WO2022070069A1 - Dihydroorotate dehydrogenase inhibitors - Google Patents

Dihydroorotate dehydrogenase inhibitors Download PDF

Info

Publication number
WO2022070069A1
WO2022070069A1 PCT/IB2021/058906 IB2021058906W WO2022070069A1 WO 2022070069 A1 WO2022070069 A1 WO 2022070069A1 IB 2021058906 W IB2021058906 W IB 2021058906W WO 2022070069 A1 WO2022070069 A1 WO 2022070069A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
pyrazol
oxy
trifluoropropan
fluoro
Prior art date
Application number
PCT/IB2021/058906
Other languages
French (fr)
Inventor
Lindsey DERATT
Scott Kuduk
Original Assignee
Janssen Biotech, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janssen Biotech, Inc. filed Critical Janssen Biotech, Inc.
Publication of WO2022070069A1 publication Critical patent/WO2022070069A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • 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
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present invention relates to novel compounds that are dihydroorotate dehydrogenase (DHODH) inhibitors. These compounds may be useful for the treatment of a disease, disorder, or medical condition where there is an advantage in inhibiting DHODH.
  • the invention also relates to pharmaceutical compositions comprising one or more of such compounds, to processes to prepare such compounds and compositions, and to the use of such compounds or pharmaceutical compositions for the method of treatment of cancer, and autoimmune and inflammatory diseases, syndromes, and disorders.
  • DHODH dihydroorotate dehydrogenase
  • AML Acute myelogenous leukemia
  • AML is a clonal disease of the blood and bone marrow resulting from mutations that occur in normal hematopoietic stem cells.
  • AML is a heterogenous disease in that it presents with a range of cytogenetic, morphological and immunophenotypic features, and is characterized by an accumulation of clonal, abnormal myeloid progenitor cells, known as myeloblasts. These cells demonstrate disruption of normal myeloid differentiation and excessive proliferation, resulting in the decreased formation of hematopoietic cells.
  • Disease remission can be achieved with standard induction chemotherapy, but refractory and relapsed disease remains a challenge due to persistence of leukemic stem cells. Therefore, AML represents an unmet medical need with >20,000 new cases per year in the US with 5-year overall survival below 30% (Stein ET et al., Health Qual Life Outcomes 16: 193, 2018).
  • Differentiation therapy is considered an attractive approach to AML treatment based on the knowledge that differentiation and loss of stem cell self-renewal are coupled in normal cells.
  • Treatment of acute promyelocytic leukemia, which represents 10-15% of all AML, with all-trans retinoic acid is the paradigm for differentiation therapy.
  • Retinoic acid targets the promyelocytic leukemia protein (PML)-retinoic acid receptor-a (RAR-a) fusion protein encoded by a t(15, 17) chromosomal translocation.
  • PML-RAR specifically lifts the transcriptionally mediated differentiation block induced by the fusion protein and early clinical trials with single agent ATRA demonstrated complete hematologic remission in all treated patients (McCulloch D et al. Onco Targets Ther 2017; 10: 1585-1601; Nowak D et al. Blood 113: 3655, 2009).
  • DHODH dihydroorotate dehydrogenase
  • DHODH is a flavin mononucleotide (FMN) flavoprotein located in the inner mitochondrial membrane that catalyzes the oxidation of dihydroorotate to orotate, the fourth step in the de novo pyrimidine biosynthesis pathway. Inhibition of DHODH leads to decreased pyrimidine synthesis important precursors for nucleotide synthesis, but also glycoprotein and phospholipid biosynthesis (Reis RAG et al., Archives Biochem Biophysics 632: 175, 2017; Vyas VK et al., Mini Rev Med Chem 11 : 1039, 2011).
  • FMN flavin mononucleotide
  • DHODH is a validated target for the treatment of autoimmune diseases with the FDA approved small molecule DHODH inhibitors leflunomide and teriflunomide for rheumatoid arthritis and multiple sclerosis, respectively (Lolli ML et al., Recent patents on Anti-Cancer Drug Discovery 13: 86, 2018).
  • DHODH inhibitors that provide a therapeutic benefit to patients suffering from cancer and/or inflammatory and immunological diseases.
  • Embodiments of the present invention relate to compounds, pharmaceutical compositions containing them, methods of making and purifying them, methods of using them as inhibitors of DHODH enzymatic activity and methods for using them in the treatment of a subject suffering from or diagnosed with a disease, disorder, or medical condition such as autoimmune or inflammatory disorders, or diseases such as cancer.
  • Embodiments of this invention include compounds of Formula (I), wherein
  • R 1 is C 1-6 alkyl or C 1-6 haloalkyl
  • R a is C 1-6 alkyl
  • R b is C 1-6 alkyl substituted with one OH member
  • R 3 is H or halo
  • R 4 is selected from the group consisting of: wherein
  • R e is selected from the group consisting of: H; halo; C 1-6 alkyl; C 1-6 alkyl substituted with one member selected from the group consisting of: OH, OCH 3 , and OCF 3 ; C 1-6 haloalkyl; C 1-6 haloalkyl substituted with one member selected from the group consisting of: OH, and OCH 3 ; and OC 1-6 alkyl;
  • R f is selected from the group consisting of: H; halo; C 1-6 alkyl; C 1-6 alkyl substituted with one member selected from the group consisting of: OH, OCH 3 , and OCF 3 ; C 1-6 haloalkyl; C 1-6 haloalkyl substituted with one member selected from the group consisting of: OH, and OCH 3 ; and OC 1-6 alkyl; and
  • R g is H or C 1-6 alkyl; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • the present invention further provides methods for treating or ameliorating a disease, syndrome, condition, or disorder in a subject, including a mammal and/or human in which the disease, syndrome, condition, or disorder is affected by the inhibition of DHODH enzymatic activity, including but not limited to, cancer and/or inflammatory or immunological diseases, using a compound of Formula (I) or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • substituted means that the specified group or moiety bears one or more substituents.
  • unsubstituted means that the specified group bears no substituents.
  • optionally substituted means that the specified group is unsubstituted or substituted by one or more substituents. Where the term “substituted” is used to describe a structural system, the substitution is meant to occur at any valency-allowed position on the system.
  • alkyl refers to a straight- or branched-chain alkyl group having from 1 to 8 carbon atoms in the chain.
  • alkyl groups include methyl (Me), ethyl (Et), n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl (tBu), pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and groups that in light of the ordinary skill in the art and the teachings provided herein would be considered equivalent to any one of the foregoing examples.
  • C 1-6 alkyl refers to straight- or branched-chain alkyl group having from 1 to 6 carbon atoms in the chain.
  • C 1-4 alkyl refers to straight- or branched-chain alkyl group having from 1 to 4 carbon atoms in the chain.
  • cycloalkyl refers to a saturated or partially saturated, monocyclic, fused polycyclic, or spiro polycyclic carbocycle having from 3 to 12 ring atoms per carbocycle.
  • C 3 - 6cycloalkyl refers to a carbocycle having from 3 to 6 ring atoms per carbocycle.
  • Illustrative examples of cycloalkyl groups include the following entities, in the form of properly bonded moieties:
  • halogen or “halo” represents chlorine, fluorine, bromine, or iodine.
  • haloalkyl refers to a straight- or branched-chain alkyl group having from 1 to 6 carbon atoms in the chain optionally substituting hydrogens with halogens.
  • C 1-6 haloalkyl refers to a straight- or branched-chain alkyl group having from 1 to 6 carbon atoms in the chain, optionally substituting hydrogens with halogens.
  • C 1-4 haloalkyl refers to a straight- or branched-chain alkyl group having from 1 to 4 carbon atoms in the chain, optionally substituting hydrogens with halogens.
  • haloalkyl groups include trifluoromethyl (CF 3 ), difluoromethyl (CF 2 H), monofluoromethyl (CH 2 F), pentafluoroethyl (CF 2 CF 3 ), tetrafluoroethyl (CHFCF 3 ), monofluoroethyl (CH 2 CH 2 F), trifluoroethyl (CH 2 CF 3 ), tetrafluorotrifluoromethylethyl (CF(CF 3 ) 2 ), and groups that in light of the ordinary skill in the art and the teachings provided herein would be considered equivalent to any one of the foregoing examples.
  • aryl refers to a monocyclic, aromatic carbocycle (ring structure having ring atoms that are all carbon) having 6 atoms per ring. (Carbon atoms in the aryl groups are sp2 hybridized.)
  • phenyl represents the following moiety:
  • heteroaryl refers to a monocyclic or fused bicyclic heterocycle (ring structure having ring atoms selected from carbon atoms and up to four heteroatoms selected from nitrogen, oxygen, and sulfur) having from 3 to 9 ring atoms per heterocycle.
  • Illustrative examples of heteroaryl groups include the following entities, in the form of properly bonded moieties: Those skilled in the art will recognize that the species of cycloalkyl, heteroaryl and aryl groups listed or illustrated above are not exhaustive, and that additional species within the scope of these defined terms may also be selected.
  • variable point of attachment means that a group is allowed to be attached at more than one alternative position in a structure.
  • the attachment will always replace a hydrogen atom on one of the ring atoms.
  • all permutations of bonding are represented by the single diagram, as shown in the illustrations below.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • the term “treat”, “treating”, or “treatment” of any disease, condition, syndrome or disorder refers, in one embodiment, to ameliorating the disease, condition, syndrome or disorder (i.e. slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof).
  • “treat”, “treating”, or “treatment” refers to alleviating or ameliorating at least one physiological or biochemical parameter associated with or causative of the disease, condition, syndrome or disorder, including those which may not be discernible by the patient.
  • “treat”, “treating”, or “treatment” refers to modulating the disease, condition, syndrome or disorder either physically (e.g. stabilization of a discernible symptom), physiologically, (e.g. stabilization of a physical parameter), or both.
  • “treat”, “treating”, or “treatment” refers to preventing or delaying the onset or development or progression of the disease, condition, syndrome or disorder.
  • subject and “patient” are used interchangeably herein and may refer to an animal, preferably a mammal, most preferably a human.
  • active compound As used herein, the terms active compound, pharmaceutical agent and active ingredient are used interchangeably to refer to a pharmaceutically active compound.
  • Other ingredients in a drug composition such as carriers, diluents or excipients, may be substantially or completely pharmaceutically inert.
  • a pharmaceutical composition (also referred to herein as a composition or formulation) may comprise the active ingredient in combination with one or more carriers and/or one or more excipients and/or one or more diluents.
  • terapéuticaally effective amount refers to an amount (e.g., of an active compound or pharmaceutical agent, such as a compound of the present invention), which elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, including reduction or inhibition of an enzyme or a protein activity, or ameliorating symptoms, alleviating conditions, slowing or delaying disease progression, or preventing a disease.
  • therapeutically effective amount may refer to an amount that, when administered to a particular subject, achieves a therapeutic effect by inhibiting, alleviating or curing a disease, condition, syndrome or disorder in the subject or by prophylactically inhibiting, preventing or delaying the onset of a disease, condition, syndrome or disorder, or symptom(s) thereof.
  • a therapeutically effective amount may be an amount which relieves to some extent one or more symptoms of a disease, condition, syndrome or disorder in a subject; and/or returns to normal either partially or completely one or more physiological or biochemical parameters associated with or causative of the disease, condition, syndrome or disorder; and/or reduces the likelihood of the onset of the disease, condition, syndrome or disorder, or symptom(s) thereof.
  • “Pharmaceutically acceptable” means that, which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary as well as human pharmaceutical use.
  • a “pharmaceutically acceptable salt” is intended to mean a salt of an acid or base of a compound represented by Formula (I) that is non-toxic, biologically tolerable, or otherwise biologically suitable for administration to the subject. See, generally, S.M. Berge, et al., “Pharmaceutical Salts”, J. Pharm. Sci., 1977, 66:1-19, and Handbook of Pharmaceutical Salts, Properties, Selection, and Use, Stahl and Wermuth, Eds., Wiley-VCH and VHCA, Zurich, 2002.
  • Preferred pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of patients without undue toxicity, irritation, or allergic response.
  • Non-limiting examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen-phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne- 1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates
  • a compound of Formula (I) may possess a sufficiently acidic group, a sufficiently basic group, or both types of functional groups, and accordingly react with a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • Compounds of Formula (I) may contain at least one nitrogen of basic character, so desired pharmaceutically acceptable salts may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, boric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, phenylacetic acid, propionic acid, stearic acid, lactic acid, ascorbic acid, maleic acid, hydroxymaleic acid, isethionic acid, succinic acid, valeric acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, oleic acid, palmitic acid, lauric acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as mandelic acid, citric acid, or
  • Compounds of Formula (I) may contain a carboxylic acid moiety, a desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide, alkaline earth metal hydroxide, any compatible mixture of bases such as those given as examples herein, and any other base and mixture thereof that are regarded as equivalents or acceptable substitutes in light of the ordinary level of skill in this technology.
  • an inorganic or organic base such as an amine (primary, secondary or tertiary), an alkali metal hydroxide, alkaline earth metal hydroxide, any compatible mixture of bases such as those given as examples herein, and any other base and mixture thereof that are regarded as equivalents or acceptable substitutes in light of the ordinary level of skill in this technology.
  • suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, carbonates, bicarbonates, primary, secondary, and tertiary amines, and cyclic amines, such as benzylamines, pyrrolidines, piperidine, morpholine, piperazine, A-methyl-glucamine and tromethamine and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
  • amino acids such as glycine and arginine
  • ammonia carbonates, bicarbonates, primary, secondary, and tertiary amines
  • cyclic amines such as benzylamines, pyrrolidines, piperidine, morpholine, piperazine, A-methyl-glucamine and tromethamine
  • inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
  • any formula given herein is intended to represent compounds having structures depicted by the structural formula as well as certain variations or forms.
  • compounds of any formula given herein may have asymmetric centers and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the general formula, and mixtures thereof, are considered within the scope of such formula.
  • the compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (A)-stereoisomers or as mixtures thereof.
  • any formula given herein is intended to represent a racemate, one or more of its enantiomeric forms, one or more of its diastereomeric forms, and mixtures thereof.
  • any formula given herein is intended to refer also to any one of: hydrates, solvates, polymorphs and of such compounds, and mixtures thereof, even if such forms are not listed explicitly.
  • R at a stereocenter designates that the stereocenter is purely of the R- configuration as defined in the art; likewise, the term “S” means that the stereocenter is purely of the 5-configuration.
  • RS refers to a stereocenter that exists as a mixture of the R- and 5-configurations.
  • Compounds containing one stereocenter drawn without a stereo bond designation are a mixture of 2 enantiomers.
  • Compounds containing 2 stereocenters both drawn without stereo bond designations are a mixture of 4 diastereomers.
  • Compounds with 2 stereocenters both labeled “RS” and drawn with stereo bond designations are a 2-component mixture with relative stereochemistry as drawn.
  • Unlabeled stereocenters drawn without stereo bond designations are a mixture of the R- and 5-configurations. For unlabeled stereocenters drawn with stereo bond designations, the absolute stereochemistry is as depicted.
  • references to a compound herein stands for a reference to any one of: (a) the recited form of such compound, and (b) any of the forms of such compound in the medium in which the compound is being considered when named.
  • reference herein to a compound such as R-COOH encompasses reference to any one of: for example, R-COOH(s), R-COOH(sol), and R-COO-(sol).
  • R-COOH(s) refers to the solid compound, as it could be for example in a tablet or some other solid pharmaceutical composition or preparation
  • R- COOH(sol) refers to the undissociated form of the compound in a solvent
  • R-COO-(sol) refers to the dissociated form of the compound in a solvent, such as the dissociated form of the compound in an aqueous environment, whether such dissociated form derives from R-COOH, from a salt thereof, or from any other entity that yields R-COO- upon dissociation in the medium being considered.
  • an expression such as “exposing an entity to compound of formula R-COOH” refers to the exposure of such entity to the form, or forms, of the compound R-COOH that exists, or exist, in the medium in which such exposure takes place.
  • an expression such as “reacting an entity with a compound of formula R-COOH” refers to the reacting of (a) such entity in the chemically relevant form, or forms, of such entity that exists, or exist, in the medium in which such reacting takes place, with (b) the chemically relevant form, or forms, of the compound R-COOH that exists, or exist, in the medium in which such reacting takes place.
  • any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number in an enriched form.
  • isotopes that can be incorporated into compounds of the invention in a form that exceeds natural abundances include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H (or chemical symbol D), 3 H (or chemical symbol T), 11 C, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, 36 C1, and 125 I, respectively.
  • Such isotopically labelled compounds are useful in metabolic studies (preferably with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques [such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT)] including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F or 11 C labeled compound may be particularly preferred for PET or SPECT studies.
  • substitution with heavier isotopes such as deuterium (i.e., 2 H, or D) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements.
  • Isotopically labeled compounds of this invention can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • C n-m alkyl refers to an aliphatic chain, whether straight or branched, with a total number N of carbon members in the chain that satisfies n ⁇ N ⁇ m, with m > n.
  • each of groups Q and R can be H or F
  • the choice of H or F for Q is made independently of the choice of H or F for R, so the choice of assignment for Q does not determine or condition the choice of assignment for R, or vice-versa, unless it is expressly indicated otherwise.
  • Illustrative claim recitation in this regard would read as “each of Q and R is independently H or F”, or “each of Q and R is independently selected from the group consisting of H and F”.
  • a zwitterionic compound would be encompassed herein by referring to a compound that is known to form a zwitterion, even if it is not explicitly named in its zwitterionic form.
  • Terms such as zwitterion, zwitterions, and their synonyms zwitterionic compound(s) are standard lUPAC-endorsed names that are well known and part of standard sets of defined scientific names.
  • the name zwitterion is assigned the name identification CHEBI:27369 by the Chemical Entities of Biological Interest (ChEBI) dictionary of molecular entities.
  • a zwitterion or zwitterionic compound is a neutral compound that has formal unit charges of opposite sign.
  • aminoethanoic acid (the amino acid glycine) has the formula H 2 NCH 2 COOH, and it exists in some media (in this case in neutral media) in the form of the zwitterion + H 3 NCH 2 COO-.
  • Zwitterions, zwitterionic compounds, inner salts and dipolar ions in the known and well- established meanings of these terms are within the scope of this invention, as would in any case be so appreciated by those of ordinary skill in the art.
  • embodiments of this invention comprise the various groupings that can be made from the listed assignments, taken independently, and equivalents thereof.
  • substituent S example is one of S 1 , S 2 , and S 3
  • this listing refers to embodiments of this invention for which S example is S 1 ; S example is S 2 ; S example is S 3 ; S example is one of S 1 and S 2 ; S example is one of S 1 and S 3 ; S example is one of S 2 and S 3 ; S example is one of S 1 , S 2 and S 3 ; and S example is any equivalent of each one of these choices.
  • C i -C j when applied herein to a class of substituents, is meant to refer to embodiments of this invention for which each and every one of the number of carbon members, from i to j including i and j, is independently realized.
  • the term C 1 -C 3 refers independently to embodiments that have one carbon member (C 1 ), embodiments that have two carbon members (C 2 ), and embodiments that have three carbon members (C 3 ).
  • Embodiments of this invention include compounds of Formula (I), wherein
  • R 1 is C 1-6 alkyl or C 1-6 haloalkyl
  • R a is C 1-6 alkyl
  • R b is C 1-6 alkyl substituted with one OH member
  • R 3 is H or halo
  • R 4 is selected from the group consisting of: wherein
  • R e is selected from the group consisting of: H; halo; C 1-6 alkyl; C 1-6 alkyl substituted with one member selected from the group consisting of: OH, OCH 3 , and OCF 3 ;
  • C 1-6 haloalkyl C 1-6 haloalkyl substituted with one member selected from the group consisting of: OH, and OCH 3 ; and OC 1-6 alkyl;
  • R f is selected from the group consisting of: H; halo; C 1-6 alkyl; C 1-6 alkyl substituted with one member selected from the group consisting of: OH, OCH 3 , and OCF 3 ;
  • C 1-6 haloalkyl C 1-6 haloalkyl substituted with one member selected from the group consisting of: OH, and OCH 3 ; and OC 1-6 alkyl; and
  • Rg is H or C 1-6 alkyl; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the invention is a compound of Formula (I) wherein wherein R 1 is C 1-4 alkyl; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the invention is a compound of Formula (I) wherein R 1 is a p armaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the invention is a compound of Formula (I) wherein R 2 is , wherein R a is CH 3 ; and R b is CH(OH)CH 3 , or C(OH)(CH 3 ) 2 ; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the invention is a compound of Formula (I) wherein R 2 is
  • An additional embodiment of the invention is a compound of Formula (I) wherein R 2 is ; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the invention is a compound of Formula (I) wherein wherein R 3 is F; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the invention is a compound of Formula (I) wherein R 4 is
  • R e and R are halo; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the invention is a compound of Formula (I) wherein R 4 is or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the invention is a compound of Formula (I) wherein R 4 is , wherein Rg is H; R e is C 1-4 alkyl, and R f is halo; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the invention is a compound of Formula (I) wherein wherein R 4 is ; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • An additional embodiment of the current invention is a compound selected from the compounds shown below in Table 1, and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof:
  • enantiomers and diastereomers of the compounds of Formula (I) are also within the scope of the invention.
  • the compounds of embodiments of the present invention can be administered alone, they will generally be administered in admixture with a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient and/or a pharmaceutically acceptable diluent selected with regard to the intended route of administration and standard pharmaceutical or veterinary practice.
  • compositions comprising compounds of Formula (I) and at least one pharmaceutically acceptable carrier, pharmaceutically acceptable excipient, and/or pharmaceutically acceptable diluent.
  • the compounds of Formula (I) may be admixed with any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilizing agent(s), and combinations thereof.
  • An embodiment of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound selected from compounds of Formula (I), and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof, in accordance with any embodiment described herein; and at least one pharmaceutically acceptable excipient.
  • An additional embodiment of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising: (A) an effective amount of at least one compound selected from compounds of Formula (I) wherein
