limitation of air flow is associated with an abnormal inflamatone response of the lungs to harmful particles or gases, caused mainly by cigarette smoking. Although COPD affects the lungs, it also produces significant systemic consequences. The term "COPD" includes chronic obstructive bronchitis, with obstruction of the small airways, and emphysema, with an enlargement of the empty spaces and the destruction of the pulmonary parenchyma, the loss of pulmonary elasticity, and the closure of the respiratory tract. little. Chronic bronchitis, by contrast, is defined by the presence of a productive cough (due to mucus hypersecretion) lasting more than three months for more than two consecutive years. There is some epidemiological evidence that mucus hypersecretion is accompanied by an obstruction of air flow, perhaps as a result of obstruction of the peripheral airways. Most patients with COPD have all three pathological conditions (chronic obstructive bronchitis, emphysema, and mucus plug formation), but the relative extent of emphysema and obstructive bronchitis within individual patients may vary, Vestbo et al, 1996; Barnes, 2004a, Barnes, 2004b; Hogg, 2004 In industrialized countries, the habit of smoking cigars is responsible for most cases of COPD, but in developing countries other environmental pollutants (in particular, with sulfur dioxide and particulates) and certain workplace chemicals (such as cadmium), are important causes. Being a passive smoker is also a risk factor.
Patients with COPD are predisposed to exacerbations, that is, an acute worsening of their respiratory symptoms. An exacerbation of COPD is an event in the natural development of the disease, characterized by a change in baseline of the patient's dyspnea, cough and / or sputum beyond a target variability, sufficient to warrant a change in treatment (Rodpguez-Roisin, 2000; Burge and Wedzicha, 2003).
It is believed that tracheobronchial infections are a common cause of the exacerbation of COPD, although there is controversy about the nature of the infectious agent, as well as its exact role (Wedzicha, 2002, White et al, 2003). In addition, COPD exacerbations are evidently associated with the levels of respirable particles and environmental pollutants in the air, and these have been associated with the proportion of hospital admissions (Rennard and Farmer, 2004).
The frequency of exacerbations is associated with the severity of the disease in COPD. Exacerbations may adversely affect the natural histone of these disorders, perhaps contributing to a greater proportion of decreased lung function, systemic effects and premature mortality. Unfortunately, to date, there is no widely accepted definition of what constitutes an exacerbation of COPD (Rodpguez-Roism, 2000). The intensity and duration of the increased symptoms required to qualify as an "exacerbation" are difficult to define. Indeed, there are vain coexisting definitions, and many clinical trials employ substantially different criteria, or poorly describe the definition (s) used to diagnose the exacerbation. The most widely cited clinical trial used in the characterization of acute exacerbation of COPD is described by Anthonisen et al. (1987). In that study, the exacerbations were divided into three groups: exacerbations of type 1 are characterized by a greater lack of breath, a greater volume of sputum, and a new or higher sputum purulence; Type 2 includes two of these symptoms; and type 3 consists of any one of these symptoms together with at least one other characteristic, including sore throat or nasal discharge in the last few days; fever sm explanation; greater whistling of the breath; more cough; or a 20% increase in respiratory or cardiac frequency compared to the baseline. These trials have been used as a reference since then, and all proposed etiologies of the exacerbation need to establish their relationship with these key characteristics.
It is also indicated that the inhibition of NF-? B is useful for treating hypoprophylactic diseases, eg, solid tumors and leukemias, on its own or in conjunction with cytostatic therapy. The inhibition of the signal chain that activates NF-? B in Several points, or interfering directly in the transcription of the gene by glucocorticoids, saccitates or gold salts, has proved useful in treating rheumatism.
Patent application WO 01/30774 discloses indole denudates, and US application No. 10 / 642,970 discloses denudates of mdol and benzimidazole which are capable of modulating NF-? B and which show a strong inhibitory effect on the cmasa I? B. In particular, the application of US Ser. No. 10 / 642,970 discloses denudates of indole and benzimidazole of formula (I), their preparation, (i) pharmaceutical compositions containing these compounds, and methods for the prophylaxis and therapy of a disease associated with increased activity of the IκB lañase kinase, which comprises administering these compounds. In addition, US Application Serial No. 10 / 642,970 describes the following compounds of formulas (B), (C) and (D): (B) (compound 43), (C) (compound 32); Y (D) (compound 48). However, US Application Ser. No. 10 / 642,970 does not specifically disclose the compound of formula (I) wherein M is N, Rl is hydrogen, R2 is carboxyl (-COOH), R3 is methyl, R4 is p? pdm-2-? lo, Rl 1 is hydrogen, and X is CH.
In view of the above, there is a need for an inhibitor of IκB kinase that acts through the selective inhibition of IKK, in particular an inhibitor of IKK-2. It would also be desirable to obtain this inhibitor which shows a localized activity, as opposed to a systemic activity. This inhibitor should be useful for treating a patient suffering from or suffering from pathological disorders (diseases) mediated by IKK-2, eg, asthma or chronic obstructive pulmonary disorder (COPD), which can be improved by targeted administration of the inhibitor.
SUMMARY OF THE INVENTION The present invention is directed to a compound having activity as an inhibitor, preferably as a selective inhibitor, of I? B (IKK), in particular IKK-2, and to a composition and methods related thereto.
In particular, the present invention is directed to the substantially pure compound of formula (A): (A) or a pharmaceutically acceptable salt, or solvate of said compound.
In addition, the present invention is directed to a pharmaceutical composition comprising a pharmaceutically effective amount of the compound of formula (A), and a pharmaceutically acceptable carrier.
In addition, the present invention is directed to the use of a compound of formula (A) as an inhibitor of kinase I? B.
BRIEF DESCRIPTION OF THE DRAWINGS FIGURE 1 shows a drawing of imaging studies in mice in the NF-? B-luc? Ferase reporter mouse model, where animals are treated with the vehicle / PBS solution. (animals with negative control), and where some animals show activation of NF-? B induced by IL-lj8 without any compound present [vehicle / IL-1/3] or with increasing doses (0.3 mpk, 1 mpk, 3 mpk and 10 mpk) of the compound (A) or compound (B).
FIGURE 2 shows a drawing of imaging studies in mice in the NF-? B-luc? Ferase reporter mouse model, showing the bioluminescence levels for animals treated with the vehicle / PBS solution (control animals negative), and where some animals have NF-? B activation induced by L-lβ without any compound present [vehicle / EL-1/3] or with increasing doses (0.3 mpk, 1 mpk, 3 mpk and 10 mpk) of the compound (A) or compound (B).
