WO2009076206A1 - Synthesis methods of histone deacetylase inhibitors (hdacis) - Google Patents

Synthesis methods of histone deacetylase inhibitors (hdacis) Download PDF

Info

Publication number
WO2009076206A1
WO2009076206A1 PCT/US2008/085684 US2008085684W WO2009076206A1 WO 2009076206 A1 WO2009076206 A1 WO 2009076206A1 US 2008085684 W US2008085684 W US 2008085684W WO 2009076206 A1 WO2009076206 A1 WO 2009076206A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
aminomethyl
histone deacetylase
yield
Prior art date
Application number
PCT/US2008/085684
Other languages
French (fr)
Inventor
Vincent C. O. Njar
Lalji K. Gediya
Original Assignee
University Of Maryland, Baltimore
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 University Of Maryland, Baltimore filed Critical University Of Maryland, Baltimore
Publication of WO2009076206A1 publication Critical patent/WO2009076206A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/28Radicals substituted by singly-bound oxygen or sulphur atoms
    • C07D213/30Oxygen atoms

Definitions

  • PCA Prostate cancer
  • Histone Deacetylases are the catalytic subunits of multiprotein complexes responsible for deacetylation of histones and nonhistone proteins. Lysine acetylation, i.e., the transfer of an acetyl moiety from acetyl-coenzyme A to the ⁇ -amino group of a specific lysine residue, has emerged as the major form of posttranslational modification of histones, and other proteins have been correlated with transcription, chromatin assembly, DNA repair, and recombinatorial events (Marks et al., Histone deacetylases and cancer, causes and therapies.
  • Histone acetylation in vivo is a dynamic, reversible process governed by the opposite actions of histone acetyltransferases (HATs) and HDACs.
  • HATs histone acetyltransferases
  • Aberrant acetylation of histone tails, emerging from either HAT mutation or abnormal recruitment of HDACs, has been linked to carcinogenesis (Pandolfi, P. P. Transcription therapy for cancer. Oncogene, 20: 3116-3127, 2001).
  • HAT histone acetyltransferases
  • HDACs histone acetyltransferases
  • HDACl is up-regulated in prostate cancer compared to benign prostatic hyperplasia (BPH) (Patra et al., Histone deacetylase and DNA methyltransferase in human prostate cancer, Biochem Biophys Res Commun, 287: 705-713, 2001).
  • Histone deacetylase inhibitors have been found to be useful for the activation of genes responsive to hormone receptors.
  • HDACIs are potent inducers of growth arrest, differentiation and/or apoptosis of several cell lines, and they constitute a novel class of chemotherapeutic agents initially identified by their ability to reverse the malignant phenotype of transformed cells. They have been shown to activate differentiation programs, inhibit cell cycle, and induce apoptosis in a wide range of tumor-derived cell lines and to block angiogenesis and stimulate the immune system in vivo (Marks et al., Histone deacetylases and cancer, causes and therapies. Nat Rev Cancer, 1: 194-202, 2001 ; Johnstone, R. W.
  • Histone-deacetylase inhibitors novel drugs for the treatment of cancer, Nat Rev Drug Discov, 1: 287-299, 2002.). Whereas the mechanisms through which HDACIs exert these anti-tumor activities have not been fully delineated, induction of histone hyperacetylation and modulation of gene transcription through chromatin remodeling are thought to be primarily responsible, leading to the selective activation of genes associated with cell growth and survival.
  • Suberoylanilide hydroxamic acid (SAHA, Vorinostat ® or Zolinza ® ) was approved in 2006 for the treatment of patients with relapsed or refractory cutaneous T-cell lymphoma (Marks et al, Dimethyl sulfoxide to vorinostat. development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol, 25: 84-90, 2007.).
  • SAHA suberanilide hydroxamic acid
  • Histone acetyltransferase and histone deacetylase have opposing effect on transcription (Ito et al., Histone acetylation and histone deacetylation. MoI Biotechnol, 20: 99-106, 2002; Kuo et al., Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays, 20: 615-626, 1998. ).
  • D ⁇ A methylation and histone deacetylation of tumor suppressor genes occur in many human cancers, leading to suppression of function of these genes thereby conferring a growth advantage for the tumor cells (Macaluso et al., A.
  • HDACIs such as SAHA, and N-(2-aminophenyl)4-[N-(pyridine-3-yl-methoxy-carbonyl)aminomethyl] benzamide (MS-275) can directly interact with the HDAC enzymes at the catalytic site and inhibit their function (Bolden et al, Anticancer activities of histone deacetylase inhibitors.
  • MS-275 is now in phase I/II clinical trials for various solid tumors and hematological malignancies (Hess-Stumpp et al., MS-275. a potent orally available inhibitor of histone deacetylases-The development of an anticancer agent. Int J Biochem Cell Biol, 39: 1388-1405, 2007.).
  • Suberoylanilide hydroxamic acid is a modest HDACI and has been used extensively in vitro and in vivo in cancer models (Butler et al., Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase. suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res, 60: 5165-5170, 2000.) and it is the only HDACI currently approved for clinical use (Marks et al, Dimethyl sulfoxide to vorinostat.
  • FIG. 1 shows structures of SAHA, MS-275 and CI-994 HDACIs and their IC50 values.
  • Suzuki et al (Suzuki et al Synthesis and histone deacetylase inhibitory activity of new benzamide derivatives, J Med Chem 1999, 42, (15), 3001-3) discloses benzamide derivatives having histone deacetylase inhibitory activity and methods of making benzamide derivatives having histone deacetylase inhibitory activity. Suzuki et al is hereby incorporated herein by reference in its entirety.
  • Ri, R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, -NHAc, -OH, alkyl, alkoxy, halogen, an amino group, a nitro group, a cyano group, an aminoalkyl group, an alkylamino group, an acyl group, an acylamino group, a thiol, thiolurea, an alkylthio group, a perfluoroalkyl group, a perfluoroalkyloxy group, a carboxyl group and an alkoxycarbonyl group.
  • the alkyl group, the alkoxy group, the aminoalkyl group, the alkylamino group, the acyl group, the acylamino group, the alkylthio group, the perfluoroalkyl group, and the perfluoroalkyloxy group substituents may contain a lower alkyl group (C 1-4).
  • the alkyl groups may be linear or branched and substituted or unsubstituted.
  • the alkyl may -CH 3 .
  • the alkoxy may be -OCH 3 .
  • the halogen may be F or Cl.
  • Ri is H, an amino group, -NHAc, or -OH. More preferably Ri is an amino group.
  • R 2 -R 4 are H, an amino group, alkyl, alkoxy, or halogen. More preferably
  • R 2 -R 4 are each H.
  • Table 1 shows some examples of benzamide derivatives of Formula M-I
  • M- 1 is of structural formula MS-275
  • Rj, R 2 , R 3 , and R 4 are each independently selected from the group consisting of H, -NHAc, -OH, alkyl, alkoxy, halogen, an amino group, a nitro group, a cyano group, an aminoalkyl group, an alkylamino group, an acyl group, an acylamino group, a thiol, thiolurea, an alkylthio group, a perfluoroalkyl group, a perfluoroalkyloxy group, a carboxyl group and an alkoxycarbonyl group.
