WO2009136001A1 - Protein kinase modulating agents - Google Patents

Protein kinase modulating agents Download PDF

Info

Publication number
WO2009136001A1
WO2009136001A1 PCT/FI2009/050365 FI2009050365W WO2009136001A1 WO 2009136001 A1 WO2009136001 A1 WO 2009136001A1 FI 2009050365 W FI2009050365 W FI 2009050365W WO 2009136001 A1 WO2009136001 A1 WO 2009136001A1
Authority
WO
WIPO (PCT)
Prior art keywords
branched
compound
linear
chosen
ring
Prior art date
Application number
PCT/FI2009/050365
Other languages
French (fr)
Inventor
Raimo Tuominen
Jari Yli-Kauhaluoma
Olli Aitio
Gustav BOIJE AF GENNÄS
Elina Ekokoski
Moshe Finel
Virpi Talman
Pia Vuorela
Anna Galkin
Janet Lord
Original Assignee
Raimo Tuominen
Jari Yli-Kauhaluoma
Olli Aitio
Boije Af Gennaes Gustav
Elina Ekokoski
Moshe Finel
Virpi Talman
Pia Vuorela
Anna Galkin
Janet Lord
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 Raimo Tuominen, Jari Yli-Kauhaluoma, Olli Aitio, Boije Af Gennaes Gustav, Elina Ekokoski, Moshe Finel, Virpi Talman, Pia Vuorela, Anna Galkin, Janet Lord filed Critical Raimo Tuominen
Publication of WO2009136001A1 publication Critical patent/WO2009136001A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/216Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acids having aromatic rings, e.g. benactizyne, clofibrate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Definitions

  • the present invention concerns new protein kinase modulating agents. Particularly, but not exclusively, the present invention concerns protein kinase C translocation inhibitors.
  • the agents described here affect signal transduction pathways regulating protein phosphorylation, which leads to modulation of apoptosis. They also possess antileukaemic and anti-inflammatory properties.
  • Apoptosis modulating agents are widely sought, because failure in regulation of apoptosis is associated with many diseases.
  • Apoptosis programmed cell death, is a genetically coded mechanism to destroy unwanted cells.
  • Apoptosis plays an important role in development, in tissue homeostasis maintenance and in defence against viral infections and mutations.
  • Apoptosis is a rapid process and causes no tissue damage, since apoptotic cells are rapidly phagocytosed by other cells.
  • Caspases a family of intracellular cysteine proteases, have a central role in apoptosis. Caspases are present in cells as inactive procaspases, which can be rapidly converted to active caspases. After pro-apoptotic signal, initiator caspases (caspase-8 and caspase-9) are initially activated, and initiator caspases then activate precursor forms of effector caspases (caspase-3, -6 and -7) to active caspases. Effector caspases cleave several cellular proteins with essential functions leading to apoptotic phenotype.
  • Apoptosis is characterized by morphological features such as chromatin condensation, nuclear fragmentation, cell membrane blebbing and formation of apoptotic bodies.
  • Caspases can be activated by distinct pathways, the death receptor pathway and the mitochondrial pathway.
  • pro-apoptotic signal-transducing molecules and pathological stimuli cause mitochondrial outer membrane permeabilization, which in turn induces apoptosis further, since caspase-activating molecules and caspase-independent death effectors are released from the space between the inner and the outer membrane of mitochondria.
  • Dissipation of the mitochondrial inner transmembrane potential ( ⁇ m) is frequently associated with the mitochondrial pathway of apoptosis.
  • Defective apoptosis regulation is associated with many diseases. Insufficient apoptosis occurs for example in cancer and autoimmune diseases. In contrast, excessive apoptosis is associated with heart failure, neurodegenerative diseases and AIDS (Acquired Immune Deficiency Syndrome). Thus, new apoptosis modulating molecules are widely sought from natural products and from synthetic compounds.
  • caspase-3 -dependent proteolytic activation of PKC ⁇ plays a key role in oxidative stress-mediated apoptosis in dopaminergic cells after exposure to an environmental neurotoxic agent.
  • PKC ⁇ protein kinase C delta
  • Activation of caspases by cytosolic cytochrome C is an early and essential step in the apoptotic-signalling pathway.
  • caspase-3 plays a major role in the regulation and execution phase of both in vitro and in vivo models of apoptosis.
  • Protein kinases play an important role in many signalling pathways in cells.
  • Protein kinase C (PKC) is a family of serine/threonine kinases that regulate various cellular processes including cell proliferation and malignant transformation.
  • isophthalic acid-derived compounds which compete with phorbol esters for binding and thus modulate PKC activity, have been designed and synthesized. They have high affinity for the diacyl glycerol binding site of PKC and modulate the function of PKC. The compounds may bind also to other site on PKC and modify phorbol ester binding indirectly via conformational change of phorbol ester binding site. Thus, it is possible for these agents to modify the activity of other protein kinases than PKC as well.
  • Phorbol esters are potent cocarcinogens, activating various PKC- isoenzymes and thereby acting as tumour promoters.
  • Phorbol myristate acetate (PMA) (typical phorbol ester)
  • the compounds of the present invention compete with, e.g., phorbol esters at the PKC binding site, thus preventing the tumour promoting activity of the phorbol esters.
  • the compounds of the invention have been tested in in vitro receptor binding, enzymatic activity assays (some inhibitors with IC50 around 10 ⁇ M), an intracellular translocation assay using confocal microscopy of HeLa cells, and in leukaemia cell lines, such as HL-60, and primary cell lines from patients with acute myeloid leukaemia (AML) or chronic lymphatic leukaemia (CLL).
  • enzymatic activity assays some inhibitors with IC50 around 10 ⁇ M
  • an intracellular translocation assay using confocal microscopy of HeLa cells and in leukaemia cell lines, such as HL-60, and primary cell lines from patients with acute myeloid leukaemia (AML) or chronic lymphatic leukaemia (CLL).
  • AML acute myeloid leukaemia
  • CLL chronic lymphatic leukaemia
  • Leukaemia is cancer of the blood cells. Like most cancer cells, leukaemic cells show a variety of alterations in genes controlling cell proliferation (e.g. Flt3, cyclinD) or apoptosis (bcl2, abl). The inventors have discovered that in particular, two isoenzymes of PKC, PKC ⁇ and PKC ⁇ , play specific roles in tumour promotion and suppression.
  • the compounds of the present invention show therapeutic potential for, among others, leukaemia, in particular for acute myeloid leukaemia (AML) and chronic lymphatic leukaemia (CLL). Further, the inventors have discovered that the compounds of the invention possess concentration dependent anti-inflammatory activity in cultured human lymphocytes.
  • the invention provides new compounds that are potent anticancer and anti-inflammatory agents and are reasonably safe and non-toxic. There is a global unmet need for such compounds.
  • the commercial potential of protein kinase modulating agents including PKC modulating, anti-inflammatory and antileukaemic agents is obvious.
  • the incidence, for example, of rheumatoid arthritis is about 1% world wide, which means there are about 60 000 000 people suffering from the disease in the world. Further, every year tens of millions of people around the world die from cancerous diseases.
  • the aim of the present invention is to provide new simple-to-synthesize phorbol-ester displacing compounds that modulate the function of protein kinases, especially that of PKC, and thus prevent unwanted cell division.
  • compounds that function as drugs and target the Cl domain of PKC isoenzymes ⁇ and ⁇ are particularly interested for the present invention.
  • the invention provides compounds that regulate protein kinase activity, such as PKC activity. These compounds are hydrophobic derivatives of isophthalic acid, particularly esters of isophthalic acid.
  • the present invention concerns compounds that modulate protein kinase activity and have the following Formula I:
  • R 1 , R 2 and R 3 may be the same or different and are each independently chosen from hydrogen, branched or linear alkyl, branched or linear hydroxyalkylene, nitro, amino,
  • composition of the present invention is characterized by what is stated in Claim 18 and the use of the present invention is characterized by what is stated in Claims 26 and 28.
  • the compounds of the present invention are useful particularly in the treatment and prevention of inflammatory and cancerous diseases by modulating the function of PKC isoenzymes ⁇ and ⁇ as well as in the treatment and prevention of Down's syndrome by inhibiting DYRKIa, but leaving casein kinase 2 (CK2) unmodulated.
  • AML acute myeloid leukaemia AML acute myeloid leukaemia
  • Nonlimiting examples of aryl groups containing one aromatic ring or two or more fused or linked aromatic rings include phenyl, naphthyl, biphenyl, diphenyl ether, diphenylamine, benzophenone, and the like.
  • Aryl groups can optionally be substituted with one or more substituents.
  • substituents include halo, nitro, cyano, linear or branched alkyl, linear or branched alkenyl, linear or branched haloalkyl, aryl, cycloalkyl, cycloalkenyl, amino, amido, carboxylate, and hydroxy.
  • the compounds of Formula I may be used as medicaments. Preferably, they are used in the manufacture of medicaments for the treatment of disorders relating to activation or inhibition of the PKC isoenzymes or other protein kinases in mammals, particularly in humans.
  • the medicaments may be used in diagnosing, treating or ameliorating various cancers or inflammatory disorders, such as leukaemia or arthritis.
  • Activation or inhibition of the PKC signaling pathway can be measured using various sensitive assay systems where the effect of a compound of interest is tested over a range of concentrations, i.e. showing a dose-response, including concentrations at which no or minimal effect is observed, through higher concentrations at which partial effect is observed, to saturating concentrations at which a maximum effect is observed.
  • the dose- response of the effect of a compound of interest is usually a sigmoidal curve expressing a degree of inhibition as a function of concentration. The curve also theoretically passes through a point at which the concentration is sufficient to reduce or enhance the activity of the PKC signaling pathway to a level that is 50% that of the difference between minimal and maximal activity in the assay.
  • This concentration is defined as the Inhibitory Concentration (50%) or IC50 value.
  • Determination of IC50 values preferably is made using conventional biochemical (acellular) assay techniques or cell based assay techniques. It is not uncommon to obtain a bell-shape dose-response curve where maximum effect is obtained in lower concentrations than the maximum concentrations used in the assay. The data obtained in dose-response assays can be used as a factor in formulating a dosage range for use in mammals and, more specifically, humans.
  • doses employed for humans typically are in the range of 0.001 mg/kg to about 1000 mg/kg per day, preferably 0.01 mg/kg to about 50 mg/kg per day, typically in a range of about 0.0005 to about 500 mg/kg per dose of activator/inhibitor, preferably from about 0.1 mg/kg to about 10 - 50 mg/kg per dose of activator/inhibitor.
  • doses range from about 0.1 to about 50 mg/kg, about 0.5 to about 40 mg/kg, about 0.7 to about 30 mg/kg, or about 1 to about 20 mg/kg. Specific doses contemplated include sub-ranges of any of the foregoing ranges in 0.1 mg/kg increments.
  • the pharmaceutical forms suitable for injectable use include, for example, sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form is preferably sterile and fluid to provide easy syringability. It is preferably stable under the conditions of manufacture and storage and is preferably preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the suspension may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the compositions may also be solutions or suspensions in non-toxic diluens or solvents, for example as a solution in 1,3-butanediol.
  • the prevention of the action of microorganisms may be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal and the like.
  • various antibacterial and antifungal agents for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions may be achieved by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions may be prepared by incorporating the active compound in the required amount, in the appropriate solvent, with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions may be prepared by incorporating the sterilized active ingredient into a sterile vehicle that contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation may include vacuum drying and the freeze drying technique, which yield a powder of the active ingredient, plus any additional desired ingredient from the previously sterile- filtered solution thereof.
  • Oral formulations include tablets, buccal tablets, troches, pills, capsules, elixirs, suspensions, syrups, wafers and the like and may further contain a binder, such as gum tragacanth, acacia, corn starch or gelatin; an excipient, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; a sweetening agent such as sucrose, lactose or saccharin; or a flavoring agent, such as peppermint, oil of wintergreen or cherry flavoring.
  • a binder such as gum tragacanth, acacia, corn starch or gelatin
  • an excipient such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as
  • polymers are poly-glycolic acid, poly- lactic acid and po Iy-JJ- lactide/glycolide.
  • Other examples for polymers are water-soluble polymers such as polyoxaalkylenes (polyoxaethylene, polyoxapropylene and mixed polymers thereof, poly- acrylamides and hydroxylalkylated polyacrylamides, poly-maleic acid and esters or - amides thereof, poly-acrylic acid and esters or -amides thereof, poly-vinylalcohol and esters or -ethers thereof, poly-vinylimidazole, poly-vinylpyrrolidon and natural polymers like chitosan.
  • Nonlimiting examples of excipients useful in a composition described herein include phosphates such as dicalcium phosphate.
  • Nonlimiting examples of lubricants used in a composition described herein include natural or synthetic oils, fats, waxes or fatty acid salts such as magnesium stearate.
  • the term "pharmaceutically acceptable salts” refers to salts or zwitterionic forms of the compounds a described above. Salts of such compounds can be prepared during the final isolation and purification of the compounds or separately by reacting the compound with an acid having a suitable cation. Suitable pharmaceutically acceptable cations include alkali metal (e.g., sodium or potassium) and alkaline earth metal (e.g., calcium or magnesium) cations. In addition, the pharmaceutically acceptable salts of the disclosed compounds that contain a basic center are acid addition salts formed with pharmaceutically acceptable acids.
  • alkali metal e.g., sodium or potassium
  • alkaline earth metal e.g., calcium or magnesium
  • acids which can be employed to form pharmaceutically acceptable salts include inorganic acids such as hydrochloric, hydrobromic, sulfuric and phosphoric, and organic acids such as oxalic, maleic, succinic, malonic and citric acid.
  • salts of compounds of the invention include, but are not limited to, hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, 2-hydroxyethanesulfonate, phosphate, hydrogen phosphate, acetate, adipate, alginate, aspartate, benzoate, butyrate, camphorate, camphorsulfonate, citrate, digluconate, glycerolphosphate, hemisulfate, heptanoate, hexanoate, formate, succinate, malonate, fumarate, maleate, methanesulfonate, mesitylenesulfonate, naphthylenesulfonate, nicotinate,
  • the pharmaceutical composition may contain formulation materials for modifying, maintaining or preserving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition.
  • formulation materials for modifying, maintaining or preserving for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition.
  • Suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogensulf ⁇ te); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates, phosphates or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or hydroxypropyl-beta-cyclodextrin); fillers; monosaccharides, disaccharides and other carbohydrates (such as glucose, mannose or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring, flavoring and diluting agents; emulsifying
  • Nonlimiting examples of sweetening agents useful in a composition described herein include sucrose, fructose, lactose or aspartame.
  • Nonlimiting examples of flavoring agents for use in a composition described herein include peppermint, oil of wintergreen or fruit flavors such as cherry or orange flavor.
  • Nonlimiting examples of coating materials for use in a composition described herein include gelatin, wax, shellac, sugar or other biological degradable polymers.
  • Nonlimiting examples of preservatives for use in a composition described herein include methyl or propylparabens, sorbic acid, chlorobutanol, phenol and thimerosal.
  • the primary vehicle or carrier in a pharmaceutical composition may be either aqueous or non-aqueous in nature.
  • a suitable vehicle or carrier may be water for injection, physiological saline solution, solution or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration.
  • Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles.
  • Other exemplary pharmaceutical compositions comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, which may further include sorbitol or a suitable substitute therefore.
  • compositions of compounds affecting protein phosphorylation pathways including PKC activators, inhibitors or modulators, in formulations for inhalation or in sustained- or controlled-delivery formulations.
  • Techniques for formulating a variety of sustained- or controlled-delivery means such as liposome carriers, bio-erodible microparticles or porous beads and depot injections, are also known to those skilled in the art. See for example, PCT Application No. PCT/US93/00829, which describes the controlled release of porous polymeric microparticles for the delivery of pharmaceutical compositions.
  • sustained-release preparations include semipermeable polymer matrices in the form of shaped articles, e.g.
  • Sustained release matrices may include polyesters, hydrogels, polylactides, copolymers of glutamic acid and gamma ethyl-L-glutamate, poly (2-hydroxyethyl- methacrylate), ethylene vinyl acetate or poly-D-3-hydroxybutyric acid.
  • Sustained-release compositions may also include liposomes.
  • composition of the present invention is preferably formulated for oral or intravenous administration. More preferably, the composition is administered orally when used in the treatment of inflammatory diseases and intravenously when used in the treatment of cancers.
  • the active agents can be formulated with or without those carriers customarily used in the compounding of solid dosage forms such as tablets and capsules.
  • a capsule may be designed to release the active portion of the formulation at the point in the gastrointestinal tract when bioavailability is maximized and pre-systemic degradation is minimized.
  • Additional agents can be included to facilitate absorption of the compounds including PKC activators, inhibitors or modulators. Diluents, flavorings, low melting point waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents and binders may also be employed.
  • the pharmaceutical composition to be used for in vivo administration typically must be sterile. This may be accomplished by filtration through sterile filtration membranes. Where the composition is lyophilized, sterilization using these methods may be conducted either prior to or following lyophilization and reconstitution.
  • the composition for parenteral administration may be stored in lyophilized form or in a solution.
  • parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. Once the pharmaceutical composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or as a dehydrated or lyophilized powder.
  • Such formulations may be stored either in a ready-to-use form or in a form (e.g., lyophilized) requiring reconstitution prior to administration.
  • the compounds of this invention can, when used in cancer therapy, be used together with other substances and compounds, such as chemotherapeutic agents, when appropriate.
  • Such compounds are, for example (according to the general classes of the compounds):
  • Alkylating agents such as cyclophosphamide, cisplatin, carboplatin, ifosfamide, chlorambucil, busulfan, thiotepa, nitrosoureas);
  • Antimetabolites such as 5-fluorouracil, fludarabine, methotrexate, azathioprine, gemcitabine (Gemzar);
  • Antitumour antibiotics such as doxorubicin, daunorubicin, epirubicin, actinomycin, bleomycin, mitomycin, plicamycin, dactinomycin, adriamycin;
  • Topoisomerase inhibitors such as irinotecan, topotecan, amsacrine, etoposide, etoposide phosphate, teniposide;
  • capecitabine Xeloda
  • podophyllotoxin amoxicillin
  • piroxicam Xeloda
  • Examples include, but are not limited to, acetoxyalkyls, acetate, formate and benzoate derivatives of alcohol, thiol and amine functional groups; and alkyl, carbocyclic, aryl and alkylaryl esters such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-bvXy ⁇ , cyclopropyl, phenyl, benzyl and phenethyl esters and the like.
  • the present invention further provides a method of regulating protein kinase activity, especially that of PKC activity in a subject comprising administering to the subject a composition comprising a compound of the invention, as described above, or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, in an amount effective to regulate the protein kinase activity.
  • the subject is a mammal, more preferably a human.
  • the compound of Formula I used when treating cancer is one of the following:
  • the compound of the invention is used in the treatment or prevention of an inflammatory disease, particularly in manufacturing a medicament for treating a subject suffering from an inflammatory disease, such as rheumatoid arthritis.
  • the compound of Formula I used when treating an inflammatory disease is the following:
  • LC-MS Liquid chromatography mass spectra
  • FT-IR spectra were recorded on a Perkin Elmer FT-IR 1725X spectrometer from a KBr tablet.
  • the 5-(hydroxymethyl)isophthalates are prepared according to the following general scheme (Scheme 2).
  • the isophthalates 15a-f are prepared according to the following general scheme (Scheme 4).
  • alkyl 3-(hydroxyr»ethy ⁇ benzoates 29a-c are prepared according to the foJ lowing general scheme (Scheme 7),
  • Step F Synthesis of 4- [(tetrahydro-2H-pyran-2-yloxy)methyl] phenol, 7c
  • Step B Synthesis of 5-(tetrahydropyran-2-yloxymethyl)isophthalic acid, 4
  • Step C - Method A Synthesis of the diester, 5al-8
  • reaction mixture was cooled down to rt and quenched by adding an ice-water- mixture (20 mL), extracted with EtOAc (3x20 mL), dried (Na 2 SO 4 ), filtered and evaporated in vacuo to give a yellowish oil, ⁇ (4-te/t-butylbenzyl)-5-(tetrahydropyran-2- yloxymethyl)isophthalate 5al.
  • the crude diesters were used in the subsequent deprotection steps without further purification.
  • Step C - Method B Synthesis of the diester, 5bl-20
  • Method A was used except that 3-(trifluoromethyl)benzyl chloride (3 equiv.) was used and the reaction was carried out in a 5 x larger scale. The reaction mixture was stirred at 80 °C for 2.5 h. A brown oil was obtained.
  • Method A was used except that 1-iodopentane was used. A yellow liquid was obtained.
  • Method B was used except that DMAP was not used but cyclohexylmethanol (2 equiv.), CDI (2 equiv.), DBU (2 equiv.) and DMF (12.5 niL) were used and the reaction was carried out in a 2 x larger scale. After all the reagents had been added, the reaction mixture was stirred at 40 °C for 19 h. DMAP (22 mg) (0.1 equiv.) was added and the reaction mixture was stirred at 40 0 C for 1 h. A clear oil was obtained after work up.
  • Method B was used except that 3-heptanol and DMF (1 mL) was used. After all the reagents had been added, the reaction mixture was stirred at 40 °C for 43 h. A yellow oil was obtained after work up.
  • Method B was used except that DMAP was not used but 3-heptanol (180 ⁇ L, 2 equiv.), DMF (5 mL) was used and the reaction was carried out in a 0.8 x smaller scale. The reaction mixture was stirred at 40 °C for 19 h. A yellowish oil was obtained after work up.
  • Method B was used except that DMAP was not used but 2-heptanol (2 equiv.), DBU (2 equiv.), CDI (2 equiv.) and DMF (12.5 mL) was used and the reaction was carried out in a 2 x larger scale.
  • the reaction mixture was stirred at 40 °C for 18 h, DMAP (22 mg) (0.1 equiv.) was added and the reaction mixture was stirred at 40 0 C for 22 h.
  • 2-heptanol 100 ⁇ L, 0.18 equiv
  • An oil was obtained after work up.
  • Method B was used except that 3-hexanol and DMF (1 mL) was used. The reaction mixture was stirred at 40 °C for 43 h. A yellowish oil was obtained after work up.
  • Method A was used except that 7b (2.2 equiv.) and DMF (10 mL) was used and the reaction mixture was stirred for 130 min.
  • the reaction mixture was cooled down to rt and quenched by adding an ice-water-mixture (40 mL), extracted with EtOAc (2x40 mL), washed with saturated NaHCO 3 -SO lution (2x 15 mL), brine (2x15 mL), dried (Na 2 SO 4 ), filtered and evaporated in vacuo to give a oil, 5b8.
  • Method B was used except that c ⁇ -decahydro-l-naphthol, DBU (3 equiv.) and DMF (1 niL) were used.
  • the reaction mixture was stirred at 40 °C for 43 h. A yellowish oil was obtained after work up.
  • Method B was used except that cyclohexanol, CDI (2 equiv.), DMAP (0.05 equiv.) and DMF (3.75 mL) were used and the reaction was carried out in a 0.6 x smaller scale. The reaction mixture was stirred at 40 °C for 41 h. A yellow liquid was obtained after work up.
  • Method B was used except that tetrahydro-3-furanylmethanol, CDI (2 equiv.), DMAP (0.05 equiv.) and DMF (3.75 mL) were used and the reaction was carried out in a 0.6 x smaller scale. The reaction mixture was stirred at 40 °C for 41 h. A clear oil was obtained after work up.
  • Method B was used except that DMAP was not used but decahydro-2-naphthol, DBU (3 equiv.), and DMF (1 mL) were used. The reaction mixture was stirred at 40 °C for 43 h. A clear oil was obtained after work up.
  • Method B was used except that 3-ethoxy-l-propanol, CDI (2.5 equiv.), DMAP (0.2 equiv.), DBU (3 equiv.) and DMF (7.5 mL) were used and the reaction was carried out in a 1.2 x larger scale. The reaction mixture was stirred at 40 °C for 24 h. A yellow liquid was obtained after work up.
  • Method B was used except that (li?)-2,3-dihydro-lH-inden-l-ol and DMF (0.8 mL) were used and the reaction was carried out in a 0.8 x smaller scale.
  • the reaction mixture was stirred at 40 °C for 24 h. A yellow liquid was obtained after work up.
  • Method B was used except that (2i?)-2-methylpentan-l-ol 13bl6, and DMF (2 mL) were used and the reaction was carried out in a 2 x larger scale. The reaction mixture was stirred at 40 °C for 21 h. A yellow oil was obtained after work up.
  • Method B was used except that (25)-2-methylpentan-l-ol 13bl7, and DMF (1 mL) were used. The reaction mixture was stirred at 40 °C for 24 h. A yellow oil was obtained after work up.
  • Method B was used except that 4-methyl-l-pentanol was used.
  • the reaction mixture was stirred at 40 °C for 18 h. A yellow oil was obtained after work up.
  • Method B was used except that 3-methyl-l-pentanol was used.
  • the reaction mixture was stirred at 40 °C for 18 h. A yellow oil was obtained after work up.
  • Step C Method C: Synthesis of tol[4-(telrahydro-2//-pyran-2- yloxy)methyl] phenyl j -5-(tetrahydropyran-2-yloxymethyl)isophtnalate, 5cl
  • reaction was quenched by adding ice-water (20 mL), extracted with EtOAc (2x25 mL), washed with saturated NaHCO 3 -solution (2x15 mL), brine (2x15 mL), dried over Na 2 SO 4 , filtered and evaporated in vacuo to give a yellow oil.
  • Step G Synthesis of (2Jf)-2-amino-3-phenylpropan-l-ol, 9bl6, a typical procedure
  • Step H Synthesis of (4 ⁇ )-4-benzyl ⁇ lJ ⁇ oxazoliclM-2-one, 10bl6, a typical procedure
  • Step J Synthesis of (4if)-4-benzj1-3- ⁇ (2/f)-2-methylpentanoyI]-l,3-oxazo ⁇ idin-2-one, 12bl6, a typical procedure
  • Sodium ⁇ / ⁇ (trimcthylsilyl)amidc (NaHDMS) (IM in THF, 95.59 niL, 1.11 cquiv.) was dropwise added during 2 h to a cooled down (-78 °C) solution of llbl ⁇ (22.5 g, 1 equiv.) in dry THF (95 mL) and stirred at that temperature for 30 min.
  • Methyl iodide 26.86 niL, 5 equiv. was dropwise added during 5 min ai -78 0 C and stirred for 4 h at that temperature.
  • LAH lithium aluminium hydride
  • Method B was used except that 1-phenylpiperazine and DMF (1 niL) were used. The reaction mixture was stirred at 40 °C for 22 h. A yellow oil was obtained after work up.
  • Method I was used except of that hexylamine and DCM (5 mL) were used and the reaction was carried out in a 0.7 ⁇ smaller scale. The reaction mixture was stirred at 40 0 C for 22.5 h and after work up a clear oil could be obtained.
  • Step A was used except of that 25 (476 mg, 1 equiv.), DHP (314 ⁇ L, 1.2 equiv.), PPTS (72 mg, 0.1 equiv.) and 1 ,2-dichloroethane (12 niL) were used, the reaction was carried out in a 0.13 x smaller scale and it was stirred at room temperature for 16 h.
  • Step B was used except of that a 10% solution of KOH (8.69 mL, 4 equiv.), 26 (716 mg, 1 equiv.) and MeOH (13.6 mL) were used and the reaction mixture was refluxed at 90 0 C for 17 h.
  • the reaction mixture was evaporated in vacuo, water (20 mL) was added to the oil, the mixture was washed with EtOAc (3 ⁇ 20 mL), cooled down on ice, pH adjusted to 4 with a 25% solution of KHSO 4 , extracted with DCM (3x30 mL), washed with brine (30 mL), dried (Na 2 SO 4 ), filtered, evaporated in vacuo to give a clear oil (452 mg, 67% yield).
  • Step P Synthesis of hexyl 3-(tetrahydropyran-2-yIoxvmethvI)benzoate, 28a
  • step C was used except of that 27 (1 equiv.), hexanol (92 ⁇ L, 1.5 equiv.), DMF (3.2 mL), CDI (93 mg, 1.2 equiv.), DBU (72 ⁇ L, 1 equiv.) and DMAP (6 mg, 0.1 equiv.) were used, the reaction was carried out in a 0.5 x smaller scale and the reaction mixture was stirred at 40 0 C for 21 h. After work up a clear oil could be obtained. The crude ester was used in the subsequent deprotection step without further purification.
  • Step P was used except of that 2-methylpentan-l-ol (90 ⁇ L, 1.5 equiv.) was used. After work up a clear oil was obtained. The crude ester was used in the subsequent deprotection step without further purification.
  • Step P was used except of that 3-heptanol (102 ⁇ L, 1.5 equiv.) was used. After work up a clear oil could be obtained. The crude ester was used in the subsequent deprotection step without further purification.
  • Method I was used except of that 31 (137 mg, 1 equiv.) and 1.1 equiv. of the rest of the reagents were used, the reaction mixture was stirred at rt for 160 min, at 40 0 C for 40 min, and the reaction was carried out in a 0.58 x smaller scale. After work up a yellowish oil could be obtained, it was used in the next reaction without further purification.
  • Step A Synthesis of methyl 3-(tetrahydropyran-2-yloxymethyl)-5-nitrobenzoate, 36
  • Step A was used except of that 35 (2.05g, 1 equiv.), DCE (20 mL), DHP (1.77 mL, 2 equiv.), PPTS (0.1 equiv.) were used, the reaction was carried out in a 0.48 ⁇ smaller scale and the reaction mixture was stirred for 25 h. The reaction was quenched by adding water (60 mL), extracted with DCM (3x60 mL), washed with saturated NaHCO 3 -solution (3x60 mL), brine (2x60 mL), dried (Na 2 SO 4 ), filtered and evaporated in vacuo to give a yellow solid (2.84 g, 99% yield).
