WO2014122222A1 - Hydroxamate derivatives bearing amide-lactams as potent hdac inhibitors and their uses as medicaments - Google Patents

Hydroxamate derivatives bearing amide-lactams as potent hdac inhibitors and their uses as medicaments Download PDF

Info

Publication number
WO2014122222A1
WO2014122222A1 PCT/EP2014/052351 EP2014052351W WO2014122222A1 WO 2014122222 A1 WO2014122222 A1 WO 2014122222A1 EP 2014052351 W EP2014052351 W EP 2014052351W WO 2014122222 A1 WO2014122222 A1 WO 2014122222A1
Authority
WO
WIPO (PCT)
Prior art keywords
oxo
amino
carbonyl
octanoyl
hydroxyamino
Prior art date
Application number
PCT/EP2014/052351
Other languages
French (fr)
Inventor
Giuseppe Giannini
Gianfranco Battistuzzi
Davide VIGNOLA
Loredana Vesci
Maurizio Taddei
Original Assignee
Sigma-Tau Industrie Farmaceutiche Riunite S.P.A.
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 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. filed Critical Sigma-Tau Industrie Farmaceutiche Riunite S.P.A.
Publication of WO2014122222A1 publication Critical patent/WO2014122222A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D205/00Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom
    • C07D205/02Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom not condensed with other rings
    • C07D205/06Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D205/08Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with one oxygen atom directly attached in position 2, e.g. beta-lactams
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • C07D207/2732-Pyrrolidones with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to other ring carbon atoms
    • C07D207/277Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/18Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • C07D207/2732-Pyrrolidones with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to other ring carbon atoms
    • C07D207/277Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D207/282-Pyrrolidone-5- carboxylic acids; Functional derivatives thereof, e.g. esters, nitriles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/68Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member
    • C07D211/72Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D211/78Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen

Definitions

  • the present invention relates to novel hydroxamate compounds and their use as medicaments.
  • the invention includes the use of such compounds and of the pharmaceutical composition containing such compounds in medicine, in relation to cancer diseases, inflammatory diseases, neuronal diseases, parasite infections (e.g., Plasmodium and/or Trypanosoma infection), as well as other diseases where an in- hibition of HDAC is responsive.
  • HDACs histone deacetylases
  • Histone deacetylases are a family of enzymes found in numerous organisms among which bacteria, fungi, plants, and animals. Such enzymes catalyze the removal of acetyl groups from ⁇ -N-acetylated lysine residues of various protein substrates including histones, transcription factors, cc-tubulin, and nuclear importers.
  • HDAC isoforms Up to date eighteen HDAC isoforms have been characterized. They are classified in four different families with regard to their DNA sequence similarity and their biological role within the cells.
  • HDAC1, HDAC2, HDAC8 and HDAC3 are members of class-I.
  • the first three isoforms are primarily found in the nucleus; meanwhile HDAC3 is also found in the cytoplasm or membrane-associated.
  • HDAC4, HDAC5, HDAC6, HDAC7, HDAC9 and HDACIO form class-II.
  • This class has been further divided in two sub-classes, class Ila (HDAC4, 5, 7 and 9) and class lib (HDAC6 and 10).
  • Class-II enzymes are expressed in a limited number of cell types and either shuttle between the nucleus and cytoplasm (i.e., class-IIa), or are mainly cytoplasmic (i.e., class-lib) (Yang X.J., et al., Mol. Cell. Biol., 2005, 25, 2873).
  • Class-IV comprises only one member (HDACll), meanwhile class-Ill, also called sirtuins, is composed of NAD + dependent enzymes.
  • HDACi HDAC inhibitors
  • HDAC inhibition was also shown to lead to the reduction of inflammation in models of autoimmune and inflammatory diseases (Leoni F., et al., Proc. Natl. Acad. Sci., 2002, 99, 2995).
  • HDAC inhibitors Use of HDAC inhibitors has also been reported in HIV patients (Archin N. M., Nature, 2012, 487, 482-486).
  • HDACi One of the first compounds to have been documented as HDACi was the well-known anti- epileptic valproic acid, which inhibits all isoforms of classes I, II and IV. Once recognized the important role of this family of enzymes in the development of cancer, many efforts directed to find potent HDACi were undertaken by numerous academic groups as well as by pharmaceutical companies.
  • Vorinostat originally known as SAHA (suberoylanilide hydroxamic acid), was the first-iZV-class small molecule hydroxamate derivative HDACi to have been approved by the FDA in 2006 to treat a rare cancer, cutaneous T-cell lymphoma in patients who have received at least one prior systemic therapy (Grant S., et al., Nature Rev.
  • SAHA suberoylanilide hydroxamic acid
  • SAHA is a potent HDACi inhibiting classes I and II as the vast majority of HDACi currently in clinical trials (Paris M., et al., J. Med. Chem., 2008, 51, 1505).
  • short chain fatty acids e.g., sodium butyrate, phenylbutyrate, pivanex (pivalo- yloxymethyl butyrate, AN-9), and valproic acid
  • short chain fatty acids e.g., sodium butyrate, phenylbutyrate, pivanex (pivalo- yloxymethyl butyrate, AN-9), and valproic acid
  • hydroxamates e.g., SAHA, belinostat (PXD101), panobinostat (LBH589), dacino- stat (LAQ-824), and tricho statin;
  • cyclic derivatives e.g., romidepsin or FK-228 which has been approved by FDA in 2009 for the treatment of cutaneous T-cell lymphoma (CTLC) and in 2011 for the treatment of peripheral T-cell lymphoma (PTCL);
  • benzamide e.g., entinostat (MS-275), mocetinostat (MGCD-0103) and acetyldi- naline (CI-994).
  • HDAC histone deacetylase inhibitor
  • standard chemothera-plastic agents e.g., docetaxel and vorinostat
  • a phase II clinical trial involving a combination of three drugs is also ongoing in high-risk myelodysplastic syndrome patients affected by acute myeloid leukemia (NCT00656617).
  • HDAC has been hypothesized as a potential target for the treatment of parasite infections (e.g., Plasmodium and/or Trypanosoma infection) about thirteen years ago.
  • HDAC2 is up- regulated in gastric cancer (Song J., et al., APMIS, 2005, 113, 264)
  • HDAC3 is up- regulated in lung cancer (Bartling B., et al., Lung Cancer, 2005, 49, 145) and there is elevated expression of HDAC6 in oral squamous cell carcinoma (Sakuma T., et al, J. Oncol, 2006, 29, 117).
  • HDAC histone deacetylase
  • the bidendate hydroxamic acid moiety is recognized to be one of the best zinc binding-group, and a multitude of HDAC inhibitors bearing such moiety has been developed (Sampath-Kumar A., et al, Bioorg. Med. Chem. Lett., 2005, 15, 8, 1969). Over the past decade, a lot of efforts have been devoted to the identification of new HDACi that could demonstrate high binding affinity toward the biological target as well as potent cellular activity, demonstrating if needs be that there is a still great need to provide new HDAC inhibitors presenting low nanomolar binding affinity toward the HDAC proteins as well as potent cellular activity. DESCRIPTION OF THE INVENTION
  • the invention provides compounds of Formula (I) or a salt, hydrate or solvate thereof, in the preparation of a com osition for inhibition of HDAC activity:
  • R 1 is H, (Ci-C 6 )-alkyl or aryl
  • R 2 is phenyl optionally substituted with halogen, benzyloxy, (Ci-C3)-alkyl or CF3; (C3-C6)-cycloalkyl; aryl-(Ci-C6)-alkyl wherein the aryl is optionally substituted with benzyloxy, (Ci-C3)-alkyl or CF3;
  • -A-E- is -(CO)-(NH)- or -(NH)-(CO)-;
  • n is an integer comprised between 0 to 2;
  • R 3 is H, or -(CO)-R 4 ;
  • R 4 is (Ci-C 6 )-alkyl, (Ci-C 6 )-cycloalkyl, aryl-(Ci-C 6 )-alkyl, aryl, -CH(R ⁇ 9(R 6 ), heteroar- yl, or -NH-(Ci-C 6 )-alkyl;
  • R 5 is H, or (Ci-C 6 )-alkyl
  • R 6 is H, (Ci-C 6 )-alkyl, aryl, or -NH(R 7 );
  • R 8 is benzyl, or i-Bu
  • the symbol means that the carbon atom bearing said symbol can adopt a R or S configuration
  • An embodiment of this invention is that of compounds of Formula (I), for use as medicaments.
  • said medicament is used for treating a subject affected by cancer diseases, inflammatory diseases, neuronal diseases, viral and parasite infections (e.g., Plasmodium and/or Trypanosoma infection).
  • the Trypanosoma infection is due to Trypanosoma Brucei rhodesiense and/or Trypanosoma cruzi
  • the invention furthermore provides a process for the preparation of compounds of Formula (I), involving conventional synthetic methods which are described underneath.
  • R 2 is as above described for compounds of Formula I,
  • R 1 is as above described for compounds of Formula I, in a polar aprotic solvent in the presence of a coupling agent well-known to those skilled in the art of peptidic coupling.
  • compounds of general Formula (I), wherein R 3 is -(CO)-R 4 , with R 4 (Ci-C 6 )-alkyl, (Ci-C 6 )-cycloalkyl, aryl-(Ci-C 6 )-alkyl, aryl, heteroaryl, or -CH(R 5 )(R 6 ) can be obtained by reacting compounds of Formula (I) wherein R 3 is H, all other substituents being as above defined, with an anhydride of formula R 4 -(CO)-0-(CO)- R 4 ; a polar aprotic solvent in the presence of an organic base.
  • compounds of general Formula (I), wherein R 3 is -(CO)-R 4 , with R 4 (Ci-C 6 )-alkyl, (Ci-C 6 )-cycloalkyl, aryl-(Ci-C 6 )-alkyl, aryl, heteroaryl, or -CH(R 5 )(R 6 ) can be obtained by reacting compounds of Formula (I) wherein R 3 is H, all other substituents being as above defined, with an acyl chloride of formula R 4 -(CO)Cl a polar aprotic solvent in the presence of an organic base.
  • R 2 is as above described for compounds of Formula I, and wherein PG refers to an amino protecting group, such as for example, i-butoxycarbonyl,
  • R 9 is a (Ci-C4)-alkyl, the latter being preferably chosen from the group consisting of methyl, ethyl or tert-butyl; in the presence of Grubbs second generation catalyst. Said reaction is then followed by hydrolysis of the ester moiety to furnish the corresponding carboxylic acid, the latter being the precursor of the hydroxamate of Formula I by means of standard coupling with substituted or unsubstituted hy- droxylamine.
  • any interfering reactive group can be protected and then deprotected according to well-established procedures described in organic chemistry (e.g., Greene T. W. and P.G.M. Wuts "Protective Groups in Organic Syn- thesis", J. Wiley & Sons, Inc., 3rd Ed., 1999) and well-known to those skilled in the art. All said transformations are only examples of well-established procedures described in organic chemistry (e.g., March J., “Advanced Organic Chemistry", J. Wiley & Sons, Inc., 4th Ed., 1992) and well-known to those skilled in the art.
  • (Ci-Cx)-alkyl and "(C3-C x )-cycloalkyl”, wherein x is an integer comprised between 2 and 6 with regard to the cycloalkyl, meanwhile it is comprised between 3 and 6 with regard to the cycloalkyl, alone or encompassed in a more complex structure, refer to linear or branched alkyl, having from 1 to 6 carbon atoms or cycloalkyl groups having from 3 to 6 carbon atoms.
  • aryl refers to an aromatic carbocyclic group of 6 to 14 carbon atoms hav- ing a single ring (e. g., phenyl) or multiple rings that may be attached in a pendent manner or may be fused.
  • Preferred aryl include phenyl, naphthyl, phenantrenyl, bi- phenyl and the like.
  • Said “aryl” may have 1 to 3 substituents chosen among hydrox- yl, halogen, haloalkyl, cyano, (Ci-Cx)-alkyl, benzyloxy, amino, aminoalkyl or alkyl- amino.
  • aryl-(Ci-C6)-alkyl refers to alkyl groups as defined above, having one aryl substituent as defined above.
  • Preferred aryl-(Ci-C6)-alkyl include benzyl, phenethyl, diphenyl methyl and the like.
  • heteroaryl refers to a monocyclic heteroaromatic group.
  • heteroaromatic groups include pyridyl, pyrrolyl, furyl, thienyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyrazoly.
  • Said “heteroaryl” may have from 1 to 3 substituents chosen among hydroxyl, hydroxyalkyl, alkyl, alkoxy, halo, haloalkyl, and amino.
  • alkoxy refers to the group -OR where R includes “alkyl” and “cycloalkyl” as above defined.
  • aminoalkyl refers to the group H2NR- where R is “alkylene”.
  • cancer means malignant neoplasm which invades and destroys the surrounding tissue and may form metastases and eventually can kill the host.
  • “Pharmaceutically acceptable salts” refers to salts of the below identified compounds of Formula (I), that retain the desired biological activity.
  • examples of such salts include, but are not restricted to acid addition salts formed with inorganic acids (e. g. hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like), and salts formed with organic acids such as acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, fumaric acid, maleic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalene sulfonic acid, toluene sulfonic acid, naphthalene disulfonic acid, methanesulfonic acid, and poly-galacturonic acid.
  • inorganic acids e. g. hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric
  • the derivatives (I) and their pharmaceutically acceptable salts, prepared according to the invention are useful agents for the treatment of disease states, disorders and pathological conditions mediated by HDACs; in particular for the treatment of cancer diseases, inflammatory diseases, neuronal diseases and parasite infections (e.g., Plasmodium and/or Trypanosoma infection).
  • compositions will contain at least one compound of Formula (I) as an active ingredient, in an amount such as to produce a significant therapeutic effect.