  • R 1 is C 1-6 alkyl or C 1-6 haloalkyl; R 2 is wherein
  • R a is C 1-6 alkyl
  • R b is C 1-6 alkyl substituted with one OH member
  • R 3 is H or halo;
  • R 4 is selected from the group consisting of: wherein
  • R e is selected from the group consisting of: H; halo; C 1-6 alkyl; C 1-6 alkyl substituted with one member selected from the group consisting of: OH, OCH 3 , and OCF 3 ; C 1-6 haloalkyl; C 1-6 haloalkyl substituted with one member selected from the group consisting of: OH, and OCH 3 ; and OC 1-6 alkyl;
  • R f is selected from the group consisting of: H; halo; C 1-6 alkyl; C 1-6 alkyl substituted with one member selected from the group consisting of: OH, OCH 3 , and OCF 3 ; C 1-6 haloalkyl; C 1-6 haloalkyl substituted with one member selected from the group consisting of: OH, and OCH 3 ; and OC 1-6 alkyl; and
  • Rg is H or C 1-6 alkyl; or pharmaceutically acceptable salts, isotopes, N-oxides, solvates, or stereoisomers thereof; and (B) at least one pharmaceutically acceptable excipient.
  • An additional embodiment of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound shown in Table 1 (e.g., a compound selected from Examples 1-7), or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer of the compound of Table 1, a pharmaceutically acceptable prodrug of the compound of Table 1, or a pharmaceutically active metabolite of the compound of Table 1; and at least one pharmaceutically acceptable excipient.
  • Solid oral dosage forms such as, tablets or capsules, containing one or more compounds of the present invention may be administered in at least one dosage form at a time, as appropriate. It is also possible to administer the compounds in sustained release formulations.
  • Additional oral forms in which the present inventive compounds may be administered include elixirs, solutions, syrups, and suspensions; each optionally containing flavoring agents and coloring agents.
  • one or more compounds of Formula (I) can be administered by inhalation (intratracheal or intranasal) or in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder.
  • they can be incorporated into a cream comprising, consisting of, and/or consisting essentially of an aqueous emulsion of polyethylene glycols or liquid paraffin.
  • An alternative means of administration includes transdermal administration by using a skin or transdermal patch.
  • compositions of the present invention can also be injected parenterally, for example, intracavernosally, intravenously, intramuscularly, subcutaneously, intradermally, or intrathecally.
  • the compositions will also include at least one of a suitable carrier, a suitable excipient, and a suitable diluent.
  • compositions of the present invention are best used in the form of a sterile aqueous solution that may contain other substances, for example, enough salts and monosaccharides to make the solution isotonic with blood.
  • compositions of the present invention may be administered in the form of tablets or lozenges, which can be formulated in a conventional manner.
  • compositions containing at least one of the compounds of Formula (I) as the active ingredient can be prepared by mixing the compound(s) with a pharmaceutically acceptable carrier, a pharmaceutically acceptable diluent, and/or a pharmaceutically acceptable excipient according to conventional pharmaceutical compounding techniques.
  • a pharmaceutically acceptable carrier e.g., benzyl alcohol, benzyl ether, benzyl ether, benzyl ether, benzyl, sulfonyl, sulfonyl, adiluent, and/or a pharmaceutically acceptable excipient according to conventional pharmaceutical compounding techniques.
  • the carrier, excipient, and diluent may take a wide variety of forms depending upon the desired route of administration (e.g., oral, parenteral, etc.).
  • suitable carriers, excipients and diluents include water, glycols, oils, alcohols, flavoring agents, preservatives, stabilizers, coloring agents and the like;
  • suitable carriers, excipients and diluents include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like.
  • Solid oral preparations also may be optionally coated with substances such as, sugars, or be enterically coated so as to modulate the major site of absorption and disintegration.
  • the carrier, excipient and diluent will usually include sterile water, and other ingredients may be added to increase solubility and preservation of the composition.
  • injectable suspensions or solutions may also be prepared utilizing aqueous carriers along with appropriate additives such as, solubilizers and preservatives.
  • a therapeutically effective amount of a compound of Formula (I) or a pharmaceutical composition thereof may comprise a dose range from about 0.1 mg to about 3000 mg, or any particular amount or range therein, in particular from about 1 mg to about 1000 mg, or any particular amount or range therein, of active ingredient in a regimen of about 1 to about (4x) per day for an average (70 kg) human; although, it is apparent to one skilled in the art that the therapeutically effective amount for a compound of Formula (I) will vary as will the diseases, syndromes, conditions, and disorders being treated.
  • An embodiment of the present invention is directed to a pharmaceutical composition for oral administration, comprising a compound of Formula (I) in an amount of from about 1 mg to about 500 mg.
  • a compound of Formula (I) may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three and (4x) daily.
  • Optimal dosages of a compound of Formula (I) to be administered may be readily determined and will vary with the particular compound used, the mode of administration, the strength of the preparation, and the advancement of the disease, syndrome, condition or disorder.
  • factors associated with the particular subject being treated including subject gender, age, weight, diet and time of administration, will result in the need to adjust the dose to achieve an appropriate therapeutic level and desired therapeutic effect.
  • the above dosages are thus exemplary of the average case. There can be, of course, individual instances wherein higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • Compounds of Formula (I) may be administered in any of the foregoing compositions and dosage regimens or by means of those compositions and dosage regimens established in the art whenever use of a compound of Formula (I) is administered to a subject in need thereof.
  • one or more compounds of Formula (I) are useful in methods for treating, ameliorating and / or preventing a disease, a syndrome, a condition or a disorder that is affected by the inhibition of DHODH enzymatic activity.
  • An additional embodiment of the invention relates to the use of compounds of Formula (I), e.g., by inhibiting dihydroorotate oxygenase enzyme activity, in treating disorders like inflammatory disorders, autoimmune disorders, or cancer; or pharmaceutically acceptable salts, isotopes, N-oxides, solvates, or stereoisomers thereof.
  • the present invention provides a method for inhibiting or altering Dihydroorotate Dehydrogenase (DHODH) enzymatic activity, the method comprising contacting DHODH with any compound of Formula (I), aspect or embodiment disclosed herein, thereby inhibiting or otherwise altering DHODH enzymatic activity.
  • DHODH Dihydroorotate Dehydrogenase
  • An additional embodiment of the present invention provides methods for treating diseases, disorders, or medical conditions mediated or otherwise affected by dihydroorotate dehydrogenase (DHODH) enzyme activity comprising administering a compound of Formula (I) to a subject in need thereof.
  • DHODH dihydroorotate dehydrogenase
  • DHODH inhibitor may refer to an agent that inhibits or reduces DHODH activity.
  • the term “therapeutically effective amount” refers to the amount of a compound of the present invention that, when administered to a subject, is effective to (1) at least partially alleviate, inhibit, prevent, and/ or ameliorate a condition, or a disorder or a disease (i) mediated by DHODH enzymatic activity; or (ii) associated with DHODH enzymatic activity; or (iii) characterized by activity (normal or abnormal) of DHODH enzyme; or (2) reduce or inhibit the activity of DHODH enzyme; or (3) reduce or inhibit the expression of DHODH; or (4) modify the protein levels of DHODH.
  • DHODH inhibitors are believed to act by inhibiting nucleic acid synthesis, cell cycle arrest or altering post-translational glycosylation of proteins involved in regulating myeloid differentiation within progenitor tumor cells.
  • An additional embodiment of the invention is a method of treating a subject suffering from or diagnosed with a disease, disorder, or medical condition mediated or otherwise affected by DHODH enzymatic activity, comprising administering to a subject in need of such treatment an effective amount of at least one compound selected from: compounds of Formula (I), such as a compound of Table 1), enantiomers and diastereomers of the compounds of Formula (I) (such as a compound of Table 1), isotopic variations of the compounds of Formula (I) (such as a compound of Table 1), and pharmaceutically acceptable salts of all of the foregoing.
  • compounds of Formula (I) such as a compound of Table 1
  • enantiomers and diastereomers of the compounds of Formula (I) such as a compound of Table 1
  • isotopic variations of the compounds of Formula (I) such as a compound of Table 1
  • a method of treating a subject suffering from or diagnosed with a disease, disorder, or medical condition, such as cancer comprises administering to the subject an effective amount of at least one compound selected from: compounds of Formula (I) (such as a compound of Table 1), and pharmaceutically acceptable salts of all the foregoing (e.g., by inhibiting or otherwise altering dihydroorotate oxygenase enzyme activity in the subject).
  • compounds of Formula (I) such as a compound of Table 1
  • pharmaceutically acceptable salts of all the foregoing e.g., by inhibiting or otherwise altering dihydroorotate oxygenase enzyme activity in the subject.
  • inhibitors of DHODH of the present invention may be used for the treatment of immunological diseases including, but not limited to, autoimmune and inflammatory disorders, e.g. arthritis, inflammatory bowel disease, gastritis, ankylosing spondylitis, ulcerative colitis, pancreatitis, Crohn’s disease, celiac disease, multiple sclerosis, systemic lupus erythematosus, lupus nephritis, rheumatic fever, gout, organ or transplant rejection, chronic allograft rejection, acute or chronic graft-versus-host disease, dermatitis including atopic, dermatomyositis, psoriasis, Behcet’s diseases, uveitis, myasthenia gravis, Grave’s disease, Hashimoto thyroiditis, Sjogren’s syndrome, blistering disorders, antibody- mediated vasculitis syndromes, immune-complex vasculitides, allergic disorders, asthma, bronchi
  • the term “affect” or “affected” when referring to a disease, disorder, or medical condition that is affected by the inhibition or alteration of DHODH enzymatic activity) includes a reduction in the frequency and / or severity of one or more symptoms or manifestations of said disease, syndrome, condition or disorder; and / or includes the prevention of the development of one or more symptoms or manifestations of said disease, syndrome, condition or disorder or the development of the disease, condition, syndrome or disorder.
  • An additional embodiment of the invention provides a method of treatment of cancer comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
  • the cancer is selected from but not limited to, lymphomas, leukemias, carcinomas, and sarcomas.
  • An additional embodiment of the invention provides the use of a compound of Formula (I), or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof, for the treatment of one or more cancer types.
  • the uses and methods of treatment described herein are directed to the treatment of cancer, wherein the cancer is selected from but not limited to: leukemias including but not limited to acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), (acute) T-cell leukemia, acute monocytic leukemia, acute promyelocytic leukemia (APL), bisphenotypic B myelomonocytic leukemia, chronic myeloid leukemia (CML), chronic myelomonocytic leukemia (CMML), large granular lymphocytic leukemia, plasma cell leukemia, and also myelodysplastic syndrome (MDS), which can develop into an acute myeloid leukemia, lymphomas including but not limited to AIDS-related lymphoma, Hodgkin lymphoma, non-Hodgkin's lymphoma (NHL), T-non-Hodgkin lymphoma (T-NHL), subtype
  • ALL acute lymph
  • cancers that may benefit from a treatment with inhibitors of DHODH of the present invention include, but are not limited to, lymphomas, leukemias, carcinomas, and sarcomas, e.g. non-Hodgkin’s lymphoma, diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), follicular lymphoma (FL), marginal zone lymphoma, T-cell lymphoma, Hodgkin’s lymphoma, Burkitt’s lymphoma, multiple myeloma, brain (gliomas), glioblastomas, breast cancer, colorectal/colon cancer, prostate cancer, lung cancer including non-small-cell, gastric cancer, endometrial cancer, melanoma, pancreatic cancer, liver cancer, kidney cancer, squamous cell carcinoma, ovarian cancer, sarcoma, osteosarcoma, thyroid cancer, bladder cancer, head & neck cancer, testicular
  • the compounds of the present invention may be employed in combination with one or more other medicinal agents, more particularly with one or more anti-cancer agents, e.g. chemotherapeutic, anti-proliferative or immunomodulating agents, or with adjuvants in cancer therapy, e.g. immunosuppressive or antiinflammatory agents.
  • anti-cancer agents e.g. chemotherapeutic, anti-proliferative or immunomodulating agents, or with adjuvants in cancer therapy, e.g. immunosuppressive or antiinflammatory agents.
  • Additional non-limiting examples of anti-cancer agents that may be administered in combination with a compound of the present invention include biologic compounds, such as monoclonal antibodies (e.g., that mediate effector function upon binding to cancer cell-associated antigens, or block interaction of a receptor expressed on cancer cells with a soluble or cell bound ligand), bispecific antibodies that mediate immune cell redirection, etc.
  • a method of treating cancer comprises administering an effective amount of a compound of the present invention (e.g., selected from compounds of Formula (I), such as a compound shown in Table 1, pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof) and an effective amount of one or more additional anticancer agents, wherein the method comprises administering the compound of the present invention and the additional anti-cancer agent(s) either simultaneously (e.g., as part of the same pharmaceutical composition) or sequentially.
  • a compound of the present invention e.g., selected from compounds of Formula (I), such as a compound shown in Table 1, pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof
  • a pharmaceutical composition comprises an effective amount of a compound of the present invention (e.g., selected from compounds of Formula (I), such as a compound shown in Table 1, pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof), an effective amount of one or more additional anti-cancer agents, and optionally one or more excipients.
  • a compound of the present invention e.g., selected from compounds of Formula (I), such as a compound shown in Table 1, pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof
  • an effective amount of one or more additional anti-cancer agents e.g., sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate,
  • An additional embodiment of the invention provides the use of a compound of Formula (I), or pharmaceutically acceptable salts, isotopes, N-oxides, solvates, or stereoisomers thereof, as part of chemotherapeutic regimens for the treatment of cancers, lymphomas and leukemias alone or in combination with classic antitumoral compounds well known by the one skilled in the art.
  • R 1 is C 1-6 alkyl or C 1-6 haloalkyl
  • a suitable base such as K 2 CO 3 , Na2CO 3 , Cs2CO 3 ,NaH, KHMDS, LiHMDS, and NaHMDS, preferably NaH
  • a suitable solvent such as THF, DMF, and ACN, preferably THF.
  • ethyl 4-methyl-1H-pyrazole-3-carboxylate is protected with a THP group employing conditions known to one skilled in the art, for example reaction with 3,4- dihydro-2H-pyran (DHP) and p-toluenesulfonic acid (TsOH) in a suitable solvent such as DCM, DCE, and the like; at room temperature; to afford ethyl 4-methyl-l-(tetrahydro-2H-pyran-2-yl)- 1H-pyrazole-3 -carboxylate.
  • DHP 3,4- dihydro-2H-pyran
  • TsOH p-toluenesulfonic acid
  • THP protecting group Deprotection of the THP protecting group is achieved employing conditions known to one skilled in the art; for example, using HC1, in a suitable solvent such as MeOH; at a temperature of about 60 °C; affords a a compound of formula (VIII), where R a is C 1-6 alkyl and R b is CH(CH 3 )(OH).
  • HC1 a suitable solvent
  • MeOH a suitable solvent
  • R a is C 1-6 alkyl
  • R b is CH(CH 3 )(OH
  • a compound of formula (VIII), where R a is C 1-6 alkyl and R b is C(CH 3 ) 2 (OH); is prepared by reaction of ethyl 4-methyl-l H-pyrazole-3 -carboxylate with MeMgBr; in a suitable solvent such as THF, ether, 1 ,4-dioxane and the like; at low temperatures, preferably 0 °C.
  • a compound of formula (VII) is reacted in a Pd-catalyzed amination reaction with benzophenone imine employing cross-coupling conditions known to one skilled in the art.
  • a compound of formula (VII) where R 1 is C 1-6 alkyl or C 1-6 haloalkyl; is reacted with a palladium catalyst such as Pd2(dba)y a suitable ligand, such as XantPhos and the like; and a suitable base, such as CS 2 CO 3 , K 2 CO 3 , NaOtBu and the like; in a solvent such as toluene, and the like; at refluxing temperatures; to provide a compound of formula (X).
  • a palladium catalyst such as Pd2(dba)y a suitable ligand, such as XantPhos and the like
  • a suitable base such as CS 2 CO 3 , K 2 CO 3 , NaOtBu and the like
  • solvent such as tolu
  • a compound of formula (XIV) is prepared in two steps by first, diazotization, employing conditions known to one skilled in the art. For example, reaction with NaNO 2 ; in an acidic aqueous solution such as AcOH in H 2 O, or other nitrite reagents; at a temperature of ⁇ 5 °C. Subsequent reaction with ethyl 3-oxobutanoate, and a suitable base, such as Na 2 CO 3 , K 2 CO 3 , and the like; in a solvent such as EtOH, MeOH and the like.
  • a triazole compound of formula (XV) is prepared by reaction of a compound of formula (XIV), with an ammonium salt such as, NH 4 OAc, and a copper catalyst, such as CuC1 2 , and the like; in a suitable solvent such as EtOH, MeOH, and the like; at elevated temperatures, preferably 90 °C.
  • R 2 is where R a is C 1-6 alkyl and R b is CH(CH 3 )(OH) or
  • Compounds of Formula (I) may be converted to their corresponding salts using methods known to one of ordinary skill in the art.
  • an amine of Formula (I) is treated with trifluoroacetic acid, HC1, or citric acid in a solvent such as Et 2 O, CH 2 CI 2 , THF, MeOH, chloroform, or isopropanol to provide the corresponding salt form.
  • trifluoroacetic acid or formic acid salts are obtained as a result of reverse phase HPLC purification conditions.
  • Crystalline forms of pharmaceutically acceptable salts of compounds of Formula (I) may be obtained in crystalline form by recrystallization from polar solvents (including mixtures of polar solvents and aqueous mixtures of polar solvents) or from non-polar solvents (including mixtures of non-polar solvents).
  • the compounds according to this invention may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention.
  • Compounds prepared according to the schemes described above may be obtained as single forms, such as single enantiomers, by form-specific synthesis, or by resolution. Compounds prepared according to the schemes above may alternately be obtained as mixtures of various forms, such as racemic (1 : 1) or non-racemic (not 1: 1) mixtures. Where racemic and non-racemic mixtures of enantiomers are obtained, single enantiomers may be isolated using conventional separation methods known to one of ordinary skill in the art, such as chiral chromatography, recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation. Where regioisomeric or diastereomeric mixtures are obtained, as applicable, single isomers may be separated using conventional methods such as chromatography or crystallization.
  • reaction mixtures were magnetically stirred at room temperature (rt) under a nitrogen atmosphere. Where solutions were “dried,” they were generally dried over a drying agent such as Na 2 SO 4 or MgSO 4 . Where mixtures, solutions, and extracts were “concentrated”, they were typically concentrated on a rotary evaporator under reduced pressure.
  • METHOD B A Gilson GX-281 semi-prep-HPLC with Phenomenex Synergi C18(10 ⁇ m, 150 x 25mm), or Boston Green ODS C18(5 ⁇ m, 150 x 30mm), and mobile phase of 5-99% ACN in water(0.1 %TFA) over 10 min and then hold at 100% ACN for 2 min, at a flow rate of 25 mL/min. or
  • Preparative supercritical fluid high performance liquid chromatography was performed either on a Thar 80 Prep-SFC system, or Waters 80Q Prep-SFC system from Waters.
  • the ABPR was set to lOObar to keep the CO 2 in SF conditions, and the flow rate may verify according to the compound characteristics, with a flow rate ranging from 50g/min to 70g/min.
  • the column temperature was ambient temperature
  • Mass spectra (MS) were obtained on a SHIMADZU LCMS-2020 MSD or Agilent 1200 ⁇ G6110AMSD using electrospray ionization (ESI) in positive mode unless otherwise indicated. Calculated (calcd.) mass corresponds to the exact mass.
  • NMR Nuclear magnetic resonance
  • s singlet
  • d doublet
  • t triplet
  • q quartet
  • dd doublet of doublets
  • ddd doublet of doublet of doublets
  • td triplet of doublets
  • dt doublet of triplets
  • spt septet
  • quin quintet
  • m multiplet
  • br broad. It will be understood that for compounds comprising an exchangeable proton, said proton may or may not be visible on an NMR spectrum depending on the choice of solvent used for running the NMR spectrum and the concentration of the compound in the solution.
  • Step A (5)-4-Bromo-5-fluoro-2-((1,1,1-trifluoropropan-2-yl)oxy)benzonitrile.
  • a mixture of (S)- 1,1,1 -trifluoropropan-2-ol (11.5 g, 101 mmol) in THF (50 mL) was added to the mixture of NaH (60% purity, 4.4 g, 110 mmol) in THF (80 mL) dropwise at 0 °C under N 2 . The mixture was stirred at room temperature for 0.5 hours.
  • Step B (S)-4-Bromo-5-fluoro-2-((l.l.1-trifluoropropan-2-yl)oxy)benzoic acid.
  • EtOH 150 mL
  • 2 M aq. NaOH 151 mL, 301 mmol
  • the mixture was stirred at 90 °C for 16 hours under N 2 .
  • the mixture was concentrated and H 2 O (200 mL) was added to the mixture.
  • Step C (M-Methyl 4-bromo-5-fluoro-2-((l.l.1-trifluoropropan-2-yl)oxy)benzoate.
  • (S)-4-bromo-5-fluoro-2-((1,1,1-trifluoropropan-2-yl)oxy)benzoic acid 27 g, 76 mmol
  • MeOH 500 mL
  • SOCl 2 11 mL, 152 mmol
  • Step A Ethyl 4-methyl- l -(yetrahvdro-2H-pyran-2-yl)- 1 H-pyrazole-3 -carboxy late.
  • DHP 4.1 g, 49 mmol
  • TsOH 558 mg, 3.2 mmol
  • DCM 50 mL
  • the reaction mixture was slowly poured into sat. aq. NaHCCh (30 mL) and extracted with DCM (20 mL x 2).
  • Step C 1-(4-Methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)ethan-1-ol.
  • Step D 1-(4-Methyl-1H-pyrazol-3-yl)ethan-1-ol.
  • 1-(4-methyl-1-(tetrahydro-2H- pyran-2-yl)-1H-pyrazol-3-yl)ethanol 1.7 g, 7.9 mmol
  • 12 M aq. HC1 3.3 mL, 39 mmol
  • the resulting mixture was stirred at 60 °C for 1 hour.
  • Step E The second eluting compound (R*)-1-(4-Methyl- 1H-pyrazol-3-yl)ethanol (206 mg, 1.6 mmol, 23% yield) as white solid.
  • Example 1 A-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-((R*)-1-hvdroxyethyl)-4-methyl-1H- pyrazol- 1 -y 1 )-2-(((X)- 1,1,1 -trifl uoropropan-2-yl )oxy)benzamide.
  • Example 3 (S)-/V-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-(2-hvdroxypropan-2-yl)-4-methyl- 1 H-pyrazol- 1 -yl)-2-(( 1 ,1,1 -trifl uoropropan-2-yl )oxy)benzamide.
  • the crude was purified by column chromatography ( SiO 2 , gradient elution: 0 - 40% ethyl acetate in petroleum ether) to give the crude product (70 mg, 122 ⁇ mol , 62% yield) as a white solid.
  • the white solid was purified by preparative reversed phase HPLC (Stationary phase: Boston Prime Cl 8, 5 ⁇ m, 150 X
  • Example 5 N-(5-Chloro-3-methyl- 1H-pyrazol-4-yl)-5-fluoro-4-(3-((S*)-l -hydroxy ethyl)-4- methyl- 1 H-pyrazol- 1 -yl)-2-(((M- 1.1.1 -trifluoropropan-2-yl )oxyibenzamide.
  • Example 6 (S)-N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-(2-hvdroxypropan-2-yl)- 4-methyl- 1 H-pyrazol- 1 -yl)-2-(( 1 , 1 , 1 -trifluoropropan-2-yl)oxy)benzamide.
  • the crude was purified by column chromatography (SiO 2 , gradient elution: 0 - 40% ethyl acetate in petroleum ether) to give the crude compound (100 mg, 177 ⁇ mol , 81% yield) as a white solid.
  • the white solid was purified by preparative reverse phase HPLC (Stationary phase: Boston Prime Cl1, 5 ⁇ m, 150 X 30 mm ; Mobile phase: water (0.05% NH 3 H 2 O + 10 mM NH 4 HCO 3 ) (A) - MeCN (B), gradient elution: 33 - 67% B in A over 7 min, flow rate: 35 mL/min) to give the title compound (60 mg, 123 ⁇ mol, 57% yield) as a white solid.
  • DCIP dichloroindophenol
  • DHO dihydroorotate
  • the assay buffer contained 50 nM HEPES, pH 7.5, 150 mM NaCl, 0.5 mM EDTA, and 0.1% Triton X-100 in MilliQ water.
  • Substrate consisting of 20 mM DHO, 5mM CoQ 6 , and ImM DCIP in assay buffer, initiates the reaction.
  • the assay is run in end-point mode by quenching the reaction with the potent DHODH inhibitor brequinar. Absorbance measurements were obtained using the BMG Phera Star plate-reading spectrophotomer.
  • Purified human DHODH was purchased from Proteros (cat. No. PR-0044). Chemicals were purchased from Sigma- Aldrich, Teknova, and Avanti Polar Lipids. Liquid handling was performed using Labcyte Echo and Formulatrix Tempest.
  • MOLM-13 cells human acute myeloid leukemia cells
  • DSMZ human acute myeloid leukemia cells
  • RPMI 1640 + Glutamax + 25mM HEPES Invitrogen, catalog number 72400
  • FBS heat inactivated fetal bovine serum