  • Step M Synthesis of methyl 3-(tetrahydropyran-2-yloxymethyl)-5-aminobenzoate, 37
  • Method I was used except of that 37 (300 mg, 1 equiv.), undecylenic acid (251 ⁇ L, 1.1 equiv.), DCM (9 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 1.25 x larger scale, and the reaction was stirred at 40 0 C for 21 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 11 : 1-1 :2) to give a clear oil (245 mg, 50% yield).
  • Method I was used except of that 37 (600 mg, 1 equiv.), nonanoic acid (434 ⁇ L, 1 equiv.), DCM (17 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 2.5 x larger scale, and the reaction was stirred at 40 0 C for 22 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 11 : 1-1 :2) to give a clear oil (531 mg, 58% yield).
  • Method I was used except of that 37 (300 mg, 1 equiv.), heptanoic acid (177 ⁇ L, 1 equiv.), DCM (9 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 1.25 x larger scale, and the reaction was stirred at 40 0 C for 22 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 11 :1-1 :2) to give a yellow oil (178 mg, 42% yield).
  • Method I was used except of that 37 (600 mg, 1 equiv.), pivalic acid (257 ⁇ L, 1 equiv.), DCM (17 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 2.5 x larger scale, and the reaction was stirred at 40 0 C for 23 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 11 : 1-0: 1) to give a yellow oil (116 mg, 15% yield).
  • Method I was used except of that 37 (300 mg, 1 equiv.), acetic acid (71 ⁇ L, 1 equiv.), DCM (9 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 1.25 x larger scale, and the reaction was stirred at 40 0 C for 21 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-0:1) to give a yellow oil (44 mg, 13% yield).
  • Step D Deprotection of compound 5, a typical procedure
  • step D The typical procedure was performed (step D) for 5a2 except that Dowex 50W ⁇ 8 (1.2 g) was used and the reaction mixture was stirred at 40 °C for 140 min. THF and EtOAc were added to the crude reaction mixture, the Dowex 50W ⁇ 8 was filtered off and the solvents were evaporated in vacuo. The crude product was recrystallised from hexane to give Ia2 (124 mg, 43% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5a3 except of that Dowex 50W ⁇ 8 (6.5 g) and MeOH (50 rnL) were used and the reaction mixture was stirred for 23 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 8%-66%) to give a white solid, Ia3 (765 mg, 42% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5a4 except that Dowex 50W ⁇ 8 (1.2 g) was used. EtOAc was added to the reaction mixture and the Dowex 50W ⁇ 8 was filtered off. The solvents were evaporated in vacuo and the crude product was recrystallised from hexane/CHCl 3 /EtOAc to give a white solid, Ia4 (146 mg, 44% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5a5 except that Dowex 50W ⁇ 8 (1.5 g) and MeOH (1OmL) were used and the reaction mixture was stirred for 6 h.
  • the crude product was purified with SiO 2 column chromatography (hexane/EtOAc, 2:1) giving a white solid, Ia5 (293 mg, 92% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5a6 but the reaction mixture was stirred for 4 h.
  • the crude product was purified with SiO 2 column chromatography (hexane/EtOAc, 2:1) giving a white solid, Ia6 (112 mg, 41% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5a7 except of that Dowex 50W ⁇ 8 (2.0 g) was used.
  • the crude product was purified first with SiO 2 column chromatography (hexane/EtOAc, 2:1) to give a white solid, Ia7 (125 mg, 29% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5bl except that Dowex 50W ⁇ 8 (1.1 g) was used.
  • the crude product was purified with SiO 2 column chromatography (hexane/EtOAc, 2:1) to give a clear oil, IbI (233 mg, 72% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5a8 except that Dowex 50W ⁇ 8 (2.0 g) and MeOH (20 mL) were used.
  • the crude product was purified with SiO 2 column chromatography (hexane/EtOAc, 2:1) to give a clear oil, Ia8 (89 mg, 39% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5b2 except of that Dowex 50W ⁇ 8 (2.5 g) and MeOH (20 mL) were used.
  • the crude product was purified with SiO 2 column chromatography (hexane/EtOAc, 2:1) to give a white solid, Ib2 (242 mg, 35% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5b3 except of that Dowex 50W ⁇ 8 (700 mg) and MeOH (8 rnL) were used and the reaction mixture was stirred at 40 0 C for 20 h.
  • step D The typical procedure was performed (step D) for 5b4 except of that Dowex 50W ⁇ 8 (1.1 g) and MeOH (10 mL) were used.
  • the crude product was purified with SiO 2 column chromatography (hexane/EtOAc, 2:1) to give a clear oil, Ib4 (81 mg, 31% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5b5 except of that Dowex 50W ⁇ 8 (1 g) and MeOH (12 mL) were used and the reaction mixture was stirred for 18 h.
  • the crude product was purified with SiO 2 column chromatography hexane-EtOAc (4:1-2:1) to give a clear oil, Ib5 (328 mg, 63% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5b6 except of that Dowex 50W ⁇ 8 (2.8 g) and MeOH (20 mL) were used.
  • the crude product was purified with SiO 2 column chromatography hexane-EtOAc (2:1) to give a clear oil, Ib6 (56 mg, 8% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5b7 except of that Dowex 50Wx8 (700 mg) and MeOH (8 mL) were used and the reaction mixture was stirred for 20 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 2:1) to give a clear oil, Ib7 (199 mg, 61% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5b8 except of that Dowex 50W ⁇ 8 (3.6 g) and MeOH (10 rnL) were used and the reaction mixture was stirred for 21 h.
  • the crude product was purified with SiO 2 column chromatography (CHCl 3 -MeOH; 10:1) to give a clear oil, Ib8 (42 mg, 17% yield for two reaction steps).
  • 1H NMR 300 MHz, CDCl 3 ) ⁇ 8.57 (app.
  • step D The typical procedure was performed (step D) for 5b9 except of that Dowex 50W ⁇ 8 (3.6 g) and MeOH (10 mL) were used and the reaction mixture was stirred for 24 h.
  • the crude product was purified with SiO 2 column chromatography (CHCl 3 -MeOH; 10:1) to give a clear oil, Ib9 (145 mg, 55% yield for two reaction steps).
  • 1H NMR 300 MHz, CDCl 3 ) ⁇ 8.54 (s, IH), 8.19 (app. t, 2H), 4.78 (s, 2H), 4.35 (t, 4H,
  • step D The typical procedure was performed (step D) for 5cl except of that Dowex 50W ⁇ 8 (3.6 g) MeOH (10 mL) and THF (5 mL) were used and the reaction mixture was stirred for 18 h.
  • the crude product was purified with SiO 2 column chromatography (CHCl 3 -MeOH; 10:1) to give a clear oil, IcI (63 mg, 22% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5b 10 except of that Dowex 50W ⁇ 8 (700 mg), MeOH (8 mL) and THF (1 mL) were used and the reaction mixture was stirred for 20 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 8:1) to give a white solid, IbIO (239 mg, 57% yield for two reaction steps).
  • 1H NMR 300 MHz, CDCl 3
  • 8.19 app.
  • step D The typical procedure was performed (step D) for 5b 11 except of that Dowex 50W ⁇ 8 (2 g), MeOH (10 mL) and DCM (2 mL) were used and the reaction mixture was stirred for 17 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 4:1) to give a clear liquid, lbll (210 mg, 54% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5bl2 except of that Dowex 50W ⁇ 8 (600 mg) and MeOH (6 mL) were used and the reaction mixture was stirred for 19 h.
  • the crude product was purified with SiO 2 column chromatography (CHCl 3 -MeOH; 20:1-4:1) to give a clear oil, Ibl2 (20 mg, 10% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5bl3 except of that Dowex 50W ⁇ 8 (700 mg), MeOH (8 mL) and THF (1 mL) were used and the reaction mixture was stirred for 20 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 8:1) to give a white solid, Ibl3 (296 mg, 71% yield for two reaction steps).
  • 1H NMR 300 MHz, CDCl 3
  • ⁇ 8.55 (app.
  • step D The typical procedure was performed (step D) for 5bl4 except of that Dowex 50W ⁇ 8 (2 g) and MeOH (10 mL) were used, the reaction mixture was stirred for 17 h and after work up a clear oil could be obtained, Ibl4.
  • step D The typical procedure was performed (step D) for 5b 15 except of that Dowex 50W ⁇ 8 (700 mg), MeOH (8 mL) and THF (2 mL) were used and the reaction mixture was stirred for 21 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 8:1) to give a white solid, Ibl5 (100 mg, 32% yield for two reaction steps).
  • 1H NMR 300 MHz, CDCl 3
  • ⁇ 8.57 app. t, IH
  • 8.18 app.
  • step D The typical procedure was performed (step D) for 5b 16 except that the reaction was carried out in a 2 x larger scale and the reaction mixture was stirred for 20 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 4:1) to give a clear oil, Ibl6 (113 mg, 39% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5b 17 and the reaction mixture was stirred for 21 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 8:1) to give a clear oil, Ibl7 (171 mg, 53% yield for two reaction steps).
  • 1 H NMR 300 MHz, CDCl 3 ) ⁇ 8.58 (app. t, IH), 8.21 (app.
  • step D The typical procedure was performed (step D) for 5b 18 except of that Dowex 50W ⁇ 8 (700 mg), MeOH (8 mL) and THF (1 mL) were used and the reaction mixture was stirred for 23 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 8:1- 4:1) to give a white solid, Ibl8 (199 mg, 54% yield for two reaction steps).
  • step D The typical procedure was performed (step D) for 5b 19 and the reaction mixture was stirred for 23 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 8:1-4:1) to give a white solid, Ibl9 (263 mg, 81% yield for two reaction steps).
  • 1H NMR 300 MHz, CDCl 3 ) ⁇ 8.59 (app.
  • step D The typical procedure was performed (step D) for 5b20 and the reaction mixture was stirred for 23 h.
  • the crude product was purified with SiO 2 column chromatography (hexane-EtOAc; 8:1-4:1) to give a white solid, Ib21 (165 mg, 51% yield for two reaction steps).
  • Step L - Method H Synthesis of ⁇ /s(2-methylpentyl)isophthalate, 15a, a typical procedure
  • Step C isophthalic acid 14a (200 mg, 1 equiv.), CDI (2 equiv.), DMF (8 niL), DBU (2 equiv.), DMAP (0.05 equiv.) and 2-methyl-pentanol (3 equiv.) were used and the reaction was carried out in a 1.3 x larger scale.
  • the reaction mixture was stirred at 40 °C for 20 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 4:1) to give clear oil (42 mg, 11% yield).
  • Step M Synthesis of f ⁇ s(2-methylpentyl)-5-aminoisophthalate, 15f
  • step D The typical procedure for deprotection was used (step D) for 16a except that the reaction mixture was stirred for 21 h. After work up, 1 M HCl- solution (20 rnL) was used to dissolve the solid, it was washed with DCM (2x20 mL), made basic with NaHCO 3 and water (20 mL) added. Extracted with DCM (3x20 mL), dried (Na 2 SO 4 ), filtered and evaporated in vacuo to give a reddish solid. It was recrystallized from ether to give a pale red solid. 1 H NMR (300 MHz, CDCl 3 ) ⁇ 7.53 (app. t, 2H), 7.38 (app.
  • step D The typical procedure for deprotection was used (step D) for 16b except that the reaction mixture was stirred for 23 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 2: 1-0: 1) to give a white solid (179 mg, 39% yield for two steps).
  • 1 H NMR 300 MHz, CD 3 OD
  • ⁇ 8.25 8.25 (app.
  • step D The typical procedure for deprotection was used (step D) for 16c except that the reaction mixture was stirred for 24 h.
  • the crude product was purified with flash column chromatography (50% EtOAc in hexane -> 100% EtOAc) to give a white solid (62 mg, 28% yield for two steps).
  • Heptanoyl chloride (410 ⁇ L, 2.2 equiv.) was dropwise added under argon to a solution of 19 (200 mg, 1 equiv.), DIPEA (453 ⁇ L, 2.2 equiv.) and DCM (2 mL) and the reaction mixture was stirred at rt for 21 h. The reaction was quenched by adding DCM (15 mL) and ice-water (10 mL), the mixture was washed with water (2x10 mL), 1 M HCl-solution
  • Step D was used for 28a except of that the reaction was carried out in a 0.4 x smaller scale and the reaction mixture was stirred at 40 0 C for 17 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-1 :1) to give clear oil (85 mg, 75% yield for two steps).
  • 1 H NMR 300 MHz, CDCl 3 ) ⁇ 8.02 (s, IH), 7.97-7.93 (m, IH), 7.57-7.55
  • Step D was used for 28b except of that the reaction was carried out in a 0.4 x smaller scale and the reaction mixture was stirred at 40 0 C for 17 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-2:1) to give clear oil (77 mg, 68% yield for two steps).
  • Step D was used for 28c except of that the reaction was carried out in a 0.4 x smaller scale and the reaction mixture was stirred at 40 0 C for 17 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-2:1) to give clear oil (78 mg, 65% yield for two steps).
  • Step B was used except of that diethyl isophthalate 2 (252 mg, 1 equiv.) was used, the reaction was carried out in a 0.1 x smaller scale and it was refluxed for 2 h.
  • the solvents were evaporated in vacuo, water (20 mL) added to the oil, washed with EtOAc (2x20 mL) and the pH of the aqueous phase was adjusted with 1 M HCl- solution to 1. Extracted with EtOAc (3x20 mL), washed with brine (4x20 mL), dried (Na 2 SO 4 ), filtered and evaporated in vacuo to give a white solid (170 mg, 87% yield).
  • Method D was used except of that 32 was used and the reaction mixture was stirred at 40 0 C for 23 h.
  • the crude product was purified with flash column chromatography (hexane- EtOAc; 2:1-0:1) to give a clear oil. NMR showed -16% ethyl ester and 84% methyl ester (71 mg, 41% yield for two steps).
  • Method D was used for 38a except of that the reaction was carried out in a 0.3 x smaller scale, and the reaction was stirred at 40 0 C for 21 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 2:1-0:1) to give a yellow solid (28 mg, 46% yield for two steps).
  • Method D was used for 38b except of that the reaction was carried out in a 0.71 x smaller scale, and the reaction was stirred at 40 0 C for 21 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 2:1-1 :1) to give a white solid (57 mg, 73% yield). ).
  • Method D was used for 38c except of that the reaction was carried out in a 0.71 x smaller scale, and the reaction was stirred at 40 0 C for 19 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-1 :1) to give a white solid (94 mg, 49% yield for two steps).
  • Method D was used for 38d except of that the reaction was carried out in a 0.71 x smaller scale, and the reaction was stirred at 40 0 C for 24 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-1 :1) to give a white solid (68 mg, 77% yield).
  • Method D was used for 38e except of that the reaction was carried out in a 0.71 x smaller scale, and the reaction was stirred at 40 0 C for 24 h.
  • the crude product was purified with flash column chromatography (hexane-EtOAc; 1 :1-0:1) to give a white solid (28 mg, 89% yield).
  • 1 H NMR 300 MHz, CD 3 OD
  • 7.81 app.
  • the ability of the compounds of invention to replace radioactively labelled phorbol-12,13- dibutyrate ([ 3 H]PDBu) from binding to recombinant PKC was assessed. Binding assays for the PKC targeted compounds of the invention were performed using five different concentrations (0.3 ⁇ M; 1 ⁇ M; 3 ⁇ M; 10 ⁇ M and 20 ⁇ M), each with three replicates on the same plate. Binding of [ H]PDBu to PKC was tested using human recombinant PKC ⁇ and PKC ⁇ that had been produced in Sf9 insect cells.
  • PKC isoforms are mainly localized in the cytosol.
  • DAG diacylglycerol
  • PKCs translocate from the cytosol to the plasma membrane or other intracellular compartments to phosphorylate their substrates.
  • Translocation is an important step in the activation process, since it takes the enzyme to close proximity with its substrates and other proteins with which it can interact. Translocation is fairly easy to visualize with confocal microscopy by overexpressing PKC constructs that contain a fluorescent tag, e.g. GFP (green fluorescent protein), in cultured cells.
  • GFP green fluorescent protein
  • the translocation of PKC was visualized in HeLa cells transfected with PKC-GFP constructs using confocal microscopy. During the translocation experiments the cells were maintained in DMEM in 37 0 C. When the effects of the compounds of the invention on PKC localization were studied, the cells were treated with the compounds and images were captured every 30 s for 30 min. Confocal images were captured with Leica TCS SP2 AOBS confocal microscope using a 63 ⁇ oil objective with an excitation wavelength at 488 nm and emission wavelength at 500-570 nm.
  • HeLa cells expressing PKC- (GFP) constructs were pretreated for 20 to 30 minutes with the compounds of the invention and stimulated with 100 nM PMA (phorbol myristate acetate). Images were captured before stimulating the cells with PMA and after PMA addition every 30 seconds for 30 minutes.
  • PMA phorbol myristate acetate
  • the translocation was quantified from confocal microscopic images captured during the experiments using Leica Confocal Software. The intensity of fluorescence was measured in a region of interest chosen within the cell cytoplasm (only one region of interest / cell). The area to be quantified was chosen not to contain any plasma membrane or intracellular structures throughout the experiment.
  • MTT test measures the enzymatic activity of mitochondria: NADH and NADPH that are formed by mitochondria reduce the yellow methyltiazoletetrazolium (MTT) into blue insoluble formazan crystals. Only the mitochondria of living or early apoptotic cells are able to metabolize MTT into the blue formazan. Therefore the cytotoxicity of a compound can be detected with MTT test already before the cell membrane is damaged.
  • LDH test measures the amount of lactate dehydrogenase released from damaged cells. LDH is an enzyme that is localized in the cytosol of all mammalian cells. In cell culture, when the cell is damaged, LDH is released into the culture medium, from which its activity can be measured.
  • the cells were plated on 96-well plates and 24 hours later the medium was changed to medium containing the test compounds. After that the cells were grown in the presence of the compounds in humidified atmosphere (37 0 C, 5 % CO 2 ) for 24 or 48 hours before the tests were carried out.
  • humidified atmosphere 37 0 C, 5 % CO 2
  • ⁇ L of culture medium was transferred from treated cells into a new 96- well plate and 50 ⁇ L of substrate solution (1.3 mM ⁇ -nicotineamide adeninedinucleotide ( ⁇ -NAD); 660 ⁇ M iodonitrotetrazolium (INT); 54 mM L(+)-lactic acid; 280 ⁇ M phenazine methosulphate in 0.2 M Tris-buffer; pH 8.2) was added to all wells. The plate was incubated at room temperature for 30 minutes and reaction was stopped by adding 50 ⁇ L of 1 M acetic acid. The absorbance was measured at 490 nm.
  • substrate solution 1.3 mM ⁇ -nicotineamide adeninedinucleotide ( ⁇ -NAD); 660 ⁇ M iodonitrotetrazolium (INT); 54 mM L(+)-lactic acid; 280 ⁇ M phenazine methosulphate in 0.2 M Tris-buffer; pH 8.2
  • MTT test MTT was added to the cells at 0.5 mg/mL concentration and the cells were allowed to metabolize it in 37 0 C, 5 % CO 2 for 2 hours. The medium was then aspirated and the blue formazan crystals were dissolved in DMSO and the absorbance was measured at the wavelength 550 nm and the absorbance at 650 nm was extracted as background.
  • the compounds of the invention showed a concentration-dependent cytotoxic effect in HeLa cells as determined with MTT test, but not with LDH test ( Figures 5 and 6). This might be due to apoptotic cell death, which would impair mitochondrial metabolism but would not damage cell membrane. Preliminary results from flow cytometric studies support apoptosis as the mechanism of cell death, but other mechanisms of cell death can not be excluded.
  • HL60 cells were cultured in RPMI 1640 medium containing 10% fetal calf serum with or without 10 ⁇ g/mL of one of the compounds Ib3, Ib5 or Ib7.
  • Apoptosis was measured after 2 days by PI staining and assessment of the percentage of cells in the Sub-Go/Gl peak by flow cytometry. All three compounds induced apoptosis to some extent but Ib5 was the most effective ( Figure 8A).
  • Cells were also assessed for induction of the cell surface marker CDl Ib, which indicates differentiation of the cells toward either monocytes or granulocytes. Compound Ib5 did not induce differentiation, but Ib3 and Ib7 were effective indcuers of the marker of differentiation CDl Ib ( Figure 8B).
  • CD34 positive leukaemic cell blasts were isolated from either blood or bone marrow of patients with acute myeloid leukaemia.
  • Cells were cultured in medium containing IL3 and SCF to maximise survival and mimic conditions in vivo.
  • Compounds IbS and Ib? were added to cultures at 1 and 10 ⁇ g/mL and apoptosis assessed after 2 days by P! staining and percentage of cells in the sub-Go/Gl peak by flow cytometry. 75% of cells were sensitive to compounds IbS and Ib7 at 10 ⁇ g/mL ( Figure 9).
  • B-CLL cells were obtained from the blood of patients with B-CLL.
  • Normal B- cclls were obtained from tonsils of patients undergoing tonsilectomy.
  • Cells were cultured in the absence of presence of a range of concentrations of compound IbS and apoptosis measured by presence of active caspasc 3 by immunostaining and flow cytometry.
  • Compound IbS induced apoptosis in the majority of B-CLL cells at 5 and 10 ⁇ g/mL (Figure 10A) but did not kill ordinary B cells (Figure 10B).
  • B-CLL cells were also cultured in the presence of stromal cells that were unaltered, or were expressing the B cell survival factor CD40L and the effect of 5 or 10 ⁇ g/rnL of compounds 1b3, 1b5 or 1b7 on apoptosis measured as above.
  • Compound IbS was able to induce apoptosis in the presence of stromal cells ( Figure 1 IA) and in the presence of stromal cells expressing CD40L ( Figure 1 I B).
  • T cells were activated and cultured in the absence (white bars) or presence of IL2 (black bars) ( Figure 12 A and 12B), which provides survival signals at sites of inflammation, and a range of concentrations of 10 different compounds.
  • Apoptosis was measured by expression of active caspase 3. Most of the compounds could induce apoptosis in T cells in the presence of 1L2, but compound IbI was the most effective, abolishing the rescue effect of IL2 at 1 ⁇ g/mL ( Figure 12A).
  • HL-60 Human promyelocytic leukaemia cells
  • mouse fibroblasts Swiss 3T3; University of Bergen, Norway
  • DNA binding stain was used to observe chromatin degradation and to count the ratio of apoptotic cells by fluorescence microscopy. Typical apoptotic events, dissipation of ⁇ m and increase of caspase-3 activity, were observed in the HL-60 cells treated with the most promising compounds.
  • Cytotoxicity by necrosis was determined using the colorimetric assay, which quantitatively measures lactate dehydrogenase (LDH), a stable cytosolic enzyme that is released upon cell lysis into the supernatants. Mutagenicity of the most promising compounds was examined with a miniaturized Ames- test.
  • LDH lactate dehydrogenase
  • HL-60 cells were maintained in RPMI 1640 supplemented with 10% heat inactivated (56 °C for 30 min) FBS, 100 ILVmL penicillin G, and 100 ⁇ g/mL streptomycin at 37 °C in an atmosphere with 5% CO 2 , 95% air and 95% relative humidity.
  • Swiss 3T3 fibroblasts were maintained in the same way but DMEM was used instead of RPMI 1640. Cells were split every two to three days as necessary.
  • HL-60 leukaemia cells and Swiss 3T3 fibroblasts were harvested from the cell culture by centrifugation, 200 rcf, 4 min (Sigma 2- 5, Germany) and resuspended into fresh medium. The number of cells was determined using a haemocytometer and the cell suspension was then diluted to get 60,000 cells/mL in fibroblast experiments and 150,000 cells/mL in leukaemia cell experiments.
  • Fibroblast cells were seeded in 48-well plates (250 ⁇ L/well) and leukaemia cells were seeded in 96- well plates (100 ⁇ L/well). Fibroblasts were incubated overnight before changing fresh media and adding 2.5 ⁇ L test compounds. In leukaemia cell experiments 1.0 ⁇ L test compounds were added to plates before cell suspension. The test compounds were dissolved into DMSO and then diluted with media so that DMSO concentration did not exceed 0.1%. Camptothecin (0.014 ⁇ M) was used as a positive control and 0.1% DMSO was used as a negative control. Cells were incubated with test compounds for 24 h at 37 °C in a humidified atmosphere with 95% air and 5% CO 2 .
  • JC-I Changes in mitochondrial membrane potential ( ⁇ m) were detected by flow cytometry with JC-I staining.
  • JC-I is a cationic and lipophilic dye, which is able to enter mitochondria and form aggregates when the ⁇ m is high. As aggregates, JC-I emits light at a wavelength of 590 nm and as a monomer at 527 nm, when excited at 490 nm.
  • HL-60 cells (1 mL, 100,000 cells/mL) were incubated with 40 ⁇ M Ia3 or 40 ⁇ M IbI for 2 h. The concentration, 40 ⁇ M, used in the experiment was selected according to preliminary experiments. Positive control cells were incubated with 10 ⁇ M valinomycin for 10 min.
  • the HL-60 cells were incubated with JC-I (10 ⁇ g/mL) for 15 min at room temperature. Then the cells were washed twice with cold PBS (5 min, 400 rcf) and resuspended in 400 ⁇ L of PBS. The fluorescence intensity of the cells was measured immediately with Becton Dickinson FACScan flow cytometer (Immunocytometry Systems, San Jose, CA) equipped with a single 488 nm argon laser. Fluorescence was collected through 530/30BP and 585/42BP filters. Forward scatter and side scatter were collected by using linear amplification and the fluorescence was collected by using log amplification. A minimum of 10,000 cells per sample were analyzed. The data was acquired in list mode and analyzed with Becton Dickinson CellQuest software.
  • caspase-3 The activity of caspase-3 was detected by commercial EnzChek Caspase-3 Assay Kit according to the manufacturer's instructions. Briefly, ImL of HL-60 cells (100,000/mL) was incubated with 50 ⁇ M Ia3 or 50 ⁇ M IbI for 0.5, 1, 1.5, and 2 h. The concentration (50 ⁇ M) for Caspase-3 assay was selected according to preliminary experiments. Positive control cells were incubated with 5 ⁇ M camptothecin. Cells were collected (5 min, 400 rcf) and washed with PBS. Then the cells were lysed in kit's lysis buffer in an ice bath for 30 min, and cellular debris was pelleted by centrifugation (5 min, 4,500 rcf).
  • Lactate dehydrogenase leakage from necrotic cells was determined using the commercial colorimetric cytotoxicity assay CytoTox 96 ® according to the manufacturer's instructions. Cells were harvested as described before. The cell density was 100,000 cells/mL in fibroblast experiments and 1 500,000 cells/mL in leukaemia cell experiments. Both cell types were incubated with test compounds for 3 h at concentrations of 20, 60, and 100 ⁇ M. A total volume of 100 ⁇ L of cell suspension was used in each well. Absorbance data were collected using a 96-well plate reader (Victor 1420 multilabel counter, PerkinElmer Life and Analytical Sciences/ Wallac Oy, Turku, Finland) at 490 nm. All assays were performed using 96-well plates with four replicates in the same plate and repeated at least two times in different days.
  • Genotoxic potential was evaluated with a miniaturized version of the Ames-test done in 6- well plates [Flamand et al, 2001] at a concentration of 370 ⁇ M. The concentration was chosen to be high to be sure that compounds are not mutagenic.
  • Genotyped Salmonella typhimurium TA98 and TAlOO (Xenometric Inc., San Diego, CA) strains were used. Histidine requirement, presence or absence of R- factor plasmid, spontaneous reversion, and rfa mutation were tested with genotyped strains before use. Strains were grown for 24 h at 37 °C in nutrient broth (Becton Dickinson, Le Pont de Claix, France) before the test.
  • 2-Aminoanthracene was used as a positive control 0.5 and 0.75 ⁇ g/well for strains TA98 and TAlOO, respectively, in the presence of 10% S9 rat liver enzyme-mix.
  • the absence of metabolic activation by S9 fraction, 2-nitrofluorene, was used as a positive control 0.1 ⁇ g/well for strain TA98 and natriumazide 0.5 ⁇ g/well for strain TAlOO.
  • Ia3 and IbI Apoptosis-inducing activity of Ia3 and IbI was studied further: Dose-response relationships of Ia3 and IbI were studied in leukaemia cells and IC50 values were determined. Ia3 showed inhibition with an IC50 of 41 ⁇ M and IbI with an IC50 of 23 ⁇ M (data not shown). Chromatin condensation in HL-60 cells incubated with 20 ⁇ M Ia3 and IbI was then observed through various time points. Compounds induced apoptosis rapidly; after 2-h exposure more than 30% of cells were apoptotic ( Figure 14). The Mini- Ames test did not show any mutagenicity of either compound. Cell growths on both bacterial strains were on the same level with test compounds as with the negative controls.
  • the compounds of the present invention have proven to be potent apoptosis inducers in leukaemia cells.
  • compounds Ia3 and IbI have IC50 values of 41 and 23 ⁇ M, respectively, according to morphological evaluation. Changes in mitochondrial membrane potential and in caspase-3 activity confirmed the results. Apoptosis was induced already during the first 2 h incubation with these compounds in HL-60 cells. Although necrosis and apoptosis have some common steps, it is general agreement that apoptosis, in contrast to necrosis, is an effective energy-requiring process. Dissipation of mitochondrial membrane potential and release of cytochrome c from mitochondria appear to be key events during apoptosis. Modulators of caspase activity are increasingly gaining interest as potential targets for drug development.
  • the compounds showing a clear apoptosis-inducing activity on cancerous HL-60 leukaemia cells also were safe to non-malignant Swiss 3T3 fibroblasts.
  • Two main problems of chemotherapy are toxicity to normal cells and failure to kill cancer cells.
  • SCID mice Animal Model Severe combined immunodeficiency mice engrafted with B cell leukaemic cells are employed for the study for the in vivo evaluation of the compounds of the invention.
  • mice are divided into 3 equal treatment groups of 10 mice each as follows: 1) a control receives placebo (1% DMSO); 2) a positive control receives rituximab at 5mg/kg; 3) the remaining group receives compound Ib5 delivered at 50mg/kg via the tail vein and maintained every other day intravenously for 2 weeks (7 injections for each mouse).
  • the body weight of the animals is measured once every week and tumour burden is assessed by in vivo imaging of the GFP expressing cells. All the animals are monitored daily for signs of illness and are killed immediately if hind- limb paralysis, respiratory distress, or 30% body weight loss occurs.
  • the endpoints of the study are tumour burden (assessed by harvesting GFP positive cells from major lymphoid organs and bone marrow) and survival defined as the time for the development of hind-limb paralysis.
  • Administration may be by any means generally understood in the art, including, but not limited to the types of administration described herein.
  • the major lymphoid organs are removed (thymus, spleen, major lymph nodes, intestinal lymph nodes, liver) and disaggregated to allow isolation of B cells by their binding to an anti- lymphoid organs.
  • CDl 9 antibody attached to Dynabeads.
  • the long bones are flushed gently with saline to remove the bone marrow and the CD 19 positive cells are isolated.
  • the fraction of leukaemic GFP positive cells in each tissue is determined by flow cytometry.
  • the disease is expected to be inhibited in its progression in those animals receiving both the rituximab and the Ib5 compound.
  • the inhibition or prevention may be based on the quantity of leukaemic cells (tumor burden) or survival time of the animals beyond inoculation with the leukaemic cells.