  • the compositions covered by the present invention are entirely conventional and are obtained with methods which are common practice in the pharmaceutical industry, such as, those illustrated in Remington's Pharmaceutical Science Handbook, Mack Pub. N.Y. - last edition. According to the administration route chosen, the compositions will be in solid or liquid form, suitable for oral, parenteral or topical administration.
  • the compositions according to the present invention contain, along with the active ingredient, at least one pharmaceutically acceptable vehicle or excipient. These may be particularly useful formulation coadjuvants, e.g. solubilis- ing agents, dispersing agents, suspension agents, and emulsifying agents.
  • the compounds of this invention are administered in a "therapeutically effective amount".
  • the amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, drug combination, age, body weight, response of the individual pa- tient, the severity of the patient's symptoms, and the like.
  • the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rats, guinea pigs, rabbits, dogs, or pigs. Animal models may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.
  • HED Human Equivalent Dose
  • an effective dose will be from 0.01 mg/kg to 100 mg/kg, preferably 0.05 mg/kg to 50 mg/kg.
  • the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rats, guinea pigs, rabbits, dogs, or pigs.
  • the precise effective dose for a human subject will depend upon the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. This amount can be determined by routine experimentation and is within the judgement of the clinician.
  • compositions may be administered individually to a patient or may be administered in combination with other agents, drugs or hormones.
  • the medicament may also contain a pharmaceutically acceptable carrier, for administration of the therapeutic agent.
  • a pharmaceutically acceptable carrier for administration of the therapeutic agent.
  • Such carriers include antibodies and other polypeptides, genes and other therapeutic agents such as liposomes, provided that the carrier does not induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity.
  • Suitable carriers may be large, slowly metabolised macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers and inactive virus particles.
  • Pharmaceutically acceptable carriers in therapeutic compositions may additionally contain liquids such as water, saline, glycerol and ethanol.
  • auxiliary substances such as wetting or emulsifying agents, pH buff- ering substances, and the like, may be present in such compositions.
  • Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for ingestion by the patient.
  • compositions of the invention can be administered directly to the subject.
  • the subjects to be treated can be animals; in particular, human subjects can be treated.
  • the medicament of this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal or transcutaneous applica- tions, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, in- travaginal or rectal means.
  • compositions for oral administration may take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, the compositions are presented in unit dosage forms to facilitate accurate dosing.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include refilled, pre- measured ampoules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • the compound of the invention is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being vari- ous vehicles or carriers and processing aids helpful for forming the desired dosing form.
  • Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • the compounds of the present invention are useful as medicaments due to their HDAC inhibiting properties for the treatment of disorders where such inhibition results in improving the health of patients, in particular, those suf- fering from cancer and inflammatory diseases.
  • compositions in question may, together with the compounds of Formula (I), contain further known active principles.
  • a further object of the invention is a process for the preparation of pharmaceutical compositions characterised by mixing one or more compounds of Formula (I) with suitable excipients, stabilizers and/or pharmaceutically acceptable diluents.
  • a preferred embodiment of this invention is that of compounds of Formula (I) described earlier, wherein R 3 is H.
  • Another preferred embodiment of this invention is that of compounds of Formula (I) described earlier, wherein R 3 is -(CO)-R 4 as described above.
  • a further preferred embodiment of this invention is that of compounds of Formula (I) described earlier, wherein R 3 is -(CO)-R 4 with R 4 being (Ci-C 6 )-alkyl, (Ci-Ce)- cycloalkyl, aryl-(Ci-C6)-alkyl, aryl, heteroaryl, or -CH(R 5 )(R 6 ).
  • R 3 is -(CO)-R 4 with R 4 being chosen from the group consisting of:
  • Another preferred embodiment of the present invention consists of the compounds selected from the group consisting of: N-[(l ⁇ S)-7-(hydroxyamino)-7-oxo-l- (phenylcarbamoyl)heptyl]-2-oxo-yrrolidine-3-carboxamide N-[(lS)-7- (hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-2-oxo-piperidine-3-carboxamide; N-[(l-S)-7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-2-oxo-4-phenyl- pyrrolidine-3-carboxamide (2 ⁇ S)-N-[(liS)-7-(hydroxyamino)-7-oxo-l- (phenylcarbamoyl)heptyl]-5-oxo-pyrrolidine-2-carboxamide; (2R)-N
  • Another preferred embodiment of the present invention consists of the compounds selected from the group consisting of: [[(7-S)-8-anilino-8-oxo-7-[(2-oxopyrrolidine-3- carbonyl)amino]octanoyl] amino] acetate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxopyrrolidine-
  • the cancer to be treated is a primary tumour, selected from the group comprising sarcoma, carcinoma, mel- anoma, bone tumour, neuroendocrine tumour, lymphoid leukaemia, mantle cell lymphoma, diffuse large B cell lymphoma (DBCLC), myeloid leukaemia, monocytic leukaemia, megakaryocytic leukaemia, acute promyelocytic leukaemia, Hodgkin's lymphoma, peripheral T-cell non Hodgkin's lymphoma and multiple myeloma.
  • sarcoma selected from the group comprising sarcoma, carcinoma, mel- anoma, bone tumour, neuroendocrine tumour, lymphoid leukaemia, mantle cell lymphoma, diffuse large B cell lymphoma (DBCLC), myeloid leukaemia, monocytic leukaemia, megakaryocytic leukaemia, acute promyelocytic
  • sarcoma and carcinoma consist of the group comprising: breast cancer; lung cancer, including noN-small cell lung cancer (NSCLC) and small-cell lung cancer (SCLC); gastrointestinal cancer, including oesophageal, gastric, small bowel, large bowel, rectal and colon cancer; glioma, including glioblastoma; ovarian cancer; cervical cancer; endometrial cancer; mesothelioma; renal cancer; prostate cancer; peritoneum cancer; pleura cancer; head and neck cancer; blad- der cancer; brain cancer; and cancer of the eyes.
  • NSCLC noN-small cell lung cancer
  • SCLC small-cell lung cancer
  • gastrointestinal cancer including oesophageal, gastric, small bowel, large bowel, rectal and colon cancer
  • glioma including glioblastoma
  • ovarian cancer cervical cancer
  • endometrial cancer mesothelioma
  • renal cancer prostate cancer
  • peritoneum cancer pleura
  • the neoplasm can also refer to a paediatric cancer.
  • paediatric cancers that can be treated or where the progression of the condition can be delayed according to the present invention are selected from the group consisting of: acute lym- phoblastic leukaemia, acute myeloid leukaemia, adrenocortical carcinoma, astrocytomas, bladder cancer, brain stem glioma, central nervous system atypical teratoid/rhabdoid cancer, brain cancer, central nervous system embryonal cancers, brain cancer, astrocytomas, craniopharyngioma, ependymoblastoma, ependymoma, childhood medulloblastoma, medulloepithelioma, pineal parenchymal cancers of intermediate differentiation, supratentorial primitive neuroectodermal cancers and pineoblastoma, breast cancer, bronchial cancers, carcinoid cancer, cervical cancer, chordoma, colorectal
  • DIPEA diisopropylethylamine
  • DMTMM 4-(4,6- dimethoxy-l,3,5-triaziN-2-yl)-4-methylmorpholinium chloride
  • EDC l-ethyl-3-(3-dimethylaminopropyl)-carbodiimide
  • NMM N-methyl morpholine
  • Visualization was performed under short-wavelength ultraviolet light and/or by dipping the plates in an aqueous H2SO4 solution of cerium sulfate/ammonium molybdate, potassium permanganate, or ethanolic solution of anisaldehyde, followed by charring with a heat gun.
  • TLC can be stained by exposing it to iodine vapour into a iodine development chamber.
  • Low- and high-resolution mass analyses were performed on AEI-MS 902 or MS-50 spectrometers using electrospray (ES) techniques. Nuclear magnetic resonance spectra were recorded on Gemini spectrometers (Varian) at 400 MHz and on Avance spectrometer (Bruker) at 300 MHz.
  • A BnONH 2 .HCI, DMTMM, NMM, THF;
  • B TFA, DCM, 0 ⁇ C to RT;
  • NMM (16.7 ⁇ ⁇ , 0.162 mmol) and DMTMM (26.9 mg, 0.097 mmol) were added at 0°C to a cold 4 ml THF solution of (2 ⁇ S)-2-amino-N'-benzyloxy-N-phenyl-octanediamide (30 mg, 0.081 mmol) and 2-oxopyrrolidine-3-carboxylic acid (6.9 mg, 0.051 mmol).
  • the reaction mixture was stirred for 12 h and the solvent was removed under reduced pressure. Standard purification by flash chromatography (CHCls/MeOH 9:1) afforded the title compound as a mixture of two diastereomers.
  • A m-CFs-aniline, PyBOP, DIPEA, DMF and DCM, RT; B: methyl acrylate, Grubbs 2 nd gen cat, DCM, RT; C: Pd/C, Ha 1 bar reaction flow 1 ml/min, MeOH, 25 C°; D: NaOH IN, MeOH, RT; E: O- benzylhydroxylamine hydrochloride, EDO, EtsN, DCM, RT; F: TFA, DCM, 0°C to RT; G: (2i?)-5- oxopyrrolidine-2-carboxylic acid, PyBOP, DIPEA, DMF and DCM, RT; H: Pd(OH) 2 /C, Ha, EtOH/AcOEt
  • STEP B methyl (£ ,7-S)-7-(tert-butoxycarbonylamino)-8-oxo-8-[3-(trifluoromethyl)- anilino]oct-2-enoate
  • O-benzylhydroxylamine hydrochloride (74 mg, 0.46 mmol), NEt3 (63 ⁇ , 0.66 mmol) in DCM (20 ml), EDC (89 mg, 0.46 mmol) were added at room temperature to a stirred solution of (7-S)-7-(tert-butoxycarbonylamino)-8-oxo-8-[3-(trifluoromethyl)- anilino]octanoic acid (190 mg, 0.44 mmol). The reaction mixture was stirred for 2 h and then diluted with DCM and washed with brine. The organic phase was concen- trated under reduced pressure and purified on silica gel (AcOEt/MeOH: 9/1).
  • Example 13 was synthesized following the procedure reported for example 12 using methyl (7-S)-7-amino-8-oxo-8-(phenethylamino)octanoate instead of methyl (7 ⁇ S)-7- amino-8-(cyclopentylamino)-8-oxo-octanoate in step A.
  • NMM (0.87 ml, 7.96 mmol) and DMTMM (1.30 g, 4.77 mmol) were added to a solution of 8-methoxy-8-oxooctanoic acid (500 mg, 2.65 mmol) and cyclopentanamine (0.40 ml mg, 3.98 mmol) in 20 ml of dry THF at 0°C.
  • the reaction mixture was then stirred for 12 h at RT and filtered, diluted with AcOEt.
  • the organic layer was washed with HC1 IN and the solvent was removed under reduced pressure.
  • the pure desired adduct was obtained after purification through flash chromatography (PE/AcOEt: 8/2 to 7/3).
  • HDAC profiling was performed against seven isolated HDAC human isoforms in the presence of a 50 ⁇ solution of the fluorogenic tetrapeptide RHKK(Ac) substrate (from p53 residues 379-382) or in the presence of a 50 ⁇ solution of its diacetylat- ed analogue RHK(Ac)K(Ac) for HDAC8.
  • Isolated human HDACs were obtained by standard purification, with the exception of HDAC3 which was a human recombinant protein as a complex of full length human HDAC3 with a C-terminal His-tag and human NCOR2 amino acids 395-489 with an N-terminal GST-tag co-expressed in baculovirus expression system.
  • the compounds of the present invention proved to be highly potent on all HDAC isoforms tested with inhibitory activity ranging in the low nanomolar scale (Table 1). Moreover, most of the compounds demonstrated binding affinity with the various HDAC isoforms higher than that of SAHA (i.e., comparison example 14).
  • the cytotoxic effect of the compounds of the present invention was evaluated on NCI-H460 noN-small cell lung carcinoma according to the method of Skehan et al. (Skehan P., et al, J. Natl. Cancer Inst., 1990, 82, 13, 1107), using SAHA (Vori- nostat) as reference compound.
  • Tumour cells were grown in RPMI 1640 medium containing 10% heat-inactivated foetal bovine serum and 50 ⁇ g/ml gentamycin sulphate and were seeded in 96-well tissue culture plates at approximately 10% confluence. They were allowed to attach and recover for at least 24 h. Varying concentrations of the compounds of the present invention were then added to each well in order to define their IC50 value (i.e., the concentration which inhibits 50% of cell survival).
  • the plates were incubated for 24 h at 37 °C, after which they were washed 3 times by removal of the supernatant and addition of PBS. The plates were then incubated for further 48 h at 37 °C. 200 ⁇ PBS and 50 ⁇ of cold 80% TCA were added and the plates were incubated on ice for at least 1 h. TCA was removed and the plates were washed 3 times by immersion in distilled-water. They were then dried on paper at 40°C for 5 min. 200 ⁇ of 0.4% sulphorodamine B in 1% acetic acid were added. The plates were incubated at room temperature for further 30 min.
  • the compounds of the present invention demonstrated a very good inhibition profile, very often much better than that observed for the reference compound SAHA. Moreover, all the compounds of the invention had a better IC50 than the respective parent analogues (Table 3). Table 3
  • cytotoxic effect of some compounds of the present invention was also evaluated against few strains of parasites to assess their efficacy in the treatment of infec- tions.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to novel amide compounds of Formula (I), and their use as anti-tumoral and pro-apoptotic agents. The invention includes the use of such compounds in medicine, in relation to cancer disease as well as other diseases where an inhibition of HDAC is responsive, and the pharmaceutical composition containing such compounds.