FIGURE 3, to the left, shows a graph of Compound compound levels (A) after an instillation í.t. 0.3 mg / kg of compound (A) in lung and plasma tissue; and on the right shows a graph of Compound compound levels (A) and compound (B) after an instillation i.t. of 0.3 mg / kg of compound (B) in lung and plasma tissue.
FIGURE 4, on the left, shows a graph of the pulmonary exposure of compound (A) after the administration of increasing doses (0.01 mpk, 0.03 mpk, 0.10 mpk and 0.30 mpk) of the compound (TO); and to the right shows a graph of the plasma exposure of the compound (A) after the administration of increasing doses (0.01 mpk, 0.03 mpk, 0.10 mpk and 0.30 mpk) of the compound (A).
FIGURE 5, to the left, shows a graph of the pulmonary exposure of compounds (A) and (B) after the administration of increasing doses (0.01 mpk, 0.03 mpk, 0.10 mpk and 0, 30 mpk) of the compound (B); and to the right shows a graph of the plasma exposure of compounds (A) and (B) after the administration of increasing doses (0.01 mpk, 0.03 mpk, 0.10 mpk and 0.30 mpk) of the compound (B) DETAILED DESCRIPTION List of abbreviations As used before and throughout the description of the invention, it will be understood that the following abbreviations, unless otherwise indicated, have the following meanings: Boc20 di-re-butyl carbonate DIEA NN-dnsopropylethylamine DMAP 4-d? Met? Lammop? Pd? Na DMF dimethylformamide DMSO dimethylsulfoxide ESI-MS electroesis ionization mass spectrometry FAB-MS fast atom bombardment mass spectrometry HATU 0- (7-azabenzotnazol-l-? l) -N hexafluorophate, N, N ', N'-tetramethyluronium HPLC high performance liquid chromatography PBS phosphate buffered saline, i.n. intranasally intra orally PO.p. mtrapeptoneal Í.t. mtratracheal microcystin-LR hepatic toxin produced by certain cyanobacteria of the genus Anabaena and Oscillatone mbr mihbares mpk mg / kg HEPES acid 4- (2-hydroxyl et? l) -1-piperazinetansulfomco DTT dithiothreitol ATP adenosm triphosphate streptavidin-conjugated HRP streptavidma-horseradish peroxidase TMB tetramethylbenzidine pfu plate-forming units MDI metered dose inhaler DPI dry powder inhaler Definitions As used before and throughout the description of the invention, it will be understood that the following terms and expressions, unless otherwise indicated, have the following meanings.
A "compound of the invention", and the equivalent expressions, mean the compound of formula (A), as described hereinbefore, the expression of which includes the pharmaceutically acceptable salt and the solvate, eg, hydrate. Similarly, the reference to intermediates, as well as whether they themselves claim or not, is intended to include salts and solvates, when the context permits. In order to increase clandad, the concrete cases in which the context permits are indicated, sometimes, in the text, but these cases are purely illustrative and do not intend to exclude other cases in which the context allows it.
"Treat" or "treatment" means prevention, partial relief or cure of the disease. The compound and the compositions of this invention are useful for treating disorders that are characterized by the activation of NF-? B and / or higher levels of cytokines and mediators that are regulated by NF-? B which include, but are not limited to TNFa and IL-lß. The inhibition or suppression of NF-? B and / or genes regulated by NF-? B, such as TNFo: can occur locally, for example, within certain tissues of the subject, or more extensively throughout the subject being treated. is treating for this disease. The inhibition or suppression of NF-? B and / or genes regulated by NF-? B, such as TNFa, can be produced by one or more mechanisms, eg, by inhibition or suppression of any stage of the pathway (s) ( s), as the inhibition of IKK. The term "NF-? B associated disorder" refers to diseases that are characterized by the activation of NF-KB in the cytoplasm (eg, after the phosphorylation of I? B). The term "disorder associated with TNFa" is a disorder characterized by higher levels of TNFa. In the present specification, the term "NF-? B associated disorder" includes a disorder associated with TNFa, but is not limited thereto, since NF-? B is involved in the activity and upregulation of other proteins and genes. promflamatonos. The term "inflmaterine or immunological diseases or disorders" is used herein to include disorders associated with NF-κB and disorders associated with TNFα, eg, any disorder, disease or condition that is associated with the release of NF. -? B and / or higher levels of TNFa, including disorders as described herein.
A "patient" includes humans and other mammals.
A "pharmaceutically effective amount" is intended to disclose an amount of a compound, composition, medicament or other active ingredient effective to produce the desired therapeutic effect.
It is intended that "substantially pure" refers to the compound that is substantially free of biological or chemical constituents, e.g., isolated from a biological or chemical composition wherein the biological or chemical components are coisolated, and wherein the analytical purity of the compound it is prehensively at least 70%. It is more preferred when the analytical purity is at least 90%; it is even more preferred when the analytical purity is at least 95%; it is also intended that "substantially pure" refers to the compound when it is substantially free of prodrugs, e.g., compound (B).
The invention also relates to a process for preparing the compound of formula (A), as shown in the following scheme.
The starting compounds for the chemical reactions are known, or can be prepared easily using methods known in the literature. U.S. Patent Application Serial No. 10 / 642,970 describes the preparation of the indolecarboxylic acid intermediate (compound 8) used in the coupling step (vi) above. The compounds of formulas (B), (C) and (D) are prepared as described in US Patent Application Serial No. 10 / 642,970, which is incorporated herein by reference.
Peptide chemistry coupling methods that are well known to those skilled in the art (see, eg, Houben-Weyl, Methoden der Organischen Chemie [Methods of Chemistry Organic], volumes 15/1 and 15/2, Georg Thieme Verlag, Stuttgart, 1974, the content of which is incorporated herein by reference), are advantageously used to condense the compounds. Compounds such as carbomldnmidazole, carbodnmides such as dicyclohexylcarbodnmide or dpsopropylcarbodiimide (DIC), tetrafluoroborate 0 - ((carbon (ethoxycarbonyl) methanol) ammo) -N, N, N ', N'-tetramethyluromone (TOTU) or poh (phosphoric acid) (PPA) are suitable for use as condensing agents or coupling reagents. The condensations can be carried out under conventional conditions. In the condensation it is necessary that the ammo groups present that do not react are protected with reversible protective groups. The same applies to carboxyl groups which are not involved in the reaction, these groups being preferably present, during the condensation, as esters of alkyl (C? -C6), benzyl esters or tert-butyl esters. No protection of the amino group is necessary if the amino groups are still present in the form of precursors, such as nitro groups or cyano groups, and are only formed by hydrogenation after condensation. After condensation, the protective groups that are present are removed in a suitable manner. For example, the groups N02 (protection of guanidino in amino acids), benzyloxycarbonyl groups and benzyl groups in benzylic esters can be removed by hydrogenation. Tere-butyl-type protecting groups are removed under acidic conditions, while the 9-fluoren-1-methyl-1-carbon radical is removed using secondary amines.