  • Compound 7A may be converted into acyimidazole using CDI in THF at room temperature and then condensed in situ with 4-(aminomethyl)benzoic acid in the presence of DBU and triethylamine in THF at room temperature afforded Compound 8A.
  • Compound 8A may be treated with CDI in THF at elevated temperature, for example, about 55-60 0 C, to form imidazolide, which may be cooled and further reacted in situ with Compound 9A in the presence of TFA to yield Compound M-I.
  • THF tetrahydrofuran
  • aprotic solvents such as aprotic solvents, electron donating solvents, such as aceonitrile (ACN) and dimethylformaide (DMF) solvents.
  • ACN aceonitrile
  • DMF dimethylformaide
  • the compounds of structural formula MS-275 may be synthesized using two transformations such as those described in Scheme 4a above.
  • Conventional column chromatography for purification may be used.
  • Applicants have also developed a simpler process for large scale purification of crude benzamide derivatives having histone deacetylase inhibitory activity of structural formula M- 1.
  • work up procedures well known in the art may be employed. For example, the solvent may be evaporated and a mixture of solvent and water added to the concentrate while stirring, for about one hour. The resulting precipitate may be filtered, washed with solvent and dried.
  • the crude product may be further stirred twice in dichloromethane to remove excess of 1,2-phenylenediamine, filtered and washed with solvent to yield purified benzamide derivatives having histone deacetylase inhibitory activity of structural formula M-I .
  • the solvents may be short-chain hydrocarbons.
  • the solvent may be pentane, hexane or ether, including pet ether, and the volume ratio of solvent to water of about 1 : 1, 1 :2, 1 :3, 1 :4, 1:5, 1 :6, 1 :7, 1 :8, 1 :9, 1 : 10, 2:1, 2:2, 2:3, 2:4, 2:5, 2:6, 2:7, 2:8, 2:9, 2: 10 or the like.
  • the yield was about 80.0% yield (>99% as determined by HPLC).
  • the overall yield for the simple and efficient production of MS-275 (4) was 72.8%.
  • the compound may be used in a pharmaceutical composition.
  • the pharmaceutical composition may be formulated for oral administration, parentral administration or for injectable administration.
  • the compound in making compositions with the compounds of the present invention, can be mixed with a pharmaceutically acceptable carrier or an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container.
  • a pharmaceutically acceptable carrier or an excipient When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the novel compound.
  • the compositions can be in the form of tablets, pills, powers, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, soft and hard gelatin capsules, and other orally ingestible formulations.
  • compositions may be in the form of a solution, suspension, tablet, capsule or the like, prepared according to methods well known in the art. It is also contemplated that administration of such compositions may be by the oral, injectable and/or parenteral routes depending upon the needs of the artisan.
  • the novel compound can be administered by nasal or oral inhalation, oral ingestion, injection (intramuscular, intravenous, and intraperitoneal), transdermal Iy, or other forms of administration.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propyl-hydroxybenzoates, sweetening agents; and flavoring agents.
  • the compositions of the present invention can also be formulated so as to provide quick, sustained or delayed release of the novel compound after administration to the patient by employing procedures known in the art.
  • pharmaceutically acceptable carrier refers to those components in the particular dosage form employed which are considered inert and are typically employed in the pharmaceutical arts to formulate a dosage form containing a particular active compound. This may include without limitation solids, liquids and gases, used to formulate the particular pharmaceutical product.
  • carriers include diluents, flavoring agents, solubilizers, suspending agents, binders or tablet disintegrating agents, encapsulating materials, penetration enhancers, solvents, emolients, thickeners, dispersants, sustained release forms, such as matrices, transdermal delivery components, buffers, stabilizers, and the like. Each of these terms is understood by those of ordinary skill.
  • Aerosol formulations for use in this invention typically include propellants, such as a fluorinated alkane, surfactants and co-solvents and may be filled into aluminum or other conventional aerosol containers which are then closed by a suitable metering valve and pressurized with propellant, producing a metered dose inhaler. Aerosol preparations are typically suitable for nasal or oral inhalation, and may be in powder or solution form, in combination with a compressed gas, typically compressed air. Additionally, aerosols may be useful topically.
  • propellants such as a fluorinated alkane, surfactants and co-solvents
  • Aerosol preparations are typically suitable for nasal or oral inhalation, and may be in powder or solution form, in combination with a compressed gas, typically compressed air. Additionally, aerosols may be useful topically.
  • the amount of the novel compound used in the treatment methods is that amount which effectively achieves the desired therapeutic result in animals.
  • the dosages of the various novel compounds will vary somewhat depending upon the parent compound, rate of in vivo hydrolysis, etc. Those skilled in the art can determine the optimal dosing of the novel compound selected based on clinical experience and the treatment indication.
  • the amount of the novel compound is 0.1 to 100 mg/kg of body weight, more preferably, 5 to 40 mg/kg.
  • Suitable solid carriers are known, e.g., magnesium carbonate, magnesium stearate, talc, lactose and the like. These carriers are typically used in oral tablets and capsules.
  • Suitable carriers for oral liquids include, e.g., water, ethanol, propylene glycol and others.
  • Topical preparations useful herein include creams, ointments, solutions, suspensions and the like. These may be formulated to enable one to apply the appropriate dosage topically to the affected area once daily, up to 3-4 times daily as appropriate. Topical sprays may be included herein as well.
  • transdermal delivery may be an option, providing a relatively steady state delivery of the medication which is preferred in some circumstances. Transdermal delivery typically involves the use of a compound in solution, with an alcoholic vehicle, optionally a penetration enhancer, such as a surfactant and other optional ingredients. Matrix and reservoir type transdermal delivery systems are examples of suitable transdermal systems. Transdermal delivery differs from conventional topical treatment in that the dosage form delivers a systemic dose of medication to the patient.
  • the compounds can also be converted into a pharmaceutically acceptable salt or pharmaceutically acceptable solvate or other physical forms (e.g., polymorphs by way of example only and not limitation) via known in the art field methods.
  • Figure 1 shows structures of SAHA, MS-275 and CI-994 HDACIs and their IC50 values.
  • CDI 1 , 1 '-Carbonyldiimidazole (C 3 H 3 N 2 )2CO)
  • TFA Trifluoroacetic acid (CF 3 CO 2 H)