Abstract

The present invention concerns a compound, which modulates protein kinase activity, having the following Formula (I) or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, wherein R1, R2 and R3 may be the same or different and are each independently chosen from hydrogen, branched or linear alkyl, branched or linear hydroxyalkylene, nitro, amino, -C(=O)OR4, -C(=O)NR5R6, and -NHC(=O)R7, as well as compositions comprising the compound as an active agent and use of the compounds and the compositions in manufacturing medicaments for regulating protein kinase activity in a subject by administering to the subject a composition comprising a compound of the invention or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, in an amount effective to regulate the protein kinase activity.

Description

PROTEIN KINASE MODULATING AGENTS
The present invention concerns new protein kinase modulating agents. Particularly, but not exclusively, the present invention concerns protein kinase C translocation inhibitors. The agents described here affect signal transduction pathways regulating protein phosphorylation, which leads to modulation of apoptosis. They also possess antileukaemic and anti-inflammatory properties.
Background
New apoptosis modulating agents are widely sought, because failure in regulation of apoptosis is associated with many diseases. Apoptosis, programmed cell death, is a genetically coded mechanism to destroy unwanted cells. Apoptosis plays an important role in development, in tissue homeostasis maintenance and in defence against viral infections and mutations. Apoptosis is a rapid process and causes no tissue damage, since apoptotic cells are rapidly phagocytosed by other cells.
Caspases, a family of intracellular cysteine proteases, have a central role in apoptosis. Caspases are present in cells as inactive procaspases, which can be rapidly converted to active caspases. After pro-apoptotic signal, initiator caspases (caspase-8 and caspase-9) are initially activated, and initiator caspases then activate precursor forms of effector caspases (caspase-3, -6 and -7) to active caspases. Effector caspases cleave several cellular proteins with essential functions leading to apoptotic phenotype. Apoptosis is characterized by morphological features such as chromatin condensation, nuclear fragmentation, cell membrane blebbing and formation of apoptotic bodies. Caspases can be activated by distinct pathways, the death receptor pathway and the mitochondrial pathway. In the mitochondrial pathway of apoptosis, pro-apoptotic signal-transducing molecules and pathological stimuli cause mitochondrial outer membrane permeabilization, which in turn induces apoptosis further, since caspase-activating molecules and caspase-independent death effectors are released from the space between the inner and the outer membrane of mitochondria. Dissipation of the mitochondrial inner transmembrane potential (ΔΨm) is frequently associated with the mitochondrial pathway of apoptosis. Defective apoptosis regulation is associated with many diseases. Insufficient apoptosis occurs for example in cancer and autoimmune diseases. In contrast, excessive apoptosis is associated with heart failure, neurodegenerative diseases and AIDS (Acquired Immune Deficiency Syndrome). Thus, new apoptosis modulating molecules are widely sought from natural products and from synthetic compounds.
It has been demonstrated that caspase-3 -dependent proteolytic activation of PKCδ plays a key role in oxidative stress-mediated apoptosis in dopaminergic cells after exposure to an environmental neurotoxic agent. Recent studies have indicated protein kinase C delta (PKCδ) to be one of the endogenous substrates for caspase-3, which cleaves the kinase. Activation of caspases by cytosolic cytochrome C is an early and essential step in the apoptotic-signalling pathway. There is also indication that caspase-3 plays a major role in the regulation and execution phase of both in vitro and in vivo models of apoptosis.
Protein kinases play an important role in many signalling pathways in cells. Protein kinase C (PKC) is a family of serine/threonine kinases that regulate various cellular processes including cell proliferation and malignant transformation.
In the present invention, using the known X-ray structure of mouse PKC delta CIb domain and the solution structure of the same domain from rat PKC gamma with bound phorbol ester as a template for molecular modelling, isophthalic acid-derived compounds, which compete with phorbol esters for binding and thus modulate PKC activity, have been designed and synthesized. They have high affinity for the diacyl glycerol binding site of PKC and modulate the function of PKC. The compounds may bind also to other site on PKC and modify phorbol ester binding indirectly via conformational change of phorbol ester binding site. Thus, it is possible for these agents to modify the activity of other protein kinases than PKC as well.
Phorbol esters, as mentioned above, are potent cocarcinogens, activating various PKC- isoenzymes and thereby acting as tumour promoters.
Figure imgf000005_0001
Figure imgf000005_0002
Phorbol myristate acetate (PMA) (typical phorbol ester)
The compounds of the present invention compete with, e.g., phorbol esters at the PKC binding site, thus preventing the tumour promoting activity of the phorbol esters.
The compounds of the invention have been tested in in vitro receptor binding, enzymatic activity assays (some inhibitors with IC50 around 10 μM), an intracellular translocation assay using confocal microscopy of HeLa cells, and in leukaemia cell lines, such as HL-60, and primary cell lines from patients with acute myeloid leukaemia (AML) or chronic lymphatic leukaemia (CLL). The compounds behave as either activators or inhibitors of the PKC isoenzymes.
Leukaemia is cancer of the blood cells. Like most cancer cells, leukaemic cells show a variety of alterations in genes controlling cell proliferation (e.g. Flt3, cyclinD) or apoptosis (bcl2, abl). The inventors have discovered that in particular, two isoenzymes of PKC, PKCα and PKCδ, play specific roles in tumour promotion and suppression. The compounds of the present invention show therapeutic potential for, among others, leukaemia, in particular for acute myeloid leukaemia (AML) and chronic lymphatic leukaemia (CLL). Further, the inventors have discovered that the compounds of the invention possess concentration dependent anti-inflammatory activity in cultured human lymphocytes.
The invention provides new compounds that are potent anticancer and anti-inflammatory agents and are reasonably safe and non-toxic. There is a global unmet need for such compounds. The commercial potential of protein kinase modulating agents including PKC modulating, anti-inflammatory and antileukaemic agents is obvious. The incidence, for example, of rheumatoid arthritis is about 1% world wide, which means there are about 60 000 000 people suffering from the disease in the world. Further, every year tens of millions of people around the world die from cancerous diseases.
The synthetic preparation of the protein kinase modulating anti- inflammatory and antileukaemic agents described herein is straightforward and relatively easy.
Summary of the invention
The aim of the present invention is to provide new simple-to-synthesize phorbol-ester displacing compounds that modulate the function of protein kinases, especially that of PKC, and thus prevent unwanted cell division. Of particular interest for the present invention are compounds that function as drugs and target the Cl domain of PKC isoenzymes α and δ.
The invention provides compounds that regulate protein kinase activity, such as PKC activity. These compounds are hydrophobic derivatives of isophthalic acid, particularly esters of isophthalic acid. In particular, the present invention concerns compounds that modulate protein kinase activity and have the following Formula I:
Figure imgf000006_0001
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, wherein
R1, R2 and R3 may be the same or different and are each independently chosen from hydrogen, branched or linear alkyl, branched or linear hydroxyalkylene, nitro, amino,
C(=O)OR4, -C(=O)NR5R6, and -NHC(=O)R7, wherein
R4 is chosen from hydrogen, a branched or linear, saturated or unsaturated Ci-Ci6 alkyl chain, a branched or linear, saturated or unsaturated Ci-Ci6 alkoxy chain, an aliphatic or aromatic, unsubstituted or substituted, mono-, di- or tricyclic ring structure connected to the -C(=O)O- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear Ci-Ci6 alkyl, branched or linear Ci-Ci6 alkoxy, branched or linear hydroxyalkylene, mono-, di- or trihalogeno- Ci-Ci6 alkylene, and wherein any of the groups R4 may possibly contain a heteroatom at any position of the group, chosen from O, S and N;
R5 and R6 may be the same or different and are each independently chosen from hydrogen, a branched or linear, saturated or unsaturated Ci-Ci6 alkyl chain, a branched or linear, saturated or unsaturated Ci-Ci6 alkoxy chain, an aliphatic or aromatic, unsubstituted or substituted, mono-, di- or tricyclic ring structure connected to the -C(=O)N- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear Ci-Ci6 alkyl, branched or linear Ci-Ci6 alkoxy, branched or linear hydroxyalkylene, mono-, di- or trihalogeno- Ci-Ci6 alkylene, wherein any of the groups R5 and R6 may possibly contain a heteroatom at any position of the group, chosen from O, S and N; alternatively, R5 and R6 may be combined to form an aliphatic or aromatic, substituted or unsubstituted, mono-, di- or tricyclic ring structure connected to the -C(=O)N- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, 0 to 3 ring(s) may be aliphatic, whereas the remaining 0 to 3 ring(s) may be aromatic, and the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the substituents are chosen from the ring substituents mentioned above, and wherein the ring structure possibly also contains a further heteroatom chosen from O, S and N; and
R7 is chosen from hydrogen, a branched or linear, saturated or unsaturated Ci-Ci6 alkyl chain, a branched or linear, saturated or unsaturated Ci-Ci6 alkoxy chain, an aliphatic or aromatic, unsubstituted or substituted, mono-, di- or tricyclic ring structure connected to the -NHC(=O)- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear Ci-Ci6 alkyl, branched or linear Ci-Ci6 alkoxy, branched or linear hydroxyalkylene, mono-, di- or trihalogeno- Ci-Ci6 alkylene, wherein any of the groups R7 may possibly contain a heteroatom at any position of the group, chosen from O, S and N; with the provisos that only one of R1, R2 and R3 may be hydrogen, only one of R1, R2 and R3 may be -C(=O)OC2H5, and if one of R1, R2 and R3 is a -C(=O)OC2H5 group, none of the other groups R1, R2 and R3 may be -C(=O)OCH3.
More specifically, the compounds of the present invention are characterized by what is stated in Claim 1.
Further, the composition of the present invention is characterized by what is stated in Claim 18 and the use of the present invention is characterized by what is stated in Claims 26 and 28.
The compounds of the present invention are useful particularly in the treatment and prevention of inflammatory and cancerous diseases by modulating the function of PKC isoenzymes α and δ as well as in the treatment and prevention of Down's syndrome by inhibiting DYRKIa, but leaving casein kinase 2 (CK2) unmodulated.
These anti- inflammatory and anticancer activities of the compounds of the invention have been demonstrated using relevant cell culture models including primary cultured cells isolated from patients suffering from cancer (leukaemic cells) and rheumatoid arthritis (inflammatory cells from affected joints). Abbreviations
ΔΨm mitochondrial membrane potential
AIDS acquired immune deficiency syndrome
AML acute myeloid leukaemia
API atmospheric pressure ionization
BuLi butyl lithium
CDI N, N '-carbony ldiimidazo Ie
CK casein kinase
CLL chronic lymphatic leukaemia
DBU l,8-diazabicyclo[5.4.0]undec-7-ene
DCM dichloromethane
DHP 3 ,4-dihydro-2H-pyran
DIPEA Λ/,Λ/-diisopropylethylamine
DMAP 4-(dimethylamino)pyridine
DMEM Dulbecco's modified Eagle medium
DMF Λ/,Λ/-dimethylformamide
DMSO dimethyl sulphoxide
EDC 1 - [3 -(dimethylamino)propyl] -3 -ethylcarbodiimide hydrochloride
EDTA ethylenediamine tetraacetic acid
EtOAc ethyl acetate
FBS foetal bovine serum
FT Fourier transform
GFP green fluorescent protein
HL-60 human promyelocytic leukaemia cells
HOBt 1 -hydroxybenzotriazo Ie
IC50 concentration yielding 50% inhibition
IL-2 interleukin-2
IR infrared spectroscopy
LAH lithium aluminium hydride
LC liquid chromatography
LDH lactate dehydrogenase
MeOH methanol
MS mass spectrum NaHDMS sodium δώ(trimethylsilyl)amide
NCE new chemical entity
NMR nuclear magnetic resonance
PBS phosphate buffered saline PDBu phorbol 12,13-dibutyrate
PEG polyethylene glycol
PK protein kinase
PMA phorbol myristate acetate
PPTS pyridinium toluene-/?-sulfonate SD standard deviation
THP tetrahydropyran-2-yl
Figures
Figure 1 is a graphical representation of the ability of the compounds of the invention to replace phorbol ester at the binding site of PKCα in a concentration-dependent manner. The results are expressed as mean ± sem from independent experiments (n = 4-7).
Figure 2 is a graphical representation of the ability of the compounds of the invention to replace phorbol ester at the binding site of PKCδ in a concentration-dependent manner. The results are expressed as mean ± sem from independent experiments (n = 4-6).
Figure 3 consists of confocal microscopy images showing the inhibition by the compounds of the invention of PMA- induced translocation of PKCα in living HeLa cells.
Figure 4 consists of confocal microscopy images showing the inhibition by the compounds of the invention of PMA- induced translocation of PKCδ in living HeLa cells.
Figure 5 is a graphical representation showing the cytotoxicity of the compounds of the invention as determined by MTT test. The results are from a single experiment.
Figure 6 is a graphical representation showing the cytotoxicity of the compounds of the invention as determined by LDH test. The results are from a single experiment. Figure 7 is an image showing the effect of Ia3 on HeLa cell morphology, where image A) shows the HeLa cells before treatment, image B) shows the HeLa cells after 72 hours of treatment with 10 μM Ia3, and image C) shows the HeLa cells after 72 hours of treatment with 20 μM Ia3.
Figure 8 shows the effect of compounds Ib3, Ib5 and Ib7 on HL60 cell apoptosis (A) and CDl Ib expression (B).
Figure 9 shows the effect of compounds Ib5 and Ib7 on primary AML cell apoptosis in a graphical representation of the percentage of viable cells after 2 days of incubation.
Figure 10 shows the concentration response of compound Ib5 on B-CLL cell apoptosis (A) and the effect of compounds Ib3 and Ib5 on normal B cell apoptosis (B).
Figure 11 shows the effect of compounds Ib3, Ib5 and Ib7 on B-CLL apoptosis in co- culture with stromal cells (A) or stromal cells expressing CD40L (B).
Figures 12A and 12B show the effect of compounds IaS, IaI , Ia3, ibl, I b3, Ib4, Ib2,
Ib5, Ibό and Ib7 on apoptosis of activated CD4 T cells cultured in the presence or absence of I L2.
Figure 13 presents the pro-apoptotic activity of some of the synthesized compounds in HL- 60 leukaemia cells (A) and in Swiss 3T3 fibroblasts (B). Columns represent the amount of apoptotic cells (percent) at concentrations of 100, 60 and 20 μM. Each data point represents an average ± SD:s (n = 6).
Figure 14 shows chromatin fragmentation in HL-60 cells treated with 20 μM Ia3 and IbI at the time points 1, 2, 3, 4 and 6 h. Each data point represents an average + SD:s.
Figure 15 shows changes in mitochondrial transmembrane potential in HL-60 cells treated for 2 h with 40 μM Ia3 or 40 μM IbI. Non-treated cells and cells treated with 10 μM valinomycin are shown as controls. Figure 16 shows the Caspase-3 activity of HL-60 cells incubated with 50 μM Ia3 and IbI. Camptothecin-treated cells are shown as positive controls. Each data point represents an average + SD:s.
Detailed description of the invention
The compounds of the present invention are 1,3,5-trisubstituted derivatives of benzene. The hydrophobic nature of these new chemical entities (NCEs) is preferred for the targets of the present invention, since the binding site of the protein kinases are hydrophobic.
Particularly, the compounds of the present invention are compounds of Formula I:
Figure imgf000012_0001
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, wherein R1, R2 and R3 may be the same or different and are each independently chosen from hydrogen, branched or linear alkyl, branched or linear hydroxyalkylene, nitro, amino, - C(=O)OR4, -C(=O)NR5R6, and -NHC(=O)R7, wherein
R4 is chosen from hydrogen, a branched or linear, saturated or unsaturated Ci-Ci6 alkyl chain, a branched or linear, saturated or unsaturated Ci-Ci6 alkoxy chain, an aliphatic or aromatic, unsubstituted or substituted, mono-, di- or tricyclic ring structure connected to the -C(=O)O- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear Ci-Ci6 alkyl, branched or linear Ci-Ci6 alkoxy, branched or linear hydroxyalkylene, mono-, di- or trihalogeno- Ci-Ci6 alkylene, and wherein any of the groups R4 may possibly contain a heteroatom at any position of the group, chosen from O, S and N; R5 and R6 may be the same or different and are each independently chosen from hydrogen, a branched or linear, saturated or unsaturated Ci-Ci6 alkyl chain, a branched or linear, saturated or unsaturated Ci-Ci6 alkoxy chain, an aliphatic or aromatic, unsubstituted or substituted, mono-, di- or tricyclic ring structure connected to the -C(=O)N- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear Ci-Ci6 alkyl, branched or linear Ci-Ci6 alkoxy, branched or linear hydroxyalkylene, mono-, di- or trihalogeno- Ci-Ci6 alkylene, wherein any of the groups R5 and R6 may possibly contain a heteroatom at any position of the group, chosen from O, S and N; alternatively, R5 and R6 may be combined to form an aliphatic or aromatic, substituted or unsubstituted, mono-, di- or tricyclic ring structure connected to the -C(=O)N- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, 0 to 3 ring(s) may be aliphatic, whereas the remaining 0 to 3 ring(s) may be aromatic, and the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the substituents are chosen from the ring substituents mentioned above, and wherein the ring structure possibly also contains a further heteroatom chosen from O, S and N; and
R7 is chosen from hydrogen, a branched or linear, saturated or unsaturated Ci-Ci6 alkyl chain, a branched or linear, saturated or unsaturated Ci-Ci6 alkoxy chain, an aliphatic or aromatic, unsubstituted or substituted, mono-, di- or tricyclic ring structure connected to the -NHC(=O)- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear Ci-Ci6 alkyl, branched or linear Ci-Ci6 alkoxy, branched or linear hydroxyalkylene, mono-, di- or trihalogeno- Ci-Ci6 alkylene, wherein any of the groups R7 may possibly contain a heteroatom at any position of the group, chosen from O, S and N; with the provisos that only one of R1, R2 and R3 may be hydrogen, only one of R1, R2 and R3 may be -C(=O)OC2H5, and if one of R1, R2 and R3 is a -C(=O)OC2H5 group, none of the other groups R1, R2 and R3 may be -C(=O)OCH3.
Preferably two of R1, R2 and R3 are identical, whereas the remaining group is different. More preferably, the remaining group is hydroxymethylene.
R5 and R6 are preferably different from each other.
According to a preferred embodiment of the present invention, the number of substituents, ootthheerr tthhaann hhyyddrrooggeenn,, oorr hheetteerrooaattoommss oonn tthhee ppoossssiibbllee rriinngg ssttrruuccttuurreess of R4, R5, R6 and R7 is limited to one, and this substituent is at position 3 or 4 of the ring.
According to another preferred embodiment of the invention, the compounds of the present invention are compounds of Formula I, or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, wherein
R1 is chosen from hydrogen, methyl, hydroxymethylene, nitro, amino and -C(=O)OR4; R2 and R3 may be the same or different and are each independently chosen from hydrogen, -C(=O)OR4, -C(=O)NR5R6, and -NHC(=O)R7, wherein R4 is chosen from hydrogen, a branched or linear, saturated Ci -C7 alkyl chain, a branched or linear, saturated hydroxy-Ci-Cs alkyl chain, an aliphatic or aromatic unsubstituted or substituted, mono- or dicyclic ring structure connected to the - C(=O)O- group either directly or through an alkylene group, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear C1-C5 alkyl, branched or linear hydroxy-Ci-Cs alkyl, branched or linear
C1-C5 alkoxy, mono-, di- or trihalogeno- C1-C5 alkylene, wherein any of the groups R4 may possibly contain a heteroatom at any position of the group, chosen from O and N;
R5 and R6 may be the same or different and are each independently chosen from a branched or linear, saturated Ci -Ci 6 alkyl chain, an aliphatic or aromatic unsubstituted or substituted, mono- or dicyclic ring structure connected to the - C(=O)N- group either directly or through an alkylene group, wherein the ring substituents are chosen from the groups hydrogen, mono-, di- or trihalogeno- C1-C5 alkylene; alternatively, R5 and R6 may be combined to form a substituted or unsubstituted, mono- or dicyclic ring structure connected to the -C(=O)N- group either directly or through an alkylene group, wherein, in the case of a dicyclic ring structure, 0 to 2 ring(s) may be aliphatic, whereas the remaining ring(s) may be aromatic, and the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the substituents are chosen from the ring substituents mentioned above, and wherein the ring structure possibly also contains a further heteroatom, which is N; and
R7 is chosen from hydrogen and a branched or linear, saturated or unsaturated Ci-Ci6 alky 1 chain; with the provisos that only one of R1, R2 and R3 may be hydrogen, only one of R1, R2 and R3 may be -C(=O)OC2H5, and if one of R1, R2 and R3 is a -C(=O)OC2H5 group, none of the other groups R1, R2 and R3 may be -C(O)OCH3.
According to a preferred aspect of the invention, at least two of R1, R2 and R3 contain an aliphatic or aromatic unsubstituted or substituted, mono- or dicyclic ring structure.
According to another preferred aspect, at least two of R1, R2 and R3 contain at least 6 carbon atoms each. These can be arranged into either a chain or a cyclic structure.
According to a further preferred embodiment of the present invention, one of R1, R2 and R3 is hydroxymethylene, while the other two have one of the following definitions: - -C(=O)OR4,
- -C(=O)NR5R6, or
- either -C(=O)OR4 or -NHC(=0)R7.
Thus, according to this embodiment, a compound of the invention will have one of the following Formulae II, III, IV and V:
Figure imgf000016_0001
Figure imgf000016_0002
Figure imgf000016_0003
and
Figure imgf000016_0004
According to a particularly preferred embodiment of the present invention, the compound of the invention is one of the following:
Figure imgf000016_0005
1 a1 1a2
Figure imgf000017_0001
1a3 1a4
Figure imgf000017_0002
1a5 1a6
Figure imgf000017_0003
1a7 1a8
Figure imgf000017_0004
1b1 1b2
Figure imgf000017_0005
1b3 1b4
Figure imgf000017_0006
1b5 1b6
Figure imgf000018_0001
1b17 1b18
Figure imgf000018_0002
1b19 1b20
Figure imgf000018_0003
Figure imgf000019_0001
15a 15b
Figure imgf000019_0002
15c 15d
Figure imgf000019_0003
15e 15f
Figure imgf000019_0004
17a 17b
Figure imgf000019_0005
Figure imgf000020_0001
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
According to a more particularly preferred embodiment of the present invention, the compound of the present invention is a compound of Formula I, wherein R4 is an aliphatic, branched or linear hydrocarbon chain containing at least 6 carbon atoms. Preferably, these compounds are chosen from:
Figure imgf000020_0002
Figure imgf000020_0004
Figure imgf000020_0003
1 b4
Figure imgf000021_0001
1b7
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
According to a more particularly preferred embodiment of the present invention, the compound of the invention is one of the following:
Figure imgf000021_0002
Figure imgf000021_0003
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
The present compounds are characterized as "small-molecular" compounds which means that they have a molecular weight of typically less than about 1500 Da, in particular less than about 1000 Da and preferably less than about 500 Da. They can be synthesized by conventional chemical reactions, as will be discussed in more detail below.
In the above Formula I, "alkyl" refers to a linear or branched saturated hydrocarbon group. Nonlimiting examples of alkyl groups are methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, octyl, amyl, and the like. "Alkoxy" represents a linear or branched saturated hydrocarbon group linked to the compound through an oxygen atom. Nonlimiting examples of alkoxy groups include methoxy, ethoxy, propoxy, and t-butoxy.
The terms "halogen", "halo" and "halide" are used in the conventional sense to refer to a chloro, bromo, fluoro or iodo substituent or corresponding ion.
As used in the present invention, an "aliphatic ring structure" is a hydrocarbon ring having 5 to 15 ring atoms and which may be unsubstituted or substituted with one or more substituents. Nonlimiting examples of substituent groups include halo, nitro, cyano, linear or branched alkyl, linear or branched alkenyl, aryl, cycloalkyl, cycloalkenyl, amino, amido, carboxylate, and hydroxy.
As used in the present invention, an "aromatic ring structure" is an aryl group generally containing 5 to 15 carbon atoms or it can be a heteroaryl group. An "aryl" group can contain a single aromatic ring or multiple aromatic rings that are fused together (i.e. contain a mutual chemical bond), directly linked (i.e. contain a mutual ring atom), or indirectly linked (i.e. the atoms of the separate aromatic rings are bound together through a bond or a common group such as a methylene or ethylene). Preferred aryl groups contain 5 to 15 carbon atoms, and particularly preferred aryl groups contain 6 to 12 carbon atoms. Nonlimiting examples of aryl groups containing one aromatic ring or two or more fused or linked aromatic rings include phenyl, naphthyl, biphenyl, diphenyl ether, diphenylamine, benzophenone, and the like. Aryl groups can optionally be substituted with one or more substituents. Nonlimiting examples of substituents include halo, nitro, cyano, linear or branched alkyl, linear or branched alkenyl, linear or branched haloalkyl, aryl, cycloalkyl, cycloalkenyl, amino, amido, carboxylate, and hydroxy.
"Heteroaryl" refers to an aromatic moiety as defined above for aryl further containing at least one ring heteroatom selected from oxygen, nitrogen and sulphur. Non-limiting examples of heteroaryl groups include pyrrolyl, pyrrolidinyl, pyridinyl, quinolinyl, indolyl, pyrimidinyl, furyl, thiophenyl, oxazolyl, azolyl, imidazolyl, triazolyl and tetrazolyl.
The compounds of Formula I may be used as medicaments. Preferably, they are used in the manufacture of medicaments for the treatment of disorders relating to activation or inhibition of the PKC isoenzymes or other protein kinases in mammals, particularly in humans. The medicaments may be used in diagnosing, treating or ameliorating various cancers or inflammatory disorders, such as leukaemia or arthritis.
Disclosed herein are compositions of the compounds as described above. The compositions comprise a therapeutically effective amount of the compounds or pharmaceutically acceptable salts, solvates, esters or prodrugs thereof and one or more of pharmaceutically acceptable carriers, diluents and adjuvants. These compositions may be used as medicaments. They may be formulated in any common way, preferably for oral or intravenous administration.
The activators and inhibitors are employed in amounts effective to achieve their intended purpose. As used herein, a "therapeutically effective amount" means an amount effective to inhibit development of, or to alleviate the existing symptoms of, the condition of the subject being treated. "Dose effective to activate" or "dose effective to inhibit" means an amount effective to activate or inhibit the PKC signaling pathway or other pathway affecting protein phosphorylation, in vivo or ex vivo. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmacological procedures in cell cultures or experimental animals, e.g., procedures for determining the ability to induce apoptosis, the cytotoxicity or the mutagenicity (see biological tests below).
Activation or inhibition of the PKC signaling pathway can be measured using various sensitive assay systems where the effect of a compound of interest is tested over a range of concentrations, i.e. showing a dose-response, including concentrations at which no or minimal effect is observed, through higher concentrations at which partial effect is observed, to saturating concentrations at which a maximum effect is observed. The dose- response of the effect of a compound of interest is usually a sigmoidal curve expressing a degree of inhibition as a function of concentration. The curve also theoretically passes through a point at which the concentration is sufficient to reduce or enhance the activity of the PKC signaling pathway to a level that is 50% that of the difference between minimal and maximal activity in the assay. This concentration is defined as the Inhibitory Concentration (50%) or IC50 value. Determination of IC50 values preferably is made using conventional biochemical (acellular) assay techniques or cell based assay techniques. It is not uncommon to obtain a bell-shape dose-response curve where maximum effect is obtained in lower concentrations than the maximum concentrations used in the assay. The data obtained in dose-response assays can be used as a factor in formulating a dosage range for use in mammals and, more specifically, humans.
The compounds employed in the methods of the present invention may be administered by any means that results in the contact of the active agent with the agent's site of action in the body of a patient. The compounds may be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. For example, they may be administered as the sole active agent in a pharmaceutical composition, or they can be used in combination with other therapeutically active ingredients.
The exact formulation, route of administration, and dosage is chosen by a subject's physician, or treating professional, in view of the subject's condition. Dosage amount and interval can be adjusted individually to provide plasma levels of the active compound that are sufficient to maintain desired therapeutic effects. In general, however, doses employed for humans typically are in the range of 0.001 mg/kg to about 1000 mg/kg per day, preferably 0.01 mg/kg to about 50 mg/kg per day, typically in a range of about 0.0005 to about 500 mg/kg per dose of activator/inhibitor, preferably from about 0.1 mg/kg to about 10 - 50 mg/kg per dose of activator/inhibitor. In some embodiments, doses range from about 0.1 to about 50 mg/kg, about 0.5 to about 40 mg/kg, about 0.7 to about 30 mg/kg, or about 1 to about 20 mg/kg. Specific doses contemplated include sub-ranges of any of the foregoing ranges in 0.1 mg/kg increments.