Description

TITLE
Hydroxamate derivatives bearing amide-lactams as potent HDAC inhibitors and their uses as medicaments
FIELD OF THE INVENTION
The present invention relates to novel hydroxamate compounds and their use as medicaments. The invention includes the use of such compounds and of the pharmaceutical composition containing such compounds in medicine, in relation to cancer diseases, inflammatory diseases, neuronal diseases, parasite infections (e.g., Plasmodium and/or Trypanosoma infection), as well as other diseases where an in- hibition of HDAC is responsive.
BACKGROUND OF THE INVENTION
Epigenetic regulation of gene expression is a dynamic and reversible process, that could contribute to human diseases, and is driven by epigenetic protein families that include the histone deacetylases (HDACs) (ArrowsmithC.H., et al., Nature, 2012, 11, 384). Histone deacetylases (HDACs) are a family of enzymes found in numerous organisms among which bacteria, fungi, plants, and animals. Such enzymes catalyze the removal of acetyl groups from ε-N-acetylated lysine residues of various protein substrates including histones, transcription factors, cc-tubulin, and nuclear importers.
Up to date eighteen HDAC isoforms have been characterized. They are classified in four different families with regard to their DNA sequence similarity and their biological role within the cells.
HDAC1, HDAC2, HDAC8 and HDAC3 are members of class-I. The first three isoforms are primarily found in the nucleus; meanwhile HDAC3 is also found in the cytoplasm or membrane-associated.
HDAC4, HDAC5, HDAC6, HDAC7, HDAC9 and HDACIO form class-II. This class has been further divided in two sub-classes, class Ila (HDAC4, 5, 7 and 9) and class lib (HDAC6 and 10). Class-II enzymes are expressed in a limited number of cell types and either shuttle between the nucleus and cytoplasm (i.e., class-IIa), or are mainly cytoplasmic (i.e., class-lib) (Yang X.J., et al., Mol. Cell. Biol., 2005, 25, 2873).
Class-IV comprises only one member (HDACll), meanwhile class-Ill, also called sirtuins, is composed of NAD+ dependent enzymes. The common feature of classes I, II and IV enzymes resides in their zinc dependent nature. HDAC inhibitors (HDACi) have been shown to be potent inducers of growth arrest, differentiation and apoptotic cell death of transformed cells in vitro and in vivo.
HDAC inhibition was also shown to lead to the reduction of inflammation in models of autoimmune and inflammatory diseases (Leoni F., et al., Proc. Natl. Acad. Sci., 2002, 99, 2995).
Use of HDAC inhibitors has also been reported in HIV patients (Archin N. M., Nature, 2012, 487, 482-486).
One of the first compounds to have been documented as HDACi was the well-known anti- epileptic valproic acid, which inhibits all isoforms of classes I, II and IV. Once recognized the important role of this family of enzymes in the development of cancer, many efforts directed to find potent HDACi were undertaken by numerous academic groups as well as by pharmaceutical companies.
Vorinostat, originally known as SAHA (suberoylanilide hydroxamic acid), was the first-iZV-class small molecule hydroxamate derivative HDACi to have been approved by the FDA in 2006 to treat a rare cancer, cutaneous T-cell lymphoma in patients who have received at least one prior systemic therapy (Grant S., et al., Nature Rev.
Drug Discov., 2007, 6, 21). SAHA is a potent HDACi inhibiting classes I and II as the vast majority of HDACi currently in clinical trials (Paris M., et al., J. Med. Chem., 2008, 51, 1505).
Actually, according to their structures, the various families of inhibitors can be grouped, in four main groups:
a) short chain fatty acids (e.g., sodium butyrate, phenylbutyrate, pivanex (pivalo- yloxymethyl butyrate, AN-9), and valproic acid);
b) hydroxamates (e.g., SAHA, belinostat (PXD101), panobinostat (LBH589), dacino- stat (LAQ-824), and tricho statin);
c) cyclic derivatives (e.g., romidepsin or FK-228 which has been approved by FDA in 2009 for the treatment of cutaneous T-cell lymphoma (CTLC) and in 2011 for the treatment of peripheral T-cell lymphoma (PTCL);
d) benzamide (e.g., entinostat (MS-275), mocetinostat (MGCD-0103) and acetyldi- naline (CI-994)).
Some clinical trials involving combination therapies have been conducted, to assess the efficacy of broad spectrum HDACi in combination with standard chemothera- peutic agents, (e.g., docetaxel and vorinostat), in patients with advanced and relapsed lung, bladder, or prostate cancer (clinical trial NCT00565227). A phase II clinical trial involving a combination of three drugs is also ongoing in high-risk myelodysplastic syndrome patients affected by acute myeloid leukemia (NCT00656617). HDAC has been hypothesized as a potential target for the treatment of parasite infections (e.g., Plasmodium and/or Trypanosoma infection) about thirteen years ago. If most efforts from the scientific community have been dedicated to the identification of selective HDACi, there is still a large medical need for pan inhibitors since it has been demonstrated that the various cancer diseases do not involve the same HDAC isoforms. Moreover, the scientific community is also divided with regard the assessment of specific HDAC isoforms to specific cancers (Gian- nini G., et al, Future Medicinal Chemistry, 2012, 4, 11, 1439-1460). Indeed, HDACI is up-regulated in prostate cancer (Halkidou K., et al., Prostate, 2004, 59, 177) and gastric cancer (Choi J.H., et al., Jpn. J. Cancer Res., 2001, 92, 1300), HDAC2 is up- regulated in gastric cancer (Song J., et al., APMIS, 2005, 113, 264), HDAC3 is up- regulated in lung cancer (Bartling B., et al., Lung Cancer, 2005, 49, 145) and there is elevated expression of HDAC6 in oral squamous cell carcinoma (Sakuma T., et al, J. Oncol, 2006, 29, 117).
The involvement of HDAC in further diseases such as neurodegenerative diseases (Chuang D.M., et al, Trends in Neuroscience, 2009, 32, 11, 591; Sleiman S.F., et al, Expert Opin. Investig. Drugs, 2009, 18, 5, 573), cardiac hypertrophy (Hamamori Y., et al, J. Clin. Invest., 2003, 112, 6, 824) has also been documented. A recent review details diseases for which HDAC inhibition is recognized as a new approach (Di- narello C.A., et al, Mol. Med., 2011, 17, 333).
The bidendate hydroxamic acid moiety is recognized to be one of the best zinc binding-group, and a multitude of HDAC inhibitors bearing such moiety has been developed (Sampath-Kumar A., et al, Bioorg. Med. Chem. Lett., 2005, 15, 8, 1969). Over the past decade, a lot of efforts have been devoted to the identification of new HDACi that could demonstrate high binding affinity toward the biological target as well as potent cellular activity, demonstrating if needs be that there is a still great need to provide new HDAC inhibitors presenting low nanomolar binding affinity toward the HDAC proteins as well as potent cellular activity. DESCRIPTION OF THE INVENTION
It has now been found that new hydroxamate derivatives are endowed with potent inhibitory activity against HDAC family of enzymes.
The invention provides compounds of Formula (I) or a salt, hydrate or solvate thereof, in the preparation of a com osition for inhibition of HDAC activity:
Figure imgf000005_0001
Formula I
wherein,
R1 is H, (Ci-C6)-alkyl or aryl;
R2 is phenyl optionally substituted with halogen, benzyloxy, (Ci-C3)-alkyl or CF3; (C3-C6)-cycloalkyl; aryl-(Ci-C6)-alkyl wherein the aryl is optionally substituted with benzyloxy, (Ci-C3)-alkyl or CF3;
-A-E- is -(CO)-(NH)- or -(NH)-(CO)-;
n is an integer comprised between 0 to 2;
R3 is H, or -(CO)-R4;
R4 is (Ci-C6)-alkyl, (Ci-C6)-cycloalkyl, aryl-(Ci-C6)-alkyl, aryl, -CH(R<9(R6), heteroar- yl, or -NH-(Ci-C6)-alkyl;
R5 is H, or (Ci-C6)-alkyl;
R6 is H, (Ci-C6)-alkyl, aryl, or -NH(R7);
Figure imgf000005_0002
R8 is benzyl, or i-Bu;
the symbol means that the carbon atom bearing said symbol can adopt a R or S configuration;
their tautomers, their geometrical isomers, their optically active forms such as en- antiomers, diastereomers and their racemate forms, as well as their pharmaceuti- cally acceptable salts thereof.
An embodiment of this invention is that of compounds of Formula (I), for use as medicaments. In a further embodiment, said medicament is used for treating a subject affected by cancer diseases, inflammatory diseases, neuronal diseases, viral and parasite infections (e.g., Plasmodium and/or Trypanosoma infection).
In a still further embodiment, the Trypanosoma infection is due to Trypanosoma Brucei rhodesiense and/or Trypanosoma cruzi
The invention furthermore provides a process for the preparation of compounds of Formula (I), involving conventional synthetic methods which are described underneath.
Compounds of general Formula (I) wherein R3 is H can be obtained by reacting compounds of Formula (II), Formula II
Figure imgf000006_0001
wherein R2 is as above described for compounds of Formula I,
with compounds of Formula (III) or of an organic salt of them, Formula III
Figure imgf000006_0002
wherein R1 is as above described for compounds of Formula I, in a polar aprotic solvent in the presence of a coupling agent well-known to those skilled in the art of peptidic coupling.
Compounds of general Formula (I), wherein R3 is -(CO)-R4, with R4 being -NH-(Ci- Ce)-alkyl can be obtained by reacting compounds of Formula (I) wherein R3 is H, all other substituents being as above defined, with an isocyanate of formula IV
0=C=N(Ci-C6)-alkyl Formula IV
in a polar solvent as described in Schlimme S., et al., ChemMealChem, 2011, 6, 1193. Compounds of general Formula (I), wherein R3 is -(CO)-R4, with R4 (Ci-Ce)-alkyl, (Ci-C6)-cycloalkyl, aryl-(Ci-Ce)-alkyl, aryl, heteroaryl, or -CH(R5)(R6) can be ob- tained by reacting compounds of Formula (I) wherein R3 is H, all other substituents being as above defined, with an acid of formula HO2CR4, using standard peptidic coupling conditions well known to those skilled in the art. Such conditions are described in patent application US2009/0023786.
Alternatively, compounds of general Formula (I), wherein R3 is -(CO)-R4, with R4 (Ci-C6)-alkyl, (Ci-C6)-cycloalkyl, aryl-(Ci-C6)-alkyl, aryl, heteroaryl, or -CH(R5)(R6) can be obtained by reacting compounds of Formula (I) wherein R3 is H, all other substituents being as above defined, with an anhydride of formula R4-(CO)-0-(CO)- R4; a polar aprotic solvent in the presence of an organic base.
Alternatively, compounds of general Formula (I), wherein R3 is -(CO)-R4, with R4 (Ci-C6)-alkyl, (Ci-C6)-cycloalkyl, aryl-(Ci-C6)-alkyl, aryl, heteroaryl, or -CH(R5)(R6) can be obtained by reacting compounds of Formula (I) wherein R3 is H, all other substituents being as above defined, with an acyl chloride of formula R4-(CO)Cl a polar aprotic solvent in the presence of an organic base.
Compounds of general Formula (II) can be obtained by reacting compounds of For- mula (V),
Formula V
Figure imgf000007_0001
wherein R2 is as above described for compounds of Formula I, and wherein PG refers to an amino protecting group, such as for example, i-butoxycarbonyl,
with compounds of Formula (VI),
Formula VI
Figure imgf000007_0002
wherein R9 is a (Ci-C4)-alkyl, the latter being preferably chosen from the group consisting of methyl, ethyl or tert-butyl; in the presence of Grubbs second generation catalyst. Said reaction is then followed by hydrolysis of the ester moiety to furnish the corresponding carboxylic acid, the latter being the precursor of the hydroxamate of Formula I by means of standard coupling with substituted or unsubstituted hy- droxylamine.
In all said transformations, any interfering reactive group can be protected and then deprotected according to well-established procedures described in organic chemistry (e.g., Greene T. W. and P.G.M. Wuts "Protective Groups in Organic Syn- thesis", J. Wiley & Sons, Inc., 3rd Ed., 1999) and well-known to those skilled in the art. All said transformations are only examples of well-established procedures described in organic chemistry (e.g., March J., "Advanced Organic Chemistry", J. Wiley & Sons, Inc., 4th Ed., 1992) and well-known to those skilled in the art.
The terms "(Ci-Cx)-alkyl" and "(C3-Cx)-cycloalkyl", wherein x is an integer comprised between 2 and 6 with regard to the cycloalkyl, meanwhile it is comprised between 3 and 6 with regard to the cycloalkyl, alone or encompassed in a more complex structure, refer to linear or branched alkyl, having from 1 to 6 carbon atoms or cycloalkyl groups having from 3 to 6 carbon atoms.
The term "aryl" refers to an aromatic carbocyclic group of 6 to 14 carbon atoms hav- ing a single ring (e. g., phenyl) or multiple rings that may be attached in a pendent manner or may be fused. Preferred aryl include phenyl, naphthyl, phenantrenyl, bi- phenyl and the like. Said "aryl" may have 1 to 3 substituents chosen among hydrox- yl, halogen, haloalkyl, cyano, (Ci-Cx)-alkyl, benzyloxy, amino, aminoalkyl or alkyl- amino.
The term "aryl-(Ci-C6)-alkyl" refers to alkyl groups as defined above, having one aryl substituent as defined above. Preferred aryl-(Ci-C6)-alkyl include benzyl, phenethyl, diphenyl methyl and the like.
The term "heteroaryl" refers to a monocyclic heteroaromatic group. Particular examples of heteroaromatic groups include pyridyl, pyrrolyl, furyl, thienyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyrazoly. Said "heteroaryl" may have from 1 to 3 substituents chosen among hydroxyl, hydroxyalkyl, alkyl, alkoxy, halo, haloalkyl, and amino.
The term "alkoxy" refers to the group -OR where R includes "alkyl" and "cycloalkyl" as above defined.
The term "aminoalkyl" refers to the group H2NR- where R is "alkylene".
The term cancer means malignant neoplasm which invades and destroys the surrounding tissue and may form metastases and eventually can kill the host.
