WO2022082174A1 - Heterocyclic compounds and uses thereof - Google Patents

Heterocyclic compounds and uses thereof Download PDF

Info

Publication number
WO2022082174A1
WO2022082174A1 PCT/US2021/071827 US2021071827W WO2022082174A1 WO 2022082174 A1 WO2022082174 A1 WO 2022082174A1 US 2021071827 W US2021071827 W US 2021071827W WO 2022082174 A1 WO2022082174 A1 WO 2022082174A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
tautomer
stereoisomer
Prior art date
Application number
PCT/US2021/071827
Other languages
French (fr)
Inventor
Son Minh Pham
Jayakanth Kankanala
Sarvajit Chakravarty
Brahmam PUJALA
Varun Kumar
Original Assignee
Nuvation Bio Operating Company 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 Nuvation Bio Operating Company Inc. filed Critical Nuvation Bio Operating Company Inc.
Publication of WO2022082174A1 publication Critical patent/WO2022082174A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • This disclosure relates generally to therapeutics engaged in inhibition of the DNA damage checkpoint kinase, Weel, which potentiates genotoxic chemotherapies by abrogating cell-cycle arrest and proper DNA repair.
  • the disclosure also provides pharmaceutically acceptable compositions comprising compounds of the present disclosure and methods of using said compositions in the treatment of diseases associated with this pathway.
  • Weel is a tyrosine kinase that phosphorylates and inactivates Cdc2 and is involved in G checkpoint signaling. More particularly, Weel is involved in G2-M checkpoint signaling. Because p53 is a key regulator in the G checkpoint, p53-deficient tumors rely only on the G checkpoint after DNA damage. More particularly, because p53 is a key regulator in the Gi-S checkpoint, p53 -deficient tumors rely only on the G2-M checkpoint after DNA damage. Hence, such tumors are selectively sensitized to DNA-damaging agents by Weel inhibition.
  • Weel belongs to a family of protein kinases involved in the terminal phosphorylation and inactivation of cyclin-dependent kinase 1 -bound cyclin B, resulting in G cell cycle arrest in response to DNA damage.
  • Weel was first identified in fission yeast, where Weel deficiency resulted in premature mitotic entry and replication of smaller-sized yeast. It is the major kinase responsible for the inhibitory phosphorylation of the tyrosine.
  • AZD-1775 (1, 2-allyl-l-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((4-(4- methylpiperazin-l-yl)phenyl)amino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one).
  • AZD-1775 exhibited antitumor activity in various preclinical studies as a monotherapy or in potentiating chemo- and radiotherapy, and is currently in phase I/II clinical trials.
  • Weel is highly expressed in several cancer types, including hepatocellular carcinoma, breast cancers, cervical cancers, lung cancers, squamous cell carcinoma, diffuse intrinsic pontine glioma (DIPG), glioblastoma, medulloblastoma, leukemia, melanoma, and ovarian cancers.
  • DIPG diffuse intrinsic pontine glioma
  • glioblastoma medulloblastoma
  • leukemia melanoma
  • ovarian cancers ovarian cancers.
  • a method of treating cancer in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing. Also provided is a method of inhibiting Weel in a cell, comprising administering a compound detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, to the cell.
  • kits comprising a compound detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, are also provided.
  • a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is also provided for the manufacture of a medicament for the treatment of cancer.
  • Alkyl refers to and includes saturated linear and branched univalent hydrocarbon structures and combination thereof, having the number of carbon atoms designated (z.e., Ci-Cio means one to ten carbons). Particular alkyl groups are those having 1 to 20 carbon atoms (a “C1-C20 alkyl”).
  • alkyl groups are those having 1 to 8 carbon atoms (a “Ci-Cs alkyl”), 3 to 8 carbon atoms (a “C3-C8 alkyl”), 1 to 6 carbon atoms (a “Ci-Ce alkyl”), 1 to 5 carbon atoms (a “C1-C5 alkyl”), or 1 to 4 carbon atoms (a “C1-C4 alkyl”).
  • alkyl examples include, but are not limited to, groups such as methyl, ethyl, n- propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec -butyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • the alkenyl group may be in “cis” or “trans” configurations, or alternatively in “E” or “Z” configurations.
  • Particular alkenyl groups are those having 2 to 20 carbon atoms (a “C2-C20 alkenyl”), having 2 to 8 carbon atoms (a “C2-C8 alkenyl”), having 2 to 6 carbon atoms (a “C2-C6 alkenyl”), or having 2 to 4 carbon atoms (a “C2-C4 alkenyl”).
  • alkenyl examples include, but are not limited to, groups such as ethenyl (or vinyl), prop-l-enyl, prop-2-enyl (or allyl), 2-methylprop-l-enyl, but-l-enyl, but-2-enyl, but-3-enyl, buta- 1,3 -dienyl, 2-methylbuta- 1,3-dienyl, homologs and isomers thereof, and the like.
  • Alkylene refers to the same residues as alkyl, but having bivalency. Particular alkylene groups are those having 1 to 6 carbon atoms (a “Ci-Ce alkylene”), 1 to 5 carbon atoms (a “C1-C5 alkylene”), 1 to 4 carbon atoms (a “C1-C4 alkylene”) or 1 to 3 carbon atoms (a “C1-C3 alkylene”). Examples of alkylene include, but are not limited to, groups such as methylene (-CH2-), ethylene (-CH2CH2-), propylene (-CH2CH2CH2-), butylene (-CH2CH2CH2CH2-), and the like.
  • Particular alkynyl groups are those having 2 to 20 carbon atoms (a “C2-C20 alkynyl”), having 2 to 8 carbon atoms (a “C2-C8 alkynyl”), having 2 to 6 carbon atoms (a “C2-C6 alkynyl”), or having 2 to 4 carbon atoms (a “C2-C4 alkynyl”).
  • alkynyl examples include, but are not limited to, groups such as ethynyl (or acetylenyl), prop-l-ynyl, prop-2-ynyl (or propargyl), but-l-ynyl, but-2-ynyl, but-3-ynyl, homologs and isomers thereof, and the like.
  • Aryl refers to and includes polyunsaturated aromatic hydrocarbon groups.
  • Aryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings.
  • the aryl group contains from 6 to 14 annular carbon atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, biphenyl, and the like.
  • Cycloalkyl refers to and includes cyclic univalent hydrocarbon structures, which may be fully saturated, mono- or polyunsaturated, but which are non-aromatic, having the number of carbon atoms designated (e.g., C1-C10 means one to ten carbons). Cycloalkyl can consist of one ring, such as cyclohexyl, or multiple rings, such as adamantyl, but excludes aryl groups. A cycloalkyl comprising more than one ring may be fused, spiro or bridged, or combinations thereof. A preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 13 annular carbon atoms.
  • a more preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a "C3-C8 cycloalkyl").
  • cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1 -cyclohexenyl, 3 -cyclohexenyl, cycloheptyl, norbornyl, and the like.
  • Halo or “halogen” refers to elements of the Group 17 series having atomic number 9 to 85.
  • Preferred halo groups include fluoro, chloro, bromo and iodo. Where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached, e.g., dihaloaryl, dihaloalkyl, trihaloaryl etc. refer to aryl and alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be but are not necessarily the same halo; thus 4-chloro-3-fluorophenyl is within the scope of dihaloaryl.
  • perhaloalkyl An alkyl group in which each hydrogen is replaced with a halo group is referred to as a “perhaloalkyl.”
  • a preferred perhaloalkyl group is trifluoroalkyl (-CF3).
  • perhaloalkoxy refers to an alkoxy group in which a halogen takes the place of each H in the hydrocarbon making up the alkyl moiety of the alkoxy group.
  • An example of a perhaloalkoxy group is trifluoromethoxy (-OCF3).
  • Hetero aryl refers to and includes unsaturated aromatic cyclic groups having from 1 to 10 annular carbon atoms and at least one annular heteroatom, including but not limited to heteroatoms such as nitrogen, oxygen and sulfur, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quatemized.
  • a heteroaryl group can be attached to the remainder of the molecule at an annular carbon or at an annular heteroatom.
  • Heteroaryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings. Examples of heteroaryl groups include, but are not limited to, pyridyl, pyrimidyl, thiophenyl, furanyl, thiazolyl, and the like.
  • Heterocycle or “heterocyclyl” refers to a saturated or an unsaturated nonaromatic group having from 1 to 10 annular carbon atoms and from 1 to 4 annular heteroatoms, such as nitrogen, sulfur or oxygen, and the like, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quatemized.
  • a heterocyclyl group may have a single ring or multiple condensed rings, but excludes heteroaryl groups.
  • a heterocycle comprising more than one ring may be fused, spiro or bridged, or any combination thereof. In fused ring s'ystems, one or more of the fused rings can be aryl or heteroaryl.
  • heterocyclyl groups include, but are not limited to, tetrahydropyranyl, dihydropyranyl, piperidinyl, piperazinyl, pyrrolidinyl, thiazolinyl, thiazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, 2,3-dihydrobenzo[b]thiophen-2-yl, 4- amino-2-oxopyrimidin-l(2H)-yl, and the like.
  • Optionally substituted unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 1, 2, 3, 4 or 5) of the substituents listed for that group in which the substituents may be the same of different.
  • an optionally substituted group has one substituent.
  • an optionally substituted group has two substituents.
  • an optionally substituted group has three substituents.
  • an optionally substituted group has four substituents.
  • an optionally substituted group has 1 to 2, 2 to 5, 3 to 5, 2 to 3, 2 to 4, 3 to 4, 1 to 3, 1 to 4 or 1 to 5 substituents.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • treatment is an approach for obtaining beneficial or desired results including clinical results.
  • beneficial or desired results include, but are not limited to, one or more of the following: decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of individuals.
  • beneficial or desired results include shrinking a tumor (reducing tumor size); decreasing the growth rate of the tumor (such as to suppress tumor growth); reducing the number of cancer cells; inhibiting, retarding or slowing to some extent and preferably stopping cancer cell infiltration into peripheral organs; inhibiting (slowing to some extent and preferably stopping) tumor metastasis; inhibiting tumor growth; preventing or delaying occurrence and/or recurrence of tumor; and/or relieving to some extent one or more of the symptoms associated with the cancer.
  • beneficial or desired results include preventing or delaying occurrence and/or recurrence, such as of unwanted cell proliferation.
  • “delaying development of a disease” means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease (such as cancer). This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. For example, a late stage cancer, such as development of metastasis, may be delayed.
  • an “effective dosage” or “effective amount” of compound or salt thereof or pharmaceutical composition is an amount sufficient to effect beneficial or desired results.
  • beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity of, or delaying the onset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • beneficial or desired results include ameliorating, palliating, lessening, delaying or decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival.
  • an effective amount comprises an amount sufficient to cause a tumor to shrink and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation.
  • an effective amount is an amount sufficient to delay development.
  • an effective amount is an amount sufficient to prevent or delay occurrence and/or recurrence.
  • an effective amount can be administered in one or more administrations, in the case of cancer, the effective amount of the drug or composition may: (i) reduce the number of cancer cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and preferably stop cancer cell infiltration into peripheral organs; (iv) inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer.
  • An effective dosage can be administered in one or more administrations.
  • an effective dosage of compound or a salt thereof, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. It is intended and understood that an effective dosage of a compound or salt thereof, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • the term “individual” is a mammal, including humans.
  • An individual includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate.
  • the individual is human.
  • the individual (such as a human) may have advanced disease or lesser extent of disease, such as low tumor burden.
  • the individual is at an early stage of a proliferative disease (such as cancer).
  • the individual is at an advanced stage of a proliferative disease (such as an advanced cancer).
  • Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”.
  • a compound described in Table 1 is provided. Also provided are salts of compounds referred to herein, such as pharmaceutically acceptable salts.
  • the disclosure also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms, and any tautomers or other forms of the compounds described.
  • a compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein.
  • Compositions comprising a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, are provided, such as compositions of substantially pure compounds.
  • a composition containing a compound as detailed herein or a salt thereof is in substantially pure form.
  • substantially pure intends a composition that contains no more than 35% impurity, wherein the impurity denotes a compound other than the compound comprising the majority of the composition or a salt thereof.
  • a composition of substantially pure compound or a salt thereof wherein the composition contains no more than 25%, 20%, 15%, 10%, or 5% impurity. In some embodiments, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 3%, 2%, 1% or 0.5% impurity. Table 1
  • provided herein is a compound described in Table 1, or a tautomer thereof, or a salt of any of the foregoing, and uses thereof. In some embodiments, provided herein is a compound described in Table 1 or a pharmaceutically acceptable salt thereof.
  • the compounds depicted herein may be present as salts even if salts are not depicted and it is understood that the present disclosure embraces all salts and solvates of the compounds depicted here, as well as the non-salt and non-solvate form of the compound, as is well understood by the skilled artisan.
  • the salts of the compounds provided herein are pharmaceutically acceptable salts. Where one or more tertiary amine moiety is present in the compound, the N-oxides are also provided and described.
  • tautomeric forms may be present for any of the compounds described herein, each and every tautomeric form is intended even though only one or some of the tautomeric forms may be explicitly depicted.
  • the tautomeric forms specifically depicted may or may not be the predominant forms in solution or when used according to the methods described herein.
  • the present disclosure also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms of the compounds described, such as the compounds of Table 1.
  • the structure or name is intended to embrace all possible stereoisomers of a compound depicted, and each unique stereoisomer has a compound number bearing a suffix “a”, “b”, etc. All forms of the compounds are also embraced by the disclosure, such as crystalline or non-crystalline forms of the compounds.
  • Compositions comprising a compound of the disclosure are also intended, such as a composition of substantially pure compound, including a specific stereochemical form thereof, or a composition comprising mixtures of compounds of the disclosure in any ratio, including two or more stereochemical forms, such as in a racemic or non-racemic mixture.
  • the disclosure also intends isotopically-labeled and/or isotopically-enriched forms of compounds described herein.
  • the compounds herein may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
  • the compound is isotopically-labeled, such as an isotopically-labeled derivative of a compound disclosed herein or variations thereof described herein, where a fraction of one or more atoms are replaced by an isotope of the same element.
  • Exemplary isotopes that can be incorporated into compounds of the disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, chlorine, such as 2 H, 3 H, n C, 13 C, 14 C, 13 N, 15 O, 17 O, 32 P, 35 S, 18 F, 36 C1.
  • Certain isotope labeled compounds e.g. 3 H and 14 C
  • is useful in compound or substrate tissue distribution studies. Incorporation of heavier isotopes such as deuterium ( 2 H) can afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life, or reduced dosage requirements and, hence may be preferred in some instances.
  • Isotopically-labeled compounds of the present disclosure can generally be prepared by standard methods and techniques known to those skilled in the art or by procedures similar to those described in the accompanying Examples substituting appropriate isotopically-labeled reagents in place of the corresponding non-labeled reagent.
  • the disclosure also includes any or all metabolites of any of the compounds described.
  • the metabolites may include any chemical species generated by a biotransformation of any of the compounds described, such as intermediates and products of metabolism of the compound, such as would be generated in vivo following administration to a human.
  • Articles of manufacture comprising a compound described herein, or a salt or solvate thereof, in a suitable container are provided.
  • the container may be a vial, jar, ampoule, preloaded syringe, i.v. bag, and the like.
  • the compounds detailed herein are orally bioavailable.
  • the compounds may also be formulated for parenteral (e.g., intravenous) administration.
  • One or several compounds described herein can be used in the preparation of a medicament by combining the compound or compounds as an active ingredient with a pharmacologically acceptable carrier, which are known in the art.
  • a pharmacologically acceptable carrier which are known in the art.
  • the carrier may be in various forms.
  • the manufacture of a medicament is for use in any of the methods disclosed herein, e.g., for the treatment of cancer.
  • the compounds of the disclosure may be prepared by a number of processes as generally described below and more specifically in the Examples hereinafter (such as the schemes provided in the Examples below).
  • a particular enantiomer of a compound this may be accomplished from a corresponding mixture of enantiomers using any suitable conventional procedure for separating or resolving enantiomers.
  • diastereomeric derivatives may be produced by reaction of a mixture of enantiomers, e.g., a racemate, and an appropriate chiral compound. The diastereomers may then be separated by any convenient means, for example by crystallization and the desired enantiomer recovered. In another resolution process, a racemate may be separated using chiral High Performance Liquid Chromatography. Alternatively, if desired a particular enantiomer may be obtained by using an appropriate chiral intermediate in one of the processes described.
  • Chromatography, recrystallization and other conventional separation procedures may also be used with intermediates or final products where it is desired to obtain a particular isomer of a compound or to otherwise purify a product of a reaction.
  • Solvates of a compound provided herein or a salt thereof are also contemplated. Solvates contain either stoichiometric or non- stoichiometric amounts of a solvent, and are often formed during the process of crystallization. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol.
  • R 1 is independently F, Cl, or methyl
  • R 2 is Ci-C 6 alkyl, C3-C6 cycloalkyl or -(C1-C3 alkylene)CF3; wherein: indicates an aromatic ring;
  • M 1 is CH, CR 3b or N;
  • M 2 is CH, CR 3b , N, or absent;
  • M 3 is CH, CR 3b , N, O, or S;
  • M 4 is CH, CR 3b , N, O or S,
  • R 3a is -CN, -OR 17 , C3-C6 cycloalkyl optionally substituted by Ci-
  • R 3b is halogen or -CN; each R 4 is independently oxo, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -C(O)R 17 , -C(O)OR 17 , -C(O)NR 17 R 18 , -CN, -Si(Ci-C 6 alkyl) 3 , -OR 17 , -NR 17 R 18 , -OC(O)NR 17 R 18 , -NR 17 C(O)R 18 , -S(O) 2 R 17 , -NR 17 S(O) 2 R 18 , -S(O) 2 NR 17 R 18 , C 3 - Ce cycloalkyl, 3- to 6-membered heterocyclyl, -(C1-C3 alkylene)CN, -(C1-C3 alkylene)OR 17 , -(C1-C3 alkylene)NR 17 R 18 ,
  • compositions of any of the compounds detailed herein are embraced by this disclosure.
  • the present disclosure includes pharmaceutical compositions comprising a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutically acceptable salt is an acid addition salt, such as a salt formed with an inorganic or organic acid.
  • Pharmaceutical compositions may take a form suitable for oral, buccal, parenteral, nasal, topical or rectal administration or a form suitable for administration by inhalation.
  • a compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein.
  • Compositions comprising a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing are provided, such as compositions of substantially pure compounds.
  • a composition containing a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is in substantially pure form.
  • the compounds herein are synthetic compounds prepared for administration to an individual.
  • compositions are provided containing a compound in substantially pure form.
  • the present disclosure embraces pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier.
  • methods of administering a compound are provided. The purified forms, pharmaceutical compositions and methods of administering the compounds are suitable for any compound or form thereof detailed herein.
  • a compound detailed herein or salt thereof may be formulated for any available delivery route, including an oral, mucosal (e.g., nasal, sublingual, vaginal, buccal or rectal), parenteral (e.g., intramuscular, subcutaneous or intravenous), topical or transdermal delivery form.
  • oral, mucosal e.g., nasal, sublingual, vaginal, buccal or rectal
  • parenteral e.g., intramuscular, subcutaneous or intravenous
  • topical or transdermal delivery form e.g., topical or transdermal delivery form.
  • a compound or salt thereof may be formulated with suitable carriers to provide delivery forms that include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultices), pastes, powders, dressings, creams, solutions, patches, aerosols (e.g., nasal spray or inhalers), gels, suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions or water-in-oil liquid emulsions), solutions and elixirs.
  • suitable carriers include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultic
  • One or several compounds described herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be used in the preparation of a formulation, such as a pharmaceutical formulation, by combining the compound or compounds, or a salt thereof, as an active ingredient with a pharmaceutically acceptable carrier, such as those mentioned above.
  • a pharmaceutically acceptable carrier such as those mentioned above.
  • the carrier may be in various forms.
  • pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants.
  • Formulations comprising the compound may also contain other substances which have valuable therapeutic properties.
  • Pharmaceutical formulations may be prepared by known pharmaceutical methods. Suitable formulations can be found, e.g., in Remington’s Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, PA, 20 th ed. (2000), which is incorporated herein by reference.
  • Compounds as described herein may be administered to individuals in a form of generally accepted oral compositions, such as tablets, coated tablets, and gel capsules in a hard or in soft shell, emulsions or suspensions.
  • carriers which may be used for the preparation of such compositions, are lactose, corn starch or its derivatives, talc, stearate or its salts, etc.
  • Acceptable carriers for gel capsules with soft shell are, for instance, plant oils, wax, fats, semisolid and liquid poly-ols, and so on.
  • pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants.
  • any of the compounds described herein can be formulated in a tablet in any dosage form described, for example, a compound as described herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be formulated as a 10 mg tablet.
  • compositions comprising a compound provided herein are also described.
  • the composition comprises a compound or salt thereof and a pharmaceutically acceptable carrier or excipient.
  • a composition of substantially pure compound is provided.
  • Compounds and compositions detailed herein such as a pharmaceutical composition containing a compound described in Table 1 provided herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier or excipient, may be used in methods of administration and treatment as provided herein.
  • the compounds and compositions may also be used in in vitro methods, such as in vitro methods of administering a compound or composition to cells for screening purposes and/or for conducting quality control assays.
  • a method of treating a disease in an individual comprising administering an effective amount of a compound described in Table 1 or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, to the individual.