The present invention also relates to a pharmaceutical composition comprising a pharmaceutically effective amount of the compound of formula (A), and a pharmaceutically acceptable carrier.
EMBODIMENTS Due to the pharmacological properties of the compound according to the invention, it is suitable for the treatment of all patients suffering or suffering from disorders that can be improved by the targeted administration of an I? B inhibitor to a site in the that the treatment is best done by localized versus systemic activity, eg, asthma or chronic obstructive pulmonary disorder (COPD).
In practice, the compound of the present invention can be administered in a pharmaceutically acceptable dosage form to humans and other animals by topical or systemic administration, including oral, inhalation, rectal, nasal, buccal, sublingual, vaginal, colonic administration , parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural), intracisternal and intrapeptoneal. It will be appreciated that the preferred route may vary, for example, according to the disorder of the recipient.
The intranasal, intratracheal or inhalation administration, as well as the aerosol formation, are specific methods of administering the compound according to the invention.
Combination therapies can improve efficacy and decrease the risk of secondary effects, compared with increasing the dose of a single agent. IKK inhibitors can be combined with bronchodilators including, but not limited to short-acting β2-agonists, long-acting β2-agonists, such as salmeterol and formoterol; anticholinergic agents, such as ipratropium bromide and tiotropium bromide. IKK inhibitors can also be combined with methylxamines, such as theophylline. IKK2 inhibitors can be combined with various anti-inflammatory therapies including, but not limited to immunomodulators directed to various stages of the inflammatory cascade, and aimed at alleviating inflammatory processes. These therapies include, but are not limited to: (A) cellular recruitment inhibitors and toxic inflammatory mediators including, but not limited to, phosphodiesterasease-4 inhibitors.; inhibitors of mitogen-activated protein kinase p38; biopharmaceutical compounds, such as the antifactor alpha of tumor necrosis, antnterleuquma-8, and monocyte chemoattractant antiprotein 1; inhibitors of adhesion molecules and chemotactic factors; and molecules that interfere with cell survival and apoptosis ehmination; (B) inhibitors of proteolytic enzymes including, but not limited to inhibitors of seroma proteases derived from neutrophils, such as neutrophil elastase; and inhibitors of matrix metalloproteinase (MMP) such as MMP-2, MMP-9 and MMP-12; (C) antioxidants including, but not limited to N-acetylcysteine, and inhibitors or reactants of reactive oxygen species; and toxic peptides such as defensins that can directly cause cell damage; (D) mucus production inhibitors that include, but are not limited to, inhibitors of mucosal genes, and also mucus elimination agents such as expectorants, mucolytics and mucokinetics; and (E) antibiotic therapy as with a cetóhdo, for example Ketek®.
The drug combinations of the present invention can be administered to a cell or cells, or to a human patient, in separately pharmaceutically acceptable formulations administered simultaneously or sequentially, formulations containing more than one therapeutic agent, or by a mixture of formulations of a single agent and multiple agents. Regardless of the form of administration, these drug combinations form a pharmaceutically effective amount of components.
The treatment regimen / dosage schedule can be modified, reasonably, throughout the course of the therapy so that the minor amounts of each of the pharmaceutically effective amounts of the compounds used in the combination, which together show an efficacy pharmaceutical satisfying, are administered, and so that the administration of this pharmaceutically effective amount of the compounds in the combination continues only the time necessary to treat the patient successfully A pharmaceutical composition according to the invention is preferentially produced and administered in dosage units, each unit containing, as an active constituent, a specific dose of the compound. Pharmaceutically acceptable salts of the compound of formula (A) are within the scope of this invention. The term "salt (s)" means addition salts of acids or bases formed with acids and bases. In addition, the term "salt (s)" includes salts of bipolar ions (internal salts), that is, the compound of formula (A) contains a basic moiety, such as an amine or a ring of pmdma or ylidazole, and an acid moiety , as carboxylic acid. Preferred are pharmaceutically acceptable (ie, physiologically acceptable, non-toxic) salts, such as, for example, acceptable metal and amine salts wherein the cation does not contribute significantly to the toxicity or biological activity of the salt. However other salts may be useful, eg, in the isolation or puncture stages that may be employed during the preparation and, therefore, it is contemplated that they are within the scope of the invention. The salts of the compounds of formula (A) can be formed, for example, by reacting a compound of formula (A) with an amount of acid or base, as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by hydration.
The acid addition salts are formed with the compound of the invention which carries a basic residue (s), such as nitrogen of ammonium, ammonium or a mono- or disubstituted group. The particular acid addition salts are the pharmaceutically acceptable acid addition salts, that is, the salts whose anion is not toxic to the patient at the pharmaceutical dose of the salt, and so that the beneficial effects inherent to the free form of the compound are not vitiated by the secondary effects of the anion. The salts are chosen optimally to be compatible with customary pharmaceutical vehicles and are adapted to an applicable administration form. The acid addition salts of the compound of this invention can be prepared by reacting the free form of the molecule carrying the rest basic with the appropriate acid, by applying or adapting known methods. For example, the acid addition salts of the compound of this invention can be prepared by dissolving the free form of the molecule bearing the basic moiety in water or an aqueous alcoholic solution or other suitable solvents containing the appropriate acid, and isolating the salt by evaporation of the solution, or by reacting the free form of the molecule carrying the basic residue and the acid in an organic solvent, in which case the salt is separated directly, or can be obtained by concentrating the solution. Some acids suitable for use in the preparation of these salts are hydrochloric acid, hydrobromic acid, phosphonic acid, sulfunic acid, various organic carboxylic and sulfonic acids, such as acetic acid, citric acid, propionic acid, succinic acid, benzoic acid, acid tartaric acid, fumaric acid, mandechic acid, ascorbic acid, malic acid, methanesulfonic acid, toluenesulfonic acid, mancheic acid, ascorbic acid, malic acid, fatty acids, adipate algmate, ascorbate, aspartate, benzenesulfonate, benzoate, cyclopentanpropionate, digluconate, dodecyl sulfate, bisulfate, butyrate, lactate, laurate, lauplsulfate, maleate, hydroiodide, 2-hydroxy ethanesulfonate, glycerophosphate, picrate, pivalate, palmoate, pectmate, persulfate, 3-phenylpropionate, thiocyanate, 2-naphthalenesulfonate, undecanoate, nicotmate, hemisulfate, heptanoate, hexanoate, camphorrate, camphor sulfonate and others.