"Pharmaceutically acceptable salts" refers to salts of the below identified compounds of Formula (I), that retain the desired biological activity. Examples of such salts include, but are not restricted to acid addition salts formed with inorganic acids (e. g. hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like), and salts formed with organic acids such as acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, fumaric acid, maleic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalene sulfonic acid, toluene sulfonic acid, naphthalene disulfonic acid, methanesulfonic acid, and poly-galacturonic acid.
We have found that the derivatives (I) and their pharmaceutically acceptable salts, prepared according to the invention, are useful agents for the treatment of disease states, disorders and pathological conditions mediated by HDACs; in particular for the treatment of cancer diseases, inflammatory diseases, neuronal diseases and parasite infections (e.g., Plasmodium and/or Trypanosoma infection).
The pharmaceutical compositions will contain at least one compound of Formula (I) as an active ingredient, in an amount such as to produce a significant therapeutic effect. The compositions covered by the present invention are entirely conventional and are obtained with methods which are common practice in the pharmaceutical industry, such as, those illustrated in Remington's Pharmaceutical Science Handbook, Mack Pub. N.Y. - last edition. According to the administration route chosen, the compositions will be in solid or liquid form, suitable for oral, parenteral or topical administration. The compositions according to the present invention contain, along with the active ingredient, at least one pharmaceutically acceptable vehicle or excipient. These may be particularly useful formulation coadjuvants, e.g. solubilis- ing agents, dispersing agents, suspension agents, and emulsifying agents.
Generally, the compounds of this invention are administered in a "therapeutically effective amount". The amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, drug combination, age, body weight, response of the individual pa- tient, the severity of the patient's symptoms, and the like. For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rats, guinea pigs, rabbits, dogs, or pigs. Animal models may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. In calculating the Human Equivalent Dose (HED) it is recommended to use the conversion table provided in Guidance for Industry and Reviewers document (2002, U.S. Food and Drug Administration, Rockville, Maryland, USA).
Generally, an effective dose will be from 0.01 mg/kg to 100 mg/kg, preferably 0.05 mg/kg to 50 mg/kg. For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models, usually mice, rats, guinea pigs, rabbits, dogs, or pigs. The precise effective dose for a human subject will depend upon the severity of the disease state, general health of the subject, age, weight, and gender of the subject, diet, time and frequency of administration, drug combination(s), reaction sensitivities, and tolerance/response to therapy. This amount can be determined by routine experimentation and is within the judgement of the clinician.
Compositions may be administered individually to a patient or may be administered in combination with other agents, drugs or hormones.
The medicament may also contain a pharmaceutically acceptable carrier, for administration of the therapeutic agent. Such carriers include antibodies and other polypeptides, genes and other therapeutic agents such as liposomes, provided that the carrier does not induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity. Suitable carriers may be large, slowly metabolised macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers and inactive virus particles.
A thorough discussion of pharmaceutically acceptable carriers is available in Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J. 1991).
Pharmaceutically acceptable carriers in therapeutic compositions may additionally contain liquids such as water, saline, glycerol and ethanol.
Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buff- ering substances, and the like, may be present in such compositions. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for ingestion by the patient.
Once formulated, the compositions of the invention can be administered directly to the subject. The subjects to be treated can be animals; in particular, human subjects can be treated.
The medicament of this invention may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, intraventricular, transdermal or transcutaneous applica- tions, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, in- travaginal or rectal means.
The compositions for oral administration may take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, the compositions are presented in unit dosage forms to facilitate accurate dosing. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include refilled, pre- measured ampoules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions. In such compositions, the compound of the invention is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being vari- ous vehicles or carriers and processing aids helpful for forming the desired dosing form. Dosage treatment may be a single dose schedule or a multiple dose schedule. As above disclosed, the compounds of the present invention are useful as medicaments due to their HDAC inhibiting properties for the treatment of disorders where such inhibition results in improving the health of patients, in particular, those suf- fering from cancer and inflammatory diseases.
The compositions in question may, together with the compounds of Formula (I), contain further known active principles.
A further object of the invention is a process for the preparation of pharmaceutical compositions characterised by mixing one or more compounds of Formula (I) with suitable excipients, stabilizers and/or pharmaceutically acceptable diluents.
A preferred embodiment of this invention is that of compounds of Formula (I) described earlier, wherein R3 is H.
Another preferred embodiment of this invention is that of compounds of Formula (I) described earlier, wherein R3 is -(CO)-R4 as described above.
A further preferred embodiment of this invention is that of compounds of Formula (I) described earlier, wherein R3 is -(CO)-R4 with R4 being (Ci-C6)-alkyl, (Ci-Ce)- cycloalkyl, aryl-(Ci-C6)-alkyl, aryl, heteroaryl, or -CH(R5)(R6).
An even more preferred embodiment of this invention is that of compounds of Formula (I) described earlier, wherein R3 is -(CO)-R4 with R4 being chosen from the group consisting of:
Figure imgf000012_0001
Another preferred embodiment of the present invention consists of the compounds selected from the group consisting of: N-[(l<S)-7-(hydroxyamino)-7-oxo-l- (phenylcarbamoyl)heptyl]-2-oxo-yrrolidine-3-carboxamide N-[(lS)-7- (hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-2-oxo-piperidine-3-carboxamide; N-[(l-S)-7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-2-oxo-4-phenyl- pyrrolidine-3-carboxamide (2<S)-N-[(liS)-7-(hydroxyamino)-7-oxo-l- (phenylcarbamoyl)heptyl]-5-oxo-pyrrolidine-2-carboxamide; (2R)-N-[(lS)-7- (hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-5-oxo-pyrrolidine-2-carboxamide; (2<S)-N-[(liS)-7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-6-oxo-piperidine-
2- carboxamide; (2R)-8-(hydroxyamino)-8-oxo-2-[2-oxo-2-[(2R)-5-oxopyrrolidiN-2- yl] ethyl] -N- [3-(trifluoromethyl)phenyl] octanamide; (2R)-N- [(IS)- 7-(hy droxyamino)- 7-oxo-l-(p-tolylcarbamoyl)heptyl]-5-oxo-pyrrolidine-2-carboxamide; (2R)-N-[(l<S)-7- (hydroxyamino)-l-(m-tolylcarbamoyl)-7-oxo-heptyl]-5-oxo-pyrrolidine-2- carboxamide; (2R)-N-[(l<S)-7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-5- oxo-pyrrolidine-2-carboxamide; (2<S)-N-[(liS)-7-(hydroxyamino)-7-oxo-l- (phenylcarbamoyl)heptyl]-4-oxo-azetidine-2-carboxamide; (2<S)-N-[(liS)-l- (cyclopentylcarbamoyl)-7-(hydroxyamino)-7-oxo-heptyl]-6-oxo-piperidine-2- carboxamide; and (2<S)-N-[(liS)-7-(hydroxyamino)-7-oxo-l-(phenethylcarbamoyl) heptyl]-6-oxo-piperidine-2-carboxamide.
Another preferred embodiment of the present invention consists of the compounds selected from the group consisting of: [[(7-S)-8-anilino-8-oxo-7-[(2-oxopyrrolidine-3- carbonyl)amino]octanoyl] amino] acetate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxopyrrolidine-
3- carbonyl)amino]octanoyl]amino] 2-methylpropanoate; [[(7S)-8-anilino-8-oxo-7- [(2- oxopyrrolidine-3-carbonyl)amino]octanoyl]amino] propanoate; [[(7-S)-8-anilino-8-oxo-
7- [(2-oxopyrrolidine-3-carbonyl)amino]octanoyl]amino] 3-methylbutanoate; [[(7-S)-8- anilino-8-oxo-7- [(2-oxopyrrolidine-3-carbonyl)amino]octanoyl]amino] benzoate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxopyrrolidine-3-carbonyl)amino]octanoyl]amino] 2- phenylacetate; [[(7<S)-8-anilino-8-oxo-7- [(2-oxopyrrolidine-3- carbonyl)amino]octanoyl] amino] pyridine-2-carboxylate; [[(7<S)-8-anilino-8-oxo-7-[(2- oxopyrrolidine-3-carbonyl)amino]octanoyl]amino] N-ethylcarbamate; [[(7<S)-8- anilino-8-oxo-7-[(2-oxopyrrolidine-3-carbonyl)amino]octanoyl]amino] N- isopropylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxopyrrolidine-3- carbonyl)amino]octanoyl] amino] N-benzylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[(2- oxopyrrolidine-3-carbonyl)amino]octanoyl]amino] 2-(tert- butoxycarbonylamino)propanoate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxopyrrolidine-3- carbonyl)amino]octanoyl] amino] 2-(benzyloxycarbonylamino)propanoate; [[(7<S)-8- anilino-8-oxo-7-[(2-oxopiperidine-3-carbonyl)amino]octanoyl]amino] acetate; [[(7<S)- 8-anilino-8-oxo-7-[(2-oxopiperidine-3-carbonyl)amino]octanoyl]amino] 2- methylpropanoate; [[(7<S)-8-anilino-8-oxo-7- [(2-oxopiperidine-3- carbonyl)amino]octanoyl] amino] propanoate; [[(7<S)-8-anilino-8-oxo-7-[(2- oxopiperidine-3-carbonyl)amino]octanoyl] amino] 3-methylbutanoate; [[(7<S)-8- anilino-8-oxo-7-[(2-oxopiperidine-3-carbonyl)amino]octanoyl]amino] benzoate; [[(7<S)- 8-anilino-8-oxo-7-[(2-oxopiperidine-3-carbonyl)amino]octanoyl]amino] 2- phenylacetate; [[(7S)-8-anilino-8-oxo-7-[(2-oxopiperidine-3- carbonyl)amino]octanoyl] amino] pyridine-2-carboxylate; [[(7<S)-8-anilino-8-oxo-7-[(2- oxopiperidine-3-carbonyl)amino]octanoyl] amino] N-ethylcarbamate; [[(7-S)-8-anilino- 8-0X0-7- [(2-oxopiperidine-3-carbonyl)amino]octanoyl]amino] N-isopropylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxopiperidine-3-carbonyl)amino]octanoyl]amino] N- benzylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxopiperidine-3- carbonyl)amino]octanoyl] amino] 2-(tert-butoxycarbonylamino)propanoate; [[(7<S)-8- anilino-8-oxo-7-[(2-oxopiperidine-3-carbonyl)amino]octanoyl] amino] 2- (benzyloxycarbonylamino)propanoate; [[(7<S)-8-anilino-8-oxo-7- [(2-oxo-4-phenyl- pyrrolidine-3-carbonyl)amino]octanoyl]amino] acetate; [[(7<S)-8-anilino-8-oxo-7- [(2- oxo-4-phenyl-pyrrolidine-3-carbonyl)amino]octanoyl] amino] 2-methylpropanoate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxo-4-phenyl-pyrrolidine-3- carbonyl)amino]octanoyl] amino] propanoate; [[(7S)-8-anilino-8-oxo-7-[(2-oxo-4- phenyl-pyrrolidine-3-carbonyl)amino]octanoyl]amino] 3-methylbutanoate; [[(7<S)-8- anilino-8-oxo-7-[(2-oxo-4-phenyl-pyrrolidine-3-carbonyl)amino]octanoyl] amino] benzoate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxo-4-phenyl-pyrrolidine-3- carbonyl)amino]octanoyl] amino] 2-phenylacetate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxo-4- phenyl-pyrrolidine-3-carbonyl)amino]octanoyl]amino] pyridine-2-carboxylate; [[(7<S)- 8-anilino-8-oxo-7-[(2-oxo-4^henyl^yrrolidine-3-carbonyl)amino]octanoyl]amino] N- ethylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxo-4-phenyl-pyrrolidine-3- carbonyl)amino] octanoyl] amino] N-isopropylcarbamate; [[(7<S)-8-anilino-8-oxo-7-[(2- oxo-4-phenyl-pyrrolidine-3-carbonyl)amino] octanoyl] amino] N-benzylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[(2-oxo-4-phenyl-pyrrolidine-3- carbonyl)amino] octanoyl] amino] 2-(tert-butoxycarbonylamino)propanoate; [[(7<S)-8- anilino-8-oxo-7-[(2-oxo-4-phenyl-pyrrolidine-3-carbonyl)amino]octanoyl] amino] 2- (benzyloxycarbonylamino)propanoate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-5- oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] acetate; [[(7<S)-8-anilino-8-oxo-7- [ [(2 S)- 5 - oxopyrroli dine- 2 -carbonyl] amino] octanoyl] amino] 2 -methylpropanoate ; [[(7-S)-8-anilino-8-oxo-7-[[(2iS)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] propanoate; [[(7<S)-8-anilino-8-oxo-7- [[(2<S)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] 3-methylbutanoate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)- 5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] benzoate; [[(7-S)-8-anilino-8-oxo-
7- [[(2<S)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2-phenylacetate; [[(7<S)-
8- anilino-8-oxo- 7- [[(2<S)-5-oxopyrrolidine-2-carbonyl]amino] octanoyl] amino] pyri- dine-2-carboxylate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] N-ethylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-5- oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] N-isopropylcarbamate; [[(7-S)-8- anilino-8-oxo-7-[[(2<S)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] N- benzylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] 2-(tert-butoxycarbonylamino)propanoate; [[(7<S)-8- anilino-8-oxo-7-[[(2<S)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2- (benzyloxycarbonylamino)propanoate; [[(7<S)-8-anilino-8-oxo-7- [[(2R)-5- oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] acetate; [[(7-S)-8-anilino-8-oxo-7- [[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2-methylpropanoate; [[(7-S)-8-anilino-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] propanoate; [[(7<S)-8-anilino-8-oxo-7- [[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] 3-methylbutanoate; [[(7-S)-8-anilino-8-oxo-7-[[(2R)- 5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] benzoate; [[(7-S)-8-anilino-8-oxo-