  • a method of treating a proliferative disease in an individual comprising administering an effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, to the individual.
  • a method of treating cancer in an individual comprising administering an effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, to the individual.
  • the compound is administered to the individual according to a dosage and/or method of administration described herein.
  • the cancer in the individual has one or more TP53 gene mutations or expresses mutant p53.
  • the cancer in the individual that has one or more TP53 gene mutations or expresses mutant p53 is glioblastoma.
  • TP53 is the human gene that encodes p53.
  • a method of treating a cancer in an individual comprising (a) selecting the individual for treatment based on (i) the presence of one or more mutations of the TP 53 gene in the cancer, or (ii) expression of mutant p53 in the cancer, and administering an effective amount of a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing,, to the individual.
  • the cancer is assayed for the expression of mutant p53.
  • the TP 53 gene of the cancer is sequenced to detect the one or more mutations.
  • the TP 53 gene is sequenced by biopsying the cancer and sequencing the TP53 gene from the biopsied cancer. In some embodiments, the TP53 gene is sequenced by sequencing circulating-tumor DNA (ctDNA) from the individual.
  • ctDNA circulating-tumor DNA
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is used to treat an individual having a proliferative disease, such as cancer as described herein.
  • the individual is at risk of developing a proliferative disease, such as cancer.
  • the individual is determined to be at risk of developing cancer based upon one or more risk factors.
  • the risk factor is a family history and/or gene associated with cancer.
  • the present compounds or salts thereof are believed to be effective for treating a variety of diseases and disorders.
  • the present compositions may be used to treat a proliferative disease, such as cancer.
  • the cancer is a solid tumor.
  • the cancer is any of adult and pediatric oncology, myxoid and round cell carcinoma, locally advanced tumors, metastatic cancer, human soft tissue sarcomas, including Ewing's sarcoma, cancer metastases, including lymphatic metastases, squamous cell carcinoma, particularly of the head and neck, esophageal squamous cell carcinoma, oral carcinoma, blood cell malignancies, including multiple myeloma, leukemias, including acute lymphocytic leukemia, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, and hairy cell leukemia, effusion lymphomas (body cavity based lymphomas), thymic lymphoma, cutaneous T cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cancer of the adrenal cortex, ACTH-producing tumors, lung cancer, including small cell carcinoma and nonsmall cell cancers, breast cancer
  • the compounds and compositions described herein suppress G2-M checkpoint in a cell (such as a cancer cell).
  • the cancer cell is a cancer cell from any of the cancer types described herein. Suppression of the G2-M DNA damage checkpoint results in premature mitosis of the cell, and consequently apoptosis.
  • a method of suppressing the G2-M DNA damage checkpoint in a cell comprising administering an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, to the cell.
  • the G2-M DNA damage checkpoint is suppressed in about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more of cells in a cell population. In some embodiments, the G2-M DNA damage checkpoint is suppressed in up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, or up to about 80% of cells in the cell population.
  • a method of inducing premature mitosis in a cell comprising administering an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, to the cell.
  • premature mitosis is induced in about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more of cells in a cell population.
  • premature mitosis is induced in up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, or up to about 80% of cells in the cell population.
  • apoptosis in some embodiments, provided herein is a method of inducing apoptosis in a cell comprising administering an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, to the cell.
  • apoptosis is induced in about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more of cells in a cell population.
  • apoptosis is induced in up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, or up to about 80% of cells in the cell population.
  • provided herein is a method of inhibiting Weel in a cell comprising administering an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, to the cell.
  • Weel is inhibited by about 10% or more, about 20% or more, about 30% or more, about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 75% or more, about 80% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more.
  • Weel is inhibited up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, up to about 80%, up to about 70%, or up to about 60%.
  • the activity of Weel is measured according to a kinase assay.
  • a method of inhibiting Weel comprising contacting Weel with an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof.
  • a compound disclosed herein or a pharmaceutically acceptable salt thereof binds to Weel with an IC50 of less than 1 pM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 50 nM, less than 10 nM, less than 5 nM, less than 1 nM, or less than 0.5 nM.
  • a compound disclosed herein or a pharmaceutically acceptable salt thereof binds to Weel with an IC50 between 0.1 nM and 1 nM, between 1 nM and 5 nM, between 5 nM and 10 nM, between 10 nM and 50 nM, between 50 nM and 100 nM, between 100 nM and 200 nM, between 200 nM and 300 nM, between 300 nM and 400 nM, between 400 nM and 500 nM, between 500 nM and 600 nM, between 600 nM and 700 nM, between 700 nM and 800 nM, between 800 nM and 900 nM, or between 900 nM and 1 pM.
  • the IC50 is measured according to a kinase assay.
  • the IC50 is measured according to a cell cytotoxicity assay.
  • a method of inhibiting the proliferation of a cell comprising contacting the cell with an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof.
  • a compound disclosed herein or a pharmaceutically acceptable salt thereof is effective in inhibiting the proliferation of the cell with an IC50 of less than 5 pM, less than 2 pM, less than 1 pM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, or less than 50 nM.
  • a compound disclosed herein or a pharmaceutically acceptable salt is effective in inhibiting the proliferation of the cell with an IC50 between 10 nM and 20 nM, between 20 nM and 50 nM, between 50 nM and 100 nM, between 100 nM and 500 nM, between 500 nM and 1 pM, between 1 pM and 2 pM, or between 2 pM and 5 pM.
  • the IC50 is measured according to a cell proliferation assay.
  • the presently disclosed compounds, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing may activate the immune system, for example by inducing apoptosis or suppressing mitosis of cancer cells. Accordingly, the present compounds, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, may be used in combination with other anti-cancer agents to enhance tumor immunotherapy.
  • a method of treating a disease in an individual comprising administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and an additional therapeutic agent to the individual.
  • the disease is a proliferative disease such as cancer.
  • the additional therapeutic agent is a cancer immunotherapy agent.
  • the additional therapeutic agent is a chemotherapeutic agent.
  • the additional therapeutic agent is an immuno stimulatory agent.
  • the additional therapeutic agent targets a checkpoint protein (for example an immune checkpoint inhibitor).
  • the additional therapeutic agent is effective to stimulate, enhance or improve an immune response against a tumor.
  • the additional chemotherapeutic agent is a DNA alkylating agent, a platinumbased chemotherapeutic agent, a kinase inhibitor or a DNA damage repair (DDR) pathway inhibitor.
  • the additional chemotherapeutic agent is a DNA alkylating agent.
  • the additional chemotherapeutic agent is a platinum-based chemotherapeutic agent. In some embodiments, the additional chemotherapeutic agent is a kinase inhibitor. In some embodiments, the additional chemotherapeutic agent is a DNA damage repair (DDR) pathway inhibitor.
  • DDR DNA damage repair
  • a combination therapy for the treatment of a disease such as cancer.
  • a method of treating a disease in an individual comprising administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, in combination with a radiation therapy.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an additional chemotherapeutic agent.
  • the chemotherapeutic agent is a kinase inhibitor or an agent that inhibits one or more DNA damage repair (DDR) pathways.
  • DDR DNA damage repair
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the additional chemotherapeutic agent.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the additional chemotherapeutic agent.
  • chemotherapeutic agents that can be used in combination with a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, include DNA-targeted agents, a DNA alkylating agent (such as cyclophosphamide, mechlorethamine, chlorambucil, melphalan, dacarbazine, temozolomide or nitrosoureas), a topoisomerase inhibitor (such as a Topoisomerase I inhibitor (e.g., irinotecan or topotecan) or a Topoisomerase II inhibitor (e.g., etoposide or teniposide)), an anthracycline (such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, or valrubicin), a histone deacetylase inhibitor (such as vorinostat), a DNA al
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a kinase inhibitor (such as bortezomib, erlotinib, gefitinib, imatinib, vemurafenib, or vismodegib).
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the kinase inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the kinase inhibitor.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DNA damaging agent.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the DNA damaging agent.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the DNA damaging agent.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DNA alkylating agent (such as cyclophosphamide, mechlorethamine, chlorambucil, melphalan, dacarbazine, temozolomide or nitrosoureas).
  • a DNA alkylating agent such as cyclophosphamide, mechlorethamine, chlorambucil, melphalan, dacarbazine, temozolomide or nitrosoureas.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the DNA alkylating agent.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the DNA alkylating agent.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a topoisomerase inhibitor (such as a Topoisomerase I inhibitor (e.g., irinotecan or topotecan) or a Topoisomerase II inhibitor (e.g., etoposide or teniposide)).
  • a topoisomerase inhibitor such as a Topoisomerase I inhibitor (e.g., irinotecan or topotecan) or a Topoisomerase II inhibitor (e.g., etoposide or teniposide)
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the topoisomerase inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the topoisomerase inhibitor.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an anthracycline (such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, or valrubicin).
  • an anthracycline such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, or valrubicin.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the anthracycline.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the anthracycline.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a histone deacetylase inhibitor (such as vorinostat or romidepsin).
  • a histone deacetylase inhibitor such as vorinostat or romidepsin.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the histone deacetylase inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the histone deacetylase inhibitor.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a taxane (such as paclitaxel or docetaxel).
  • a taxane such as paclitaxel or docetaxel.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the taxane.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the taxane.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a nucleotide analog or precursor analog (such as azacitidine, azathioprine, capecitabine, cytarabine, doxifluridine, 5-fluorouracil, gemcitabine, hydroxyurea, mercaptopurine, methotrexate, or tioguanine).
  • a nucleotide analog or precursor analog such as azacitidine, azathioprine, capecitabine, cytarabine, doxifluridine, 5-fluorouracil, gemcitabine, hydroxyurea, mercaptopurine, methotrexate, or tioguanine.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the nucleotide analog or precursor analog.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the nucleotide analog or precursor analog.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a platinum-based chemotherapeutic agent (such as cisplatin, carboplatin, or oxaliplatin).
  • a platinum-based chemotherapeutic agent such as cisplatin, carboplatin, or oxaliplatin.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the platinum-based chemotherapeutic agent.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the platinum-based chemotherapeutic agent.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of pemetrexed.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the pemetrexed.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the pemetrexed.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DDR pathway inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the DDR pathway inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the DDR pathway inhibitor.
  • inhibitors of the DDR pathway include poly(ADP-ribose) polymerase (PARP) inhibitors (such as olaparib, rucaparib, niraparib, or talazoparib), ataxia telangiectasia mutated (ATM) protein inhibitors, ataxia telangiectasia and Rad3 -related (ATR) protein inhibitors, checkpoint kinase 1 (Chkl) inhibitors, or combinations thereof.
  • PARP poly(ADP-ribose) polymerase
  • ATM telangiectasia mutated
  • ATR ataxia telangiectasia and Rad3 -related
  • Chkl checkpoint kinase 1
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a PARP inhibitor (such as olaparib, rucaparib, niraparib, or talazoparib).
  • a PARP inhibitor such as olaparib, rucaparib, niraparib, or talazoparib.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the PARP inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the PARP inhibitor.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATM protein inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the ATM protein inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the ATM protein inhibitor.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATR protein inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the ATR protein inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the ATR protein inhibitor.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an Chkl inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered prior to, after, or simultaneously co-administered with the Chkl inhibitor.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the Chkl inhibitor.
  • a combination therapy in which a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is coadministered (which may be separately or simultaneously) with one or more additional agents that are effective in stimulating immune responses to thereby further enhance, stimulate or upregulate immune responses in a subject.
  • a method for stimulating an immune response in a subject comprising administering to the subject a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and one or more immuno stimulatory antibodies, such as an anti-PD-1 antibody, an anti-PD-Ll antibody and/or an anti-CTLA-4 antibody, such that an immune response is stimulated in the subject, for example to inhibit tumor growth.
  • the subject is administered a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-PD-1 antibody.
  • the subject is administered a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-PD-Ll antibody.
  • the subject is administered a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-CTLA-4 antibody.
  • the immuno stimulatory antibody e.g., anti-PD-1, anti-PD-Ll and/or anti-CTLA-4 antibody
  • the immunostimulatory antibody can be, for example, a chimeric or humanized antibody (e.g., prepared from a mouse anti-PD-1, anti-PD-Ll and/or anti-CTLA-4 antibody).
  • the present disclosure provides a method for treating a proliferative disease (e.g., cancer), comprising administering a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-PD-1 antibody to a subject.
  • a compound disclosed herein or , or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered at a subtherapeutic dose
  • the anti-PD-1 antibody is administered at a subtherapeutic dose
  • both are administered at a subtherapeutic dose.
  • the present disclosure provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a subtherapeutic dose of anti- PD-1 antibody to a subject.
  • the subject is human.
  • the anti-PD-1 antibody is a human sequence monoclonal antibody.
  • the present disclosure provides a method for treating a hyperproliferative disease (e.g., cancer), comprising administering a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-PD-Ll antibody to a subject.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered at a subtherapeutic dose
  • the anti-PD-Ll antibody is administered at a subtherapeutic dose
  • both are administered at a subtherapeutic dose.
  • the present disclosure provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a subtherapeutic dose of anti- PD-Ll antibody to a subject.
  • the subject is human.
  • the anti-PD-Ll antibody is a human sequence monoclonal antibody.
  • the combination of therapeutic agents discussed herein can be administered concurrently as a single composition in a pharmaceutically acceptable carrier, or concurrently as separate compositions each in a pharmaceutically acceptable carrier. In another embodiment, the combination of therapeutic agents can be administered sequentially.
  • an anti-CTLA-4 antibody and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing can be administered sequentially, such as anti-CTLA-4 antibody being administered first and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, second, or a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, being administered first and anti-CTLA-4 antibody second.
  • an anti-PD-1 antibody and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing can be administered sequentially, such as anti-PD-1 antibody being administered first and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, second, or a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, being administered first and anti-PD-1 antibody second.
  • an anti-PD-Ll antibody and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing can be administered sequentially, such as anti-PD-Ll antibody being administered first and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, second, or a compound disclosed, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, being administered first and anti- PD-Ll antibody second.
  • the combination of a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing can be further combined with an immunogenic agent, such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines.
  • an immunogenic agent such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing can also be further combined with standard cancer treatments.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing can be effectively combined with chemotherapeutic regimes. In these instances, it is possible to reduce the dose of other chemotherapeutic reagent administered with the combination of the instant disclosure (Mokyr et al. (1998) Cancer Research 58: 5301-5304).
  • combination therapies with a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing include radiation, surgery, or hormone deprivation.
  • Angiogenesis inhibitors can also be combined with a compound disclosed herein or a salt thereof. Inhibition of angiogenesis leads to tumor cell death, which can be a source of tumor antigen fed into host antigen presentation pathways.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing can be used in conjunction with anti-neoplastic antibodies.
  • treatment with an anti-cancer antibody or an anti-cancer antibody conjugated to a toxin can lead to cancer cell death (e.g., tumor cells) which would potentiate an immune response mediated by CTLA-4, PD-1, PD-L1 or a compound disclosed herein or a salt thereof.
  • a treatment of a hyperproliferative disease can include an anti-cancer antibody in combination with a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and anti-CTLA-4 and/or anti-PD-1 and/or anti-PD-Ll antibodies, concurrently or sequentially or any combination thereof, which can potentiate anti-tumor immune responses by the host.
  • anti-CTLA-4 and/or anti-PD-1 and/or anti-PD-Ll antibodies concurrently or sequentially or any combination thereof, which can potentiate anti-tumor immune responses by the host.
  • Other antibodies that can be used to activate host immune responsiveness can be further used in combination with a compound disclosed herein or a salt thereof.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing can be combined with an anti- CD73 therapy, such as an anti-CD73 antibody.
  • a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing is administered in combination with another Weel inhibitor.
  • the dose of a compound administered to an individual may vary with the particular compound or salt thereof, the method of administration, and the particular disease, such as type and stage of cancer, being treated.
  • the amount of the compound or salt thereof is a therapeutically effective amount.
  • the effective amount of the compound may in one aspect be a dose of between about 0.01 and about 100 mg/kg.
  • Effective amounts or doses of the compounds of the disclosure may be ascertained by routine methods, such as modeling, dose escalation, or clinical trials, taking into account routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the disease to be treated, the subject’s health status, condition, and weight.
  • An exemplary dose is in the range of about from about 0.7 mg to 7 g daily, or about 7 mg to 350 mg daily, or about 350 mg to 1.75 g daily, or about 1.75 to 7 g daily.
  • Any of the methods provided herein may in one aspect comprise administering to an individual a pharmaceutical composition that contains an effective amount of a compound provided herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable excipient.
  • a compound or composition of the disclosure may be administered to an individual in accordance with an effective dosing regimen for a desired period of time or duration, such as at least about one month, at least about 2 months, at least about 3 months, at least about 6 months, or at least about 12 months or longer, which in some variations may be for the duration of the individual’s life.
  • the compound is administered on a daily or intermittent schedule.
  • the compound can be administered to an individual continuously (for example, at least once daily) over a period of time.
  • the dosing frequency can also be less than once daily, e.g., about a once weekly dosing.
  • the dosing frequency can be more than once daily, e.g., twice or three times daily.
  • the dosing frequency can also be intermittent, including a ‘drug holiday’ (e.g., once daily dosing for 7 days followed by no doses for 7 days, repeated for any 14 day time period, such as about 2 months, about 4 months, about 6 months or more). Any of the dosing frequencies can employ any of the compounds described herein together with any of the dosages described herein.
  • a drug holiday e.g., once daily dosing for 7 days followed by no doses for 7 days, repeated for any 14 day time period, such as about 2 months, about 4 months, about 6 months or more.
  • the compounds provided herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, may be administered to an individual via various routes, including, e.g., intravenous, intramuscular, subcutaneous, oral and transdermal.
  • a compound provided herein can be administered frequently at low doses, known as 'metronomic therapy,' or as part of a maintenance therapy using compound alone or in combination with one or more additional drugs.
  • Metronomic therapy or maintenance therapy can comprise administration of a compound provided herein in cycles.
  • Metronomic therapy or maintenance therapy can comprise intra-tumoral administration of a compound provided herein.
  • the disclosure provides a method of treating cancer in an individual by parenterally administering to the individual (e.g., a human) an effective amount of a compound or salt thereof.
  • the route of administration is intravenous, intra-arterial, intramuscular, or subcutaneous.
  • the route of administration is oral.
  • the route of administration is transdermal.
  • compositions including pharmaceutical compositions as described herein for the use in treating, preventing, and/or delaying the onset and/or development of cancer and other methods described herein.
  • the composition comprises a pharmaceutical formulation which is present in a unit dosage form.
  • articles of manufacture comprising a compound of the disclosure or a salt thereof, composition, and unit dosages described herein in suitable packaging for use in the methods described herein.