Abstract

Simple and efficient procedures for the synthesis of histone deacetylase inhibitors. The procedure may provide MS-275 in 72% overall yields.

Description

TITLE OF INVENTION:
SYNTHESIS METHODS OF HISTONE DEACETYLASE INHIBITORS (HDACIs) CROSS REFERENCE TO RELATED APPLICATIONS:
[01] This Application claims benefit to U.S. Provisional Application 61/012,269, filed
December 7, 2007 and claims benefit to U.S. Provisional Application 61/012,709, filed
December 10, 2007, each of which are hereby incorporated by reference in their entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR
DEVELOPMENT:
[02] This invention was made with the support of the U.S. government under Grant
Number CAl 17991 from the National Institutes of Health and the National cancer Institute
(NCI) and Grant Number W81XWH-04-1-0101 from the U.S. Department of Defense. The
U.S. government has certain rights in this invention.
NAMES OF PARTIES OF A JOINT RESEARCH AGREEMENT:
[03] Not Applicable
INCORPORATION BY REFERENCE OF MATERIAL SUBMITTED ON A
COMPACT DISC:
[04] Not Applicable
BACKGROUND OF THE INVENTION
Prostate Cancer
[05] Prostate cancer (PCA) is the most common malignancy and age-related cause of cancer death worldwide. Apart from lung cancer, PCA is the most common form of cancer in men and the second leading cause of death in American men. In the United States in 2007, an estimated 218,890 new case of prostate cancer will be diagnosed and about 27,050 men will die of this disease. (Jemal et al, Cancer statistics, CA Cancer J Clin 2007, 57, (1), 43-66).
The growth of most prostate tumors depends on androgens during the initial stages of tumor development, and thus, anti-hormonal therapy by surgical or medical suppression of androgen action remains a major treatment option of the disease. (Denmeade and Isaacs, A history of prostate cancer treatment. Nat Rev Cancer 2002, 2, (5), 389-96). Although this treatment may be initially successful, most tumors eventually recur due to the expansion of an androgen-refractory population of PCA cells. (Barry et al, A nationwide survey of practicing urologists: current management of benign prostatic hyperplasia and clinically localized prostate cancer. J Urol 1997, 158, (2), 488-91; discussion 492). Treatment of androgen-independent tumors with cytotoxic agents is generally unsatisfactory, and, at this stage, the disease is usually fatal to the patient (Singh et al., Combinatorial androgen receptor targeted therapy for prostate cancer. Endocr Relat Cancer, 13: 653-666, 2006.). Metastatic disease that develops even after potentially curative surgery remains a major clinical challenge. Therapeutic treatments for patients with metastatic PCA are limited because current chemotherapeutic and radiotherapeutic regimens are largely ineffective. (Feldman and Feldman, The development of androgen-independent prostate cancer. Na/ Rev Cancer 2001, l, (l), 34-45).
[06] Since androgen-independent, metastatic prostate cancer is incurable up to now, there exist a need for treatments that block proliferation and induce differentiation and/or apoptosis of prostate cancer cells.
[07] There is an urgent need to develop new therapeutic agents with defined targets to prevent and treat PCA. [08] Histone Deacetylases
[09] Histone Deacetylases (HDACs) are the catalytic subunits of multiprotein complexes responsible for deacetylation of histones and nonhistone proteins. Lysine acetylation, i.e., the transfer of an acetyl moiety from acetyl-coenzyme A to the ε-amino group of a specific lysine residue, has emerged as the major form of posttranslational modification of histones, and other proteins have been correlated with transcription, chromatin assembly, DNA repair, and recombinatorial events (Marks et al., Histone deacetylases and cancer, causes and therapies. Nat Rev Cancer, 1: 194-202, 2001.)- Histone acetylation in vivo is a dynamic, reversible process governed by the opposite actions of histone acetyltransferases (HATs) and HDACs. Aberrant acetylation of histone tails, emerging from either HAT mutation or abnormal recruitment of HDACs, has been linked to carcinogenesis (Pandolfi, P. P. Transcription therapy for cancer. Oncogene, 20: 3116-3127, 2001). In various cases, altered HAT or HDAC activity has been identified in a variety of cancers. It has recently been demonstrated that the expression and activity of HDACl is up-regulated in prostate cancer compared to benign prostatic hyperplasia (BPH) (Patra et al., Histone deacetylase and DNA methyltransferase in human prostate cancer, Biochem Biophys Res Commun, 287: 705-713, 2001).
[10] Histone deacetylase inhibitors (HDACIs) have been found to be useful for the activation of genes responsive to hormone receptors. HDACIs are potent inducers of growth arrest, differentiation and/or apoptosis of several cell lines, and they constitute a novel class of chemotherapeutic agents initially identified by their ability to reverse the malignant phenotype of transformed cells. They have been shown to activate differentiation programs, inhibit cell cycle, and induce apoptosis in a wide range of tumor-derived cell lines and to block angiogenesis and stimulate the immune system in vivo (Marks et al., Histone deacetylases and cancer, causes and therapies. Nat Rev Cancer, 1: 194-202, 2001 ; Johnstone, R. W. Histone-deacetylase inhibitors, novel drugs for the treatment of cancer, Nat Rev Drug Discov, 1: 287-299, 2002.). Whereas the mechanisms through which HDACIs exert these anti-tumor activities have not been fully delineated, induction of histone hyperacetylation and modulation of gene transcription through chromatin remodeling are thought to be primarily responsible, leading to the selective activation of genes associated with cell growth and survival. Suberoylanilide hydroxamic acid (SAHA, Vorinostat® or Zolinza®) was approved in 2006 for the treatment of patients with relapsed or refractory cutaneous T-cell lymphoma (Marks et al, Dimethyl sulfoxide to vorinostat. development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol, 25: 84-90, 2007.).