The acid addition salts of the compound of this invention can also be used to regenerate the parent compound of the invention from the salts by the application or adaptation of known methods. For example, the ongen compound of the invention can be regenerated from its acid addition salts by treatment with an alkali, eg, an aqueous solution of sodium bicarbonate or an aqueous solution of ammonia.
The base addition salts are formed with the compounds of the invention which carry the carboxy moiety. The addition salts of specific bases are the pharmaceutically acceptable base addition salts, ie the salts whose cation is not toxic to the patient at the pharmaceutical dose of the salt, so that the beneficial effects inherent to the free form of the The compounds are not vitiated by the secondary effects of the anion. The salts are chosen optimally to be compatible with customary pharmaceutical carriers and are adapted to an applicable form of administration. The base addition salts of the compound of this invention can be prepared by reacting the free form of the molecule carrying the acid moiety with the appropriate base, by the application or adaptation of known methods. For example, the base addition salts of the compound of this invention can be prepared by dissolving the free form of the molecule carrying the acid moiety in water or an alcoholic aqueous solution or other suitable solvents containing the appropriate base, and isolating the salt by evaporating the solution, or reacting the free form of the molecule carrying the acid moiety and the base in an organic solvent, in which case the salt is separated directly or can be obtained by concentrating the solution. Some suitable bases to be used in the preparation of these salts are those derived from alkaline earth metal salts or amines, such as sodium hydride, sodium hydroxide, potassium hydroxide, calcium hydroxide, aluminum hydroxide, lithium hydroxide, hydroxide. magnesium, eme hydroxide, ammonia, ethylendiamma, N-methylglucamma, hsma, argimna, ormtma, colma, N, N'-d? benc? let? lend? am? na, chloroprocaine, diethanolamm, procaine, N-benzylphenethylamine, diethylamine, piperazine, tns (hydroxylmethyl) ammonethane, tetramethylammonium hydroxide and the like.
The base addition salts of the compound of this invention can also be used to regenerate the parent compound of the invention from the salts by the application or adaptation of known methods. For example, the ongen compound of the invention can be regenerated from its base addition salts by treatment with an acid, eg, hydrochloride acid.
In practice, the compound of the present invention is administered in a suitable formulation to patients, so that its activity is specifically localized. It will be appreciated that the prefended route will depend on the site of the disorder to which the administration is directed The pharmaceutically acceptable dosage forms refer to the dosage forms of the compound of the invention and include, for example, powders, suspensions, powders, inhalants, tablets, emulsions and solutions particularly suitable for inhalation. The techniques and formulations can be found, in general, in Remmgton's Pharmaceutical Sciences, Mack Pubhshmg Co, Easton, PA, latest edition. If desired, and for a more efficient distribution, the compound can be microencapsulated within, or linked to slow-release or targeted delivery systems, such as biodegradable and biocompatible polymer matrices (eg, pol? (Co-ghcohdo of d, l-lactate), hposomes and microspheres, and injected subcutaneously or intramuscularly by a technique called subcutaneous or intramuscular depot, to provide a continuous slow release of the compound (s) over a period of 2 weeks or longer .
The compound can also be sterilized, for example, by filtration through a filter that retains bactenas, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile medium immediately before use.
Suitable formulations for nasal or tracheal administration mean formulations that are in a form suitable to be administered nasally or by inhalation to the patient. The formulation may contain a carrier, in powder form, having a particle size, for example, in the range of 1 to 500 microns (including particle sizes in a range between 20 and 500 microns in 5 micron increments, like 30 microns, 35 microns, etc.). Suitable formulations in which the carrier is a liquid, for administration, for example, as a nasal spray or nasal drops, include aqueous or oily solutions of the active ingredient. Formulations suitable for aerosol administration can be prepared according to conventional methods, and can be administered with other therapeutic agents. MDI and DPI are feasible means for performing inhalation therapy by administering a dosage form of the compound of the present invention.
The actual dosage levels of the active ingredient (s) in the compositions of the invention can be varied to obtain an amount of the active ingredient (s) that is (are) effective (effective) to obtain a desired therapeutic response for a particular composition and method of administration to the patient. A selected dosage level for any particular patient, therefore, depends on a variety of factors, including the desired therapeutic effect, the route of administration, the desired duration of treatment, the etiology and severity of the disease, the patient's condition, weight, sex, diet and age, the type and potency of each active ingredient, the rates of absorption, metabolism and / or excretion, and other factors A total target dose of the compounds of this invention administered to a patient in unit or divided doses is about 1000 mg, more particularly about 50 mg to 300 mg, and even more particularly about 10 mg to 100 mg. However, higher or lower target doses may also be appropriate. The target dose may be administered by a single administration in the form of a unit dosage unit, or by several smaller dosage units, or by multiple administration of subdivided doses at predetermined intervals. The percentage of active ingredient in a composition can go, although it should constitute a proportion so that an adequate dosage is obtained. Obviously, unitary dosage forms can be administered at about the same time. A dosage can be administered as often as necessary to obtain the desired therapeutic effect. Some patients may respond quickly to a higher or lower dose, and may find a much weaker maintenance dose adequate. For other patients it may be necessary to perform long-term treatments with a ratio of 1 to 4 target doses, according to the physiological requirements of each concrete patient. It is understood that, for other patients, it will be necessary to prescribe no more than one or two doses per day.
The formulations can be prepared in a unit dosage form by any method known in the pharmacy art. These methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers. , or both, and then, if necessary, mold the product.
Experimental part To analyze mass spectroscopic methods (FAB-MS, ESI-MS) were used. Temperatures are indicated in degrees Celsius; TA indicates ambient temperature (from 22 ° C to 26 ° C). The abbreviations used are explained or correspond to conventions customary for those skilled in the art.