The formulation components are present in concentrations that are acceptable to the site of administration. For example, buffers are used to maintain the composition at physiological pH or at a slightly lower pH, typically within a pH range of from about 5 to about 8. Compounds of the present invention can be administered to a mammalian subject in a variety of forms adapted to the chosen route of administration, e.g., orally or parenterally. Parenteral administration in this respect includes administration by the following routes: intravenous, intramuscular, subcutaneous, rectal, intraocular, intrasynovial, transepithelial including transdermal, ophthalmic, sublingual and buccal; topically including ophthalmic, dermal, ocular, rectal, and nasal inhalation via insufflation aerosol. Also administration into the brain tissue or cerebral ventricles is possible. Parenteral compositions usually contain a buffering agent and, optionally, a stabilizing agent.
Solutions of the active compound as a free base or a pharmacologically acceptable salt can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. A dispersion can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
The pharmaceutical forms suitable for injectable use include, for example, sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form is preferably sterile and fluid to provide easy syringability. It is preferably stable under the conditions of manufacture and storage and is preferably preserved against the contaminating action of microorganisms such as bacteria and fungi. The suspension may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The compositions may also be solutions or suspensions in non-toxic diluens or solvents, for example as a solution in 1,3-butanediol. The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), suitable mixtures thereof and vegetable oils. In addition, fixed oils may be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of a dispersion and by the use of surfactants. The prevention of the action of microorganisms may be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions may be achieved by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions may be prepared by incorporating the active compound in the required amount, in the appropriate solvent, with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions may be prepared by incorporating the sterilized active ingredient into a sterile vehicle that contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation may include vacuum drying and the freeze drying technique, which yield a powder of the active ingredient, plus any additional desired ingredient from the previously sterile- filtered solution thereof.
Oral formulations include tablets, buccal tablets, troches, pills, capsules, elixirs, suspensions, syrups, wafers and the like and may further contain a binder, such as gum tragacanth, acacia, corn starch or gelatin; an excipient, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; a sweetening agent such as sucrose, lactose or saccharin; or a flavoring agent, such as peppermint, oil of wintergreen or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills or capsules may be coated with shellac, sugar or both. A syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propyl parabens as preservatives, a dye and flavoring, such as cherry or orange flavor. The active compound may be enclosed in hard or soft shell gelatin capsules, or it may be incorporated into sustained-release preparations and formulations.
Pharmaceutically acceptable ingredients are well known for the various types of formulation and may be for example binders such as natural or synthetic polymers, excipients, lubricants, surfactants, sweetening and flavouring agents, coating materials, preservatives, dyes, thickeners, adjuvants, antimicrobial agents, antioxidants and carriers for the various formulation types. Nonlimiting examples of binders useful in a composition described herein include gum tragacanth, acacia, starch, gelatine, and biological degradable polymers such as homo- or co-polyesters of dicarboxylic acids, alkylene glycols, polyalkylene glycols and aliphatic hydroxyl carboxylic acids; homo- or co-polyamides of dicarboxylic acids, alkylene diamines, and aliphatic amino carboxylic acids; corresponding polyester-polyamide-co-polymers, polyanhydrides, polyorthoesters, polyphosphazene and polycarbonates. The biological degradable polymers may be linear, branched or crosslinked. Specific examples are poly-glycolic acid, poly- lactic acid and po Iy-JJ- lactide/glycolide. Other examples for polymers are water-soluble polymers such as polyoxaalkylenes (polyoxaethylene, polyoxapropylene and mixed polymers thereof, poly- acrylamides and hydroxylalkylated polyacrylamides, poly-maleic acid and esters or - amides thereof, poly-acrylic acid and esters or -amides thereof, poly-vinylalcohol and esters or -ethers thereof, poly-vinylimidazole, poly-vinylpyrrolidon and natural polymers like chitosan.
Nonlimiting examples of excipients useful in a composition described herein include phosphates such as dicalcium phosphate. Nonlimiting examples of lubricants used in a composition described herein include natural or synthetic oils, fats, waxes or fatty acid salts such as magnesium stearate.
As used herein, the term "pharmaceutically acceptable salts" refers to salts or zwitterionic forms of the compounds a described above. Salts of such compounds can be prepared during the final isolation and purification of the compounds or separately by reacting the compound with an acid having a suitable cation. Suitable pharmaceutically acceptable cations include alkali metal (e.g., sodium or potassium) and alkaline earth metal (e.g., calcium or magnesium) cations. In addition, the pharmaceutically acceptable salts of the disclosed compounds that contain a basic center are acid addition salts formed with pharmaceutically acceptable acids. Examples of acids which can be employed to form pharmaceutically acceptable salts include inorganic acids such as hydrochloric, hydrobromic, sulfuric and phosphoric, and organic acids such as oxalic, maleic, succinic, malonic and citric acid. Nonlimiting examples of salts of compounds of the invention include, but are not limited to, hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, 2-hydroxyethanesulfonate, phosphate, hydrogen phosphate, acetate, adipate, alginate, aspartate, benzoate, butyrate, camphorate, camphorsulfonate, citrate, digluconate, glycerolphosphate, hemisulfate, heptanoate, hexanoate, formate, succinate, malonate, fumarate, maleate, methanesulfonate, mesitylenesulfonate, naphthylenesulfonate, nicotinate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, trichloroacetate, trifluoroacetate, glutamate, bicarbonate, undecanoate, lactate, citrate, tartrate, gluconate, benzene sulphonate, and/?-toluenesulphonate salts. In addition, available amino groups present in the compounds of the invention can be quaternized with methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dimethyl, diethyl, dibutyl and diamyl sulfates; decyl, lauryl, myristyl and steryl chlorides, bromides and iodides; and benzyl and phenethyl bromides. In light of the foregoing, any reference to compounds appearing herein is intended to include compounds disclosed herein as well as pharmaceutically acceptable salts, solvates (e.g., hydrates), esters or prodrugs thereof.
The pharmaceutical composition may contain formulation materials for modifying, maintaining or preserving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition. Suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogensulfϊte); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates, phosphates or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or hydroxypropyl-beta-cyclodextrin); fillers; monosaccharides, disaccharides and other carbohydrates (such as glucose, mannose or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring, flavoring and diluting agents; emulsifying agents; hydrophilic polymers (such as polyvinylpyrrolidone); low molecular weight polypeptides; salt-forming counterions (such as sodium); preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine, sorbic acid or hydrogen peroxide); solvents (such as glycerin, propylene glycol or polyethylene glycol); sugar alcohols (such as mannitol or sorbitol); suspending agents; surfactants or wetting agents (such as pluronics, PEG, sorbitan esters, polysorbates such as polysorbate 20, polysorbate 80, triton, tromethamine, lecithin, cholesterol, tyloxapal); stability enhancing agents (such as sucrose or sorbitol); tonicity enhancing agents (such as alkali metal halides (preferably sodium or potassium chloride); delivery vehicles; diluents; excipients and pharmaceutical adjuvants (Remington's Pharmaceutical Sciences, 18th Edition, A.R. Gennaro, ed., Mack Publishing Company (1990)).
Surfactants for use in a composition described herein can be anionic, anionic, amphoteric or neutral. Nonlimiting examples of surfactants useful in a composition described herein include lecithin, phospholipids, octyl sulfate, decyl sulfate, dodecyl sulfate, tetradecyl sulfate, hexadecyl sulfate and octadecyl sulfate, sodium oleate or sodium caprate, 1- acylaminoethane-2-sulfonic acids, such as l-octanoylaminoethane-2-sulfonic acid, 1- decanoylaminoethane-2-sulfonic acid, l-dodecanoylaminoethane-2-sulfonic acid, 1- tetradecanoylaminoethane-2-sulfonic acid, l-hexadecanoylaminoethane-2-sulfonic acid and l-octadecanoylaminoethane-2-sulfonic acid, and taurocholic acid and taurodeoxycholic acid, bile acids and their salts, such as cholic acid, deoxycholic acid and sodium glycocholates, sodium caprate or sodium laurate, sodium oleate, sodium lauryl sulphate, sodium cetyl sulphate, sulfated castor oil and sodium dioctylsulfosuccinate, cocamidopropylbetaine and laurylbetaine, fatty alcohols, cholesterols, glycerol mono- or - distearate, glycerol mono- or -dioleate and glycerol mono- or -dipalmitate and polyoxyethylene stearate.
Nonlimiting examples of sweetening agents useful in a composition described herein include sucrose, fructose, lactose or aspartame. Nonlimiting examples of flavoring agents for use in a composition described herein include peppermint, oil of wintergreen or fruit flavors such as cherry or orange flavor. Nonlimiting examples of coating materials for use in a composition described herein include gelatin, wax, shellac, sugar or other biological degradable polymers. Nonlimiting examples of preservatives for use in a composition described herein include methyl or propylparabens, sorbic acid, chlorobutanol, phenol and thimerosal.
The primary vehicle or carrier in a pharmaceutical composition may be either aqueous or non-aqueous in nature. For example, a suitable vehicle or carrier may be water for injection, physiological saline solution, solution or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles. Other exemplary pharmaceutical compositions comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, which may further include sorbitol or a suitable substitute therefore.
Such compositions and preparations should preferably contain at least about 0.1% by weight of active compound. The dosage of the compounds of the present invention that will be most suitable will vary with the form of administration, the particular compound chosen and the physiological characteristics of the particular patient under treatment. In some cases, the compositions or preparations contain a compound of Formula I in the range of about 2% to about 6%. The amount of active compound in the compositions or preparations may be selected so as to provide a suitable dosage for the disorder, disease or diagnostic application. Compositions or preparations according to the present invention may be prepared so that an oral dosage unit form contains from about 0.1 to about 1000 mg of active compound and all combinations and subcombinations of ranges and specific amounts therein.
The specific contents and amounts in the optimal pharmaceutical composition will be determined by one skilled in the art depending upon, for example, the intended route of administration, delivery format and desired dosage. See, for example, Remington's Pharmaceutical Sciences, supra. Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the PKC activator, inhibitor or modulator.
Additional pharmaceutical compositions will be evident to those skilled in the art, including compositions of compounds affecting protein phosphorylation pathways, involving PKC activators, inhibitors or modulators, in formulations for inhalation or in sustained- or controlled-delivery formulations. Techniques for formulating a variety of sustained- or controlled-delivery means, such as liposome carriers, bio-erodible microparticles or porous beads and depot injections, are also known to those skilled in the art. See for example, PCT Application No. PCT/US93/00829, which describes the controlled release of porous polymeric microparticles for the delivery of pharmaceutical compositions. Additional examples of sustained-release preparations include semipermeable polymer matrices in the form of shaped articles, e.g. films, or microcapsules. Sustained release matrices may include polyesters, hydrogels, polylactides, copolymers of glutamic acid and gamma ethyl-L-glutamate, poly (2-hydroxyethyl- methacrylate), ethylene vinyl acetate or poly-D-3-hydroxybutyric acid. Sustained-release compositions may also include liposomes.
The composition of the present invention is preferably formulated for oral or intravenous administration. More preferably, the composition is administered orally when used in the treatment of inflammatory diseases and intravenously when used in the treatment of cancers.
When intravenous administration is contemplated, the therapeutic compositions for use in this invention may be in the form of a pyrogen- free, parenterally acceptable aqueous solution comprising the active compound in a pharmaceutically acceptable vehicle. A particularly suitable vehicle for parenteral injection is sterile distilled water in which the active compound is formulated as a sterile, isotonic solution, properly preserved.
When oral administration is contemplated, the active agents can be formulated with or without those carriers customarily used in the compounding of solid dosage forms such as tablets and capsules. For example, a capsule may be designed to release the active portion of the formulation at the point in the gastrointestinal tract when bioavailability is maximized and pre-systemic degradation is minimized. Additional agents can be included to facilitate absorption of the compounds including PKC activators, inhibitors or modulators. Diluents, flavorings, low melting point waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents and binders may also be employed.
The pharmaceutical composition to be used for in vivo administration typically must be sterile. This may be accomplished by filtration through sterile filtration membranes. Where the composition is lyophilized, sterilization using these methods may be conducted either prior to or following lyophilization and reconstitution. The composition for parenteral administration may be stored in lyophilized form or in a solution. In addition, parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. Once the pharmaceutical composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or as a dehydrated or lyophilized powder. Such formulations may be stored either in a ready-to-use form or in a form (e.g., lyophilized) requiring reconstitution prior to administration. Furthermore, the compounds of this invention can, when used in cancer therapy, be used together with other substances and compounds, such as chemotherapeutic agents, when appropriate. Such compounds are, for example (according to the general classes of the compounds):
Alkylating agents, such as cyclophosphamide, cisplatin, carboplatin, ifosfamide, chlorambucil, busulfan, thiotepa, nitrosoureas);
Antimetabolites, such as 5-fluorouracil, fludarabine, methotrexate, azathioprine, gemcitabine (Gemzar); Antitumour antibiotics, such as doxorubicin, daunorubicin, epirubicin, actinomycin, bleomycin, mitomycin, plicamycin, dactinomycin, adriamycin;
Hormonal therapy agents, such as steroids, finasteride, aromatase inhibitors, tamoxifen, goserelin;
Taxanes, such as paclitaxel (Taxol), docetaxel (Taxotere); (antimicrotubule) ;
Topoisomerase inhibitors, such as irinotecan, topotecan, amsacrine, etoposide, etoposide phosphate, teniposide;
Vinca alkaloids, such as vincristine, vinblastine, vinorelbine, vindesine;
(antimiotic agents) capecitabine (Xeloda), podophyllotoxin, amoxicillin, and piroxicam.
In addition to the above, there are several new compounds disclosed in pending patent applications, e.g.: Epothilones (US 2005244413), serratamolide (US 2005239694), indol derivatives (US 2005239752), various plant extracts: extract of sea buckthorn - Hippophae rhamnoides (US 2005214394), extracts of Ganoderma lucidum, Salvia miltiorrhiza and Scutellaria barbata (US 2005208070), the contents of the afore-mentioned US Patent Applications are herewith incorporated by reference.
When necessary, in order to promote penetration of the blood-brain-barrier, the active compounds can be administered by using various strategies for gaining drug access to the brain. These include, e.g., the transcellular lipophilic pathway, which allows small, lipophilic compounds to cross the blood-brain barrier and "receptor-mediated endocytosis". The compounds employed in the uses and methods of the present invention may exist in prodrug form. As used herein, the term "prodrug" is intended to include any covalently bonded carriers which release the active parent drug or other formulae or compounds employed in the methods of the present invention in vivo when such prodrug is administered to a mammalian subject. Since prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability, manufacturing, etc.), the compounds employed in the present methods may, if desired, be delivered in prodrug form. Thus, the present invention contemplates methods of delivering prodrugs. Prodrugs of the compounds employed in the present invention may be prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound.
Accordingly, prodrugs include, for example, compounds described herein in which a hydroxy, thiol, amino or carboxy group is bonded to any group that, when the prodrug is administered to a mammalian subject, cleaves to form a free hydroxyl, thiol, free amino or carboxylic acid, respectively. Examples include, but are not limited to, acetoxyalkyls, acetate, formate and benzoate derivatives of alcohol, thiol and amine functional groups; and alkyl, carbocyclic, aryl and alkylaryl esters such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-bvXy\, cyclopropyl, phenyl, benzyl and phenethyl esters and the like.
The present invention further provides a method of regulating protein kinase activity, especially that of PKC activity in a subject comprising administering to the subject a composition comprising a compound of the invention, as described above, or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, in an amount effective to regulate the protein kinase activity. Preferably the subject is a mammal, more preferably a human.
According to one embodiment of the present invention, the compound of the invention is used in the treatment or prevention of cancer, particularly in manufacturing a medicament for treating a subject suffering from cancer, such as prostate cancer, pancreatic cancer, lymphoma, lung cancer, thyroid cancer, malignant glioma, testicular cancer, cervical cancer, uterine cancer, stomach cancer, leukaemia, melanoma, ovarian cancer, renal cancer, cancer of the large intestine, rectal cancer and breast cancer, preferably leukaemia, particularly acute myeloid leukaemia or chronic lymphatic leukaemia.
Preferably the compound of Formula I used when treating cancer is one of the following:
Figure imgf000034_0001
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
According to another embodiment of the present invention, the compound of the invention is used in the treatment or prevention of an inflammatory disease, particularly in manufacturing a medicament for treating a subject suffering from an inflammatory disease, such as rheumatoid arthritis.
Preferably the compound of Formula I used when treating an inflammatory disease is the following:
Figure imgf000034_0002
1 b1
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
Further utility of the present compounds is in the field of biological research as chemicals, including reagents for testing of biological models. EXAMPLES
Example 1 Synthetic procedures
Synthetic reactions were monitored using Merck silica gel 60 (230-400 mesh) neutral thin layer chromatography plates. Flash column chromatography was performed with Merck silica gel 60 (230-400 mesh) and the mobile phase indicated.
1H NMR (300 MHz) and 13C NMR (75.4 MHz) spectra were acquired on a Varian
Mercury 300 Plus spectrometer in the solvents indicated. Chemical shifts for 1H NMR spectra are reported relative to internal standards CHCI3 in CDCI3 (7.26 ppm) and (CHs)2SO in (CDs)2SO (2.50 ppm). In 13C NMR spectra, peaks are referenced to the central line of the signal arising from the solvent, e.g. CDCI3 (77.21 ppm) and (CDs)2SO (39.52 ppm). Coupling constants are reported in Hertz (Hz). Spectral splitting patterns are designated as follows: s, singlet; d, doublet; dd, doublet of doublets; m, multiplet; q, quartet; qv, quintet. Deuterated solvents were purchased from Aldrich.
Liquid chromatography mass spectra (LC-MS) were acquired on a Perkin Elmer Sciex Instruments API 3000 quadrupole LC/MS/MS mass spectrometer. FT-IR spectra were recorded on a Perkin Elmer FT-IR 1725X spectrometer from a KBr tablet.
Some intermediates (7a-c) were synthesized using the following scheme (Scheme 1).
Scheme 1
Figure imgf000035_0001
6a
Figure imgf000035_0002
6a-c 7a-c
Figure imgf000036_0001
The 5-(hydroxymethyl)isophthalates are prepared according to the following general scheme (Scheme 2).
Scheme 2
Figure imgf000036_0002
Figure imgf000036_0003
5a 1-8 1a1 -8
5b1-20 1 b1-20
5c1 1c1
Figure imgf000036_0004
Ia6 CH2Ph Ibl3 decahydronaphthalen-2-yl
Ia7 CH2(W-OMe-Ph) Ibl4 3-ethoxypropyl
Ia8 CH2(CH2)3CH3 Ibl5 (1 R)-2,3-dihydro-1 H-inden-1 -yl
IbI CH2(CH2)4CH3 Ibl6 (2R)-2-methylpentyl
Ib2 CH2(c-Hex) Ibl7 (2S)-2-methylpentyl
Ib3 CH(CH2CH3)((CH2)3CH3) Ibl8 bicyclo[2.2.1 ]hept-2-ylmethyl
Ib4 CH(CH3)((CH2)3CH3) Ibl9 4-methylpentyl
Ib5 CH2CH(CH3)((CH2)2CH3) lb20 3-methylpentyl
Ib6 CH(CH3)((CH2)4CH3) IcI P-CH2OH-Ph
Ib7 CH(CH2CH3)((CH2)2CH3)
Chiral intermediates 13bl6-17 were synthesized using the following general procedure (Scheme 3).
Scheme 3
Figure imgf000037_0001
The isophthalates 15a-f are prepared according to the following general scheme (Scheme 4).
Figure imgf000038_0001
14a-e 15a-e 15f
Y=H, Me, NO2, NH2 Compound Y R1
15a H CH2CH(CH3)CH2CH2CH3 CH2CH(CH3)CH2CH2CH3
15b CH3 CH2CH(CH3)CH2CH2CH3 CH2CH(CH3)CH2CH2CH3
15c NO2 CH2CH(CH3)CH2CH2CH3 CH2CH(CH3)CH2CH2CH3
15d NO2 CH2(m -CF3-Ph) CH2(m -CF3-Ph)
15e H CH3 CH2CH(CH3)CH2CH2CH3
15f NH2 Crl2Crl(Crl3)Crl2Crl2Crl3 CH2CH(CH3)CH2CH2CH3
The 5-(hydroxymethyl)isophthalamides 17a-c are prepared according to the following general scheme (Scheme 5).
Scheme 5
Figure imgf000038_0002
16a -c 17a-c
Compound R
17a Λr-[(4-Λr-Phenyl)piperazin-l-yl
17b 7V-(3-(trifluoromethyl)benzyl
17c Λ-faexyl The amides 20 and 22 are prepared according to the following scheme (Scheme 6).
Scheme 6
Figure imgf000039_0001
*2 HCI
Figure imgf000039_0002
21 22
The alkyl 3-(hydroxyr«ethyπbenzoates 29a-c are prepared according to the foJ lowing general scheme (Scheme 7),
Scheme 7
Figure imgf000039_0003
23 24 25
Figure imgf000039_0004
26 27 28a-c
Figure imgf000039_0005
29a-c Methyl 3-(hydroxymethyl)-5-[[[3-(trifluoromethyl)phenyl]acetyl]amino]benzoate 33 is prepared according to the following scheme (Scheme 8).
Scheme 8
Figure imgf000040_0001
The methyl 3-(hydroκymethyl)-5-(alkanoylamino)benzoates 39a~e are prepared according Io the following general scheme (Scheme 9).
Scheme 9
Figure imgf000040_0002
34 35 36
Figure imgf000040_0003
37 38a-e 39a-e Compound R
39a (CH2)8CH=CH2
39b (CH2)7CH3
39c (CH2)5CH3
39d C(CHs)3
39e CH3
Example 2 Synthesis of intermediates
Example 2a
Synthesis of intermediates - Scheme 1
Step E: Synthesis of 5-chloropentan-l-ol, 6a
A solution of methyl 5 -chloro valerate (1 mL, 1 equiv.) in dry THF (23 rnL) was dropwise added to a solution of LAH (285 mg, 1.1 equiv.) in THF (8 mL) and was stirred for 75 min. The reaction mixture was cooled down on a ice-bath, 1 M H2SO4 (5.8 mL) was added to the reaction mixture, the solution stirred for 30 min, filtered, the solid was washed with ether (25 mL), the organic phase dried (Na2SO4), filtered and evaporated in vacuo to give a liquid (570 mg, 67% yield). 1H NMR (300 MHz, CDCl3) δ 3.59 (dt, 4H, J=6.3 and 26.7 Hz), 1.85-1.76 (m, 2H), 1.64-1.48 (m, 6H); 13C NMR (75.4 MHz, CDCl3) δ 62.5, 45.1, 32.5, 32.0, 23.3.
Step F: Synthesis of 2-[(5-chloropentyl)oxy]tetrahydro-2H-pyran, 7a
DΗP (627 μL, 1.5 equiv.) was added to a solution of 6a (570 mg, 1 equiv.), PPTS (23 mg, 0.02 equiv.) and DCM (17 mL) and stirred at rt for 22 h. DCM (40 mL) was added to the reaction mixture, it was washed with brine (20 mL), water (2x20 mL), dried (Na2SO4), filtered and evaporated in vacuo to give a clear liquid, (911 mg, 95% yield). The 7a was used in the subsequent esterification step without further purification. Step F: Synthesis of 2-(4-chlorobutoxy)tetrahydro-2H-pyran, 7b
DΗP (1832 μL, 2 equiv.) was added to a solution of 4-chloro-l-butanol (1 niL, 1 equiv.), PPTS (23 mg, 0.03 equiv.) and DCM (15 niL) and stirred at rt for 19 h. The reaction mixture was washed with water (3χ5 niL), the organic phase dried (Na2SO4), filtered and evaporated in vacuo to give a clear liquid, 7b (1.93 g, 100% yield). The 7b was used in the subsequent esterification step without further purification.
Step F: Synthesis of 4- [(tetrahydro-2H-pyran-2-yloxy)methyl] phenol, 7c
DΗP (613 μL, 1.67 equiv.) was added to a solution of 4-(hydroxymethyl)phenol (500 mg, 1 equiv.), PPTS (23 mg, 0.02 equiv.) and TΗF (7.5 mL) and stirred at rt for 18 h. TΗF (15 mL) was added to the reaction mixture, it was washed with 1 M NaOH (10 mL), water (80 0C, 3 x 7.5 mL) and the organic phase evaporated in vacuo to give a clear liquid, 7c (414 mg, 49% yield). 7c was used in the subsequent esterification step without further purification.
Example 2b
Synthesis of intermediates - Scheme 2
Step A: Synthesis of diethyl 5-(tetrahydropyran-2-yloxymethyl)isophthalate, 3
A mixture of 2 (5.72 g, 1 equiv.), 3,4-dihydro-2H-pyran (DΗP) (5.54 mL, 2.7 equiv.), pyridinium toluene-/?-sulfonate (PPTS) (285 mg, 0.05 equiv.) and 1 ,2-dichloroethane (50 mL) was stirred at room temperature for 6 h. The reaction mixture was quenched by adding cold water, extracted with EtOAc, washed with a saturated solution of NaHCO3 and brine. The organic phases were dried with Na2SO4, filtered and evaporated in vacuo to give a yellow oil 3 (7.4 Ig, 97% yield). This oil was used without further purification.
Step B: Synthesis of 5-(tetrahydropyran-2-yloxymethyl)isophthalic acid, 4
A mixture of 3 (3.46 g, 1 equiv.), a 10% solution of KOH (46.2 mL, 8 equiv.) and MeOH (50 mL) was refluxed for 1 h. The solvents were evaporated in vacuo and the pH was adjusted with a 25% solution OfKHSO4 to 4.00. The white precipitate was filtered, washed with water, dissolved into a solution of EtOAc and THF (1 :1, 3χ50 mL), filtered, dried (Na2SO4), filtered and evaporated in vacuo to give a white solid 4 (2.45 g, 85% yield). This solid was used without further purification. 1H NMR (300 MHz, DMSO-J6) δ 13.3 (bs, 2H), 8.39 (t, 1H, J=1.5 Hz), 8.13 (d, 2H, J=1.5 Hz), 4.80 (d, 1H, J=12.6 Hz), 4.72 (t, IH, J=3.0 Hz), 4.59 (d, 1H, J=12.6 Hz), 3.82-3.75 (m, IH), 3.52-3.45 (m, IH), 1.80-1.62 (m, 2H), 1.58-1.48 (m, 4H); 13C NMR (75.4 MHz, DMSO-J6) δ 166.5, 139.8, 132.1, 131.3, 129.0, 97.7, 67.3, 61.4, 30.1, 25.0, 19.1.
Step C - Method A: Synthesis of the diester, 5al-8
A mixture of 4 (200 mg, 1 equiv.), 4-te/t-butylbenzyl bromide (394 mg, 3 equiv.), K2CO3 (493 mg, 5 equiv.), KI (130 mg, 1.1 equiv.) and dry DMF (4 mL) was heated at 110 °C for 2 h. The reaction mixture was cooled down to rt and quenched by adding an ice-water- mixture (20 mL), extracted with EtOAc (3x20 mL), dried (Na2SO4), filtered and evaporated in vacuo to give a yellowish oil, δώ(4-te/t-butylbenzyl)-5-(tetrahydropyran-2- yloxymethyl)isophthalate 5al. The crude diesters were used in the subsequent deprotection steps without further purification.
Step C - Method B: Synthesis of the diester, 5bl-20
A mixture of 4 (250 mg, 1 equiv.), 1,1-carbonyldiimidazole (CDI) (318 mg, 2.2 equiv.) and dry DMF (2 mL) was stirred under argon at room temperature for 1 h. 1-Hexanol (336 μL, 3 equiv.), 4-(dimethylamino)pyridine (DMAP) (l lmg, 0.1 equiv.) and 1,8- diazabicyclo[5.4.0]undec-7-ene (DBU) (332 μL, 2 equiv.) were added to the reaction mixture that was stirred at 40 °C for 22 h. The reaction was quenched with ice-water (10 mL) and extracted with EtOAc (3χ20 mL). The combined organic phases were washed with a saturated solution OfNaHCO3 (2x20 mL), brine (20 mL), dried (Na2SO4), filtered and evaporated in vacuo to give a yellow oil, dihexyl-5-(tetrahydropyran-2- yloxymethyl)isophthalate 5bl. The crude diesters were used in the subsequent deprotection step without further purification. Synthesis of 6/s(3-cyanobenzyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5a2
Method A was used except that 3 equiv. of 3-cyanobenzyl bromide was used and the reaction mixture was stirred at 85 °C overnight. A yellow oil was obtained.
Synthesis of 6/s(3-trifluoromethylbenzyl)-5-(tetrahydropyran-2-yloxymethyl)- isophthalate, 5a3
Method A was used except that 3-(trifluoromethyl)benzyl chloride (3 equiv.) was used and the reaction was carried out in a 5 x larger scale. The reaction mixture was stirred at 80 °C for 2.5 h. A brown oil was obtained.
Synthesis of 6/s(3-nitrobenzyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5a4
Method A was used except that 3 equiv. of 3-nitrobenzyl chloride was used and the reaction mixture was stirred at 80 °C for 1 h. A yellow solid was obtained.
Synthesis of 6/s(3-chlorobenzyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5a5
Method A was used except that 3-chlorobenzyl bromide (2.2 equiv.) was used and the reaction mixture was stirred for 100 min. A red oil was obtained.
Synthesis of dibenzyl-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5a6
Method A was used except that benzyl bromide was used. A yellow oil was obtained.
Synthesis of ό/s(3-methoxybenzyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate,
5a7
Method A was used except of that 3-methoxybenzyl chloride was used. A brown liquid was obtained. Synthesis of dipentyl-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5a8
Method A was used except that 1-iodopentane was used. A yellow liquid was obtained.
Synthesis of dicyclohexylmethyl-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5b2
Method B was used except that DMAP was not used but cyclohexylmethanol (2 equiv.), CDI (2 equiv.), DBU (2 equiv.) and DMF (12.5 niL) were used and the reaction was carried out in a 2 x larger scale. After all the reagents had been added, the reaction mixture was stirred at 40 °C for 19 h. DMAP (22 mg) (0.1 equiv.) was added and the reaction mixture was stirred at 40 0C for 1 h. A clear oil was obtained after work up.
Synthesis of 6/s(l-ethylpentyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5b3
Method B was used except that 3-heptanol and DMF (1 mL) was used. After all the reagents had been added, the reaction mixture was stirred at 40 °C for 43 h. A yellow oil was obtained after work up.
Synthesis of 6/s(l-methylpentyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5b4
Method B was used except that DMAP was not used but 3-heptanol (180 μL, 2 equiv.), DMF (5 mL) was used and the reaction was carried out in a 0.8 x smaller scale. The reaction mixture was stirred at 40 °C for 19 h. A yellowish oil was obtained after work up.
Synthesis of 6/s(2-methylpentyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5b5
