WO2020210383A1 - Heterocyclic compounds and uses thereof - Google Patents

Heterocyclic compounds and uses thereof Download PDF

Info

Publication number
WO2020210383A1
WO2020210383A1 PCT/US2020/027308 US2020027308W WO2020210383A1 WO 2020210383 A1 WO2020210383 A1 WO 2020210383A1 US 2020027308 W US2020027308 W US 2020027308W WO 2020210383 A1 WO2020210383 A1 WO 2020210383A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
salt
alkylene
optionally substituted
halogen
Prior art date
Application number
PCT/US2020/027308
Other languages
French (fr)
Inventor
Sarvajit Chakravarty
Son Minh Pham
Jayakanth Kankanala
Brahmam PUJALA
Varun Kumar
Original Assignee
Nuvation Bio 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 Inc. filed Critical Nuvation Bio Inc.
Priority to US17/594,299 priority Critical patent/US20220168313A1/en
Priority to EP20787952.9A priority patent/EP3952878A4/en
Publication of WO2020210383A1 publication Critical patent/WO2020210383A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/542Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • 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
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems

Definitions

  • This disclosure relates generally to therapeutics engaged in inhibition of the DNA damage checkpoint kinase, Wee1, which potentiates genotoxic chemotherapies by abrogating cell-cycle arrest and proper DNA repair.
  • the invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of diseases associated with this pathway.
  • Wee1 is a tyrosine kinase that phosphorylates and inactivates Cdc2 and is involved in G checkpoint signaling. More particularly, Wee1 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 G1-S checkpoint, p53-deficient tumors rely only on the G2-M checkpoint after DNA damage. Hence, such tumors are selectively sensitized to DNA-dam ging agents by Wee1 inhibition.
  • Wee1 belongs to a family of protein kinases involved in the terminal
  • Wee1 was first identified in fission yeast, where Wee1 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-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((4- (4methylpiperazin-1-yl)phenyl)amino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one).
  • AZD-1775 exhibited antitumor activity in various preclinical studies in potentiating chemo- and radiotherapy and is currently in phase I/II clinical trials.
  • Wee1 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.
  • the compound of Formula (I) or a salt thereof is of Formula (Ia), (Ia-1), (Ia-2), (I-b), (Ib-1) or (Ib-2), as detailed herein.
  • a method of treating cancer in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound detailed herein, such as a compound of Formula (I), or a pharmaceutically acceptable salt thereof. Also provided is a method of inhibiting Wee1 in a cell, comprising administering a compound detailed herein, or a salt thereof, to the cell.
  • compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier or excipient.
  • Kits comprising a compound detailed herein or a pharmaceutically acceptable salt thereof are also provided.
  • Compounds as detailed herein or a pharmaceutically acceptable salt thereof are 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 (i.e., C 1 -C 10 means one to ten carbons). Particular alkyl groups are those having 1 to 20 carbon atoms (a“C 1 -C 20 alkyl”).
  • alkyl groups are those having 1 to 8 carbon atoms (a“C 1 -C 8 alkyl”), 3 to 8 carbon atoms (a“C 3 -C 8 alkyl”), 1 to 6 carbon atoms (a “C 1 -C 6 alkyl”), 1 to 5 carbon atoms (a“C 1 -C 5 alkyl”), or 1 to 4 carbon atoms (a“C 1 -C 4 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.
  • 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-1-enyl, prop-2-enyl (or allyl), 2-methylprop-1-enyl, but-1-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“C1-C6 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“C 1 -C 3 alkylene”). Examples of alkylene include, but are not limited to, groups such as methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), butylene (-CH 2 CH 2 CH 2 CH 2 -), and the like.
  • Alkynyl refers to an unsaturated linear or branched univalent hydrocarbon chain or combination thereof, having at least one site of acetylenic unsaturation (i.e., having at least one moiety of the formula C ⁇ C) and having the number of carbon atoms designated (i.e., C 2 -C 10 means two to ten carbon atoms).