[11] One of the early HDACIs discovered is N-hydroxy-N'-phenylactanediamide, also called suberanilide hydroxamic acid (SAHA). (Richon et al., Second generation hybrid polar compounds are potent inducers of transformed cell differentiation, Proc Natl Acad Sci USA 1996, 93, (12), 5705-8; Kelly et al., Phase I clinical trial of histone deacetylase inhibitor, suberoylanilide hydroxamic acid administered intravenously, Clin Cancer Res 2003, 9, (10 Pt 1), 3578-88.) This compound (trade name: Vorinostat®) was approved in 2006 by the U.S. Food and Drug Administration (FDA) for the treatment of advanced cutaneous T-cell- lymphoma. (Bolden et al., Anticancer activities of histone deacetylase inhibitors. Nat Rev DrugDiscov 2006, 5, (9), 769-84.)
[12] Histone acetyltransferase and histone deacetylase (HDAC) have opposing effect on transcription (Ito et al., Histone acetylation and histone deacetylation. MoI Biotechnol, 20: 99-106, 2002; Kuo et al., Roles of histone acetyltransferases and deacetylases in gene regulation. Bioessays, 20: 615-626, 1998. ). Often, DΝA methylation and histone deacetylation of tumor suppressor genes occur in many human cancers, leading to suppression of function of these genes thereby conferring a growth advantage for the tumor cells (Macaluso et al., A. How does DΝA methylation mark the fate of cells?. Tumori, 90: 367-372, 2004; Robertson et al. DΝA methylation: past, present and future directions. Carcinogenesis, 21: 461-467, 2000.). It has recently been demonstrated that the expression and activity of HDACl is up-regulated (2 -4-fold) in prostate cancer compared to benign prostatic hyperplasia (Patra et al., Histone deacetylase and DΝA methyltransferase in human prostate cancer. Biochem Biophys Res Commun, 287: 705-713, 2001.). HDACIs, such as SAHA, and N-(2-aminophenyl)4-[N-(pyridine-3-yl-methoxy-carbonyl)aminomethyl] benzamide (MS-275) can directly interact with the HDAC enzymes at the catalytic site and inhibit their function (Bolden et al, Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov, 5: 769-784, 2006; Marks et al., Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells, J Natl Cancer Inst, 92: 1210-1216, 2000; Minucci et al., Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer, Nat Rev Cancer, 6: 38-51, 2006.). This leads to acetylation of histones which opens-up the chromatin structure allowing transcription of anti-growth and pro- apoptotic genes to occur. MS-275 is now in phase I/II clinical trials for various solid tumors and hematological malignancies (Hess-Stumpp et al., MS-275. a potent orally available inhibitor of histone deacetylases-The development of an anticancer agent. Int J Biochem Cell Biol, 39: 1388-1405, 2007.).
[13] Suberoylanilide hydroxamic acid (SAHA) is a modest HDACI and has been used extensively in vitro and in vivo in cancer models (Butler et al., Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase. suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res, 60: 5165-5170, 2000.) and it is the only HDACI currently approved for clinical use (Marks et al, Dimethyl sulfoxide to vorinostat. development of this histone deacetylase inhibitor as an anticancer drug, Nat Biotechnol, 25: 84-90, 2007; Richon et al., Histone deacetylase inhibitors, development of suberoylanilide hydroxamic acid fSAHA) for the treatment of cancers. Blood Cells MoI Dis, 27: 260-264, 2001.). The HDACI benzamide compound MS-275 is in several clinical trials as a potential therapy for a variety of cancers (Hess-Stumpp et al, MS-275, a potent orally available inhibitor of histone deacetylases— the development of an anticancer agent, Int J Biochem Cell Biol, 39: 1388- 1405, 2007.). [14] Figure 1 shows structures of SAHA, MS-275 and CI-994 HDACIs and their IC50 values.
[15] Synthesis Methods
[16] Synthesis of benzamide derivatives having histone deacetylase inhibitory activity
[17] Suzuki et al (Suzuki et al Synthesis and histone deacetylase inhibitory activity of new benzamide derivatives, J Med Chem 1999, 42, (15), 3001-3) discloses benzamide derivatives having histone deacetylase inhibitory activity and methods of making benzamide derivatives having histone deacetylase inhibitory activity. Suzuki et al is hereby incorporated herein by reference in its entirety.
[18] An example of the synthesis method of Suzuki et al to produce MS-275 via a three- step procedure in 50.96% overall yield is outlined in Scheme 3 below.
Scheme 3: Previous Procedure for Synthesis of MS-275 en rt, 4h
(used without purification)
Figure imgf000007_0001
[Overall yield: 0.91 x 0.56 x 100 = 50.96%;
Figure imgf000007_0002
MS-275 [19] In addition to the modest overall yield, the procedure of Suzuki et al has other disadvantages, such as a tedious method for the preparation of an acid chloride using oxalyl chloride and requiring the use of column chromatography for purification.
[20] Thus, there continues to be a need for efficient and high yield production of HDACIs.
BRIEF SUMMARY OF THE INVENTION
[21] METHODS OF SYNTHESES
[22] Applicants have discovered improved syntheses of HDACIs.
Synthesis of benzamide derivatives having histone deacetylase inhibitory activity
[23] Applicants have developed a new two-step procedure for preparation of benzamide derivatives having histone deacetylase inhibitory activity of structural formula M-I :
Figure imgf000008_0001
M-I
[24] Where Ri, R2, R3, and R4 are each independently selected from the group consisting of H, -NHAc, -OH, alkyl, alkoxy, halogen, an amino group, a nitro group, a cyano group, an aminoalkyl group, an alkylamino group, an acyl group, an acylamino group, a thiol, thiolurea, an alkylthio group, a perfluoroalkyl group, a perfluoroalkyloxy group, a carboxyl group and an alkoxycarbonyl group. [25] The alkyl group, the alkoxy group, the aminoalkyl group, the alkylamino group, the acyl group, the acylamino group, the alkylthio group, the perfluoroalkyl group, and the perfluoroalkyloxy group substituents may contain a lower alkyl group (C 1-4). The alkyl groups may be linear or branched and substituted or unsubstituted.
[26] The alkyl may -CH3. The alkoxy may be -OCH3. The halogen may be F or Cl.
[27] Preferably Ri is H, an amino group, -NHAc, or -OH. More preferably Ri is an amino group.
[28] Preferably R2-R4 are H, an amino group, alkyl, alkoxy, or halogen. More preferably