7- [[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2-phenylacetate; [[(7-S)-
8- anilino-8-oxo- 7- [[(2R)-5-oxopyrrolidine-2-carbonyl]amino] octanoyl] amino] pyri- dine-2-carboxylate; [[(7-S)-8-anilino-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] N-ethylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[[(2R)-5- oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] N-isopropylcarbamate; [[(7-S)-8- anilino-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] N- benzylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] 2-(tert-butoxycarbonylamino)propanoate; [[(7<S)-8- anilino-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2- (benzyloxycarbonylamino)propanoate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-6- oxopiperidine-2-carbonyl]amino]octanoyl]amino] acetate; [[(7<S)-8-anilino-8-oxo-7- [[(2<S)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] 2-methylpropanoate; [[(7-¾-8-anilino-8-oxo-7-[[(2iS)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] propanoate; [[(7<S)-8-anilino-8-oxo-7- [[(2<S)-6-oxopiperidine-2- carbonyl] amino] octanoyl] amino] 3-methylbutanoate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-
6- oxopiperidine-2-carbonyl]amino]octanoyl]amino] benzoate; [[(7<S)-8-anilino-8-oxo- 7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] 2-phenylacetate; [[(7-S)-
8-anilino-8-oxo-7-[[(2S)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] pyridine- 2-carboxylate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-6-oxopiperidine-2- carbonyl] amino] octanoyl] amino] N-ethylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-6- oxopiperidine-2-carbonyl]amino]octanoyl]amino] N-isopropylcarbamate; [[(7<S)-8- anilino-8-oxo-7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] N- benzylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-6-oxopiperidine-2- carbonyl] amino] octanoyl] amino] 2-(tert-butoxycarbonylamino)propanoate; [[(7<S)-8- anilino-8-oxo-7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] 2- (benzyloxycarbonylamino)propanoate; [[(7S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl]amino]-8-[3-(trifluoromethyl)anilino]octanoyl]amino] acetate; [[(7<S)-8-oxo-
7- [[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8-[3-
(trifluoromethyl)anilino] octanoyl] amino] 2-methylpropanoate; [[(7<S)-8-oxo-7-[[(2R)- 5-oxopyrrolidine-2-carbonyl]amino]-8-[3-(trifluoromethyl)anilino]octanoyl]amino] propanoate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8-[3- (trifluoromethyl)anilino] octanoyl] amino] 3-methylbutanoate; [[(7<S)-8-oxo-7-[[(2R)-5- oxopyrrolidine-2-carbonyl]amino]-8-[3-(trifluoromethyl)anilino]octanoyl]amino] benzoate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8-[3- (trifluoromethyl)anilino] octanoyl] amino] 2-phenylacetate; [[(7<S)-8-oxo-7-[[(2R)-5- oxopyrrolidine-2-carbonyl]amino]-8-[3-(trifluoromethyl)anilino]octanoyl]amim pyr- idine-2-carboxylate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8-[3- (trifluoromethyl)anilino] octanoyl] amino] N-ethylcarbamate; [[(7<S)-8-oxo-7-[[(2R)-5- oxopyrrolidine-2-carbonyl]amino]-8-[3-(trifluoromethyl)anilino]octanoyl]amino] N- isopropylcarbamate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8-[3- (trifluoromethyl)anilino] octanoyl] amino] N-benzylcarbamate; [[(7<S)-8-oxo-7-[[(2R)-5- oxopyrrolidine-2-carbonyl]amino]-8-[3-(trifluoromethyl)anilino]octanoyl]amino] 2- (tert-butoxycarbonylamino)propanoate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl]amino]-8-[3-(trifluoromethyl)anilino]octanoyl]amino] 2- (benzyloxycarbonylamino)propanoate; [[(7-S)-8-(4-methylanilino)-8-oxo-7- [[(2R)-5- oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] acetate; [[(7-S)-8-(4- methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2- methylpropanoate; [[(7-S)-8-(4-methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] propanoate; [[(7-S)-8-(4-methylanilino)-8-oxo-7- [ [(2R)- 5-oxopyrrolidine-2-carbonyl] amino] octanoyl] amino] 3-methylbutanoate; [[(7S)-8-(4-methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] benzoate; [[(7-S)-8-(4-methylanilino)-8-oxo-7-[[(2R)- 5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2-phenylacetate; [[(7<S)-8-(4- methylanilino)- 8-oxo- 7- [ [(2R)- 5-oxopyrrolidine-2-carbonyl] amino] octanoyl] amino] pyridine-2-carboxylate; [[(7-S)-8-(4-methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] N-ethylcarbamate; [[(7-S)-8-(4-methylanilino)-8- oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] N- isopropylcarbamate; [[(7-S)-8-(4-methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] N-benzylcarbamate; [[(7-S)-8-(4-methylanilino)-8- oxo- 7- [ [(2R)- 5-oxopyrrolidine-2-carbonyl] amino] octanoyl] amino] 2- (tert- butoxycarbonylamino)propanoate; [[(7-S)-8-(4-methylanilino)-8-oxo-7-[[(2R)-5- oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2- (benzyloxycarbonylamino)propanoate; [[(7-S)-8-(3-methylanilino)-8-oxo-7-[[(2R)-5- oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] acetate; [[(7<S)-8-(3- methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2- methylpropanoate; [[(7-S)-8-(3-methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] propanoate; [[(7-S)-8-(3-methylanilino)-8-oxo-7- [[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 3-methylbutanoate; [[(7S)-8-(3-methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] benzoate; [[(7-S)-8-(3-methylanilino)-8-oxo-7-[[(2R)- 5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2-phenylacetate; [[(7<S)-8-(3- methylanilino)- 8-oxo- 7- [ [(2R)- 5-oxopyrrolidine-2-carbonyl] amino] octanoyl] amino] pyridine-2-carboxylate; [[(7-S)-8-(3-methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] N-ethylcarbamate; [[(7-S)-8-(3-methylanilino)-8- oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] N- isopropylcarbamate; [[(7-S)-8-(3-methylanilino)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl] amino] octanoyl] amino] N-benzylcarbamate; [[(7S)-8-(3-methylanilino)-8- oxo- 7- [ [(2R)- 5-oxopyrrolidine-2-carbonyl] amino] octanoyl] amino] 2- (tert- butoxycarbonylamino)propanoate; [[(7-S)-8-(3-methylanilino)-8-oxo-7-[[(2R)-5- oxopyrrolidine-2-carbonyl]amino]octanoyl]amino] 2- (benzyloxycarbonylamino)propanoate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl]amino]-8-(phenethylamino)octanoyl]amino] acetate; [[(7<S)-8-oxo-7-[[(2R)-5- oxopyrrolidine-2-carbonyl] amino] -8-(phenethylamino)octanoyl] amino] 2- methylpropanoate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8- (phenethylamino)octanoyl] amino] propanoate; [[(7<S)-8-oxo-7- [[(2R)-5- oxopyrrolidine-2-carbonyl] amino] -8-(phenethylamino)octanoyl] amino] 3- methylbutanoate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8- (phenethylamino)octanoyl] amino] benzoate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl]amino]-8-(phenethylamino)octanoyl]amino] 2-phenylacetate; [[(7-S)-8-oxo- 7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8-(phenethylamino)octanoyl]amino] pyridine-2-carboxylate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8- (phenethylamino)octanoyl] amino] N-ethylcarbamate; [[(7<S)-8-oxo-7- [[(2R)-5- oxopyrrolidine-2-carbonyl] amino] -8-(phenethylamino)octanoyl] amino] N- isopropylcarbamate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2-carbonyl]amino]-8- (phenethylamino)octanoyl] amino] N-benzylcarbamate; [[(7<S)-8-oxo-7- [[(2R)-5- oxopyrrolidine-2-carbonyl] amino] -8-(phenethylamino)octanoyl] amino] 2-(tert- butoxycarbonylamino)propanoate; [[(7-S)-8-oxo-7-[[(2R)-5-oxopyrrolidine-2- carbonyl]amino]-8-(phenethylamino)octanoyl]amino] 2- (benzyloxycarbonylamino)propanoate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-4-oxoazetidine- 2 -carbonyl] amino] octanoyl] amino] acetate; [[(7S)-8-anilino-8-oxo-7-[[(2S)-4- oxoazetidine-2-carbonyl]amino]octanoyl]amino] 2-methylpropanoate; [[(7<S)-8- anilino-8-oxo-7-[[(2<¾-4-oxoazetidine-2-carbonyl]amino]octanoyl]amino] propanoate; [[(7<¾-8-anilino-8-oxo-7-[[(2<¾-4-oxoazetidine 3- methylbutanoate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-4-oxoazetidine-2- carbonyl] amino] octanoyl] amino] benzoate; [[(7-S)-8-anilino-8-oxo-7-[[(2S)-4- oxoazetidine-2-carbonyl]amino]octanoyl]amino] 2-phenylacetate; [[(7-S)-8-anilino-8- oxo-7-[[(2<S)-4-oxoazetidine-2-carbonyl]amino]octanoyl]amino] pyridine-2- carboxylate; [[(7<S)-8-anilino-8-oxo-7- [[(2<S)-4-oxoazetidine-2- carbonyl] amino] octanoyl] amino] N-ethylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-4- oxoazetidine-2-carbonyl]amino]octanoyl]amino] N-isopropylcarbamate; [[(7<S)-8- anilino-8-oxo-7-[[(2<S)-4-oxoazetidine-2-carbonyl]amino]octanoyl]amino] N- benzylcarbamate; [[(7-S)-8-anilino-8-oxo-7-[[(2<S)-4-oxoazetidine-2- carbonyl] amino] octanoyl] amino] 2- (tert-butoxy carbonylamino)propanoate; [[(7<S)-8- anilino- 8-oxo- 7- [[(2 S - 4-oxoazetidine-2-carbonyl] amino] octanoyl] amino] 2 -
(benzyloxycarbonylamino)propanoate; [[(7-S)-8-(cyclopentylamino)-8-oxo-7-[[(2<S)-6- oxopiperidine-2-carbonyl]amino]octanoyl]amino] acetate; [[(7-S)-8-
(cyclopentylamino)-8-oxo-7-[[(2iS)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] 2-methylpropanoate; [[(7-S)-8-(cyclopentylamino)-8-oxo-7-[[(2<S)-6-oxopiperidine-2- carbonyl] amino] octanoyl] amino] propanoate; [ [(7<S)-8- (cyclopentylamino)- 8-oxo- 7- [[(2<S)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] 3-methylbutanoate; [[(7<S)- 8-(cyclopentylamino)-8-oxo-7-[[(2iS)-6-oxopiperidine-2- carbonyl] amino] octanoyl] amino] benzoate; [ [(7<S)-8- (cyclopentylamino)- 8-oxo- 7- [[(2<S)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] 2-phenylacetate; [[(7-S)-8- (cyclopentylamino)-8-oxo-7-[[(2iS)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] pyridine-2-carboxylate; [[(7-S)-8-(cyclopentylamino)-8-oxo-7-[[(2<S)-6-oxopiperidine-2- carbonyl] amino] octanoyl] amino] N-ethylcarbamate; [[(7-S)-8-(cyclopentylamino)-8- oxo-7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] N- isopropylcarbamate; [[(7-S)-8-(cyclopentylamino)-8-oxo-7-[[(2<S)-6-oxopiperidine-2- carbonyl] amino] octanoyl] amino] N-benzylcarbamate; [ [(7-S)-8- (cyclopentylamino)- 8- oxo-7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]octanoyl]amino] 2-(tert- butoxycarbonylamino)propanoate; [[(7-S)-8-(cyclopentylamino)-8-oxo-7-[[(2<S)-6- oxopiperidine-2-carbonyl]amino] octanoyl] amino] 2- (benzyloxycarbonylamino)propanoate; [[(7-S)-8-oxo-7-[[(2<S)-6-oxopiperidine-2- carbonyl]amino]-8-(phenethylamino)octanoyl]amino] acetate; [[(7<S)-8-oxo-7-[[(2<S)-6- oxopiperidine-2-carbonyl]amino]-8-(phenethylamino)octanoyl]amino] 2- methylpropanoate; [[(7-S)-8-oxo-7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]-8- (phenethylamino)octanoyl]amino] propanoate; [[(7-S)-8-oxo-7-[[(2<S)-6-oxopiperidine- 2 - carbonyl] amino] - 8- (phenethylamino)octanoyl] amino] 3- methylbutanoate ; [ [(7 S - 8- oxo-7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]-8-(phenethylamino)octanoyl]amino] benzoate; [[(7-S)-8-oxo-7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]-8- (phenethylamino)octanoyl] amino] 2-phenylacetate; [[(7<S)-8-oxo-7-[[(2<S)-6- oxopiperidine-2-carbonyl]amino]-8-(phenethylamino)octanoyl]amino] pyridine-2- carboxylate; [[(7-S)-8-oxo-7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]-8- (phenethylamino)octanoyl] amino] N-ethylcarbamate; [[(7<S)-8-oxo-7-[[(2<S)-6- oxopiperidine-2-carbonyl]amino]-8-(phenethylamino)octanoyl]amino] N- isopropylcarbamate; [[(7-S)-8-oxo-7-[[(2<S)-6-oxopiperidine-2-carbonyl]amino]-8- (phenethylamino)octanoyl] amino] N-benzylcarbamate; [[(7<S)-8-oxo-7-[[(2<S)-6- oxopiperidine-2-carbonyl]amino]-8-(phenethylamino)octanoyl]amino] 2-(tert- butoxycarbonylamino)propanoate; and [[(7-S)-8-oxo-7-[[(2<S)-6-oxopiperidine-2- carbonyl] amino] - 8- (phenethylamino)octanoyl] amino] 2 - (benzyloxy carbonylamino) propanoate.
According to another embodiment of the present invention the cancer to be treated is a primary tumour, selected from the group comprising sarcoma, carcinoma, mel- anoma, bone tumour, neuroendocrine tumour, lymphoid leukaemia, mantle cell lymphoma, diffuse large B cell lymphoma (DBCLC), myeloid leukaemia, monocytic leukaemia, megakaryocytic leukaemia, acute promyelocytic leukaemia, Hodgkin's lymphoma, peripheral T-cell non Hodgkin's lymphoma and multiple myeloma.
The above mentioned sarcoma and carcinoma consist of the group comprising: breast cancer; lung cancer, including noN-small cell lung cancer (NSCLC) and small-cell lung cancer (SCLC); gastrointestinal cancer, including oesophageal, gastric, small bowel, large bowel, rectal and colon cancer; glioma, including glioblastoma; ovarian cancer; cervical cancer; endometrial cancer; mesothelioma; renal cancer; prostate cancer; peritoneum cancer; pleura cancer; head and neck cancer; blad- der cancer; brain cancer; and cancer of the eyes.
The neoplasm can also refer to a paediatric cancer. For example paediatric cancers that can be treated or where the progression of the condition can be delayed according to the present invention are selected from the group consisting of: acute lym- phoblastic leukaemia, acute myeloid leukaemia, adrenocortical carcinoma, astrocytomas, bladder cancer, brain stem glioma, central nervous system atypical teratoid/rhabdoid cancer, brain cancer, central nervous system embryonal cancers, brain cancer, astrocytomas, craniopharyngioma, ependymoblastoma, ependymoma, childhood medulloblastoma, medulloepithelioma, pineal parenchymal cancers of intermediate differentiation, supratentorial primitive neuroectodermal cancers and pineoblastoma, breast cancer, bronchial cancers, carcinoid cancer, cervical cancer, chordoma, colorectal cancer, oesophageal cancer, extra cranial germ cell cancer, gastric cancer, glioma, hepatocellular (liver) cancer, Hodgkin lymphoma, kidney cancer, laryngeal cancer, leukaemia, acute lymphoblastic/myeloid leukaemia, liver cancer, noN-Hodgkin lymphoma, medulloblastoma, mesothelioma, multiple endocrine neoplasia syndrome, nasopharyngeal cancer, oral cancer, ovarian cancer, pancreatic cancer, papillomatosis, renal cell cancer, rhabdomyosarcoma, salivary gland cancer, sarcoma, skin cancer, thymoma and thymic carcinoma, thyroid cancer and vaginal cancer.