  • Particular alkynyl groups are those having 2 to 20 carbon atoms (a“C 2 -C 20 alkynyl”), having 2 to 8 carbon atoms (a“C 2 -C 8 alkynyl”), having 2 to 6 carbon atoms (a“C 2 -C 6 alkynyl”), or having 2 to 4 carbon atoms (a “C 2 -C 4 alkynyl”).
  • alkynyl examples include, but are not limited to, groups such as ethynyl (or acetylenyl), prop-1-ynyl, prop-2-ynyl (or propargyl), but-1-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 "C 3 -C 8 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.
  • a“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 (-CF 3 ).
  • “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).
  • Heteroaryl 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 quaternized.
  • 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 non- aromatic 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 quaternized.
  • 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 systems, 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-1(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
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • 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. 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.
  • 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”.
  • U is O or S
  • W is A or AB, wherein A and B are fused together;
  • A is phenyl or 5- or 6-membered heteroaryl, each of which is optionally substituted
  • R 17a wherein A and R 17a together are and n is 0, 1, 2, 3, or 4;
  • B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl, each of which is optionally substituted with R 17b , wherein A, B, R 17a , and R 17b
  • n are independently 0, 1, 2, 3, or 4;
  • X is hydrogen or C 1 -C 6 alkyl
  • Y is N or CR 1 ;
  • Z is N or CR 2 ;
  • R 1 and R 2 are independently hydrogen or R 17a ;
  • R 3a and R 3b are independently hydrogen or R 17a , or R 3a and R 3b are taken together with the carbon to which they are attached to form a C 3 -C 6 cycloalkyl;
  • each R 17b is independently oxo or R 17a , or
  • any two R 17b groups when bound to the same carbon atom or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R 10 ;
  • each R 17a is independently C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, halogen, -CN, -OR 10 , -SR 10 , -NR 11 R 12 , -C(O)R 10 , -C(O)OR 10 , -Si(C 1 -C 6 alkyl) 3 , -C(O)NR 11 R 12 ,
  • each R 17a is independently optionally substituted by halogen, oxo, -CN, -OR 13 , -NR 13 R 14 , -C(O)R 13 , -C(O)NR 13 R 14 , -NR 13 C(O)R 14 , -S(O) 2 R 13 , -NR 13 S(O) 2 R 14 , -S(O) 2 NR 13 R 14 , -(C 1 -C 3 alkylene)C(O)NR 13 R 14 , -(C 1 - C3 alkylene)NR 13 C(O)R 14 , -(C1-C3 alkylene)S(O)2R 13 , -(C1-C3 alkylene)NR 13 S(O)2R 14 , -(C1-C3 alkylene)NR 13 S(O)2R 14 , -(C1- C3 alkylene)S(O)2NR 13 R 14 , -(C1-C3
  • R 4 is 5- to 10-membered heteroaryl or phenyl, wherein the 5- to 10-membered heteroaryl and phenyl of R 4 are each independently optionally substituted by halogen, oxo, -OR 13 , -NR 13 R 14 , - C(O)NR 13 R 14 , -C(O)R 13 , -CN, C3-C8 cycloalkyl, or C1-C6 alkyl optionally substituted by oxo, -CN, -OH or halogen;
  • R 10 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C 6 -C 14 aryl, 5- to 6-membered heteroaryl or 3- to 6-membered heterocyclyl, wherein the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C
  • R 11 and R 12 are each independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl or 3- to 6-membered heterocyclyl, wherein the C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, C 6 -C 14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl of R 11 and R 12 are independently optionally substituted by halogen, oxo, -CN, -OR 15 , -NR 15 R 16 or C 1 -C 6 alkyl optionally substituted by halogen, -OH, or oxo,
  • R 11 and R 12 are taken together with the atom to which they attached to form a 3- to 6- membered heterocyclyl optionally substituted by halogen, oxo, or C 1 -C 6 alkyl optionally substituted by halogen;
  • R 13 and R 14 are each independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C3-C6 cycloalkyl, or 3- to 6-membered heterocyclyl of R 13 and R 14 are optionally substituted by halogen, -CN, -OR 15 , -NR 15 R 16 , or oxo,
  • R 13 and R 14 are taken together with the atom to which they attached to form a 3- to 6- membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by halogen or oxo;
  • R 15 and R 16 are each independently hydrogen, C 1 -C 6 alkyl optionally substituted by halogen or oxo, C2-C6 alkenyl optionally substituted by halogen or oxo, or C2-C6 alkynyl optionally substituted by halogen or oxo,
  • R 15 and R 16 are taken together with the atom to which they attached to form a 3- to 6- membered heterocyclyl optionally substituted by halogen, oxo or C 1 -C 6 alkyl optionally substituted by oxo or halogen.