R2-R4 are each H.
[29] Table 1 shows some examples of benzamide derivatives of Formula M-I
[30] Table 1
Figure imgf000009_0001
= Concentration required to inhibit by 50%. [31 ] Preferably M- 1 is of structural formula MS-275
Figure imgf000010_0001
MS-275
[32] The new two-step procedure is outlined in Scheme 4a.
Figure imgf000010_0002
where in formulae 7A, 8A and 9A, Rj, R2, R3, and R4 are each independently selected from the group consisting of H, -NHAc, -OH, alkyl, alkoxy, halogen, an amino group, a nitro group, a cyano group, an aminoalkyl group, an alkylamino group, an acyl group, an acylamino group, a thiol, thiolurea, an alkylthio group, a perfluoroalkyl group, a perfluoroalkyloxy group, a carboxyl group and an alkoxycarbonyl group. [33] Compound 7A may be converted into acyimidazole using CDI in THF at room temperature and then condensed in situ with 4-(aminomethyl)benzoic acid in the presence of DBU and triethylamine in THF at room temperature afforded Compound 8A. Compound 8A may be treated with CDI in THF at elevated temperature, for example, about 55-600C, to form imidazolide, which may be cooled and further reacted in situ with Compound 9A in the presence of TFA to yield Compound M-I.
Other known solvents may be used in place of THF (tetrahydrofuran), such as aprotic solvents, electron donating solvents, such as aceonitrile (ACN) and dimethylformaide (DMF) solvents. Known weak acids may also be used.
Other known reagents in organic synthesis, such as peptide type coupling agents, may be used in place of CDI.
[34] The compounds of structural formula M-I may be synthesized using two transformations such as those described in Scheme 4a above.
[35] While Applicants' new synthesis method may be preformed in two-steps, the synthesis may include other steps.
[36] The synthesis of MS-275 is shown below in Scheme 4 as an example of Applicants invention of a two-step procedure: [37] Scheme 4: Preparation of MS-275
Figure imgf000012_0001
Scheme 4: New Synthesis of MS-275 (4)
Figure imgf000013_0001
Condensation of 3-(hydroxymethyl)pyridine (7) and 4-(aminomethyl)benzoic in the presence of CDI gave 4-[N-(pyridin-3-ylmethoxycarbonyl)aminomethyl]benzoic Acid (8) in 91.0% yield. In the previous method of Suzuki et ah, the carboxylic acid derivative 8 was first converted into acyl chloride hydrochloride by treatment of oxalyl chloride in toluene and then reacted with imidazole to form the acylimidazole intermediate. (Suzuki et al., Synthesis and histone deacetylase inhibitory activity of new benzamide derivatives. J Med Chem 1999, 42, (15), 3001-3.). However, Applicants synthesized the imidazolide of intermediate 8 by treatment with CDI at about 55-60 0C in THF. The imidazolide was cooled to ambient and further reacted in situ with 1,2-phenylenediamine in the presence of TFA to afford MS-275
(4).
[38] While the reagents disclosed herein are named specifically, the skilled artisan is aware of equivalent reagents that function in substantially the same manner as the reagents disclosed hererin. A non-limiting example is that although CDI is employed, any compound that converts alcohols and amines into carbamates, esters and/or ureas is contemplated.
[39] The compounds of structural formula MS-275 may be synthesized using two transformations such as those described in Scheme 4a above. [40] Conventional column chromatography for purification may be used. However, Applicants have also developed a simpler process for large scale purification of crude benzamide derivatives having histone deacetylase inhibitory activity of structural formula M- 1. At or after completion, work up procedures well known in the art may be employed. For example, the solvent may be evaporated and a mixture of solvent and water added to the concentrate while stirring, for about one hour. The resulting precipitate may be filtered, washed with solvent and dried. The crude product may be further stirred twice in dichloromethane to remove excess of 1,2-phenylenediamine, filtered and washed with solvent to yield purified benzamide derivatives having histone deacetylase inhibitory activity of structural formula M-I .
[41] The solvents may be short-chain hydrocarbons. The solvent may be pentane, hexane or ether, including pet ether, and the volume ratio of solvent to water of about 1 : 1, 1 :2, 1 :3, 1 :4, 1:5, 1 :6, 1 :7, 1 :8, 1 :9, 1 : 10, 2:1, 2:2, 2:3, 2:4, 2:5, 2:6, 2:7, 2:8, 2:9, 2: 10 or the like. [42] For purification of MS-275 using dichloromethane as an exemplary solvent, the yield was about 80.0% yield (>99% as determined by HPLC). The overall yield for the simple and efficient production of MS-275 (4) was 72.8%.
[43] The compound may be used in a pharmaceutical composition. The pharmaceutical composition may be formulated for oral administration, parentral administration or for injectable administration.
[44] In making compositions with the compounds of the present invention, the compound can be mixed with a pharmaceutically acceptable carrier or an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier, or medium for the novel compound. Thus, the compositions can be in the form of tablets, pills, powers, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, soft and hard gelatin capsules, and other orally ingestible formulations.
[45] The pharmaceutical compositions may be in the form of a solution, suspension, tablet, capsule or the like, prepared according to methods well known in the art. It is also contemplated that administration of such compositions may be by the oral, injectable and/or parenteral routes depending upon the needs of the artisan. The novel compound can be administered by nasal or oral inhalation, oral ingestion, injection (intramuscular, intravenous, and intraperitoneal), transdermal Iy, or other forms of administration. [46] Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propyl-hydroxybenzoates, sweetening agents; and flavoring agents. The compositions of the present invention can also be formulated so as to provide quick, sustained or delayed release of the novel compound after administration to the patient by employing procedures known in the art.