Abstract

Disclosed are compounds, compositions and methods for treating diseases, disorders, or medical conditions that are affected by the modulation of DHODH. Such compounds are represented by Formula (I) as follows, wherein R1, R2, R3, and R4 are defined herein.

Description

DIHYDROOROTATE DEHYDROGENASE INHIBITORS
CROSS REFERENCE TO RELATED APPLICATIONS
This application is entitled to priority pursuant to 35 U.S.C. § 119(e) to U.S. Provisional Patent Application No. 63/085,314, filed on September 30, 2020, the disclosure of which is incorporated by reference herein in its entirety.
FIELD OF THE INVENTION
The present invention relates to novel compounds that are dihydroorotate dehydrogenase (DHODH) inhibitors. These compounds may be useful for the treatment of a disease, disorder, or medical condition where there is an advantage in inhibiting DHODH. The invention also relates to pharmaceutical compositions comprising one or more of such compounds, to processes to prepare such compounds and compositions, and to the use of such compounds or pharmaceutical compositions for the method of treatment of cancer, and autoimmune and inflammatory diseases, syndromes, and disorders.
BACKGROUND OF THE INVENTION
Acute myelogenous leukemia (AML) is a clonal disease of the blood and bone marrow resulting from mutations that occur in normal hematopoietic stem cells. AML is a heterogenous disease in that it presents with a range of cytogenetic, morphological and immunophenotypic features, and is characterized by an accumulation of clonal, abnormal myeloid progenitor cells, known as myeloblasts. These cells demonstrate disruption of normal myeloid differentiation and excessive proliferation, resulting in the decreased formation of hematopoietic cells. Disease remission can be achieved with standard induction chemotherapy, but refractory and relapsed disease remains a challenge due to persistence of leukemic stem cells. Therefore, AML represents an unmet medical need with >20,000 new cases per year in the US with 5-year overall survival below 30% (Stein ET et al., Health Qual Life Outcomes 16: 193, 2018).
Differentiation therapy is considered an attractive approach to AML treatment based on the knowledge that differentiation and loss of stem cell self-renewal are coupled in normal cells. Treatment of acute promyelocytic leukemia, which represents 10-15% of all AML, with all-trans retinoic acid is the paradigm for differentiation therapy. Retinoic acid targets the promyelocytic leukemia protein (PML)-retinoic acid receptor-a (RAR-a) fusion protein encoded by a t(15, 17) chromosomal translocation. Targeting PML-RAR specifically lifts the transcriptionally mediated differentiation block induced by the fusion protein and early clinical trials with single agent ATRA demonstrated complete hematologic remission in all treated patients (McCulloch D et al. Onco Targets Ther 2017; 10: 1585-1601; Nowak D et al. Blood 113: 3655, 2009).
Although differentiation therapy is successful, it is only applicable to a small population of AML patients. Research efforts have aimed at identifying additional differentiation inducing agents, but with limited success. Recently dihydroorotate dehydrogenase (DHODH) emerged as a potentially more broadly applicable differentiation target in a phenotypic screen aimed at identifying small molecules that overcome blockade of the maturation of primary murine bone marrow cells expressing the homeobox protein HoxA9. This protein is a key transcription factor involved in balancing stem cell maintenance/differentiation and is normally expressed in hematopoietic progenitor cells and downregulated upon induction of differentiation and has been found to be widely overexpressed in AML (Sykes et al., Cell 167: 171, 2016).
DHODH is a flavin mononucleotide (FMN) flavoprotein located in the inner mitochondrial membrane that catalyzes the oxidation of dihydroorotate to orotate, the fourth step in the de novo pyrimidine biosynthesis pathway. Inhibition of DHODH leads to decreased pyrimidine synthesis important precursors for nucleotide synthesis, but also glycoprotein and phospholipid biosynthesis (Reis RAG et al., Archives Biochem Biophysics 632: 175, 2017; Vyas VK et al., Mini Rev Med Chem 11 : 1039, 2011). DHODH is a validated target for the treatment of autoimmune diseases with the FDA approved small molecule DHODH inhibitors leflunomide and teriflunomide for rheumatoid arthritis and multiple sclerosis, respectively (Lolli ML et al., Recent patents on Anti-Cancer Drug Discovery 13: 86, 2018).
Since the first observation by Sykes et al. demonstrating that DHODH inhibition drives AML differentiation in vitro, as evidenced by upregulation of the differentiation markers CDllb and CD 14, and results in dose dependent anti-leukemic effects, decreased leukemic stem cells and prolonged survival in vivo, additional evidence emerged demonstrating that small molecule DHODH inhibitors mediate antiproliferative activity against AML cells with concomitant cell cycle arrest, upregulation of CDllb and CD 14, and induction of apoptosis (Wu D et al.. Haematologica 103: 1472, 2018; Sainas S et al., J Med Chem 61: 6034, 2018; Cao L et al., Mol Cancer Ther, October 23rd Epub ahead of print). Moreover, preclinical solid tumor in vitro and in vivo models demonstrated effectiveness of DHODH inhibition and DHODH was identified as a synthetic lethality in PTEN and KRAS mutant solid tumors (Pharmacology and Therapeutics, Epub October 19th, 2018; Mathur D et al., Cancer Discovery 7: 1, 2017; Cell Chemical Biology 25: 1, 2018).
Thus, there remains a need for DHODH inhibitors that provide a therapeutic benefit to patients suffering from cancer and/or inflammatory and immunological diseases.
SUMMARY OF THE INVENTION
Embodiments of the present invention relate to compounds, pharmaceutical compositions containing them, methods of making and purifying them, methods of using them as inhibitors of DHODH enzymatic activity and methods for using them in the treatment of a subject suffering from or diagnosed with a disease, disorder, or medical condition such as autoimmune or inflammatory disorders, or diseases such as cancer.
Embodiments of this invention include compounds of Formula (I),
Figure imgf000004_0001
wherein
R1 is C1-6alkyl or C1-6haloalkyl;
Figure imgf000004_0002
wherein
Ra is C1-6alkyl;
Rb is C1-6alkyl substituted with one OH member;
R3 is H or halo;
R4 is selected from the group consisting of:
Figure imgf000004_0003
Figure imgf000005_0001
wherein
Re is selected from the group consisting of: H; halo; C1-6alkyl; C1-6alkyl substituted with one member selected from the group consisting of: OH, OCH3, and OCF3; C1-6haloalkyl; C1-6haloalkyl substituted with one member selected from the group consisting of: OH, and OCH3; and OC1-6alkyl;
Rf is selected from the group consisting of: H; halo; C1-6alkyl; C1-6alkyl substituted with one member selected from the group consisting of: OH, OCH3, and OCF3; C1-6haloalkyl; C1-6haloalkyl substituted with one member selected from the group consisting of: OH, and OCH3; and OC1-6alkyl; and
Rg is H or C1-6alkyl; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
The present invention further provides methods for treating or ameliorating a disease, syndrome, condition, or disorder in a subject, including a mammal and/or human in which the disease, syndrome, condition, or disorder is affected by the inhibition of DHODH enzymatic activity, including but not limited to, cancer and/or inflammatory or immunological diseases, using a compound of Formula (I) or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
Additional embodiments, features, and advantages of the invention will be apparent from the following detailed description and through practice of the invention.
DETAILED DESCRIPTION OF THE INVENTION
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in art. As used in the specification and the appended claims, unless specified to the contrary, the following terms have the meaning indicated in order to facilitate the understanding of the present invention.
The singular forms "a", "an" and "the" encompass plural references unless the context clearly indicates otherwise.
With reference to substituents, the term “independently” refers to the situation where when more than one substituent is possible, the substituents may be the same or different from each other.
The term “substituted” means that the specified group or moiety bears one or more substituents. The term "unsubstituted" means that the specified group bears no substituents. The term “optionally substituted” means that the specified group is unsubstituted or substituted by one or more substituents. Where the term “substituted” is used to describe a structural system, the substitution is meant to occur at any valency-allowed position on the system.
Unless qualified specifically in particular instances of use, the term “alkyl” refers to a straight- or branched-chain alkyl group having from 1 to 8 carbon atoms in the chain. Examples of alkyl groups include methyl (Me), ethyl (Et), n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl (tBu), pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and groups that in light of the ordinary skill in the art and the teachings provided herein would be considered equivalent to any one of the foregoing examples. “C1-6alkyl” refers to straight- or branched-chain alkyl group having from 1 to 6 carbon atoms in the chain. “C1-4alkyl” refers to straight- or branched-chain alkyl group having from 1 to 4 carbon atoms in the chain.
The term “cycloalkyl” refers to a saturated or partially saturated, monocyclic, fused polycyclic, or spiro polycyclic carbocycle having from 3 to 12 ring atoms per carbocycle. “C3- 6cycloalkyl” refers to a carbocycle having from 3 to 6 ring atoms per carbocycle. Illustrative examples of cycloalkyl groups include the following entities, in the form of properly bonded moieties:
Figure imgf000006_0001
The term “halogen” or “halo” represents chlorine, fluorine, bromine, or iodine.
The term “haloalkyl” refers to a straight- or branched-chain alkyl group having from 1 to 6 carbon atoms in the chain optionally substituting hydrogens with halogens. The term “C1-6 haloalkyl” as used here refers to a straight- or branched-chain alkyl group having from 1 to 6 carbon atoms in the chain, optionally substituting hydrogens with halogens. The term “C1-4 haloalkyl” as used here refers to a straight- or branched-chain alkyl group having from 1 to 4 carbon atoms in the chain, optionally substituting hydrogens with halogens. Examples of “haloalkyl” groups include trifluoromethyl (CF3), difluoromethyl (CF2H), monofluoromethyl (CH2F), pentafluoroethyl (CF2CF3), tetrafluoroethyl (CHFCF3), monofluoroethyl (CH2CH2F), trifluoroethyl (CH2CF3), tetrafluorotrifluoromethylethyl (CF(CF3)2), and groups that in light of the ordinary skill in the art and the teachings provided herein would be considered equivalent to any one of the foregoing examples.
The term “aryl” refers to a monocyclic, aromatic carbocycle (ring structure having ring atoms that are all carbon) having 6 atoms per ring. (Carbon atoms in the aryl groups are sp2 hybridized.)
The term “phenyl” represents the following moiety:
Figure imgf000007_0001
The term “heteroaryl” refers to a monocyclic or fused bicyclic heterocycle (ring structure having ring atoms selected from carbon atoms and up to four heteroatoms selected from nitrogen, oxygen, and sulfur) having from 3 to 9 ring atoms per heterocycle. Illustrative examples of heteroaryl groups include the following entities, in the form of properly bonded moieties:
Figure imgf000007_0002
Those skilled in the art will recognize that the species of cycloalkyl, heteroaryl and aryl groups listed or illustrated above are not exhaustive, and that additional species within the scope of these defined terms may also be selected.
The term “variable point of attachment” means that a group is allowed to be attached at more than one alternative position in a structure. The attachment will always replace a hydrogen atom on one of the ring atoms. In other words, all permutations of bonding are represented by the single diagram, as shown in the illustrations below.
Figure imgf000007_0003
Those skilled in the art will recognize that that if more than one such substituent is present for a given ring, the bonding of each substituent is independent of all of the others. The groups listed or illustrated above are not exhaustive.
As used herein, the term "or" means "and/or" unless stated otherwise.
As used herein, the terms "including", "containing" and “comprising” are used in their open, non-limiting sense.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
As used herein, the term “treat”, “treating”, or “treatment” of any disease, condition, syndrome or disorder refers, in one embodiment, to ameliorating the disease, condition, syndrome or disorder (i.e. slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment, “treat”, “treating”, or “treatment” refers to alleviating or ameliorating at least one physiological or biochemical parameter associated with or causative of the disease, condition, syndrome or disorder, including those which may not be discernible by the patient. In a further embodiment, “treat”, “treating”, or “treatment” refers to modulating the disease, condition, syndrome or disorder either physically (e.g. stabilization of a discernible symptom), physiologically, (e.g. stabilization of a physical parameter), or both. In yet another embodiment, “treat”, “treating”, or “treatment” refers to preventing or delaying the onset or development or progression of the disease, condition, syndrome or disorder.
The terms “subject” and “patient” are used interchangeably herein and may refer to an animal, preferably a mammal, most preferably a human.
As used herein, the terms active compound, pharmaceutical agent and active ingredient are used interchangeably to refer to a pharmaceutically active compound. Other ingredients in a drug composition, such as carriers, diluents or excipients, may be substantially or completely pharmaceutically inert. A pharmaceutical composition (also referred to herein as a composition or formulation) may comprise the active ingredient in combination with one or more carriers and/or one or more excipients and/or one or more diluents.
The term “therapeutically effective amount” (used interchangeably herein with “effective amount”) refers to an amount (e.g., of an active compound or pharmaceutical agent, such as a compound of the present invention), which elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, including reduction or inhibition of an enzyme or a protein activity, or ameliorating symptoms, alleviating conditions, slowing or delaying disease progression, or preventing a disease. Stated another way, the term therapeutically effective amount may refer to an amount that, when administered to a particular subject, achieves a therapeutic effect by inhibiting, alleviating or curing a disease, condition, syndrome or disorder in the subject or by prophylactically inhibiting, preventing or delaying the onset of a disease, condition, syndrome or disorder, or symptom(s) thereof. A therapeutically effective amount may be an amount which relieves to some extent one or more symptoms of a disease, condition, syndrome or disorder in a subject; and/or returns to normal either partially or completely one or more physiological or biochemical parameters associated with or causative of the disease, condition, syndrome or disorder; and/or reduces the likelihood of the onset of the disease, condition, syndrome or disorder, or symptom(s) thereof. "Pharmaceutically acceptable" means that, which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary as well as human pharmaceutical use. A "pharmaceutically acceptable salt” is intended to mean a salt of an acid or base of a compound represented by Formula (I) that is non-toxic, biologically tolerable, or otherwise biologically suitable for administration to the subject. See, generally, S.M. Berge, et al., “Pharmaceutical Salts”, J. Pharm. Sci., 1977, 66:1-19, and Handbook of Pharmaceutical Salts, Properties, Selection, and Use, Stahl and Wermuth, Eds., Wiley-VCH and VHCA, Zurich, 2002. Preferred pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of patients without undue toxicity, irritation, or allergic response. Non-limiting examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen-phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne- 1,4-dioates, hexyne-1,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, γ-hydroxybutyrates, glycolates, tartrates, methane-sulf onates, propanesulfonates, naphthalene- 1 -sulfonates, naphthalene-2-sulfonates, and mandelates.
A compound of Formula (I) may possess a sufficiently acidic group, a sufficiently basic group, or both types of functional groups, and accordingly react with a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
Compounds of Formula (I) may contain at least one nitrogen of basic character, so desired pharmaceutically acceptable salts may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, boric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, phenylacetic acid, propionic acid, stearic acid, lactic acid, ascorbic acid, maleic acid, hydroxymaleic acid, isethionic acid, succinic acid, valeric acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, oleic acid, palmitic acid, lauric acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as mandelic acid, citric acid, or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid, 2-acetoxybenzoic acid, naphthoic acid, or cinnamic acid, a sulfonic acid, such as laurylsulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, ethanesulfonic acid, any compatible mixture of acids such as those given as examples herein, and any other acid and mixture thereof that are regarded as equivalents.
Compounds of Formula (I) may contain a carboxylic acid moiety, a desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide, alkaline earth metal hydroxide, any compatible mixture of bases such as those given as examples herein, and any other base and mixture thereof that are regarded as equivalents or acceptable substitutes in light of the ordinary level of skill in this technology. Illustrative examples of suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, carbonates, bicarbonates, primary, secondary, and tertiary amines, and cyclic amines, such as benzylamines, pyrrolidines, piperidine, morpholine, piperazine, A-methyl-glucamine and tromethamine and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
Each compound used herein may be discussed interchangeably with respect to its chemical formula, chemical name, abbreviation, etc.
Any formula given herein is intended to represent compounds having structures depicted by the structural formula as well as certain variations or forms. In particular, compounds of any formula given herein may have asymmetric centers and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the general formula, and mixtures thereof, are considered within the scope of such formula. The compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (A)-stereoisomers or as mixtures thereof. Thus, any formula given herein is intended to represent a racemate, one or more of its enantiomeric forms, one or more of its diastereomeric forms, and mixtures thereof. Additionally, any formula given herein is intended to refer also to any one of: hydrates, solvates, polymorphs and of such compounds, and mixtures thereof, even if such forms are not listed explicitly.