  • suitable packaging is known in the art and includes, for example, vials, vessels, ampules, bottles, jars, flexible packaging and the like.
  • An article of manufacture may further be sterilized and/or sealed.
  • kits for carrying out the methods of the disclosure which comprises one or more compounds described herein or a composition comprising a compound described herein.
  • the kits may employ any of the compounds disclosed herein.
  • the kit employs a compound described herein or a salt thereof.
  • the kits may be used for any one or more of the uses described herein, and, accordingly, may contain instructions for the treatment of cancer.
  • Kits generally comprise suitable packaging.
  • the kits may comprise one or more containers comprising any compound described herein.
  • Each component if there is more than one component
  • kits may be in unit dosage forms, bulk packages (e.g., multi-dose packages) or sub-unit doses.
  • kits may be provided that contain sufficient dosages of a compound as disclosed herein and/or an additional pharmaceutically active compound useful for a disease detailed herein to provide effective treatment of an individual for an extended period, such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 7 months, 8 months, 9 months, or more.
  • Kits may also include multiple unit doses of the compounds and instructions for use and be packaged in quantities sufficient for storage and use in pharmacies (e.g., hospital pharmacies and compounding pharmacies).
  • kits may optionally include a set of instructions, generally written instructions, although electronic storage media (e.g.. magnetic diskette or optical disk) containing instructions are also acceptable, relating to the use of component(s) of the methods of the present disclosure.
  • the instructions included with the kit generally include information as to the components and their administration to an individual.
  • Step-1 Synthesis of l-(4-fhioro-3-methylphenyl)-2-ilsopropyl-6-(methylthio)- lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of 2-isopropyl-6- (methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (250 mg, 1.11 mmol, 1.0 eq) and 4-fluoro-3-methylphenylboronic acid (206 mg, 1.34 mmol, 1.2 eq) in acetonitrile (10 mL) was added 2,2-bipyridine (260 mg, 1.66 mmol, 1.5 eq), copper acetate (400 mg, 2.22 mmol, 2.0 eq) and Na2COs (552 mg, 3.3 mmol, 3.0 eq) and DCM (30 mL).
  • reaction mixture was stirred at RT for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by silica gel column chromatography to afford the desired product (50 mg, 13.7%).
  • Step-2 Synthesis of tert-butyl 7-(l-(4-fhioro-3-methylphenyl)-2-isopropyl-3- oxo-2, 3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline- 2(lH)-carboxylate: To a stirred solution l-(4-fluoro-3-methylphenyl)-2-isopropyl-6- (methylthio)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one (50 mg, 0.15 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (60 mg, 0.3 mmol, 2.0 eq) and allowed to stir at RT for 1 h.
  • m-CPBA 60 mg, 0.3 mmol, 2.0 eq
  • Step-3 Synthesis of l-(4-fhioro-3-methylphenyl)-2-isopropyl-6-(l, 2,3,4- tetrahydroisoquinolin-7-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one: tert-Butyl 7- (l-(4-fluoro-3-methylphenyl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin- 6-ylamino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (50 mg, 0.09 mmol, 1.0 eq) was dissolved in dioxane (0.5 mL), followed by dropwise addition of 4.0 M-HC1 (0.5 mL) and allowed to stir at RT for 1 h.
  • Example S-2 Synthesis of 6-((2-(cyclopropylmethyl)-l ,2,3,4-tetrahydroisoquinolin-6- yl)amino)-l-(5-fluoro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-l,2-dihydro-3H- pyrazolo[3,4-d]pyrimidin-3-one (Compound No. 1.2)
  • reaction mixture was diluted with sodium bicarbonate and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by reverse phase chromatography to afford the desired compound (54 mg, 48.5%).
  • Example S-3 Synthesis of l-(3-tert-butyl-5-fluorophenyl)-2-isopropyl-6-(l, 2,3,4- tetrahydroisoquinolin-7-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one ( Compound No. 1.3)
  • Step-1 Synthesis of 2-(3-(tert-butyl)-5-fhiorophenyl)-4,4,5,5-tetramethyl- l,3,2-dioxaborolane:To a stirred solution of l-bromo-3-(tert-butyl)-5-fluorobenzene (300 mg, 1.29 mmol, 1.0 eq) and 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (394.8 mg, 1.55 mmol, 1.2 eq) in dioxane (30 mL) was added KO Ac (252 mg, 2.59 mmol, 2.0 eq) at rt.
  • Step-2 Synthesis of l-(3-(tert-butyl)-5-fhiorophenyl)-2-isopropyl-6- (methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (580 mg, 2.59 mmol, 1.0 eq) and 2-(3-(tert-butyl)-5-fluorophenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (720 mg, 2.59 mmol, 1.0 eq) in (50 mL) of CH2CI2 was added Na2COs (23 mg, 7.77 mmol, 3.0 eq), CU(OAC)2 (2.09 g, 5.18 mmol, 2.0
  • Step-3 Synthesis of tert-butyl 7-((l-(3-(tert-butyl)-5-fhiorophenyl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(3-(tert-butyl)-5- fluorophenyl)-2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (81 mg, 0.216 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (103 mg, 0.43 mmol, 2.0 eq) and allowed to stir at RT for 30 minutes.
  • m-CPBA 103 mg, 0.43
  • Step-4 Synthesis of 1 l-(3-(tert-butyl)-5-fhiorophenyl)-2-isopropyl-6- ((l,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3- one,: tert-Butyl 7-((l-(3-(tert-butyl)-5-fluorophenyl)-2-isopropyl-3-oxo-2,3-dihydro-lH- pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (35 mg, 0.06 mmol, 1.0 eq) was dissolved in 4.0 M-HC1 (0.5 mL) and allowed to stir at RT for 2 h.
  • Example S-4 Synthesis of7-((l-(5-fluoro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinolin-l(2H)-one (Compound No. 1.4)
  • Example S-5 Synthesis ofl-(5-fluoro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(2-(2- hydroxyacetyl)-l,2,3,4-tetrahydroisoquinolin-6-ylamino)-2-isopropyl-lH-pyrazolo[3,4- d]pyrimidin-3(2H)-one (Compound No. 1.5)
  • Example S-6 Synthesis of4-(2-isopropyl-3-oxo-6-((l,2,3,4-tetrahydroisoquinolin-6- yl)amino)-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)-2-(trifluoromethyl)benzonitrile (Compound No. 1.6)
  • Step-1 Synthesis of 4-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-lH- pyrazolo[3,4-d]pyrimidin-l-yl)-2-(trifhioromethyl)benzonitrile: To a stirred solution of 2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (224 mg , 1.0 mmol, 1.0 eq) and (4-cyano-3-(trifluoromethyl)phenyl)boronic acid (214.9 mg, 1.0 mmol, 1.2 eq) in ACN (40 mL) was added 2,2-bipyridine (312.3 mg, 2.0 mmol, 2.0 eq), copper acetate (364 mg, 2.0 mmol, 2.0 eq) and Na2COs (318 mg, 3.0 mmol, 3.0 eq).
  • reaction mixture was stirred at RT for 48h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by silica gel column chromatography to afford the desired product (185 mg, 47.1%).
  • Step-2 Synthesis of tert-butyl 6-((l-(4-cyano-3-(trifhioromethyl)phenyl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution 4-(2-isopropyl-6- (methylthio)-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)-2-
  • Step-3 Synthesis of 4-(2-isopropyl-3-oxo-6-((l,2,3,4-tetrahydroisoquinolin-6- yl)amino)-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)-2-
  • Example S-7 Synthesis ofl-(3-(6-((l,l-dimethyl-l,2,3,4-tetrahydroisoquinolin-6-yl)amino)- 2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)phenyl)cyclopropane-l- carbonitrile (Compound No. 1.7)
  • Step-1 Synthesis of l-(3-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-lH- pyrazolo[3,4-d]pyrimidin-l-yl)phenyl)cyclopropane-l-carbonitrile: To a stirred solution of 2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (224 mg , 1.0 mmol, 1.0 eq) and l-(3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)cyclopropane-l- carbonitrile (269.15 mg, 1.0 mmol, 1.0 eq) in ACN (15 mL) was added 2,2-bipyridine (312.3 mg, 2.0 mmol, 2.0 eq), copper acetate (364 mg, 2.0
  • reaction mixture was stirred at RT for 48 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated, and purified by silica gel column chromatography to afford the desired product (80 mg, 21.9%).
  • Step-2 Synthesis of tert-butyl 6-((l-(3-(l-cyanocyclopropyl)phenyl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-l,l-dimethyl- 3,4-dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution l-(3-(2-isopropyl-6- (methylthio)-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)phenyl)cyclopropane-l- carbonitrile (80 mg, 0.22 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (97 mg, 0.43 mmol, 2.0 eq) and allowed to stir at RT for 1
  • Step-3 Synthesis of l-(3-(6-((l,l-dimethyl-l,2,3,4-tetrahydroisoquinolin-6- yl)amino)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l- yl)phenyl)cyclopropane-l-carbonitrile: tert-Butyl 6-((l-(3-(l-cyanocyclopropyl)phenyl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-l,l-dimethyl-3,4- dihydroisoquinoline-2(lH)-carboxylate (40 mg, 0.07 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition
  • Example S-8 Synthesis of2-fluoro-5-(2-isopropyl-3-oxo-6-((l,2,3,4-tetrahydroisoquinolin-7- yl)amino)-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)benzonitrile (Compound No 1.8)
  • Step-1 Synthesis of 2-fluoro-5-(2-isopropyl-6-(methylthio)-3-oxo-2,3- dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)benzonitrile: To a stirred solution of 2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (250 mg, 1.11 mmol, 1.0 eq) and 3-cyano-4-fluorophenylboronic acid (220 mg, 1.34 mmol, 1.2 eq) in acetonitrile (10 mL) was added 2,2-bipyridine (260 mg, 1.66 mmol, 1.5 eq), copper acetate (400 mg, 2.22 mmol, 2.0 eq) and Na2COs (552 mg, 3.3 mmol, 3.0 eq).
  • reaction mixture was stirred at RT for 24h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated, and purified by silica gel column chromatography to afford the desired product (70 mg, 18.4%).
  • Step-2 Synthesis of tert-butyl 7-(l-(3-cyano-4-fhiorophenyl)-2-isopropyl-3- oxo-2, 3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline- 2(lH)-carboxylate: To a stirred solution 2-fluoro-5-(2-isopropyl-6-(methylthio)-3-oxo-2,3- dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)benzonitrile (60 mg, 0.17 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (75 mg, 0.35 mmol, 2.0 eq) and allowed to stir at RT for 1 h.
  • Step-3 Synthesis of 2-fluoro-5-(2-isopropyl-3-oxo-6-((l, 2,3,4- tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l- yl)benzonitrile hydrochloride tert-Butyl 7-(l-(3-cyano-4-fluorophenyl)-2-isopropyl-3-oxo- 2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(lH)- carboxylate (60 mg, 0.11 mmol, 1.0 eq) was stirred in 2M HC1 in ether (2 mL).
  • Step-1 Synthesis of l-(6-bromo-3-fhioropyridin-2-yl)cyclobutan-l-ol: To a stirred solution of 2,6-dibromo-3-fluoropyridine (1 g, 3.92 mmol, 1.0 eq) in in diethyl ether (20 mL) was added n-BuLi (1.72 ml, 4.31 mmol, 1.1 eq) allowed to stirred at -78 °C for 1 hour, cyclobutanone (0.6 ml, 7.84 mmol, 2.0 eq) was added and allowed to stir at same temp for 15 min.
  • Step-2 Synthesis of l-(5-fhioro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of l-(6-bromo-3-fluoropyridin-2-yl)cyclobutan-l-ol (387 mg, 1.57 mmol, 1.0 eq) and 2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (352 mg, 1.57 mmol, 1.0 eq) in (20 mL) of dioxane were added potassium carbonate (434 mg, 3.14 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (59.8 mg
  • Step-3 Synthesis of tert-butyl 6-((l-(5-fhioro-6-(l- hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4- d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-l,2- dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (197.5 mg, 0.51 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (240 mg, 1,01 mmol, 2.0 eq) and allowed to stir at
  • Step-4 Synthesis of l-(5-fhioro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one dihydrochloride: tert-Butyl 6-((l-(5-fluoro-6-(l- hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4- d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (65 mg, 0.11 mmol, 1.0 eq) was dissolved in dioxane (1.5 mL), followed by dropwise addition
  • Example S-10 Synthesis ofl-(5-fluoro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-6- ((1 ,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l ,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (Compound No 1.10)
  • Step-1 Synthesis of tert-butyl 7-((l-(5-fhioro-6-(l-hydroxycydobutyl)pyridin- 2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro-6-(l- hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one (197.5 mg, 0.51 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (240 mg, 1,01 mmol, 2.0 eq) and allowed to
  • Step-2 Synthesis of l-(5-fhioro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one: tert-Butyl-7-((l-(5-fluoro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate (76 mg, 0.11 mmol, 1.0 eq) was dissolved in dioxane (1.5
  • Example S-ll Synthesis ofl-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one (Compound No 1.11)
  • Step-1 Synthesis of l-(6-bromo-3-fhioropyridin-2-yl)-l-cyclopropylethan-l- ol: To a stirred solution of 2,6-dibromo-3-fluoropyridine (500 mg, 1.96 mmol, 1.0 eq) in (10 mL) of diethylether was added n-BuLi (1.96 ml, 4.90 mmol, 2.5 eq ) allowed to stirred at -78 °C for 1 hour. 1-Cyclopropylethan-l-one (0.2 ml ,2.35 mmol, 1.2 eq) was added and allowed to stir at RT for overnight.
  • Step-2 Synthesis of l-(6-(l-cydopropyl-l-hydroxyethyl)-5-fhioropyridin-2- yl)-2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of l-(6-bromo-3-fluoropyridin-2-yl)-l-cyclopropylethan-l-ol (280 mg, 1.07 mmol, 1.0 eq) and 2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3- one (241 mg, 1.07 mmol, 1.0 eq) in (20 mL) of dioxane were added potassium carbonate (259 mg, 2.14 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min
  • Step-3 Synthesis of tert-butyl 6-((l-(6-(l-cyclopropyl-l-hydroxyethyl)-5- fhioropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6- yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(6-( 1- cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-l,2-dihydro- 3H-pyrazolo[3,4-d]pyrimidin-3-one (172.5 mg, 0.43 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (178 mg,
  • Step-4 Synthesis of l-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fhioropyridin-2- yl)-2-isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H- pyrazolo[3,4-d]pyrimidin-3-one: tert-Butyl 6-((l-(6-(l-cyclopropyl-l-hydroxyethyl)-5- fluoropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6- yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (188 mg, 0.31 mmol, 1.0
  • Example S-12 Synthesis ofl-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2-yl)-2- isopropyl-6-((l ,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l ,2-dihydro-3H-pyrazolo[3,4-
  • Step-1 Synthesis of tert-butyl 7-((l-(6-(l-cyclopropyl-l-hydroxyethyl)-5- fhioropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6- yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate : To a stirred solution of l-(6-( 1- cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-lH- pyrazolo[3,4-d]pyrimidin-3(2H)-one (172.5 mg, 0.43 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (178 mg, 0.8
  • Step-2 Synthesis of l-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fhioropyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one: tert-Butyl 7-((l-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2- yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate (132 mg, 0.31 mmol, 1.0 eq) was dissolved in dioxane (2 mL), followed by
  • Example S-13 Synthesis ofl-(5-fluoro-6-(3-fluorooxetan-3-yl)pyridin-2-yl)-2-isopropyl-6- ((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Step-1 Synthesis of 3-(6-bromo-3-fhioropyridin-2-yl)oxetan-3-: To a stirred solution of 2,6-dibromo-3-fluoropyridine (1 g, 3.92 mmol, 1.0 eq) in (20 mL) of diethyl ether was added n-Buli (1.72 ml, 4.31 mmol, 1.1 eq) allowed to stirred at -78 °C for 1 h. Oxetan-3- one (0.5 ml ,7.84 mmol, 2.0 eq) were added and allowed to stir at same temp for 15 min.
  • Step-2 Synthesis of l-(5-fhioro-6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 3-(6-bromo-3-fluoropyridin-2-yl)oxetan-3-ol (386 mg, 1.55 mmol, 1.0 eq) and 2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (349 mg, 1.55 mmol, 1.0 eq) in (20 mL) of dioxane were added potassium carbonate (430 mg, 3.11 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (59 mg
  • Step-3 Synthesis of l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin-2-yl)-2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of l-(5-fluoro-6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)- l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (260 mg, 0.664 mmol, 1.0 eq) in (10 mL) of DCM was added DAST (0.13 ml, 0.996 mmol, 1.1 eq) allowed to stirred at -78 °C for 1 h .
  • Step-4 Synthesis of tert-butyl 6-((l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin- 2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro-6-(3- fluorooxetan-3-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one (229 mg, 0.58 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (276 mg, 1.16 m
  • Step-5 Synthesis of l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one: tert-Butyl-6-((l-(5-fluoro-6-(3-fluorooxetan-3-yl)pyridin-2-yl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate (174 mg, 0.29 mmol, 1.0 eq) was dissolved in DCM (10 mL), followed by
  • Example S-14 Synthesis ofl-(5-fluoro-6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2-isopropyl-6- (l,2,3,4-tetrahydroisoquinolin-6-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one (Compound No 1.14)
  • Step-1 Synthesis of tert-butyl 6-(l-(5-fhioro-6-(3-hydroxyoxetan-3- yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6- ylamino)-3,4-dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro- 6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-lH-pyrazolo[3,4- d]pyrimidin-3(2H)-one (200 mg, 0.51 mmol, 1.0 eq) in (3.0 mL) of toluene was added m- CPBA (242 mg, 1.02 mmol, 2.0 eq) and
  • Step-2 Synthesis of l-(5-fhioro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one: tert-Butyl-6-(l-(5-fluoro-6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4- dihydroisoquinoline-2(lH)-carboxylate (80 mg, 0.135 mmol, 1.0 eq) was dissolved in DCM (10 mL), followed by dropwise addition of TFA (1 mL) and
  • Step-1 Synthesis of tert-butyl 7-(l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin- 2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro-6-(3- hydroxyoxetan-3-yl)
  • Step-2 Synthesis of l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin-2-yl)-2-isopropyl-6- (l,2,3,4-tetrahydroisoquinolin-7-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one: tert- Butyl 7-(l-(5-fluoro-6-(3-fluorooxetan-3-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH- pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (70 mg, 0.117 mmol, 1.0 eq) was dissolved in DCM (10 mL), followed by dropwise addition of TFA (1 mL) and
  • IC50 values of compounds against WEE1 kinase enzyme were determined by LanthaScreenTM Terbium Labeled TR-FRET assay.
  • Kinase assays were performed in IX kinase buffer (#PV6135, Invitrogen, Life Technologies Grand Island, NY) where total reaction volume was 10 pL in low-volume 384-well plates (#4511, Corning).
  • Serially diluted compounds (3-fold) were incubated with WEE1 Enzyme (1 nM) (#PR7373A, Invitrogen, Life Technologies, Grand Island, NY) for 10 min; a mixture of ATP (10 pM) (#A1852, Sigma, St.
  • Test compounds in 100% DMSO were added into the kinase reaction mixture by Acoustic technology (Echo550; nanoliter range) and incubated for 20 min at room temperature.
  • 33 P- ATP was delivered into the reaction mixture to initiate the reaction. Reactions were carried out at 10 pM ATP. After a 2 hour incubation at room temperature, kinase activity was detected by P81 filter-binding method.
  • Compounds were tested in 10-dose IC50 mode with a 3-fold serial dilution.
  • a nonlinear regression model with a sigmoidal dose response and variable slope within GraphPad Prism GraphPad Software, San Diego, CA was used to calculate the IC50 value of individual test compounds. The results are shown in Table 3.
  • Example B Determination of potency of compounds in cytotoxicity assay inA427 cell line
  • A427 (HTB-53; ATCC), a lung epithelial cell line, was seeded in medium (MEM,
  • Example B Determination of potency of compounds in cell proliferation assay in selected cancer cell lines and cellular PD effects.
  • test compounds are studied in additional cell lines with various histotypes, such as LoVo colorectal adenocarcinoma, NCI-H460 large-cell lung carcinoma, HCT-116 colorectal carcinoma, and A2780 ovarian cancer cells.
  • the cancer cells are harvested during the logarithmic growth period and counted. Cell concentrations are adjusted to the appropriate number with suitable medium, and 90 pL cell suspensions are added to 96- well plates. After cells are seeded, the plates are shaken gently to distribute cells evenly and incubated at 37 °C, 5% CO2 on day 1.
  • Cells are treated with test compounds at 9 concentrations within a desired concentration range (e.g. 1.5 nM - 10 pM) on day 2 by series diluting the test compound stock solution (10 mM in DMSO) with culture medium. Cell viability is assessed by Cell Titer-Gio® as recommended by Promega (Cat. No.: G7572, Promega) typically 72 h posttreatment.
  • a desired concentration range e.g. 1.5 nM - 10 pM
  • Test compounds may be studied in the same and/or other cancer cell lines with varying sensitivities to reported Weel inhibiting compounds using similar proliferation methods with possible variations in cell seeding densities and/or incubation durations.
  • Example B5. Determination of potency of compounds by assay of cellular PD effects.
  • pCDC2 and y-H2AX are two clinically relevant biomarkers associated with Weel inhibition. CDC2Y 15 phosphorylation in cells was reported to be abolished by Weel inhibitors (Gavory G et. al., Almac Discovery, AACR poster, 2016). y-H2AX, a DNA double-strand break marker, was upregulated by Weel treatment in Weel sensitive cell lines (Guertin AD et al., Molecular Cancer Therapeutics, 2013). The effects of selected test compound on pCDC2 and y-H2AX are assessed in selected cancer cell lines post 24 or 48 hr treatment using Western blotting methods with selective antibodies (Guertin AD et al., Molecular Cancer Therapeutics, 2013).
  • A427 cells or AsPC-1 cells were plated in 6-well plates and cultured for 24 hr to approximately 80-90% confluency. Medium was replaced, and the cells were treated with the vehicle control or the test compound at several different concentrations. After incubation of treated cells in cell culture conditions for a specified time (e.g., 24 hr), cells were rinsed with ice-cold PBS and lysed in IX cell lysis buffer containing protease inhibitors and phosphatase inhibitors.
  • ELISA enzyme-linked immunosorbent assay
  • the cells were scraped from the plate with a cell scraper after a brief incubation on ice and transferred to a centrifuge tube, and then subjected to three freeze-thaw cycles in liquid nitrogen and a 37°C water bath for further lysis.
  • the lysates were centrifuged to pellet cell debris (using, for example, a 10 min centrifugation of 2000 X g at 4°C) and the supernatants transferred to fresh tubes on ice.
  • the protein concentrations of the samples were estimated by the Bradford method or equivalent.
  • the ELISA was carried out with the PathScan® Phospho- CDC2 (Tyrl5) Sandwich ELISA Kit (Cat. #7176, Cell Signaling Technology, Danvers, MA) according to the manufacturer’s instructions. Results are shown in Table 5.
  • Changes in the levels of phospho-CDC2 are alternatively or additionally analyzed by Western blotting of the samples using a primary antibody to phospho-CDC2 such as phospho-CDC2 (Tyrl5) (10A11) rabbit mAb (Cat. #4539, Cell Signaling Technology) or rabbit polyclonal anti-CDKl (phospho Y15) antibody (Cat. #ab47594, Abeam, Cambridge, United Kingdom).
  • a primary antibody to phospho-CDC2 such as phospho-CDC2 (Tyrl5) (10A11) rabbit mAb (Cat. #4539, Cell Signaling Technology) or rabbit polyclonal anti-CDKl (phospho Y15) antibody (Cat. #ab47594, Abeam, Cambridge, United Kingdom).
  • Example B6 Evaluation of test compound in mouse xenograft models
  • test compound (as a single agent and in combination with other agents such as gemcitabine, nab-paclitaxel and temozomide)
  • tumor growth experiments are performed in a cell line xenograft model and/or a PDX model.
  • the cell line is chosen based on the in vitro studies described above.
  • the PDX model to be used is established from a tumor taken directly from a patient with, for example, pancreatic ductal adenocarcinoma (PDAC) or glioblastoma.
  • PDAC pancreatic ductal adenocarcinoma
  • Tumors or tumor chucks are implanted subcutaneously into the flanks of nude mice and allowed to grow until the tumor size reaches 200 mm 3 .
  • the treatment groups are, for example: vehicle control, gemcitabine + nab-paclitaxel, test compound alone, gemcitabine + nab-paclitaxel + test compound at 10 mice per group.
  • the treatment groups are alternatively, for example: vehicle control, temozolomide, test compound alone, temozolomide + test compound.
  • the exact treatment groups, drug dose, and dosing schedule are determined specifically for each study according to standard practice. Tumor growth is monitored, and volume recorded at regular intervals. When the individual tumor of each mouse reaches an approximate end-point (tumor volume >1,500 mm 3 ), the mouse is sacrificed with regulated CO2.
  • the tumor growth inhibition (TGI) is calculated by comparing the control group’s tumor measurements with the other study groups once the predetermined endpoint is reached in the control group. Alternatively, cells are implanted orthotopically and overall survival is measured.