The invention is exemplified by the following examples.
EXAMPLES PREPARATION 1 Synthesis of (3- (N-phen? N-2-p? Nd? L) ammo) -2- (d? -terc-butox? Carbon? Lammo) methyl propionate (scheme 1, compound 3 ) Scheme 1 2- (di-fe / r-butoxycarbonylamino) methyl acrylate (scheme 1, compound 2) 50 g (0.228 mol) of (eerc-butoxycarbonyl) sepna (V) were dissolved in 300 ml of acetone. 107 g (0.493 mol) of di-butyl-butyl dicarbonate and 2.64 g (22 mmol) of 4- (d? Met? Lam? No) p? Pd? Na (DMAP) were added. The mixture was stirred at room temperature overnight, after which the solvent was removed under reduced pressure, and the residue was suspended in 500 ml of ethyl acetate. The organic phase was washed with 500 ml of 1 N HCl, dried using magnesium sulfate and the organic solvents were removed under reduced pressure. 23 g of acplate 2 were obtained by crystallization of the residue in 200 ml of heptane at -30 ° C and then filtering with suction. The mother liquor was concentrated and the residue was dissolved in 140 ml of acetonitrile. 31 g (0.142 mol) of di-ert-butyl dicarbonate and 1.26 g (10 mmol) of DMAP were added. After heating the mixture at 50 ° C for 8 h, the solvent was removed in vacuo and the residue was suspended in 500 ml of ethyl acetate. The organic phase was washed with 400 ml of 1 N HCl and dried over magnesium sulfate. After the solvent had been removed in vacuo, an additional 31.5 g of acplate 2 were obtained by cystenation in heptane. Performance. 54.5 g (0.181 mol), 79%.
Empirical formula C14H23N06; P.M. = 301.34; MS ((2M *) + Na +) 625.7. NMR of? (DMSO-), 1.40 (s, 18 H), 3.74 (s, 3 H), 5.85 (s, 1 H), 6.28 (s, 1 H).
(Methyl 3 - (γ-phenyl-β-2-pyridyl) a-nino) -2- (di-tert-butoxycarbonylamino) propionate (scheme 1, compound 3) 4.96 g (16 g) were mixed ( , 5 mmol) of acnlate 2 with 5.6 g (33 mmol) of 2-an? Hnop? Nd? Na and 32.16 g (98.7 mmol) of cesium carbonate. 50 ml of acetonite was added and the mixture was stirred at 45 ° C for 2 days. The solid was removed by filtration with suction through diatomite and washed 3 times with 100 ml portions of acetone. The combined organic phases were evaporated and the residue was chromatographed on silica gel, using heptane / diethyl ether 1: 1. 5.66 g (73%) of the ester 3 were obtained. Empirical formula C ^^ NBOS; P M. = 471.56; MS (M + H) 472.2 Separation of the enantiomers (scheme 1, compound 3 (S) and compound 3TR.}.) The racemic amino ester 3 was prepared from the corresponding acrylic ester 2 and then separated into the 3 (S) and 3 (R) enantiomers by Preparative HPLC using chiral stationary phases, such as Chiralpak AD (Daicel) 100 x 380, at room temperature, flow rate 300 ml / mm. The purity of the enantiomers was determined by analytical HPLC, such as Chiralpak-AD-H (Daicel) 4.6 x 250, 30 ° C, flow rate 1 ml / min, room temperature.
PREPARATION 2 Synthesis of 2- (2-meth? Lam? Nop? Nm? D? N-4-? L) -lH-mdol-5-carboxylic acid (8) (Scheme 2, compound 8) Scheme 2 l-dimethylamino-4,4-dimethoxy-pent-l-en-3-one (scheme 2, compound 6) 100 g (0.76 mol) of 3,3-d? methox? -2 -butanone (4) were stirred. together with 90.2 g (0.76 mol) of N, N-dimethylformamide dimethyl acetal (5) at 120 ° C for 48 h. The methanol formed in the reaction was continuously removed from the reaction solution by distillation. The reaction occurred spontaneously when the solution was cooled, and the crystallization was completed by the addition of a small amount of heptane. This produced 128.24 g of the crude product 6 (yield 90%), which was reacted without further purification Empirical formula C9H | 7N03; P.M. = 187.24; MS (M + H) 188.2. NMR of? (DMSO-6 6), 1.22 (s, 3 H), 2.80 (s, 3 H), 3.10 (s, 9 H), 5.39 (d, J = 15 Hz, 1 H ), 7.59 (d, J = 15 Hz, 1 H). [4- (1, 1-dimethoxyethyl) pyrimidin-2-yl] methylamine (scheme 2, compound 7) 1.22 g (53 mmol) of sodium was dissolved in 100 ml of absolute ethanol. 5.8 g (53 mmol) hydrochloride and 10 metilguanidma g (53 mmol) of ld? Met? Lammo-4,4-d? MetOx? Pent-l-en-3-one (6) they were added to this solution while stirring, and everything was heated to reflux for 4 h. To finish the reaction, the ethanol was evaporated. The resulting product 7 was used for the subsequent reaction without further purification. Yield: 11.5 g (58 mmol, quantitative); Empirical formula C9H15N3O2; P.M. = 197.24, MS (M + H) 198.2 NMR of? (DMSO-¿6), 1.45 (s, 3 H), 2.78 (s, 3 H), 3.10 (s, 6 H), 6.75 (d, J = 3 Hz, 1 H ), 7.0-7.1 (s (a), 1 H), 8.30 (d, J = 3 Hz, 1 H). 2- (2-Methylaminopyrimidm-4-yl) -l / -indole-5-carboxylic acid (scheme 2, compound 8). 5 g (25 mmol) of [4- (1, 1-methox? Et? ) p? nm? d? n-2-? l] met? lam? na (7) and 3.85 g of 4-h? drazmobenzoic acid, at room temperature and while stirring, to 150 ml of acid 50% sulfun, and the mixture was heated at 130 ° C for 4 h. The methanol that formed in the reaction was continuously removed from the reaction solution by distillation. After cooling to 10 ° C, the reaction mixture was poured into 200 ml of ice and adjusted to a pH of about 5.5 with a concentrated sodium hydroxide solution. The precipitate formed from sodium sulfate and product mixture was removed by filtration, and the residue from the filter was extracted several times with methanol. The combined methanol extracts were concentrated, and product 8 was purified by flash chromatography (CH2Cl2 / methanol 9: 1). Yield: 0.76 g (11%); Empirical formula C14H12N402; P.M. = 268.28; MS (M + H) 269.1. NMR of? (DMSO-Í 6), 2.95 (s, 3 H), 6.90-7.10 (s (a), 1 H), 7.18 (d, J = 3 Hz, 1 H), 7.4 (s, 1 H), 7.58 (d, J = 4.5 Hz, 1 H), 7.80 (d, J = 4.5 Hz, 1 H), 8.30 (s, 1 H), 8.38 (d, J = 3 Hz, 1 H), 11.85 (s, 1 H), 12.40-12.60 (s) (a), 1 H).
EXAMPLE 1 Synthesis of acid 2-. { [2- (2-met? Lammop? R? M? D? N-4-? L) -lH-mdol-5-carboml] am? No} -3- (fen? Lp? R? Dm-2-? Lammo) prop? On? Co (A) Scheme 3 Acid methyl ester 2-. { [2- (2-methylaminopyrimidin-4-yl) -lH-indol-5-carbonyl] amino} -3- (phenylpyridin-2-ylamino) -propionic acid (Scheme 3, compound 10) 2.9 g of the S enantiomer of 3 (3 (SY) in 30 ml of dioxane were dissolved, and the solution was cooled to 0 ° C. 30 ml of 4N HCl in dioxane were added, after which the mixture allowed to reach room temperature and then stirred for 12 h. the solvent was removed in vacuo. the residue was suspended in 30 ml of DMF (solution A) 2.47 g (9.2 mmol) of acid 8 were dissolved in 50 ml of DMF and cooled to 0 ° C. 4.21 g of HATU and 6.4 ml of DIEA were added. the mixture was stirred at 0 ° C for 45 minutes, allowed to reach room temperature and solution A was added. The mixture was stirred at room temperature for 12 h.The solvent was removed in vacuo and the residue was distributed among 300 ml of a saturated solution of NaHCO 3 and 300 ml of ethyl acetate The aqueous phase was extracted three times with 100 ml portions of ethyl acetate and the phases The combined organic extracts were washed with 400 ml of a saturated NaCl solution. The organic phase was dried with magnesium sulfate. The solvents were removed under reduced pressure and the residue was chromatographed on silica gel using heptane / ethyl acetate 1: 3. 1.78 g (55%) of ester K was obtained). Empirical formula C29H27N703; P.M. = 521.58; MS (M + H) 522.2.