Method B was used except that 2-methyl-l-pentanol was used and the reaction was carried out in a 1.6 x larger scale. The reaction mixture was stirred at 40 °C for 23 h. A clear oil was obtained after work up. Synthesis of føs(l-methylhexyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5b6
Method B was used except that DMAP was not used but 2-heptanol (2 equiv.), DBU (2 equiv.), CDI (2 equiv.) and DMF (12.5 mL) was used and the reaction was carried out in a 2 x larger scale. The reaction mixture was stirred at 40 °C for 18 h, DMAP (22 mg) (0.1 equiv.) was added and the reaction mixture was stirred at 40 0C for 22 h. 2-heptanol (100 μL, 0.18 equiv) added and stirred for another 22 h. An oil was obtained after work up.
Synthesis of føs(l-ethylbutyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5b7
Method B was used except that 3-hexanol and DMF (1 mL) was used. The reaction mixture was stirred at 40 °C for 43 h. A yellowish oil was obtained after work up.
Synthesis of 6/s[4-{tetrahydro-2/l-ρyra!i-2-yIoxy)bi!tyI]-5-(tetrahydropyran-2- yloxymethyl)isophthalate, 5b8
Method A was used except that 7b (2.2 equiv.) and DMF (10 mL) was used and the reaction mixture was stirred for 130 min. The reaction mixture was cooled down to rt and quenched by adding an ice-water-mixture (40 mL), extracted with EtOAc (2x40 mL), washed with saturated NaHCO3-SO lution (2x 15 mL), brine (2x15 mL), dried (Na2SO4), filtered and evaporated in vacuo to give a oil, 5b8.
Synthesis of 6/s[5-(tetrahydro-2f/-pyraii-2~yIoxy)pentyJ]-5-(tetrahydropyran-2- yloxymethyl)isophthalate, 5b9
Method A was used as for 5b8 except that 7a was used and the reaction mixture was stirred 95 min. Work up was done as in 5b8 and a reddish oil was obtained. Synthesis of fe/sf(4aS',8aS)~decahydronaphthaIen-l-yI]-5-(tetrahydropyran-2-
Method B was used except that cώ-decahydro-l-naphthol, DBU (3 equiv.) and DMF (1 niL) were used. The reaction mixture was stirred at 40 °C for 43 h. A yellowish oil was obtained after work up.
Synthesis of dicyclohexyl-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5b 11
Method B was used except that cyclohexanol, CDI (2 equiv.), DMAP (0.05 equiv.) and DMF (3.75 mL) were used and the reaction was carried out in a 0.6 x smaller scale. The reaction mixture was stirred at 40 °C for 41 h. A yellow liquid was obtained after work up.
\loxymethyl)isophthalate. 5b12
Method B was used except that tetrahydro-3-furanylmethanol, CDI (2 equiv.), DMAP (0.05 equiv.) and DMF (3.75 mL) were used and the reaction was carried out in a 0.6 x smaller scale. The reaction mixture was stirred at 40 °C for 41 h. A clear oil was obtained after work up.
'dron
Method B was used except that DMAP was not used but decahydro-2-naphthol, DBU (3 equiv.), and DMF (1 mL) were used. The reaction mixture was stirred at 40 °C for 43 h. A clear oil was obtained after work up.
Synthesis of 6/s(3-ethoxypropyl)-5-(tetrahydropyran-2-yϊoxymethyϊ)isophthalate,
Method B was used except that 3-ethoxy-l-propanol, CDI (2.5 equiv.), DMAP (0.2 equiv.), DBU (3 equiv.) and DMF (7.5 mL) were used and the reaction was carried out in a 1.2 x larger scale. The reaction mixture was stirred at 40 °C for 24 h. A yellow liquid was obtained after work up.
Synthesis of 6is|(lA)-23-dihydπ>-l//-inden-l-yl
Method B was used except that (li?)-2,3-dihydro-lH-inden-l-ol and DMF (0.8 mL) were used and the reaction was carried out in a 0.8 x smaller scale. The reaction mixture was stirred at 40 °C for 24 h. A yellow liquid was obtained after work up.
Synthesis of føs[(2R)-2-methylpentyl]-5-(tetrahydropyran-2- yloxymethyl)isophthalate, 5b 16
Method B was used except that (2i?)-2-methylpentan-l-ol 13bl6, and DMF (2 mL) were used and the reaction was carried out in a 2 x larger scale. The reaction mixture was stirred at 40 °C for 21 h. A yellow oil was obtained after work up.
Synthesis of 6/s[(2S)-2-methylpentyl] -5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5bl7
Method B was used except that (25)-2-methylpentan-l-ol 13bl7, and DMF (1 mL) were used. The reaction mixture was stirred at 40 °C for 24 h. A yellow oil was obtained after work up.
Synthesis of bis(bicyclo[2.2.1]hept-2-ylmethyl)-5-(tetrahydropyran-2- yloxymethyl)isophthalate, 5b 18
Method B was used except that 2-norbornanemethanol was used. The reaction mixture was stirred at 40 °C for 18 h. A yellow oil was obtained after work up. Synthesis of 6/s(4-methylpentyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5bl9
Method B was used except that 4-methyl-l-pentanol was used. The reaction mixture was stirred at 40 °C for 18 h. A yellow oil was obtained after work up.
Synthesis of 6/s(3-methylpentyl)-5-(tetrahydropyran-2-yloxymethyl)isophthalate, 5b20
Method B was used except that 3-methyl-l-pentanol was used. The reaction mixture was stirred at 40 °C for 18 h. A yellow oil was obtained after work up.
Step C - Method C: Synthesis of tol[4-(telrahydro-2//-pyran-2- yloxy)methyl] phenyl j -5-(tetrahydropyran-2-yloxymethyl)isophtnalate, 5cl
A solution of 7c (327 mg, 1 equiv.) in dry DMF (1 mL) was added to a mixture of 4 (200 mg, 1 equiv.), Λ/-(3-dimethylaminopropyl)-Λf'-ethylcarbodiimide hydrochloride (EDC) (301 mg, 2.2 equiv.), 1-hydroxybenzotriazole (HOBt) (212 mg, 2.2 equiv.), N- ethyldiisopropylamine (DIPEA) (273 μL, 2.2 equiv.) and dry DMF (1 mL) and the reaction mixture was stirred under argon at room temperature for 22 h. The reaction was quenched by adding ice-water (20 mL), extracted with EtOAc (2x25 mL), washed with saturated NaHCO3-solution (2x15 mL), brine (2x15 mL), dried over Na2SO4, filtered and evaporated in vacuo to give a yellow oil.
Example 2c
Synthesis of chiral intermediates - Scheme 3
Step G: Synthesis of (2Jf)-2-amino-3-phenylpropan-l-ol, 9bl6, a typical procedure
A solution of D-phenylalanine 8bl6 (16.52 g, 1 equiv.) and THF (200 mL) was cooled on ice and NaBH4 (9.08 g, 2.4 equiv.) was portionwise added. A solution of I2 (25.38 g, 1 equiv.) in THF (50 mL) was drop wise added to the cooled down reaction mixture during 135 min, the reaction mixture was stirred for 60 min during warming up to rt, re fluxed over night (19 h) and cooled down on ice. MeOH (50 niL) was dropwise added to the reaction mixture during 30 min, it was stirred at rt for 1 h, evaporated in vacuo to a grey oil, a solution of KOH (40 g) in water (200 mL) was added and it was stirred at rt for 3 h. Water (40 mL) was added to the reaction mixture and it was extracted with DCM (3χ 100 mL), washed with water (50 mL), brine (50 mL), dried (Na2SO4), filtered and evaporated in vacuo. Recrystallized from toluene gave a white solid (11.4 g, 75% yield). mp=89°C. 1H NMR (300 MHz, CDCl3) δ 7.31 (t, 2H, J=4.5 Hz), 7.23 (t, IH, J=4.5 Hz), 7.19 (d, 2H, J=4.5 Hz), 3.64 (dd, IH, J=2.4 and 6.6 Hz), 3.39 (dd, IH, J=4.5 and 6.3 Hz), 3.13 (m, IH), 2.80 (dd, IH, J=3.3 and 8.1 Hz), 2.54 (dd, IH, J=5.4 and 8.1 Hz), 1.88 (bs, 3H); 13C NMR (75.4 MHz, CDCl3) δ 138.8, 129.4, 128.8, 126.7, 66.6, 54.4, 41.2.
Synthesis of (2iy)-2-amino-3-phenylpropan-l-ol, 9bl7
The procedure for the synthesis of (2i?)-2-amino-3-phenylpropan-l-ol 9bl6, using Step G, was used except of that D-phenylalanine 8b 17 was used and the reaction was carried out in a 2 x larger scale. 9bl7 was obtained as a clear oil which was used in next reaction without further purification. 1H NMR (300 MHz, CDCl3) δ 7.35-18 (m, 5H), 3.64 (dd, IH, J=4.2 and 10.8 Hz), 3.40 (dd, IH, J=7.2 and 10.8 Hz), 3.17 (m, IH), 2.80 (dd, IH, J=5.1 and 13.5 Hz), 2.53 (dd, IH, J=9.0 and 13.5 Hz), 2.04 (bs, 3H); 13C NMR (75.4 MHz, CDCl3) δ 138.9, 129.4, 128.8, 126.6, 66.4, 54.4, 41.0.
Step H: Synthesis of (4^)-4-benzyl~lJ~oxazoliclM-2-one, 10bl6, a typical procedure
9bl6 (16.64 g, 1 equiv.), diethyl carbonate (26.67, 2 equiv.) and potassium carbonate (1.52 g, 0.1 equiv.) were mixed and heated at 130 0C using a distillation apparatus until no more EtOH was distilled off (2 h). The reaction mixture was cooled on ice, DCM (150 mL) and 1 M NaHCO3-solution (75 mL) were added, the organic phase was collected, a saturated solution OfNaHCO3 (75 mL) was added to the organic phase and the mixture was stirred at rt for 1 h. The organic phase was collected, washed with brine (75 mL), dried (Na2SO4), filtered, evaporated in vacuo, recrystallized from hexane and EtOAc to give a white solid (16.52 g, 85% yield). mp=86°C. 1H NMR (300 MHz, CDCl3) δ 7.34 ({. 211, J=4.5 Hz). 7.28 (t, IH, J=4.5 Hz), 7.18 (d, 2H, J=5.2 Hz), 5.31 (bs, IH), 4.47 (t, IH, .7=5.1 Hz), 4.16 (dd, IR j=3.3 and 5.1 Hz), 4.0Q (m. IH), 2.92-2.84 (in, 2FI). 13C NMR (75.4 MFIz. CDCl3) δ 144.2, 136.2, 129.3, 129.1, 127.5. 69.9. 54.0, 41.7. Synthesis of (4A>4-benzyl~lJ~oxazolicli8i-2-O8ie, !0b17
The procedure for the synthesis of (4i?)-4- benzyl- 1 ,3-oxazolidin-2-one 10bl6, using step H, was used except that 9M7 was used and the reaction was carried out in a 1.8 x larger scale. After evaporated in vacuo toluene (200 mL) was added and the mixture was put to fridge to recrystallizc. After recrystallisation from hexane and EtOAc the product was obtained as a while solid (21.87 g, 62% yield). 1-1 NMR (300 MHz, CDCl3) δ 7.37-7.25 (m. 3H). 7.19-7.15 (m, 2H), 5.73 (bs, IH), 4.47 (app. t, IH). 4.17-4.04 (m, 2H), 2.88 (d, 2H, J=6.6 Hz). 13C NMR (75.4 MHz, CDCl3) δ 159.6, 136.1. 129.2. 127.4, 69.8, 54.0, 41.6; DEPT: 129.2 (CH), 127.4 (CH ), 69.8 (CH2), 54.0 (CH), 41.6 (CH2).
Sfep T: Synthesis of (4i?)-4-benz\I-3-pentanoyI-i,3-oxaz;oIidin-2-one, I IMό, a typical
BuLi (2.5 M in hexane, 37.30 mL, 2.5 equiv.) was added dropwise during 45 min to a cooled down (-78 °C) solution of 10b16 (16.52 g, 1 equiv.) in dry THF (200 mL). Pentanoyl chloride ( 13.28 mL, 1.2 equiv.) was added drop wise to the reaction mixture at - 78 0C during 5 min and the mixture was stirred for 15 min. then allowed to warm up to rt and stirred for 1 h. A saturated solution Of NaHCO3 (200 mL) was added to the reaction mixture and the organic phase was collected. The aqueous phase was extracted with EtOAc (2 100 mL), the organic phases were combined, dried (NaSO4), filtered and evaporated in vacuo to give a clear oil (22.50 g, 92% yield). 1H NMR (300 MHz, CDCl3) δ 7.33 (t, 2H, .7=4.5 Hz). 7.27 (t, I H, . /==4.5 Hz), 7.20 (d, 2H, J===4.5 Hz), 4.70-4.65 (m. IH), 4.21 -4.15 (ra, 2H), 3.30 (dd, 1H, J=1.8 and 8.1 Hz), 3.01-2.8 (m, 2H ), 2.77 (dd, IH, .7=13.2 Hz and 6.6 Hz). 1.71-1.65 (m, 2H), 1.42 (sext., 2H, J==4.5 Hz), 0.96 (t. 3H, .7=4.5 Hz); 13C NMR (75.4 MHz, CDCl3) δ 173.6, 153.6, 135.5, 129.6, 129.1, 127.5, 66.3, 55.3, 38.1 , 35.4, 26.6, 22.5, 14.0
Synthesis of (4S)-4-benzyl-3-pentanoyI-l,3-oxazolidin-2-one, 11M7
The procedure for the synthesis of (4i?)-4-benzyl-3-pentanoyl- l ,3-oxazolidin-2-one Ilbl6, using step 1. was used except that (4S)-4~benzyl-l ,3-oxazolidin-2~one I θbl7 (1 equiv.), pentanoic anhydride (1 equiv.), BuLi (1.5 M in hexane, 1 equiv.) and THF (100 mL) were used and the reaction was carried out in a 0.3 x smaller scale. The crude product was purified with flash chromatography using hexane-EtOAc (10:1-5:1) as eluent, and a clear oil was obtained (4.82 g, 65% yield). 1H NMR (300 MHz, CDCl1) δ 7.33 (t, 2H, J=4,5 Hz), 7.27 (t, IH, ./=4.5 Hz), 7.20 (d, 2H, J=4.5 Hz), 4.70-4.65 (m, I H), 4.21-4.15 (m, 2H), 3.30 IH, J=1.8 and 8.1 Hz), 3.01-2.87 (m. 2H), 2.77 (dd, 1H, J=13.2 Hz and 6.6 Hz), 1.71-
1.65 (m, 2H), 1.42 (sext., 2H, J=4.5 Hz), 0.96 (t, 3H, J=4.5 Hz); 13C NMR (75.4 MHz, CDCl3) δ 173.6, 153.6, 135.5, 129.6, 129.1 , 127.5, 66.3, 55.3, 38.1, 35.4, 26.6, 22.5, 14.0
Step J: Synthesis of (4if)-4-benzj1-3-{(2/f)-2-methylpentanoyI]-l,3-oxazoϊidin-2-one, 12bl6, a typical procedure
Sodium έ/ϊ(trimcthylsilyl)amidc (NaHDMS) (IM in THF, 95.59 niL, 1.11 cquiv.) was dropwise added during 2 h to a cooled down (-78 °C) solution of llblό (22.5 g, 1 equiv.) in dry THF (95 mL) and stirred at that temperature for 30 min. Methyl iodide (26.86 niL, 5 equiv.) was dropwise added during 5 min ai -78 0C and stirred for 4 h at that temperature. The reaction mixture was let to warm up to rt while saturated solution of NH4CI (165 mL) was added, the reaction mixture was evaporated in vacuo, extracted with DCM (2x150 mL) and washed with 1 M KHSCVsolution (3x100 mL). The aqueous phases were combined and were extracted with DCM (3x150), the organic phase was washed with 1 M K.HSO j-solution (3x100 mL), added to the first collected organic phase, dried (Na2SO-I), filtered and evaporated in vacuo. The crude product was purified with flash column chromatography using hexane-EtOAc (10:1) as eluent to give a clear oil (17.53 g, 74% yield). !H NMR (300 MHz, CDCh) δ 7.36-7.20 (m, 5H). 4.71-4.64 (m, IH), 4.23-4.14 (m. 2H), 3.79-3.68 (in, IH), 3.27 (dd, 1H. J=3.3 and 13.2 Hz), 2.77 (dd, 1H. J-9.6 Hz and 13.5 Hz). 1.79-1.68 (m, IH), 1.46-1.26 (m. 3H), 1.22 (d, 3H, J=6.9 Hz), 0.91 (1 3H, J=7.2 Hz): 13C NMR (75.4 MFIz, CDCl3) δ 177.5, 153.2. 135.5. 129.6, 129.1, 127.5, 66.2, 55.5, 38.1. 37.6, 35.8, 20.6, 17.5, 14.3
Synthesis of (4S)-4-benzyl-3-[(2S)-2-methylpentanoyl]-l,3-oxazolidin-2-one, 12bl7
The procedure for the synthesis of (4i?)~4-benzyl-3-[(2/?)-2-methylpentanoyl]-l ,3- oxazolidin-2-one 12bl6, using step j, was used except that Ilbl7 was used, after methyl iodide was added the reaction mixture was stirred at -78 °C for 5.5 h and the reaction was carried out in a 0.13 x smaller scale. The crude product was purified with flash column chromatography using hexanc-ElOAc (15-20% of FfOAc) as cluent to give a clear oil (2.37 g, 75% yield). 1H NMR (300 MHz, (1DCk) δ 7.36-7.20 (m, 5H), 4.71-4.64 (ra, III ), 4.23-4.14 (m, 2H). 3.79-3.67 (m, IH), 3.27 (dd, IH../=3.3 and 13.5 Hz), 2.77 (dd, IH, J 9.3 H/ and 13.5 Hz), 1.79-1.68 (ra, I II), 1.46-1.26 (m, 311), 1.22 Ul, 311, J 6.9 H/). 0.91
(t. 3H, J=7.2 Hz): HC NMR (75.4 MHz, CDCl,) ό 177.5, 153.3, 135.6, 129.6, 129.1. 127.5, 66.2. 55.6, 38.1, 37.6, 35.8, 20.6, 17.5, 14.3
A solution of lithium aluminium hydride (LAH) (3.38 g, 1.4 cquiv.) in dry ether (250 niL) was drop wise added under argon during 30 mm to a cooled down (ice bath) solution of 12bl6 (17.53 g. 1 equiv.) in dry ether (200 mL) and stirred for 2 h at that temperature. The reaction was quenched by adding brine (100 mL) dropwise during 20 rain and stirred for 30 miπ on ice. The reaction mixture was filtered, the precipitate wrashed with ether, the filtrate dried (Na2SO4), filtered, evaporated in vacuo, cooled down in the fridge, filtered and distilled two times to give a clear liquid, bp=28 °C (22 mbar). 1H NMR (300 MHz, CD2CJ2) 6 3.48-3.30 (m, 2H), 1.63-1.52 (in, 111), 1.41-1.25 (m. 3H), 1.16-1.01 (ra, 111). 0.90-0.85 (m, 6H); 1 V NMIl (75.4 MH/, CD2Cl2) δ 68.2, 35.6, 35.5, 20.2, 16.4, 14.2
Synthesis of (2S)-2-methylpentan- ! -ol, 13bl 7
The procedure for the synthesis of (2i?)~2-methylpentan~l-ol ObI 6, using Step K, was used except that (4lS)-4-benzyl-3-[(2S)-2-mcthylpentanoyll-l,3-oxazolidin-2-onc 12bl7, was used and the reaction was carried out in a 0.11 χ smaller scale. The crude product was purified by precipitating off the by-product (4S)-4-benzyl-l,3-oxazolidin-2-onc with ether, the solid was washed with pentanc and put to fridge. This procedure was repeated twice to give a clear liquid (560 mg, 75% yield). 1H NMIl (300 MHz, CD2(1I2) δ 3.47-3.30 (ra, 2H), 1.63-1.52 (m, IH), 1.41-1.25 (m, 3H). 1.16-1.01 (m, IH), 0.90-0.85 (m, 6H); πC NMR 575.4 MH/, (1D2Cl2) δ 68.2, 35.6, 35.5, 20.2, 16.4, 14.2 Example 2d
Synthesis of intermediates - Scheme 5
Synthesis of 6ιs-7V,7V-[(4-7V-phenyl)piperazin-l-yl]-5-(tetrahydropyran-2- yloxymethyl)isophthalamide, 16a
Method B was used except that 1-phenylpiperazine and DMF (1 niL) were used. The reaction mixture was stirred at 40 °C for 22 h. A yellow oil was obtained after work up.
Method I: Synthesis of TVjTV-fosP-CtrifluoromethylJbenzyll-S-Ctetrahydropyran^- yloxymethyl)isophthalamide, 16b, a typical procedure
To a solution of 4 (250 mg, 1 equiv.), DIPEA (336 μL, 2.2 equiv.) in dry DCM (15 mL) was added EDC (376 mg, 2.2 equiv.), HOBt (265 mg, 2.2 equiv.) and the reaction mixture was stirred under argon at rt for 30 min. 3-(Trifluoromethyl)benzylamine (281 μL, 2.2 equiv.) was added, the reaction mixture was stirred at rt for 3 h and at 40 0C for 40 min. DCM (30 mL) added and the reaction mixture washed with saturated NaHCO3-solution (3x 15 mL), 1 M HCl-solution (2x10 mL), water (10 mL), brine (10 mL), dried (Na2SO4), filtered and evaporated in vacuo to give a yellow oil.
Synthesis of 7V,7V-diheχyl-5-(tetrahydropyran-2-yloxymethyl)isophthalamide, 16c
Method I was used except of that hexylamine and DCM (5 mL) were used and the reaction was carried out in a 0.7 χ smaller scale. The reaction mixture was stirred at 40 0C for 22.5 h and after work up a clear oil could be obtained.
Example 2e
Synthesis of intermediates - Scheme 6
Synthesis of methyl 3,5-diaminobenzoate, 19
A solution of methyl 3,5-dinitrobenzoate 18 (904 mg), Pd/C (10%), EtOAc (20 mL) and EtOH (10 mL) was hydrogenated for 22 h. The reaction mixture was filtered through a pad of Celite 545, it was washed with MeOH and EtOAc, evaporated in vacuo to give a brown solid (656 mg, 99% yield). 1H NMR (300 MHz, DMSO-J6) δ 6.42 (d, 2H, J=I.2 Hz), 6.32 (t, IH, J=I.2 Hz), 3.82 (s, 3H); 13C NMR (75.4 MHz, DMSO-J6) δ 167.3, 149.3, 130.5, 103.6, 51.5; DEPT 103.6 (CH), 51.5 (CH3).
Example 2f
Synthesis of intermediates - Scheme 7
Step N: Synthesis of mono-methylisophthalate, 24
A mixture of NaOH (432 mg, 1.05 equiv.) in MeOH (4 mL) was dropwise added during 10 min to a solution of dimethyl isophthalate 23 (2 g, 1 equiv.) and acetone (20 mL) and stirred at rt for 21 h. NaOH (43 mg, 0.1 equiv.) was added to the reaction mixture and it was stirred for 4 h at rt. The solvents were evaporated in vacuo, water (40 mL) added, pH adjusted to 1 with cone. HCl- solution, the precipitate filtered with sinter, washed with water (4x10 mL) and dried in vacuo to give a white solid (1.81 g, 98% yield). 1H NMR (300 MHz, DMSO-J6) δ 13.32 (bs, IH), 8.49 (app. t, IH), 8.21-8.17 (m, 2H), 7.67 (app. t, IH), 3.89 (s, 3H); 13C NMR (75.4 MHz, DMSO-J6) δ 166.4, 165.5, 133.8, 133.2, 131.3, 130.0, 129.7, 129.4, 52.4
Step O: Synthesis of methyl 3-(hydroxymethyl)benzoate, 25
A solution of borane dimethyl sulphide complex (BH3-SMe2) (4.19 mL, 5.5 equiv.) in dry THF (28 mL) was dropwise added during 1 h to a cooled down (0 0C) solution of 24 (1.81 g, 1 equiv.) in dry THF (45 mL), stirred at 0 0C for 15 min, at rt for 4 h and cooled down to 0 0C. Ice (100 mL) added, washed with brine (20 mL), extracted with ether (3x30 mL), the organic phase washed with a 3% solution OfH2O2, a saturated solution OfNaHCO3 (3><15 mL), brine (2x20 mL), dried (Na2SO4), filtered and evaporated in vacuo. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-2:1) to give a clear oil (486 mg, 29% yield). 1H NMR (300 MHz, CDCl3) δ 8.03 (t, IH, J=I.8 Hz), 7.97- 7.94 (m, IH), 7.59-7.56 (app. d, IH), 7.46-7.41 (app. t, IH), 4.75 (s, 2H), 3.92 (s, 3H); 13C NMR (75.4 MHz, CDCl3) δ 167.2, 141.4, 131.6, 130.6, 129.0, 128.9, 128.2, 65.0, 52.4 Step A: Synthesis of methyl 3~(tetrahydropyran~2-yloxymethyl)benzoate, 26
Step A was used except of that 25 (476 mg, 1 equiv.), DHP (314 μL, 1.2 equiv.), PPTS (72 mg, 0.1 equiv.) and 1 ,2-dichloroethane (12 niL) were used, the reaction was carried out in a 0.13 x smaller scale and it was stirred at room temperature for 16 h. The reaction mixture was evaporated in vacuo, DCM (50 mL) added, water (50 mL) added, extracted with DCM (50 mL), washed with a saturated solution OfNaHCO3 (3x30 mL), brine (3x20 mL), dried (NaSO4), filtered and evaporated in vacuo to give a clear oil (710 mg, 99% yield). This oil was used in the next reaction without further purification. 1H NMR (300 MHz, CDCl3) δ 8.04-8.03 (m, IH), 7.97-7.94 (m, IH), 7.59-7.56 (app. d, IH), 7.45-7.39 (app. t, IH), 4.82 (d, IH, J=12.3 Hz), 4.72 (t, IH, J=3.6 Hz), 4.54 (d, IH, J=12.0 Hz), 3.95-3.84 (m, 4H), 3.59-3.52 (m, IH), 1.94-1.51 (m, 6H); 13C NMR (75.4 MHz, CDCl3) δ 167.2, 139.0, 132.4, 130.5, 129.0, 128.9, 128.6, 98.1, 94.8, 68.5, 63.6, 63.1, 62.3, 52.3, 31.1, 30.9, 30.7, 25.6, 20.0, 19.5.
Step B: Synthesis of 3-(tetrahydropyran~2-yloxymelhyI)benzoic acid, 27
Step B was used except of that a 10% solution of KOH (8.69 mL, 4 equiv.), 26 (716 mg, 1 equiv.) and MeOH (13.6 mL) were used and the reaction mixture was refluxed at 90 0C for 17 h. The reaction mixture was evaporated in vacuo, water (20 mL) was added to the oil, the mixture was washed with EtOAc (3χ20 mL), cooled down on ice, pH adjusted to 4 with a 25% solution of KHSO4, extracted with DCM (3x30 mL), washed with brine (30 mL), dried (Na2SO4), filtered, evaporated in vacuo to give a clear oil (452 mg, 67% yield). 1H NMR (300 MHz, CDCl3) δ 8.11 (s, IH), 8.03 (d, IH, J=7.5 Hz), 7.63 (d, IH, J=7.8 Hz) 7.46 (t, IH, J=7.8 Hz), 4.85 (d, IH, J=12.3 Hz), 4.75 (t, IH, J=3.6 Hz), 4.56 (d, IH, J=12.0 Hz), 3.97-3.89 (m, IH), 3.61-3.54 (m, IH), 1.92-1.53 (m, 6H); 13C NMR (75.4 MHz, CDCl3) δ 171.9, 139.1, 133.2, 129.7, 129.6, 129.5, 128.7, 98.2, 68.5, 62.3, 30.7, 25.6, 21.2, 19.4
Step P: Synthesis of hexyl 3-(tetrahydropyran-2-yIoxvmethvI)benzoate, 28a
Method B (step C) was used except of that 27 (1 equiv.), hexanol (92 μL, 1.5 equiv.), DMF (3.2 mL), CDI (93 mg, 1.2 equiv.), DBU (72 μL, 1 equiv.) and DMAP (6 mg, 0.1 equiv.) were used, the reaction was carried out in a 0.5 x smaller scale and the reaction mixture was stirred at 40 0C for 21 h. After work up a clear oil could be obtained. The crude ester was used in the subsequent deprotection step without further purification.
Synthesis of 2-methylpentyl 3-(tetrahydropvran-2-vloxymethvl)benzoate. 28b
Step P was used except of that 2-methylpentan-l-ol (90 μL, 1.5 equiv.) was used. After work up a clear oil was obtained. The crude ester was used in the subsequent deprotection step without further purification.
Synthesis of 1-ethylpentyl 3-(telrahydropyran-2-yloxymethvl)benzoate, 28c
Step P was used except of that 3-heptanol (102 μL, 1.5 equiv.) was used. After work up a clear oil could be obtained. The crude ester was used in the subsequent deprotection step without further purification.
Example 2g
Synthesis of intermediates - Scheme 8
Step Q: Synthesis of monomethyl 5-(tetrahydropyran-2-yloxymethyl)isophthalate, 31
A mixture of 2 (231 mg, 1 equiv.), KOH (39 mg, 1 equiv.) and MeOH (3 mL) was stirred at 40 0C for 20 h. The solvent was evaporated in vacuo, water (50 mL) added to the oil, it was washed with DCM (50 mL), cooled down on ice and pH adjusted to 4 with 25% KHSO4-solution. The mixture was extracted with EtOAc (2x50 mL) and evaporated in vacuo to give an oil (137 mg, 65% yield). NMR studies showed a mixture of ethyl and methyl ester (-15% and 85% respectively). This was used in the next reaction without further purification. 1H NMR (300 MHz, CDCl3) δ 11.76 (bs, IH), 8.67 (app. t, IH), 8.28 (dd, 2H, J=I.5 and 4.8 Hz), 4.88 (d, IH, J=12.3 Hz), 4.77 (t, IH, J=3.3 Hz), 4.56 (d, IH, J=12.6 Hz), 4.41 (q, 0.37H, J=7.2 Hz), 3.95 (s, 3H), 3.93-3.88 (m, IH), 3.62-3.55 (m, IH), 1.93-1.54 (m, 6H), 1.42 (t, 0.51H, J=7.2 Hz). Synthesis of methyl 3-(tetrahydropyran-2-yloxymethyl)-5-[[[3- (trifluoromethyl)phenyl] acetyl] amino] benzoate, 32
Method I was used except of that 31 (137 mg, 1 equiv.) and 1.1 equiv. of the rest of the reagents were used, the reaction mixture was stirred at rt for 160 min, at 40 0C for 40 min, and the reaction was carried out in a 0.58 x smaller scale. After work up a yellowish oil could be obtained, it was used in the next reaction without further purification.
Example 2h Synthesis of intermediates - Scheme 9
Step R: Synthesis of methyl 3-(hydroxymethyl)-5-nitrobenzoate, 35
To a cooled down (0 0C) solution of 5-nitroisophthalic acid monomethyl ester 34 (3 g, 1 equiv.) in dry THF (75 mL) was BH3-SMe2 (2.53 mL, 2 equiv.) dropwise added under argon during 10 min, the reaction mixture was let to stir at rt for 19 h, it was heated at 60 0C for 2 h, it was quenched with a solution of acetic acid (400 μL) and water (400 μL) and stirred for 40 min. A saturated solution OfNaHCO3 (5 mL) was added to the mixture, it was evaporated in vacuo, EtOAc (50 mL) was added to it and it was washed with saturated NaHCO3-solution (3x25 mL), brine (2x20 mL), dried (Na2SO4), filtered and evaporated in vacuo. The crude product was purified with flash column chromatography (hexane-EtOAc; 1 :1) to give a white solid (2.41 g, 86% yield). 1H NMR (300 MHz, CDCl3) δ 8.76 (dd, IH, J=I.5 and 2.1 Hz), 8.45-8.44 (m, IH), 8.36-8.34 (m, IH), 4.89 (d, 2H, J=0.6 Hz), 3.98 (s, 3H), 2.02 (bs, IH); 13C NMR (75.4 MHz, CDCl3) δ 165.2, 148.7, 143.7, 133.3, 132.1, 125.5, 123.7, 63.7, 53.1; DEPT 148.7, 143.7, 133.3 (CH), 132.1, 125.5 (CH), 123.7 (CH), 63.7 (CH2), 53.1 (CH3)
Step A: Synthesis of methyl 3-(tetrahydropyran-2-yloxymethyl)-5-nitrobenzoate, 36
Step A was used except of that 35 (2.05g, 1 equiv.), DCE (20 mL), DHP (1.77 mL, 2 equiv.), PPTS (0.1 equiv.) were used, the reaction was carried out in a 0.48 χ smaller scale and the reaction mixture was stirred for 25 h. The reaction was quenched by adding water (60 mL), extracted with DCM (3x60 mL), washed with saturated NaHCO3-solution (3x60 mL), brine (2x60 mL), dried (Na2SO4), filtered and evaporated in vacuo to give a yellow solid (2.84 g, 99% yield). 1H NMR (300 MHz, CDCl3) δ 8.74 (app. t, IH), 8.42-8.41 (m, IH), 8.32 (s, IH), 4.90 (d, IH, J=13.2 Hz), 4.74 (t, IH, J=3.3 Hz), 4.61 (d, IH, J=12.6 Hz), 3.97 (s, 3H), 3.90-3.83 (m, IH), 3.60-3.52 (m, IH), 1.89-1.50 (m, 6H); 13C NMR (75.4 MHz, CDCl3) δ 165.1, 148.6, 141.6, 134.1, 131.9, 126.3, 123.6, 98.6, 94.8, 67.4, 63.1, 62.5, 53.0, 30.8, 30.5, 25.6, 25.5, 19.9, 19.4; DEPT 134.1 (CH), 131.9, 126.3 (CH), 123.6 (CH), 98.6 (CH), 94.8 (CH), 67.4 (CH2), 63.1 (CH2), 62.5 (CH2), 53.0 (CH3), 30.8 (CH2), 30.5 (CH2), 25.6 (CH2), 25.4 (CH2), 19.9 (CH2), 19.4 (CH2).
Step M: Synthesis of methyl 3-(tetrahydropyran-2-yloxymethyl)-5-aminobenzoate, 37
A solution of 36 (2.80 g) in EtOH (20 mL) and THF (10 niL) was hydrogenated with Pd/C (10%) for 23 h at rt and filtered through a pad of Celite 545, it was washed with MeOH and THF and evaporated in vacuo to give a brownish liquid (2.51 g, 100% yield). The amine was used in the next reactions without further purification. 1H NMR (500 MHz, CDCl3) δ 7.44 (s, IH), 7.30 (s, IH), 6.93 (s, IH), 4.77 (d, IH, J=12 Hz), 4.74 (t, IH, J=3.5 Hz), 4.49 (d, IH, J=12.5 Hz), 3.96-3.94 (m, IH), 3.92 (s, 3H), 3.83 (bs, 2H), 3.61-3.57 (m, IH), 1.94-1.57 (m, 6H); 13C NMR (125 MHz, CDCl3) δ 167.3, 146.8, 139.8, 131.4, 119.3, 118.8, 115.2, 98.1, 68.6, 62.4, 52.3, 30.7, 25.7, 19.6; DEPT 119.2 (CH), 118.8 (CH), 115.2 (CH), 98.0 (CH), 68.6 (CH2), 62.4 (CH2), 52.4 (CH3), 30.7 (CH2), 25.6 (CH2), 19.5 (CH2)
Synthesis of methyl 3-(undec-10-enoylamino)-5-(tetrahydropyran-2- yloxymethyl)benzoate, 38a
Method I was used except of that 37 (300 mg, 1 equiv.), undecylenic acid (251 μL, 1.1 equiv.), DCM (9 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 1.25 x larger scale, and the reaction was stirred at 40 0C for 21 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 11 : 1-1 :2) to give a clear oil (245 mg, 50% yield). 1H NMR (300 MHz, CDCl3) δ 8.01 (s, IH), 7.89 (s, IH), 7.80 (s, IH), 7.74 (s, IH), 5.85-5.71 (m, 2H), 5.00-4.88 (m, 2H), 4.76 (d, 1H, J=12.6 Hz), 4.69 (t, IH, J=3.3 Hz), 4.48 (d, IH, J=12.6 Hz), 3.92-3.84 (m, 4H), 3.56-3.49 (m, IH), 2.38-2.29 (m, 2H), 2.05-1.97 (m, 2H), 1.88-1.48 (m, 6H), 1.34-1.25 (m, 12H); 13C NMR (75.4 MHz, CDCl3) δ 171.7, 166.9, 140.0, 139.3, 138.6, 131.0, 124.4, 123.6, 119.9, 114.3, 98.2, 68.4, 62.3, 52.4, 37.8, 34.0, 33.9, 30.6, 29.5, 29.4, 29.4, 29.3, 29.2, 29.0, 25.7, 25.5, 24.9, 19.4 4 mg of methyl 3-(undec-10-enoylamino)-5-methylbenzoate was separated with the flash column chromatography purification as a white solid. 1H NMR (300 MHz, CDCI3) δ 7.78 (bs, 2H), 7.60 (s, IH), 7.14 (s, IH), 5.88-5.74 (m, 2H), 5.03-4.90 (m, 2H), 3.90 (s, 3H), 2.39-2.33 (m, 5H), 2.07-2.00 (m, 2H), 1.78-1.68 (m, 2H), 1.44-1.25 (m, 12H); 13C NMR (75.4 MHz, CDCl3) δ 171.7, 167.1, 139.6, 139.4, 138.2, 130.9, 126.2, 125.2, 118.0, 114.4, 52.4, 38.0, 34.0, 30.3, 29.5, 29.5, 29.4, 29.3, 29.1, 25.7, 21.7
Synthesis of methyl 3-(nonanoylamino)-5-(tetrahydropyran-2-yloxymethyl)benzoate, 38b
Method I was used except of that 37 (600 mg, 1 equiv.), nonanoic acid (434 μL, 1 equiv.), DCM (17 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 2.5 x larger scale, and the reaction was stirred at 40 0C for 22 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 11 : 1-1 :2) to give a clear oil (531 mg, 58% yield). 1H NMR (300 MHz, CDCl3) δ 8.01 (s, IH), 7.88 (s, IH), 7.75 (s, IH), 7.63 (s, IH), 4.77 (d, IH, J=12.3 Hz), 4.69 (t, IH, J=3.6 Hz), 4.49 (d, IH, J=12.3 Hz), 3.93-3.85 (m, 4H), 3.57-3.50 (m, IH), 2.38-2.30 (m, 2H), 1.90-1.50 (m, 8H), 1.30-1.25 (m, 10H), 0.86 (t, 3H, J=6.9 Hz); 13C NMR (75.4 MHz, CDCl3) δ 171.9, 166.9, 140.0, 138.6, 131.0, 124.4, 123.6, 119.9, 98.2, 68.4, 62.4, 52.4, 37.9, 34.0, 32.0, 30.7, 29.5, 29.4, 29.4, 29.3, 29.3, 25.7, 25.6, 24.9, 22.8, 19.5, 14.2; DEPT 140.0, 138.6, 131.0, 124.4 (CH), 123.6 (CH), 119.9 (CH), 98.2 (CH), 68.4 (CH2), 62.4 (CH2), 52.4 (CH3), 37.9 (CH2), 34.0 (CH2), 32.0 (CH2), 30.7 (CH2), 29.5 (CH2), 29.4 (CH2), 29.4 (CH2), 29.3 (CH2), 29.3 (CH2), 25.7 (CH2), 25.6 (CH2), 24.9 (CH2), 22.8 (CH2), 19.5 (CH2), 14.2 (CH3); 37 mg of methyl 3-(nonanoylamino)-5-methylbenzoate was separated at the flash column chromatography purification as a brownish solid. 1H NMR (300 MHz, CDCl3) δ 7.79 (bs, 2H), 7.58 (s, IH), 7.46 (s, IH), 3.88 (s, 3H), 2.38-2.31 (m, 5H), 1.76-1.58 (m, 2H), 1.26 (bs, 10H), 0.87 (t, 3H, J=6.3 Hz).
Synthesis of methyl 3-(heptanoylamino)-5-(tetrahydropyran-2-yloxymethyl)benzoate, 38c
Method I was used except of that 37 (300 mg, 1 equiv.), heptanoic acid (177 μL, 1 equiv.), DCM (9 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 1.25 x larger scale, and the reaction was stirred at 40 0C for 22 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 11 :1-1 :2) to give a yellow oil (178 mg, 42% yield). 1H NMR (300 MHz, CDCl3) δ 8.00 (s, IH), 7.88 (s, IH), 7.77 (s, IH), 7.28 (s, IH), 4.79 (d, 1H, J=12.3 Hz), 4.71 (t, 1H, J=3.6 Hz), 4.51 (d, IH, J=12.3 Hz), 3.94-3.87 (m, 4H), 3.59-3.52 (m, IH), 2.37 (t, 2H, J=7.5 Hz), 1.91-1.52 (m, 8H), 1.42-1.26 (m, 6H), 0.89 (t, 3H, J=6.9 Hz); 13C NMR (75.4 MHz, CDCl3) δ 171.7, 166.9, 140.1, 138.5, 131.1, 124.6, 123.5, 119.9, 98.3, 68.4, 62.4, 52.4, 38.0, 31.7, 30.7, 29.1, 25.7, 25.6, 22.7, 19.5, 14.2.
8 mg of methyl 3-(heptanoylamino)-5-methylbenzoate was separated at the flash column chromatography purification as a yellow solid. 1H NMR (300 MHz, CDCl3) δ 7.78 (bs, 2H), 7.59 (s, IH), 7.24 (s, IH), 3.89 (s, 3H), 2.38-2.31 (m, 5H), 1.77-1.67 (m, 2H), 1.42- 1.28 (bs, 6H), 0.89 (t, 3H, J=6.6 Hz);13C NMR (75.4 MHz, CDCl3) δ 171.8, 167.1, 139.5, 138.2, 130.8, 126.1, 125.2, 118.0, 52.4, 38.0, 31.8, 29.1,26.0, 22.7, 21.6, 14.2
Synthesis of methyl 3-(2,2-dimethylpropanoylamino)-5-(tetrahydropyran-2- yloxymethyl)benzoate, 38d
Method I was used except of that 37 (600 mg, 1 equiv.), pivalic acid (257 μL, 1 equiv.), DCM (17 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 2.5 x larger scale, and the reaction was stirred at 40 0C for 23 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 11 : 1-0: 1) to give a yellow oil (116 mg, 15% yield). 1H NMR (300 MHz, CDCl3) δ 8.03 (t, IH, J=I.8 Hz), 7.88 (app. t, IH), 7.78 (s, IH), 7.42 (bs, IH), 4.80 (d, IH, J=12.0 Hz), 4.71 (t, IH, J=3.6 Hz), 4.51 (d, 1H, J=12.O Hz), 3.94-3.87 (m, 4H), 3.59-3.52 (m, IH), 1.91-1.52 (m, 6H), 1.32 (s, 9H);13C NMR (75.4 MHz, CDCl3) δ 177.0, 166.9, 140.0, 138.5, 131.0, 124.6, 123.8, 120.2, 98.2, 68.4, 62.4, 52.4, 39.8, 30.6, 27.7, 25.5, 19.5
Synthesis of methyl 3-(acetylamino)-5-(tetrahydropyran-2-yloxymethyl)benzoate, 38e
Method I was used except of that 37 (300 mg, 1 equiv.), acetic acid (71 μL, 1 equiv.), DCM (9 mL), 1.1 equiv. of the rest of the reagents were used, the reaction was carried out in a 1.25 x larger scale, and the reaction was stirred at 40 0C for 21 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-0:1) to give a yellow oil (44 mg, 13% yield). 1H NMR (300 MHz, CDCl3) δ 7.89 (s, 2H), 7.88 (s, IH), 7.74 (s, IH), 4.77 (d, IH, J=12.6 Hz), 4.69 (t, IH, J=3.9 Hz), 4.48 (d, IH, J=12.3 Hz), 3.93-3.85 (m, 4H), 3.57-3.50 (m, IH), 2.17 (s, 3H), 1.88-1.49 (m, 6H); 13C NMR (75.4 MHz, CDCl3) δ 169.0, 166.9, 140.1, 138.6, 131.0, 124.5, 123.7, 120.0, 98.3, 68.4, 62.4, 52.4, 30.7, 25.5, 24.6, 19.5