Landscapes

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

Abstract

Heterocyclic compounds as Weel inhibitors are provided. The compounds may find use as therapeutic agents for the treatment of diseases and may find particular use in oncology.

Description

HETEROCYCLIC COMPOUNDS AND USES THEREOF
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority benefit of United States Provisional Patent Application No. 63/090,634, filed October 12, 2020. The entire contents of that patent application are incorporated by reference herein.
FIELD
[0002] This disclosure relates generally to therapeutics engaged in inhibition of the DNA damage checkpoint kinase, Weel, which potentiates genotoxic chemotherapies by abrogating cell-cycle arrest and proper DNA repair. The disclosure also provides pharmaceutically acceptable compositions comprising compounds of the present disclosure and methods of using said compositions in the treatment of diseases associated with this pathway.
BACKGROUND
[0003] Weel is a tyrosine kinase that phosphorylates and inactivates Cdc2 and is involved in G checkpoint signaling. More particularly, Weel is involved in G2-M checkpoint signaling. Because p53 is a key regulator in the G checkpoint, p53-deficient tumors rely only on the G checkpoint after DNA damage. More particularly, because p53 is a key regulator in the Gi-S checkpoint, p53 -deficient tumors rely only on the G2-M checkpoint after DNA damage. Hence, such tumors are selectively sensitized to DNA-damaging agents by Weel inhibition.
[0004] Weel belongs to a family of protein kinases involved in the terminal phosphorylation and inactivation of cyclin-dependent kinase 1 -bound cyclin B, resulting in G cell cycle arrest in response to DNA damage. Weel was first identified in fission yeast, where Weel deficiency resulted in premature mitotic entry and replication of smaller-sized yeast. It is the major kinase responsible for the inhibitory phosphorylation of the tyrosine.
[0005] Before cells undergo mitosis, they progress through a tightly controlled cascade of Gi-S, intra-S, and G2-M checkpoints. Weel kinase has emerged as a key G2-M checkpoint regulator. This tyrosine kinase negatively regulates entry into mitosis by catalyzing an inhibitory phosphorylation of Cdc2 (the human homolog of cyclin-dependent kinase 1 (CDK1) on tyrosine- 15 (Y15). This results in inactivation of the Cdc2/cyclin B complex, which arrests cells in G2-M, allowing for DNA repair. Such inhibition also occurs through Chkl-mediated inhibition of Cdc25 phosphatases, which remove the inhibitory phosphorylation on Cdc2. Thus, entry into mitosis rests on a balance between the opposing activities of Weel and Chkl/Cdc25. Weel inhibition is thus expected to abrogate G2-M arrest and propel cells into premature mitosis, a hypothesis confirmed by studies documenting that Weel inhibition by either small molecule inhibitors or small interference RNA leads to premature entry into mitosis and consequent cell death through mitotic catastrophe or apoptosis. (S. Muller, J. Clinical. Oncology, 2015).
[0006] Recently, a few classes of Weel inhibitors have been disclosed. Among them is a selective inhibitor, AZD-1775 (1, 2-allyl-l-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((4-(4- methylpiperazin-l-yl)phenyl)amino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one). AZD-1775 exhibited antitumor activity in various preclinical studies as a monotherapy or in potentiating chemo- and radiotherapy, and is currently in phase I/II clinical trials.
[0007] Weel is highly expressed in several cancer types, including hepatocellular carcinoma, breast cancers, cervical cancers, lung cancers, squamous cell carcinoma, diffuse intrinsic pontine glioma (DIPG), glioblastoma, medulloblastoma, leukemia, melanoma, and ovarian cancers. (P. Reigan et al., Trends in Pharmacol. Sci., 2016).
[0008] There are few Weel inhibitors in clinical development. There is scope to improve Weel inhibitor selectivity and the properties of the inhibitors to permit targeting of specific cancer types.
BRIEF SUMMARY
[0009] In one aspect, provided is a compound selected from the compounds in Table 1, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing.
[0010] In another aspect, provided is a method of treating cancer in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing. Also provided is a method of inhibiting Weel in a cell, comprising administering a compound detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, to the cell.
[0011] In another aspect, provided is a pharmaceutical composition comprising a compound detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier or excipient. Kits comprising a compound detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, are also provided. A compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is also provided for the manufacture of a medicament for the treatment of cancer.
DETAILED DESCRIPTION
Definitions
[0012] “Alkyl” refers to and includes saturated linear and branched univalent hydrocarbon structures and combination thereof, having the number of carbon atoms designated (z.e., Ci-Cio means one to ten carbons). Particular alkyl groups are those having 1 to 20 carbon atoms (a “C1-C20 alkyl”). More particular alkyl groups are those having 1 to 8 carbon atoms (a “Ci-Cs alkyl”), 3 to 8 carbon atoms (a “C3-C8 alkyl”), 1 to 6 carbon atoms (a “Ci-Ce alkyl”), 1 to 5 carbon atoms (a “C1-C5 alkyl”), or 1 to 4 carbon atoms (a “C1-C4 alkyl”). Examples of alkyl include, but are not limited to, groups such as methyl, ethyl, n- propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec -butyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
[0013] “Alkenyl” as used herein refers to an unsaturated linear or branched univalent hydrocarbon chain or combination thereof, having at least one site of olefinic unsaturation (z.e., having at least one moiety of the formula C=C) and having the number of carbon atoms designated (z.e., C2-C10 means two to ten carbon atoms). The alkenyl group may be in “cis” or “trans” configurations, or alternatively in “E” or “Z” configurations. Particular alkenyl groups are those having 2 to 20 carbon atoms (a “C2-C20 alkenyl”), having 2 to 8 carbon atoms (a “C2-C8 alkenyl”), having 2 to 6 carbon atoms (a “C2-C6 alkenyl”), or having 2 to 4 carbon atoms (a “C2-C4 alkenyl”). Examples of alkenyl include, but are not limited to, groups such as ethenyl (or vinyl), prop-l-enyl, prop-2-enyl (or allyl), 2-methylprop-l-enyl, but-l-enyl, but-2-enyl, but-3-enyl, buta- 1,3 -dienyl, 2-methylbuta- 1,3-dienyl, homologs and isomers thereof, and the like.
[0014] “Alkylene” as used herein refers to the same residues as alkyl, but having bivalency. Particular alkylene groups are those having 1 to 6 carbon atoms (a “Ci-Ce alkylene”), 1 to 5 carbon atoms (a “C1-C5 alkylene”), 1 to 4 carbon atoms (a “C1-C4 alkylene”) or 1 to 3 carbon atoms (a “C1-C3 alkylene”). Examples of alkylene include, but are not limited to, groups such as methylene (-CH2-), ethylene (-CH2CH2-), propylene (-CH2CH2CH2-), butylene (-CH2CH2CH2CH2-), and the like. [0015] “Alkynyl” as used herein refers to an unsaturated linear or branched univalent hydrocarbon chain or combination thereof, having at least one site of acetylenic unsaturation (z.e., having at least one moiety of the formula C=C) and having the number of carbon atoms designated (z.e., C2-C10 means two to ten carbon atoms). Particular alkynyl groups are those having 2 to 20 carbon atoms (a “C2-C20 alkynyl”), having 2 to 8 carbon atoms (a “C2-C8 alkynyl”), having 2 to 6 carbon atoms (a “C2-C6 alkynyl”), or having 2 to 4 carbon atoms (a “C2-C4 alkynyl”). Examples of alkynyl include, but are not limited to, groups such as ethynyl (or acetylenyl), prop-l-ynyl, prop-2-ynyl (or propargyl), but-l-ynyl, but-2-ynyl, but-3-ynyl, homologs and isomers thereof, and the like.
[0016] “Aryl” refers to and includes polyunsaturated aromatic hydrocarbon groups. Aryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings. In one variation, the aryl group contains from 6 to 14 annular carbon atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, biphenyl, and the like.
[0017] “Carbonyl” refers to the group C=O.
[0018] “Cycloalkyl” refers to and includes cyclic univalent hydrocarbon structures, which may be fully saturated, mono- or polyunsaturated, but which are non-aromatic, having the number of carbon atoms designated (e.g., C1-C10 means one to ten carbons). Cycloalkyl can consist of one ring, such as cyclohexyl, or multiple rings, such as adamantyl, but excludes aryl groups. A cycloalkyl comprising more than one ring may be fused, spiro or bridged, or combinations thereof. A preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 13 annular carbon atoms. A more preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a "C3-C8 cycloalkyl"). Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1 -cyclohexenyl, 3 -cyclohexenyl, cycloheptyl, norbornyl, and the like.
[0019] “Halo” or “halogen” refers to elements of the Group 17 series having atomic number 9 to 85. Preferred halo groups include fluoro, chloro, bromo and iodo. Where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached, e.g., dihaloaryl, dihaloalkyl, trihaloaryl etc. refer to aryl and alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be but are not necessarily the same halo; thus 4-chloro-3-fluorophenyl is within the scope of dihaloaryl. An alkyl group in which each hydrogen is replaced with a halo group is referred to as a “perhaloalkyl.” A preferred perhaloalkyl group is trifluoroalkyl (-CF3). Similarly, “perhaloalkoxy” refers to an alkoxy group in which a halogen takes the place of each H in the hydrocarbon making up the alkyl moiety of the alkoxy group. An example of a perhaloalkoxy group is trifluoromethoxy (-OCF3).
[0020] “Hetero aryl” refers to and includes unsaturated aromatic cyclic groups having from 1 to 10 annular carbon atoms and at least one annular heteroatom, including but not limited to heteroatoms such as nitrogen, oxygen and sulfur, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quatemized. A heteroaryl group can be attached to the remainder of the molecule at an annular carbon or at an annular heteroatom. Heteroaryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings. Examples of heteroaryl groups include, but are not limited to, pyridyl, pyrimidyl, thiophenyl, furanyl, thiazolyl, and the like.
[0021] “Heterocycle” or “heterocyclyl” refers to a saturated or an unsaturated nonaromatic group having from 1 to 10 annular carbon atoms and from 1 to 4 annular heteroatoms, such as nitrogen, sulfur or oxygen, and the like, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quatemized. A heterocyclyl group may have a single ring or multiple condensed rings, but excludes heteroaryl groups. A heterocycle comprising more than one ring may be fused, spiro or bridged, or any combination thereof. In fused ring s'ystems, one or more of the fused rings can be aryl or heteroaryl. Examples of heterocyclyl groups include, but are not limited to, tetrahydropyranyl, dihydropyranyl, piperidinyl, piperazinyl, pyrrolidinyl, thiazolinyl, thiazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, 2,3-dihydrobenzo[b]thiophen-2-yl, 4- amino-2-oxopyrimidin-l(2H)-yl, and the like.
[0022] “Oxo” refers to the moiety =0.
[0023] “Optionally substituted” unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 1, 2, 3, 4 or 5) of the substituents listed for that group in which the substituents may be the same of different. In one embodiment, an optionally substituted group has one substituent. In another embodiment, an optionally substituted group has two substituents. In another embodiment, an optionally substituted group has three substituents. In another embodiment, an optionally substituted group has four substituents. In some embodiments, an optionally substituted group has 1 to 2, 2 to 5, 3 to 5, 2 to 3, 2 to 4, 3 to 4, 1 to 3, 1 to 4 or 1 to 5 substituents.
[0024] A “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
[0025] As used herein, “treatment” or “treating” is an approach for obtaining beneficial or desired results including clinical results. For example, beneficial or desired results include, but are not limited to, one or more of the following: decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of individuals. In reference to cancers or other unwanted cell proliferation, beneficial or desired results include shrinking a tumor (reducing tumor size); decreasing the growth rate of the tumor (such as to suppress tumor growth); reducing the number of cancer cells; inhibiting, retarding or slowing to some extent and preferably stopping cancer cell infiltration into peripheral organs; inhibiting (slowing to some extent and preferably stopping) tumor metastasis; inhibiting tumor growth; preventing or delaying occurrence and/or recurrence of tumor; and/or relieving to some extent one or more of the symptoms associated with the cancer. In some embodiments, beneficial or desired results include preventing or delaying occurrence and/or recurrence, such as of unwanted cell proliferation.
[0026] As used herein, “delaying development of a disease” means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease (such as cancer). This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. For example, a late stage cancer, such as development of metastasis, may be delayed.
[0027] As used herein, an “effective dosage” or “effective amount” of compound or salt thereof or pharmaceutical composition is an amount sufficient to effect beneficial or desired results. For prophylactic use, beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity of, or delaying the onset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use, beneficial or desired results include ameliorating, palliating, lessening, delaying or decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival. In reference to cancers or other unwanted cell proliferation, an effective amount comprises an amount sufficient to cause a tumor to shrink and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation. In some embodiments, an effective amount is an amount sufficient to delay development. In some embodiments, an effective amount is an amount sufficient to prevent or delay occurrence and/or recurrence. An effective amount can be administered in one or more administrations, in the case of cancer, the effective amount of the drug or composition may: (i) reduce the number of cancer cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and preferably stop cancer cell infiltration into peripheral organs; (iv) inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer. An effective dosage can be administered in one or more administrations. For purposes of this disclosure, an effective dosage of compound or a salt thereof, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. It is intended and understood that an effective dosage of a compound or salt thereof, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
[0028] As used herein, the term “individual” is a mammal, including humans. An individual includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate. In some embodiments, the individual is human. The individual (such as a human) may have advanced disease or lesser extent of disease, such as low tumor burden. In some embodiments, the individual is at an early stage of a proliferative disease (such as cancer). In some embodiments, the individual is at an advanced stage of a proliferative disease (such as an advanced cancer).
[0029] Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”.
[0030] It is understood that aspects and variations described herein also include “consisting” and/or “consisting essentially of’ aspects and variations.
Compounds
[0031] In one aspect, provided is a compound described in Table 1. Also provided are salts of compounds referred to herein, such as pharmaceutically acceptable salts. The disclosure also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms, and any tautomers or other forms of the compounds described.
[0032] A compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein. Compositions comprising a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, are provided, such as compositions of substantially pure compounds. In some embodiments, a composition containing a compound as detailed herein or a salt thereof is in substantially pure form. Unless otherwise stated, “substantially pure” intends a composition that contains no more than 35% impurity, wherein the impurity denotes a compound other than the compound comprising the majority of the composition or a salt thereof. In some embodiments, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 25%, 20%, 15%, 10%, or 5% impurity. In some embodiments, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 3%, 2%, 1% or 0.5% impurity. Table 1
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
[0033] In some embodiments, provided herein is a compound described in Table 1, or a tautomer thereof, or a salt of any of the foregoing, and uses thereof. In some embodiments, provided herein is a compound described in Table 1 or a pharmaceutically acceptable salt thereof.
[0034] The embodiments and variations described herein are suitable for any compound described in Table 1 detailed herein, where applicable.
[0035] Representative examples of compounds detailed herein, including intermediates and final compounds according to the present disclosure are depicted herein. It is understood that in one aspect, any of the compounds may be used in the methods detailed herein, including, where applicable, intermediate compounds that may be isolated and administered to an individual.
[0036] The compounds depicted herein may be present as salts even if salts are not depicted and it is understood that the present disclosure embraces all salts and solvates of the compounds depicted here, as well as the non-salt and non-solvate form of the compound, as is well understood by the skilled artisan. In some embodiments, the salts of the compounds provided herein are pharmaceutically acceptable salts. Where one or more tertiary amine moiety is present in the compound, the N-oxides are also provided and described.
[0037] Where tautomeric forms may be present for any of the compounds described herein, each and every tautomeric form is intended even though only one or some of the tautomeric forms may be explicitly depicted. The tautomeric forms specifically depicted may or may not be the predominant forms in solution or when used according to the methods described herein.
[0038] The present disclosure also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms of the compounds described, such as the compounds of Table 1. The structure or name is intended to embrace all possible stereoisomers of a compound depicted, and each unique stereoisomer has a compound number bearing a suffix “a”, “b”, etc. All forms of the compounds are also embraced by the disclosure, such as crystalline or non-crystalline forms of the compounds. Compositions comprising a compound of the disclosure are also intended, such as a composition of substantially pure compound, including a specific stereochemical form thereof, or a composition comprising mixtures of compounds of the disclosure in any ratio, including two or more stereochemical forms, such as in a racemic or non-racemic mixture.
[0039] The disclosure also intends isotopically-labeled and/or isotopically-enriched forms of compounds described herein. The compounds herein may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. In some embodiments, the compound is isotopically-labeled, such as an isotopically-labeled derivative of a compound disclosed herein or variations thereof described herein, where a fraction of one or more atoms are replaced by an isotope of the same element. Exemplary isotopes that can be incorporated into compounds of the disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, chlorine, such as 2H, 3H, nC, 13C, 14C, 13N, 15O, 17O, 32P, 35S, 18F, 36C1. Certain isotope labeled compounds (e.g. 3H and 14C) is useful in compound or substrate tissue distribution studies. Incorporation of heavier isotopes such as deuterium (2H) can afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life, or reduced dosage requirements and, hence may be preferred in some instances.
[0040] Isotopically-labeled compounds of the present disclosure can generally be prepared by standard methods and techniques known to those skilled in the art or by procedures similar to those described in the accompanying Examples substituting appropriate isotopically-labeled reagents in place of the corresponding non-labeled reagent.
[0041] The disclosure also includes any or all metabolites of any of the compounds described. The metabolites may include any chemical species generated by a biotransformation of any of the compounds described, such as intermediates and products of metabolism of the compound, such as would be generated in vivo following administration to a human.
[0042] Articles of manufacture comprising a compound described herein, or a salt or solvate thereof, in a suitable container are provided. The container may be a vial, jar, ampoule, preloaded syringe, i.v. bag, and the like. [0043] Preferably, the compounds detailed herein are orally bioavailable. However, the compounds may also be formulated for parenteral (e.g., intravenous) administration.
[0044] One or several compounds described herein can be used in the preparation of a medicament by combining the compound or compounds as an active ingredient with a pharmacologically acceptable carrier, which are known in the art. Depending on the therapeutic form of the medication, the carrier may be in various forms. In one variation, the manufacture of a medicament is for use in any of the methods disclosed herein, e.g., for the treatment of cancer.
General synthetic methods
[0045] The compounds of the disclosure may be prepared by a number of processes as generally described below and more specifically in the Examples hereinafter (such as the schemes provided in the Examples below).
[0046] Where it is desired to obtain a particular enantiomer of a compound, this may be accomplished from a corresponding mixture of enantiomers using any suitable conventional procedure for separating or resolving enantiomers. Thus, for example, diastereomeric derivatives may be produced by reaction of a mixture of enantiomers, e.g., a racemate, and an appropriate chiral compound. The diastereomers may then be separated by any convenient means, for example by crystallization and the desired enantiomer recovered. In another resolution process, a racemate may be separated using chiral High Performance Liquid Chromatography. Alternatively, if desired a particular enantiomer may be obtained by using an appropriate chiral intermediate in one of the processes described.
[0047] Chromatography, recrystallization and other conventional separation procedures may also be used with intermediates or final products where it is desired to obtain a particular isomer of a compound or to otherwise purify a product of a reaction.
[0048] Solvates of a compound provided herein or a salt thereof are also contemplated. Solvates contain either stoichiometric or non- stoichiometric amounts of a solvent, and are often formed during the process of crystallization. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol.
[0049] In some embodiments, compounds disclosed herein are synthesized according to Scheme 1 to Scheme 5. Scheme 1
Figure imgf000034_0002
Scheme 3
Figure imgf000034_0001
Scheme 4
Figure imgf000035_0002
In Schemes 1-5:
Y is hydrogen or R4; m is 0, 1, 2, or 3; n is 0, 1, 2, 3, or 4;
R1 is independently F, Cl, or methyl;
R2 is Ci-C6 alkyl, C3-C6 cycloalkyl or -(C1-C3 alkylene)CF3;
Figure imgf000035_0001
wherein:
Figure imgf000036_0001
indicates an aromatic ring;
M1 is CH, CR3b or N;
M2 is CH, CR3b, N, or absent;
M3 is CH, CR3b, N, O, or S;
M4 is CH, CR3b, N, O or S,
R3a is -CN, -OR17, C3-C6 cycloalkyl optionally substituted by Ci-
Ce haloalkyl, -(C1-C3 alkylene)OH, -OH or -CN, 3- to 6-membered heterocyclyl substituted by halogen or -OH, or Ci-Ce alkyl optionally substituted by C3-C6 cycloalkyl, halogen, -OH or -CN, provided that when R3ais Ci-Ce alkyl optionally substituted by halogen, -OH or -CN, C3-C6 cycloalkyl substituted by -(C1-C3 alkylene)OH or -OH, or 3- to 6-membered heterocyclyl substituted by halogen or -OH then at least one of M1, M2, M3, and M4 is CR3b;
R3b is halogen or -CN; each R4 is independently oxo, Ci-Ce alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -C(O)R17, -C(O)OR17, -C(O)NR17R18, -CN, -Si(Ci-C6 alkyl)3, -OR17, -NR17R18, -OC(O)NR17R18, -NR17C(O)R18, -S(O)2R17, -NR17S(O)2R18, -S(O)2NR17R18, C3- Ce cycloalkyl, 3- to 6-membered heterocyclyl, -(C1-C3 alkylene)CN, -(C1-C3 alkylene)OR17, -(C1-C3 alkylene)NR17R18, -(C1-C3 alkylene)CF3, -(C1-C3 alkylene)C(O)R17, -(Ci- C3 alkylene)C(O)NR17R18, -(C1-C3 alkylene)NR17C(O)R18, -(C1-C3 alkylene)S(O)2R17, -(Ci- C3 alkylene)NR17S(O)2R18, -(C1-C3 alkylene)S(O)2NR17R18, -(C1-C3 alkylene)(C3-C6 cycloalkyl) or -(C1-C3 alkylene)(3- to 6-membered heterocyclyl) , wherein each R4 is independently optionally substituted by halogen, oxo, -OR19, -NR19R20, or -C(O)R19, or two R4, when bound to the same carbon are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, each is optionally substituted by R19; each R17, R18, R19, and R20 is independently hydrogen, C3-C6 cycloalkyl, 3-6 membered heterocyclyl or Ci-Ce alkyl, each of which is optionally substituted by halogen, oxo or -OH, or R17 and R18 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or -OH. Particular examples are provided in the Example Section below. Pharmaceutical Compositions and Formulations
[0050] Pharmaceutical compositions of any of the compounds detailed herein are embraced by this disclosure. Thus, the present disclosure includes pharmaceutical compositions comprising a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier or excipient. In one aspect, the pharmaceutically acceptable salt is an acid addition salt, such as a salt formed with an inorganic or organic acid. Pharmaceutical compositions may take a form suitable for oral, buccal, parenteral, nasal, topical or rectal administration or a form suitable for administration by inhalation.