  • U is S.
  • U is O.
  • Y is N.
  • Y is CR 1 . In some embodiments, Y is CR 1 and R 1 is hydrogen. [0035] In some embodiments of a compound of Formula (I), Z is N. In some embodiments of a compound of Formula (I), Z is N. In some embodiments of
  • Z is CR 2 .
  • Y is CR 2 and R 2 is hydrogen.
  • Y and Z are N.
  • Y is N and Z is N.
  • Y is CR 1 and Z is N.
  • Y is CH and Z is N.
  • Y is N and Z is CR 2 .
  • Y is N and Z is CH.
  • X is hydrogen. In some embodiments, X is C1-C6 alkyl. In some embodiments, X is methyl.
  • R 3a and R 3b are
  • R 3a and R 3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl.
  • R 3a and R 3b are independently hydrogen or C1-C6 alkyl; or R 3a and R 3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl.
  • R 3a is hydrogen and R 3b is C1-C6 alkyl. In some embodiments, R 3a is hydrogen and R 3b is hydrogen. In some embodiments, R 3a is C1-C6 alkyl and R 3b is C1-C6 alkyl. In some embodiments, R 3a is methyl and R 3b is hydrogen. In some embodiments, R 3a is methyl and R 3b is methyl. In some embodiments, R 3a and R 3b are taken together with the carbon to which they are attached to form a C 3 -C 6 cycloalkyl.
  • X is hydrogen; R 3a and R 3b are independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -CN, -OR 10 , -SR 10 , -NR 11 R 12 , -C(O)R 10 , -C(O)NR 11 R 12 ; or R 3a and R 3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl.
  • X is C1- C6 alkyl
  • R 3a and R 3b are independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -CN, -OR 10 , -SR 10 , -NR 11 R 12 , -C(O)R 10 , -C(O)NR 11 R 12 ; or R 3a and R 3b are taken together with the carbon to which they are attached to form a C 3 -C 6 cycloalkyl.
  • X is hydrogen;
  • R 3a and R 3b are independently hydrogen, C 1 -C 6 alkyl, C 2 - C 6 alkenyl, C 2 -C 6 alkynyl, halogen, -CN, -OR 10 , -SR 10 , -NR 11 R 12 , -C(O)R 10 , -C(O)NR 11 R 12 ; or R 3a and R 3b are taken together with the carbon to which they are attached to form a C3- C6 cycloalkyl.
  • R 4 is phenyl optionally substituted by halogen, oxo, -OR 13 , -NR 13 R 14 , -C(O)NR 13 R 14 , -C(O)R 13 , -CN, C 3 -C 8 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by oxo, -CN, -OH or halogen.
  • R 4 is phenyl optionally substituted by halogen, oxo, -OR 13 , -NR 13 R 14 , -C(O)NR 13 R 14 , -C(O)R 13 , -CN, C 3 -C 8 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by oxo, -CN, -OH or halogen.