EXAMPLES
Abbreviations:
AcOEt: ethyl acetate
ESI-MS: electrospray ionization mass spectrometry
CHC13: chloroform
DCM: dichloromethane
DIPEA: diisopropylethylamine
DMF: dimethylformamide
DMTMM: 4-(4,6- dimethoxy-l,3,5-triaziN-2-yl)-4-methylmorpholinium chloride EDC: l-ethyl-3-(3-dimethylaminopropyl)-carbodiimide
Et20: diethyl ether
EtOH: ethanol
HC1: hydrochloric acid
HPLC: high-performance liquid chromatography
MeOH: methanol
NaHC03: sodium bicarbonate
Na2C03: sodium carbonate
NaOH: sodium hydroxide NEt3: triethylamine
NMM: N-methyl morpholine
Pd/C: palladium on charcoal
Pd(OH)2/C palladium hydroxyde on charcoal
PE petroleum ether
PyBOP: (benzotriazole-l-yl-oxy-tris-(dimethylamino)-phosphonium hexafluoro- phosphate)
RT: room temperature
TFA: trifuoroacetic acid
THF: tetrahydrofuran
TLC: thin layer chromatography
General Remarks: All non-aqueous reactions were run in flame-dried glassware under a positive pressure of argon with exclusion of moisture from reagents and glassware using standard techniques for manipulating air-sensitive compounds. Anhydrous THF, toluene, Et20 and DCM were obtained by filtration through drying columns (Solvent Delivery System); other solvents were distilled under positive pressure of dry argon before use and dried by standard methods. Commercial grade reagents were used without further purification. Flash chromatography was performed on 230-400 mesh silica gel with the indicated solvent systems. TLC was per- formed on pre-coated, glass-backed silica gel plates (Merck 6OF254). Visualization was performed under short-wavelength ultraviolet light and/or by dipping the plates in an aqueous H2SO4 solution of cerium sulfate/ammonium molybdate, potassium permanganate, or ethanolic solution of anisaldehyde, followed by charring with a heat gun. Alternatively, TLC can be stained by exposing it to iodine vapour into a iodine development chamber. Low- and high-resolution mass analyses were performed on AEI-MS 902 or MS-50 spectrometers using electrospray (ES) techniques. Nuclear magnetic resonance spectra were recorded on Gemini spectrometers (Varian) at 400 MHz and on Avance spectrometer (Bruker) at 300 MHz. Mass analyses were performed on Waters ZQ2000 spectrometer using electrospray (ES) technique. LCMS analyses were performed on a LC-Waters apparatus (HPLC Waters Alliance 2695, ZQ2000 MS and PDA-UV detector 2996). EXAMPLE 1
N-r(l5^-7-(hvdroxyamino)-7-oxo-l-(phenylcarbam
carboxamide
Figure imgf000022_0001
A: BnONH2.HCI, DMTMM, NMM, THF; B: TFA, DCM, 0<C to RT;
C: (S)-4-oxo-azetidine-2-carboxylic acid, PyBOP, NEt3, DMF and DCM, RT; D: Pd/C, H2, CHCI3
Scheme 1
STEP A: (tert-butyl N-[(l<S)-7-(benzyloxyamino)-7-oxo-l-(phenylcarbamoyl)- heptyl] carbamate
DMTMM (408 mg, 1.48 mmol) and NMM (271 μΐ, 249 mg, 1.77 mmol), were added to a 4 ml solution of (<S)-methyl 7-(£er£-butoxycarbonylamino)-8-oxo-8- (phenylamino)octanoate (300 mg, 0.82 mmol) and O-benzylhydroxylamine hydro- chloride (196 mg, 1.48 mmol) in dry THF at 0°C. The reaction mixture was then stirred for 12 h at RT. The crude reaction mixture was filtered, diluted with AcOEt and the organic layer washed with HCl IN. Removal of the solvent removed under reduced pressure followed by purification by flash chromatography (PE/AcOEt: 70:30) afforded the title compound as an oil.
Yield: 65% (250 mg).
Ή NMR (400 MHz, CDCI3) δ: 1.01-1.48 (m, 15H), 1.89 (m, 3H), 2.34 (m, 1H), 4.26 (s, 1H), 4.84 (s, 2H), 5.56 (s, 1H), 7.02 (d, 1H), 7.13-7.42 (m, 7H), 7.49 (d, 2H), 8.99 (s, 1H), 9.23 (s, 1H).
STEP B: (2<S)-2-amino-N'-benzyloxy-N-phenyl-octanediamide
TFA was added (100 μΐ) were added to a 900 μΐ DCM solution of (tert-butyl N-[(1S - 7-(benzyloxyamino)-7-oxo-l-(phenylcarbamoyl)-heptyl]carbamate (250 mg, 0.31 mmol). The reaction mixture was stirred for 12 h and the solvent was removed under reduced pressure. Standard purification by flash chromatography (DCM/MeOH 9:1) afforded the title compound as oil.
Yield: 79% (90 mg). Ή NMR (400 MHz, MeOD-d4) δ: 1.21-1.64 (m, 6H), 1.84 (dq, 2H), 2.03 (t, 2H), 3.86 (t, 1H), 4.80 (s, 3H), 7.09 (t, 1H), 7.32 (dt, 7H), 7.57 (d, 2H).
STEP C: (2<¾-A^-benzyloxy-2-[(2-oxopyrrolidine-3-carbonyl)amino]-N-phenyl- octanediamide
NMM (16.7 μΐ^, 0.162 mmol) and DMTMM (26.9 mg, 0.097 mmol) were added at 0°C to a cold 4 ml THF solution of (2<S)-2-amino-N'-benzyloxy-N-phenyl-octanediamide (30 mg, 0.081 mmol) and 2-oxopyrrolidine-3-carboxylic acid (6.9 mg, 0.051 mmol). The reaction mixture was stirred for 12 h and the solvent was removed under reduced pressure. Standard purification by flash chromatography (CHCls/MeOH 9:1) afforded the title compound as a mixture of two diastereomers.
Yield: 78% (30 mg).
Ή NMR (400 MHz, MeOD-d4) δ: 1.15-1.88 (m, 8H), 2.03 (t, 2H), 2.17-2.63 (m, 2H), 3.21-3.57 (m, 4H), 4.39-4.67 (m, 2H), 7.00-7.65 (m, 10H).
STEP D: N-[(l<S)-7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-2-oxo- pyrrolidine-3-carboxamide
A purged 4 ml CHCI3 solution of (2<S)-N'-benzyloxy-2-[(2-oxopyrrolidine-3- carbonyl)amino]-N-phenyl-octanediamide (20 mg, 0.042 mmol) was stirred under 1 atm. of H2 in the presence of Pd/C 10% w/w catalyst. The reaction mixture was stirred for 12 h and the solvent was removed under reduced pressure. Standard pu- rification by flash chromatography (CHCls/MeOH 8:2) afforded the title compound as a mixture of two diastereomers.
Yield: 50% (8.2 mg).
Ή NMR (400 MHz, MeOD-d4) δ: 1.13-1.90 (m, 10 H), 2.07 (t, 3H) 2.23-2.64 (m, 2H), 3.30 - 3.61 (m, 3H), 4.45 (dt, 1H), 6.97-7.13 (m, 1H), 7.27 (q, 2H), 7.57 (m, 2H).
Examples 2 to 6 were synthesized following the procedure depicted in scheme 1 using the appropriate acid in step C.
EXAMPLE 2
N-[(l8)-7-(hvdroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyll-2-oxo-piperidine-3- carboxamide
The title compound was obtained as white solid after purification by flash chromatography (CHCls/MeOH 8:2) as a mixture of two diastereomers.
Yield: 55%. Ή NMR (400 MHz, MeOD-d4) δ: 1.14-1.55 (m, 7H), 1.54-1.81 (m, 4H), 1.81-2.21 (m, 6H), 3.41 (m, 4H), 4.42 (dt, 1H), 6.94-7.13 (m, 1H), 7.26 (q, 2H), 7.59 (m 2H).
EXAMPLE 3
N-[(lS^-7-(hvdroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyll-2-oxo-4-phenyl- pyrrolidine-3-carboxamide
The title compound was obtained as white solid after purification by flash chromatography (CHCl3/MeOH 8:2) as a mixture of two diastereomers.
Yield: 52%.
Ή NMR (400 MHz, MeOD-d4) δ: 1.12-1.76 (m, 8H), 2.03 (dt, 2H), 3.42 (dt, 1H), 3.69- 3.82 (m, 2H), 4.00-4.22 (m, 1H), 4.47 (dd, 1H), 7.01-7.13 (m, 1H), 7.14-7.37 (m, 7H), 7.47 (d, 1H), 7.63 (d, 1H).
EXAMPLE 4
(28)-N-[(l,S)-7-(hvdroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyll-5-oxo-pyrrolidine- 2-carboxamide
The title compound was obtained as white solid after purification by flash chromatography (CHCls/MeOH 8:2).
Yield: 65%.
Ή NMR (400 MHz, MeOD-d4) δ: 1.37 (dt, 5H), 1.60 (s, 2H), 1.67-1.93 (m, 2H), 2.06 (t, 3H), 2.23-2.56 (m, 2H), 4.25 (dd, 1H), 4.45 (dd, 1H), 7.07 (t, 1H), 7.27 (t, 2H), 7.52 (d, 2H).
EXAMPLE 5
(2R)-N-[(lS)-7-(hvdroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyll-5-oxo-pyrrolidine- 2-carboxamide
The title compound was obtained as white solid after purification by flash chroma- tography (CHCl3/MeOH 8:2).
Yield: 63%.
Ή NMR (400 MHz, MeOD-d4) δ: 1.37 (dt, 5H), 1.60 (s, 2H), 1.67-1.93 (m, 2H), 2.06 (t, 3H), 2.23-2.56 (m, 2H), 4.25 (dd, 1H), 4.45 (dd, 1H), 7.07 (t, 1H), 7.27 (t, 2H), 7.52 (d, 2H).
EXAMPLE 6
(28)-N-[(l,S)-7-(hvdroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyll-6-oxo-piperidine-
2-carboxamide
Yield: 57%. 1H NMR (400 MHz, MeOD-d4) δ: 1.39 (dt, 4H), 1.60 (s, 2H), 1.65-1.96 (m, 5H), 2.06 (t, 3H), 2.30 (td, 2H), 4.10 (t, 1H), 4.46 (dd, 1H), 7.07 (t, 1H), 7.27 (t, 2H), 7.51 (d, 2H).
EXAMPLE 7
(2R)-8-(hvdroxyamino)-8-oxo-2-r2-oxo-2-r(2R)-5-oxopyrrolidiN-2-yllethyll-N-r3- (trifluoromethyl)phenylloctanamide
Figure imgf000025_0001
A: m-CFs-aniline, PyBOP, DIPEA, DMF and DCM, RT; B: methyl acrylate, Grubbs 2nd gen cat, DCM, RT; C: Pd/C, Ha 1 bar reaction flow 1 ml/min, MeOH, 25 C°; D: NaOH IN, MeOH, RT; E: O- benzylhydroxylamine hydrochloride, EDO, EtsN, DCM, RT; F: TFA, DCM, 0°C to RT; G: (2i?)-5- oxopyrrolidine-2-carboxylic acid, PyBOP, DIPEA, DMF and DCM, RT; H: Pd(OH)2/C, Ha, EtOH/AcOEt
Scheme 2
STEP A: tert-butyl N-[(lS)-l-[[3-(trifluoromethyl)phenyl]carbamoyl]hex-5- enyl] carbamate
A solution of (<S)-2-tert-butoxycarbonylamino-hept-6-enoic acid (200 mg, 0.82 mmol), DIPEA (416 μΐ, 2.5 mmol) and m-CF3-aniline (145 mg, 0.90 mmol) was stirred at RT in DCM (20 ml) for 20 minutes before adding PyBOP (428 mg, 0.82 mmol) and anhydrous DMF (1 ml). The reaction mixture was then stirred for 2 hours at RT. The solvent was removed under reduced pressure and the crude reaction mixture was diluted with AcOEt, washed with 5% Na2CO3, H2O and then with 5% aqueous citric acid and finally with brine. After removal of the solvent under reduced pressure and purification on silica gel (N-hexane/AcOEt: 9/1) the desired adduct was obtained. Yield: 85%.
ESI-MS m/z 409.3 (M+Na)+; ESI-MS m/z 385.4 (M-H)- Ή NMR (300 MHz, DMSO-ds) δ: 1.33 (m, 2H), 1.38 (s, 9H), 1.61 (m, 2H), 2.03 (m, 2H), 4.05 (m, 1H), 4.97 (m, 2H), 5.77 (m, 1H), 7.11 (d, 1H), 7.40 (d, 1H), 7.55 (t, 1H), 7.80 (d, 1H), 8.09 (s, 1H), 10.31 (s, 1H).
STEP B: methyl (£ ,7-S)-7-(tert-butoxycarbonylamino)-8-oxo-8-[3-(trifluoromethyl)- anilino]oct-2-enoate
To a stirred solution of tert-butyl N-[(l<S)-l-[[3-(trifluoromethyl)phenyl] carbamoyl] - hex-5-enyl] carbamate (260 mg, 0.67 mmol) and methyl acrylate (724 μΐ, 8.04 mmol) at RT in DCM was added Grubbs 2nd generation catalyst (34 mg, 0.04 mmol). The reaction mixture was stirred overnight and then concentrated under reduced pres- sure. The resulting crude product was purified on silica gel (N-hexane / AcOEt: 9/1). Yield: 71 %.
ESI-MS m/z 467.0 (M+Na)+.
Ή NMR (300 MHz, DMSO-ds) δ: 1.22-1.72 (m, 4H), 1.37 (s, 9H), 2.21 (m, 2H), 3.63 (s, 3H), 4.07 (m, 1H), 5.86 (d, 1H), 6.88 (dt, 1H), 7.13 (d, 1H), 7.38 (d, 1H), 7.54 (t, 1H), 7.79 (d, 1H), 8.07 (s, 1H), 10.30 (s, 1H).
STEP C: methyl (7-S)-7-(tert-butoxycarbonylamino)-8-oxo-8-[3-(trifluoromethyl)- anilino] octanoate
Methyl (£ ,7-S)-7-(tert-butoxycarbonylamino)-8-oxo-8-[3-(trifluoromethyl)anilino]oct- 2-enoate (200 mg, 0.44 mmol) was dissolved in MeOH (50 ml). The solution was fluxed in a H-Cube system at 1 ml/min with a 10% Pd/C 30X4 mm cartridge and p¾ = 1 bar. The solvent was removed under reduced pressure and the product obtained was used without any further purification in the next step.
Yield: 100%.
ESI-MS m/z 469.0 (M+Na)+.
Ή NMR (300 MHz, DMSO-de) δ: 1.28 (m, 4H), 1.37 (s, 9H), 1.47 (m, 2H), 1.57 (m, 2H), 2.27 (t, 2H), 3.56 (s, 3H), 4.01 (m, 1H), 7.07 (d, 1H), 7.38 (d, 1H), 7.54 (t, 1H), 7.79 (d, 1H), 8.07 (s, 1H), 10.28 (s, 1H).
STEP D: (7-S)-7-(tert-butoxycarbonylamino)-8-oxo-8-[3-(trifluoromethyl)anilino]- octanoic acid
NaOH IN (582 μΐ, 0.58 mmol) was added to a 2 ml MeOH solution of methyl (7S)-7- (tert-butoxycarbonylamino)-8-oxo-8- [3- (trifluoromethyl)anilino] octanoate (200 mg, 0.44 mmol) at room temperature. The reaction mixture was stirred for 12 h, then acidified to pH=4 with HCl IN and extracted with AcOEt. The solvent was removed under reduced pressure and the resulting product was used without any purification in the next step.
Yield: 100%.
ESI-MS m/z 454.9 (M+Na)+.
Ή NMR (300 MHz, DMSO-ds) δ: 1.28 (m, 4H), 1.37 (s, 9H), 1.47 (m, 2H), 1.62 (m, 2H), 2.17 (t, 2H), 4.02 (m, 1H), 7.07 (d, 1H), 7.38 (d, 1H), 7.53 (t, 1H), 7.79 (d, 1H), 8.08 (s, 1H), 10.29 (s, 1H), 11.95 (bs, 1H).
STEP E: tert-butyl N-[(lS)-7-(benzyloxyamino)-7-oxo-l-[[3-(trifluoromethyl)- phenyl] carbamoyl] hep tyl] carbamate
O-benzylhydroxylamine hydrochloride (74 mg, 0.46 mmol), NEt3 (63 μΐ, 0.66 mmol) in DCM (20 ml), EDC (89 mg, 0.46 mmol) were added at room temperature to a stirred solution of (7-S)-7-(tert-butoxycarbonylamino)-8-oxo-8-[3-(trifluoromethyl)- anilino]octanoic acid (190 mg, 0.44 mmol). The reaction mixture was stirred for 2 h and then diluted with DCM and washed with brine. The organic phase was concen- trated under reduced pressure and purified on silica gel (AcOEt/MeOH: 9/1).
Yield: 68 %.
ESI-MS m/z 560.0 (M+Na)+.
Ή NMR (300 MHz, DMSO-ds) δ: 1.24 (m, 4H), 1.37 (s, 9H), 1.44 (m, 2H), 1.57 (m, 2H), 1.92 (t, 2H), 4.02 (m, 1H), 4.77 (s, 2H), 7.07 (d, 1H), 7.36 (m, 6H), 7.54 (t, 1H), 7.80 (d, 1H), 8.08 (s, 1H), 10.28 (s, 1H), 10.90 (s, 1H).
STEP F: (2<S)-2-amino-N'-benzyloxy-N-[3-(trifluoromethyl)phenyl]octanediamide TFA (240 μΐ, 3 mmol) was added slowly to a stirred solution in DCM (5 ml) of tert- butyl N- [(1 S - 7- (benzyloxyamino)- 7-oxo- 1 - [ [3- (trifluoromethyl)phenyl] carbamoyl] - heptyl] carbamate (160 mg, 0.30 mmol) at 0°C. The reaction mixture was then al- lowed to warm to RT and further stirred for 3 h. The solvent was removed under reduced pressure to afford the desired adduct as the trifluoroacetate salt which was used without any purification in the next step.
ESI-MS m/z 438.17 (M+H)+; ESI-MS m/z 460.16 (M+Na)+.
STEP G: (2R)-N'-benzyloxy-2-[2-oxo-2-[(2R)-5-oxopyrrolidiN-2-yl]ethyl]-N-[3- (trifluoromethyl)phenyl]octanediamide