[0051] A compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein. Compositions comprising a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, are provided, such as compositions of substantially pure compounds. In some embodiments, a composition containing a compound as detailed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is in substantially pure form.
[0052] In one variation, the compounds herein are synthetic compounds prepared for administration to an individual. In another variation, compositions are provided containing a compound in substantially pure form. In another variation, the present disclosure embraces pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier. In another variation, methods of administering a compound are provided. The purified forms, pharmaceutical compositions and methods of administering the compounds are suitable for any compound or form thereof detailed herein.
[0053] A compound detailed herein or salt thereof may be formulated for any available delivery route, including an oral, mucosal (e.g., nasal, sublingual, vaginal, buccal or rectal), parenteral (e.g., intramuscular, subcutaneous or intravenous), topical or transdermal delivery form. A compound or salt thereof may be formulated with suitable carriers to provide delivery forms that include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultices), pastes, powders, dressings, creams, solutions, patches, aerosols (e.g., nasal spray or inhalers), gels, suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions or water-in-oil liquid emulsions), solutions and elixirs.
[0054] One or several compounds described herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be used in the preparation of a formulation, such as a pharmaceutical formulation, by combining the compound or compounds, or a salt thereof, as an active ingredient with a pharmaceutically acceptable carrier, such as those mentioned above. Depending on the therapeutic form of the system (e.g., transdermal patch vs. oral tablet), the carrier may be in various forms. In addition, pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants. Formulations comprising the compound may also contain other substances which have valuable therapeutic properties. Pharmaceutical formulations may be prepared by known pharmaceutical methods. Suitable formulations can be found, e.g., in Remington’s Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, PA, 20th ed. (2000), which is incorporated herein by reference.
[0055] Compounds as described herein may be administered to individuals in a form of generally accepted oral compositions, such as tablets, coated tablets, and gel capsules in a hard or in soft shell, emulsions or suspensions. Examples of carriers, which may be used for the preparation of such compositions, are lactose, corn starch or its derivatives, talc, stearate or its salts, etc. Acceptable carriers for gel capsules with soft shell are, for instance, plant oils, wax, fats, semisolid and liquid poly-ols, and so on. In addition, pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants.
[0056] Any of the compounds described herein can be formulated in a tablet in any dosage form described, for example, a compound as described herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be formulated as a 10 mg tablet.
[0057] Compositions comprising a compound provided herein are also described. In one variation, the composition comprises a compound or salt thereof and a pharmaceutically acceptable carrier or excipient. In another variation, a composition of substantially pure compound is provided. Methods of Use
[0058] Compounds and compositions detailed herein, such as a pharmaceutical composition containing a compound described in Table 1 provided herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier or excipient, may be used in methods of administration and treatment as provided herein. The compounds and compositions may also be used in in vitro methods, such as in vitro methods of administering a compound or composition to cells for screening purposes and/or for conducting quality control assays.
[0059] Provided herein is a method of treating a disease in an individual comprising administering an effective amount of a compound described in Table 1 or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, to the individual. Further provided herein is a method of treating a proliferative disease in an individual, comprising administering an effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, to the individual. Also provided herein is a method of treating cancer in an individual comprising administering an effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, to the individual. In some embodiments, the compound is administered to the individual according to a dosage and/or method of administration described herein.
[0060] In some embodiments, the cancer in the individual has one or more TP53 gene mutations or expresses mutant p53. In some embodiments, the cancer in the individual that has one or more TP53 gene mutations or expresses mutant p53 is glioblastoma. TP53 is the human gene that encodes p53. In some embodiments, provided herein is a method of treating a cancer in an individual, comprising (a) selecting the individual for treatment based on (i) the presence of one or more mutations of the TP 53 gene in the cancer, or (ii) expression of mutant p53 in the cancer, and administering an effective amount of a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing,, to the individual. In some embodiments, the cancer is assayed for the expression of mutant p53. In some embodiments, the TP 53 gene of the cancer is sequenced to detect the one or more mutations. In some embodiments, the TP 53 gene is sequenced by biopsying the cancer and sequencing the TP53 gene from the biopsied cancer. In some embodiments, the TP53 gene is sequenced by sequencing circulating-tumor DNA (ctDNA) from the individual. [0061] In some embodiments, provided herein is a method of using a compound disclosed herein or any embodiment in the manufacture of a medicament for treatment of a disease. In some embodiments, provided herein is a method of using a compound disclosed herein or any embodiment in the manufacture of a medicament for treatment of cancer.
[0062] In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is used to treat an individual having a proliferative disease, such as cancer as described herein. In some embodiments, the individual is at risk of developing a proliferative disease, such as cancer. In some of these embodiments, the individual is determined to be at risk of developing cancer based upon one or more risk factors. In some of these embodiments, the risk factor is a family history and/or gene associated with cancer.
[0063] The present compounds or salts thereof are believed to be effective for treating a variety of diseases and disorders. For example, in some embodiments, the present compositions may be used to treat a proliferative disease, such as cancer. In some embodiments the cancer is a solid tumor. In some embodiments the cancer is any of adult and pediatric oncology, myxoid and round cell carcinoma, locally advanced tumors, metastatic cancer, human soft tissue sarcomas, including Ewing's sarcoma, cancer metastases, including lymphatic metastases, squamous cell carcinoma, particularly of the head and neck, esophageal squamous cell carcinoma, oral carcinoma, blood cell malignancies, including multiple myeloma, leukemias, including acute lymphocytic leukemia, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, and hairy cell leukemia, effusion lymphomas (body cavity based lymphomas), thymic lymphoma, cutaneous T cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cancer of the adrenal cortex, ACTH-producing tumors, lung cancer, including small cell carcinoma and nonsmall cell cancers, breast cancer, including small cell carcinoma and ductal carcinoma, gastrointestinal cancers, including stomach cancer, colon cancer, colorectal cancer, polyps associated with colorectal neoplasia, pancreatic cancer, liver cancer, urological cancers, including bladder cancer, including primary superficial bladder tumors, invasive transitional cell carcinoma of the bladder, and muscle-invasive bladder cancer, prostate cancer, malignancies of the female genital tract, including ovarian carcinoma, primary peritoneal epithelial neoplasms, cervical carcinoma, uterine endometrial cancers, vaginal cancer, cancer of the vulva, uterine cancer and solid tumors in the ovarian follicle, malignancies of the male genital tract, including testicular cancer and penile cancer, kidney cancer, including renal cell carcinoma, brain cancer, including intrinsic brain tumors, neuroblastoma, astrocytic brain tumors, gliomas, glioblastoma, metastatic tumor cell invasion in the central nervous system, bone cancers, including osteomas and osteosarcomas, skin cancers, including melanoma, tumor progression of human skin keratinocytes, squamous cell cancer, thyroid cancer, retinoblastoma, neuroblastoma, peritoneal effusion, malignant pleural effusion, mesothelioma, Wilms's tumors, gall bladder cancer, trophoblastic neoplasms, hemangiopericytoma, and Kaposi's sarcoma.
[0064] In some embodiments, the compounds and compositions described herein suppress G2-M checkpoint in a cell (such as a cancer cell). In some embodiments, the cancer cell is a cancer cell from any of the cancer types described herein. Suppression of the G2-M DNA damage checkpoint results in premature mitosis of the cell, and consequently apoptosis. In some embodiments, provided herein is a method of suppressing the G2-M DNA damage checkpoint in a cell comprising administering an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, to the cell. In some embodiments, the G2-M DNA damage checkpoint is suppressed in about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more of cells in a cell population. In some embodiments, the G2-M DNA damage checkpoint is suppressed in up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, or up to about 80% of cells in the cell population.
[0065] In some embodiments, provided herein is a method of inducing premature mitosis in a cell comprising administering an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, to the cell. In some embodiments, premature mitosis is induced in about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more of cells in a cell population. In some embodiments, premature mitosis is induced in up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, or up to about 80% of cells in the cell population.
[0066] In some embodiments, provided herein is a method of inducing apoptosis in a cell comprising administering an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, to the cell. In some embodiments, apoptosis is induced in about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more of cells in a cell population. In some embodiments, apoptosis is induced in up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, or up to about 80% of cells in the cell population.
[0067] In some embodiments, provided herein is a method of inhibiting Weel in a cell comprising administering an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof, to the cell. In some embodiments, Weel is inhibited by about 10% or more, about 20% or more, about 30% or more, about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 75% or more, about 80% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more. In some embodiments, Weel is inhibited up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, up to about 80%, up to about 70%, or up to about 60%. In some embodiments, the activity of Weel is measured according to a kinase assay.
[0068] In some embodiments, provided herein is a method of inhibiting Weel comprising contacting Weel with an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof. In some embodiments, a compound disclosed herein or a pharmaceutically acceptable salt thereof binds to Weel with an IC50 of less than 1 pM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 50 nM, less than 10 nM, less than 5 nM, less than 1 nM, or less than 0.5 nM. In some embodiments, a compound disclosed herein or a pharmaceutically acceptable salt thereof binds to Weel with an IC50 between 0.1 nM and 1 nM, between 1 nM and 5 nM, between 5 nM and 10 nM, between 10 nM and 50 nM, between 50 nM and 100 nM, between 100 nM and 200 nM, between 200 nM and 300 nM, between 300 nM and 400 nM, between 400 nM and 500 nM, between 500 nM and 600 nM, between 600 nM and 700 nM, between 700 nM and 800 nM, between 800 nM and 900 nM, or between 900 nM and 1 pM. In some embodiments, the IC50 is measured according to a kinase assay. In some embodiments, the IC50 is measured according to a cell cytotoxicity assay.
[0069] In some embodiments, provided herein is a method of inhibiting the proliferation of a cell, comprising contacting the cell with an effective amount of a compound disclosed herein or a pharmaceutically acceptable salt thereof. In some embodiments, a compound disclosed herein or a pharmaceutically acceptable salt thereof is effective in inhibiting the proliferation of the cell with an IC50 of less than 5 pM, less than 2 pM, less than 1 pM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, or less than 50 nM. In some embodiments, a compound disclosed herein or a pharmaceutically acceptable salt is effective in inhibiting the proliferation of the cell with an IC50 between 10 nM and 20 nM, between 20 nM and 50 nM, between 50 nM and 100 nM, between 100 nM and 500 nM, between 500 nM and 1 pM, between 1 pM and 2 pM, or between 2 pM and 5 pM. In some embodiments, the IC50 is measured according to a cell proliferation assay.
Combination Therapy
[0070] As provided herein, the presently disclosed compounds, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, may activate the immune system, for example by inducing apoptosis or suppressing mitosis of cancer cells. Accordingly, the present compounds, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, may be used in combination with other anti-cancer agents to enhance tumor immunotherapy. In some embodiments, provided herein is a method of treating a disease in an individual comprising administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and an additional therapeutic agent to the individual. In some embodiments, the disease is a proliferative disease such as cancer.
[0071] In some embodiments, the additional therapeutic agent is a cancer immunotherapy agent. In some embodiments, the additional therapeutic agent is a chemotherapeutic agent. In some embodiments, the additional therapeutic agent is an immuno stimulatory agent. In some embodiments, the additional therapeutic agent targets a checkpoint protein (for example an immune checkpoint inhibitor). In some embodiments, the additional therapeutic agent is effective to stimulate, enhance or improve an immune response against a tumor. In some embodiments, the additional chemotherapeutic agent is a DNA alkylating agent, a platinumbased chemotherapeutic agent, a kinase inhibitor or a DNA damage repair (DDR) pathway inhibitor. In some embodiments, the additional chemotherapeutic agent is a DNA alkylating agent. In some embodiments, the additional chemotherapeutic agent is a platinum-based chemotherapeutic agent. In some embodiments, the additional chemotherapeutic agent is a kinase inhibitor. In some embodiments, the additional chemotherapeutic agent is a DNA damage repair (DDR) pathway inhibitor.
[0072] In another aspect, provided herein is a combination therapy for the treatment of a disease, such as cancer. In some embodiments, provided herein is a method of treating a disease in an individual comprising administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, in combination with a radiation therapy.
[0073] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an additional chemotherapeutic agent. In some embodiments, the chemotherapeutic agent is a kinase inhibitor or an agent that inhibits one or more DNA damage repair (DDR) pathways. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the additional chemotherapeutic agent. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the additional chemotherapeutic agent.
[0074] Examples of chemotherapeutic agents that can be used in combination with a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, include DNA-targeted agents, a DNA alkylating agent (such as cyclophosphamide, mechlorethamine, chlorambucil, melphalan, dacarbazine, temozolomide or nitrosoureas), a topoisomerase inhibitor (such as a Topoisomerase I inhibitor (e.g., irinotecan or topotecan) or a Topoisomerase II inhibitor (e.g., etoposide or teniposide)), an anthracycline (such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, or valrubicin), a histone deacetylase inhibitor (such as vorinostat or romidepsin), a bromodomain inhibitor, other epigenetic inhibitors, a taxane (such as paclitaxel or docetaxel), a kinase inhibitor (such as bortezomib, erlotinib, gefitinib, imatinib, vemurafenib, or vismodegib), an anti- angiogenic inhibitor, a nucleotide analog or precursor analog (such as azacitidine, azathioprine, capecitabine, cytarabine, doxifluridine, 5-fluorouracil, gemcitabine, hydroxyurea, mercaptopurine, methotrexate, or tioguanine), or a platinum-based chemotherapeutic agent (such as cisplatin, carboplatin, or oxaliplatin), pemetrexed, or a combination thereof. In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a kinase inhibitor (such as bortezomib, erlotinib, gefitinib, imatinib, vemurafenib, or vismodegib). In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the kinase inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the kinase inhibitor.
[0075] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DNA damaging agent. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the DNA damaging agent. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the DNA damaging agent.
[0076] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DNA alkylating agent (such as cyclophosphamide, mechlorethamine, chlorambucil, melphalan, dacarbazine, temozolomide or nitrosoureas). In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the DNA alkylating agent. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the DNA alkylating agent.
[0077] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a topoisomerase inhibitor (such as a Topoisomerase I inhibitor (e.g., irinotecan or topotecan) or a Topoisomerase II inhibitor (e.g., etoposide or teniposide)). In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the topoisomerase inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the topoisomerase inhibitor.
[0078] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an anthracycline (such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, or valrubicin). In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the anthracycline. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the anthracycline.
[0079] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a histone deacetylase inhibitor (such as vorinostat or romidepsin). In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the histone deacetylase inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the histone deacetylase inhibitor.
[0080] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a taxane (such as paclitaxel or docetaxel). In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the taxane. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the taxane.
[0081] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a nucleotide analog or precursor analog (such as azacitidine, azathioprine, capecitabine, cytarabine, doxifluridine, 5-fluorouracil, gemcitabine, hydroxyurea, mercaptopurine, methotrexate, or tioguanine). In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the nucleotide analog or precursor analog. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the nucleotide analog or precursor analog.
[0082] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a platinum-based chemotherapeutic agent (such as cisplatin, carboplatin, or oxaliplatin). In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the platinum-based chemotherapeutic agent. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the platinum-based chemotherapeutic agent.
[0083] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of pemetrexed. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the pemetrexed. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the pemetrexed.
[0084] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DDR pathway inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the DDR pathway inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the DDR pathway inhibitor. Examples of inhibitors of the DDR pathway include poly(ADP-ribose) polymerase (PARP) inhibitors (such as olaparib, rucaparib, niraparib, or talazoparib), ataxia telangiectasia mutated (ATM) protein inhibitors, ataxia telangiectasia and Rad3 -related (ATR) protein inhibitors, checkpoint kinase 1 (Chkl) inhibitors, or combinations thereof. [0085] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a PARP inhibitor (such as olaparib, rucaparib, niraparib, or talazoparib). In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the PARP inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the PARP inhibitor.
[0086] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATM protein inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the ATM protein inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the ATM protein inhibitor.
[0087] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATR protein inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the ATR protein inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the ATR protein inhibitor. [0088] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound disclosed herein or any embodiment, variation or aspect thereof or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an Chkl inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered prior to, after, or simultaneously co-administered with the Chkl inhibitor. In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the Chkl inhibitor.
[0089] In another aspect, provided herein is a combination therapy in which a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is coadministered (which may be separately or simultaneously) with one or more additional agents that are effective in stimulating immune responses to thereby further enhance, stimulate or upregulate immune responses in a subject. For example, provided is a method for stimulating an immune response in a subject comprising administering to the subject a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and one or more immuno stimulatory antibodies, such as an anti-PD-1 antibody, an anti-PD-Ll antibody and/or an anti-CTLA-4 antibody, such that an immune response is stimulated in the subject, for example to inhibit tumor growth. In one embodiment, the subject is administered a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-PD-1 antibody. In another embodiment, the subject is administered a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-PD-Ll antibody. In yet another embodiment, the subject is administered a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-CTLA-4 antibody. In another embodiment, the immuno stimulatory antibody (e.g., anti-PD-1, anti-PD-Ll and/or anti-CTLA-4 antibody) is a human antibody. Alternatively, the immunostimulatory antibody can be, for example, a chimeric or humanized antibody (e.g., prepared from a mouse anti-PD-1, anti-PD-Ll and/or anti-CTLA-4 antibody).
[0090] In one embodiment, the present disclosure provides a method for treating a proliferative disease (e.g., cancer), comprising administering a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-PD-1 antibody to a subject. In further embodiments, a compound disclosed herein or , or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered at a subtherapeutic dose, the anti-PD-1 antibody is administered at a subtherapeutic dose, or both are administered at a subtherapeutic dose. In another embodiment, the present disclosure provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a subtherapeutic dose of anti- PD-1 antibody to a subject. In certain embodiments, the subject is human. In certain embodiments, the anti-PD-1 antibody is a human sequence monoclonal antibody.
[0091] In one embodiment, the present disclosure provides a method for treating a hyperproliferative disease (e.g., cancer), comprising administering a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and an anti-PD-Ll antibody to a subject. In further embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered at a subtherapeutic dose, the anti-PD-Ll antibody is administered at a subtherapeutic dose, or both are administered at a subtherapeutic dose. In another embodiment, the present disclosure provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a subtherapeutic dose of anti- PD-Ll antibody to a subject. In certain embodiments, the subject is human. In certain embodiments, the anti-PD-Ll antibody is a human sequence monoclonal antibody.
[0092] In certain embodiments, the combination of therapeutic agents discussed herein can be administered concurrently as a single composition in a pharmaceutically acceptable carrier, or concurrently as separate compositions each in a pharmaceutically acceptable carrier. In another embodiment, the combination of therapeutic agents can be administered sequentially. For example, an anti-CTLA-4 antibody and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be administered sequentially, such as anti-CTLA-4 antibody being administered first and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, second, or a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, being administered first and anti-CTLA-4 antibody second. Additionally or alternatively, an anti-PD-1 antibody and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be administered sequentially, such as anti-PD-1 antibody being administered first and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, second, or a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, being administered first and anti-PD-1 antibody second. Additionally or alternatively, an anti-PD-Ll antibody and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be administered sequentially, such as anti-PD-Ll antibody being administered first and a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, second, or a compound disclosed, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, being administered first and anti- PD-Ll antibody second.