Acid 2-. { [2- (2-methylaminopyrimidin-4-yl) -l ^ 1 -indole-5-carbonyl] amino} -3- (phenylpyridin-2-ylamino) propionic (scheme 3, compound A) 2.0 g (3.8 mmol) of the methyl ester K) was dissolved in 200 ml of methanol. 1 ml of 2N aqueous NaOH was added and the mixture was stirred at room temperature for 12 h. After evaporating the solvents, the residue was dissolved in water and the pH was adjusted to about 5 using a saturated solution of? AH2P04. The resulting precipitate was removed by filtration and washed with water. After drying under reduced pressure of about 1 mbar at 40 ° C, 1.95 g (quantitative yield) of the acid was isolated A. Empirical formula C28H25? 7? 3; P.M. = 507.56; MS (M + H) 508.3. RM? from ? (DMSO-? 6), 2.95 (s, 3 H), 4.22-4.50 (m, 2 H), 4.65-4.72 (m, 1 H), 6.29 -6 , 36 (d, 1 H), 6.70 - 6.79 (m, 1 H), 6.90 - 7.10 (sa, 1 H), 7.13-7.19 (m, 1 H) , 7.22-7.38 (m, 5 H), 7.40-7.48 (m, 3 H), 7.50-7.55 (m, 1 H), 7.57-7.60 (m, 1 H), 7.96 (sa, 1 H), 8.34 - 8.40 (m, 2 H), 8.80 - 8.90 (d, 1 H), 11.80 (s) , 1 HOUR).
IN VITRO TEST PROCEDURE ELISA of the IKK enzyme The assay buffer had the following composition (50 mM HEPES, 10 mM MgC12, 10 mM β-glycerophosphate, 2 μM microcystin-LR, 0.01% NP-40, DTT 5 mM). The IKK enzyme preparation was diluted 1:50 (preparation prepared in situ) plus test compound in DMSO (final concentration in the well: 2%). The assay procedure is as follows: incubation of enzyme and compound for 30 min; addition of 1 mM ATP or 50 μM ATP; peptide pSer36-IkB (substrate): 40 μM; incubation for 45 min and addition of anti-peptide antibody pSer32-pSer36-IkB; incubation for 45 min and transfer to a plate coated with protein G; incubation for 90 min, followed by 3x washes; addition of streptavidin-HRP, then incubation for 45 min, followed by 6x washes; addition of TMB and incubation for 15 min; and stopping and reading solution using a photometer.
The results of the in vitro analysis appear in Table I below. Table I In the ELISA of the IKK enzyme described above, at 1 mM ATP, the compound of formula (A) shows an IC50 of the kinase I? B 705, 4.725 and 47.5 times higher than the compounds (B), (C) and (D), respectively. These data demonstrate unexpected and significantly higher activity of compound (A) relative to compounds (B), (C) and (D).
COMPARISON THROUGH IN VIVO TEST PROCEDURES BETWEEN COMPOUNDS A AND B Gene expression induced by NF-? B contributes significantly to the pathogenesis of inflammatory diseases, such as asthma and arthritis. I? B kinase (IKK) is the convergent point for the activation of NF-? B by a broad spectrum of inflammatory agonists.
IKK is a complex of multiple subunits that contains two catalytic subunits, D K-1 (also called D K-a) and D K-2 (also called KK-β), and the regulatory subunit IKK-β. Gene knockout studies have clearly shown that the IKK-2 or KK-β subunits of the IKK complex are necessary for the activation of NF-? B by all known proinflammatory stimuli, including lipopolysaccharide (LPS), and IL -1/3. Accordingly, cells deficient in IKK-β are deficient in the activation of IKK and NF-βB in response to tumor necrosis factor alpha (TNFo :) or interleukin-lβ (IL-lß).
In situ bioimaging data have also shown that NF-αB activity induced by IL-1β in the lung is inhibited by the administration of a dominant negative form of IKKβ (Adv-KK-2 DN). Therefore, a selective inhibitor of IKK-β would not only be of great interest as a potential anti-inflammatory agent, but would also be a valuable tool for understanding the mechanisms that regulate the activation of NF-βB by these inflammatory agonists.
A. Mouse model of luciferase-NF-? B indicator Methods used for imaging studies in mice with compound (A) and compound (B).
General description The compounds were used as nanotuned suspensions in 0.2% Tween 80 in PBS, and dosed via the intranasal route.
Administration of the drug by intranasal route and inflammatory stimuli: The mice were anesthetized with 4% isoflurane gas in oxygen. A volume of 25 μl was applied to each nasal tissue and the mice were allowed to aspirate the suspension.
Balb / c female mice of 6-8 weeks were used for the studies, in which an indicator of AdV-NF? B-luc? Ferase was instilled into the lung. To obtain the images of the mice, they were anesthetized with 4% isoflurane in oxygen. Lucifenin was administered i.p. at a dose of 150 mg / kg. Ten minutes after the injection of luciferma, the images of the animals were formed in an IVIS200 (Xenogen) system with a one-minute bioluminescent exposure. Alternatively, at 10-15 minutes after the administration of luciferma, mice were rapidly euthanized and internal tissues dissected and ex vivo images were formed.
B. Determination of dose-response 1-2 x 108 pfu of adenovirus-NF? B-luc? Ferase was administered intranasally 3-5 days before stimulation (.n.) with an inflammatory stimulus (IL). -lß or LPS). The animals were dosed i.n. with 0.3-10 mg / kg of compound 30 m? n-1 h before exposure to 0.5 μg of LPS or 50 ng of IL-lß Then an image of the animal was formed one or several times from 1 h to 24 h after administering the inflammatory stimulus.
The results of the live procedures are as follows. The effects of the compound (A) and the compound (B) on the activation of NF-? B induced by IL-lβ are shown in figures 1 and 2. Although both compounds inhibited the activity NF-? B in a manner dependent on At dose, compound (A) showed superior efficacy with an estimated ED50 of approximately 1 mg / kg.
C. Pharmacokinetic procedures Male Hartley guinea pigs (450-550 g), previously sensitized with ovalbumin, were used for the determination of lung and plasma compound levels. Nano-stressed suspensions of compound (A) and compound (B) were dosed by intratracheal instillation at 0.01, 0.03, 0.1 and 0.3mg / kg. One hour after dosing, animals were euthanized (Euthasol), and blood samples of 1 ml were obtained by cardiac puncture and collected in hepanna coated jenngas. Plasma was separated from the cellular component of blood by centrifugation, and stored at -80 ° C until assayed. The lungs were removed by dissection, blotted, weighed and stored in 20-25 ml vidne vials individually at -80 ° C until assayed for compound levels.
The key differences between the pharmacokinetic profiles of compound (A) and compound (B) are illustrated in figure 3 using the findings of the highest dose group (0.3 mg / kg) Figure 3 demonstrates that pulmonary exposure to compound (A) or compound (B) after instillation i.t. of the compound (B) is low relative to the compound (A) after the t.t. administration, which suggests that the compound (B) is rapidly absorbed from the lung. Figures 3 to 5 also demonstrate that compound (B) would be a weaker candidate for inhalation, because 1) it is distributed very systemically after exposure, when it is administered by tracheal route; 2) the compound (B) is a prodrug of the compound (A) having a different exposure profile; and 3) the compound (B) produces a reduced exposure to the compound (A) in the lung than that obtained by directly dosing the compound (A).
Compound (A) is a stronger candidate for inhalation than compound (B), because 1) compound (A) has a low systemic exposure after i.t. and oral; and 2) compound (A) should have a longer pulmonary residence time.
In addition, there are supports to affirm that compound (B) is very systematically available when administered orally.
The ratio of lung to plasma for compound (A) varies between 143 and 284 (depending on the dose), while the ratio between lung to plasma for compound (B) varies between 13 and 44 (depending on the dose). These ratios were obtained by dividing the lung levels of the compound between the corresponding plasma levels at the same dose.