A solution of (2<S)-2-amino-N'-benzyloxy-N-[3-(trifluoromethyl)phenyl]- octanediamide (148 mg, 0.34 mmol), DIPEA (284 μΐ, 1.69 mmol) and (R)- pyroglutamic acid (167 mg, 0.34 mmol) was stirred at RT in DCM (20 ml) for 20 mi- nutes before adding PyBOP (177 mg, 0.34 mmol) and anhydrous DMF (1 ml). The reaction mixture was further stirred for 2 hours at RT. The solvent was removed under reduced pressure and the crude reaction mixture was diluted with AcOEt, washed with 5% Na2C03, water and then with 5% aqueous citric acid and finally with brine. After removal of the solvent under reduced pressure and purification on silica gel (AcOEt/MeOH: 9/1) the desired adduct was obtained.
Yield: 60%.
ESI-MS m/z 571.16 (M+Na)+.
Ή NMR (300 MHz, DMSO-ds) δ: 1.23 (m, 4H), 1.47 (m, 2H), 1.67 (m, 2H), 1.77-2.33 (m, 6H), 4.10 (m, 1H), 4.38 (m, 1H), 4.76 (s, 2H), 7.35 (m, 6H), 7.54 (t, 1H), 7.79 (d, 1H), 7.81 (s, 1H), 8.09 (s, 1H), 8.29 (d, 1H), 10.45 (s, 1H), 10.92 (s, 1H).
STEP H: (2R)-8-(hydroxyamino)-8-oxo-2-[2-oxo-2-[(2R)-5-oxopyrrolidiN-2-yl]ethyl]- N- [3- (trifluoromethyl)phenyl] octanamide
(2<S)-N'-benzyloxy-2-[[(2i2)-5-oxopyrrolidine-2-carbonyl]amino]-N-[3- (trifluoromethyl)phenyl]octanediamide (30 mg, 0.05 mmol) was dissolved in MeOH
(40 ml). The solution was passed in a H-Cube system at 1 ml/min within a 20
%Pd(OH)2/C 30X4 mm cartridge and ¾ generated in full ¾ mode. The solvent was removed under reduced pressure to obtain the desired adduct.
Yield: 95%.
ESI-MS m/z 459.2 (M+H)+.
Ή NMR (300 MHz, DMSO-ds) δ: 1.25 (m, 4H), 1.48 (m, 2H), 1.70 (m, 2H), 1.77-2.33
(m, 6H), 4.11 (m, 1H), 4.40 (m, 1H), 7.42 (d, 1H), 7.56 (t, 1H), 7.82 (s, 1H), 7.79 (d,
1H), 8.10 (s, 1H), 8.25 (d, 1H), 8.63 (s, 1H), 10.31 (s, 1H), 10.43 (s, 1H).
Examples 8 to 11 were synthesized following the procedure depicted in scheme 2 us- ing the appropriate amino starting material in step A and the adequate acid in step
D.
EXAMPLE 8
(2R)-N-[(lS)-7-(hvdroxyamino)-7-oxo-l-(p-tolylcarbamoyl)heptyll-5-oxo-pyrrolidine- 2-carboxamide
Yield: 94%.
Ή NMR (300 MHz, DMSO-ds) δ: 1.28 (m, 4H), 1.47 (m, 2H), 2.11 (m, 2H), 2.13-2.25 (m, 6H), 2.25 (s, 3H), 4.11 (m, 1H), 4.42 (m, 1H), 7.11 (d, 2H), 7.48 (d, 2H), 7.82 (s, 1H), 8.16 (d, 1H), 8.64 (s, 1H), 9.97 (s, 1H), 10.31 (s, 1H). ESI-MS m/z 405.16 (M+H)+.
EXAMPLE 9
(2R)-N- \(1S)- 7-(hydroxyamino)- 1 - (m-tolylcarbamoyl)- 7-oxo-heptvH - 5-oxo- pyrrolidine-2-carboxamide
Yield: 96%.
Ή NMR (300 MHz, DMSO-ds) δ: 1.25 (m, 4H), 1.47 (m, 2H), 2.11 (m, 2H), 2.13-2.26 (m, 6H), 2.27 (s, 3H), 4.12 (m, 1H), 4.42 (m, 1H), 6.88 (d, 1H), 7.20 (t, 1H), 7.43 (m, 2H), 7.82 (s, 1H), 8.15 (d, 1H), 8.63 (s, 1H), 9.97 (s, 1H), 10.31 (s, 1H).
ESI-MS m/z 405.37 (M+H)+.
EXAMPLE 10
(2R)-N-[(lS )-7-(hvdroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyll-5-oxo-pyrrolidine-
2-carboxamide
Yield: 96%.
Ή NMR (300 MHz, DMSO-ds) δ: 1.17 (m, 4H), 1.43 (m, 2H), 1.52 (m, 2H), 1.72-1.88 (m, 1H), 1.90 (t, 2H), 1.98-2.32 (m, 3H), 2.69 (t, 2H), 3.16-3.40 (m, 2H), 4.06 (m, 1H), 4.18 (m, 1H), 7.19 (m, 3H), 7.27 (m, 2H), 7.79 (s, 1H), 7.95 (d, 1H), 8.02 (t, 1H), 8.63 (s, 1H), 10.30 (s, 1H).
ESI-MS m/z 419.2 (M+H)+; ESI-MS m/z 417.2 (M-H)-.
EXAMPLE 11
(2S )-N-[(lSr)-7-(hvdroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyll-4-oxo-azetidine-2- carboxamide
Yield: 97%.
ESI-MS m/z 376.9 (M+H)+; ESI-MS m/z 375.0 (M-H)-.
Ή NMR (300 MHz, DMSO-ds) δ: 1.23 (m, 4H), 1.47 (m, 2H), 1.64 (m, 2H), 1.92 (t, 2H), 2.68 (dt, 1H), 3.10 (dd, 1H), 4.07 (m, 1H), 4.43 (m, 1H), 7.04 (t, 1H), 7.29 (t, 2H), 7.58 (d, 2H), 8.13 (s, 1H), 8.38 (d, 1H), 8.62 (s, 1H), 10.07 (s, 1H), 10.30 (s, 1H).
EXAMPLE 12
(2S )-N-[(lSr)-l-(cvclopentylcarbamoyl)-7-(hvdroxyamino)-7-oxo-heptyll-6-oxo- piperidine-2-carboxamide
STEP A: methyl (7S)-8-(cyclopentylamino)-8-oxo-7- [[(2S)-6-oxopiperidine-2- carbonyl] amino] octanoate
DMTMM (140 mg, 0.51 mmol) and NMM (92.3 μΐ, 0.84 mmol), were added to a 3 ml solution of methyl (7-S)-7-amino-8-(cyclopentylamino)-8-oxo-octanoate (101 mg, 0.37 mmol) in dry THF at 0°C. The reaction mixture was then stirred for 12 h at RT. The crude reaction mixture was filtered, diluted with AcOEt and the organic layer washed with HCl IN. Removal of the solvent removed under reduced pressure followed by purification by flash chromatography (CHCls/MeOH: 94/6) afforded the ti- tie compound.
Yield: 63% (70 mg).
Ή NMR (400 MHz, CDC13) δ: 1.09- 1.44 (m, 8H), 1.43-2.11 (m, 13H), 2.11-2.49 (m,
4H), 3.60 (s, 3H), 4.04 (dd, 2H), 4.38 (q, 1H), 6.63 (d, 1H), 8.20 (d, 1H).
STEP B: (2S)-N- [(lS)-l-(cyclopentylcarbamoyl)-7-(hydroxyamino)-7-oxo-heptyl] -6- oxo-piperidine-2-carboxamide
HONH2 (50% w/w aq. solution, 165 μΐ, 2.70 mmol) was added to 3 ml solution of methyl (7-S)-8-(cyclopentylamino)-8-oxo-7- [[(2iS)-6-oxopiperidine-2-carbonyl]amino] - octanoate (70 mg, 0.18 mmol in THF (1.8 ml) at 0°C followed by careful addition of IN NaOH (10 eq). The mixture was then allowed to reach RT in 4 h, and was stirred overnight at RT. The reaction mixture was then cooled back to 0°C and diluted with AcOEt before adding HCl IN (8 equiv). The resulting mixture was concentrated by azeotropic evaporation with toluene and the resulting crude reaction mixture was purified by filtration of the residual salts after having been dissolved in MeOH. Yield: 77% (55 mg).
Ή NMR (400 MHz, DMSO-de) δ: 0.94-2.00 (m, 20H), 1.90-2.26 (m, 4H), 3.82-4.01 (m, 2H), 4.14 (d, 1H).
Example 13 was synthesized following the procedure reported for example 12 using methyl (7-S)-7-amino-8-oxo-8-(phenethylamino)octanoate instead of methyl (7<S)-7- amino-8-(cyclopentylamino)-8-oxo-octanoate in step A.
EXAMPLE 13
(28)-N-[(l,S)-7-(hvdroxyamino)-7-oxo-l-(phenethylcarbamoyl)heptyll-6-oxo- piperidine-2-carboxamide
The title compound was obtained as a white solid after purification by flash chromatography (CHCVMeOH: 8/2).
Yield: 52%.
Ή NMR (400 MHz, MeOD-d4) δ: 1.30 (s, 4H), 1.49- 1.95 (m, 8H), 2.06 (q, 2H), 2.32 (t, 2H), 2.79 (t, 2H), 3.35-3.57 (m, 2H), 4.08 (t, 1H), 4.26 (dd, 1H), 7.11-7.33 (m, 5H). COMPARISON EXAMPLES
The following examples which do not present the lactam-carbonylamino moiety on the side chain of the scaffold were tested in vitro as comparison examples. Examples 14-18 have already been reported in the literature and are not part of the pre- sent invention (i.e., Oger F., et al, J. Med. Chem., 2010, 53, 1937; Salmi-Smail C, et al, J. Med. Chem., 2010, 53, 3038).
EXAMPLE 14
8-(hvdroxyamino)-8-oxo-N-phenyl-octanamide
EXAMPLE 15
8-(hydroxyamino)-8-oxo-N-(iP-tolyl)octanamide
EXAMPLE 16
8-(hvdroxyamino)-8-oxo-N-(m-tolyl)octanamide
EXAMPLE 17
8-(hvdroxyamino)-8-oxo-N-[3-(trifluoromethyl)phenylloctanamide
EXAMPLE 18
8-(hvdroxyamino)-8-oxo-N-phenethyl-octanamide
EXAMPLE 19
N-cvclopentyl-8-(hydroxyamino)-8-oxo-octanamide
STEP A: methyl 8-(cyclopentylamino)-8-oxo-octanoate
NMM (0.87 ml, 7.96 mmol) and DMTMM (1.30 g, 4.77 mmol) were added to a solution of 8-methoxy-8-oxooctanoic acid (500 mg, 2.65 mmol) and cyclopentanamine (0.40 ml mg, 3.98 mmol) in 20 ml of dry THF at 0°C. The reaction mixture was then stirred for 12 h at RT and filtered, diluted with AcOEt. The organic layer was washed with HC1 IN and the solvent was removed under reduced pressure. The pure desired adduct was obtained after purification through flash chromatography (PE/AcOEt: 8/2 to 7/3).
Yield: 46% (312 mg).
Ή NMR (400 MHz, CDC13) δ: 1.29-1.13 (m, 4H), 1.47 (q, 4H), 1.63-1.74 (m, 2H), 1.74-1.85 (m, 2H), 2.11 (dt, 4H), 3.26 (m, 5H), 3.49 (s, 3H). BIOLOGY
EXAMPLE 20
HDAC enzymatic assay
HDAC profiling was performed against seven isolated HDAC human isoforms in the presence of a 50 μΜ solution of the fluorogenic tetrapeptide RHKK(Ac) substrate (from p53 residues 379-382) or in the presence of a 50 μΜ solution of its diacetylat- ed analogue RHK(Ac)K(Ac) for HDAC8. Isolated human HDACs were obtained by standard purification, with the exception of HDAC3 which was a human recombinant protein as a complex of full length human HDAC3 with a C-terminal His-tag and human NCOR2 amino acids 395-489 with an N-terminal GST-tag co-expressed in baculovirus expression system. Each compound was dissolved in DMSO, and progressively diluted solutions were used for testing. SAHA was used as reference compound. Upon its deacetylation, the fluorophore was released given rise to fluorescence emission which was detected by a fluorimeter, and the IC50 values of the compounds were determined by analyzing dose-response inhibition curves.
Table 1
Figure imgf000032_0001
IC50 < 150 nM: ++++; 150 nM < IC50 < 250 nM: +++; 250 nM < IC50 < 500 nM: ++; 500 nM < IC50 < 1000 nM: +; NA: Not Active Results
The compounds of the present invention proved to be highly potent on all HDAC isoforms tested with inhibitory activity ranging in the low nanomolar scale (Table 1). Moreover, most of the compounds demonstrated binding affinity with the various HDAC isoforms higher than that of SAHA (i.e., comparison example 14).
Such higher affinity was also observed regardless the nature of the CAP unit as it can be seen from Table 2 wherein the various comparison examples are reported together with the compounds of the present invention.
Table 2
Figure imgf000033_0001
IC50 < 150 nM: ++++; 150 nM < IC50 < 300 nM: +++; 300 nM < IC50 < 500 nM: ++; 500 nM < IC50 < 1000 nM: +; NA: Not Active EXAMPLE 21
Cytotoxicity
The cytotoxic effect of the compounds of the present invention was evaluated on NCI-H460 noN-small cell lung carcinoma according to the method of Skehan et al. (Skehan P., et al, J. Natl. Cancer Inst., 1990, 82, 13, 1107), using SAHA (Vori- nostat) as reference compound.
Tumour cells were grown in RPMI 1640 medium containing 10% heat-inactivated foetal bovine serum and 50 μg/ml gentamycin sulphate and were seeded in 96-well tissue culture plates at approximately 10% confluence. They were allowed to attach and recover for at least 24 h. Varying concentrations of the compounds of the present invention were then added to each well in order to define their IC50 value (i.e., the concentration which inhibits 50% of cell survival).
The plates were incubated for 24 h at 37 °C, after which they were washed 3 times by removal of the supernatant and addition of PBS. The plates were then incubated for further 48 h at 37 °C. 200 μΐ PBS and 50 μΐ of cold 80% TCA were added and the plates were incubated on ice for at least 1 h. TCA was removed and the plates were washed 3 times by immersion in distilled-water. They were then dried on paper at 40°C for 5 min. 200 μΐ of 0.4% sulphorodamine B in 1% acetic acid were added. The plates were incubated at room temperature for further 30 min. Sulphorodamine B was removed, and the plates were washed 3 times by immersion in 1% acetic acid and were dried on paper and at 40 °C for 5 min. Then 200 μΐ Tris 10 mM were added. The plates were kept under magnetic stirring for 20 min. Cell survival was determined by means of optical density by a Multiskan spectrofluorimeter at 540 nm. The amount of cells killed was calculated as the percentage decrease in sulphoro- damine B binding compared to control cultures. The IC50 values reported in Table 3 were calculated with the "ALLFIT" program.
Results
The compounds of the present invention demonstrated a very good inhibition profile, very often much better than that observed for the reference compound SAHA. Moreover, all the compounds of the invention had a better IC50 than the respective parent analogues (Table 3). Table 3
Figure imgf000035_0001
EXAMPLE 22
Cytotoxicity
The cytotoxic effect of some compounds of the present invention was also evaluated against few strains of parasites to assess their efficacy in the treatment of infec- tions.
Plasmodium falciparum
In vitro activity against erythrocytic stages of Plasmodium falciparum (NF54, Schiphol Airport, Netherlands; Ponnudurai T., et al., Trop. Geogr. Med., 1981, 33, 1, 50) was determined using a 3H-hypoxanthine incorporation assay (Desjardins R.E., et al., Antimicrob Agents Chemother., 1979, 16, 6, 710; Matile H., et al., I. Lefkovits and B. Pernis (ed.), Immunological Methods. Academic Press, San Diego, 1990), using the chloroquine as the control drug. Compounds were dissolved in DMSO at 10 mg/ml and added to parasite cultures incubated in RPMI 1640 medium without hy- poxanthine, supplemented with HEPES (5.94 g/1), NaHC03 (2.1 g/1), neomycin (100 U/ml), AlbumaxR (5 g/1) and washed human red cells A+ at 2.5% haematocrit (0.3% parasitaemia). Serial drug dilutions of eleven 3-fold dilution steps covering a range from 100 to 0.002 μg/ml were prepared. The 96-well plates were incubated in a humidified atmosphere at 37°C; 4% C02, 3% 02, 93% N2. After 48 h, 50 μΐ of 3H- hypoxanthine (corresponding to 0.5 μϋί) were added to each well of the plates. The plates were incubated for further 24 h under the same conditions. The plates were then harvested with a Betaplate™ cell harvester (Wallac, Zurich, Switzerland), and the red blood cells transferred onto a glass fibre filter prior to being washed with distilled water. The dried filters were inserted into a plastic foil with 10 ml of scintillation fluid, and counted in a Betaplate™ liquid scintillation counter (Wallac, Zurich, Switzerland). IC50 values were calculated from sigmoidal inhibition curves by linear regression (Huber C, et al., Acta Trop., 1993, 55, 257) using Microsoft Excel software.
Trypanosoma brucei rhodesiense and Trypanosoma cruzi
Experimental conditions used to assess biological activity of the compounds of the invention against these strains are the ones reported in Orhan I., et al., Mar. Drugs, 2010, 8, 47. As evidenced from the results reported in Table 4 underneath, the compounds of the present invention are equipotent or even more active than reference compound d SAHA.
Table 4
Figure imgf000037_0001
Trypanosoma
Examples
Brucei Plasmodium
Cruzi
rhodesiense falciparum
14 3 10 0.02
7 4 1 0.003
9 5 1 0.006
11 6 NT 0.02