The term “R” at a stereocenter designates that the stereocenter is purely of the R- configuration as defined in the art; likewise, the term “S” means that the stereocenter is purely of the 5-configuration. As used herein, the term “RS” refers to a stereocenter that exists as a mixture of the R- and 5-configurations.
Compounds containing one stereocenter drawn without a stereo bond designation are a mixture of 2 enantiomers. Compounds containing 2 stereocenters both drawn without stereo bond designations are a mixture of 4 diastereomers. Compounds with 2 stereocenters both labeled “RS” and drawn with stereo bond designations are a 2-component mixture with relative stereochemistry as drawn. Unlabeled stereocenters drawn without stereo bond designations are a mixture of the R- and 5-configurations. For unlabeled stereocenters drawn with stereo bond designations, the absolute stereochemistry is as depicted.
Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof. The methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art.
Reference to a compound herein stands for a reference to any one of: (a) the recited form of such compound, and (b) any of the forms of such compound in the medium in which the compound is being considered when named. For example, reference herein to a compound such as R-COOH, encompasses reference to any one of: for example, R-COOH(s), R-COOH(sol), and R-COO-(sol). In this example, R-COOH(s) refers to the solid compound, as it could be for example in a tablet or some other solid pharmaceutical composition or preparation; R- COOH(sol) refers to the undissociated form of the compound in a solvent; and R-COO-(sol) refers to the dissociated form of the compound in a solvent, such as the dissociated form of the compound in an aqueous environment, whether such dissociated form derives from R-COOH, from a salt thereof, or from any other entity that yields R-COO- upon dissociation in the medium being considered. In another example, an expression such as “exposing an entity to compound of formula R-COOH” refers to the exposure of such entity to the form, or forms, of the compound R-COOH that exists, or exist, in the medium in which such exposure takes place. In still another example, an expression such as “reacting an entity with a compound of formula R-COOH” refers to the reacting of (a) such entity in the chemically relevant form, or forms, of such entity that exists, or exist, in the medium in which such reacting takes place, with (b) the chemically relevant form, or forms, of the compound R-COOH that exists, or exist, in the medium in which such reacting takes place. In this regard, if such entity is for example in an aqueous environment, it is understood that the compound R-COOH is in such same medium, and therefore the entity is being exposed to species such as R-COOH(aq) and/or R-COO-(aq), where the subscript “(aq)” stands for “aqueous” according to its conventional meaning in chemistry and biochemistry. A carboxylic acid functional group has been chosen in these nomenclature examples; this choice is not intended, however, as a limitation but it is merely an illustration. It is understood that analogous examples can be provided in terms of other functional groups, including but not limited to hydroxyl, basic nitrogen members, such as those in amines, and any other group that interacts or transforms according to known manners in the medium that contains the compound. Such interactions and transformations include, but are not limited to, dissociation, association, tautomerism, solvolysis, including hydrolysis, solvation, including hydration, protonation, and deprotonation. No further examples in this regard are provided herein because these interactions and transformations in a given medium are known by any one of ordinary skill in the art.
Any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number in an enriched form. Examples of isotopes that can be incorporated into compounds of the invention in a form that exceeds natural abundances include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H (or chemical symbol D), 3H (or chemical symbol T), 11C, 13C, 14C, 15N, 18O, 17O, 31P, 32P, 35S, 18F, 36C1, and 125I, respectively. Such isotopically labelled compounds are useful in metabolic studies (preferably with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques [such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT)] including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18F or 11C labeled compound may be particularly preferred for PET or SPECT studies. Further, substitution with heavier isotopes such as deuterium (i.e., 2H, or D) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements. Isotopically labeled compounds of this invention can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
The term Cn-m alkyl refers to an aliphatic chain, whether straight or branched, with a total number N of carbon members in the chain that satisfies n ≤ N ≤ m, with m > n.
When the same plurality of substituents is assigned to various groups, the specific individual substituent assignment to each of such groups is meant to be independently made with respect to the specific individual substituent assignments to the remaining groups. By way of illustration, but not as a limitation, if each of groups Q and R can be H or F, the choice of H or F for Q is made independently of the choice of H or F for R, so the choice of assignment for Q does not determine or condition the choice of assignment for R, or vice-versa, unless it is expressly indicated otherwise. Illustrative claim recitation in this regard would read as “each of Q and R is independently H or F”, or “each of Q and R is independently selected from the group consisting of H and F”.
In another example, a zwitterionic compound would be encompassed herein by referring to a compound that is known to form a zwitterion, even if it is not explicitly named in its zwitterionic form. Terms such as zwitterion, zwitterions, and their synonyms zwitterionic compound(s) are standard lUPAC-endorsed names that are well known and part of standard sets of defined scientific names. In this regard, the name zwitterion is assigned the name identification CHEBI:27369 by the Chemical Entities of Biological Interest (ChEBI) dictionary of molecular entities. As generally well known, a zwitterion or zwitterionic compound is a neutral compound that has formal unit charges of opposite sign. Sometimes these compounds are referred to by the term “inner salts”. Other sources refer to these compounds as “dipolar ions”, although the latter term is regarded by still other sources as a misnomer. As a specific example, aminoethanoic acid (the amino acid glycine) has the formula H2NCH2COOH, and it exists in some media (in this case in neutral media) in the form of the zwitterion +H3NCH2COO-. Zwitterions, zwitterionic compounds, inner salts and dipolar ions in the known and well- established meanings of these terms are within the scope of this invention, as would in any case be so appreciated by those of ordinary skill in the art. Because there is no need to name each and every embodiment that would be recognized by those of ordinary skill in the art, no structures of the zwitterionic compounds that are associated with the compounds of this invention are given explicitly herein. They are, however, part of the embodiments of this invention. No further examples in this regard are provided herein because the interactions and transformations in a given medium that lead to the various forms of a given compound are known by any one of ordinary skill in the art.
When referring to any formula given herein, the selection of a particular moiety from a list of possible species for a specified variable is not intended to define the same choice of the species for the variable appearing elsewhere. In other words, where a variable appears more than once, the choice of the species from a specified list is independent of the choice of the species for the same variable elsewhere in the formula, unless stated otherwise.
By way of a first example on substituent terminology, if substituent S1 example is one of S1 and S2, and substituent S2 example is one of S3 and S4, then these assignments refer to embodiments of this invention given according to the choices S1 example is S1 and S2 example is S3; S1 example is S1 and S2 example is S4; S1 example is S2 and S2 example is S3; S1 example is S2 and S2 example is S4; and equivalents of each one of such choices. The shorter terminology “ S1 example is one of S1 and S2, and S2 example is one of S3 and S4” is accordingly used herein for the sake of brevity, but not by way of limitation. The foregoing first example on substituent terminology, which is stated in generic terms, is meant to illustrate the various substituent assignments described herein.
Furthermore, when more than one assignment is given for any member or substituent, embodiments of this invention comprise the various groupings that can be made from the listed assignments, taken independently, and equivalents thereof. By way of a second example on substituent terminology, if it is herein described that substituent Sexample is one of S1, S2, and S3, this listing refers to embodiments of this invention for which Sexample is S1; Sexample is S2; Sexample is S3; Sexample is one of S1 and S2; Sexample is one of S1 and S3; Sexample is one of S2 and S3; Sexample is one of S1, S2 and S3; and Sexample is any equivalent of each one of these choices. The shorter terminology “Sexample is one of S1, S2, and S3” is accordingly used herein for the sake of brevity, but not by way of limitation. The foregoing second example on substituent terminology, which is stated in generic terms, is meant to illustrate the various substituent assignments described herein.
The nomenclature “Ci-Cj” with j > i, when applied herein to a class of substituents, is meant to refer to embodiments of this invention for which each and every one of the number of carbon members, from i to j including i and j, is independently realized. By way of example, the term C1-C3 refers independently to embodiments that have one carbon member (C1), embodiments that have two carbon members (C2), and embodiments that have three carbon members (C3).
Embodiments of this invention include compounds of Formula (I),
Figure imgf000015_0001
wherein
R1 is C1-6alkyl or C1-6haloalkyl;
Figure imgf000015_0002
wherein
Ra is C1-6alkyl;
Rb is C1-6alkyl substituted with one OH member;
R3 is H or halo;
R4 is selected from the group consisting of:
Figure imgf000015_0003
Figure imgf000016_0001
wherein
Re is selected from the group consisting of: H; halo; C1-6alkyl; C1-6alkyl substituted with one member selected from the group consisting of: OH, OCH3, and OCF3;
C1-6haloalkyl; C1-6haloalkyl substituted with one member selected from the group consisting of: OH, and OCH3; and OC1-6alkyl;
Rf is selected from the group consisting of: H; halo; C1-6alkyl; C1-6alkyl substituted with one member selected from the group consisting of: OH, OCH3, and OCF3;
C1-6haloalkyl; C1-6haloalkyl substituted with one member selected from the group consisting of: OH, and OCH3; and OC1-6alkyl; and
Rgis H or C1-6alkyl; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein wherein R1 is C1-4alkyl; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein R1 is a p armaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer
Figure imgf000016_0002
thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein R2 is
Figure imgf000016_0003
, wherein Ra is CH3; and Rb is CH(OH)CH3, or C(OH)(CH3)2; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein R2 is
; or a pharmaceutically acceptable salt,
Figure imgf000016_0004
isotope, N-oxide, solvate, or stereoisomer thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein R2 is
Figure imgf000017_0001
; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein wherein R3 is F; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein R4 is
, where Re and R are halo; or a pharmaceutically acceptable salt, isotope, N-oxide,
Figure imgf000017_0002
solvate, or stereoisomer thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein R4 is or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer
Figure imgf000017_0003
thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein R4 is , wherein Rg is H; Re is C1-4alkyl, and Rf is halo; or a pharmaceutically acceptable
Figure imgf000017_0004
salt, isotope, N-oxide, solvate, or stereoisomer thereof.
An additional embodiment of the invention is a compound of Formula (I) wherein wherein R4 is
Figure imgf000017_0005
; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
An additional embodiment of the current invention is a compound selected from the compounds shown below in Table 1, and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof:
Table 1
Figure imgf000018_0001
Also within the scope of the invention are enantiomers and diastereomers of the compounds of Formula (I). Also within the scope of the invention are the pharmaceutically acceptable salts, N-oxides or solvates of the compounds of Formula (I). Also within the scope of the invention are the pharmaceutically acceptable prodrugs of compounds of Formula (I), and pharmaceutically active metabolites of the compounds of Formula (I).
Also within the scope of the invention are isotopic variations of compounds of Formula (I), such as, e.g., deuterated compounds of Formula (I). Also within the scope of the invention are the pharmaceutically acceptable salts, N-oxides or solvates of the isotopic variations of the compounds of Formula (I). Also within the scope of the invention are the pharmaceutically acceptable prodrugs of the isotopic variations of the compounds of Formula (I), and pharmaceutically active metabolites of the isotopic variations of the compounds of Formula (I). Even though the compounds of embodiments of the present invention (including their pharmaceutically acceptable salts and pharmaceutically acceptable solvates) can be administered alone, they will generally be administered in admixture with a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient and/or a pharmaceutically acceptable diluent selected with regard to the intended route of administration and standard pharmaceutical or veterinary practice.
Thus, particular embodiments of the present invention are directed to pharmaceutical and veterinary compositions comprising compounds of Formula (I) and at least one pharmaceutically acceptable carrier, pharmaceutically acceptable excipient, and/or pharmaceutically acceptable diluent. By way of example, in the pharmaceutical compositions of embodiments of the present invention, the compounds of Formula (I) may be admixed with any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilizing agent(s), and combinations thereof.
An embodiment of the invention relates to a pharmaceutical composition comprising an effective amount of at least one compound selected from compounds of Formula (I), and pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof, in accordance with any embodiment described herein; and at least one pharmaceutically acceptable excipient.
An additional embodiment of the invention is a pharmaceutical composition comprising: (A) an effective amount of at least one compound selected from compounds of Formula (I)
Figure imgf000019_0001
wherein
R1 is C1-6alkyl or C1-6haloalkyl; R2 is
Figure imgf000019_0002
wherein
Ra is C1-6alkyl;
Rb is C1-6alkyl substituted with one OH member;
R3 is H or halo; R4 is selected from the group consisting of:
Figure imgf000020_0001
Figure imgf000020_0002
wherein
Re is selected from the group consisting of: H; halo; C1-6alkyl; C1-6alkyl substituted with one member selected from the group consisting of: OH, OCH3, and OCF3; C1-6haloalkyl; C1-6haloalkyl substituted with one member selected from the group consisting of: OH, and OCH3; and OC1-6alkyl;
Rf is selected from the group consisting of: H; halo; C1-6alkyl; C1-6alkyl substituted with one member selected from the group consisting of: OH, OCH3, and OCF3; C1-6haloalkyl; C1-6haloalkyl substituted with one member selected from the group consisting of: OH, and OCH3; and OC1-6alkyl; and
Rgis H or C1-6alkyl; or pharmaceutically acceptable salts, isotopes, N-oxides, solvates, or stereoisomers thereof; and (B) at least one pharmaceutically acceptable excipient.
An additional embodiment of the invention is a pharmaceutical composition comprising an effective amount of a compound shown in Table 1 (e.g., a compound selected from Examples 1-7), or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer of the compound of Table 1, a pharmaceutically acceptable prodrug of the compound of Table 1, or a pharmaceutically active metabolite of the compound of Table 1; and at least one pharmaceutically acceptable excipient.
Solid oral dosage forms such as, tablets or capsules, containing one or more compounds of the present invention may be administered in at least one dosage form at a time, as appropriate. It is also possible to administer the compounds in sustained release formulations.
Additional oral forms in which the present inventive compounds may be administered include elixirs, solutions, syrups, and suspensions; each optionally containing flavoring agents and coloring agents. Alternatively, one or more compounds of Formula (I) can be administered by inhalation (intratracheal or intranasal) or in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder. For example, they can be incorporated into a cream comprising, consisting of, and/or consisting essentially of an aqueous emulsion of polyethylene glycols or liquid paraffin. They can also be incorporated, at a concentration of between about 1 % and about 10 % by weight of the cream, into an ointment comprising, consisting of, and/or consisting essentially of a wax or soft paraffin base together with any stabilizers and preservatives as may be required. An alternative means of administration includes transdermal administration by using a skin or transdermal patch.
The pharmaceutical compositions of the present invention (as well as the compounds of the present invention alone) can also be injected parenterally, for example, intracavernosally, intravenously, intramuscularly, subcutaneously, intradermally, or intrathecally. In this case, the compositions will also include at least one of a suitable carrier, a suitable excipient, and a suitable diluent.
For parenteral administration, the pharmaceutical compositions of the present invention are best used in the form of a sterile aqueous solution that may contain other substances, for example, enough salts and monosaccharides to make the solution isotonic with blood.