Example 3
Synthesis of the compounds of the invention
Synthesis of the 5-(hydroxymethyϊ)isophthalates of Formula I - Scheme 2
Step D: Deprotection of compound 5, a typical procedure
Dowex 50Wx 8 (700 mg) and MeOH (8 niL) were added to the crude 5al and the reaction mixture was stirred at 40 °C overnight. The Dowex 50Wχ8 was filtered off, and the filtrate was evaporated in vacuo. The crude IaI was purified with SiO2 column chromatography (hexane/EtOAc, 8:1-2:1) to give a yellow solid, bis(4-tert-buty\bmzyl)-5- (hydroxymethyl)isophthalate IaI (157 mg, 45% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.68-8.67 (m, IH, ArH between carbonyl groups), 8.26- 8.25 (m, 2H, 2 x ArH adjacent to the hydroxymethyl group), 7.45-7.38 (m, 8H, ArH in the p-t-butylbenzyl groups), 5.37 (s, 4 H, 2 x CO2CH2Ar), 4.80 (d, 2H, J=6.0Hz, CH2OH), 1.79 (t, 1 H, J=6.0Hz, CH2OH), 1.34 (s, 18H, 6 x CH3); 13C NMR (75.4 MHz, CDCl3) δ 165.8, 164.9, 152.1, 151.7, 141.9, 132.9, 132.4, 131.2, 130.4, 128.5, 125.8, 67.2, 64.5, 34.8, 31.5
Synthesis of ό/s(3-cyanobenzyl)-5-(hydroxymethyl)isophthalate, Ia2
The typical procedure was performed (step D) for 5a2 except that Dowex 50Wχ8 (1.2 g) was used and the reaction mixture was stirred at 40 °C for 140 min. THF and EtOAc were added to the crude reaction mixture, the Dowex 50Wχ8 was filtered off and the solvents were evaporated in vacuo. The crude product was recrystallised from hexane to give Ia2 (124 mg, 43% yield for two reaction steps).
1H NMR (300 MHz, CDCl3) δ 8.65 (app. t, IH, J=I.5 and 1.8 Hz, ArH between carbonyl groups), 8.29 (app. t, 2H, J=O.6 and 0.9 Hz, 2 x ArH adjacent to the hydroxymethyl group), 7.76 (m, 2H, 2-H-Ar in the 3-cyanobenzyl groups), 7.68 (m, 4H, 4-H-Ar and 6-H-Ar in the 3-cyanobenzyl groups), 7.53 (t, 2H, J=7.5 Hz, 5-H-Ar in the 3-cyanobenzyl groups), 5.42 (s, 4H, 2 x CO2CH2Ar), 4.85 (s, 2H, CH2OH), 1.92 (br s, IH, CH2OH); 13C NMR (75.4 MHz, CDCl3) δ 165.4, 142.5, 137.5, 132.8, 132.7, 132.3, 131.9, 130.7, 130.3, 129.8, 118.6, 113.2, 66.0, 64.3
Synthesis of 6/s(3-trifluoromethylbenzyl)-5-(hydroxymethyl)isophthalate, Ia3
The typical procedure was performed (step D) for 5a3 except of that Dowex 50Wχ8 (6.5 g) and MeOH (50 rnL) were used and the reaction mixture was stirred for 23 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 8%-66%) to give a white solid, Ia3 (765 mg, 42% yield for two reaction steps).
1H NMR (300 MHz, CDCl3) δ 8.67 (s, IH, ArH between carbonyl groups), 8.28 (s, 2H, 2 x ArH adjacent to the hydroxymethyl group), 7.71 (s, 2H, 2-H-Ar in the 3-trifluoromethyl- benzyl groups), 7.66-7.62 (m, 4H, 4-H-Ar and 6-H-Ar in the 3-trifluoromethylbenzyl groups), 7.53 (t, 2H, J=7.8 Hz, 5-H-Ar in the 3-trifluoromethylbenzyl groups), 5.44 (s, 4H, 2 x CO2CH2Ar), 4.83 (d, 2H, J=5.5 Hz, CH2OH), 1.87 (t, IH, J=5.5 Hz, CH2OH); 13C NMR (75.4 MHz, CDCl3) δ 165.5, 142.3, 136.8, 132.6, 131.9, 131.3 (q, J=32.5 Hz), 130.8, 130.3, 129.4, 125.5 (q, J=3.74 Hz), 125.3 (q, J=3.85 Hz), 124.1 (q, J=273 Hz), 66.5, 64.3
Synthesis of ό/s(3-nitrobenzyl)-5-(hydroxymethyl)isophthalate, Ia4
The typical procedure was performed (step D) for 5a4 except that Dowex 50Wχ8 (1.2 g) was used. EtOAc was added to the reaction mixture and the Dowex 50Wχ8 was filtered off. The solvents were evaporated in vacuo and the crude product was recrystallised from hexane/CHCl3/EtOAc to give a white solid, Ia4 (146 mg, 44% yield for two reaction steps).
1H NMR (300 MHz, (CD3)2SO) δ 8.43 (app. t, IH, J=1.5 and 1.8 Hz, ArH between carbonyl groups) 8.36 (app. t, 2H, J=I.5 and 1.8 Hz, 2 x ArH adjacent to the hydroxymethyl group), 8.25-8.21 (m, 4H, 2-H-Ar and 4-H-Ar in the 3-nitrobenzyl groups), 7.95 (d, 2H, J=8.1 Hz, 6-H-Ar in the 3-nitrobenzyl groups), 7.71 (t, 2H, J=8.1 Hz, 5-H-Ar in the 3-nitrobenzyl groups), 5.54 (t, IH, J=5.7 Hz, CH2OH), 5.53 (s, 4H, 2 x CO2CH2Ar), 4.64 (d, 2H, J=5.7 Hz, CH2OH); 13C NMR (75.4 MHz, (CD3)2SO) δ 164.8, 147.8, 144.7, 138.1, 134.8, 131.6, 130.2, 129.9, 128.2, 123.2, 122.8, 65.5, 61.8 Synthesis of føs(3-chlorobenzyl)-5-(hydroxymethyl)isophthalate, Ia5
The typical procedure was performed (step D) for 5a5 except that Dowex 50Wχ8 (1.5 g) and MeOH (1OmL) were used and the reaction mixture was stirred for 6 h. The crude product was purified with SiO2 column chromatography (hexane/EtOAc, 2:1) giving a white solid, Ia5 (293 mg, 92% yield for two reaction steps).
1H NMR (300 MHz, CDCl3) δ 8.66 (s, IH, ArH between carbonyl groups), 8.28 (s, 2H, 2 x ArH adjacent to the hydroxymethyl group), 7.45 (s, 2H, 2-H-Ar in the 3-chlorobenzyl groups), 7.34 (obsc. s, 6H, 4-H-Ar, 5-H-Ar, and 6-H-Ar in the 3-chlorobenzyl groups), 5.37 (s, 4H, 2 x CO2CH2Ar), 4.83 (s, 2H, CH2OH), 1.86 (br s, IH, CH2OH); 13C NMR
(75.4 MHz, CDCl3) δ 165.5, 142.2, 137.4, 134.7, 132.6, 130.9, 130.3, 130.2, 128.8, 128.6, 126.6, 66.5, 64.4
Synthesis of dibenzyl-5-(hydroxymethyl)isophthalate, Ia6
The typical procedure was performed (step D) for 5a6 but the reaction mixture was stirred for 4 h. The crude product was purified with SiO2 column chromatography (hexane/EtOAc, 2:1) giving a white solid, Ia6 (112 mg, 41% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.68 (s, IH, ArH between carbonyl groups), 8.26 (s, 2H, 2 x ArH adjacent to the hydroxymethyl group), 7.47-7.36 (m, 10 H, 10 x ArH in the benzyl groups), 5.40 (s, 4H, 2 x CO2CH2Ar), 4.80 (d, 2H, J=6.0 Hz, CH2OH), 1.84 (br s, IH, CH2OH); 13C NMR (75.4 MHz, CDCl3) δ 165.7, 142.1, 135.9, 132.4, 131.2, 130.3, 128.9, 128.6, 128.6, 67.3, 64.5
Synthesis of føs(3-methoxybenzyl)-5-(hydroxymethyl)isophthalate, Ia7
The typical procedure was performed (step D) for 5a7 except of that Dowex 50Wχ8 (2.0 g) was used. The crude product was purified first with SiO2 column chromatography (hexane/EtOAc, 2:1) to give a white solid, Ia7 (125 mg, 29% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.68 (s, IH, ArH between carbonyl groups), 8.27 (s, 2H, 2 x ArH adjacent to the hydroxymethyl group), 7.32 (t, 2H, J=8.5 Hz, 5-H-Ar in the 3- methoxybenzyl groups), 7.04 (d, 2H, J=7.0 Hz, 6-H-Ar in the 3-methoxybenzyl groups), 6.99 (s, 2H, 2-H-Ar in the 3-methoxybenzyl groups), 6.89 (dd, 2H, J=2.5 Hz, 8.5 Hz, 4-H- Ar in the 3-methoxybenzyl groups), 5.37 (s, 4H, 2 x CO2CH2Ar), 4.80 (d, 2H, J=5.5 Hz, CH2OH), 3.83 (s, 6H, OCH3), 1.84 (t, IH, J=5.5 Hz, CH2OH); 13C NMR (75.4 MHz, CDCl3) δ 165.7, 160.0, 142.1, 137.4, 132.4, 131.1, 130.3, 129.9, 120.7, 114.1, 114.0, 67.2, 64.4, 55.5
Synthesis of dihexyl-5-(hydroxymethyl)isophthalate, IbI
The typical procedure was performed (step D) for 5bl except that Dowex 50Wχ8 (1.1 g) was used. The crude product was purified with SiO2 column chromatography (hexane/EtOAc, 2:1) to give a clear oil, IbI (233 mg, 72% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.61 (s, IH, ArH between carbonyl groups), 8.24 (s, 2H, ArH adjacent to the hydroxymethyl group), 4.83 (d, 2H, J=6.0 Hz, CH2OH), 4.35 (app. t, 4H, J=7.0 Hz, CO2CH2), 1.89 (app. t, IH, J=6.0 Hz, CH2OH), 1.82-1.77 (m, 4H, OCH2CH2), 1.47-1.43 (m, 4H, CH2CH2CH2CH3), 1.38-1.35 (m, 8H, CH2CH2CH3), 0.92 (app. t, 6H, J=6.5 Hz, CH3); 13C NMR (75 MHz, CDCl3) δ 166.1, 141.9, 132.1, 131.4, 130.0, 65.8, 64.5, 31.7, 28.8, 25.9, 22.8, 14.2
Synthesis of dipentyl-5-(hydroxymethyl)isophthalate, Ia8
The typical procedure was performed (step D) for 5a8 except that Dowex 50Wχ8 (2.0 g) and MeOH (20 mL) were used. The crude product was purified with SiO2 column chromatography (hexane/EtOAc, 2:1) to give a clear oil, Ia8 (89 mg, 39% yield for two reaction steps).
1H NMR (300 MHz, CDCl3) δ 8.61 (s, IH, ArH between carbonyl groups), 8.24 (s, 2H, 2 x
ArH adjacent to the hydroxymethyl group), 4.83 (d, 2H, J=5.0 Hz, CH2OH), 4.36 (t, 4H, J=7.0 Hz, CO2CH2), 1.91 (app. t, IH, J=5.0 Hz, CH2OH), 1.83-1.77 (m, 4H, OCH2CH2),
1.46-1.38 (m, 8H, CH2CH2CH3), 0.96-0.93 (app. t, 6H, J=7.5 Hz, CH3); 13C NMR (75.4
MHz, CDCl3) δ 166.1, 142.1, 132.1, 131.3, 129.8, 65.8, 64.3, 28.5, 28.3, 22.5, 14.1
Synthesis of dicyclohexylmethyl-5-(hydroxymethyl)isophthalate, Ib2
The typical procedure was performed (step D) for 5b2 except of that Dowex 50Wχ8 (2.5 g) and MeOH (20 mL) were used. The crude product was purified with SiO2 column chromatography (hexane/EtOAc, 2:1) to give a white solid, Ib2 (242 mg, 35% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.61 (s, IH, ArH between carbonyl groups), 8.23 (s, 2H, 2 x ArH adjacent to the hydroxymethyl group), 4.83 (s, 2H, CH2OH), 4.17 (d, 4H, J=6.5 Hz, CO2CH2-C-HeX), 1.86-1.70 (m, 8H, c-hexyl-H), 1.34-1.20 (m, 8H, c-hexyl-H), 1.12-1.07 (m, 4H, c-hexyl-H), 0.98-0.90 (m, 2H, c-hexyl-H); 13C NMR (75.4 MHz, CDCl3) δ 166.0, 142.0, 132.1, 131.5, 130.0, 70.7, 64.6, 37.4, 30.0, 26.6, 25.9
Synthesis of føs(l-ethylpentyl)-5-(hydroxymethyl)isophthalate, Ib3
The typical procedure was performed (step D) for 5b3 except of that Dowex 50Wχ8 (700 mg) and MeOH (8 rnL) were used and the reaction mixture was stirred at 40 0C for 20 h.
The crude product was purified with SiO2 column chromatography (hexane/EtOAc, 2:1) to give clear oil, Ib3 (213 mg, 61% yield for two reaction steps).
1H NMR (300 MHz, CDCl3) δ 8.61 (app. tr., IH, ArH between carbonyl groups), 8.23 (m,
2H, 2 x ArH adjacent to the hydroxymethyl group), 5.10 (quin., 2H, CH, J=6.0Hz), 4.82 (s, 2H, CH2OH), 1.95 (bs, IH, OH), 1.76-1.65 (m, 8H, 2XCH2CHCH2), 1.38-1.30 (m, 8H,
2x(CH2)2CH3), 0.95 (t, 6H, J=7.5Hz, 2xCH3), 0.89 (t, 6Η, J=7.2Hz, 2xCH3); 13C NMR
(75.4 MHz, CDCl3) δ 165.8, 141.8, 132.1, 131.8, 130.1, 77.1, 64.6, 33.6, 27.7, 27.3, 22.8,
14.2, 9.9.
Synthesis of 6/s(l-methylpentyl)-5-(hydroxymethyl)isophthalate, Ib4
The typical procedure was performed (step D) for 5b4 except of that Dowex 50Wχ8 (1.1 g) and MeOH (10 mL) were used. The crude product was purified with SiO2 column chromatography (hexane/EtOAc, 2:1) to give a clear oil, Ib4 (81 mg, 31% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.56 (s, IH, ArH between carbonyl groups), 8.19 (s, 2H, 2 x ArH adjacent to the hydroxymethyl group), 5.15 (sext., 2H, J=6.3 Hz, CH), 4.80 (s, 2H, CH2OH), 2.38 (bs, IH, OH), 1.80-1.53 (m, 4H, 2xCHCH2), 1.42-1.24 (m, 14Η, 2xCH2(CH2)2CH3 and 2xCHCH3), 0.89 (t, 6Η, J=6.9 Hz); 13C NMR (75.4 MHz, CDCl3) δ 165.7, 141.9, 132.0, 131.7, 129.9, 72.6, 64.4, 35.8, 27.8, 22.7, 20.2, 14.1
Synthesis of 6/s(2-methylpentyl)-5-(hydroxymethyl)isophthalate, Ib5
The typical procedure was performed (step D) for 5b5 except of that Dowex 50Wχ8 (1 g) and MeOH (12 mL) were used and the reaction mixture was stirred for 18 h. The crude product was purified with SiO2 column chromatography hexane-EtOAc (4:1-2:1) to give a clear oil, Ib5 (328 mg, 63% yield for two reaction steps).
1H NMR (300 MHz, CDCl3) δ 8.60 (app. t, IH, ArH between carbonyl groups), 8.23 (m, 2H, 2 x ArH adjacent to the hydroxymethyl group), 4.82 (d, 2H, J=0.9 Hz, CH2OH), 4.24 (dd, 2H, J=5.4 and 10.5 Hz, CO2CH2), 4.13 (dd, 2Η, J=6.9 and 10.5 Hz, CO2CH2), 2.02- 1.91 (m, 2Η, 2χCH), 1.83 (bs, 1Η, OH), 1.51-1.20 (m, 8Η, 2χ(CH2)2CH3), 1.02 (d, 6H, J=6.6 Hz, 2xCHCH3 ), 0.92 (app.t, 6Η, J=7.2 Hz, 2xCH2CH3); 13C NMR (75.4 MHz, CDCl3) δ 166.0, 142.0, 132.1, 131.5, 129.9, 70.5, 64.5, 35.9, 32.6, 20.2, 17.2, 14.5
Synthesis of 6/s(l-methylhexyl)-5-(hydroxymethyl)isophthalate, Ib6
The typical procedure was performed (step D) for 5b6 except of that Dowex 50Wχ8 (2.8 g) and MeOH (20 mL) were used. The crude product was purified with SiO2 column chromatography hexane-EtOAc (2:1) to give a clear oil, Ib6 (56 mg, 8% yield for two reaction steps).
1U NMR (300 MHz, CDCl3) δ 8.58 (app. t, IH, ArHbetween carbonyl groups), 8.21 (app d, 2Η, 2 x ArH adjacent to the hydroxymethyl group), 5.24-5.13 (sext., 2Η, J=6.3Hz, 2H, 2xCH), 4.81 (s, 2Η, CH2OH), 1.89 (bs, IH, OH), 1.82-1.55 (m, 5Η, 2x(CH2)2), 1.44-1.27 (m, 18Η, 2x(CH2)3 and 2xCΗCH3, J=6.3Ηz), 0.88 (t, 6H, J=6.8 Hz, 2xCH2CH3); 13C NMR (75.4 MHz, CDCl3) δ 165.6, 141.8, 132.0, 131.8, 130.0, 72.6, 64.6, 36.1, 31.8, 25.3, 22.7, 20.2, 14.2.
Synthesis of ό/s(l-ethylbutyl)-5-(hydroxymethyl)isophthalate, Ib7
The typical procedure was performed (step D) for 5b7 except of that Dowex 50Wx8 (700 mg) and MeOH (8 mL) were used and the reaction mixture was stirred for 20 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 2:1) to give a clear oil, Ib7 (199 mg, 61% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.62 (app. t, IH, J=I.5 Hz, ArHbetween carbonyl groups), 8.22 (app. d, 2Η, J=1.2 Hz, 2 x ArH adjacent to the hydroxymethyl group), 5.16-5.08 (m, 2Η, 2χCH), 4.82 (s, 2Η, CH2OH), 1.86 (bs, IH, OH), 1.76-1.56 (m, 8Η, 2χCH(CH2)2), 1.48-1.30 (m, 4Η, 2xCHCH2CH3 ), 0.98-0.91 (m, 12H, 4xCH3); 13C NMR (75.4 MHz, CDCl3) δ 165.8, 141.8, 132.0, 131.8, 130.1, 76.8, 64.6, 36.1, 27.3, 18.9, 14.2, 9.9; DEPT 132.0 (CH), 130.1 (CH), 76.8 (CH), 64.6 (CH2), 36.1 (CH2), 27.3 (CH2), 18.9 (CH2), 14.2 (CH3), 9.9 (CH3).
Synthesis of 6/s(4-hydroxybutyl)-5-(hydroxymethyl)isophthalate, Ib8
The typical procedure was performed (step D) for 5b8 except of that Dowex 50Wχ8 (3.6 g) and MeOH (10 rnL) were used and the reaction mixture was stirred for 21 h. The crude product was purified with SiO2 column chromatography (CHCl3-MeOH; 10:1) to give a clear oil, Ib8 (42 mg, 17% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.57 (app. t, IH), 8.22 (s, 2H), 4.81 (s, 2H), 4.39 (t, 4H, J=6.0 Hz), 3.74 (t, 4H, J=6.0 Hz), 1.93-1.84 (m, 4H), 1.79-1.73 (m, 4H); 13C NMR (75.4 MHz, CDCl3) δ 166.0, 142.1, 132.2, 131.3, 129.9, 65.5, 64.4, 62.6. 29.5, 25.4
Synthesis of 6/s(5-hydroxypentyI)-5-(hydroxymethyl)isophthalate, Ib9
The typical procedure was performed (step D) for 5b9 except of that Dowex 50Wχ8 (3.6 g) and MeOH (10 mL) were used and the reaction mixture was stirred for 24 h. The crude product was purified with SiO2 column chromatography (CHCl3-MeOH; 10:1) to give a clear oil, Ib9 (145 mg, 55% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.54 (s, IH), 8.19 (app. t, 2H), 4.78 (s, 2H), 4.35 (t, 4H,
J=6.6 Hz), 3.67 (t, 4H, J=6.3 Hz), 2.16 (bs, 3H) 1.81 (app. quint., 4H), 1.70-1.48 (m, 8H);
13C NMR (75.4 MHz, CDCl3) δ 166.0, 142.2, 132.1, 131.2, 129.9, 65.6, 64.3, 62.8, 32.4,
28.6, 22.6
Synthesis of itøs(4-hydroxybenzyl)-5-(hydroxymethyI)isophthalate, I cI
The typical procedure was performed (step D) for 5cl except of that Dowex 50Wχ8 (3.6 g) MeOH (10 mL) and THF (5 mL) were used and the reaction mixture was stirred for 18 h. The crude product was purified with SiO2 column chromatography (CHCl3-MeOH; 10:1) to give a clear oil, IcI (63 mg, 22% yield for two reaction steps).
1H NMR (300 MHz, CDCl3) δ 8.93 (s, IH), 8.50 (app. t, 2H), 7.46 (d, 4H, J=8.7 Hz), 7.24 (d, 4H, J=8.7 Hz), 4.91 (s, 2H), 4.74 (s, 4H); 13C NMR (75.4 MHz, DMSO-J6) δ 165.6, 151.4, 145.2, 140.8, 133.8, 131.7, 131.0, 129.1, 122.6, 64.6, 63.9. Synthesis of fe/sf(4aS',8aS)~decahydronaphlhaIen-l-yI]-5-(hydroxymethyI)-
The typical procedure was performed (step D) for 5b 10 except of that Dowex 50Wχ8 (700 mg), MeOH (8 mL) and THF (1 mL) were used and the reaction mixture was stirred for 20 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 8:1) to give a white solid, IbIO (239 mg, 57% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.56 (app. t, IH, J=1.5 Hz), 8.19 (app. d, 2H, J=0.9), 5.08- 5.02 (m, 2H), 4.80 (s, 2H), 2.11 (bs, 2H), 1.87-1.35 (m, 26H), 1.26-1.21 (m, 4H); 13C NMR (75.4 MHz, CDCl3) δ 165.3, 141.9, 132.0, 131.8, 130.0, 64.5, 40.3, 40.3, 35.7, 31.8, 26.3, 26.1, 24.7, 24.3, 21.5, 20.3
Figure imgf000069_0001
The typical procedure was performed (step D) for 5b 11 except of that Dowex 50Wχ8 (2 g), MeOH (10 mL) and DCM (2 mL) were used and the reaction mixture was stirred for 17 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 4:1) to give a clear liquid, lbll (210 mg, 54% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.92 (t, IH, J=I.8 Hz), 8.21-8.20 (m, 2H), 5.08-5.00 (m, 2H), 4.81 (s, 2H), 1.98-1.32 (m, 20H);
Synthesis of 6is(telrah\drofuran-3-yImel'h\I)-5-(hydroxyraethyI)isophthaIate, 1 bl 2
The typical procedure was performed (step D) for 5bl2 except of that Dowex 50Wχ8 (600 mg) and MeOH (6 mL) were used and the reaction mixture was stirred for 19 h. The crude product was purified with SiO2 column chromatography (CHCl3-MeOH; 20:1-4:1) to give a clear oil, Ibl2 (20 mg, 10% yield for two reaction steps).
1H NMR (300 MHz, CDCl3) δ 8.55 (t, IH, J=I.8 Hz) , 8.22-8.21 (m, 2H), 4.81 (s, 2H),
4.36 (dd, 2H, J=6.6 and 10.8 Hz), 4.25 (dd, 2H, J=7.8 and 10.8 Hz), 3.95-3.88 (m, 4H), 3.83-3.75 (m, 2H), 3.71-3.66 (m, 2H), 2.81-2.67 (m, 2H), 2.36 (bs, IH), 2.18-1.68 (m, 2H),
1.79-1.68 (m, 2H); 13C NMR (75.4 MHz, CDCl3) δ 165.8, 142.3, 132.3, 131.0, 130.0, 70.7,
67.9, 67.0, 64.3, 38.5, 29.2 Synthesis of 6/s{decahydroiiaphthale5i~2-yI)-5-{hydroxymethyI)isophthalafe, IbO
The typical procedure was performed (step D) for 5bl3 except of that Dowex 50Wχ8 (700 mg), MeOH (8 mL) and THF (1 mL) were used and the reaction mixture was stirred for 20 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 8:1) to give a white solid, Ibl3 (296 mg, 71% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.55 (app. d, IH), 8.18 (s, 2H), 5.21 (bs, IH), 4.99-4.92 (m, IH), 4.78 (s, 2H), 2.11-0.89 (m, 32H); 13C NMR (75.4 MHz, CDCl3) δ 165.5, 141.9, 132.0, 131.9, 130.0, 74.7, 72.0, 64.6, 43.0, 42.5, 41.3, 39.3, 37.8, 37.6, 35.5, 35.1, 34.8, 33.9, 33.4, 32.1, 31.9, 31.8, 31.6, 30.5, 28.7, 27.1, 26.8, 26.4, 25.5, 22.9, 14.3
The typical procedure was performed (step D) for 5bl4 except of that Dowex 50Wχ8 (2 g) and MeOH (10 mL) were used, the reaction mixture was stirred for 17 h and after work up a clear oil could be obtained, Ibl4.
1H NMR (300 MHz, CDCl3) δ 8.56 (app. t, IH), 8.21 (app. t, 2H), 4.79 (s, 2H), 4.44 (t, 4H, J=6.3 Hz), 3.57 (t, 4H, J=6.3 Hz), 3.49 (q, J=7.2 Hz), 2.31 (bs, IH), 2.05 (quin, 4H, J=6.3 Hz), 1.19 (t, 6H, J=7.2); 13C NMR (75.4 MHz, CDCl3) δ 165.9, 142.1, 132.2, 131.7, 129.9, 67.2, 66.5, 64.4, 63.0, 29.3, 15.4
Synthesis of d/sI(lR)-2,3-dihydro-li/-inden-i-ylI-5-(hydroxyniethyl)isophthaIate,
The typical procedure was performed (step D) for 5b 15 except of that Dowex 50Wχ8 (700 mg), MeOH (8 mL) and THF (2 mL) were used and the reaction mixture was stirred for 21 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 8:1) to give a white solid, Ibl5 (100 mg, 32% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.57 (app. t, IH), 8.18 (app. t, 2H), 7.47 (d, 2H, J=7.5 Hz), 7.31 (d, 4H, J=3.6 Hz), 7.26-7.19 (m, 2H), 6.45 (dd, 2H, J=4.2 and 6.9 Hz), 4.73 (s, 2H), 3.24-3.14 (m, 2H), 2.98-2.89 (m, 2H), 2.67-2.55 (m, 2H), 2.29-2.19 (m, 2H); 13C NMR (75.4 MHz, CDCl3) δ 165.9, 144.7, 141.9, 141.0, 132.2, 131.3, 130.2, 129.3, 126.9, 125.9, 125.0, 79.6, 64.3, 32.5, 30.4 Synthesis of 6/s[(2J?)-methyIpentj'I]-5-(hydroxymeth\I)isophthaIate, I biό
The typical procedure was performed (step D) for 5b 16 except that the reaction was carried out in a 2 x larger scale and the reaction mixture was stirred for 20 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 4:1) to give a clear oil, Ibl6 (113 mg, 39% yield for two reaction steps).
1H NMR (500 MHz, CDCl3) δ 8.61 (s, IH), 8.23 (m, 2H), 4.82 (s, 2H), 4.24 (dd, 2H, J=6.0 and 10.5 Hz), 4.13 (dd, 2H, J=6.9 and 10.5 Hz), 2.02-1.91 (m, 2H), 1.80 (bs, IH), 1.50- 1.22 (m, 8H), 1.02 (d, 6H, J=6.6 Hz), 0.92 (t, 6H, J=7.2 Hz); 13C NMR (75.4 MHz, CDCl3) δ 166.0, 142.0, 132.1, 131.5, 129.9, 70.5, 64.5, 35.9, 32.6, 20.2, 17.2, 14.4
The typical procedure was performed (step D) for 5b 17 and the reaction mixture was stirred for 21 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 8:1) to give a clear oil, Ibl7 (171 mg, 53% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.58 (app. t, IH), 8.21 (app. d, 2H), 4.80 (s, 2H), 4.23 (dd, 2H, J=5.7 and 10.8 Hz), 4.12 (dd, 2H, J=6.9 and 10.8 Hz), 2.22 (bs, IH), 2.01-1.90 (m, 2H), 1.50-1.21 (m, 8H), 1.01 (d, 6H, J=6.9 Hz), 0.92 (t, 6H, J=6.9 Hz); 13C NMR (75.4 MHz, CDCl3) δ 166.0, 142.1, 132.1, 131.4, 129.9, 70.5, 64.5, 35.8, 32.6, 20.2, 17.2, 14.4
Svnthesis of djs(bicyclo[2.2.1]hept-2-ylmethyl)-5-(hydroxv5Bethyl)isoρhthalate, IbI S
The typical procedure was performed (step D) for 5b 18 except of that Dowex 50Wχ8 (700 mg), MeOH (8 mL) and THF (1 mL) were used and the reaction mixture was stirred for 23 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 8:1- 4:1) to give a white solid, Ibl8 (199 mg, 54% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.54 (app. t, IH), 8.18 (app. t, 2H), 4.78 (s, 2H), 4.36 (dd, 2H, J=6.6 and 11.1 Hz), 4.19 (app. dd, 2H, J=9.3 and 11.1 Hz), 4.05 (d, IH, 7.5 Hz), 2.46 (s, IH), 2.36-2.22 (m, 4H) 1.97-1.88 (m, IH), 1.82-1.72 (m, 2H), 1.59-1.09 (m, 12H), 0.81- 0.73 (m, 2H); 13C NMR (75.4 MHz, CDCl3) δ 166.1, 166.0, 142.1, 132.1, 131.3, 129.9, 68.8, 67.5, 64.4, 41.2, 39.9, 38.9, 38.7, 38.6, 36.8, 36.4, 35.4, 34.2, 33.7, 30.0, 29.8, 29.0, 22.8; DEPT: 132.1 (CH), 129.9 (CH), 68.8 (CH2), 67.5 (CH2), 64.4 (CH2), 41.2 (CH), 39.9 (CH2), 38.9 (CH), 38.7 (CH), 38.6 (CH), 36.8 (CH), 36.4 (CH), 35.4 (CH2), 34.2 (CH2), 33.7 (CH2), 30.0 (CH2), 29.8 (CH2), 29.0 (CH2), 22.8 (CH2)
Synthesis of 6js(4-methylpentyl)-5~(hyclroxymef hyl)isophthalate, 1 b!9
The typical procedure was performed (step D) for 5b 19 and the reaction mixture was stirred for 23 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 8:1-4:1) to give a white solid, Ibl9 (263 mg, 81% yield for two reaction steps). 1H NMR (300 MHz, CDCl3) δ 8.59 (app. t, IH), 8.22 (m, 2H), 4.81 (s, 2H), 4.33 (t, 4H, J=6.9 Hz), 1.86 (bs, IH), 1.84-1.74 (m, 4H), 1.69-1.55 (m, 2H), 1.35-1.28 (m, 4H), 0.92 (d, 12H, J=6.6 Hz); 13C NMR (75.4 MHz, CDCl3) δ 166.0, 141.9, 132.1, 131.4, 130.0, 66.1, 64.5, 35.2, 27.9, 26.8, 22.7; DEPT: 132.1 (CH), 130.0 (CH), 66.1 (CH2), 64.5 (CH2), 35.2 (CH2), 27.9 (CH), 26.8 (CH), 22.7 (CH3)
Synthesis of itøs(3-methylpentyl)-5-(hydroxvraethyl)isophthaIate, 1 b20
The typical procedure was performed (step D) for 5b20 and the reaction mixture was stirred for 23 h. The crude product was purified with SiO2 column chromatography (hexane-EtOAc; 8:1-4:1) to give a white solid, Ib21 (165 mg, 51% yield for two reaction steps).
1H NMR (300 MHz, CDCl3) δ 8.54 (app. t, IH), 8.19 (app. t, 2H), 4.78 (s, 2H), 4.42-4.29 (m, 4H), 2.41 (s, IH), 1.86-1.74 (m, 2H), 1.63-1.49 (m, 4H), 1.47-1.34 (m, 2H), 1.30-1.15 (m, 2H), 0.94 (d, 6H, J=6.3 Hz), 0.89 (t, 6H, J=7.2 Hz); 13C NMR (75.4 MHz, CDCl3) δ 166.0, 142.1, 132.1, 131.3, 129.8, 64.4, 64.3, 35.2, 31.7, 29.5, 19.3, 11.4; DEPT: 132.1 (CH), 129.8 (CH), 64.4 (CH2), 64.3 (CH2), 35.2 (CH2), 31.7 (CH), 29.6 (CH2), 19.3 (CH3), 11.4 (CH3) Example 3b
Synthesis of the isophthalates of Formula I - Scheme 4
Step L - Method H: Synthesis of ό/s(2-methylpentyl)isophthalate, 15a, a typical procedure
Method B (step C) was used except that isophthalic acid 14a (200 mg, 1 equiv.), CDI (2 equiv.), DMF (8 niL), DBU (2 equiv.), DMAP (0.05 equiv.) and 2-methyl-pentanol (3 equiv.) were used and the reaction was carried out in a 1.3 x larger scale. The reaction mixture was stirred at 40 °C for 20 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1) to give clear oil (42 mg, 11% yield). 1H NMR (300 MHz, CDCl3) δ 8.69 (t, IH, J=I.8 Hz), 8.30 (dd, 2H, J=I.8 and 7.8 Hz), 7.54 (t, 1H,J=7.8 Hz), 4.24 (dd, 2H, J=5.7 and 10.5 Hz), 4.14 (dd, 2H, J=6.6 and 10.5 Hz), 2.02-1.91 (m, 2H), 1.52-1.20 (m, 8H), 1.03 (d, 6H, J=6.6 Hz), 0.93 (app. t, 6H, J=7.2 Hz); 13C NMR (75.4 MHz, CDCl3) δ 166.1, 133.9, 131.2, 130.8, 128.8, 70.4, 35.9, 32.7, 20.2, 17.2, 14.5
Synthesis of føs(2-methylpentyl)-5-methylisophthalate, 15b
Method H was used except that 5-methylisophthalic acid 14b (217 mg, 1 equiv.) was used. The crude product was purified with flash column chromatography (hexane-EtOAc; 4: 1) to give clear oil (116 mg, 28% yield). 1H NMR (300 MHz, CDCl3) δ 8.48 (bs, IH), 8.03 (app. t, 2H), 4.23 (dd, 2H, J=5.7 and 10.8 Hz), 4.12 (dd, 2H, J=6.6 and 10.8 Hz), 2.46 (s, 3H),
2.01-1.91 (m, 2H), 1.51-1.19 (m, 8H), 1.02 (d, 6H, J=7.2 Hz), 0.92 (app. t, 6H, J=7.2 Hz);
13C NMR (75.4 MHz, CDCl3) δ 166.3, 138.8, 134.5, 131.1, 128.1, 70.4, 35.9, 32.6, 21.4, 20.2, 17.2, 14.5
Synthesis of føs(2-methylpentyl)-5-nitroisophthalate, 15c
Method H was used except that 5-nitroisophthalic acid 14c (508 mg, 1 equiv.) was used and the reaction was carried out in a 2 x larger scale. The reaction mixture was stirred for 21 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1) to give a yellow oil (329 mg, 36% yield). 1H NMR (300 MHz, CDCl3) δ 9.01 (d, IH, J=I.5 Hz), 8.97 (app. t, 2H), 4.30 (dd, 2H, J=6.0 and 10.8 Hz), 4.20 (dd, 2H, J=6.9 and 10.8 Hz), 2.05-1.95 (m, 2H), 1.48-1.21 (m, 8H), 1.04 (d, 6H, J=6.6 Hz), 0.94 (app. t, 6H, J=I.2 Hz); 13C NMR (75.4 MHz, CDCl3) δ 164.0, 148.7, 135.9, 133.1, 128.2, 71.4, 35.8, 32.6, 20.2, 17.2, 14.4
Synthesis of 6/s(3-trifluoromethylbenzyl)-5-nitroisophthalate, 15d
Method A was used except that 5-nitroisophthalic acid 14d (508 mg, 1 equiv.), 3- (trifluoromethyl)benzyl chloride (3 equiv.) and DMF (9 rnL) were used, the reaction was carried out in a 3.4 x larger scale and stirred for 2 h at 80 0C. The crude product was purified with flash column chromatography (hexane-EtOAc; 5:1-4:1) to give a yellow liquid (433 mg, 34% yield). 1H NMR (300 MHz, CDCl3) δ 9.05 (app. t, IH), 9.02 (app. d, 2H, J=I.5 Hz), 7.72 (s, 2H), 7.66 (app. t, 4H), 7.55 (app. t, 2H), 5.49 (s, 4H);
Synthesis of methyl (2-methylpropyl)isophthalate, 15e
Method H was used except that mono methyl isophthalate (217 mg, 1 equiv.) was used and the reaction mixture was stirred for 19 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 19:1-3:2) to give a clear oil (240 mg, 76% yield). 1H NMR (300 MHz, CDCl3) δ 8.69 (app. t, IH, J=I.8 Hz), 8.23 (app. dd, 2H, J=I.8 and 7.8 Hz), 7.53 (app. t, IH, J=7.8 Hz), 4.24 (dd, 2H, J=6.0 and 10.8 Hz), 4.13 (dd, 2H, J=6.9 and 10.8 Hz), 3.95 (s, 3H), 2.02-1.92 (m, IH), 1.49-1.19 (m, 4H), 1.02 (d, 3H, J=6.9 Hz), 0.93 (app. t, 6H, J=6.9 Hz); 13C NMR (75.4 MHz, CDCl3) δ 166.5, 166.1, 134.0, 133.9, 131.2, 130.9, 130.8, 128.8, 70.5, 52.6, 35.9, 32.7, 20.2, 17.2, 14.5; DEPT 134.0 (CH), 133.9 (CH), 130.9 (CH), 128.8 (CH), 70.5 (CH2), 52.6 (CH3), 35.9 (CH2), 32.7 (CH), 20.2 (CH2), 17.2 (CH3), 14.5 (CH3).
Step M: Synthesis of føs(2-methylpentyl)-5-aminoisophthalate, 15f
A solution of 15c (80 mg), Pd/ C (10%) in EtOH (5 mL) was hydrogenated at rt for 18 h. The catalyst was filtered off with Celite 545, it was washed with MeOH, EtOAc and DCM and evaporated in vacuo. The crude product was purified with flash column chromatography (hexane-EtOAc; 19:1-3:2) to give a yellow solid (57 mg, 77% yield). 1H NMR (300 MHz, CDCl3) δ 8.06 (t, IH, J=I.5 Hz), 7.51 (d, 2H, J=I.5 Hz), 4.20 (dd, 2H, J=5.7 and 10.8 Hz), 4.10 (dd, 2H, J=6.9 and 10.8 Hz), 3.93 (bs, 2H), 2.00-1.89 (m, 2H), 1.51-1.19 (m, 8H), 1.01 (d, 6H, J=6.9 Hz), 0.92 (app. t, 6H, J=7.2 Hz); 13C NMR (75.4 MHz, CDCl3) δ 166.3, 146.9, 132.0, 120.8, 119.9, 70.3, 35.9, 32.6, 20.2, 17.2, 14.4
Example 3c Synthesis of the 5-(hydroxymethyl)isophthalamides of Formula I - Scheme 5
Synthesis of ό/s-7V,7V-[(4-7V-phenyl)piperazin-l-yl]-5-(hydroxymethyl)isophthalamide, 17a
The typical procedure for deprotection was used (step D) for 16a except that the reaction mixture was stirred for 21 h. After work up, 1 M HCl- solution (20 rnL) was used to dissolve the solid, it was washed with DCM (2x20 mL), made basic with NaHCO3 and water (20 mL) added. Extracted with DCM (3x20 mL), dried (Na2SO4), filtered and evaporated in vacuo to give a reddish solid. It was recrystallized from ether to give a pale red solid. 1H NMR (300 MHz, CDCl3) δ 7.53 (app. t, 2H), 7.38 (app. t, IH), 7.32-7.27 (m, 4H), 6.96-6.91 (m, 6H),4.78 (s, 2H), 3.95 (bs, 4H), 3.61 (bs, 4H), 3.26 (bs, 4H), 3.13 (bs, 4H), 2.16 (bs, IH); 13C NMR (75.4 MHz, CDCl3) δ 169.6, 151.0, 143.1, 136.3, 129.4, 126.7, 124.4, 121.0, 117.0, 64.1, 49.8, 47.8, 42.3
Synthesis of 7V,7V-bis [3-(trifluoromethyl)benzyl] -5-(hydroxymethyl)isophthalamide, 17b
The typical procedure for deprotection was used (step D) for 16b except that the reaction mixture was stirred for 23 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 2: 1-0: 1) to give a white solid (179 mg, 39% yield for two steps). 1H NMR (300 MHz, CD3OD) δ 8.25 (app. t, IH), 8.03 (t, 2H, J=0.9 Hz), 7.67 (s, 2H), 7.63 (d, 2H, J=7.2 Hz), 7.58-7.49 (m, 4H), 4.72 (s, 2H), 4.64 (s, 4H); 13C NMR (75.4 MHz, CD3OD) δ 169.4, 144.4, 141.6, 136.1, 132.4, 130.4, 129.7, 126.2, 125.3, 125.0, 125.0, 64.3, 44.2
Synthesis of 7V,7V-dihexyl-5-(hydroxymethyl)isophthalamide, 17c
The typical procedure for deprotection was used (step D) for 16c except that the reaction mixture was stirred for 24 h. The crude product was purified with flash column chromatography (50% EtOAc in hexane -> 100% EtOAc) to give a white solid (62 mg, 28% yield for two steps). 1H NMR (300 MHz, CDCl3) δ 7.91 (bs, IH), 7.72 (d, 2H, J=I.5 Hz), 6.74 (t, 2H, J=5.7 Hz), 4.60 (s, 2H), 3.39 (dd, 4H, J=7.2 and 13.2 Hz), 3.02 (bs, IH), 1.63-1.54 (m, 4H), 1.40-1.25 (m, 12H), 0.88 (t, 6H, J=6.9 Hz); 13C NMR (75.4 MHz, CDCl3) δ 167.3, 142.5, 135.2, 128.1, 124.4, 64.1, 40.5, 31.7, 29.7, 26.9, 22.8, 14.2
Example 3d
Synthesis of the amides of Formula I - Scheme 6
Synthesis of methyl 3,5-diheptanoylaminobenzoate, 20
Heptanoyl chloride (410 μL, 2.2 equiv.) was dropwise added under argon to a solution of 19 (200 mg, 1 equiv.), DIPEA (453 μL, 2.2 equiv.) and DCM (2 mL) and the reaction mixture was stirred at rt for 21 h. The reaction was quenched by adding DCM (15 mL) and ice-water (10 mL), the mixture was washed with water (2x10 mL), 1 M HCl-solution
(3x10 mL), saturated NaHCO3-solution (3x10 mL), brine (20 mL), dried (NaSO4), filtered and evaporated in vacuo. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-2:1) to give yellow oil (226 mg, 48% yield). 1H NMR (500 MHz, CDCl3) δ 8.40 (bs, 2H), 8.03 (s, IH), 7.89 (s, 2H), 3.81 (s, 3H), 2.32 (t, 4H, J=7.5 Hz), 1.65 (qv, 4H, J=7.5 Hz), 1.32-1.24 (m, 12H), 0.83 (t, 6H, J=7.0 Hz); 13C NMR (125 MHz, CDCl3) δ 172.6, 167.0, 138.9, 131.1, 117.0, 116.4, 52.4, 37.7, 31.7, 29.1, 25.7, 22.6, 14.1
Synthesis of.'V.iV-fS-ChydroxymethyObesizesie-iJ-diyljdlhexiinaralde, 22
To a mixture of 3,5-aminobenzyl alcohol dihydrochloride 21 (211 mg, 1 equiv.), dry pyridine (10 mL) and dry DCM (10 mL) was added heptanoyl chloride (310 μL, 2 equiv.) and the reaction mixture was stirred under argon for 20 h. The reaction mixture was evaporated in vacuo, EtOAc (30 mL) added to the oil and it was washed with 4 M HCl- solution (3x30 mL), saturated NaHCO3-solution (3x30 mL), brine (2x20 mL), dried (Na2SO4), filtered and evaporated in vacuo. The crude product was purified with flash column chromatography using hexane-EtOAc (2:1) as eluent to give a white solid. 1H NMR (300 MHz, CDCl3) δ 7.76 (s, 3H), 7.52 (s, 2H), 7.29 (app. d, 2H), 5.00 (s, 2H), 2.30 (m, 6H), 1.73-1.56 (m, 6H), 1.39-1.23 (m, 18H), 0.90-0.84 (m, 9H); 13C NMR (75.4 MHz, CDCl3) δ 174.0, 172.0, 138.9, 137.9, 115.0, 110.8, 100.3, 65.9, 38.0, 34.4, 31.7, 31.6, 29.1, 29.0, 25.7, 25.0, 22.7, 14.2.
131 mg of it was hydrolysed using EtOH (10 mL) and a 10% KOH-solution (310 μL), stirred at rt for 3.5 h and evaporated in vacuo. EtOAc (40 mL) and a saturated solution of NaHCO3 (20 mL) was added, the organic phase washed with brine (20 mL), dried (Na2SO4), filtered and evaporated in vacuo to give a white solid (124 mg, 95% yield for the last step). 1H NMR (300 MHz, CD3OD) δ 7.77 (t, IH, J=I.8 Hz), 7.32 (d, 2H, J=I.8 Hz), 4.56 (s, 2H), 2.36 (t, 4H, J=7.5 Hz), 1.74-1.64 (quin., 4H, J=7.5 Hz), 1.44-1.29 (m, 12H), 0.92 (t, 6H, J=6.9 Hz); 13C NMR (75.4 MHz, CD3OD) δ 174.8, 144.1, 140.3, 115.4, 112.1, 65.0, 38.0, 32.7, 30.0, 26.9, 23.6, 14.4; DEPT 115.4 (CH), 112.1 (CH), 65.0 (CH2), 38.0 (CH2), 32.7 (CH2), 30.0 (CH2), 26.9 (CH2), 23.6 (CH2), 14.4 (CH3).
Example 3e Synthesis of the alkyl 3-(hydroxymethyl)benzoates of Formula I - Scheme 7
Synthesis of hexyl 3-(hydroxymethyI)benzoate, 29a