Claims

1. A compound having the eneral Formula (I)
Figure imgf000038_0001
Formula I
wherein,
R1 is H, (Ci-C6)-alkyl or aryl;
R2 is phenyl optionally substituted with halogen, benzyloxy, (Ci-C3)-alkyl or CF3; (C3-C6)-cycloalkyl; aryl-(Ci-C6)-alkyl wherein the aryl is optionally substituted with benzyloxy, (Ci-C3)-alkyl or CF3;
-A-E- is -(CO)-(NH)- or -(NH)-(CO)-;
n is an integer comprised between 0 to 2;
R3 is H, or -(CO)-R4;
R4 is (Ci-C6)-alkyl, (Ci-C6)-cycloalkyl, aryl-(Ci-C6)-alkyl, aryl, -CH(R<9(R6), heteroar- yl, or -NH-(Ci-C6)-alkyl;
R5 is H, or (Ci-C6)-alkyl;
R6 is H, (Ci-C6)-alkyl, aryl, or -NH(R7);
Figure imgf000038_0002
R8 is benzyl, or i-Bu;
the symbol means that the carbon atom bearing said symbol can adopt a R or S configuration;
their tautomers, their geometrical isomers, their optically active forms such as en- antiomers, diastereomers and their racemate forms, as well as their pharmaceutically acceptable salts thereof.
2. The compound according to claim 1, wherein -A-E- is -(CO)-(NH)-.
3. The compound according to claim 1, wherein R2 is phenyl.
4. The compound according to claim 3, wherein said phenyl is substituted in position meta.
5. The compound according to claim 1 selected from the group consisting of: N-[(l-S)- 7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-2-oxo-yrrolidine-3- carboxamide N-[(lS)-7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-2-oxo- piperidine-3-carboxamide; N-[(l<S)-7-(hydroxyamino)-7-oxo-l- (phenylcarbamoyl)heptyl]-2-oxo-4-phenyl-pyrrolidine-3-carboxamide (2<S)-N- [(l-¾-7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-5-oxo^yrrolidine-2- carboxamide; (2R)-N-[(l<¾-7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-5- oxo-pyrrolidine-2-carboxamide; (2<¾-N-[(l<¾-7-(hydroxyamino)-7-oxo-l- (phenylcarbamoyl)heptyl]-6-oxo-piperidine-2-carboxamide; (2R)-8- (hydroxyamino)-8-oxo-2- [2-0X0-2- [(2R)-5-oxopyrrolidiN-2-yl]ethyl]-N- [3- (trifluoromethyl)phenyl]octanamide; (2R)-N-[(l<S)-7-(hydroxyamino)-7-oxo-l-(p- tolylcarbamoyl)heptyl]-5-oxo-pyrrolidine-2-carboxamide; (2R)-N-[(lS)-7- (hydroxyamino)-l-(m-tolylcarbamoyl)-7-oxo-heptyl]-5-oxo-pyrrolidine-2- carboxamide; (2R)-N-[(l<¾-7-(hydroxyamino)-7-oxo-l-(phenylcarbamoyl)heptyl]-5- oxo-pyrrolidine-2-carboxamide; (2<S)-N-[(l-S)-7-(hydroxyamino)-7-oxo-l- (phenylcarbamoyl)heptyl]-4-oxo-azetidine-2-carboxamide; (2<S)-N-[(1-S)-1- (cyclopentylcarbamoyl)-7-(hydroxyamino)-7-oxo-heptyl]-6-oxo-piperidine-2- carboxamide; (2<¾-N-[(l-¾-7-(hydroxyamino)-7-oxo-l-(phenethylcarbamoyl) hep- tyl]-6-oxo-piperidine-2-carboxamide; and their corresponding acetate, 2- methylpropanoate, propanoate, 3-methylbutanoate, benzoate, 2-phenylacetate, pyridine-2-carboxylate, N-ethylcarbamate, N-isopropylcarbamate, N- benzylcarbamate, and 2-(tert-butoxycarbonylamino)propanoate, 2- (benzyloxycarbonylamino)propanoate prodrugs.
6. A pharmaceutical composition containing at least one compound according to claims 1-5 as the active ingredient in admixture with at least one pharmaceutically acceptable vehicle and/or excipient.
7. The compound according to any of claims 1-5 for use as a medicament.
8. The compound according to claim 7 wherein the medicament is useful for the treatment of a pathological state for which the modulation of HDAC activity would result at improving the health of the patient.
9. The compound according to claim 8 where the pathological state is a cancer dis- neurodegenerative disease, an inflammatory disease, stroke, ischemia or viral or Plasmodium and/or Trypanosoma infections.
10. The compound according to claim 9 where the cancer disease is cancer of the breasts, pancreas, lung, colon, pleura, peritoneum, face and neck, kidney, bladder, brain, prostate, ovaries or eyes.
11. The compound according to claim 10 where the cancer is a metastatic form of cancer.
12. The compound according to claim 9 where the inflammatory disease is rheumatoid arthritis.
13. The compound according to claim 9 where the neurodegenerative disease is Huntington's disease, Parkinson's disease, or amyotrophic lateral sclerosis.
14. A process for preparing the pharmaceutical composition of claim 6, comprising mixing at least one compound of Formula (I) with at least one pharmaceutically acceptable vehicle and/or excipient.
15. A compound of Formula II
Formula II
Figure imgf000040_0001
wherein,
R2 is as defined in compound of Formula I of claim 1 for use as intermediate in the synthesis of compound of Formula I of claim 1 and Bn is a benzyl group.
16. A method of treatment of a patient affected by a cancer disease comprising the administration of a compound according to claims 1-5.
PCT/EP2014/052351 2013-02-08 2014-02-06 Hydroxamate derivatives bearing amide-lactams as potent hdac inhibitors and their uses as medicaments WO2014122222A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP13154686 2013-02-08
EP13154686.3 2013-02-08

Publications (1)

Publication Number Publication Date
WO2014122222A1 true WO2014122222A1 (en) 2014-08-14

Family

ID=47681785

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/052351 WO2014122222A1 (en) 2013-02-08 2014-02-06 Hydroxamate derivatives bearing amide-lactams as potent hdac inhibitors and their uses as medicaments

Country Status (3)

Country Link
AR (1) AR094731A1 (en)
TW (1) TW201443016A (en)
WO (1) WO2014122222A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2608476A1 (en) * 2017-02-20 2017-04-11 Universidad De Granada Compounds for the treatment of diseases caused by parasites of the gender leishmania (Machine-translation by Google Translate, not legally binding)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006005955A1 (en) * 2004-07-12 2006-01-19 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Amide derivatives as inhibitors of histone deacetylase
US20090023786A1 (en) 2004-04-05 2009-01-22 Alton Pharma, Inc. Histone Deacetylase Inhibitor Prodrugs

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090023786A1 (en) 2004-04-05 2009-01-22 Alton Pharma, Inc. Histone Deacetylase Inhibitor Prodrugs
WO2006005955A1 (en) * 2004-07-12 2006-01-19 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Amide derivatives as inhibitors of histone deacetylase

Non-Patent Citations (32)

* Cited by examiner, † Cited by third party
Title
"Guidance for Industry and Reviewers", 2002, U.S. FOOD AND DRUG ADMINISTRATION
"Re- mington's Pharmaceutical Sciences", 1991, MACK PUB. CO.
"Remington's Pharmaceutical Science Handbook", MACK PUB
ARCHIN N. M., NATURE, vol. 487, 2012, pages 482 - 486
ARROWSMITHC.H. ET AL., NATURE, vol. 11, 2012, pages 384
BARTLING B. ET AL., LUNG CANCER, vol. 49, 2005, pages 145
BELVEDERE ET AL: "Aminosuberoyl hydroxamic acids (ASHAs): A potent new class of HDAC inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, GB, vol. 17, no. 14, 19 June 2007 (2007-06-19), pages 3969 - 3971, XP022119991, ISSN: 0960-894X, DOI: 10.1016/J.BMCL.2007.04.089 *
CHOI J.H. ET AL., JPN. J. CANCER RES., vol. 92, 2001, pages 1300
CHUANG D.M. ET AL., TRENDS IN NEUROSCIENCE, vol. 32, no. 11, 2009, pages 591
DESJARDINS R.E. ET AL., ANTIMICROB AGENTS CHEMOTHER., vol. 16, no. 6, 1979, pages 710
DI- NARELLO C.A. ET AL., MOL. MED., vol. 17, 2011, pages 333
GIAN- NINI G. ET AL., FUTURE MEDICINAL CHEMISTRY, vol. 4, no. 11, 2012, pages 1439 - 1460
GRANT S. ET AL., NATURE REV. DRUG DISCOV., vol. 6, 2007, pages 21
GREENE T. W; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1999, J. WILEY & SONS, INC.
HALKIDOU K. ET AL., PROSTATE, vol. 59, 2004, pages 177
HAMAMORI Y. ET AL., J. CLIN. INVEST., vol. 112, no. 6, 2003, pages 824
HUBER C. ET AL., ACTA TROP., vol. 55, 1993, pages 257
LEONI F. ET AL., PROC. NATL. ACAD. SCI., vol. 99, 2002, pages 2995
MARCH J.: "Advanced Organic Chemistry", 1992, J. WILEY & SONS, INC.
MATILE H. ET AL.: "Immunological Methods", 1990, ACADEMIC PRESS
OGER F. ET AL., J. MED. CHEM., vol. 53, 2010, pages 1937
ORHAN I. ET AL., MAR. DRUGS, vol. 8, 2010, pages 47
PARIS M. ET AL., J. MED. CHEM., vol. 51, 2008, pages 1505
PONNUDURAI T. ET AL., TROP. GEOGR. MED., vol. 33, no. 1, 1981, pages 50
SAKUMA T. ET AL., J. ONCOL., vol. 29, 2006, pages 117
SALMI-SMAIL C. ET AL., J. MED. CHEM., vol. 53, 2010, pages 3038
SAMPATH-KUMAR A. ET AL., BIOORG. MED. CHEM. LETT., vol. 15, no. 8, 2005, pages 1969
SCHLIMME S. ET AL., CHEMMEDCHEM, vol. 6, 2011, pages 1193
SKEHAN P. ET AL., J. NATL. CANCER INST., vol. 82, no. 13, 1990, pages 1107
SLEIMAN S.F. ET AL., EXPERT OPIN. INVESTIG. DRUGS, vol. 18, no. 5, 2009, pages 573
SONG J. ET AL., APMIS, vol. 113, 2005, pages 264
YANG X.J. ET AL., MOL. CELL. BIOL., vol. 25, 2005, pages 2873

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2608476A1 (en) * 2017-02-20 2017-04-11 Universidad De Granada Compounds for the treatment of diseases caused by parasites of the gender leishmania (Machine-translation by Google Translate, not legally binding)

Also Published As

Publication number Publication date
TW201443016A (en) 2014-11-16
AR094731A1 (en) 2015-08-26

Similar Documents

Publication Publication Date Title
DK2860174T3 (en) Benzamide derivative as histone deacetylase inhibitor with potent differentiation and antiproliferative activity
SK49194A3 (en) Urethanes and ureas which are inducing production of cytokine
AU2004226215A1 (en) Oxime derivatives and their use as pharmaceutically active agents
EP3062784A1 (en) Antimicrobial compounds
JPH05504968A (en) Glycinamide derivatives, their production and pharmaceuticals containing them
DK2758371T3 (en) NEW LACTAMBÆRENDE thio derivatives as potent HDAC INHIBITORS AND USES THEREOF AS MEDICINES
US20120258993A1 (en) Non-natural macrocyclic amide hdac6 inhibitor compounds and their uses as therapeutic agents
CA2774569A1 (en) Novel n-benzylamide substituted derivatives of 2-(acylamido)acetic acid and 2-(acylamido)propionic acids: potent neurological agents
WO2014122222A1 (en) Hydroxamate derivatives bearing amide-lactams as potent hdac inhibitors and their uses as medicaments
US11795144B2 (en) Inhibitors of RAC1 and uses thereof for treating cancers
Dusek et al. Synthesis and antiproliferative activities of novel O-benzyl salicylamide derivatives
RU2737433C2 (en) Benzo-n-hydroxyamide compounds having anti-tumor activity
US20180305404A1 (en) Novel ligands for prevention of neurotoxicity of the alzheimer&#39;s disease related amyloid-beta peptide
US11149062B2 (en) HDAC inhibitors and methods of treatment using the same
WO2015200349A2 (en) Elastase inhibitors
CA2181986A1 (en) Carbamoylmethylurea derivatives
Soyer et al. Synthesis and anticonvulsant activity of some N-(benzoyl) glycinanilide derivatives

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: 14702884

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: 14702884

Country of ref document: EP

Kind code of ref document: A1