For buccal or sublingual administration, the pharmaceutical compositions of the present invention may be administered in the form of tablets or lozenges, which can be formulated in a conventional manner.
By way of further example, pharmaceutical compositions containing at least one of the compounds of Formula (I) as the active ingredient can be prepared by mixing the compound(s) with a pharmaceutically acceptable carrier, a pharmaceutically acceptable diluent, and/or a pharmaceutically acceptable excipient according to conventional pharmaceutical compounding techniques. The carrier, excipient, and diluent may take a wide variety of forms depending upon the desired route of administration (e.g., oral, parenteral, etc.). Thus, for liquid oral preparations such as, suspensions, syrups, elixirs and solutions, suitable carriers, excipients and diluents include water, glycols, oils, alcohols, flavoring agents, preservatives, stabilizers, coloring agents and the like; for solid oral preparations such as, powders, capsules, and tablets, suitable carriers, excipients and diluents include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like. Solid oral preparations also may be optionally coated with substances such as, sugars, or be enterically coated so as to modulate the major site of absorption and disintegration. For parenteral administration, the carrier, excipient and diluent will usually include sterile water, and other ingredients may be added to increase solubility and preservation of the composition. Injectable suspensions or solutions may also be prepared utilizing aqueous carriers along with appropriate additives such as, solubilizers and preservatives.
According to particular embodiments, a therapeutically effective amount of a compound of Formula (I) or a pharmaceutical composition thereof may comprise a dose range from about 0.1 mg to about 3000 mg, or any particular amount or range therein, in particular from about 1 mg to about 1000 mg, or any particular amount or range therein, of active ingredient in a regimen of about 1 to about (4x) per day for an average (70 kg) human; although, it is apparent to one skilled in the art that the therapeutically effective amount for a compound of Formula (I) will vary as will the diseases, syndromes, conditions, and disorders being treated.
An embodiment of the present invention is directed to a pharmaceutical composition for oral administration, comprising a compound of Formula (I) in an amount of from about 1 mg to about 500 mg.
Advantageously, a compound of Formula (I) may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three and (4x) daily.
Optimal dosages of a compound of Formula (I) to be administered may be readily determined and will vary with the particular compound used, the mode of administration, the strength of the preparation, and the advancement of the disease, syndrome, condition or disorder. In addition, factors associated with the particular subject being treated, including subject gender, age, weight, diet and time of administration, will result in the need to adjust the dose to achieve an appropriate therapeutic level and desired therapeutic effect. The above dosages are thus exemplary of the average case. There can be, of course, individual instances wherein higher or lower dosage ranges are merited, and such are within the scope of this invention.
Compounds of Formula (I) may be administered in any of the foregoing compositions and dosage regimens or by means of those compositions and dosage regimens established in the art whenever use of a compound of Formula (I) is administered to a subject in need thereof.
According to particular embodiments, one or more compounds of Formula (I) are useful in methods for treating, ameliorating and / or preventing a disease, a syndrome, a condition or a disorder that is affected by the inhibition of DHODH enzymatic activity. An additional embodiment of the invention relates to the use of compounds of Formula (I), e.g., by inhibiting dihydroorotate oxygenase enzyme activity, in treating disorders like inflammatory disorders, autoimmune disorders, or cancer; or pharmaceutically acceptable salts, isotopes, N-oxides, solvates, or stereoisomers thereof.
In a further aspect the present invention provides a method for inhibiting or altering Dihydroorotate Dehydrogenase (DHODH) enzymatic activity, the method comprising contacting DHODH with any compound of Formula (I), aspect or embodiment disclosed herein, thereby inhibiting or otherwise altering DHODH enzymatic activity.
An additional embodiment of the present invention provides methods for treating diseases, disorders, or medical conditions mediated or otherwise affected by dihydroorotate dehydrogenase (DHODH) enzyme activity comprising administering a compound of Formula (I) to a subject in need thereof.
As used herein, the term "DHODH inhibitor" may refer to an agent that inhibits or reduces DHODH activity.
In one embodiment, the term “therapeutically effective amount” (or “effective amount”) refers to the amount of a compound of the present invention that, when administered to a subject, is effective to (1) at least partially alleviate, inhibit, prevent, and/ or ameliorate a condition, or a disorder or a disease (i) mediated by DHODH enzymatic activity; or (ii) associated with DHODH enzymatic activity; or (iii) characterized by activity (normal or abnormal) of DHODH enzyme; or (2) reduce or inhibit the activity of DHODH enzyme; or (3) reduce or inhibit the expression of DHODH; or (4) modify the protein levels of DHODH. Without being bound by a particular theory, DHODH inhibitors are believed to act by inhibiting nucleic acid synthesis, cell cycle arrest or altering post-translational glycosylation of proteins involved in regulating myeloid differentiation within progenitor tumor cells.
An additional embodiment of the invention is a method of treating a subject suffering from or diagnosed with a disease, disorder, or medical condition mediated or otherwise affected by DHODH enzymatic activity, comprising administering to a subject in need of such treatment an effective amount of at least one compound selected from: compounds of Formula (I), such as a compound of Table 1), enantiomers and diastereomers of the compounds of Formula (I) (such as a compound of Table 1), isotopic variations of the compounds of Formula (I) (such as a compound of Table 1), and pharmaceutically acceptable salts of all of the foregoing. Stated another way, according to an embodiment, a method of treating a subject suffering from or diagnosed with a disease, disorder, or medical condition, such as cancer, comprises administering to the subject an effective amount of at least one compound selected from: compounds of Formula (I) (such as a compound of Table 1), and pharmaceutically acceptable salts of all the foregoing (e.g., by inhibiting or otherwise altering dihydroorotate oxygenase enzyme activity in the subject).
In another embodiment, inhibitors of DHODH of the present invention may be used for the treatment of immunological diseases including, but not limited to, autoimmune and inflammatory disorders, e.g. arthritis, inflammatory bowel disease, gastritis, ankylosing spondylitis, ulcerative colitis, pancreatitis, Crohn’s disease, celiac disease, multiple sclerosis, systemic lupus erythematosus, lupus nephritis, rheumatic fever, gout, organ or transplant rejection, chronic allograft rejection, acute or chronic graft-versus-host disease, dermatitis including atopic, dermatomyositis, psoriasis, Behcet’s diseases, uveitis, myasthenia gravis, Grave’s disease, Hashimoto thyroiditis, Sjogren’s syndrome, blistering disorders, antibody- mediated vasculitis syndromes, immune-complex vasculitides, allergic disorders, asthma, bronchitis, chronic obstructive pulmonary disease (COPD), cystic fibrosis, pneumonia, pulmonary diseases including edema, embolism, fibrosis, sarcoidosis, hypertension and emphysema, silicosis, respiratory failure, acute respiratory distress syndrome, BENTA disease, berylliosis, and polymyositis.
As used herein, unless otherwise noted, the term “affect” or “affected” (when referring to a disease, disorder, or medical condition that is affected by the inhibition or alteration of DHODH enzymatic activity) includes a reduction in the frequency and / or severity of one or more symptoms or manifestations of said disease, syndrome, condition or disorder; and / or includes the prevention of the development of one or more symptoms or manifestations of said disease, syndrome, condition or disorder or the development of the disease, condition, syndrome or disorder.
An additional embodiment of the invention provides a method of treatment of cancer comprising administering to a subject in need thereof, a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof. According to an embodiment, the cancer is selected from but not limited to, lymphomas, leukemias, carcinomas, and sarcomas.
An additional embodiment of the invention provides the use of a compound of Formula (I), or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof, for the treatment of one or more cancer types.
According to particular embodiments, the uses and methods of treatment described herein are directed to the treatment of cancer, wherein the cancer is selected from but not limited to: leukemias including but not limited to acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), (acute) T-cell leukemia, acute monocytic leukemia, acute promyelocytic leukemia (APL), bisphenotypic B myelomonocytic leukemia, chronic myeloid leukemia (CML), chronic myelomonocytic leukemia (CMML), large granular lymphocytic leukemia, plasma cell leukemia, and also myelodysplastic syndrome (MDS), which can develop into an acute myeloid leukemia, lymphomas including but not limited to AIDS-related lymphoma, Hodgkin lymphoma, non-Hodgkin's lymphoma (NHL), T-non-Hodgkin lymphoma (T-NHL), subtypes of NHL such as Diffuse Large Cell Lymphoma (DLBCL), activated B-cell DLBCL, germinal center B-cell DLBCL, double-hit lymphoma and double-expressor lymphoma; anaplastic large cell lymphoma, marginal B cell lymphoma and primary mediastinal B-cell lymphoma, immunoblastic large cell lymphoma, Burkitt lymphoma, follicular lymphoma, hairy cell leukemia, Hodgkin's disease, mantle cell lymphoma (MCL), lymphoplasmatic lymphoma, precursor B -lymphoblastic lymphoma, lymphoma of the central nervous system, small lymphocytic lymphoma (SLL) and chronic lymphocytic leukemia (CLL); T-cell NHL such as precursor T- lymphoblastic lymphoma/leukemia, peripheral T-cell lymphoma (PTCL), cutaneous T-cell lymphoma (CTCL), angioimmunoblastic T-cell lymphoma, extranodal natural killer T-cell lymphoma, enteropathy type T-cell lymphoma, subcutaneous panniculitis-like T-cell lymphoma, anaplastic large cell lymphoma sarcomas including but not limited to sarcoma of the soft tissue, gliosarcoma, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma; and other cancers, such as solid tumors, including but not limited to breast cancer, colorectal carcinoma, gastric cancer, gliosarcoma, head & neck cancer, hepatocellular carcinoma, lung cancer, multiple myeloma, neuroblastoma, ovarian cancer, pancreatic cancer, prostate cancer, renal cell carcinoma and sarcoma.
In an embodiment, cancers that may benefit from a treatment with inhibitors of DHODH of the present invention include, but are not limited to, lymphomas, leukemias, carcinomas, and sarcomas, e.g. non-Hodgkin’s lymphoma, diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), follicular lymphoma (FL), marginal zone lymphoma, T-cell lymphoma, Hodgkin’s lymphoma, Burkitt’s lymphoma, multiple myeloma, brain (gliomas), glioblastomas, breast cancer, colorectal/colon cancer, prostate cancer, lung cancer including non-small-cell, gastric cancer, endometrial cancer, melanoma, pancreatic cancer, liver cancer, kidney cancer, squamous cell carcinoma, ovarian cancer, sarcoma, osteosarcoma, thyroid cancer, bladder cancer, head & neck cancer, testicular cancer, Ewing’s sarcoma, rhabdomyosarcoma, medulloblastoma, neuroblastoma, cervical cancer, renal cancer, urothelial cancer, vulval cancer, esophageal cancer, salivary gland cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, and GIST (gastrointestinal stromal tumor).
In another embodiment of the present invention, the compounds of the present invention may be employed in combination with one or more other medicinal agents, more particularly with one or more anti-cancer agents, e.g. chemotherapeutic, anti-proliferative or immunomodulating agents, or with adjuvants in cancer therapy, e.g. immunosuppressive or antiinflammatory agents. Additional non-limiting examples of anti-cancer agents that may be administered in combination with a compound of the present invention include biologic compounds, such as monoclonal antibodies (e.g., that mediate effector function upon binding to cancer cell-associated antigens, or block interaction of a receptor expressed on cancer cells with a soluble or cell bound ligand), bispecific antibodies that mediate immune cell redirection, etc. According to an embodiment, a method of treating cancer comprises administering an effective amount of a compound of the present invention (e.g., selected from compounds of Formula (I), such as a compound shown in Table 1, pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof) and an effective amount of one or more additional anticancer agents, wherein the method comprises administering the compound of the present invention and the additional anti-cancer agent(s) either simultaneously (e.g., as part of the same pharmaceutical composition) or sequentially. According to an embodiment, a pharmaceutical composition comprises an effective amount of a compound of the present invention (e.g., selected from compounds of Formula (I), such as a compound shown in Table 1, pharmaceutically acceptable salts, isotopes, N-oxides, solvates, and stereoisomers thereof), an effective amount of one or more additional anti-cancer agents, and optionally one or more excipients.
An additional embodiment of the invention provides the use of a compound of Formula (I), or pharmaceutically acceptable salts, isotopes, N-oxides, solvates, or stereoisomers thereof, as part of chemotherapeutic regimens for the treatment of cancers, lymphomas and leukemias alone or in combination with classic antitumoral compounds well known by the one skilled in the art.
GENERAL SYNTHETIC METHODS
Exemplary compounds useful in methods of the invention will now be described by reference to the illustrative synthetic schemes for their general preparation below and the specific examples that follow. Artisans will recognize that, to obtain the various compounds herein, starting materials may be suitably selected so that the ultimately desired substituents will be carried through the reaction scheme with or without protection as appropriate to yield the desired product. Alternatively, it may be necessary or desirable to employ, in the place of the ultimately desired substituent, a suitable group that may be carried through the reaction scheme and replaced as appropriate with the desired substituent. Unless otherwise specified, the variables are as defined above in reference to Formula (I). Reactions may be performed between the melting point and the reflux temperature of the solvent, and preferably between 0 °C and the reflux temperature of the solvent. Reactions may be heated employing conventional heating or microwave heating. Reactions may also be conducted in sealed pressure vessels above the normal reflux temperature of the solvent. Abbreviations used in the instant specification, particularly the schemes and examples, are as follows in Table 2:
Table 2.
Figure imgf000028_0001
Figure imgf000029_0001
PREPARATIVE EXAMPLES
Exemplary compounds useful in methods of the invention will now be described by reference to the illustrative synthetic schemes for their general preparation below and the specific examples to follow.
SCHEME 1
Figure imgf000030_0001
According to SCHEME 1, 4-bromo-2,5-difluorobenzonitrile is reacted with an alcohol compound of formula (V), where R1 is C1-6alkyl or C1-6haloalkyl; a suitable base such as K2CO3, Na2CO3, Cs2CO3,NaH, KHMDS, LiHMDS, and NaHMDS, preferably NaH; in a suitable solvent such as THF, DMF, and ACN, preferably THF. Subsequent hydrolysis in the presence of a base such as NaOH, LiOH, and KOH, preferably NaOH; in a suitable solvent such as EtOH, THF, DMF, water, and ACN, or a mixture thereof, preferably in EtOH; affords an aryl acid compound of formula (VI). A compound of formula (VI) is reacted with thionyl chloride, oxalyl chloride, and the like; in a suitable solvent such as MeOH; provides a methyl ester compound of formula (VII). Ullmann-type aromatic amination reaction of compound of formula (VII) with a nucleophilic heteroaromatic compound of formula (VIII), where Ra and Rb are as defined in claim 1 ; in the presence of catalytic Cui; a diamine ligand such as trans-dimethylcyclohexane- 1,2-diamine; and a base such as K3PO4, K2CO3, CS2CO3, NaHCO3, triethylamine, and the like, preferably K2CO3; in a suitable solvent such as 1,4-dioxane, DMSO, DMF, THF, ACN, and the like, preferably 1,4-dioxane; provides a compound of formula (IX).
SCHEME 2
Figure imgf000030_0002
According to SCHEME 2, ethyl 4-methyl-1H-pyrazole-3-carboxylate is protected with a THP group employing conditions known to one skilled in the art, for example reaction with 3,4- dihydro-2H-pyran (DHP) and p-toluenesulfonic acid (TsOH) in a suitable solvent such as DCM, DCE, and the like; at room temperature; to afford ethyl 4-methyl-l-(tetrahydro-2H-pyran-2-yl)- 1H-pyrazole-3 -carboxylate. Reduction of ethyl 4-methyl-l-(tetrahydro-2H-pyran-2-yl)-1H- pyrazole-3 -carboxylate, using a suitable reducing agent such as DIBAL-H; in a suitable solvent such as toluene, and the like; at temperatures ranging from -78 °C to room temperature; affords 4-methy 1- 1 -(tetrahydro-2H-pyran-2-yl)- 1 H-pyrazole-3 -carbaldehyde. 4-Methy 1- 1 -(tetrahydro- 2H-pyran-2-yl)-1H-pyrazole-3-carbaldehyde is reacted in a Grignard addition, using MeMgBr; in a suitable solvent, such as THF, ether, 1,4-dioxane, and the like; at 0 °C; provides l-(4- methyl-l-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)ethan-l-ol. Deprotection of the THP protecting group is achieved employing conditions known to one skilled in the art; for example, using HC1, in a suitable solvent such as MeOH; at a temperature of about 60 °C; affords a a compound of formula (VIII), where Ra is C1-6alkyl and Rb is CH(CH3)(OH). Final chiral separation using SFC affords the two resolved enantiomers.
SCHEME 3
Figure imgf000031_0001
According to SCHEME 3, a compound of formula (VIII), where Ra is C1-6alkyl and Rb is C(CH3)2(OH); is prepared by reaction of ethyl 4-methyl-l H-pyrazole-3 -carboxylate with MeMgBr; in a suitable solvent such as THF, ether, 1 ,4-dioxane and the like; at low temperatures, preferably 0 °C.
SCHEME 4
Figure imgf000032_0001