  • R 4 is , , , . In some embodiments,
  • R 4 is . In some embodiments, R 4 is . In some embodiments, R 4 is . In some embodiments, R 4 is . In some embodiments, R 4 is .
  • W is A, wherein A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted with R 17a .
  • W is AB, wherein A and B are fused together; A is phenyl or 5- to 6- membered heteroaryl, each of which is optionally substituted with R 17a ; and B is C3- C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl, each of which is optionally substituted with R 17b .
  • W is A, wherein A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted with R 17a , and wherein A and R 17a together are and n is 0, 1, 2, 3, or 4.
  • W is AB, wherein A and B are fused together; A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted with R 17a ; B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl, each of which is optionally substituted
  • W is A, wherein A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted with R 17a . In some embodiments, W is A, wherein A is phenyl or 5- to 6-membered heteroaryl, each of which is
  • R 17a optionally substituted with R 17a , wherein A and R 17a together are and n is 0, 1, 2, 3,
  • R 17a is independently 3- to 12- membered heterocyclyl, -(C1-C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by halogen or -OH, -C(O)NR 11 R 12 , -(C1-C3 alkylene)NR 11 R 12 , -CN, halogen, -NR 11 R 12 , C3-C6 cycloalkyl, or -OR 10 , wherein the 3- to 12-membered heterocyclyl of R 17a is optionally substituted with C1-C6 alkyl optionally substituted by halogen or - OH, -C(O)R 13 , -(C1-C3 alkylene)OR 13 , -S(O)2R 13 , C3-C8 cycloalkyl,
  • R 17a is independently C 1 -C 6 alkyl optionally substituted by halogen or -OH, halogen, -CN, or 3- to 12-membered heterocyclyl optionally substituted with C 1 -C 6 alkyl optionally substituted by halogen or -OH.
  • W is A, wherein A is phenyl optionally substituted with R 17a , wherein R 17a is independently 3- to 12-membered heterocyclyl, -(C1-C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by halogen or -OH, -C(O)NR 11 R 12 , -(C1-C3 alkylene)NR 11 R 12 , -CN, halogen, -NR 11 R 12 , C3-C6 cycloalkyl, or -OR 10 , wherein the 3- to 12-membered heterocyclyl of R 17a is optionally substituted with C1-C6 alkyl optionally substituted by halogen or - OH, -C(O)R 13 , -(C1-C3 alkylene)OR 13 , -S(O)2R 13 , C3-C8 cycloalkyl, oxo, halogen, or -OR 13
  • W is A wherein A is phenyl optionally substituted with R 17a , wherein R 17a is independently C 1 -C 6 alkyl optionally substituted by halogen or -OH, halogen, -CN, or 3- to 12-membered heterocyclyl optionally substituted with C 1 -C 6 alkyl optionally substituted by halogen or -OH.
  • W is A, wherein A is pyridinyl optionally substituted with R 17a .
  • W is A, wherein A is pyridinyl optionally substituted with R 17a , wherein R 17a is independently 3- to 12-membered heterocyclyl, -(C 1 -C 3 alkylene)OR 10 , C 1 -C 6 alkyl optionally substituted by halogen or - OH, -C(O)NR 11 R 12 , -(C 1 -C 3 alkylene)NR 11 R 12 , -CN, halogen, -NR 11 R 12 , C 3 -C 6 cycloalkyl, or -OR 10 , wherein the 3- to 12-membered heterocyclyl of R 17a is optionally substituted with C1-C6 alkyl optionally substituted by halogen or - OH, -C(O)R 13 , -(C1
  • W is A, wherein A is pyridinyl optionally substituted with R 17a , wherein R 17a is independently C1-C6 alkyl optionally substituted by halogen or -OH, halogen, -CN, or 3- to 12-membered heterocyclyl optionally substituted with C1-C6 alkyl optionally substituted by halogen or -OH.