Claims (14)

    REGVGNDIC ATIONS:
  1. A substantially pure compound of formula (A) (A) or its pharmaceutically acceptable salt or solvate.
  2. 2. A pharmaceutical composition comprising a pharmaceutically effective amount of a compound of formula (A) and a pharmaceutically acceptable carrier.
  3. 3. A method for treating a patient suffering or suffering from a pathological disorder that can be improved by the inhibition of IKK-2, which comprises administering to said patient a pharmaceutically effective amount of the compound according to claim 1.
  4. 4. The method according to claim 3, wherein the administration is performed to produce a localized activity.
  5. 5. The method according to claim 3, wherein the pathological disorder is asthma, nnitis, chronic obstructive pulmonary disorder, or exacerbations of chronic obstructive pulmonary disorder.
  6. 6. The method according to claim 3, wherein the administration is via tracheal, intranasal, by inhalation, or by administration of an aerosol.
  7. A method for treating a patient suffering from asthma, comprising administering to the patient a pharmaceutically effective amount of a compound of claim 1.
  8. 8. A method for treating a patient suffering from nnitis, comprising administering to the patient a pharmaceutically effective amount of a compound of claim 1.
  9. 9. A method for treating a patient suffering from chronic obstructive pulmonary disorder, comprising administering to the patient a pharmaceutically effective amount of a compound of claim 1.
  10. 10. A method for treating a patient suffering from chronic obstructive pulmonary exacerbation disorder, comprising administering to the patient a pharmaceutically effective amount of a compound of claim 1.
  11. 11. The pharmaceutical composition according to claim 2, further comprising a pharmaceutically effective amount of a compound selected from the group consisting of a bronchodilator, a long-acting beta-2-agonist, an anticolmer agent, a methylxanthine, and an antimflamatone therapy. , mixed with a pharmaceutically acceptable vehicle
  12. The pharmaceutical composition according to claim 11, wherein the bronchodilator is a short-acting beta-2-agomsta; the long-acting beta-2-agomsta is selected from salmeterol and formoterol; the anticholmergic agent is selected from ipratropium bromide and tiotropium bromide; Methylxanthine is theophylline; and the anti-inflammatory therapy is selected from inhibitors of cellular recruitment and toxic inflammatory mediators, inhibitors of proteolytic enzymes, antioxidants, inhibitors of mucus production, and antibiotic therapy.
  13. 13. A method for treatment according to claim 3, further comprising administering a pharmaceutically effective amount of a compound selected from the group consisting of a bronchodilator, a long-acting beta-2-agonist, an anticholinergic agent, a methylxanthine, and a antimflamatone therapy, mixed with a pharmaceutically acceptable vehicle.
  14. 14. A method for treatment according to claim 13, wherein the bronchodilator is a short-acting beta-2-agonist; long acting beta-2-agonists are selected from salmeterol and formoterol; the anticholmergic agent is selected from ipratropium bromide and tiotropium bromide; Methylxanthine is teofihna; and the antiinflamatone therapy is selected from inhibitors of cellular recruitment and toxic inflammatory mediators, inhibitors of proteolytic enzymes, antioxidants, inhibitors of mucus production, and antibiotic therapy.
MXPA06012870A 2004-05-12 2005-05-11 Substantially pure 2-{[2-(2- methylamino -pyrimidin-4- yl)-1h- indole-5- carbonyl] -amino}-3 -(phenylpyridin -2-yl-amino)- propionic acid as an ikb kinase inhibitor. MXPA06012870A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57014604P 2004-05-12 2004-05-12
PCT/US2005/016381 WO2005113544A1 (en) 2004-05-12 2005-05-11 Substantially pure 2-{[2-(2-methylamino-pyrimidin-4-yl)-1h-indole-5-carbonyl]-amino}-3-(phenylpyridin-2-yl-amino)-propionic acid as an ikb kinase inhibitor