[47] The term "pharmaceutically acceptable carrier" refers to those components in the particular dosage form employed which are considered inert and are typically employed in the pharmaceutical arts to formulate a dosage form containing a particular active compound. This may include without limitation solids, liquids and gases, used to formulate the particular pharmaceutical product. Examples of carriers include diluents, flavoring agents, solubilizers, suspending agents, binders or tablet disintegrating agents, encapsulating materials, penetration enhancers, solvents, emolients, thickeners, dispersants, sustained release forms, such as matrices, transdermal delivery components, buffers, stabilizers, and the like. Each of these terms is understood by those of ordinary skill.
[48] Aerosol formulations for use in this invention typically include propellants, such as a fluorinated alkane, surfactants and co-solvents and may be filled into aluminum or other conventional aerosol containers which are then closed by a suitable metering valve and pressurized with propellant, producing a metered dose inhaler. Aerosol preparations are typically suitable for nasal or oral inhalation, and may be in powder or solution form, in combination with a compressed gas, typically compressed air. Additionally, aerosols may be useful topically.
[49] Generally, the amount of the novel compound used in the treatment methods is that amount which effectively achieves the desired therapeutic result in animals. Naturally, the dosages of the various novel compounds will vary somewhat depending upon the parent compound, rate of in vivo hydrolysis, etc. Those skilled in the art can determine the optimal dosing of the novel compound selected based on clinical experience and the treatment indication. Preferably the amount of the novel compound is 0.1 to 100 mg/kg of body weight, more preferably, 5 to 40 mg/kg.
[50] Suitable solid carriers are known, e.g., magnesium carbonate, magnesium stearate, talc, lactose and the like. These carriers are typically used in oral tablets and capsules.
[51] Suitable carriers for oral liquids include, e.g., water, ethanol, propylene glycol and others.
[52] Topical preparations useful herein include creams, ointments, solutions, suspensions and the like. These may be formulated to enable one to apply the appropriate dosage topically to the affected area once daily, up to 3-4 times daily as appropriate. Topical sprays may be included herein as well. [53] Depending upon the particular compound selected, transdermal delivery may be an option, providing a relatively steady state delivery of the medication which is preferred in some circumstances. Transdermal delivery typically involves the use of a compound in solution, with an alcoholic vehicle, optionally a penetration enhancer, such as a surfactant and other optional ingredients. Matrix and reservoir type transdermal delivery systems are examples of suitable transdermal systems. Transdermal delivery differs from conventional topical treatment in that the dosage form delivers a systemic dose of medication to the patient.
[54] The compounds can also be converted into a pharmaceutically acceptable salt or pharmaceutically acceptable solvate or other physical forms (e.g., polymorphs by way of example only and not limitation) via known in the art field methods.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS:
[55] Figure 1 shows structures of SAHA, MS-275 and CI-994 HDACIs and their IC50 values.
DETAILED DESCRIPTION OF THE INVENTION
[56] ABBREVIATIONS
[57] CDI = 1 , 1 '-Carbonyldiimidazole (C3H3N2)2CO)
[58] THF = Tetrahydrofuran (C4H8O)
[59] DBU = l,8-Diazabicyclo[5.4.0]undec-7-ene
[60] TFA = Trifluoroacetic acid (CF3CO2H)
[61 ] Et3N = triethylamine
Experimental Section
[62] iV-(2-Aminophenyl)-4-[iV-(pyridin-3-ylmethoxycarbonyl) aminomethyl] benzamide (4, MS-275).
[63] To a suspension of 4-[N-(Pyridin-3-ylmethoxycarbonyl)aminomethyl]benzoic
Acid (5.0 g, 0.017 mol) in THF (100 mL) was added CDI (3.12 g, 0.019 mol), and the mixture stirred for 3 h at 60 0C. After formation of acylimidazole the clear solution was cooled to room temperature (rt). To this was added 1,2-phenylenediamine (15.11 g, 0.14 mmol) and trifluoroacetic acid (1.2 mL, 0.015 mol) and then stirred for 16 h. The reaction mixture was evaporated to remove THF and crude product was stirred in a mixture of hexane and water (2:5, v/v) for 1 h and filtered and dried. The residue was stirred in dichloromethane twice to afford pure MS-275 (4) as off white powder 5.25 g, 80% yield: mp 159-160 * C; IR (KBr) 3295, 1648, 1541, 1508, 1457, 1309, 1183, 742 cm"1. 1H NMR (DMSO-J6) δ 4.28 (d, 2H, J = 5.9 Hz), 4.86 (s, 2H), 5.10 (s, 2H), 6.60 (t, IH, J = 7.3 Hz), 6.78 (d, IH, J = 7 Hz), 6.97 (t, IH, J= 7 Hz), 7.17 (d, IH, J= 8 Hz), 7.3-7.5(m, 3H), 7.78 (d, IH, J= 8 Hz), 7.93 (d, 2H, J = 8 Hz), 8.53 (d, IH, J = 3.7 Hz), 8.59 (s, IH), 9.61 (s, IH); HRMS: calcd 376.1560 (C2iH2oN4θ3), found 376.1558. These spectral and analytical data are as previously reported in J Med Chem 1999, 42, (15), 3001-3.
[64] 4-[7V-(Pyridin-3-ylmethoxycarbonyI)aminomethyl] benzoic Acid (8) may be prepared as follows. To a suspension of l, l'-carbonyldiimidazole (CDI, 25.6 g, 158 mmol) in THF (120 mL) was added 3-pyridinemethanol (7, 17.3 g, 158 mmol) in THF (50 mL) at 10 0C, and the mixture stirred for 1 h at rt. The resulting solution was added to a suspension of 4-(aminomethyl)benzoic acid (22.6 g, 158 mmol), DBU (24.3 g, 158 mmol), and triethylamine (22.2 mL, 158 mmol) in THF (250 mL). After stirring for 5 h at rt, the mixture was evaporated to remove THF and then dissolved in water (300 mL). The solution was acidified with HCl (pH 5) to precipitate a white solid which was collected by filtration, washed with water (300 mL) and methanol (50 mL), respectively, and dried to yield pure 8 (41.1 g, 91% yield): mp 207-208 0 C; IR (KBr) 3043, 1718, 1568, 1434, 1266, 1 108, 1037, 984, 756 cm4; 1H NMR (DMSO-^6) δ 4.28 (d, 2H, J= 5.9 Hz), 5.10 (s, 2H), 7.3-7.5 (m, 3H), 7.7-8.1 (m, 4H), 8.5-8.7 (m, 2H). These spectral and analytical data are as previously reported in Suzuki et al, J Med Chem 1999, 42, (15), 3001-3.