Step D was used for 28a except of that the reaction was carried out in a 0.4 x smaller scale and the reaction mixture was stirred at 40 0C for 17 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-1 :1) to give clear oil (85 mg, 75% yield for two steps). 1H NMR (300 MHz, CDCl3) δ 8.02 (s, IH), 7.97-7.93 (m, IH), 7.57-7.55
(m, IH) 7.42 (t, IH, J=7.8 Hz), 4.73 (s, 2H), 4.30 (t, 2H, J=6.9 Hz), 1.80-1.71 (m, 2H),
1.46-1.22 (m, 6H), 0.87 (t, 3H, J=6.9 Hz); 13C NMR (75.4 MHz, CDCl3) δ 171.9, 139.1, 133.2, 129.7, 129.6, 129.5, 128.7, 98.2, 68.5, 62.3, 30.7, 25.6, 21.2, 19.4
Synthesis of 2-methylpentyl 3-(hydroxvmethyI)benzoate, 29b
Step D was used for 28b except of that the reaction was carried out in a 0.4 x smaller scale and the reaction mixture was stirred at 40 0C for 17 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-2:1) to give clear oil (77 mg, 68% yield for two steps). 1H NMR (300 MHz, CDCl3) δ 7.99 (s, IH), 7.95-7.16 (m, IH), 7.54 (d, IH, J=7.5 Hz), 7.40 (t, IH, J=7.5 Hz), 4.71 (s, 2H), 4.18 (dd, IH, J=5.7 and 10.8 Hz), 4.07 (dd, IH, J=6.9 and 10.8 Hz), 1.98-1.87 (m, 2H), 1.47- 1.14 (m, 4H), 1.00 (d, 3H, J=6.6 Hz), 0.91 (t, 3H, J=6.9 Hz); 13C NMR (75.4 MHz, CDCl3) δ 166.9, 141.5, 131.5, 130.8, 128.8, 128.7, 128.0, 70.2, 64.8, 35.8, 32.6, 20.1, 17.1, 14.4
Synthesis of 1-ethylpentyl 3-(hydroxymeth\I)benzoate, 29c
Step D was used for 28c except of that the reaction was carried out in a 0.4 x smaller scale and the reaction mixture was stirred at 40 0C for 17 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-2:1) to give clear oil (78 mg, 65% yield for two steps). 1H NMR (300 MHz, CDCl3) δ 7.81-7.94 (m, 3H), 7.56-7.53 (m, IH), 7.41 (t, IH, J=7.8 Hz), 5.10-5.02 (m, IH), 4.72 (s, 2H), 2.45 (bs, IH), 1.73-1.64 (m, 4H), 1.33- 1.31 (m, 4H), 0.95-0.85 (m, 6H); 13C NMR (75.4 MHz, CDCl3) δ 166.6, 141.4, 131.4, 131.2, 128.9, 128.7, 128.0, 76.5, 64.9, 33.5, 27.7, 27.2, 22.8, 14.1, 9.8
Example 3f Synthesis of 5-hydroxymethylisophthalic acid, 30
Step B was used except of that diethyl isophthalate 2 (252 mg, 1 equiv.) was used, the reaction was carried out in a 0.1 x smaller scale and it was refluxed for 2 h. The solvents were evaporated in vacuo, water (20 mL) added to the oil, washed with EtOAc (2x20 mL) and the pH of the aqueous phase was adjusted with 1 M HCl- solution to 1. Extracted with EtOAc (3x20 mL), washed with brine (4x20 mL), dried (Na2SO4), filtered and evaporated in vacuo to give a white solid (170 mg, 87% yield). 1H NMR (300 MHz, DMSO-J6) δ 13.20 (bs, 2H), 8.35 (s, IH), 8.12 (s, 2H, J=1.5 Hz), 5.45 (bs, IH), 4.62 (s, 2H); 13C NMR (75.4 MHz, CDCl3) δ 166.7, 143.9, 131.1, 131.1, 128.4, 62.0
Example 3g
Synthesis of methyl 3-(hydroxymethyl)-5-[[[3-(trifluoromethyl)phenyl] acetyl] - amino] benzoate, 33 - Scheme 8
Method D was used except of that 32 was used and the reaction mixture was stirred at 40 0C for 23 h. The crude product was purified with flash column chromatography (hexane- EtOAc; 2:1-0:1) to give a clear oil. NMR showed -16% ethyl ester and 84% methyl ester (71 mg, 41% yield for two steps). 1H NMR (300 MHz, CDCl3) δ 8.22-8.20 (m, IH), 8.02 (s, IH), 7.97 (s, IH), 7.56 (s, IH), 7.51 (d, 2H, J=7.8 Hz), 7.42 (t, 1H, J=7.5 Hz), 7.25-7.23 (m, IH), 4.65 (s, 2H), 4.62 (s, 2H), 4.32 (q, 0.45H, J=7.2 Hz), 3.86 (s, 2.44H), 3.15 (bs, IH), 1.35 (t, 0.57H, J=7.2 Hz); 13C NMR (75.4 MHz, CDCl3) δ 167.0, 166.5, 166.0, 142.6,
142.5, 139.2, 134. 6, 131.4, 131.1, 130.9, 130.7, 130.1, 130.0, 129.4, 126.9, 125.9, 124.8, 124.7, 124.7, 124.6, 124.6, 124.5, 122.3, 118.7, 64.0, 61.7, 52.6, 43.8
Example 3h
Synthesis of the methyl 3-(hydroxymethyl)-5-(alkanoylamino)benzoates of Formula I
- Scheme 9
Synthesis of methyl 3-(undec-10-enoylamino)-5-(hydroxymethyl)benzoate, 39a
Method D was used for 38a except of that the reaction was carried out in a 0.3 x smaller scale, and the reaction was stirred at 40 0C for 21 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 2:1-0:1) to give a yellow solid (28 mg, 46% yield for two steps). 1H NMR (500 MHz, CDCl3) δ 7.94 (s, IH), 7.85 (s, IH), 7.75 (s, IH), 7.71 (s, IH), 5.84-5.75 (m, IH), 5.00-4.96 (m, IH), 4.93-4.91 (m, IH), 4.66 (s, 2H), 3.88 (s, 3H), 2.43 (bs, IH), 2.35 (t, 2H, J=7.5 Hz), 2.02 (q, 2H, J=7.0 Hz), 1.70 (qv, 2H, J=7.5 Hz), 1.36-1.28 (m, 10H); 13C NMR (75.4 MHz, CDCl3) δ 172.0, 166.9, 142.6, 139.3,
138.6, 131.2, 123.7, 122.8, 120.0, 114.4, 64.7, 52.5, 37.9, 34.0, 29.5, 29.5, 29.4, 29.3, 29.1, 25.7; DEPT 139.3 (CH), 123.7 (CH), 122.8 (CH), 120.0 (CH), 114.4 (CH2), 64.7 (CH2),
52.5 (CH3), 37.9 (CH2), 34.0 (CH2), 29.5 (CH2), 29.5 (CH2), 29.4 (CH2), 29.3 (CH2), 29.1 (CH2), 25.7 (CH2).
Synthesis of methyl 3-(nonanoylamino)-5-(hydroxymethyl)benzoate, 39b
Method D was used for 38b except of that the reaction was carried out in a 0.71 x smaller scale, and the reaction was stirred at 40 0C for 21 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 2:1-1 :1) to give a white solid (57 mg, 73% yield). ). 1H NMR (300 MHz, CDCl3) δ 7.99 (s, IH), 7.95 (s, IH), 7.82 (s, IH), 7.67 (s, IH), 4.63 (s, 2H), 3.86 (s, 3H), 2.72 (bs, IH), 2.34 (t, 2H, J=7.8 Hz), 1.69 (qv, 2H, J=7.5 Hz), 1.40-1.18 (m, 10H), 0.86 (t, 3H, J=6.6 Hz); 13C NMR (75.4 MHz, CDCl3) δ 172.4, 167.0, 142.6, 138.6, 130.9, 123.6, 123.0, 120.1, 64.5, 52.5, 37.8, 32.0, 29.5, 29.5, 29.3, 25.7, 22.8, 14.2; DEPT 123.7 (CH), 123.1 (CH), 120.2 (CH), 62.6 (CH2), 52.5 (CH3), 37.8 (CH2), 32.0 (CH2), 29.6 (CH2), 29.5 (CH2), 29.4 (CH2), 25.8 (CH2), 22.9 (CH2), 14.2 (CH3) Synthesis of methyl 3-(heptanoylamino)-5-(hydroxymethyl)benzoate, 39c
Method D was used for 38c except of that the reaction was carried out in a 0.71 x smaller scale, and the reaction was stirred at 40 0C for 19 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-1 :1) to give a white solid (94 mg, 49% yield for two steps). 1H NMR (300 MHz, CDCl3) δ 8.08 (s, IH), 7.95 (s, IH), 7.80 (s, IH), 7.66 (s, IH), 4.61 (s, 2H), 3.86 (s, 3H), 2.85 (bs, IH), 2.34 (t, 2H, J=7.5 Hz), 1.68 (qv, 2H, J=7.5 Hz), 1.37-1.25 (m, 6H), 0.86 (t, 3H, J=6.9 Hz); 13C NMR (75.4 MHz, CDCl3) δ 172.5, 167.1, 142.6, 138.6, 130.9, 123.6, 123.0, 120.1, 64.5, 52.5, 37.7, 31.7, 29.1, 25.7, 22.7, 14.2
Synthesis of methyl 3-(2,2-dimethylpropanoylamino)-5-(hydroxymethyl)benzoate, 39d
Method D was used for 38d except of that the reaction was carried out in a 0.71 x smaller scale, and the reaction was stirred at 40 0C for 24 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 4:1-1 :1) to give a white solid (68 mg, 77% yield). 1H NMR (300 MHz, CDCl3) δ 7.92 (s, IH), 7.85 (s, IH), 7.71-7.70 (m, IH), 7.62 (bs, IH), 4.66 (s, 2H), 3.88 (s, 3H), 2.60 (bs, IH), 1.31 (s, 9H); 13C NMR (75.4 MHz, CDCl3) δ 177.3, 166.9, 142.6, 138.5, 131.0, 123.7, 123.2, 120.4, 64.6, 52.4, 39.8, 27.7
Synthesis of methyl 3-(acetylamino)-5-(hydroxymethyl)benzoate, 39e
Method D was used for 38e except of that the reaction was carried out in a 0.71 x smaller scale, and the reaction was stirred at 40 0C for 24 h. The crude product was purified with flash column chromatography (hexane-EtOAc; 1 :1-0:1) to give a white solid (28 mg, 89% yield). 1H NMR (300 MHz, CD3OD) δ 8.14 (t, IH, J=I.8 Hz), 7.81 (app. t, IH), 7.75 (t, IH, J=1.8 Hz), 4.63 (s, 2H), 3.90 (s, 3H), 2.14 (s, 3H); 13C NMR (75.4 MHz, CD3OD) δ 171.8, 168.3, 144.4, 140.4, 132.0, 124.2, 123.7, 120.7, 64.5, 52.7, 23.8; DEPT 124.2 (CH), 123.7 (CH), 120.7 (CH), 64.5 (CH2), 52.7 (CH3), 23.8 (CH3). Example 4 Biological tests
Example 4a Binding assays
The ability of the compounds of invention to replace radioactively labelled phorbol-12,13- dibutyrate ([3H]PDBu) from binding to recombinant PKC was assessed. Binding assays for the PKC targeted compounds of the invention were performed using five different concentrations (0.3 μM; 1 μM; 3 μM; 10 μM and 20 μM), each with three replicates on the same plate. Binding of [ H]PDBu to PKC was tested using human recombinant PKCα and PKCδ that had been produced in Sf9 insect cells. Twenty micrograms (20 μg) of protein from Sf9 cell lysate per well was incubated in a reaction mixture containing calcium, phosphatidylserine, [3H]PDBu (final cone. 25 nM) and the compounds to be tested, for 10 min in a 96-well filterplate. Poly(ethyleneglycol) was then added to precipitate the proteins and after 15 min the filters were washed and radioactivity was counted in Microbeta scintillation counter. The experiments were repeated three to seven times for each compound (Figures 1 and 2, Table I).
Table I. Inhibition of phorbol ester binding to PKCα and PKCδ by the compounds of the invention. Results are mean from 3 independent experiments.
Figure imgf000082_0001
Figure imgf000082_0002
Figure imgf000083_0001
Figure imgf000083_0002
nd = not determined nd = not determined
* = Promising binding activity * = promising binding activity
** = Some binding activity ** = some binding activity
Example 4b
Effects on PKC translocation in living cells
In the inactive state PKC isoforms are mainly localized in the cytosol. Upon activation by diacylglycerol (DAG) or phorbol esters, PKCs translocate from the cytosol to the plasma membrane or other intracellular compartments to phosphorylate their substrates. Translocation is an important step in the activation process, since it takes the enzyme to close proximity with its substrates and other proteins with which it can interact. Translocation is fairly easy to visualize with confocal microscopy by overexpressing PKC constructs that contain a fluorescent tag, e.g. GFP (green fluorescent protein), in cultured cells. Since the compounds of the invention inhibited phorbol ester binding to recombinant PKCs α and δ in vitro, we studied the effects of some of the compounds on the cellular localization of PKCs α and δ and their ability to inhibit phorbol ester induced PKC translocation in living HeLa cells (human cervical cancer cells, ATCC code CCL-2).
Method
The translocation of PKC was visualized in HeLa cells transfected with PKC-GFP constructs using confocal microscopy. During the translocation experiments the cells were maintained in DMEM in 37 0C. When the effects of the compounds of the invention on PKC localization were studied, the cells were treated with the compounds and images were captured every 30 s for 30 min. Confocal images were captured with Leica TCS SP2 AOBS confocal microscope using a 63 χ oil objective with an excitation wavelength at 488 nm and emission wavelength at 500-570 nm. In translocation inhibition assays HeLa cells expressing PKC- (GFP) constructs were pretreated for 20 to 30 minutes with the compounds of the invention and stimulated with 100 nM PMA (phorbol myristate acetate). Images were captured before stimulating the cells with PMA and after PMA addition every 30 seconds for 30 minutes. The translocation was quantified from confocal microscopic images captured during the experiments using Leica Confocal Software. The intensity of fluorescence was measured in a region of interest chosen within the cell cytoplasm (only one region of interest / cell). The area to be quantified was chosen not to contain any plasma membrane or intracellular structures throughout the experiment.
Seven of the compounds were studied in translocation experiments. Six of them had an inhibitory effect on PMA- induced PKC translocation in micromolar concentrations (Figures 3 and 4). One of the compounds (Ib7) stimulated translocation of PKCα without phorbol ester addition (not shown). These results show that the compounds of the invention are able to modulate activation / translocation of PKC in physiological conditions in living cells.
Example 4c
Effects on HeLa cell viability and proliferation
Methods The effects of nine of the compounds of the invention on HeLa cell viability and proliferation was studied using standard methylthiazoletetrazolium (MTT) and lactate dehydrogenase (LDH) tests and a continuous cell culturing platform with integrated optics and informatics (Cell-IQ®, Chip-man Technologies Ltd, Tampere, Finland). In all of the studies, normal culturing medium (DMEM, 10 % FBS, penicillin 100 U/ml and streptomycin 100 μg/ml) was used.
MTT test measures the enzymatic activity of mitochondria: NADH and NADPH that are formed by mitochondria reduce the yellow methyltiazoletetrazolium (MTT) into blue insoluble formazan crystals. Only the mitochondria of living or early apoptotic cells are able to metabolize MTT into the blue formazan. Therefore the cytotoxicity of a compound can be detected with MTT test already before the cell membrane is damaged. LDH test measures the amount of lactate dehydrogenase released from damaged cells. LDH is an enzyme that is localized in the cytosol of all mammalian cells. In cell culture, when the cell is damaged, LDH is released into the culture medium, from which its activity can be measured.
For MTT and LDH tests the cells were plated on 96-well plates and 24 hours later the medium was changed to medium containing the test compounds. After that the cells were grown in the presence of the compounds in humidified atmosphere (37 0C, 5 % CO2) for 24 or 48 hours before the tests were carried out.
For LDH test 50 μL of culture medium was transferred from treated cells into a new 96- well plate and 50 μL of substrate solution (1.3 mM β-nicotineamide adeninedinucleotide (β-NAD); 660 μM iodonitrotetrazolium (INT); 54 mM L(+)-lactic acid; 280 μM phenazine methosulphate in 0.2 M Tris-buffer; pH 8.2) was added to all wells. The plate was incubated at room temperature for 30 minutes and reaction was stopped by adding 50 μL of 1 M acetic acid. The absorbance was measured at 490 nm.
For MTT test MTT was added to the cells at 0.5 mg/mL concentration and the cells were allowed to metabolize it in 37 0C, 5 % CO2 for 2 hours. The medium was then aspirated and the blue formazan crystals were dissolved in DMSO and the absorbance was measured at the wavelength 550 nm and the absorbance at 650 nm was extracted as background.
For Cell-IQ® experiments HeLa cells were plated on 48 well plates and approximately 24 hours later medium was changed to medium containing the test compounds. The cells were then grown in Cell-IQ® (37 0C, 5 % CO2, humidified atmosphere) for 72 h and images were captured automatically from 3-4 positions in every well at one hour intervals. After the experiment total cell number in each image was counted using Cell-IQ analyzer® software. The effects of the compounds of the invention to HeLa cell proliferation were assessed based on the analysis results and the effects on HeLa cell morphology were assessed visually from images captured during the experiments.
Results The compounds of the invention showed a concentration-dependent cytotoxic effect in HeLa cells as determined with MTT test, but not with LDH test (Figures 5 and 6). This might be due to apoptotic cell death, which would impair mitochondrial metabolism but would not damage cell membrane. Preliminary results from flow cytometric studies support apoptosis as the mechanism of cell death, but other mechanisms of cell death can not be excluded.
Cell-IQ experiments showed that the compounds of the invention inhibited HeLa cell proliferation in a concentration-dependent manner. Small concentrations (0.1 μM) seemed to have no effect on proliferation, but bigger concentrations inhibited cell proliferation and caused cell death. The compounds also had an effect on morphology of HeLa cells (Figure
V). Example 4d Apoptosis
Example 4d.l Induction of apoptosis in an acute myeloid leukaemia (AML) cell line HL60
HL60 cells were cultured in RPMI 1640 medium containing 10% fetal calf serum with or without 10 μg/mL of one of the compounds Ib3, Ib5 or Ib7. Apoptosis was measured after 2 days by PI staining and assessment of the percentage of cells in the Sub-Go/Gl peak by flow cytometry. All three compounds induced apoptosis to some extent but Ib5 was the most effective (Figure 8A). Cells were also assessed for induction of the cell surface marker CDl Ib, which indicates differentiation of the cells toward either monocytes or granulocytes. Compound Ib5 did not induce differentiation, but Ib3 and Ib7 were effective indcuers of the marker of differentiation CDl Ib (Figure 8B).
Example 4d.2
Induction of apoptosis in Leukaemic blasts from patients with AML
CD34 positive leukaemic cell blasts were isolated from either blood or bone marrow of patients with acute myeloid leukaemia. Cells were cultured in medium containing IL3 and SCF to maximise survival and mimic conditions in vivo. Compounds IbS and Ib? were added to cultures at 1 and 10 μg/mL and apoptosis assessed after 2 days by P! staining and percentage of cells in the sub-Go/Gl peak by flow cytometry. 75% of cells were sensitive to compounds IbS and Ib7 at 10 μg/mL (Figure 9).
OSlS Ml
Primary B-CLL cells were obtained from the blood of patients with B-CLL. Normal B- cclls were obtained from tonsils of patients undergoing tonsilectomy. Cells were cultured in the absence of presence of a range of concentrations of compound IbS and apoptosis measured by presence of active caspasc 3 by immunostaining and flow cytometry. Compound IbS induced apoptosis in the majority of B-CLL cells at 5 and 10 μg/mL (Figure 10A) but did not kill ordinary B cells (Figure 10B).
To mimic the in vivo situation B-CLL cells were also cultured in the presence of stromal cells that were unaltered, or were expressing the B cell survival factor CD40L and the effect of 5 or 10 μg/rnL of compounds 1b3, 1b5 or 1b7 on apoptosis measured as above. Compound IbS was able to induce apoptosis in the presence of stromal cells (Figure 1 IA) and in the presence of stromal cells expressing CD40L (Figure 1 I B).
Example 4d.4
Effect of the compounds on apoptosis of activated T cells MI the presence of absence of
To determine if the compounds could have some anti-inflammatory properties their effects on the survival of human CD4 T cells activated with PHA was determined. T cells were activated and cultured in the absence (white bars) or presence of IL2 (black bars) (Figure 12 A and 12B), which provides survival signals at sites of inflammation, and a range of concentrations of 10 different compounds. Apoptosis was measured by expression of active caspase 3. Most of the compounds could induce apoptosis in T cells in the presence of 1L2, but compound IbI was the most effective, abolishing the rescue effect of IL2 at 1 μg/mL (Figure 12A).
Example 4d.S
Induction of apoptosis in promyelocytic leukaemia cells (HL-60) and mouse fibroblasts (Swiss 3T3) and in vitro toxicity of compounds Ia3 and IaI.
Cell Culture
Human promyelocytic leukaemia cells (HL-60; University of Bergen, Norway) and mouse fibroblasts (Swiss 3T3; University of Bergen, Norway) were used to examine the ability of the novel synthesized compounds to induce apoptosis since for a compound to be medically useful apoptosis induction should be selective for cancerous versus noncancerous cells. DNA binding stain was used to observe chromatin degradation and to count the ratio of apoptotic cells by fluorescence microscopy. Typical apoptotic events, dissipation of ΔΨm and increase of caspase-3 activity, were observed in the HL-60 cells treated with the most promising compounds. Cytotoxicity by necrosis was determined using the colorimetric assay, which quantitatively measures lactate dehydrogenase (LDH), a stable cytosolic enzyme that is released upon cell lysis into the supernatants. Mutagenicity of the most promising compounds was examined with a miniaturized Ames- test.
HL-60 cells were maintained in RPMI 1640 supplemented with 10% heat inactivated (56 °C for 30 min) FBS, 100 ILVmL penicillin G, and 100 μg/mL streptomycin at 37 °C in an atmosphere with 5% CO2, 95% air and 95% relative humidity. Swiss 3T3 fibroblasts were maintained in the same way but DMEM was used instead of RPMI 1640. Cells were split every two to three days as necessary.
Determination of Apoptosis Evaluation of chromatin condensation by fluorescence microscopy
To observe chromatin condensation, cells were stained with DNA-binding HOECHST 33342 fluorochrome, and the ratio of apoptotic cells was then counted using fluorescence microscopy (Nikon, Type 108, Japan). At first HL-60 leukaemia cells and Swiss 3T3 fibroblasts were harvested from the cell culture by centrifugation, 200 rcf, 4 min (Sigma 2- 5, Germany) and resuspended into fresh medium. The number of cells was determined using a haemocytometer and the cell suspension was then diluted to get 60,000 cells/mL in fibroblast experiments and 150,000 cells/mL in leukaemia cell experiments. Fibroblast cells were seeded in 48-well plates (250 μL/well) and leukaemia cells were seeded in 96- well plates (100 μL/well). Fibroblasts were incubated overnight before changing fresh media and adding 2.5 μL test compounds. In leukaemia cell experiments 1.0 μL test compounds were added to plates before cell suspension. The test compounds were dissolved into DMSO and then diluted with media so that DMSO concentration did not exceed 0.1%. Camptothecin (0.014 μM) was used as a positive control and 0.1% DMSO was used as a negative control. Cells were incubated with test compounds for 24 h at 37 °C in a humidified atmosphere with 95% air and 5% CO2. Cells were fixed with 2% formaldehyde in PBS-solution (pH 7.4) with 1 μg/mL of the HOECHST 33342 indicator. A minimum of 200 cells was evaluated with fluorescence microscopy (Nikon, type 108, Japan) from each well and the number of apoptotic cells was quantified. The first experiments were performed at concentrations of 20, 60, and 100 μM with three replicates in the same plate and repeated at least two times in different days. Apoptosis-inducing activity of Ia3 and IbI was studied further at concentrations of 10, 20, 60, 100 μM, and 10, 20, and 40 μM, respectively, in leukaemia cells. The concentrations yielding 50% inhibition (IC50) were determined by fitting the data into four parameter logistic curves using SigmaPlot2002 for Windows Version 8.0 software (SPSS Inc., Chicago, IL).
Flow cytometric analysis of changes in ΔΨm by JC-I staining
Changes in mitochondrial membrane potential (ΔΨm) were detected by flow cytometry with JC-I staining. JC-I is a cationic and lipophilic dye, which is able to enter mitochondria and form aggregates when the ΔΨm is high. As aggregates, JC-I emits light at a wavelength of 590 nm and as a monomer at 527 nm, when excited at 490 nm. HL-60 cells (1 mL, 100,000 cells/mL) were incubated with 40 μM Ia3 or 40 μM IbI for 2 h. The concentration, 40 μM, used in the experiment was selected according to preliminary experiments. Positive control cells were incubated with 10 μM valinomycin for 10 min. After exposure to test compounds, the HL-60 cells were incubated with JC-I (10 μg/mL) for 15 min at room temperature. Then the cells were washed twice with cold PBS (5 min, 400 rcf) and resuspended in 400 μL of PBS. The fluorescence intensity of the cells was measured immediately with Becton Dickinson FACScan flow cytometer (Immunocytometry Systems, San Jose, CA) equipped with a single 488 nm argon laser. Fluorescence was collected through 530/30BP and 585/42BP filters. Forward scatter and side scatter were collected by using linear amplification and the fluorescence was collected by using log amplification. A minimum of 10,000 cells per sample were analyzed. The data was acquired in list mode and analyzed with Becton Dickinson CellQuest software.
Determination of Caspase-3 activity
The activity of caspase-3 was detected by commercial EnzChek Caspase-3 Assay Kit according to the manufacturer's instructions. Briefly, ImL of HL-60 cells (100,000/mL) was incubated with 50 μM Ia3 or 50 μM IbI for 0.5, 1, 1.5, and 2 h. The concentration (50 μM) for Caspase-3 assay was selected according to preliminary experiments. Positive control cells were incubated with 5μM camptothecin. Cells were collected (5 min, 400 rcf) and washed with PBS. Then the cells were lysed in kit's lysis buffer in an ice bath for 30 min, and cellular debris was pelleted by centrifugation (5 min, 4,500 rcf). Fifty microliters of supernatant, containing caspase-3 released from the cells, was transferred to 96-well plates, and non- fluorescent substrate Z-DEVD-Rl 10 was added. Cleavage of Z-DEVD- Rl 10 to fluorescent monoamide and Rl 10 by caspase-3 activity was measured after 30 min incubation with a fluorescence microplate reader (Fluoroscan Ascent, Labsystems, Helsinki, Finland) with excitation and emission wavelengths of 485 and 538 nm, respectively.
Figure imgf000091_0001
Lactate dehydrogenase leakage from necrotic cells was determined using the commercial colorimetric cytotoxicity assay CytoTox 96® according to the manufacturer's instructions. Cells were harvested as described before. The cell density was 100,000 cells/mL in fibroblast experiments and 1 500,000 cells/mL in leukaemia cell experiments. Both cell types were incubated with test compounds for 3 h at concentrations of 20, 60, and 100 μM. A total volume of 100 μL of cell suspension was used in each well. Absorbance data were collected using a 96-well plate reader (Victor 1420 multilabel counter, PerkinElmer Life and Analytical Sciences/ Wallac Oy, Turku, Finland) at 490 nm. All assays were performed using 96-well plates with four replicates in the same plate and repeated at least two times in different days.
M ini- Ames Mutagen ici ty-Test
Genotoxic potential was evaluated with a miniaturized version of the Ames-test done in 6- well plates [Flamand et al, 2001] at a concentration of 370 μM. The concentration was chosen to be high to be sure that compounds are not mutagenic. Genotyped Salmonella typhimurium TA98 and TAlOO (Xenometric Inc., San Diego, CA) strains were used. Histidine requirement, presence or absence of R- factor plasmid, spontaneous reversion, and rfa mutation were tested with genotyped strains before use. Strains were grown for 24 h at 37 °C in nutrient broth (Becton Dickinson, Le Pont de Claix, France) before the test. 2-Aminoanthracene was used as a positive control 0.5 and 0.75 μg/well for strains TA98 and TAlOO, respectively, in the presence of 10% S9 rat liver enzyme-mix. The absence of metabolic activation by S9 fraction, 2-nitrofluorene, was used as a positive control 0.1 μg/well for strain TA98 and natriumazide 0.5 μg/well for strain TAlOO.
After incubation, the revertant colonies were counted manually. Toxicity of the test substances to the bacteria was scored as colonies against the slight background growth, and the number of revertant colonies on the plates was compared to that of the control plates. Results
Chromatin Fragmentation and Degradation, LDH Leakage, and Mutagenicity HL-60 and 3T3 Swiss cells were incubated with test compounds for 24 h and then labelled with DNA-binding HOECHST 33342 stain to observe chromatin degradation and to count the ratio of apoptotic cells by fluorescence microscopy. The results are presented in Figures 13a and b. None of the compounds were cytotoxic to either cell lines according to the LDH assay (data not shown).
Apoptosis-inducing activity of Ia3 and IbI was studied further: Dose-response relationships of Ia3 and IbI were studied in leukaemia cells and IC50 values were determined. Ia3 showed inhibition with an IC50 of 41 μM and IbI with an IC50 of 23 μM (data not shown). Chromatin condensation in HL-60 cells incubated with 20 μM Ia3 and IbI was then observed through various time points. Compounds induced apoptosis rapidly; after 2-h exposure more than 30% of cells were apoptotic (Figure 14). The Mini- Ames test did not show any mutagenicity of either compound. Cell growths on both bacterial strains were on the same level with test compounds as with the negative controls.
Changes in δψm and Caspase-3 Activity
Changes in ΔΨm, connected to induction of apoptosis by mitochondrial pathway, were analyzed by flow cytometry with JC-I staining. A clear loss of ΔΨm was observed in HL- 60 cells after 2 h incubation with 40 μM of Ia3 and IbI (Figure 15).
A time-dependent increase in the activity of caspase-3, one of the major effector caspases, was seen during the first 2 h incubation in HL-60 cells treated with 50 μM of Ia3 or IbI (Figure 16). The caspase-3 activity was clearly increased already after 1 h of incubation. An increase of caspase-3 activity was higher in cells treated with compound IbI, which is consistent with other results.
Discussion
The compounds of the present invention have proven to be potent apoptosis inducers in leukaemia cells. For example, compounds Ia3 and IbI have IC50 values of 41 and 23 μM, respectively, according to morphological evaluation. Changes in mitochondrial membrane potential and in caspase-3 activity confirmed the results. Apoptosis was induced already during the first 2 h incubation with these compounds in HL-60 cells. Although necrosis and apoptosis have some common steps, it is general agreement that apoptosis, in contrast to necrosis, is an effective energy-requiring process. Dissipation of mitochondrial membrane potential and release of cytochrome c from mitochondria appear to be key events during apoptosis. Modulators of caspase activity are increasingly gaining interest as potential targets for drug development.
The compounds showing a clear apoptosis-inducing activity on cancerous HL-60 leukaemia cells also were safe to non-malignant Swiss 3T3 fibroblasts. For a compound to be medically useful, it is critical that apoptosis induction is selective for cancerous versus non-cancerous cells. Two main problems of chemotherapy are toxicity to normal cells and failure to kill cancer cells. These results indicate also that the new hydrophobic 5- (hydroxymethyl)isophthalic acid derivatives are neither cytotoxic nor mutagenic and on this basis these compounds are promising drug candidates.
Example 5 In vivo testing
Example 5a
Use of a PKC-Cl domain binding compound, Ib5, to treat Chronic Lymphocytic Leukaemia (CLL)
Animal Model Severe combined immunodeficiency (SCID) mice engrafted with B cell leukaemic cells are employed for the study for the in vivo evaluation of the compounds of the invention. The human Raji B-cell line transfected with a plasmid expressing green fluorescent protein (2 x 106 cells) is injected intravenously into female 6-8 week old C.B.-17 SCID mice. At 72 hours after inoculation, the animals are divided into 3 equal treatment groups of 10 mice each as follows: 1) a control receives placebo (1% DMSO); 2) a positive control receives rituximab at 5mg/kg; 3) the remaining group receives compound Ib5 delivered at 50mg/kg via the tail vein and maintained every other day intravenously for 2 weeks (7 injections for each mouse). The body weight of the animals is measured once every week and tumour burden is assessed by in vivo imaging of the GFP expressing cells. All the animals are monitored daily for signs of illness and are killed immediately if hind- limb paralysis, respiratory distress, or 30% body weight loss occurs. The endpoints of the study are tumour burden (assessed by harvesting GFP positive cells from major lymphoid organs and bone marrow) and survival defined as the time for the development of hind-limb paralysis.
Administration of Experimental and Control Medications
Administration may be by any means generally understood in the art, including, but not limited to the types of administration described herein. Immediately after sacrifice, the major lymphoid organs are removed (thymus, spleen, major lymph nodes, intestinal lymph nodes, liver) and disaggregated to allow isolation of B cells by their binding to an anti-
CDl 9 antibody attached to Dynabeads. In addition, the long bones are flushed gently with saline to remove the bone marrow and the CD 19 positive cells are isolated. The fraction of leukaemic GFP positive cells in each tissue is determined by flow cytometry.
Results
The disease is expected to be inhibited in its progression in those animals receiving both the rituximab and the Ib5 compound. The inhibition or prevention may be based on the quantity of leukaemic cells (tumor burden) or survival time of the animals beyond inoculation with the leukaemic cells.