[0093] Furthermore, if more than one dose of the combination therapy is administered sequentially, the order of the sequential administration can be reversed or kept in the same order at each time point of administration, sequential administrations can be combined with concurrent administrations, or any combination thereof.
[0094] Optionally, the combination of a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be further combined with an immunogenic agent, such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines.
[0095] A compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can also be further combined with standard cancer treatments. For example, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be effectively combined with chemotherapeutic regimes. In these instances, it is possible to reduce the dose of other chemotherapeutic reagent administered with the combination of the instant disclosure (Mokyr et al. (1998) Cancer Research 58: 5301-5304). Other combination therapies with a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, include radiation, surgery, or hormone deprivation. Angiogenesis inhibitors can also be combined with a compound disclosed herein or a salt thereof. Inhibition of angiogenesis leads to tumor cell death, which can be a source of tumor antigen fed into host antigen presentation pathways.
[0096] In another example, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be used in conjunction with anti-neoplastic antibodies. By way of example and not wishing to be bound by theory, treatment with an anti-cancer antibody or an anti-cancer antibody conjugated to a toxin can lead to cancer cell death (e.g., tumor cells) which would potentiate an immune response mediated by CTLA-4, PD-1, PD-L1 or a compound disclosed herein or a salt thereof. In an exemplary embodiment, a treatment of a hyperproliferative disease (e.g., a cancer tumor) can include an anti-cancer antibody in combination with a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and anti-CTLA-4 and/or anti-PD-1 and/or anti-PD-Ll antibodies, concurrently or sequentially or any combination thereof, which can potentiate anti-tumor immune responses by the host. Other antibodies that can be used to activate host immune responsiveness can be further used in combination with a compound disclosed herein or a salt thereof.
[0097] In some embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, can be combined with an anti- CD73 therapy, such as an anti-CD73 antibody.
[0098] In yet further embodiments, a compound disclosed herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, is administered in combination with another Weel inhibitor.
Dosing and Method of Administration
[0099] The dose of a compound administered to an individual (such as a human) may vary with the particular compound or salt thereof, the method of administration, and the particular disease, such as type and stage of cancer, being treated. In some embodiments, the amount of the compound or salt thereof is a therapeutically effective amount.
[0100] The effective amount of the compound may in one aspect be a dose of between about 0.01 and about 100 mg/kg. Effective amounts or doses of the compounds of the disclosure may be ascertained by routine methods, such as modeling, dose escalation, or clinical trials, taking into account routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the disease to be treated, the subject’s health status, condition, and weight. An exemplary dose is in the range of about from about 0.7 mg to 7 g daily, or about 7 mg to 350 mg daily, or about 350 mg to 1.75 g daily, or about 1.75 to 7 g daily.
[0101] Any of the methods provided herein may in one aspect comprise administering to an individual a pharmaceutical composition that contains an effective amount of a compound provided herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable excipient.
[0102] A compound or composition of the disclosure may be administered to an individual in accordance with an effective dosing regimen for a desired period of time or duration, such as at least about one month, at least about 2 months, at least about 3 months, at least about 6 months, or at least about 12 months or longer, which in some variations may be for the duration of the individual’s life. In one variation, the compound is administered on a daily or intermittent schedule. The compound can be administered to an individual continuously (for example, at least once daily) over a period of time. The dosing frequency can also be less than once daily, e.g., about a once weekly dosing. The dosing frequency can be more than once daily, e.g., twice or three times daily. The dosing frequency can also be intermittent, including a ‘drug holiday’ (e.g., once daily dosing for 7 days followed by no doses for 7 days, repeated for any 14 day time period, such as about 2 months, about 4 months, about 6 months or more). Any of the dosing frequencies can employ any of the compounds described herein together with any of the dosages described herein.
[0103] The compounds provided herein, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, may be administered to an individual via various routes, including, e.g., intravenous, intramuscular, subcutaneous, oral and transdermal. A compound provided herein can be administered frequently at low doses, known as 'metronomic therapy,' or as part of a maintenance therapy using compound alone or in combination with one or more additional drugs. Metronomic therapy or maintenance therapy can comprise administration of a compound provided herein in cycles. Metronomic therapy or maintenance therapy can comprise intra-tumoral administration of a compound provided herein.
[0104] In one aspect, the disclosure provides a method of treating cancer in an individual by parenterally administering to the individual (e.g., a human) an effective amount of a compound or salt thereof. In some embodiments, the route of administration is intravenous, intra-arterial, intramuscular, or subcutaneous. In some embodiments, the route of administration is oral. In still other embodiments, the route of administration is transdermal.
[0105] The disclosure also provides compositions (including pharmaceutical compositions) as described herein for the use in treating, preventing, and/or delaying the onset and/or development of cancer and other methods described herein. In certain embodiments, the composition comprises a pharmaceutical formulation which is present in a unit dosage form.
[0106] Also provided are articles of manufacture comprising a compound of the disclosure or a salt thereof, composition, and unit dosages described herein in suitable packaging for use in the methods described herein. Suitable packaging is known in the art and includes, for example, vials, vessels, ampules, bottles, jars, flexible packaging and the like. An article of manufacture may further be sterilized and/or sealed.
Kits
[0107] The present disclosure further provides kits for carrying out the methods of the disclosure, which comprises one or more compounds described herein or a composition comprising a compound described herein. The kits may employ any of the compounds disclosed herein. In one variation, the kit employs a compound described herein or a salt thereof. The kits may be used for any one or more of the uses described herein, and, accordingly, may contain instructions for the treatment of cancer.
[0108] Kits generally comprise suitable packaging. The kits may comprise one or more containers comprising any compound described herein. Each component (if there is more than one component) can be packaged in separate containers or some components can be combined in one container where cross-reactivity and shelf life permit.
[0109] The kits may be in unit dosage forms, bulk packages (e.g., multi-dose packages) or sub-unit doses. For example, kits may be provided that contain sufficient dosages of a compound as disclosed herein and/or an additional pharmaceutically active compound useful for a disease detailed herein to provide effective treatment of an individual for an extended period, such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 7 months, 8 months, 9 months, or more. Kits may also include multiple unit doses of the compounds and instructions for use and be packaged in quantities sufficient for storage and use in pharmacies (e.g., hospital pharmacies and compounding pharmacies). [0110] The kits may optionally include a set of instructions, generally written instructions, although electronic storage media (e.g.. magnetic diskette or optical disk) containing instructions are also acceptable, relating to the use of component(s) of the methods of the present disclosure. The instructions included with the kit generally include information as to the components and their administration to an individual.
[0111] The disclosure can be further understood by reference to the following examples, which are provided by way of illustration and are not meant to be limiting.
EXAMPLES
Example S-l: Synthesis ofl-(4-fluoro-3-methylphenyl)-2-isopropyl-6-(l,2,3,4- tetrahydroisoquinolin-7-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one (Compound No. 1-1)
Figure imgf000056_0001
[0112] Step-1: Synthesis of l-(4-fhioro-3-methylphenyl)-2-ilsopropyl-6-(methylthio)- lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of 2-isopropyl-6- (methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (250 mg, 1.11 mmol, 1.0 eq) and 4-fluoro-3-methylphenylboronic acid (206 mg, 1.34 mmol, 1.2 eq) in acetonitrile (10 mL) was added 2,2-bipyridine (260 mg, 1.66 mmol, 1.5 eq), copper acetate (400 mg, 2.22 mmol, 2.0 eq) and Na2COs (552 mg, 3.3 mmol, 3.0 eq) and DCM (30 mL). The reaction mixture was stirred at RT for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by silica gel column chromatography to afford the desired product (50 mg, 13.7%). LCMS: 333.1 (M+l)+.
[0113] Step-2: Synthesis of tert-butyl 7-(l-(4-fhioro-3-methylphenyl)-2-isopropyl-3- oxo-2, 3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline- 2(lH)-carboxylate: To a stirred solution l-(4-fluoro-3-methylphenyl)-2-isopropyl-6- (methylthio)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one (50 mg, 0.15 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (60 mg, 0.3 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert-Butyl 7-amino-3,4-dihydroisoquinoline-2(lH)-carboxylate (41 mg, 0.16 mmol, 1.1 eq) and Na2COs (64 mg, 0.6 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (50 mg, 62.6%). LCMS: 533.4 (M+l) +.
[0114] Step-3: Synthesis of l-(4-fhioro-3-methylphenyl)-2-isopropyl-6-(l, 2,3,4- tetrahydroisoquinolin-7-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one: tert-Butyl 7- (l-(4-fluoro-3-methylphenyl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin- 6-ylamino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (50 mg, 0.09 mmol, 1.0 eq) was dissolved in dioxane (0.5 mL), followed by dropwise addition of 4.0 M-HC1 (0.5 mL) and allowed to stir at RT for 1 h. After completion of reaction, the reaction mixture was filtered and triturated with diethyl ether to afford title compound (30 mg, 75%). LCMS: 433.5 (M+l)+ ; 'H NMR (400 MHz, DMSO- 6): d ppm 10.25 (br s, 1H) 9.36 (br s, 1H) 8.82 (s, 1 H) 7.64 (br s, 1H) 7.43 - 7.58 (m, 3 H) 7.27 - 7.43 (m, 1H) 7.11 (d, J = 7.5 Hz, 1H) 3.86 - 4.16 (m, 3H) 3.34 (br s, 2H) 2.93 (br s, 2H) 2.33 (s, 3H) 1.26 (d, J = 6.5 Hz, 6H).
Example S-2: Synthesis of 6-((2-(cyclopropylmethyl)-l ,2,3,4-tetrahydroisoquinolin-6- yl)amino)-l-(5-fluoro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-l,2-dihydro-3H- pyrazolo[3,4-d]pyrimidin-3-one (Compound No. 1.2)
Figure imgf000058_0001
[0115] To a stirred solution l-(5-fluoro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one (100 mg, 0.21 mmol , 1.0 eq) and cyclopropanecarbaldehyde (0.023 mL, 0.315 mmol, 1.5 eq) in IPA (2 mL) was added one drop of acetic acid, followed by addition of sodium cyanoborohydride (52 mg, 0.86 mmol, 4.0 eq) and the reaction mixture was stirred at RT for overnight. After completion of reaction, the reaction mixture was diluted with sodium bicarbonate and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by reverse phase chromatography to afford the desired compound (54 mg, 48.5%). LCMS: 532.28 (M+l) +; 'H NMR (400 MHz, DMSO- 6): 3 ppm 10.18 (br s, 1H) 8.81 (s, 1 H) 7.93 - 8.06 (m, 1H) 7.78 - 7.89 (m, 1H) 7.55 (br s, 1H) 7.40 (d, J = 7.0 Hz, 1H) 7.01 (d, J = 9.2 Hz, 1H) 5.22 (s, 1 H) 4.14 - 4.24 (m, 1H) 3.59 (br s, 2H) 2.80 (br s, 2H) 2.38 (br s, 2H) 2.33 (br s, 1H) 1.52 (s, 6 H) 1.32 (d, J = 6.5 Hz, 6H) 0.53 (br s, 2H) 0.17 (br s, 2H).
Example S-3: Synthesis of l-(3-tert-butyl-5-fluorophenyl)-2-isopropyl-6-(l, 2,3,4- tetrahydroisoquinolin-7-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one ( Compound No. 1.3)
Figure imgf000059_0001
[0116] Step-1: Synthesis of 2-(3-(tert-butyl)-5-fhiorophenyl)-4,4,5,5-tetramethyl- l,3,2-dioxaborolane:To a stirred solution of l-bromo-3-(tert-butyl)-5-fluorobenzene (300 mg, 1.29 mmol, 1.0 eq) and 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (394.8 mg, 1.55 mmol, 1.2 eq) in dioxane (30 mL) was added KO Ac (252 mg, 2.59 mmol, 2.0 eq) at rt. The resulting mixture was purged with N2 for 10 min. followed by addition of Pd(dppf)Ch.DCM (21 mg, 0.026 mmol, 0.02 eq) and again purged with N2 for 10 min. The resulting mixture was stirred at 70°C for overnight. The progress of reaction was monitored by LCMS. The reaction mixture was diluted with water (50 mL) extracted with EtOAc (2x50 mL). The combined organic extracts were washed with water (50 mL), with brine (50 mL) dried over Na2SO4 and concentrated under reduced pressure to afford the desired compound (1.13 g). LCMS: 279.19 (M+l)+.
[0117] Step-2: Synthesis of l-(3-(tert-butyl)-5-fhiorophenyl)-2-isopropyl-6- (methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (580 mg, 2.59 mmol, 1.0 eq) and 2-(3-(tert-butyl)-5-fluorophenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (720 mg, 2.59 mmol, 1.0 eq) in (50 mL) of CH2CI2 was added Na2COs (23 mg, 7.77 mmol, 3.0 eq), CU(OAC)2 (2.09 g, 5.18 mmol, 2.0 eq), Py2 (1.80 g, 5.18 mmol, 2.0 eq), and the resulting mixture was stirred at RT for 72 h. After completion of reaction, the reaction mixture was diluted with water (50.0 mL) and extracted with EtOAc (50 mL x 2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (250 mg, 25.8%). LCMS: 377.14(M+1)+.
[0118] Step-3: Synthesis of tert-butyl 7-((l-(3-(tert-butyl)-5-fhiorophenyl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(3-(tert-butyl)-5- fluorophenyl)-2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (81 mg, 0.216 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (103 mg, 0.43 mmol, 2.0 eq) and allowed to stir at RT for 30 minutes. tert-Butyl 7-amino-3,4- dihydroisoquinoline-2(lH)-carboxylate (54 mg, 0.216 mmol, 1.0 eq) and Na2COs (92 mg, 0.86 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (20 mL x 2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (35 mg, 28.0%). LCMS: 575.31(M+1)+.
[0119] Step-4: Synthesis of 1 l-(3-(tert-butyl)-5-fhiorophenyl)-2-isopropyl-6- ((l,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3- one,: tert-Butyl 7-((l-(3-(tert-butyl)-5-fluorophenyl)-2-isopropyl-3-oxo-2,3-dihydro-lH- pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (35 mg, 0.06 mmol, 1.0 eq) was dissolved in 4.0 M-HC1 (0.5 mL) and allowed to stir at RT for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure and purified product was obtained by reverse phase chromatography (13 mg, 41.1%). LCMS: (475.25 M+l)+; 'H NMR (400 MHz, DMSO- 6): d ppm 10.20 (br s, 1H) 8.81 (s, 1 H) 8.21 (s, 1 H) 7.49 (br s, 1H) 7.34 - 7.46 (m, 1H) 7.16 - 7.34 (m, 2H) 6.91 - 7.03 (m, 1H) 3.98 - 4.11 (m, 1H) 3.90 (s, 2 H) 3.04 (t, J = 5.7 Hz, 2H) 2.59 - 2.79 (m, 2H) 1.26 - 1.34 (m, 15H).
Example S-4: Synthesis of7-((l-(5-fluoro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinolin-l(2H)-one (Compound No. 1.4)
Figure imgf000061_0001
[0120] To a stirred solution of l-(5-fluoro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (140 mg, 0.37 mmol, 1.0 eq) in (4.0 mL) of toluene was added m-CPBA (163.4 mg, 0.74 mmol, 2.0 eq) and allowed to stir at RT for 60 minutes. 7-Amino-3,4-dihydroisoquinolin-l(2H)-one (60.16 mg, 0.37 mmol, 1.0 eq) and Na2COs (156.8 mg, 1.48 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (30 mg, 16.5%). LCMS: 492.58 (M+l) +; 'H NMR (400 MHz, DMSO- 6): Jppm 10.39 (br s, 1H) 8.83 (s, 1H) 8.49 (br s, 1H) 7.87 - 8.16 (m, 3H) 7.66 (d, J = 8.3 Hz, 1H) 7.25 (d, J = 8.3 Hz, 1H) 5.22 (s, 1H) 4.09 - 4.27 (m, 1H) 3.37 (br s, 2H) 2.86 (t, J = 6.5 Hz, 2H) 1.52 (s, 6H) 1.29 - 1.42 (m, 6H).
Example S-5: Synthesis ofl-(5-fluoro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(2-(2- hydroxyacetyl)-l,2,3,4-tetrahydroisoquinolin-6-ylamino)-2-isopropyl-lH-pyrazolo[3,4- d]pyrimidin-3(2H)-one (Compound No. 1.5)
Figure imgf000061_0002
[0121] To a stirred solution of glycolic acid (22 mg, 0.28 mmol, 1.2 eq) in DMF (5 mL) was added EDCI (66 mg, 0.34 mmol, 1.5 eq) followed by HOBT (46 mg, 0.35 mmol, 1.5 eq) and stirred for 15 min. To this solution was added l-(5-fluoro-6-(2-hydroxypropan-2- yl)pyridin-2-yl)-2-isopropyl-6-(l,2,3,4-tetrahydroisoquinolin-6-ylamino)-lH-pyrazolo[3,4- d]pyrimidin-3(2H)-one (110 mg, 0.23 mmol, 1.0 eq) and DIPEA (0.2 mL, 1.15 mmol, 5.0 eq) and stirred at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude obtained was purified by reverse phase chromatography to furnish the title compound (32 mg, 23%). LCMS: 536.3 (M+l)+; 'H NMR (400 MHz, DMSO- 6): Jppm 10.24 (br s, 1H) 8.82 (s, 1H) 7.92 - 8.10 (m, 1H) 7.84 (d, J = 7.02 Hz, 1H) 7.62 (br s, 1H) 7.44 (br s, 1H) 7.13 (dd, J = 17.5, 8.3 Hz, 1H) 5.23 (s, 1H) 4.50-4.57 (m, 3H) 4.08 - 4.28 (m, 3H) 3.57-3.70 (m, 2H), 2.61-2.82 (m, 2H), 1.52 (s, 6H) 1.32 (d, J = 6.5 Hz, 6H).
Example S-6: Synthesis of4-(2-isopropyl-3-oxo-6-((l,2,3,4-tetrahydroisoquinolin-6- yl)amino)-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)-2-(trifluoromethyl)benzonitrile (Compound No. 1.6)
Figure imgf000062_0001
[0122] Step-1: Synthesis of 4-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-lH- pyrazolo[3,4-d]pyrimidin-l-yl)-2-(trifhioromethyl)benzonitrile: To a stirred solution of 2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (224 mg , 1.0 mmol, 1.0 eq) and (4-cyano-3-(trifluoromethyl)phenyl)boronic acid (214.9 mg, 1.0 mmol, 1.2 eq) in ACN (40 mL) was added 2,2-bipyridine (312.3 mg, 2.0 mmol, 2.0 eq), copper acetate (364 mg, 2.0 mmol, 2.0 eq) and Na2COs (318 mg, 3.0 mmol, 3.0 eq). The reaction mixture was stirred at RT for 48h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by silica gel column chromatography to afford the desired product (185 mg, 47.1%). LCMS: 394.39 (M+l)+.
[0123] Step-2: Synthesis of tert-butyl 6-((l-(4-cyano-3-(trifhioromethyl)phenyl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution 4-(2-isopropyl-6- (methylthio)-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)-2-
(trifluoromethyl)benzonitrile (137 mg, 0.34 mmol, 1.0 eq) in (4.0 mL) of toluene was added m-CPBA (155 mg, 0.69 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert-Butyl 6-amino- 3,4-dihydroisoquinoline-2(lH)-carboxylate (86.4 mg, 0.34 mmol, 1.0 eq) and Na2COs (148 mg, 1.39 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (60 mg, 29.0%). LCMS: 594.39 (M+l) +.
[0124] Step-3: Synthesis of 4-(2-isopropyl-3-oxo-6-((l,2,3,4-tetrahydroisoquinolin-6- yl)amino)-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)-2-
(trifhioromethyl)benzonitrile: tert-Butyl 6-((l-(4-cyano-3-(trifluoromethyl)phenyl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate (60 mg, 0.10 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HC1 (1 mL) and allowed to stir at RT for 1 h. After completion of reaction, the reaction mixture was filtered and purified by reverse phase purification to afford title compound (30 mg, 56.0%). LCMS: 494.5 (M+l)+; 'H NMR (400 MHz, DMSO-^6) 8 ppm 10.49 (br s, 1H) 9.10 (br s, 2H) 8.89 (br s, 1H) 8.36 (d, J = 7.8 Hz, 1H) 8.17 (br s, 1H) 8.04 (d, J = 7.8 Hz, 1H) 7.64 (br s, 1H) 7.47 (br s, 1H) 7.14 (d, J = 7.8 Hz, 1H) 4.24 (br s, 2H) 3.98 (br s, 1H) 3.39 (br s, 2H) 2.97 (br s, 2H) 1.33 (d, J = 5.7 Hz, 6H).
Example S-7: Synthesis ofl-(3-(6-((l,l-dimethyl-l,2,3,4-tetrahydroisoquinolin-6-yl)amino)- 2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)phenyl)cyclopropane-l- carbonitrile (Compound No. 1.7)
Figure imgf000064_0001
[0125] Step-1: Synthesis of l-(3-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-lH- pyrazolo[3,4-d]pyrimidin-l-yl)phenyl)cyclopropane-l-carbonitrile: To a stirred solution of 2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (224 mg , 1.0 mmol, 1.0 eq) and l-(3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)cyclopropane-l- carbonitrile (269.15 mg, 1.0 mmol, 1.0 eq) in ACN (15 mL) was added 2,2-bipyridine (312.3 mg, 2.0 mmol, 2.0 eq), copper acetate (364 mg, 2.0 mmol, 2.0 eq) and Na2COs (318 mg, 3.0 mmol, 3.0 eq). The reaction mixture was stirred at RT for 48 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated, and purified by silica gel column chromatography to afford the desired product (80 mg, 21.9%). LCMS: 366.39 (M+l)+.
[0126] Step-2: Synthesis of tert-butyl 6-((l-(3-(l-cyanocyclopropyl)phenyl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-l,l-dimethyl- 3,4-dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution l-(3-(2-isopropyl-6- (methylthio)-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)phenyl)cyclopropane-l- carbonitrile (80 mg, 0.22 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (97 mg, 0.43 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert-Butyl 6-amino-l,l-dimethyl-3,4- dihydroisoquinoline-2(lH)-carboxylate (60.5 mg, 0.22 mmol, 1.0 eq) and DIPEA (0.15 mL, 0.87 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (40 mg, 30.8%). LCMS: 594.39 (M+l) +.
[0127] Step-3: Synthesis of l-(3-(6-((l,l-dimethyl-l,2,3,4-tetrahydroisoquinolin-6- yl)amino)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l- yl)phenyl)cyclopropane-l-carbonitrile: tert-Butyl 6-((l-(3-(l-cyanocyclopropyl)phenyl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-l,l-dimethyl-3,4- dihydroisoquinoline-2(lH)-carboxylate (40 mg, 0.07 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HC1 (1 mL) and allowed to stir at RT for 1 h. After completion of reaction, the precipitate formed was collected by to afford title compound (15 mg, 42.0%). LCMS: 494.5(M+1)+; 'H NMR (400 MHz, DMSO- e): <7 ppm 10.36 (br s, 1H) 9.34 (br s, 2H) 8.84 (s, 1H) 7.47 - 7.67 (m, 3H) 7.44 (br s, 1H) 7.23 - 7.41 (m, 2H) 3.97 - 4.14 (m, 1H) 3.40 (br s, 2H) 2.95 (br s, 2H) 1.78 - 1.92 (m, 2H) 1.53 - 1.69 (m, 8H) 1.17 - 1.35 (m, 6H).
Example S-8: Synthesis of2-fluoro-5-(2-isopropyl-3-oxo-6-((l,2,3,4-tetrahydroisoquinolin-7- yl)amino)-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)benzonitrile (Compound No 1.8)
Figure imgf000065_0001
[0128] Step-1: Synthesis of 2-fluoro-5-(2-isopropyl-6-(methylthio)-3-oxo-2,3- dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)benzonitrile: To a stirred solution of 2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (250 mg, 1.11 mmol, 1.0 eq) and 3-cyano-4-fluorophenylboronic acid (220 mg, 1.34 mmol, 1.2 eq) in acetonitrile (10 mL) was added 2,2-bipyridine (260 mg, 1.66 mmol, 1.5 eq), copper acetate (400 mg, 2.22 mmol, 2.0 eq) and Na2COs (552 mg, 3.3 mmol, 3.0 eq). The reaction mixture was stirred at RT for 24h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated, and purified by silica gel column chromatography to afford the desired product (70 mg, 18.4%). LCMS: 344.0 (M+l)+.
[0129] Step-2: Synthesis of tert-butyl 7-(l-(3-cyano-4-fhiorophenyl)-2-isopropyl-3- oxo-2, 3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline- 2(lH)-carboxylate: To a stirred solution 2-fluoro-5-(2-isopropyl-6-(methylthio)-3-oxo-2,3- dihydro-lH-pyrazolo[3,4-d]pyrimidin-l-yl)benzonitrile (60 mg, 0.17 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (75 mg, 0.35 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert-Butyl-7-amino-3,4-dihydroisoquinoline-2(lH)-carboxylate (48 mg, 0.19 mmol, 1.1 eq) and NaHCCh (61 mg, 0.7 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (60 mg, 63.1%). LCMS: 544.3(M+1) +.
[0130] Step-3: Synthesis of 2-fluoro-5-(2-isopropyl-3-oxo-6-((l, 2,3,4- tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-l- yl)benzonitrile hydrochloride tert-Butyl 7-(l-(3-cyano-4-fluorophenyl)-2-isopropyl-3-oxo- 2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(lH)- carboxylate (60 mg, 0.11 mmol, 1.0 eq) was stirred in 2M HC1 in ether (2 mL). After completion of reaction as monitored by LCMS analysis, solution was concentrated and triturated with diethyl ether to afford title compound (35 mg, 71.8%) as off white solid. LCMS: 444.5 (M+l)+; 'H NMR (400 MHz, DMSO- 6): d ppm 10.37 (br s, 1H) 9.25 (br s, 2H) 8.85 (s, 1 H) 8.19 - 8.28 (m, 1H) 7.97 (d, J = 3.5 Hz, 1H) 7.73 (t, J = 8.9 Hz, 1H) 7.35 - 7.61 (m, 2H) 7.15 (d, J = 8.3 Hz, 1H) 4.23 (br s, 2H) 4.07 (dd, J = 13.3, 6.80 Hz, 2H) 3.35 (br s, 2H) 2.84-3.00 (m, 2H) 1.28 (d, J = 7.02 Hz, 6 H). Example S-9: Synthesis ofl-(5-fluoro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-6- (l,2,3,4-tetrahydroisoquinolin-6-ylamino)-lEl-pyrazolo[3,4-d]pyrimidin-3(2E[)-one
Figure imgf000067_0001
[0131] Step-1: Synthesis of l-(6-bromo-3-fhioropyridin-2-yl)cyclobutan-l-ol: To a stirred solution of 2,6-dibromo-3-fluoropyridine (1 g, 3.92 mmol, 1.0 eq) in in diethyl ether (20 mL) was added n-BuLi (1.72 ml, 4.31 mmol, 1.1 eq) allowed to stirred at -78 °C for 1 hour, cyclobutanone (0.6 ml, 7.84 mmol, 2.0 eq) was added and allowed to stir at same temp for 15 min. After completion of reaction, the reaction mixture was basified with NH4CI and extracted with EtOAc (150 mL x 2). Combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (444 mg, 46.0%). LCMS: 245.99 (M+l) +.
[0132] Step-2: Synthesis of l-(5-fhioro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of l-(6-bromo-3-fluoropyridin-2-yl)cyclobutan-l-ol (387 mg, 1.57 mmol, 1.0 eq) and 2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (352 mg, 1.57 mmol, 1.0 eq) in (20 mL) of dioxane were added potassium carbonate (434 mg, 3.14 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (59.8 mg, 0.31 mmol, 0.2 eq), and N,N '-dimethylethylenediamine (DMEDA) (0.07 ml, 0.63 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 130 °C for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (100 mL x 2). The combined organic layers were washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (395 mg 64.5%). LCMS: 390.13 (M+l) +.