Claims

We Claim:
Claim 1. A method of making a benzamide derivative having histone deacetylase inhibitory activity of structural formula M-I
Figure imgf000019_0001
M-I
said method comprising condensing Compound 7A and 4-(aminomethyl)benzoic acid to yield imidazole Compound 8A:
Figure imgf000019_0002
reacting Compound 8A with Compound 9A to yield a compound of structural formula
M-I,
Figure imgf000020_0001
wherein in formulae 7A, 8A, 9A, and M-I, Ri, R2, R3, and R4 are each independently selected from the group consisting of H, -NHAc, -OH, alkyl, alkoxy, halogen, an amino group, a nitro group, a cyano group, an aminoalkyl group, an alkylamino group, an acyl group, an acylamino group, a thiol, thiolurea, an alkylthio group, a perfluoroalkyl group, a perfluoroalkyloxy group, a carboxyl group and an alkoxycarbonyl group.
Claim 2. The method of claim 1, wherein Ri is an amino group.
Claim 3. The method of claim 1, wherein R2-R4 are each independently H, an amino group, alkyl, alkoxy, or halogen.
Claim 4. The method of claim 1, wherein R2-R4 are each independently F or H.
Claim 5. The method of claim 1, wherein R2-R4 are each H.
Claim 6. The method of claim 1, wherein Compound 9A is 1,2-phenylenediamine and structural formula M-I is:
Figure imgf000021_0001
Claim 7. The method of claim 1, wherein the compound 7A is converted to acylimidazole at ambient temperature and then condensed in situ with 4- (aminomethyl)benzoic acid at ambient temperature to form Compound 8A.
Claim 8. The method of of claim 1, further comprising converting Compound 8A into an imidazolide derivative at elevated temperature and the reacting step is conducted in situ with Compound 9A to yield Compound M-I .
Claim 9. A method of making a benzamide derivative having histone deacetylase inhibitory activity of structural formula M-275, comprising condensing 3-(hydroxymethyl)pyridine (7) and 4-(aminomethyl)benzoic to give 4-[N- (pyridin-3-ylmethoxycarbonyl)aminomethyl]benzoic Acid; synthesizing an imidazolide derivative of said 4-[N-(pyridin-3- ylmethoxycarbonyl)aminomethyl]benzoic Acid by treatment with CDI at elevated temperature in THF; cooling said imidazolide derivative to ambient; and reacting said imidazolide derivative in situ with 1,2-phenylenediamine to yield:
Figure imgf000022_0001
(MS-275).
PCT/US2008/085684 2007-12-07 2008-12-05 Synthesis methods of histone deacetylase inhibitors (hdacis) WO2009076206A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US1226907P 2007-12-07 2007-12-07
US61/012,269 2007-12-07
US1270907P 2007-12-10 2007-12-10
US61/012,709 2007-12-10