  • W is selected from the group consisting of:
  • W is selected from the group consisting of:
  • W is AB, wherein A and B are fused together; A is phenyl or 5- or 6-membered heteroaryl, each of which is optionally substituted with R 17a ; B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7- membered heteroaryl, or C6 aryl, each of which is optionally substituted with R 17b ; and
  • W is AB wherein A and B are fused together; A is phenyl or 6-membered heteroaryl, each of which is optionally substituted with
  • R 17a wherein A and R 17a together are and n is 0, 1, 2, 3, or 4;
  • B is C 3 -C 6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C 6 aryl, each of which is optionally substituted with R 17b , wherein A, B, R 17a , and R 17b together are and m and n are independently 0, 1, 2, 3, or 4.
  • n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4.
  • A, B, R 17a and R 17b together are ; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl; each R 17a is independently -(C1-C3 alkylene)OR 10 , C1- C6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C1-C3 alkylene)NR 11 R 12 , -CN, halogen, -NR 11 R 12 , C3-C6 cycloalkyl, or -OR 10 ; each R 17b is independently oxo, -(C1- C 3 alkylene)OR 10 , C 1 -C 6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C 1 - C 3 alkylene)NR 11 R 12 , -CN, halogen,
  • each R 17b is independently oxo, -(C1-C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C 1 -C 3 alkylene)NR 11 R 12 , -CN, halogen, - NR 11 R 12 , C 3 -C 6 cycloalkyl, or -OR 10 , or any two groups R 17b , when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C 3 - C 6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • R 17a and R 17b together are ; wherein B is 3- to 7-membered heterocyclyl; each R 17a is independently halogen or C1-C6 alkyl optionally substituted by halogen; each R 17b is independently C1-C6 alkyl optionally substituted by halogen, or any two groups R 17b , when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C 3 -C 6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments of a compound of Formula (I), n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4.
  • A, B, R 17a and R 17b together are ; wherein B is C 3 -C 6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C 6 aryl; each R 17a is independently -(C 1 -C 3 alkylene)OR 10 , C 1 - C 6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C 1 -C 3 alkylene)NR 11 R 12 , -CN, halogen, -NR 11 R 12 , C 3 -C 6 cycloalkyl, or -OR 10 ; each R 17b is independently oxo, -(C 1 - C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C1- C3 alkylene)NR 11 R 12 , -CN,
  • A, B, R 17a and R 17b together are ; wherein B is 3- to 7-membered heterocyclyl; each R 17a is independently -(C1-C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by
  • each R 17b is independently oxo, -(C 1 -C 3 alkylene)OR 10 , C 1 -C 6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C 1 -C 3 alkylene)NR 11 R 12 , -CN, halogen, - NR 11 R 12 , C 3 -C 6 cycloalkyl, or -OR 10 , or any two groups R 17b , when bound to the same carbon atom are taken together with the carbon to which they are attached to form a C 3 - C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • R 17a and R 17b together are ; wherein B is 3- to 7-membered heterocyclyl; each R 17a is independently halogen or C 1 -C 6 alkyl optionally substituted by halogen; each R 17b is independently C 1 -C 6 alkyl optionally substituted by halogen, or any two groups R 17b , when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C 3 -C 6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments of a compound of Formula (I), n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4.
  • A, B, R 17a and R 17b together are ; wherein B is C 3 -C 6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl; each R 17a is independently -(C1-C3 alkylene)OR 10 , C1- C6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C1-C3 alkylene)NR 11 R 12 , -CN, halogen, -NR 11 R 12 , C3-C6 cycloalkyl, or -OR 10 ; each R 17b is independently oxo, -(C1- C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C1- C3 alkylene)NR 11 R 12 , -CN, halogen, -
  • A, B, R 17a and R 17b together are ; wherein B is 3- to 7-membered heterocyclyl; each R 17a is independently -(C1-C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by
  • each R 17b is independently oxo, -(C 1 -C 3 alkylene)OR 10 , C 1 -C 6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C 1 -C 3 alkylene)NR 11 R 12 , -CN, halogen, - NR 11 R 12 , C 3 -C 6 cycloalkyl, or -OR 10 , or any two groups R 17b , when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C 3 - C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • R 17a and R 17b together are ; wherein B is 3- to 7-membered heterocyclyl; each R 17a is independently halogen or C1-C6 alkyl optionally substituted by halogen; each R 17b is independently C1-C6 alkyl optionally substituted by halogen, or any two groups R 17b , when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4.