[0133] Step-3: Synthesis of tert-butyl 6-((l-(5-fhioro-6-(l- hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4- d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-l,2- dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (197.5 mg, 0.51 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (240 mg, 1,01 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert-Butyl 6-amino-3,4-dihydroisoquinoline-2(lH)-carboxylate (126 mg, 0.51 mmol, 1.2 eq) and Na2COs (215 mg, 2.03 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (155 mg, 51.8%). LCMS: 590.28 (M+l) +.
[0134] Step-4: Synthesis of l-(5-fhioro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one dihydrochloride: tert-Butyl 6-((l-(5-fluoro-6-(l- hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4- d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (65 mg, 0.11 mmol, 1.0 eq) was dissolved in dioxane (1.5 mL), followed by dropwise addition of 4.0 M-HC1 (4 mL) and allowed to stir at RT for 2h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the title compound (24 mg, 38.7%).
LCMS: 490.23 (M+l) +; 'H NMR (400 MHz, DMSO- 6): Jppm 10.31 (br s, 1H) 9.19 (br s, 1H) 8.84 (s, 1H) 7.93 - 8.09 (m, 1H) 7.88 (d, J = 5.7 Hz, 1H) 7.70 (br s, 1H) 7.47 (d, J = 8.3 Hz, 1H) 7.15 (d, J = 8.3 Hz, 1H) 4.20 (br s, 2H) 3.37 (br s, 2H) 2.88 - 3.04 (m, 2H) 2.69 (d, J = 15.8 Hz, 2H) 2.18 - 2.33 (m, 2H) 1.84 (br s, 1H) 1.38 - 1.55 (m, 2H) 1.34 (d, J = 6.6 Hz, 6H).
Example S-10: Synthesis ofl-(5-fluoro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-6- ((1 ,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l ,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (Compound No 1.10)
Figure imgf000069_0001
[0135] Step-1: Synthesis of tert-butyl 7-((l-(5-fhioro-6-(l-hydroxycydobutyl)pyridin- 2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro-6-(l- hydroxycyclobutyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one (197.5 mg, 0.51 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (240 mg, 1,01 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert-Butyl 7-amino-3,4- dihydroisoquinoline-2(lH)-carboxylate (126 mg, 0.51 mmol, 1.2 eq) and Na2COs (215 mg, 2.03 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (141 mg, 47.2%). LCMS: 590.28 (M+l) +. [0136] Step-2: Synthesis of l-(5-fhioro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one: tert-Butyl-7-((l-(5-fluoro-6-(l-hydroxycyclobutyl)pyridin-2-yl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate (76 mg, 0.11 mmol, 1.0 eq) was dissolved in dioxane (1.5 mL), followed by dropwise addition of 4.0 M-HC1 (4 mL) and allowed to stir at RT for 2h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound (30 mg, 41.4%). LCMS: 490.23 (M+l)+; 'H NMR (400 MHz, DMSO-^6): Jppm 10.35 (br s, 1H) 9.07 (br s, 2H) 8.84 (s, 1 H) 8.01 (t, J = 9.43 Hz, 1H) 7.90 (d, J = 6.1 Hz, 1H) 7.68 (br s, 1H) 7.48 (d, J = 7.45 Hz, 1H) 7.17 (d, J = 9.2 Hz, 1H) 4.26 (br s, 2H) 4.19 (br s, 1H) 3.37 (br s, 2H) 2.95 (d, J = 5.26 Hz, 2H) 2.64 - 2.78 (m, 2H) 2.25 (d, J = 10.5 Hz, 2H) 1.85 (m 2H) 1.34 (d, J = 7.0 Hz, 6H).
Example S-ll: Synthesis ofl-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one (Compound No 1.11)
Figure imgf000070_0001
[0137] Step-1: Synthesis of l-(6-bromo-3-fhioropyridin-2-yl)-l-cyclopropylethan-l- ol: To a stirred solution of 2,6-dibromo-3-fluoropyridine (500 mg, 1.96 mmol, 1.0 eq) in (10 mL) of diethylether was added n-BuLi (1.96 ml, 4.90 mmol, 2.5 eq ) allowed to stirred at -78 °C for 1 hour. 1-Cyclopropylethan-l-one (0.2 ml ,2.35 mmol, 1.2 eq) was added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was basified with NH4CI and extracted with EtOAc (150 mL x 2). Combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (280 mg, 54.8%). LCMS: 260 (M+l) +
[0138] Step-2: Synthesis of l-(6-(l-cydopropyl-l-hydroxyethyl)-5-fhioropyridin-2- yl)-2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of l-(6-bromo-3-fluoropyridin-2-yl)-l-cyclopropylethan-l-ol (280 mg, 1.07 mmol, 1.0 eq) and 2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3- one (241 mg, 1.07 mmol, 1.0 eq) in (20 mL) of dioxane were added potassium carbonate (259 mg, 2.14 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (40 mg, 0.21 mmol, 0.2 eq), and N,N'- dimethylethylenediamine (DMEDA) (0.05 ml, 0.22 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 130 °C for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (100 mL x 2). The combined organic layers were washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound (345 mg 79.4%). LCMS: 404.15 (M+l) +.
[0139] Step-3: Synthesis of tert-butyl 6-((l-(6-(l-cyclopropyl-l-hydroxyethyl)-5- fhioropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6- yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(6-( 1- cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-l,2-dihydro- 3H-pyrazolo[3,4-d]pyrimidin-3-one (172.5 mg, 0.43 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (178 mg, 0.854 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert- Butyl 6-amino-3,4-dihydroisoquinoline-2(lH)-carboxylate (117 mg, 0.470 mmol, 1.2 eq) and Na2CC>3 (181 mg, 1.71 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (188 mg, 72.8%). LCMS: 604.30 (M+l) +. [0140] Step-4: Synthesis of l-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fhioropyridin-2- yl)-2-isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H- pyrazolo[3,4-d]pyrimidin-3-one: tert-Butyl 6-((l-(6-(l-cyclopropyl-l-hydroxyethyl)-5- fluoropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6- yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (188 mg, 0.31 mmol, 1.0 eq) was dissolved in dioxane (2 mL), followed by dropwise addition of 4.0 M-HC1 (4 mL) and allowed to stir at RT for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound (30 mg, 19.1%). LCMS: 504.24 (M+l) +; 'H NMR (400 MHz, DMSO- 6): 8 ppm 10.33 (br s, 1H) 8.97 (br s, 1H) 8.86 (s, 1H) 7.97 - 8.08 (m, 1H) 7.88 (d, J = 9.2 Hz, 1H) 7.68 (br s, 1H) 7.48 (d, J = 8.7 Hz, 1H) 7.15 (d, J = 8.3 Hz, 1H) 4.89 (s, 1 H) 4.10 - 4.33 (m, 2H) 3.42 (m, 2H) 2.96 (br s, 2H) 1.51 (s, 3H) 1.31 (t, J = 6.1 Hz, 6H) 0.42 (br s, 2H) 0.23 (br s, 2H).
Example S-12: Synthesis ofl-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2-yl)-2- isopropyl-6-((l ,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l ,2-dihydro-3H-pyrazolo[3,4-
Figure imgf000072_0001
[0141] Step-1: Synthesis of tert-butyl 7-((l-(6-(l-cyclopropyl-l-hydroxyethyl)-5- fhioropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6- yl)amino)-3,4-dihydroisoquinoline-2(lH)-carboxylate : To a stirred solution of l-(6-( 1- cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-lH- pyrazolo[3,4-d]pyrimidin-3(2H)-one (172.5 mg, 0.43 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (178 mg, 0.854 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert- Butyl 7-amino-3,4-dihydroisoquinoline-2(lH)-carboxylate (117 mg, 0.470 mmol, 1.2 eq) and Na2CC>3 (181 mg, 1.71 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (132 mg, 51.2%). LCMS: 604.30 (M+l) +.
Step-2: Synthesis of l-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fhioropyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-7-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one: tert-Butyl 7-((l-(6-(l-cyclopropyl-l-hydroxyethyl)-5-fluoropyridin-2- yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate (132 mg, 0.31 mmol, 1.0 eq) was dissolved in dioxane (2 mL), followed by dropwise addition of 4.0 M-HC1 (4 mL) and allowed to stir at RT for 2h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound (40 mg, 25.5%). LCMS: 504.24 (M+l) +; 'H NMR (400 MHz, DMSO- 6): 3 ppm 10.22 (br s, 1H), 8.82 (s, 1 H) 7.97 - 8.05 (m, 1H) 7.86 (d, J = 5.7 Hz, 1H) 7.51 (br s, 1H) 7.39 (d, J = 9.6 Hz, 1H) 7.05 (d, J = 8.3 Hz, 1H) 4.89 (s, 1 H) 4.18 (d, J = 7.5 Hz, 1H) 3.95 (br s, 2H) 3.08 (br s, 2H) 2.73 (br s, 2H) 2.67 (br s, 2H) 1.51 (s, 3H) 1.31 (m, J = 6.4 Hz, 6H) 0.42 (m 2H) 0.35 (m 2H).
Example S-13: Synthesis ofl-(5-fluoro-6-(3-fluorooxetan-3-yl)pyridin-2-yl)-2-isopropyl-6- ((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
(Compound No 1.13)
Figure imgf000074_0001
[0142] Step-1: Synthesis of 3-(6-bromo-3-fhioropyridin-2-yl)oxetan-3-: To a stirred solution of 2,6-dibromo-3-fluoropyridine (1 g, 3.92 mmol, 1.0 eq) in (20 mL) of diethyl ether was added n-Buli (1.72 ml, 4.31 mmol, 1.1 eq) allowed to stirred at -78 °C for 1 h. Oxetan-3- one (0.5 ml ,7.84 mmol, 2.0 eq) were added and allowed to stir at same temp for 15 min. After completion of reaction, the reaction mixture was basified with NH4CI and extracted with EtOAc (150 mL x 2). Combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (800 mg, 82.9%). LCMS: 247.96(M+1) +.
[0143] Step-2: Synthesis of l-(5-fhioro-6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 3-(6-bromo-3-fluoropyridin-2-yl)oxetan-3-ol (386 mg, 1.55 mmol, 1.0 eq) and 2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (349 mg, 1.55 mmol, 1.0 eq) in (20 mL) of dioxane were added potassium carbonate (430 mg, 3.11 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (59 mg, 0.31 mmol, 0.2 eq), and N,N'-dimethylethylenediamine (DMEDA) (0.06 ml, 0.62 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 130 °C for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (100 mL x 2). The combined organic layers were washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound (260 mg 42.7%). LCMS: 392.11 (M+l) +.
[0144] Step-3: Synthesis of l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin-2-yl)-2- isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of l-(5-fluoro-6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)- l,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (260 mg, 0.664 mmol, 1.0 eq) in (10 mL) of DCM was added DAST (0.13 ml, 0.996 mmol, 1.1 eq) allowed to stirred at -78 °C for 1 h . After completion of reaction, the reaction mixture was basified with NaHCOs and extracted with EtOAc (50 mL x 2). Combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (229 mg, 87.6%). LCMS: 394.11 (M+l) +.
[0145] Step-4: Synthesis of tert-butyl 6-((l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin- 2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro-6-(3- fluorooxetan-3-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one (229 mg, 0.58 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (276 mg, 1.16 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert-Butyl 6-amino-3,4- dihydroisoquinoline-2(lH)-carboxylate (144.5 mg, 0.58 mmol, 1.2 eq) and Na2COs (246.76 mg, 2.33 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (174 mg, 50.4%). LCMS: 594.26 (M+l) +.
[0146] Step-5: Synthesis of l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one: tert-Butyl-6-((l-(5-fluoro-6-(3-fluorooxetan-3-yl)pyridin-2-yl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4- dihydroisoquinoline-2(lH)-carboxylate (174 mg, 0.29 mmol, 1.0 eq) was dissolved in DCM (10 mL), followed by dropwise addition of TFA (1 mL) and allowed to stir at RT for 3 h. After completion of reaction, the reaction mixture was washed with diethyl ether (2 x 10 mL), filtered, and dried under reduced pressure to afford the desired compound (90 mg, 51.9%). LCMS: 494.21 (M+l) +; 'H NMR (400 MHz, DMSO-76): 8 ppm 10.38 (br s, 1H), 8.96 (br s, 2H), 8.87 (s, 1 H), 8.18 - 8.28 (m, 1H), 8.06 - 8.13 (m, 1H), 7.67 (br s, 1H), 7.50 (d, 7 = 7.8 Hz, 1H), 7.17 (d, J = 8.3 Hz, 1H), 5.13 - 5.27 (m, 2H), 4.87 - 5.03 (m, 2H), 4.08 - 4.28 (m, 3H), 2.98 (t, J = 6.1 Hz, 2H) 1.34 (d, J = 6.6 Hz, 6H).
Example S-14: Synthesis ofl-(5-fluoro-6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2-isopropyl-6- (l,2,3,4-tetrahydroisoquinolin-6-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one (Compound No 1.14)
Figure imgf000076_0001
[0147] Step-1: Synthesis of tert-butyl 6-(l-(5-fhioro-6-(3-hydroxyoxetan-3- yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6- ylamino)-3,4-dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro- 6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-lH-pyrazolo[3,4- d]pyrimidin-3(2H)-one (200 mg, 0.51 mmol, 1.0 eq) in (3.0 mL) of toluene was added m- CPBA (242 mg, 1.02 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert-Butyl 6-amino-3,4- dihydroisoquinoline-2(lH)-carboxylate (127 mg, 0.51 mmol, 1.2 eq) and Na2COs (216.24 mg, 2.04 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (80 mg, 26.5%). LCMS: 592.26 (M+l) +.
[0148] Step-2: Synthesis of l-(5-fhioro-6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2- isopropyl-6-((l,2,3,4-tetrahydroisoquinolin-6-yl)amino)-l,2-dihydro-3H-pyrazolo[3,4- d]pyrimidin-3-one: tert-Butyl-6-(l-(5-fluoro-6-(3-hydroxyoxetan-3-yl)pyridin-2-yl)-2- isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4- dihydroisoquinoline-2(lH)-carboxylate (80 mg, 0.135 mmol, 1.0 eq) was dissolved in DCM (10 mL), followed by dropwise addition of TFA (1 mL) and allowed to stir at RT for 3h. After completion of reaction, the reaction mixture was washed with diethyl ether (2 x 10 mL), filtered and dried under reduced pressure to afford the desired compound, (19 mg, 26.4%). LCMS: 492.21 (M+l) +; 'H NMR (400 MHz, DMSO-76): Jppm 10.19 (br s, 1H), 8.81 (s, 1H), 8.44 (br s, 3H), 8.08 (t, J = 9.2 Hz, 1H), 7.93 (d, J = 5.7 Hz, 1H), 7.49 (br s, 1H), 7.38 (d, J = 8.3 Hz, 1H), 6.97 (d, J = 7.8 Hz, 1H), 5.06 (d, 7 = 6.1 Hz, 2H), 4.70 (d, J = 6.6 Hz, 2H), 4.17 (d, 7 = 7.0 Hz, 1H), 3.81 (br s, 2H), 2.96 (br s, 2H), 2.72 (m, H), 1.33 (d, 7 = 6.1 Hz, 6H).
Example S-15: Synthesis ofl-(5-fluoro-6-(3-fluorooxetan-3-yl)pyridin-2-yl)-2-isopropyl-6- (l,2,3,4-tetrahydroisoquinolin-7-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one
Figure imgf000077_0001
[0149] Step-1: Synthesis of tert-butyl 7-(l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin- 2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4- dihydroisoquinoline-2(lH)-carboxylate: To a stirred solution of l-(5-fluoro-6-(3- hydroxyoxetan-3-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-lH-pyrazolo[3,4-d]pyrimidin- 3(2H)-one (130 mg, 0.330 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (156 mg, 0.66 mmol, 2.0 eq) and allowed to stir at RT for 1 h. tert-Butyl 7-amino-3,4- dihydroisoquinoline-2(lH)-carboxylate (82 mg, 0.330 mmol, 1.2 eq) and Na2COs (140 mg, 2.04 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL x 2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by silica gel column chromatography to afford the desired product (70 mg, 35.7%). LCMS: 594.26 (M+l) +.
Step-2: Synthesis of l-(5-fhioro-6-(3-fhiorooxetan-3-yl)pyridin-2-yl)-2-isopropyl-6- (l,2,3,4-tetrahydroisoquinolin-7-ylamino)-lH-pyrazolo[3,4-d]pyrimidin-3(2H)-one: tert- Butyl 7-(l-(5-fluoro-6-(3-fluorooxetan-3-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-lH- pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(lH)-carboxylate (70 mg, 0.117 mmol, 1.0 eq) was dissolved in DCM (10 mL), followed by dropwise addition of TFA (1 mL) and allowed to stir at RT for 3 h. After completion of reaction, the reaction mixture was washed with diethyl ether (2 x 10 mL), filtered and dried under reduced pressure to afford the desired compound (3 mg, 5.4%). LCMS: 494.20 (M+l) +; 'H NMR (400 MHz, DMSO-^6): 3 10.40 (br s, 1H), 8.76 - 8.91 (m, 1H), 8.17 - 8.27 (m, 1H), 8.12 (d, J = 10.1 Hz, 1H), 7.64 (br s, 1H), 7.48 (d, J = 8.7 Hz, 1H), 7.19 (d, J = 7.9 Hz, 1H), 5.14 - 5.29 (m, 2H), 4.91 - 5.06 (m, 2H), 4.26 (br s, 2H), 4.18 (d, J = 7.5 Hz, 1H), 3.32 (br s, 2H), 2.95 (br s, 2H), 1.35 (d, J = 6.6 Hz, 6 H).
[0150] The compounds disclosed therein are prepared according to the experimental details exemplified in Examples S1-S15 and Scheme 1 to Scheme 5, using the appropriate starting materials and reagents. Biological Examples
Example BL WEE1 IC50 Determination
[0151] IC50 values of compounds against WEE1 kinase enzyme were determined by LanthaScreen™ Terbium Labeled TR-FRET assay. Kinase assays were performed in IX kinase buffer (#PV6135, Invitrogen, Life Technologies Grand Island, NY) where total reaction volume was 10 pL in low-volume 384-well plates (#4511, Corning). Serially diluted compounds (3-fold) were incubated with WEE1 Enzyme (1 nM) (#PR7373A, Invitrogen, Life Technologies, Grand Island, NY) for 10 min; a mixture of ATP (10 pM) (#A1852, Sigma, St. Louis, MO) and fluorescent-PolyGT substrate (200 nM) (#PV3610, Invitrogen, Life Technologies Grand Island, NY) was added and incubated in dark at room temperature for 1 h. After 1 h, 10 pL stop solution containing Terbium labeled antibody (4 nM) (#PV3529, Invitrogen, Life Technologies Grand Island, NY) and EDTA (#E5134, Sigma, St. Louis, MO) (20 mM) in TR-FRET dilution buffer (# PV3574, Invitrogen, Life Technologies Grand Island, NY) was added. Readings were taken in a Synergy Neo Plate reader (BioTek, Winooski, VT) at single excitation of 340 nm and dual emission at 495 nm and 520 nm respectively.
[0152] The % activity of test samples was calculated as (Sample - Min)*100/(Max - Min). [Max: DMSO control, complete reaction with enzyme & DMSO and Min: No enzyme & DMSO]. Percent inhibition (100 -% activity) was fitted to the “four-parameter logistic model” in XLfit for determination of IC50 values. The results are shown in Table 2.
Table 2
Figure imgf000079_0001
Figure imgf000080_0001
Example B2. PKMYT1 IC50 Determination
[0153] Inhibition of PKMYT1 kinase activity by test compounds was measured by the HotSpot Kinase Assay at Reaction Biology Corporation (Malvern, PA). Briefly, Myelin Basic Protein substrate was prepared in Reaction Buffer (20 mM Hepes (pH 7.5), 10 mM MgCh, 1 mM EGTA, 0.01% Brij35, 0.02 mg/ml BSA, 0.1 mM Na3VO4, 2 mM DTT, 1% DMSO). PKMYT1 kinase was delivered into the substrate solution and gently mixed. Test compounds in 100% DMSO were added into the kinase reaction mixture by Acoustic technology (Echo550; nanoliter range) and incubated for 20 min at room temperature. 33P- ATP was delivered into the reaction mixture to initiate the reaction. Reactions were carried out at 10 pM ATP. After a 2 hour incubation at room temperature, kinase activity was detected by P81 filter-binding method. Compounds were tested in 10-dose IC50 mode with a 3-fold serial dilution. A nonlinear regression model with a sigmoidal dose response and variable slope within GraphPad Prism (GraphPad Software, San Diego, CA) was used to calculate the IC50 value of individual test compounds. The results are shown in Table 3.
Table 3
Figure imgf000080_0002
Figure imgf000081_0001
Example B3. Determination of potency of compounds in cytotoxicity assay inA427 cell line
[0154] A427 (HTB-53; ATCC), a lung epithelial cell line, was seeded in medium (MEM,
41090101; Gibco) at a cell count of 1500 cells per 100 pL per well in a 96 well edge plate (167425; ThermoFisher). Cells were allowed to grow at 37 °C for 24 hr in 5% CO2 environment (culture conditions) in a Nuaire incubator (humidified). Serially diluted test compounds (100 pF) within the desired testing concentration ranges were added to the culture plate and the cells were further incubated in culture conditions for 72 hr. The experiment was terminated at the designated incubation time by replacing the medium with 100 pL of 1 mM of resazurin (R7017; Sigma) prepared in culture medium, and the plates were further incubated in culture conditions for 4-6 hr. Fluorescence was recorded using a multimodal plate reader (Biotek Synergy Neo) at an excitation wavelength of 535 nm and emission wavelength of 590 nm to obtain relative fluorescence units. Data were analysed as follows: the background fluorescence (blank containing only medium) value was subtracted from each reading and normalized with the vehicle control (DMSO treated cells) to obtain percent survival/proliferation. Percent survival was subtracted from 100 to get the percent inhibition of proliferation which was used to calculate IC50 values. Potency of compounds in other cell lines (such as A549, AsPc-1, Pane 10.05, A172, U-87MG) may be determined in an analogous manner. The results are shown in Table 4. Table 4
Figure imgf000082_0001
Example B4. Determination of potency of compounds in cell proliferation assay in selected cancer cell lines and cellular PD effects.
[0155] The effects of test compounds are studied in additional cell lines with various histotypes, such as LoVo colorectal adenocarcinoma, NCI-H460 large-cell lung carcinoma, HCT-116 colorectal carcinoma, and A2780 ovarian cancer cells. The cancer cells are harvested during the logarithmic growth period and counted. Cell concentrations are adjusted to the appropriate number with suitable medium, and 90 pL cell suspensions are added to 96- well plates. After cells are seeded, the plates are shaken gently to distribute cells evenly and incubated at 37 °C, 5% CO2 on day 1.
[0156] Cells are treated with test compounds at 9 concentrations within a desired concentration range (e.g. 1.5 nM - 10 pM) on day 2 by series diluting the test compound stock solution (10 mM in DMSO) with culture medium. Cell viability is assessed by Cell Titer-Gio® as recommended by Promega (Cat. No.: G7572, Promega) typically 72 h posttreatment.
[0157] Cell viability data are plotted using GraphPad Prism (version 5, GraphPad Software, Inc., San Diego, CA). In addition, a nonlinear regression model with a sigmoidal dose response and variable slope within GraphPad Prism is used to calculate the IC50 value of individual test compounds.
[0158] Test compounds may be studied in the same and/or other cancer cell lines with varying sensitivities to reported Weel inhibiting compounds using similar proliferation methods with possible variations in cell seeding densities and/or incubation durations.
Example B5. Determination of potency of compounds by assay of cellular PD effects.
[0159] pCDC2 and y-H2AX are two clinically relevant biomarkers associated with Weel inhibition. CDC2Y 15 phosphorylation in cells was reported to be abolished by Weel inhibitors (Gavory G et. al., Almac Discovery, AACR poster, 2016). y-H2AX, a DNA double-strand break marker, was upregulated by Weel treatment in Weel sensitive cell lines (Guertin AD et al., Molecular Cancer Therapeutics, 2013). The effects of selected test compound on pCDC2 and y-H2AX are assessed in selected cancer cell lines post 24 or 48 hr treatment using Western blotting methods with selective antibodies (Guertin AD et al., Molecular Cancer Therapeutics, 2013).
[0160] Changes in the levels of phospho-CDC2 following treatment of cells with test compound were assessed by enzyme-linked immunosorbent assay (ELISA). A427 cells or AsPC-1 cells were plated in 6-well plates and cultured for 24 hr to approximately 80-90% confluency. Medium was replaced, and the cells were treated with the vehicle control or the test compound at several different concentrations. After incubation of treated cells in cell culture conditions for a specified time (e.g., 24 hr), cells were rinsed with ice-cold PBS and lysed in IX cell lysis buffer containing protease inhibitors and phosphatase inhibitors. The cells were scraped from the plate with a cell scraper after a brief incubation on ice and transferred to a centrifuge tube, and then subjected to three freeze-thaw cycles in liquid nitrogen and a 37°C water bath for further lysis. The lysates were centrifuged to pellet cell debris (using, for example, a 10 min centrifugation of 2000 X g at 4°C) and the supernatants transferred to fresh tubes on ice. The protein concentrations of the samples were estimated by the Bradford method or equivalent. The ELISA was carried out with the PathScan® Phospho- CDC2 (Tyrl5) Sandwich ELISA Kit (Cat. #7176, Cell Signaling Technology, Danvers, MA) according to the manufacturer’s instructions. Results are shown in Table 5.
Table 5
Figure imgf000084_0001
ND: Not Determined
[0161] Changes in the levels of phospho-CDC2 are alternatively or additionally analyzed by Western blotting of the samples using a primary antibody to phospho-CDC2 such as phospho-CDC2 (Tyrl5) (10A11) rabbit mAb (Cat. #4539, Cell Signaling Technology) or rabbit polyclonal anti-CDKl (phospho Y15) antibody (Cat. #ab47594, Abeam, Cambridge, United Kingdom).
Example B6. Evaluation of test compound in mouse xenograft models
[0162] To examine the in vivo antitumor activity of test compound (as a single agent and in combination with other agents such as gemcitabine, nab-paclitaxel and temozomide), tumor growth experiments are performed in a cell line xenograft model and/or a PDX model. The cell line is chosen based on the in vitro studies described above. The PDX model to be used is established from a tumor taken directly from a patient with, for example, pancreatic ductal adenocarcinoma (PDAC) or glioblastoma.
[0163] Cells or tumor chucks are implanted subcutaneously into the flanks of nude mice and allowed to grow until the tumor size reaches 200 mm3. Tumors are measured using a caliper and tumor volumes calculated using the formula: Tumor volume = (a x b2/2) where ‘b’ is the smallest diameter and ‘a’ is the largest diameter. Once the established tumors reach approximately 200 mm3, the mice are then stratified into treatment groups. The treatment groups are, for example: vehicle control, gemcitabine + nab-paclitaxel, test compound alone, gemcitabine + nab-paclitaxel + test compound at 10 mice per group. The treatment groups are alternatively, for example: vehicle control, temozolomide, test compound alone, temozolomide + test compound. The exact treatment groups, drug dose, and dosing schedule are determined specifically for each study according to standard practice. Tumor growth is monitored, and volume recorded at regular intervals. When the individual tumor of each mouse reaches an approximate end-point (tumor volume >1,500 mm3), the mouse is sacrificed with regulated CO2. The tumor growth inhibition (TGI) is calculated by comparing the control group’s tumor measurements with the other study groups once the predetermined endpoint is reached in the control group. Alternatively, cells are implanted orthotopically and overall survival is measured.
[0164] Although the foregoing disclosure has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is apparent to those skilled in the art that certain minor changes and modifications will be practiced in light of the above teaching. Therefore, the description and examples should not be construed as limiting the scope of the disclosure.