According to SCHEME 4, a compound of formula (VII) is reacted in a Pd-catalyzed amination reaction with benzophenone imine employing cross-coupling conditions known to one skilled in the art. For example, a compound of formula (VII), where R1 is C1-6alkyl or C1-6haloalkyl; is reacted with a palladium catalyst such as Pd2(dba)y a suitable ligand, such as XantPhos and the like; and a suitable base, such as CS2CO3, K2CO3, NaOtBu and the like; in a solvent such as toluene, and the like; at refluxing temperatures; to provide a compound of formula (X). Reaction of commercially available or synthetically accessible amine compounds of formula (XI), where R3 is as defined in claim 1; with a suitable Lewis acid such as A1Me3, A1C13, and the like; in a suitable aprotic solvent such as DCM, toluene, and the like; at elevated temperatures, preferably 90 °C; affords a compound of formula (XII). Imine cleavage is achieved employing methods known to one skilled in the art. For example, treatment of a compound of formula (XII), with HC1 in MeOH, EtOH, and the like; at room temperature for a period of 16 h; affords a compound of formula (XIII). A compound of formula (XIV) is prepared in two steps by first, diazotization, employing conditions known to one skilled in the art. For example, reaction with NaNO2; in an acidic aqueous solution such as AcOH in H2O, or other nitrite reagents; at a temperature of <5 °C. Subsequent reaction with ethyl 3-oxobutanoate, and a suitable base, such as Na2CO3, K2CO3, and the like; in a solvent such as EtOH, MeOH and the like. A triazole compound of formula (XV), is prepared by reaction of a compound of formula (XIV), with an ammonium salt such as, NH4OAc, and a copper catalyst, such as CuC12, and the like; in a suitable solvent such as EtOH, MeOH, and the like; at elevated temperatures, preferably 90 °C.
SCHEME 5
Figure imgf000033_0001
According to SCHEME 5, a compound of formula (IX), where R1 is C1-6alkyl or
C1-6haloalkyl, and R2 is where Ra is C1-6alkyl and Rb is CH(CH3)(OH) or
Figure imgf000033_0002
C(CH3)2(OH), is reacted with a commercially available or synthetically accessible amine compound of formula (XI), where R4 is as defined in claim 1; a suitable Lewis acid such as A1Me3, A1C13, and the like; in a suitable aprotic solvent such as DCM, toluene, and the like; at elevated temperatures, preferably 90 °C; affords compounds of Formula (I).
SCHEME 6
Figure imgf000033_0003
According to SCHEME 6, a compound of formula (XV), where R1 is C1-6alkyl or C1-6haloalkyl, and R4 is as described in claim 1; is reacted in a Grignard addition reaction employing MeMgBr; in a suitable solvent, such as THF, ether, 1,4-dioxane, and the like; at low temperatures; to provide compound of Formula (I), R2 is , where Ra is C1-6alkyl; and
Figure imgf000033_0004
Rb is C1-6alkyl substituted with one OH member.
Compounds of Formula (I) may be converted to their corresponding salts using methods known to one of ordinary skill in the art. For example, an amine of Formula (I) is treated with trifluoroacetic acid, HC1, or citric acid in a solvent such as Et2O, CH2CI2, THF, MeOH, chloroform, or isopropanol to provide the corresponding salt form. Alternately, trifluoroacetic acid or formic acid salts are obtained as a result of reverse phase HPLC purification conditions. Crystalline forms of pharmaceutically acceptable salts of compounds of Formula (I) may be obtained in crystalline form by recrystallization from polar solvents (including mixtures of polar solvents and aqueous mixtures of polar solvents) or from non-polar solvents (including mixtures of non-polar solvents).
Where the compounds according to this invention have at least one chiral center, they may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention.
Compounds prepared according to the schemes described above may be obtained as single forms, such as single enantiomers, by form-specific synthesis, or by resolution. Compounds prepared according to the schemes above may alternately be obtained as mixtures of various forms, such as racemic (1 : 1) or non-racemic (not 1: 1) mixtures. Where racemic and non-racemic mixtures of enantiomers are obtained, single enantiomers may be isolated using conventional separation methods known to one of ordinary skill in the art, such as chiral chromatography, recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation. Where regioisomeric or diastereomeric mixtures are obtained, as applicable, single isomers may be separated using conventional methods such as chromatography or crystallization.
The following specific examples are provided to further illustrate the invention and various preferred embodiments.
EXAMPLES
In obtaining the compounds described in the examples below and the corresponding analytical data, the following experimental and analytical protocols were followed unless otherwise indicated.
Unless otherwise stated, reaction mixtures were magnetically stirred at room temperature (rt) under a nitrogen atmosphere. Where solutions were “dried,” they were generally dried over a drying agent such as Na2SO4 or MgSO4. Where mixtures, solutions, and extracts were “concentrated”, they were typically concentrated on a rotary evaporator under reduced pressure.
Normal-phase silica gel chromatography (FCC) was performed on silica gel (SiO2) using prepacked cartridges. Preparative reverse-phase high performance liquid chromatography (RP HPLC) was performed on either:
METHOD A. A Gilson GX-281 semi-prep-HPLC with Phenomenex Synergi C18(10μm, 150 x 25mm), or Boston Green ODS C18(5μm, 150 x 30mm), and mobile phase of 5-99% ACN in water (with 0.225%FA) over 10 min and then hold at 100% ACN for 2 min, at a flow rate of 25 mL/min. or
METHOD B. A Gilson GX-281 semi-prep-HPLC with Phenomenex Synergi C18(10μm, 150 x 25mm), or Boston Green ODS C18(5μm, 150 x 30mm), and mobile phase of 5-99% ACN in water(0.1 %TFA) over 10 min and then hold at 100% ACN for 2 min, at a flow rate of 25 mL/min. or
METHOD C. A Gilson GX-281 semi-prep-HPLC with Phenomenex Synergi C18(10μm, 150 x 25mm), or Boston Green ODS C18(5μm, 150 x 30mm), and mobile phase of 5-99% ACN in water(0.05%HCl) over 10 min and then hold at 100% ACN for 2 min, at a flow rate of 25 mL/min. or
METHOD D. a Gilson GX-281 semi-prep-HPLC with Phenomenex Gemini C18 (10μm, 150 x 25mm), AD(10μm, 250mm x 30mm), or Waters XBridge C18 column (5μm, 150 x 30mm), mobile phase of 0-99% ACN in water (with 0.05% ammonia hydroxide v/v) over 10 min and then hold at 100% ACN for 2 min, at a flow rate of 25 mL/min. or
METHOD E. a Gilson GX-281 semi-prep-HPLC with Phenomenex Gemini Cl 8 (10μm, 150 x 25mm), or Waters XBridge Cl 8 column (5μm, 150 x 30mm), mobile phase of 5-99% ACN in water(10mM NH4HCO3) over 10 min and then hold at 100% ACN for 2 min, at a flow rate of 25 mL/min.
Preparative supercritical fluid high performance liquid chromatography (SFC) was performed either on a Thar 80 Prep-SFC system, or Waters 80Q Prep-SFC system from Waters. The ABPR was set to lOObar to keep the CO2 in SF conditions, and the flow rate may verify according to the compound characteristics, with a flow rate ranging from 50g/min to 70g/min. The column temperature was ambient temperature Mass spectra (MS) were obtained on a SHIMADZU LCMS-2020 MSD or Agilent 1200\G6110AMSD using electrospray ionization (ESI) in positive mode unless otherwise indicated. Calculated (calcd.) mass corresponds to the exact mass.
Nuclear magnetic resonance (NMR) spectra were obtained on Bruker model AVIII 400 spectrometers. Definitions for multiplicity are as follows: s = singlet, d = doublet, t= triplet, q = quartet, dd = doublet of doublets, ddd = doublet of doublet of doublets, td = triplet of doublets, dt = doublet of triplets, spt = septet, quin = quintet, m = multiplet, br = broad. It will be understood that for compounds comprising an exchangeable proton, said proton may or may not be visible on an NMR spectrum depending on the choice of solvent used for running the NMR spectrum and the concentration of the compound in the solution.
Chemical names were generated using ChemDraw Ultra 17.1 (CambridgeSoft Corp., Cambridge, MA) or OEMetaChem Vl.4.0.4 (Open Eye).
Compounds designated as R* or S* are enantiopure compounds where the absolute configuration was not determined.
Intermediate 1 : (M-5-Fluoro-4-(4.4.5.5-tetramethyl- 1.3.2-dioxaborolan-2-yl)-A-(o-tolyl)-2- ((1.1.1- trifluoropropan-2-yl)oxy)benzamide.
Figure imgf000036_0001
Step A, (5)-4-Bromo-5-fluoro-2-((1,1,1-trifluoropropan-2-yl)oxy)benzonitrile. A mixture of (S)- 1,1,1 -trifluoropropan-2-ol (11.5 g, 101 mmol) in THF (50 mL) was added to the mixture of NaH (60% purity, 4.4 g, 110 mmol) in THF (80 mL) dropwise at 0 °C under N2. The mixture was stirred at room temperature for 0.5 hours. 4-Bromo-2,5-difluorobenzonitrile (20 g, 91.7 mmol) in THF (70 mL) was added to the mixture dropwise at 0 °C under N2. The mixture was stirred at room temperature for 12 hours. Sat. aq. NH4CI (100 mL) was added to the mixture dropwise at 0 °C. The mixture was extracted with ethyl acetate (100 mL x 3). The combined organic layers were washed with brine (200 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (S 1O2, petroleum ether/ethyl acetate =1/0 to 30/1) to give the title compound (26 g, 81.4 mmol, 89% yield) as a white solid. MS (ESI): mass calcd. for C10H6BrF4NO, 311.0; m/z found, 314.0 [M+H]+. 1 H NMR (400MHz, CDCl3) δ = 7.37 (d, J= 7.3 Hz, 1H), 7.29 (d, J= 5.3 Hz, 1H), 4.67 (spt, J= 6.1 Hz, 1H), 1.62 (d, J= 6.5 Hz, 3H); 19F NMR (376MHz, CDCl3) δ = -78.19 (s, 3F), -111.75 (s, IF).
Step B, (S)-4-Bromo-5-fluoro-2-((l.l.1-trifluoropropan-2-yl)oxy)benzoic acid. To a mixture of (S)-4-bromo-5-fluoro-2-((1,1,1-trifluoropropan-2-yl)oxy)benzonitrile (26 g, 81.4 mmol) in EtOH (150 mL) was added 2 M aq. NaOH (151 mL, 301 mmol ) under N2. The mixture was stirred at 90 °C for 16 hours under N2. The mixture was concentrated and H2O (200 mL) was added to the mixture. The mixture was adjusted to pH=3 with 2 M aq. HC1 at 0 °C. The mixture was filtered. The filter cake was washed with H2O (150 mL x 3). The filter cake was collected and dried under vacuum to give the title compound (27 g, 75.8 mmol, 93% yield) as a yellow solid. MS (ESI): mass calcd. for C10H7BrF4O3, 330.0; m/z found, 332.9 [M+H]+. 1 H NMR (400MHz, CDCl3) δ = 7.84 (d, J= 8.3 Hz, 1H), 7.30 (d, J= 5.3 Hz, 1H), 4.69 - 4.82 (m, 1H), 1.61 (d, J = 6.3 Hz, 3H); 19F NMR (376MHz, CDCl3) δ = -78.11 (s, 3F), -112.52 (br s, 1F).
Step C. (M-Methyl 4-bromo-5-fluoro-2-((l.l.1-trifluoropropan-2-yl)oxy)benzoate. To a solution of (S)-4-bromo-5-fluoro-2-((1,1,1-trifluoropropan-2-yl)oxy)benzoic acid (27 g, 76 mmol) in MeOH (500 mL) was added SOCl2 (11 mL, 152 mmol) dropwise at 0 °C over 15 minutes. The mixture was stirred at 70 °C for 4 hours. The reaction mixture was concentrated under reduced pressure. The residue was diluted with H2O (150 mL) and extracted with ethyl acetate (100 mL x 3). The combined organic layers were washed with brine (300 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The crude was purified by column chromatography (SiO2 gradient elution: 0 - 1.5% ethyl acetate in petroleum ether) to give the title compound (26 g, 73.2 mmol, 97% yield) as a yellow oil. MS (ESI): mass calcd. for C11H9BrF4O3, 344.0; m/z found, 344.8 [M+H]+. 1H NMR (400MHz, CDCl3) δ = 7.62 (d, J= 8.5 Hz, 1H), 7.26 (d, J= 5.5 Hz, 1H), 4.61 (spt, J= 6.2 Hz, 1H), 3.91 (s, 3H), 1.55 (d, J= 6.5 Hz, 3H); 19F NMR (376MHz, CDCl3) δ = -78.13 (s, 3F), -113.57 (s, IF).
Intermediate 2, (5*)-1-(4-Methyl-l H-pyrazol-3-yl)ethan-1-ol.
Figure imgf000037_0001
Step A, Ethyl 4-methyl- l -(yetrahvdro-2H-pyran-2-yl)- 1 H-pyrazole-3 -carboxy late. At 0 °C, DHP (4.1 g, 49 mmol) was added to a mixture of ethyl 4-methyl-l H-pyrazole-3 -carboxylate (5.0 g, 32 mmol) and TsOH (558 mg, 3.2 mmol) in DCM (50 mL). The resulting mixture was stirred at 20 °C for 15 hours under an atmosphere of nitrogen. The reaction mixture was slowly poured into sat. aq. NaHCCh (30 mL) and extracted with DCM (20 mL x 2). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude was purified by column chromatography (SiO2, gradient elution: 0 - 20% ethyl acetate in petroleum ether) to give the title compound (8.5 g, 32 mmol, 99% yield) as a yellow oil. MS (ESI): mass calcd. for C12H18N2O3, 238.1; m/z found, 239.1 [M+H]+ 1H NMR (400 MHz, CDCl3) δ 7.45 (s, 1H), 5.42 (dd, J = 2.5, 9.8 Hz, 1H), 4.39 (q, J= 7.3 Hz, 2H), 4.10 - 4.01 (m, 1H), 3.67 (dt, J = 2.8, 11.3 Hz, 1H), 2.29 (s, 3H), 2.08 - 1.96 (m, 3H), 1.74 - 1.58 (m, 3H), 1.39 (t, J = 7.2 Hz, 3H) ppm.
B. 4-Methyl-1-(tetrahydro-2H-pyran-2-ypl)-1H-pyrazole-3-carbaldehyde . At 78 °C, and under an atmosphere of nitrogen, DIBAL-H (1 M solution in toluene, 95 mL, 95 mmol) was added dropwise to a mixture of ethyl 4-methyl-1- (tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3- carboxylate (8.4 g, 32 mmol) in DCM (160 mL). The reaction mixture was stirred at -78 °C for 1 hour. The reaction mixture was quenched by addition of 1 M aq. HC1 (20 mL), then the mixture was filtered. The filtrate was extracted with DCM (100 mL x 3). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude was purified by column chromatography (SiO2, gradient elution: 0 - 20% ethyl acetate in petroleum ether) to give the title compound (4.2 g, 22 mmol, 68% yield) as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 10.03 (d, J= 0.7 Hz, 1H), 7.44 (s, 1H), 5.39 (dd, J= 3.5, 8.5 Hz, 1H), 4.09 - 4.02 (m, 1H), 3.77 - 3.65 (m, 1H), 2.30 (d, J= 0.7 Hz, 3H), 2.19 - 2.04 (m, 3H), 1.79 - 1.61 (m, 3H) ppm.
Step C. 1-(4-Methyl-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazol-3-yl)ethan-1-ol. To a solution of
4-methyl- 1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazole-3-carbaldehyde (2.0 g, 10.3 mmol) in THF (40 mL) at 0 °C, was added MeMgBr (3 M solution in ether, 10 mL, 31 mmol) dropwise. The resulting mixture was stirred at 0 °C for 3 hours. The reaction mixture was slowly poured into sat. aq. NH4CI (30 mL) and extracted with DCM (30 mL x 2). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude was purified by column chromatography (SiO2, gradient elution: 0 - 60% ethyl acetate in petroleum ether) to give the title compound (1.8 g, 8.4 mmol, 81% yield) as a colorless oil. MS (ESI): mass calcd. for C11H18N2O2, 210.1; m/z found, 211.0 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 7.33 (s, 1H), 5.34 - 5.23 (m, 1H), 4.94 (q, J= 6.4 Hz, 1H), 4.06 (br d, J= 11.5 Hz, 1H), 3.73 - 3.59 (m, 1H), 2.03 (br d, J= 13.1 Hz, 6H), 1.71 - 1.58 (m, 3H), 1.53 (d, J= 6.5 Hz, 3H) ppm.
Step D. 1-(4-Methyl-1H-pyrazol-3-yl)ethan-1-ol. To a solution of 1-(4-methyl-1-(tetrahydro-2H- pyran-2-yl)-1H-pyrazol-3-yl)ethanol (1.7 g, 7.9 mmol) in MeOH (17 mL) was added 12 M aq. HC1 (3.3 mL, 39 mmol). Then the resulting mixture was stirred at 60 °C for 1 hour. To the reaction mixture was added sat. aq. Na2CO2 dropwise to pH=12 and extracted with DCM/MeOH (v/v, 10/1, 10 mL x 4). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude was purified by column chromatography (SiO2, gradient elution: 0 - 100% ethyl acetate in petroleum ether) to give the title compound (900 mg, 7.0 mmol, 89% yield) as colorless oil. MS (ESI): mass calcd. for C6H10N2O, 126.1; m/z found, 127.2 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 12.24 (br s, 1H), 7.24 (br s, 1H), 4.99 (br d, J = 1.5 Hz, 1H), 4.82 - 4.71 (m, 1H), 2.00 (s, 3H), 1.35 (d, J= 6.5 Hz, 3H) ppm. SFC: purity 49.8% / 50.2%, retention time 3.580 min / 3.865 min; method: AD-3_EtOH(DEA)_5_40_2,5ML_7MIN. Step E, (S*)-1-(4-Methyl-1H-pyrazol-3-yl)ethanol. The racemic 1-(4-methyl-1H-pyrazol-3- yl)ethanol (900 mg, 7.0 mmol) was separated by SFC (Stationary phase: DAICEL CHIRALPAK AD-H, 5 μm 250 mm x 30 mm); Mobile phase: Supercritical CO2 (A) - EtOH (0.1% NH3.H2O) (B), gradient elution: 20% B in A at 60 mL/min) to give the first eluting compound (S*)-1-(4- methyl-1H -pyrazol-3-yl)ethanol (299 mg, 2.4 mmol, 33% yield) as yellow oil. MS (ESI): mass calcd. for C6H10N2O, 126.1; m/z found, 127.1 [M+H]+. 1H NMR (400 MHz, DMSO-t/e) δ 7.28 (s, 1H), 5.09 - 4.93 (m, 1H), 2.06 (s, 3H), 1.52 (d, J= 6.5 Hz, 3H) ppm. SFC: purity 99.6%, retention time 3.584 min; method: AD-3_EtOH(DEA)_5_40_2,5ML_7MIN.
Intermediate 3: (R*)-1-(4-Methyl-17/-pyrazol-3-yl)ethanol.
Figure imgf000039_0001
From preparation of Intermediate 2, Step E. The second eluting compound (R*)-1-(4-Methyl- 1H-pyrazol-3-yl)ethanol (206 mg, 1.6 mmol, 23% yield) as white solid. MS (ESI): mass calcd. for C6H10N2O, 126.1; m/z found, 127.1 [M+H]+. 1H NMR (400MHz, DMSO-d6 ) δ 7.33 - 7.29 (m, 1H), 5.02 (d, J= 6.5 Hz, 1H), 2.08 (s, 3H), 1.54 (d, J= 6.8 Hz, 3H) ppm. SFC: purity 99.4%, retention time 3.862 min; method: AD-3_EtOH(DEA)_5_40_2.5ML 7MIN. Intermediate 4: 2-(4-Methyl-1H-pyrazol-3-yl)propan-2-ol.
Figure imgf000040_0001
To solution of ethyl 4-methyl-1H -pyrazole-3-carboxylate (2.0 g, 13 mmol) in THF (40 mL) was added dropwise MeMgBr (3 M solution in ether, 22 mL, 65 mmol) at 0 °C. The resulting mixture was stirred at 0 °C for 2 hours. The reaction mixture was slowly poured into sat. aq. NH4CI (30 mL) and extracted with DCM (30 mL x 2). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified by column chromatography (SiO2, gradient elution: 0 - 60% ethyl acetate in petroleum ether) to give the title compound (1.4 g, 9.8 mmol, 76% yield) as a white solid. MS (ESI): mass calcd. for C7H12N2O, 140.1; m/z found, 141.1 [M+H]+. 1H NMR (400MHz, CDCI3) δ = 7.27 (s, 1H), 2.10 (s, 3H), 1.58 (s, 6H).
Intermediate 5: Methyl 5-fluoro-4-(3-((S*)-1-hvdroxyethyl)-4-methyl-1H -pyrazol-1-yl)-2-(((S)-1,1,1 -trifluoropropan-2-yl)oxy)benzoate.
Figure imgf000040_0002
To a mixture of (S)-methyl 4-bromo-5-fluoro-2-((1,1,1-trifluoropropan-2-yl)oxy)benzoate (Intermediate 1, 250 mg, 720 μmol), (S*)-1-(4-methyl-1H -pyrazol-3-yl)ethanol (Intermediate 2, 101 mg, 780 μmol ) and K2CO3 (200 mg, 1.5 mmol) in dioxane (4 mL) was added CuI (69 mg, 360μmol ) and trans-(1R,2R)-N1,N2 -dimethylcyclohexane-1,2-diamine (62 mg, 430μmol ) at room temperature. The mixture was stirred at 120 °C for 32 hours under N2. The mixture was quenched by addition of H2O (10 mL) and extracted with ethyl acetate (10 mL x 3). The combined organic layers were dried, filtered and concentrated under vacuum. The crude was purified by column chromatography (SiO2, gradient elution: 0 - 20% ethyl acetate in petroleum ether) to give the title compound (90 mg, 230μmol , 32% yield) as an off-white solid. MS (ESI): mass calcd for C17H18F4N2O4, 390.1; m/z found, 391.1 [M+H]+. 1H NMR (400MHz, CDCl3) δ = 7.91 - 7.87 (m, 1H), 7.75 (d, J= 12.5 Hz, 1H), 7.68 (d, J= 6.6 Hz, 1H), 5.10 - 5.00 (m, 1H), 4.76 (spt, J= 6.2 Hz, 1H), 3.92 (s, 3H), 2.41 (d, J= 5.5 Hz, 1H), 2.18 (d, J= 0.8 Hz, 3H), 1.61 (d, J = 6.6 Hz, 3H), 1.59 (s, 3H).
Intermediate 6: Methyl 5-fluoro-4-(3-((R*)-1-hydroxyethyl)-4-methyl-1H -pyrazol-1-yl)-2-(((N)-
1,1,1 -trifluoropropan-2-yl)oxy)benzoate.
Figure imgf000041_0001
The title compound was prepared in manner analogous to Intermediate 5 except using (R *)- 1 -(4- methyl- 1H -pyrazol-3-yl)ethanol (Intermediate 3) instead of (S*)-1-(4-methyl-1H-pyrazol-3- yl)ethan-1-ol (Intermediate 2). The crude was purified by column chromatography (SiO2, gradient elution: 0 - 20% ethyl acetate in petroleum ether) to give the title compound (90 mg, 230μmol , 27% yield) as an off-white solid. MS (ESI): mass calcd for C17H18F4N2O4, 390.1; m/z found, 391.1 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 7.89 (d, J= 2.0 Hz, 1H), 7.75 (d, J= 12.5 Hz, 1H), 7.68 (d, J= 6.6 Hz, 1H), 5.10 - 5.00 (m, 1H), 4.76 (spt, J= 6.2 Hz, 1H), 3.92 (s, 3H), 2.42 (d, J= 5.5 Hz, 1H), 2.17 (d, J= 0.7 Hz, 3H), 1.61 (d, J= 6.6 Hz, 3H), 1.59 - 1.56 (m, 3H); 19F NMR (376MHz, CDCl3) δ -78.28 (3F), -131.24 (IF) ppm.