Publications (1)

Publication Number Publication Date
MXPA06012870A true MXPA06012870A (en) 2007-02-15

Family

ID=34969825

Family Applications (1)

Application Number Title Priority Date Filing Date
MXPA06012870A MXPA06012870A (en) 2004-05-12 2005-05-11 Substantially pure 2-{[2-(2- methylamino -pyrimidin-4- yl)-1h- indole-5- carbonyl] -amino}-3 -(phenylpyridin -2-yl-amino)- propionic acid as an ikb kinase inhibitor.

Country Status (23)

Country Link
EP (1) EP1747215A1 (en)
JP (1) JP2007537266A (en)
KR (1) KR20070011483A (en)
CN (1) CN1950359A (en)
AR (1) AR049274A1 (en)
AU (1) AU2005245834A1 (en)
BR (1) BRPI0511029A (en)
CA (1) CA2566213A1 (en)
EC (1) ECSP066992A (en)
GT (1) GT200500111A (en)
IL (1) IL178992A0 (en)
MA (1) MA28553B1 (en)
MX (1) MXPA06012870A (en)
NO (1) NO20065719L (en)
PA (1) PA8633101A1 (en)
PE (1) PE20060269A1 (en)
RU (1) RU2006143758A (en)
SV (1) SV2006002111A (en)
TN (1) TNSN06338A1 (en)
TW (1) TW200605881A (en)
UY (1) UY28897A1 (en)
WO (1) WO2005113544A1 (en)
ZA (1) ZA200608712B (en)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19951360A1 (en) 1999-10-26 2001-05-03 Aventis Pharma Gmbh Substituted indoles
DE102004033406A1 (en) 2004-07-10 2006-02-16 Sanofi-Aventis Deutschland Gmbh Process for the preparation of the enantiomeric forms of 2,3-diaminopropionic acid derivatives
EP2025674A1 (en) 2007-08-15 2009-02-18 sanofi-aventis Substituted tetra hydro naphthalines, method for their manufacture and their use as drugs
US11969501B2 (en) 2008-04-21 2024-04-30 Dompé Farmaceutici S.P.A. Auris formulations for treating otic diseases and conditions
KR20130097813A (en) 2008-04-21 2013-09-03 오토노미, 인코포레이티드 Auris formulations for treating otic diseases and conditions
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
CA2797428A1 (en) * 2010-04-27 2011-11-03 Hutchison Medipharma Limited Pyrimidinyl indole compounds
EP2582709B1 (en) 2010-06-18 2018-01-24 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
TW201221505A (en) 2010-07-05 2012-06-01 Sanofi Sa Aryloxyalkylene-substituted hydroxyphenylhexynoic acids, process for preparation thereof and use thereof as a medicament
TW201215388A (en) 2010-07-05 2012-04-16 Sanofi Sa (2-aryloxyacetylamino)phenylpropionic acid derivatives, processes for preparation thereof and use thereof as medicaments
TW201215387A (en) 2010-07-05 2012-04-16 Sanofi Aventis Spirocyclically substituted 1,3-propane dioxide derivatives, processes for preparation thereof and use thereof as a medicament
US8828995B2 (en) 2011-03-08 2014-09-09 Sanofi Branched oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
US8871758B2 (en) 2011-03-08 2014-10-28 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120052A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives substituted with carbocycles or heterocycles, method for producing same, drugs containing said compounds, and use thereof
US8828994B2 (en) 2011-03-08 2014-09-09 Sanofi Di- and tri-substituted oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120055A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
JP7033789B2 (en) 2016-06-29 2022-03-11 オトノミー,インク. Triglyceride ear preparation and its use
CN106588803A (en) * 2016-11-16 2017-04-26 西南科技大学 Novel method for preparing 5-acetylisoxazole

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10237722A1 (en) * 2002-08-17 2004-08-19 Aventis Pharma Deutschland Gmbh Indole or benzimidazole derivatives for the modulation of IKappaB kinase

Also Published As

Publication number Publication date
PA8633101A1 (en) 2006-01-23
TNSN06338A1 (en) 2008-02-22
NO20065719L (en) 2006-12-12
TW200605881A (en) 2006-02-16
SV2006002111A (en) 2006-01-30
MA28553B1 (en) 2007-04-03
UY28897A1 (en) 2005-12-30
CA2566213A1 (en) 2005-12-11
ZA200608712B (en) 2008-06-25
IL178992A0 (en) 2007-03-08
KR20070011483A (en) 2007-01-24
WO2005113544A1 (en) 2005-12-01
ECSP066992A (en) 2006-12-29
EP1747215A1 (en) 2007-01-31
JP2007537266A (en) 2007-12-20
BRPI0511029A (en) 2007-11-27
CN1950359A (en) 2007-04-18
GT200500111A (en) 2006-05-09
AU2005245834A1 (en) 2005-12-01
PE20060269A1 (en) 2006-05-11
RU2006143758A (en) 2008-06-27
AR049274A1 (en) 2006-07-12

Similar Documents

Publication Publication Date Title
MXPA06012870A (en) Substantially pure 2-{[2-(2- methylamino -pyrimidin-4- yl)-1h- indole-5- carbonyl] -amino}-3 -(phenylpyridin -2-yl-amino)- propionic acid as an ikb kinase inhibitor.
US8293748B2 (en) p38 MAP kinase inhibitors
ES2428917T3 (en) P38 MAP kinase inhibitors
JP5855113B2 (en) Novel substituted quinoline compounds as S-nitrosoglutathione reductase inhibitors
ES2359467T3 (en) USEFUL COMPOUNDS AS PROTEIN QUINASAS INHIBITORS.
ES2302033T3 (en) DERIVATIVES OF QUINOZALINE AS INHIBITORS OF ELASTASA DE LOS NEUROFILOS AND ITS USE.
ES2525756T3 (en) Imidazo [1,2-a] pyridines as JNK modulators
US20220185792A1 (en) Prmt5 inhibitor compounds
RU2661895C2 (en) Guanidinobenzoic acid ester compound
JP4393197B2 (en) Use of oxindole derivatives in the treatment of dementia related diseases, Alzheimer's disease and glycogen synthase kinase-3 related symptoms
WO2021193756A1 (en) Novel benzimidazole derivative
KR20140105459A (en) Kinase inhibitors
JP2018048143A (en) Kcnq2-5 channel activator
WO2012002502A1 (en) Dihydropyrimidinone derivative and pharmaceutical use thereof
WO1999024407A1 (en) Compounds which inhibit tryptase activity
JP6441830B2 (en) Substituted 2-aza-bicyclo [2.2.1] heptane-3-carboxylic acid (cyano-methyl) -amide inhibitors of cathepsin C
US20070142417A1 (en) Substantially Pure 2-{[2-(2-Methylamino-Pyrimidin-4-YL)-1H-Indole-5-Carbonyl]-Amino}-3-Phenylpyridin-2-YL-Amino)-Propionic Acid as an IkB Kinase Inhibitor
CN115003381A (en) Compounds and their use for the treatment of alpha 1-antitrypsin deficiency
US20200283396A1 (en) Caspase inhibitors and methods of use thereof
JPWO2003011812A1 (en) Novel amine derivative having human-β-tryptase inhibitory activity and medicament containing the same
US11447497B2 (en) (S)-3-(2-(4-(benzyl)-3-oxopiperazin-1-yl)acetamido)-4-oxo-5-(2,3,5,6-tetrafluorophenoxy)pentanoic acid derivatives and related compounds as caspase inhibitors for treating cardiovascular diseases
JP2012512822A (en) Pyrimidin-2-ylamino derivatives and their use for treating inflammation
US8445529B2 (en) Indole derivative having, carbamoyl group, ureido group and substituted oxy group
CN115286583B (en) Diphenylamino pyrimidine-containing compound, preparation and application thereof as HDACs enzyme inhibitor
WO2006076318A1 (en) Monopotassium salt of an ikb kinase inhibitor

Legal Events

Date Code Title Description
FA Abandonment or withdrawal