Claims

1. A compound selected from the group consisting of the compounds in Table 1, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing.
2. The compound of claim 1, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, wherein the compound is selected from the group consisting of:
Figure imgf000086_0001
Figure imgf000087_0001
3. The compound of claim 1 or 2, or a pharmaceutically acceptable salt thereof.
4. A pharmaceutical composition comprising the compound of claim 1 or 2, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier.
5. A method of treating a cancer in an individual in need thereof comprising administering to the individual a therapeutically effective amount of the compound of claim 1 or 2, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing.
6. The method of claim 5, further comprising administering a radiation therapy to the individual.
7. The method of claim 5 or 6, further comprising administering to the individual a therapeutically effective amount of an additional therapeutic agent.
8. The method of claim 7, wherein the additional therapeutic agent is a cancer immunotherapy agent or a chemotherapeutic agent.
9. The method of claim 7 or 8, wherein the additional therapeutic agent is a DNA alkylating agent, a platinum-based chemotherapeutic agent, a kinase inhibitor or a DNA damage repair (DDR) pathway inhibitor.
10. The method of any one of claims 5-9, wherein the cancer comprises a mutant TP53 gene.
11. The method of any one of claims 5-10, comprising selecting the individual for treatment based on (i) the presence of one or more mutations in the TP53 gene in the cancer, or (ii) expression of mutant p53 in the cancer.
12. A method of suppressing a G2-M checkpoint in a cell, comprising administering the compound of claim 1 or 2, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, to the cell.
13. A method of inducing premature mitosis in a cell, comprising administering the compound of claim 1 or 2, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, to the cell.
14. A method of inducing apoptosis in a cell, comprising administering the compound of claim 1 or 2, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, to the cell.
15. A method of inhibiting Weel in a cell, comprising administering the compound of claim 1 or 2, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, to the cell.
16. A method of inhibiting Weel, comprising contacting Weel with the compound of claim 1 or 2, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing.
17. The method of claim 16, wherein the inhibitor binds to Weel with an IC50 of less than 1 pM according to a kinase assay.
18. Use of the compound of claim 1 or 2, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing, in the manufacture of a medicament for treatment of cancer.
19. A kit comprising the compound of claim 1 or 2, or a stereoisomer, tautomer, or a pharmaceutically acceptable salt of any of the foregoing.
87
PCT/US2021/071827 2020-10-12 2021-10-12 Heterocyclic compounds and uses thereof WO2022082174A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063090634P 2020-10-12 2020-10-12
US63/090,634 2020-10-12

Publications (1)

Publication Number Publication Date
WO2022082174A1 true WO2022082174A1 (en) 2022-04-21

Family

ID=81209438

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/071827 WO2022082174A1 (en) 2020-10-12 2021-10-12 Heterocyclic compounds and uses thereof

Country Status (1)

Country Link
WO (1) WO2022082174A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022247641A1 (en) * 2021-05-28 2022-12-01 江苏天士力帝益药业有限公司 Wee1 inhibitor and use thereof
WO2023045942A1 (en) * 2021-09-22 2023-03-30 微境生物医药科技(上海)有限公司 1,2-dihydro-3h-pyrazole[3,4-d]pyrimidin-3-one compound serving as wee-1 inhibitor

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100221211A1 (en) * 2007-10-23 2010-09-02 Hidetomo Furuyama Pyridone-substituted-dihydropyrazolopyrimidinone derivative
WO2019074979A1 (en) * 2017-10-09 2019-04-18 Girafpharma, Llc Heterocyclic compounds and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100221211A1 (en) * 2007-10-23 2010-09-02 Hidetomo Furuyama Pyridone-substituted-dihydropyrazolopyrimidinone derivative
WO2019074979A1 (en) * 2017-10-09 2019-04-18 Girafpharma, Llc Heterocyclic compounds and uses thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KIM, HY ET AL.: "Targeting the WEE1 kinase as a molecular targeted therapy for gastric cancer", ONCOTARGET, vol. 7, no. 31, 2 August 2016 (2016-08-02) - 23 June 2016 (2016-06-23), pages 49902 - 49916, XP055699271, DOI: 10.18632/oncotarget.10231 *
VAN LINDEN A A; BATURIN D; FORD J B; FOSMIRE S P; GARDNER L; KORCH C; REIGAN P; PORTER C C: "Inhibition of Wee1 Sensitizes Cancer Cells to Antimetabolite Chemotherapeutics In Vitro and In Vivo, Independent of p53 Functionality", MOLECULAR CANCER THERAPEUTICS, vol. 12, no. 12, December 2013 (2013-12-01) - 11 October 2013 (2013-10-11), pages 2675 - 2684, XP055435258, DOI: 10.1158/1535-7163 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022247641A1 (en) * 2021-05-28 2022-12-01 江苏天士力帝益药业有限公司 Wee1 inhibitor and use thereof
WO2023045942A1 (en) * 2021-09-22 2023-03-30 微境生物医药科技(上海)有限公司 1,2-dihydro-3h-pyrazole[3,4-d]pyrimidin-3-one compound serving as wee-1 inhibitor

Similar Documents

Publication Publication Date Title
US11299493B2 (en) Heterocyclic compounds and uses thereof
US10807994B2 (en) Heterocyclic compounds and uses thereof
AU2020329288A1 (en) Heterocyclic compounds as kinase inhibitors
WO2020210381A1 (en) Heterocyclic compounds and uses thereof
US11332473B2 (en) Substituted pyrazolo[3,4-d]pyrimidines as Wee1 inhibitors
WO2020210380A1 (en) Heterocyclic compounds and uses thereof
WO2022082174A1 (en) Heterocyclic compounds and uses thereof
WO2019018583A1 (en) 1,8-naphthyridinone compounds and uses thereof
AU2020207952A1 (en) Heterocyclic compounds as adenosine antagonists
WO2021146631A1 (en) Heterocyclic compounds as adenosine antagonists
WO2020210383A1 (en) Heterocyclic compounds and uses thereof
WO2020210377A1 (en) Heterocyclic compounds and uses thereof
WO2022236256A1 (en) Heterocyclic compounds as kinase inhibitors
WO2021030620A1 (en) Heterocyclic compounds as kinase inhibitors
WO2021146629A1 (en) Heterocyclic compounds as adenosine antagonists
WO2022236257A1 (en) Heterocyclic compounds as kinase inhibitors
WO2022236253A1 (en) Heterocyclic compounds as kinase inhibitors
WO2022236255A2 (en) Heterocyclic compounds as kinase inhibitors

Legal Events

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

Ref document number: 21881302

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: 21881302

Country of ref document: EP

Kind code of ref document: A1