Publications (1)

Publication Number Publication Date
WO2009076206A1 true WO2009076206A1 (en) 2009-06-18

Family

ID=40755835

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/085684 WO2009076206A1 (en) 2007-12-07 2008-12-05 Synthesis methods of histone deacetylase inhibitors (hdacis)

Country Status (1)

Country Link
WO (1) WO2009076206A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104610133A (en) * 2015-01-26 2015-05-13 亿腾药业(泰州)有限公司 Method for synthesizing novel anticancer medicine entinostat
CN104876857A (en) * 2015-05-12 2015-09-02 亿腾药业(泰州)有限公司 Preparation of benzamide histone deacetylase inhibitor with differentiation and anti-proliferation activity
EP3168210A1 (en) 2015-11-13 2017-05-17 Sandoz Ag Crystalline forms of entinostat

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0847992B1 (en) * 1996-09-30 2004-06-23 Schering Aktiengesellschaft Benzamide derivatives, useful as cell differentiation inducers
US7244751B2 (en) * 2003-02-14 2007-07-17 Shenzhen Chipscreen Biosciences Ltd. Histone deacetylase inhibitors of novel benzamide derivatives with potent differentiation and anti-proliferation activity

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0847992B1 (en) * 1996-09-30 2004-06-23 Schering Aktiengesellschaft Benzamide derivatives, useful as cell differentiation inducers
US7244751B2 (en) * 2003-02-14 2007-07-17 Shenzhen Chipscreen Biosciences Ltd. Histone deacetylase inhibitors of novel benzamide derivatives with potent differentiation and anti-proliferation activity

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MAI A: "Histone deacetylation in epigenetics: an attractive target for anticancer therapy", MED RES REV., vol. 25, no. 3, May 2005 (2005-05-01), pages 261 - 309 *
SUZUKI T ET AL.: "Synthesis and histone deacetylase inhibitory activity of new benzamide derivatives", J MED CHEM., vol. 42, no. 15, 29 July 1999 (1999-07-29), pages 3001 - 3003 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104610133A (en) * 2015-01-26 2015-05-13 亿腾药业(泰州)有限公司 Method for synthesizing novel anticancer medicine entinostat
CN104876857A (en) * 2015-05-12 2015-09-02 亿腾药业(泰州)有限公司 Preparation of benzamide histone deacetylase inhibitor with differentiation and anti-proliferation activity
EP3168210A1 (en) 2015-11-13 2017-05-17 Sandoz Ag Crystalline forms of entinostat
WO2017081278A1 (en) 2015-11-13 2017-05-18 Sandoz Ag Crystalline forms of entinostat

Similar Documents

Publication Publication Date Title
JP4405602B2 (en) Histone deacetylase inhibitor
ES2359564T3 (en) USEFUL BENZAMIDE DERIVATIVES AS CELLULAR DIFFERENTIATION INDUCERS.
KR100655808B1 (en) Colchinol derivatives as vascular damaging agents, their use and method for preparation thereof
US5589490A (en) Benzoic acid substituted derivatives having cardiovascular activity
JP2006526590A (en) Matrix metalloproteinase inhibitor
Wen et al. Identification of N-(6-mercaptohexyl)-3-(4-pyridyl)-1H-pyrazole-5-carboxamide and its disulfide prodrug as potent histone deacetylase inhibitors with in vitro and in vivo anti-tumor efficacy
JP2011500783A (en) Histone deacetylase inhibitor
JP2000505424A (en) Nitrated and nitrosylated α-adrenergic receptor antagonist compounds, compositions and uses thereof
Liu et al. Synthesis and structure-activity relationship study of water-soluble carbazole sulfonamide derivatives as new anticancer agents
KR20120006027A (en) Plasminogen activator inhibitor-1 inhibitor
WO2011146855A1 (en) Selective hdac inhibitors
WO2009076234A2 (en) Synthesis methods of histone deacetylase inhibitors (hdacis)
AU2008309269B2 (en) Novel histone deacetylase inhibitors
TW201443001A (en) Bicyclic analgesic compounds
JP2018501268A (en) Tizoxanide phosphate and alkane sulfonate and pharmaceutical use thereof
WO2009076206A1 (en) Synthesis methods of histone deacetylase inhibitors (hdacis)
JP7423655B2 (en) Quinolyl-containing compounds, pharmaceutical compositions and uses thereof
PL175707B1 (en) Novel 1-aryloxy-3-alkylamine 2-propanol esters and pharmaceutical agent
CN109293660B (en) rutaecarpine-NO donor conjugate and application thereof
CN114539237B (en) IDO inhibitor, preparation method, pharmaceutical composition and application
JP2007506664A (en) Inhibitors of matrix metalloproteinases
CN111349077B (en) Pyridazine derivative and preparation method and medical application thereof
WO2015027959A1 (en) Cyclic peptide compound, and preparation method, pharmaceutical composition and use thereof
WO2018082567A1 (en) Highly-efficient ido/tdo dual inhibitor in nitrogen-containing heterocyclic helical structure
EP1156999B1 (en) Method for the preparation of a chiral-beta-amino ester

Legal Events

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

Ref document number: 08860072

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08860072

Country of ref document: EP

Kind code of ref document: A1