Claims

Claims
1. A compound, which modulates protein kinase activity, having the following Formula I:
Figure imgf000095_0001
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, wherein R1, R2 and R3 may be the same or different and are each independently chosen from hydrogen, branched or linear alkyl, branched or linear hydroxyalkylene, nitro, amino, - C(=O)OR4, -C(=O)NR5R6, and -NHC(=O)R7, wherein
R4 is chosen from hydrogen, a branched or linear, saturated or unsaturated Ci-Ci6 alkyl chain, a branched or linear, saturated or unsaturated Ci-Ci6 alkoxy chain, an aliphatic or aromatic, unsubstituted or substituted, mono-, di- or tricyclic ring structure connected to the -C(=O)O- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear Ci-Ci6 alkyl, branched or linear Ci-Ci6 alkoxy, branched or linear hydroxyalkylene, mono-, di- or trihalogeno- Ci-Ci6 alkylene, and wherein any of the groups R4 may possibly contain a heteroatom at any position of the group, chosen from O, S and N; R5 and R6 may be the same or different and are each independently chosen from hydrogen, a branched or linear, saturated or unsaturated Ci-Ci6 alkyl chain, a branched or linear, saturated or unsaturated Ci-Ci6 alkoxy chain, an aliphatic or aromatic, unsubstituted or substituted, mono-, di- or tricyclic ring structure connected to the -C(=O)N- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear Ci-Ci6 alkyl, branched or linear Ci-Ci6 alkoxy, branched or linear hydroxyalkylene, mono-, di- or trihalogeno- Ci-Ci6 alkylene, wherein any of the groups R5 and R6 may possibly contain a heteroatom at any position of the group, chosen from O, S and N; alternatively, R5 and R6 may be combined to form an aliphatic or aromatic, substituted or unsubstituted, mono-, di- or tricyclic ring structure connected to the -C(=O)N- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, 0 to 3 ring(s) may be aliphatic, whereas the remaining 0 to 3 ring(s) may be aromatic, and the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the substituents are chosen from the ring substituents mentioned above, and wherein the ring structure possibly also contains a further heteroatom chosen from O, S and N; and R7 is chosen from hydrogen, a branched or linear, saturated or unsaturated Ci-Ci6 alkyl chain, a branched or linear, saturated or unsaturated Ci-Ci6 alkoxy chain, an aliphatic or aromatic, unsubstituted or substituted, mono-, di- or tricyclic ring structure connected to the -NHC(=O)- group either directly or through an alkylene group, wherein, in the case of a di- or tricyclic ring structure, the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear Ci-Ci6 alkyl, branched or linear Ci-Ci6 alkoxy, branched or linear hydroxyalkylene, mono-, di- or trihalogeno- Ci-Ci6 alkylene, wherein any of the groups R7 may possibly contain a heteroatom at any position of the group, chosen from O, S and N; with the provisos that only one of R1, R2 and R3 may be hydrogen, only one of R1, R2 and R3 may be -C(=O)OC2H5, and if one of R1, R2 and R3 is a -C(=O)OC2H5 group, none of the other groups R1, R2 and R3 may be -C(=O)OCH3.
2. The compound of Claim 1, wherein one of R1, R2 and R3 is hydroxymethyl, whereas the other groups have the meanings indicated in Claim 1.
3. The compound of Claim 1, wherein two of R1, R2 and R3 are identical.
4. The compound of Claim 1, wherein R5 and R6 are different from each other.
5. The compound of any of Claims 1 to 4, wherein the number of substituents, other than hydrogen, or heteroatoms on the possible ring structures of R4, R5, R6 and R7 is limited to one.
6. The compound of any of Claims 1 to 5, wherein the substituents on the possible ring structures of R4, R5, R6 and R7 are at position 3 or 4 of the ring.
7. The compound of any of Claims 1 to 6, wherein
R1 is chosen from hydrogen, methyl, hydroxymethylene, nitro, amino and -C(=O)OR4; R2 and R3 may be the same or different and are each independently chosen from hydrogen,
-C(=O)OR4, -C(=O)NR5R6, and -NHC(=O)R7, wherein
R4 is chosen from hydrogen, a branched or linear, saturated Ci -C7 alkyl chain, a branched or linear, saturated hydroxy-Ci-Cs alkyl chain, an aliphatic or aromatic unsubstituted or substituted, mono- or dicyclic ring structure connected to the - C(=O)O- group either directly or through an alkylene group, wherein the ring substituents are chosen from the groups hydrogen, halogen, cyano, nitro, branched or linear C1-C5 alkyl, branched or linear hydroxy-Ci-Cs alkyl, branched or linear C1-C5 alkoxy, mono-, di- or trihalogeno- C1-C5 alkylene, wherein any of the groups R4 may possibly contain a heteroatom at any position of the group, chosen from O and N; R5 and R6 may be the same or different and are each independently chosen from a branched or linear, saturated Ci -Ci 6 alkyl chain, an aliphatic or aromatic unsubstituted or substituted, mono- or dicyclic ring structure connected to the - C(=O)N- group either directly or through an alkylene group, wherein the ring substituents are chosen from the groups hydrogen, mono-, di- or trihalogeno- C1-C5 alkylene; alternatively, R5 and R6 may be combined to form a substituted or unsubstituted, mono- or dicyclic ring structure connected to the -C(=O)N- group either directly or through an alkylene group, wherein, in the case of a dicyclic ring structure, 0 to 2 ring(s) may be aliphatic, whereas the remaining ring(s) may be aromatic, and the rings are either connected to each other through a bond or share a mutual ring bond or ring atom, wherein the substituents are chosen from the ring substituents mentioned above, and wherein the ring structure possibly also contains a further heteroatom, which is N; and
R7 is chosen from hydrogen and a branched or linear, saturated or unsaturated C1-C16 alkyl chain; with the provisos that only one of R1, R2 and R3 may be hydrogen, only one of R1, R2 and R3 may be -C(=O)OC2H5, and if one of R1, R2 and R3 is a -C(=O)OC2H5 group, none of the other groups R1, R2 and R3 may be -C(=O)OCH3.
8. The compound of any of claims 1 to 7, wherein at least two of R1, R2 and R3 contain an aliphatic or aromatic unsubstituted or substituted, mono- or dicyclic ring structure.
9. The compound of any of claims 1 to 7, wherein at least two of R1, R2 and R3 contain at least 6 carbon atoms.
10. The compound of any of claims 1 to 9, wherein one of R1, R2 and R3 is hydroxymethylene, while the other two are -C(=O)OR4, wherein R4 has the meaning defined in claim 1.
11. The compound of any of claims 1 to 9, wherein one of R1, R2 and R3 is hhyyddrrooxxyymmeetthhyylleennee,, wwhhiillee tthhee other two are -C(=O)NR5R6, wherein R5 and R6 have the meanings defined in claim 1.
12. The compound of any of claims 1 to 9, wherein one of R1, R2 and R3 is hydroxymethylene, while the other two may be the same or different, and are independently chosen from -C(=O)OR4 and -NHC(=O)R7, wherein R4 and R7 have the meanings defined in claim 1.
13. The compound of any of claims 1 to 12, chosen from the following compounds:
Figure imgf000099_0001
Figure imgf000100_0001
1a3 1a4
Figure imgf000100_0002
1a5 1a6
Figure imgf000100_0003
1a7 1a8
Figure imgf000100_0004
1b1 1b2
Figure imgf000100_0005
1b3 1b4
Figure imgf000100_0006
1b5 1b6
Figure imgf000101_0001
1b17 1b18
Figure imgf000101_0002
1b19 1b20
Figure imgf000101_0003
Figure imgf000102_0001
Figure imgf000103_0001
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
14. The compound of any preceding claim, wherein R > 4 is an aliphatic, branched or linear hydrocarbon chain containing at least 6 carbon atoms.
15. The compound of any preceding claim, chosen from the following compounds:
Figure imgf000103_0002
1a8 1 b1
Figure imgf000103_0003
1 b3 1 b4
Figure imgf000104_0001
Figure imgf000104_0002
1 b7 or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
16. The compound of any preceding claim, chosen from the following compounds:
Figure imgf000104_0003
1b7
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
17. The compound of any of claims 1 to 16 for use as a medicament.
18. A composition containing a therapeutically effective amount of a compound of any of claims 1 to 16 or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof and one or more pharmaceutically acceptable carriers, diluents or adjuvants.
19. The composition of Claim 18 formulated for oral administration.
20. The composition of Claim 18 formulated for intravenous administration.
21. The composition of any of claims 18 to 20 for use as a medicament.
22. The compound of any of claims 1 to 16 for regulating protein kinase activity in a subject.
23. The compound of Claim 22, wherein the subject is a mammal, particularly a human.
24. The compound of Claim 22 or 23, wherein the subject suffers from cancer, particularly leukaemia, more particularly acute myeloid leukaemia or chronic lymphatic leukaemia.
25. The compound of Claims 22 or 23, wherein the subject suffers from an inflammatory disease, particularly rheumatoid arthritis.
26. Use of the composition of any of Claims 18 to 20, containing the compound of any of Claims 1 to 16, or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, in manufacturing a medicament for the treatment or prevention of cancer.
27. The use of Claim 26, wherein the compound is one of the following:
Figure imgf000105_0001
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
28. Use of the composition of any of Claims 18 to 20, containing the compound of any of Claims 1 to 16, in manufacturing a medicament for the treatment or prevention of an inflammatory disease.
29. The use of Claim 28, wherein the compound is:
Figure imgf000106_0001
or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.
PCT/FI2009/050365 2008-05-06 2009-05-06 Protein kinase modulating agents WO2009136001A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
FI20085415 2008-05-06
FI20085415A FI20085415A0 (en) 2008-05-06 2008-05-06 Protein kinase modifying agents

Publications (1)

Publication Number Publication Date
WO2009136001A1 true WO2009136001A1 (en) 2009-11-12

Family

ID=39523070

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/FI2009/050365 WO2009136001A1 (en) 2008-05-06 2009-05-06 Protein kinase modulating agents

Country Status (2)

Country Link
FI (1) FI20085415A0 (en)
WO (1) WO2009136001A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018089433A1 (en) * 2016-11-08 2018-05-17 Navitor Pharmaceuticals, Inc. PHENYL mTORC INHIBITORS AND USES THEREOF

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002064547A2 (en) * 2001-02-14 2002-08-22 Warner-Lambert Company Llc Isophthalic acid derivatives as matrix metalloproteinase inhibitors
WO2005051914A1 (en) * 2003-11-24 2005-06-09 Merck & Co., Inc. Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
WO2007067444A1 (en) * 2005-12-08 2007-06-14 Millennium Pharmaceuticals, Inc. Bicyclic compounds with kinase inhibitory activity
WO2008004798A1 (en) * 2006-07-04 2008-01-10 Korea Research Institute Of Bioscience And Biotechnology Compounds that inhibit hif-1 activity, the method for preparation thereof and the pharmaceutical composition containing them as an effective component

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002064547A2 (en) * 2001-02-14 2002-08-22 Warner-Lambert Company Llc Isophthalic acid derivatives as matrix metalloproteinase inhibitors
WO2005051914A1 (en) * 2003-11-24 2005-06-09 Merck & Co., Inc. Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
WO2007067444A1 (en) * 2005-12-08 2007-06-14 Millennium Pharmaceuticals, Inc. Bicyclic compounds with kinase inhibitory activity
WO2008004798A1 (en) * 2006-07-04 2008-01-10 Korea Research Institute Of Bioscience And Biotechnology Compounds that inhibit hif-1 activity, the method for preparation thereof and the pharmaceutical composition containing them as an effective component

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BARKER ET AL: "Synthesis of non-symmetrical 3,5-diamidobenzyl amines, ethers and sulfides", TETRAHEDRON LETTERS, ELSEVIER, AMSTERDAM, vol. 49, no. 10, 8 January 2008 (2008-01-08), pages 1660 - 1664, XP022457903, ISSN: 0040-4039 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018089433A1 (en) * 2016-11-08 2018-05-17 Navitor Pharmaceuticals, Inc. PHENYL mTORC INHIBITORS AND USES THEREOF

Also Published As

Publication number Publication date
FI20085415A0 (en) 2008-05-06

Similar Documents

Publication Publication Date Title
EP1924548B1 (en) Ester prodrugs of prostratin and related phorbol compounds
CN101072764B (en) (-)-epigallocatechin gallate derivatives for inhibiting proteasome
EA023804B1 (en) Oxabicycloheptanes, their preparation and use
CN103781783A (en) Piperazine-substituted benzothiophene derivatives as antipsychotic agents
CA2599352A1 (en) Novel lipoxygenase inhibitors
Gao et al. Design, synthesis and evaluation of novel phenothiazine derivatives as inhibitors of breast cancer stem cells
CN104024213A (en) Synthetic epigallocatechin gallafe (EGGG) analogs
WO2000037422A2 (en) Ion channel modulating agents
JP5401652B2 (en) Protease inhibitor
US7728039B2 (en) Biphenyl and Naphthyl-Phenyl Hydroxamic Acid Derivatives
WO2021086610A1 (en) Compositions and methods of use
KR102001431B1 (en) Indoleamide compounds as an ihnibitor of cellular necrosis
EP3129014B1 (en) Novel organoselenium compounds, method for producing same, and pharmaceutical uses thereof as antitumour agents
WO2009136001A1 (en) Protein kinase modulating agents
CN115043820B (en) PAR-1 inhibitor and preparation method of PAR-1 inhibitor analogue and application of PAR-1 inhibitor and analogue in preventing and treating thrombotic diseases
CN115477608B (en) Tubulin inhibitor and preparation method and application thereof
WO2010059142A1 (en) Anthraquinone dioximes and uses thereof
US5981577A (en) α-methylene peperomins and halogenated derivatives thereof
EP2142548B1 (en) Acylated derivative of the purinic antiviral agent aciclovir and use thereof as a potent anticancer agent
JP6404220B2 (en) Condensation product of theanine derivative and carboxylic acid coumarin derivative, its intermediate, preparation method, and use thereof
JP2022500502A (en) Borate-based drugs and their use
WO2022028353A1 (en) Heterocyclic compound as bcl-2 inhibitor
JP2018537526A (en) Bortezomib complex and method of use
TWI542569B (en) Analogs of 3,5-dihydroxypentanoate for bone formation
CN101115734A (en) Spiro derivatives as lipoxygenase inhibitors

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: 09742201

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: 09742201

Country of ref document: EP

Kind code of ref document: A1