Intermediate 7: CO-Methyl 5-fluoro-4-(3-(2- -2-yl)-4-methyl-1H -pyrazol-1-yl)-2-
((1,1,1 -trifluoropropan-2-yl)oxy)benzoate.
Figure imgf000041_0002
To a mixture of fS')-methyl 4-bromo-5-fluoro-2-((1,1,1-trifluoropropan-2-yl)oxy)benzoate (Intermediate 1, 200 mg, 580μmol ), 2-(4-methyl-1H-pyrazol-3-yl)propan-2-ol (Intermediate 4, 89 mg, 640 μmol) and K2CO3 (160 mg, 1.2 mmol) in dioxane (4 mL) was added Cui (55 mg, 290 μmol) and trans-(1R,2R)-N1,N2-dimethylcyclohexane-1,2-diamine (49 mg, 350 μmol) at room temperature. The mixture was stirred at 125 °C for 16 hour under N2. The mixture was quenched by addition of H2O (10 mL) and extracted with ethyl acetate (10 mL x 3). The combined organic layers were dried, filtered and concentrated under vacuum. The crude was purified by column chromatography (SiO2, gradient elution: 0 - 20% ethyl acetate in petroleum ether) to give the title compound (55 mg, 130 μmol, 22% yield) as a white solid. MS (ESI): mass calcd for C18H20F4N2O4, 404.1 ; m/z found, 405.1 [M+H]+. 1 H NMR (400MHz, CDCI3) δ = 7.90 (d, J= 2.0 Hz, 1H), 7.74 (d, J= 12.5 Hz, 1H), 7.67 (d, J= 6.6 Hz, 1H), 4.74 (spt, J= 6.3 Hz, 1H), 3.92 (s, 3H), 3.02 (s, 1H), 2.24 (s, 3H), 1.65 (s, 6H), 1.57 (s, 3H); 19F NMR (376MHz, CDCI3) δ = -78.26 (s, IL), -131.26 (s, IL).
Example 1: A-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-((R*)-1-hvdroxyethyl)-4-methyl-1H- pyrazol- 1 -y 1 )-2-(((X)- 1,1,1 -trifl uoropropan-2-yl )oxy)benzamide.
Figure imgf000042_0001
2-Chloro-6-fluoroaniline (50 mg, 350 μmol) was dissolved in DCE (1 mL), and AlMe3 (2 M solution in toluene, 0.17 mL, 350 μmol) was added under N2. After stirring 0.5 hour, the mixture of methyl 5-fluoro-4-(3 -((R *)- 1 -hydroxy ethyl)-4-methyl- 1 H -pyrazol - 1 -yl)-2-(( (S)- 1 ,1,1- trifluoropropan-2-yl)oxy)benzoate (Intermediate 6, 45 mg, 115 μmol) in DCE (1 mL) was added. The mixture was stirred at 90 °C overnight. 1 M aq. HC1 (0.6 mL) was added and stirred at room temperature for 2 minutes. The mixture was poured into water (20 mL) and extracted with ethyl acetate (10 mL X 2). The combined organic phase was dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The crude product was purified by preparative reversed phase HPLC (Stationary phase: Boston Prime Cl 8, 5μm, 150 X 30 mm ; Mobile phase: water (0.05% NH3H2O + 10 mM NH4HCO3) (A) - MeCN (B), gradient elution: 60 - 90% B in A over 7 min, flow rate: 25 mL/min) to give the title compound (15 mg, 30 μmol, 26% yield) as a white powder. MS (ESI): mass calcd. For C22H19CIF5N3O3, 503.9; m/z found, 504.0 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 9.07 (s, 1H), 8.18 (d, J= 13.1 Hz, 1H), 7.94 (d, J= 1.9 Hz, 1H), 7.75 (d, J = 6.1 Hz, 1H), 7.32 - 7.28 (m, 1H), 7.26 -7.19 (m, 1H), 7.18 - 7.08 (m, 1H), 5.23 - 4.96 (m, 2H), 2.54 (d, J = 5.2 Hz, 1H), 2.18 (s, 3H), 1.67 (d, J= 6.6 Hz, 3H), 1.62 (d, J= 6.6 Hz, 3H) ; 19F NMR (376MHz, CDCI3) δ -78.18 (3F), -113.66 (IF), -130.60 (IF) ppm. SFC: purity 100.00%, retention time 0.972 min, method: AD_ETOH_DEA_5_40_4ML_4MIN_5CM.
Figure imgf000043_0002
Figure imgf000043_0001
Prepared in an analogous fashion to Example 1, except using methyl 5-fluoro-4-(3-((X*)- l - hydroxyethyl)-4-methyl- 1 H -pyrazol- 1 -y 1 )-2- ( ( (A) - 1 ,1,1 -trifluoropropan-2-yl)oxy)benzoate (Intermediate 5) instead of methyl 5-fluoro-4-(3-((R*)-1-hydroxyethyl)-4-methyl-17/-pyrazol-1- yl)-2-(((S)-1,,,1-trifluoropropan-2-yl)oxy)benzoate (Intermediate 6). The crude product was purified by preparative reverse phase HPLC (Stationary phase: Phenomenex Gemini NX-C18, 3μm, 75 X 30 mm ; Mobile phase: water (0.05% NH3H2O + 10 mM NH4HCO3) (A) - MeCN (B), gradient elution: 53 - 83% B in A over 6 min, flow rate: 25 mL/min) to give the title compound (15 mg, 30 μmol, 26% yield) as a white powder. MS (ESI): mass calcd. for C22H19CIF5N3O3, 503.9; m/z found, 504.0 [M+H]+. 1H NMR (400MHz, CDCI3) δ = 9.07 (s, 1H),
8.19 (d, J= 13.1 Hz, 1H), 7.95 (d, J= 1.9 Hz, 1H), 7.74 (d, J= 6.2 Hz, 1H), 7.33 - 7.28 (m, 1H), 7.26 - 7.20 (m, 1H), 7.14 (dt, J = 1.5, 8.8 Hz, 1H), 5.15 - 4.91 (m, 2H), 2.44 (d, J = 5.2 Hz, 1H),
2.19 (s, 3H), 1.68 (d, J= 6.4 Hz, 3H), 1.61 (d, J= 6.9 Hz, 3H); 19F NMR (376MHz, CDCI3) δ = - 78.18 (s, IF), -113.67 (br s, IF), -130.60 (s, IF).
Example 3 : (S)-/V-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-(2-hvdroxypropan-2-yl)-4-methyl- 1 H-pyrazol- 1 -yl)-2-(( 1 ,1,1 -trifl uoropropan-2-yl )oxy)benzamide.
Figure imgf000044_0001
Prepared in an analogous fashion to Example 1, except using (S) -methyl 5-fluoro-4-(3-(2- hydroxypropan-2-yl)-4-methyl- 1 /7-pyrazol- 1 -yl)-2-((l , 1 , 1 -trifluoropropan-2-yl)oxy)benzoate (Intermediate 7) instead of methyl 5-fluoro-4-(3-((R*)-1-hydroxyethyl)-4-methyl-17/-pyrazol-1- yl)-2-(((S)-1,1,1-trifluoropropan-2-yl)oxy)benzoate (Intermediate 6). The crude was purified by column chromatography ( SiO2, gradient elution: 0 - 40% ethyl acetate in petroleum ether) to give the crude product (70 mg, 122μmol , 62% yield) as a white solid. The white solid was purified by preparative reversed phase HPLC (Stationary phase: Boston Prime Cl 8, 5μm, 150 X
30 mm ; Mobile phase: water (0.05% NH3H2O + 10 mM NH4HCO3) (A) - MeCN (B), gradient elution: 55 - 85% B in A over 7 min, flow rate: 35 mL/min) to give the title compound (51 mg, 98μmol , 49% yield) as white solid. MS (ESI): mass calcd. for C23H21CIF5N3O3, 517.1; m/z found, 518.0 [M+H]+. 1H NMR (400MHZ, CDCl3) δ = 9.05 (br s, 1H), 8.18 (br d, J= 13.1 Hz, 1H), 7.95 (br s, 1H), 7.73 (br d, J= 6.1 Hz, 1H), 7.33 - 7.28 (m, 1H), 7.26 - 7.20 (m, 1H), 7.14 (br t, J= 8.6 Hz, 1H), 5.04 (td, J= 6.1, 12.1 Hz, 1H), 3.02 (s, 1H), 2.25 (s, 3H), 1.69 (br s, 3H), 1.66 (s, 6H); 19F NMR (376MHz, CDCl3) δ = -78.17 (s, IF), -113.68 (br s, IF), -130.64 (s, IF). SFC: purity 100.0 %, retention time 4.770 min; method: OD_3_EtOH_DEA_5_40_25ML_8MIN.
Figure imgf000044_0003
Figure imgf000044_0002
Prepared in an analogous fashion to Example 1, except using 5-chloro-3-methyl-1H-pyrazol-4- amine instead of 2-chloro-6-fluoroaniline. The crude product was purified by preparative reverse phase HPLC (Stationary phase: Boston Prime C18, 5μm, 150 X 30 mm; Mobile phase: water (0.05% NH3H2O + 10 mM NH4HCO3) (A) - MeCN (B), gradient elution: 40 - 70% B in A over 7 min, flow rate: 25 mL/min) to give the title compound (30 mg, 61μmol , 53% yield) as a white powder. MS (ESI): mass calcd. for C20H20CIF4N5O3, 489.8; m/z found, 490.0 [M+H]+. 1H NMR (400 MHz, CDCI3) d 11.28 (br s, 1H), 8.75 (s, 1H), 8.13 (d, J= 13.1 Hz, 1H), 7.93 (d, J= 2.0 Hz, 1H), 7.73 (d, J= 6.0 Hz, 1H), 5.21 -4.94 (m, 2H), 2.78 (br s, 1H), 2.29 (s, 3H), 2.18 (s, 3H), 1.68 (d, J= 6.5 Hz, 3H), 1.62 (d, J= 6.5 Hz, 3H) ; 19F NMR (376 MHz, CDCI3) δ -78.22 (3F), - 130.61 (IF) ppm. SFC: purity 100.00%, retention time 1.999 min, method: AS_ETOH_DEA_5_40_28ML_8MIN.
Example 5: N-(5-Chloro-3-methyl- 1H-pyrazol-4-yl)-5-fluoro-4-(3-((S*)-l -hydroxy ethyl)-4- methyl- 1 H-pyrazol- 1 -yl)-2-(((M- 1.1.1 -trifluoropropan-2-yl )oxyibenzamide.
Figure imgf000045_0001
Prepared in an analogous fashion to Example 1, except using methyl 5-fluoro-4-(3-((S*)- l - hydroxy ethyl)-4-methyl- 1 H -pyrazol- 1 -yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzoate (Intermediate 5) instead of methyl 5-fluoro-4-(3-((R*)-1-hydroxyethyl)-4-methyl-1H -pyrazol-1- yl)-2-(((S)- 1,1,1-trifluoropropan-2-yl)oxy)benzoate (Intermediate 6) and 5-chloro-3-methyl-1H- pyrazol-4-amine instead of 2-chloro-6-fluoroaniline. The crude product was purified by preparative reverse phase HPLC (Stationary phase: Phenomenex Gemini NX-C18, 3μm, 75 x 30 mm ; Mobile phase: water (0.05% NH3H2O + 10 mM NH4HCO3) (A) - MeCN (B), gradient elution: 36 - 66% B in A over 6 min, flow rate: 25 mL/min) to give the title compound (30 mg,
61μmol , 53% yield) as a white powder. MS (ESI): mass calcd. For C20H20CIF4N5O3, 489.8; m/z found, 490.2 [M+H]+. 1H NMR (400MHz, CDCI3) δ = 10.19 (br s, 1H), 8.75 (s, 1H), 8.15 (d, J = 13.1 Hz, 1H), 7.94 (d, J= 1.8 Hz, 1H), 7.73 (d, J= 6.2 Hz, 1H), 5.19 - 4.97 (m, 2H), 2.48 (br d, J = 5.1 Hz, 1H), 2.31 (s, 3H), 2.19 (s, 3H), 1.70 (d, J= 6.4 Hz, 3H), 1.62 (s, 3H); 19F NMR (376MHz, CDCl3) δ = -78.20 (s, IF), -130.60 (s, IF).
Example 6: (S)-N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-(2-hvdroxypropan-2-yl)- 4-methyl- 1 H-pyrazol- 1 -yl)-2-(( 1 , 1 , 1 -trifluoropropan-2-yl)oxy)benzamide.
Figure imgf000046_0001
Prepared in an analogous fashion to Example 1, except using (S)-methyl 5-fluoro-4-(3-(2- hydroxypropan-2-yl)-4-methyl- 1 /7-pyrazol- 1 -yl)-2-((l , 1 , 1 -trifluoropropan-2-yl)oxy)benzoate (Intermediate 7) instead of methyl 5-fluoro-4-(3-((RR)-l -hydroxyethyl)-4-methyl-1H -pyrazol-1- yl)-2-(((S)-1,1,1-trifluoropropan-2-yl)oxy)benzoate (Intermediate 6) and 5-chloro-3-methyl-1H- pyrazol-4-amine instead of 2-chloro-6-fluoroaniline. The crude was purified by column chromatography (SiO2, gradient elution: 0 - 40% ethyl acetate in petroleum ether) to give the crude compound (100 mg, 177μmol , 81% yield) as a white solid. The white solid was purified by preparative reverse phase HPLC (Stationary phase: Boston Prime Cl1, 5μm, 150 X 30 mm ; Mobile phase: water (0.05% NH3H2O + 10 mM NH4HCO3) (A) - MeCN (B), gradient elution: 33 - 67% B in A over 7 min, flow rate: 35 mL/min) to give the title compound (60 mg, 123 μmol, 57% yield) as a white solid. MS (ESI): mass calcd. for C21H22CIF4N5O3, 503.1; m/z found, 503.9 [M+H]+. 1H NMR (400MHz, CDCl3) δ = 9.86 (br s, 1H), 8.75 (s, 1H), 8.15 (d, J= 13.1 Hz, 1H), 7.95 (d, J= 2.0 Hz, 1H), 7.72 (d, J= 6.0 Hz, 1H), 5.09 - 4.95 (m, 1H), 3.04 (br s, 1H), 2.32 (s, 3H), 2.25 (s, 3H), 1.70 (d, J= 6.5 Hz, 3H), 1.66 (s, 6H); 19F NMR (376MHz, CDCl3) δ = -78.18 (s, IF), -130.64 (s, IF). SFC: purity 100%, retention time 3.50 min; method: OD_ETOH_DEA_5_40_28ML_8MIN.
Figure imgf000046_0002
Figure imgf000047_0001
Figure imgf000047_0002
Figure imgf000047_0003
A mixture of (A)-methyl 4-bromo-5-fluoro-2-((1,1,1-trifluoropropan-2- yl)oxy)benzoate (Intermediate 1, 300 mg, 870 μmol), diphenylmethanimine (189 mg, 1.0 mmol), and CS2CO3 (425 mg, 1.3 mmol) in toluene (10 mL) was purged with N2. Then Xantphos (100 mg, 174μmol ) and Pd2(dba)3 (80 mg, 87μmol ) were added into the mixture reaction. The reaction mixture was purged with N2 and heated at 110 °C overnight. The mixture was diluted H2O (20 mL) and extracted with ethyl acetate (20 mL x 3). The organic layer was dried over Na2SO4, filtered and evaporated under vacuum. The residue was purified by column chromatography (SiO2, gradient elution: 0-50% ethyl acetate in petroleum ether) to give the title compound (363 mg, 665μmol , 77% yield) as a light yellow oil. MS (ESI): mass cal cd for C24H19F4NO3, 445.13; m/z found, 446.1 [M+H]+. 1H NMR (400 MHz, CDCI3) δ 7.83 - 7.73 (m, 2H), 7.69 - 7.60 (m, 1H), 7.52 (br d, J= 6.8 Hz, 1H), 7.50 - 7.34 (m, 5H), 7.19 - 7.08 (m, 2H), 6.43 (d, J= 6.6 Hz, 1H), 4.40 (spt, J= 6.2 Hz, 1H), 3.84 (s, 3H), 1.38 (d, J= 6.4 Hz, 3H); 19F NMR (376 MHz, CDCI3) δ -74.90 - -81.31 (3 F), -130.97 (IF) ppm.
Figure imgf000047_0004
Figure imgf000047_0005
To a solution of 5-chloro-3-methyl-1H-pyrazol-4- amine (210 mg, 1.6 mmol) in DCE (4 mL) was added AlMe3 (2M in toluene, 0.93 mL, 1.9 mmol) dropwise under N2 at 0 °C. After 30 minutes, (S)-methyl 4-((diphenylmethylene)amino)- 5 -fluor o-2- ((1,1,1- trifluoropropan-2-yl)oxy)benzoate (290 mg, 531μmol ) in DCE (3 mL) was added. The mixture was stirred at 90 °C for 16 hours. The reaction mixture was diluted with DCM (20 mL), and quenched by H2O (10 mL), then extracted with DCM (30 mL x 2). The organic layers were dried over anhydrous Na2SO4, concentrated under vacuum. The residue was purified by column chromatography (SiO2, gradient elution: 0 - 80 % petroleum ether in ethyl acetate) to give the title compound (410 mg, 616μmol , 95% yield) as yellow solid. MS (ESI): mass calcd for C27H21CIF4N4O2, 544.13; m/z found, 567.0 [M+Na]+. 1H NMR (400 MHz, CDCI3) δ 7.77 (d, J= 7.39 Hz, 2 H), 7.54 - 7.59 (m, 1 H), 7.45 - 7.51 (m, 3 H), 7.38 (dd, J = 5.01, 1.91 Hz, 3 H), 7.23 (br d, J= 3.46 Hz, 2 H), 6.75 (d, J= 6.32 Hz, 1 H), 5.02 (dt, J= 12.52, 6.38 Hz, 1 H), 2.20 (s, 3 H), 1.40 (d, J= 6.32 Hz, 3 H); 19F NMR (376 MHz, CDCl3) δ -79.87 (3F) -133.86 (IF) ppm.
Figure imgf000048_0001
HC1 (4 M solution in MeOH, 33 mL) was added to (S)-7V-(5-chloro-3-
Figure imgf000048_0002
methyl-1H-pyrazol -4-yl)-4-((diphenylmethylene)amino)-5-fluoro-2-((l , 1 , 1 -trifluoropropan-2- yl)oxy)benzamide (400 mg, 601 μmol). The mixture was stirred at r.t. overnight. The reaction mixture was concentrated under vacuum. The residue was diluted with ethyl acetate (30 mL) and alkalized with sat. aq. NaHCO3 (10 mL), then extracted with ethyl acetate (20 mL). The organic layers were dried over anhydrous Na2SO4and concentrated under vacuum. The crude product was purified by column chromatography (SiO2, gradient elution: 0 - 80 % petroleum ether in ethyl acetate) to give the title product (200 mg, 525 μmo, l 72% yield) as a yellow solid. MS (ESI): mass calcd for C14H13CIF4N4O2, 380.07; m/z found, 381.0 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 7.59 (d, J= 12.05 Hz, 1 H), 6.62 (d, J= 7.03 Hz, 1 H), 5.14 (quin, J= 6.34 Hz, 1 H), 2.21 (s, 3 H), 1.59 (d, J= 6.27 Hz, 3 H); 19F NMR (376 MHz, CDCl3) δ -79.97 (3F), -144.73 (1F) ppm.
Figure imgf000048_0003
A solution of (S)-4-amino-N -(5-
Figure imgf000048_0004
chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-2-((l, 1,1 -trifluoro propan-2-yl)oxy)benzamide (200 mg, 525 μmol) in AcOH (1.2 mL), H2O (2 mL) and cone. HC1 (219 μL, 2.6 mmol) was cooled to
<5 °C and a solution of NaNO2 (45 mg, 646 μmo)l in H2O (2 mL) was added slowly. The mixture was slowly added to a mixture of ethyl 3-oxobutanoate (67 uL, 2.6 mmol) and AcONa (216 mg, 2.6 mmol) in Na2CO3 (1M, 2 mL, 2.6 mmol) and EtOH (2 mL) at 0 °C. The mixture was stirred for 2 hours. The reaction mixture was diluted with ethyl acetate (20 mL), then washed with H2O (10 mL), and extracted with ethyl acetate (20 mL). The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum pressure to give the title product (380 mg, crude) as a brown gum. MS (ESI): mass calcd for C20H20CIF4N5O5, 521.11; m/z found, 522.0 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 7.78 - 7.69 (m, 2H), 7.58 (d, J= 6.2 Hz, 1H), 5.38 - 5.31 (m, 1H), 4.44 - 4.36 (m, 2H), 2.61 - 2.54 (m, 3H), 2.25 - 2.23 (m, 3H), 1.65 - 1.60 (m, 3H), 1.44 - 1.36 (m, 3H); 19F NMR (376 MHz, CDCl3) δ -79.37 - -80.00 (3F), -142.42 (IF) ppm.
Figure imgf000049_0001
Figure imgf000049_0002
To a solution of (S)- ethyl 2-(2-(4-((5-chloro-3 -methyl- 1H-pyrazol-4-yl)carbamoyl)-2-fluoro -5-((l , 1 , 1 - trifluoropropan-2-yl)oxy)phenyl)hydrazono)-3-oxobutanoate (380 mg, crude) in EtOH (10 mL) was added CuCh ( 215 mg, 1.6 mmol) and AcONEh (561 mg, 7.3 mmol). The mixture was refluxed for 16 hours. The reaction mixture was filtered and concentrated under vacuum. The residue was purified by column chromatography (SiO2, gradient elution: 0 - 80 % petroleum ether in ethyl acetate) to give the title compound (230 mg, 434 μmol, 84% yield over two steps) as a yellow solid. MS (ESI): mass calcd for C20H19CIF4N6O4, 518.11; m/z found, 519.0 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 7.85 (d, J= 5.8 Hz, 1H), 7.77 (d, J= 10.7 Hz, 1H), 5.31 (spt, J = 6.2 Hz, 1H), 4.45 (q, J= 7.1 Hz, 2H), 2.62 (s, 3H), 2.27 (s, 3H), 1.61 (d, J= 6.3 Hz, 3H), 1.43 (t, J= 7.2 Hz, 3H); 19F NMR (376 MHz, CDCl3) δ -79.75 (3F), -130.08 (IF) ppm.
Figure imgf000049_0003
Figure imgf000049_0004
A solution of (S)- ethyl 2-(4-((5-chloro-3-methyl-1H-pyrazol-4-yl)carbamoyl)-2-fluoro-5- (( 1,1,1 -trifluoropropan- 2-yl)oxy)phenyl)-5-methyl-2H-1,2,3-triazole-4-carboxylate (230 mg, 434 μmol) in THF (10 mL) was cooled to 0 °C, then MeMgBr (3 M in Et2O, 1.5 mL, 4.3 mmol) was added dropwise. The resulting mixture was stirred at 0 °C for 2 hours. The reaction mixture was quenched with sat. aq. NH4CI (5 mL), diluted with ethyl acetate (20 mL), then washed with H2O (10 mL), and extracted with ethyl acetate (20 mL). The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by preparative high- performance liquid chromatography: (Stationary phase: Boston Prime C18, 5μm, 150 X 30 mm ; Mobile phase: water (0.05% NH3H2O + 10 mM NH4HCO3) (A) - MeCN (B), gradient elution: 40 - 70% B in A over 7 min, flow rate: 25 mL/min) to give the title compound (106 mg, 210 μmol, 47% yield) as a white solid. MS (ESI): mass calcd. for C20H21CIF4N6O3, 504.13; m/z found, 505.1 [M+H]+. 1H NMR (400 MHz, CDCl3) δ 7.76 (s, 1H), 7.74 (d, J= 4.6 Hz, 1H), 5.28 (td, J= 6.1, 12.4 Hz, 1H), 2.53 (s, 3H), 2.26 (s, 3H), 1.65 (s, 6H), 1.61 (d, J= 6.4 Hz, 3H); 19F NMR (376 MHz, CDCl3) δ -79.73 (3F), -130.34 (IF) ppm. SFC: purity 100%, retention time 1.381 mm; method: OD_ETOH_DEA_5_40_4ML_4MIN_5CM.
Biological Data DHODH inhibitory activities of the compounds of Examples 1-7 were assessed using the following assays. The half maximal inhibitory concentration values (IC50) are summarized in Table 3.
Biological Assays
In vitro Assay: DHODH enzymatic assay
To detect DHODH enzyme activities, dichloroindophenol (DCIP) is added as the final electron acceptor in the assay. DCIP can accept electrons from the reduced coenzyme Q generated in the assay, or from dihydroorotate (DHO) via FMN by binding presumably to the ubiquinone pocket. DCIP solutions are blue, with an intense absorbance around 600 nm, but becomes colorless upon reduction (J. Biol. Chem. (1986) 261, 11386). The assay buffer contained 50 nM HEPES, pH 7.5, 150 mM NaCl, 0.5 mM EDTA, and 0.1% Triton X-100 in MilliQ water. Substrate consisting of 20 mM DHO, 5mM CoQ6, and ImM DCIP in assay buffer, initiates the reaction. The assay is run in end-point mode by quenching the reaction with the potent DHODH inhibitor brequinar. Absorbance measurements were obtained using the BMG Phera Star plate-reading spectrophotomer. Purified human DHODH was purchased from Proteros (cat. No. PR-0044). Chemicals were purchased from Sigma- Aldrich, Teknova, and Avanti Polar Lipids. Liquid handling was performed using Labcyte Echo and Formulatrix Tempest.
In vitro Assay: MOLM-13 Cellular Assay
MOLM-13 cells (human acute myeloid leukemia cells) were obtained from DSMZ and were maintained in RPMI 1640 + Glutamax + 25mM HEPES (Invitrogen, catalog number 72400) supplemented with 10% heat inactivated fetal bovine serum (FBS; Invitrogen, catalog number 16140). The day prior to assay set-up, cells were pelleted, resuspended in fresh media, counted, and cells were plated at 0.4 x 106 cell/mL in a T150 flask. On the day of the assay, cells were pelleted, resuspend in fresh media, counted and seeded at 5,000 cells/well in white opaque 96- well tissue culture treated microplates (Perkin Elmer, catalog number 6005680). Cells were exposed to different concentrations of test compounds at 37 °C, 5% CO2 for 72 hours immediately after seeding. Cell viability was acquired on a Perkin Elmer Envision 2104 multilabel reader using the CellTiter-Glo assay (Promega) according to the manufacturer’s instructions.
Table 3,
Figure imgf000051_0001
NT means Not Tested

Claims

What is claimed is:
1. A compound having the structure of Formula (I): wherein
Figure imgf000052_0001
R1 is C1-6alkyl or C1-6haloalkyl;
Figure imgf000052_0002
wherein
Ra is C1-6alkyl; Rb is C1-6alkyl substituted with one OH member;
R3 is H or halo; R4 is selected from the group consisting of:
Figure imgf000052_0003
Figure imgf000052_0004
wherein
Re is selected from the group consisting of: H; halo; C1-6alkyl; C1-6alkyl substituted with one member selected from the group consisting of: OH, OCH3, and OCF3;
C1-6haloalkyl; C1-6haloalkyl substituted with one member selected from the group consisting of: OH, and OCH3; and OC1-6alkyl;
Rf is selected from the group consisting of: H; halo; C1-6alkyl; C1-6alkyl substituted with one member selected from the group consisting of: OH, OCH3, and OCF3; C1-6haloalkyl; C1-6haloalkyl substituted with one member selected from the group consisting of: OH, and OCH3; and OC1-6alkyl; and Rgis H or C1-6alkyl; or a pharmaceutically acceptable salt, isotope, N-oxide, solvate, or stereoisomer thereof.
2. The compound according to claim 1, wherein R1 is Ci-4alkyl; or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof.
3. The compound according to claim 1, wherein R1 is or a pharmaceutically
Figure imgf000053_0001
acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof.
4. The compound according to any of claims 1-3, wherein R2 is wherein
Figure imgf000053_0002
Ra is CH3; and Rb is CH(OH)CH3, or C(OH)(CH3)2; or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof.
The compound according to any of claims 1-3, wherein R2 is
Figure imgf000053_0003
Figure imgf000053_0004
or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof.
6. The compound according to any of claims 1-3, wherein R2 is
Figure imgf000053_0005
or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof.
7. The compound according to any of claims 1-6, wherein R3 is F. The compound according to any of claims 1-7, wherein R4 is , where Re and Rf
Figure imgf000054_0001
are halo; or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N- oxide thereof. The compound according to any of claims 1-7, wherein R4 is
Figure imgf000054_0002
; or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof.
Figure imgf000054_0003
Re is Ci-4alkyl, and Rf is halo; or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof. The compound according to any of claims 1-7, wherein R4 is
Figure imgf000054_0004
; or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof. A compound selected from the group consisting of:
N-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-((R*)-l -hydroxy ethyl)-4-methyl-1H-pyrazol- 1- yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
N-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-((S*)-1-hydroxyethyl)-4-methyl-1H-pyrazo1-1- yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
(S)-N-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-(2-hydroxypropan-2-yl)-4-methyl-1H- pyrazol-1 -y l)-2- (( 1 , 1 , 1 -trifluoropropan-2-yl)oxy)benzamide;
N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-((R*)-l -hydroxy ethyl)-4-methyl-1H- pyrazol- 1 -yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-((S*)-1-hydroxyethyl)-4-methyl-1H- pyrazol- 1 -yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
(S)-N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-(2-hydroxypropan-2-yl)-4- methyl- 1 H-pyrazol- 1 -yl)-2-(( 1,1,1 -trifluoropropan-2-yl)oxy)benzamide; and (S)-N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(4-(2-hydroxypropan-2-yl)-5- methyl-2H- 1 ,2,3 -triazol-2-yl)-2-(( 1 ,1,1 -trifluoropropan-2-yl)oxy)benzamide; or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof. A pharmaceutical composition comprising: (A) a compound according to any of claims 1-12, or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof; and (B) at least one pharmaceutically acceptable excipient. A pharmaceutical composition comprising an effective amount of: N-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-((R*)-l -hydroxy ethyl)-4-methyl-1H-pyrazol- 1- yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
N-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-((S*)-1-hydroxyethyl)-4-methyl-1H-pyrazol-1- yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
(S)-N-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-(2-hydroxypropan-2-yl)-4-methyl-1H- pyrazol-1 -y l)-2- (( 1 , 1 , 1 -trifluoropropan-2-yl)oxy)benzamide;
N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-((R*)-l -hydroxy ethyl)-4-methyl-1H- pyrazol- 1 -yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-((S*)-1-hydroxyethyl)-4-methyl-1H- pyrazol- 1 -yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
(S)-N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-(2-hydroxypropan-2-yl)-4- methyl- 1 H-pyrazol- 1 -yl)-2-(( 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
(S)-N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(4-(2-hydroxypropan-2-yl)-5- methyl-2H- 1 ,2,3 -triazol-2-yl)-2-(( 1 ,1,1 -trifluoropropan-2-yl)oxy)benzamide; or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof; and at least one pharmaceutically acceptable excipient. A method of treating a subject suffering from or diagnosed with a disease, disorder, or medical condition comprising inhibiting or altering dihydroorotate oxygenase enzyme activity in the subject by administering to the subject an effective amount of at least one compound according to any of claims 1-12, or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof. The method according to claim 15, wherein the disorder, disease or medical condition is selected from the group consisting of: inflammatory disorders and autoimmune disorders. The method according to claim 15, wherein the disorder, disease or medical condition is cancer. The method according to claim 15, wherein the disorder, disease or medical condition is selected from the group consisting of: lymphomas, leukemias, carcinomas, and sarcomas. The method according to claim 15, wherein the disorder, disease or medical condition is selected from the group consisting of: acute lymphoblastic leukemia, acute myeloid leukemia, (acute) T-cell leukemia, acute lymphoblastic leukemia, acute lymphocytic leukemia, acute monocytic leukemia, acute promyelocytic leukemia, bisphenotypic B myelomonocytic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, chronic myeloid leukemia, chronic myelomonocytic leukemia, large granular lymphocytic leukemia, plasma cell leukemia, and also myelodysplastic syndrome, which can develop into an acute myeloid leukemia. The method according to claim 15, wherein the disorder, disease or medical condition is acute myeloid leukemia. The method according to any of claims 15-20, wherein the at least one compound is: N-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-((R*)-l -hydroxy ethyl)-4-methyl-1H-pyrazol- 1- yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
N-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-((S*)-1-hydroxyethyl)-4-methyl-1H-pyrazol-1- yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
(S)-N-(2-Chloro-6-fluorophenyl)-5-fluoro-4-(3-(2-hydroxypropan-2-yl)-4-methyl-1H- pyrazol-1 -y l)-2- (( 1 , 1 , 1 -trifluoropropan-2-yl)oxy)benzamide;
N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-((R*)-l -hydroxy ethyl)-4-methyl-1H- pyrazol- 1 -yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-((S*)-1-hydroxyethyl)-4-methyl-1H- pyrazol- 1 -yl)-2-(((S)- 1,1,1 -trifluoropropan-2-yl)oxy)benzamide;
(S)-N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(3-(2-hydroxypropan-2-yl)-4- methyl- 1 H-pyrazol- 1 -yl)-2-(( 1,1,1 -trifluoropropan-2-yl)oxy)benzamide; and
(S)-N-(5-Chloro-3-methyl-1H-pyrazol-4-yl)-5-fluoro-4-(4-(2-hydroxypropan-2-yl)-5- methyl-2H- 1 ,2,3 -triazol-2-yl)-2-(( 1 ,1,1 -trifluoropropan-2-yl)oxy)benzamide; or a pharmaceutically acceptable salt, solvate, stereoisomer, isotopic variant, or N-oxide thereof.
PCT/IB2021/058906 2020-09-30 2021-09-29 Dihydroorotate dehydrogenase inhibitors WO2022070069A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063085314P 2020-09-30 2020-09-30
US63/085,314 2020-09-30

Publications (1)

Publication Number Publication Date
WO2022070069A1 true WO2022070069A1 (en) 2022-04-07

Family

ID=78080405

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2021/058906 WO2022070069A1 (en) 2020-09-30 2021-09-29 Dihydroorotate dehydrogenase inhibitors

Country Status (1)

Country Link
WO (1) WO2022070069A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018077944A2 (en) * 2016-10-27 2018-05-03 Bayer Aktiengesellschaft 4,5-annulated 1,2,4-triazolones
WO2018077923A1 (en) * 2016-10-27 2018-05-03 Bayer Aktiengesellschaft 2,4,5-trisubstituted 1,2,4-triazolones useful as inhibitors of dhodh
WO2020144638A1 (en) * 2019-01-11 2020-07-16 Janssen Biotech, Inc. Dihydroorotate dehydrogenase inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018077944A2 (en) * 2016-10-27 2018-05-03 Bayer Aktiengesellschaft 4,5-annulated 1,2,4-triazolones
WO2018077923A1 (en) * 2016-10-27 2018-05-03 Bayer Aktiengesellschaft 2,4,5-trisubstituted 1,2,4-triazolones useful as inhibitors of dhodh
WO2020144638A1 (en) * 2019-01-11 2020-07-16 Janssen Biotech, Inc. Dihydroorotate dehydrogenase inhibitors

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
"Handbook of Pharmaceutical Salts, Properties, Selection, and Use", 2002, WILEY-VCH
CAO L ET AL., MOL CANCER THER
CELL CHEMICAL BIOLOGY, vol. 25, 2018, pages 1
HÉLÈNE MUNIER-LEHMANN ET AL: "On Dihydroorotate Dehydrogenases and Their Inhibitors and Uses", JOURNAL OF MEDICINAL CHEMISTRY, vol. 56, no. 8, 25 April 2013 (2013-04-25), pages 3148 - 3167, XP055148746, ISSN: 0022-2623, DOI: 10.1021/jm301848w *
J. BIOL. CHEM., vol. 261, 1986, pages 11386
LOLLI ML ET AL., RECENT PATENTS ON ANTI-CANCER DRUG DISCOVERY, vol. 13, 2018, pages 86
MATHUR D ET AL., CANCER DISCOVERY, vol. 7, 2017, pages 1
MCCULLOCH D ET AL., ONCO TARGETS THER, vol. 10, 2017, pages 1585 - 1601
NOWAK D ET AL., BLOOD, vol. 113, 2009, pages 3655
PHARMACOLOGY AND THERAPEUTICS, 19 October 2018 (2018-10-19)
REIS RAG ET AL., ARCHIVES BIOCHEM BIOPHYSICS, vol. 632, 2017, pages 175
S.M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: 10.1002/jps.2600660104
SAINAS S ET AL., J MED CHEM, vol. 61, 2018, pages 6034
STEIN ET ET AL., HEALTH QUAL LIFE OUTCOMES, vol. 16, 2018, pages 193
SYKES ET AL., CELL, vol. 167, 2016, pages 171
VYAS VK ET AL., MINI REV MED CHEM, vol. 11, 2011, pages 1039
WU D ET AL., HAEMATOLOGICA, vol. 103, 2018, pages 1472

Similar Documents

Publication Publication Date Title
US11753393B2 (en) Dihydroorotate dehydrogenase inhibitors
US10150766B2 (en) P2X7 modulators
US20220081422A1 (en) Dihydroorotate dehydrogenase inhibitors
EP3908579B1 (en) Dihydroorotate dehydrogenase inhibitors
EP4021886A1 (en) Substituted urea dihydroorotate dehydrogenase inhibitors
WO2021084498A1 (en) Fluorinated quinoline, quinoxaline and benzo[b][1,4]oxazine derivatives as dihydroorotate dehydrogenase (dhodh) inhibitors for the treatment of cancer, autoimmune and inflammatory diseases
US20230037448A1 (en) Fluorinated quinoline and quinoxaline derivatives as dihydroorotate dehydrogenase (dhodh) inhibitors for the treatment of cancer, autoimmune and inflammatory diseases
WO2021156787A1 (en) Heterocyclic compounds as dihydroorotate dehydrogenase inhibitors
WO2021240429A1 (en) Benzofuran and benzopyran dihydroorotate dehydrogenase inhibitors
WO2021240424A1 (en) Indazole and benzoisoxazole dihydroorotate dehydrogenase inhibitors
WO2021240423A1 (en) Dihydroorotate dehydrogenase inhibitors
WO2022070069A1 (en) Dihydroorotate dehydrogenase inhibitors
AU2020363021A1 (en) Bi-aryl dihydroorotate dehydrogenase inhibitors
WO2022070071A1 (en) Dihydroorotate dehydrogenase inhibitors
WO2022070068A1 (en) Dihydroorotate dehydrogenase inhibitors
WO2022074534A1 (en) Combinations of dihydroorotate dehydrogenase inhibitors and hypomethylating agents

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: 21787062

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: 21787062

Country of ref document: EP

Kind code of ref document: A1