  • A, B, R 17a and R 17b together are ; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl; each R 17a is independently -(C1-C3 alkylene)OR 10 , C1- C6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C1-C3 alkylene)NR 11 R 12 , -CN, halogen, -NR 11 R 12 , C3-C6 cycloalkyl, or -OR 10 ; each R 17b is independently oxo, -(C1- C 3 alkylene)OR 10 , C 1 -C 6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C 1 - C 3 alkylene)NR 11 R 12 , -CN, halogen,
  • A, B, R 17a and R 17b together are ; wherein B is 3- to 7-membered heterocyclyl; each R 17a is independently -(C1-C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by
  • each R 17b is independently oxo, -(C1-C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C1-C3 alkylene)NR 11 R 12 , -CN, halogen, - NR 11 R 12 , C3-C6 cycloalkyl, or -OR 10 , or any two groups R 17b , when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3- C 6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • R 17a and R 17b together are ; wherein B is 3- to 7-membered heterocyclyl; each R 17a is independently halogen or C 1 -C 6 alkyl optionally substituted by halogen; each R 17b is independently C 1 -C 6 alkyl optionally substituted by halogen, or any two groups R 17b , when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C 3 -C 6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I) n is 2 In some embodiments n is 3 In some embodiments, n is 4.
  • A, B, R 17a and R 17b together are ; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C 6 aryl; each R 17a is independently -(C 1 -C 3 alkylene)OR 10 , C 1 - C 6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C 1 -C 3 alkylene)NR 11 R 12 , -CN, halogen, -NR 11 R 12 , C 3 -C 6 cycloalkyl, or -OR 10 ; each R 17b is independently oxo, -(C 1 - C 3 alkylene)OR 10 , C 1 -C 6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C 1 - C 3 alkylene)NR 11 R 12 , -CN,
  • A, B, R 17a and R 17b together are ; wherein B is 3- to 7-membered heterocyclyl; each R 17a is independently -(C1-C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by
  • each R 17b is independently oxo, -(C1-C3 alkylene)OR 10 , C1-C6 alkyl optionally substituted by halogen, -C(O)NR 11 R 12 , -(C 1 -C 3 alkylene)NR 11 R 12 , -CN, halogen, - NR 11 R 12 , C 3 -C 6 cycloalkyl, or -OR 10 , or any two groups R 17b , when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C 3 - C 6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • R 17a and R 17b together are ; wherein B is 3- to 7-membered heterocyclyl; each R 17a is independently halogen or C 1 -C 6 alkyl optionally substituted by halogen; each R 17b is independently C1-C6 alkyl optionally substituted by halogen, or any two groups R 17b , when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
  • W is AB ,wherein A and B are fused together and AB is selected from the group consisting of:
  • AB is or wherein the wavy line denotes attachment point to the parent molecule and AB is optionally substituted by R 17a and R 17b .
  • AB is , wherein the wavy line denotes attachment points to the parent molecule and AB is substituted by C1-C6 alkyl.
  • AB is , wherein the wavy line denotes attachment points to the parent molecule and AB is substituted by C1-C6 alkyl.
  • W is selected from the group consisting of:
  • the compound has one or more of the following features:
  • R 4 is phenyl optionally substituted by halogen, oxo, -OR 13 , -NR 13 R 14 , - C(O)NR 13 R 14 , -C(O)R 13 , -CN, C3-C8 cycloalkyl, or C1-C6 alkyl optionally substituted by oxo, -CN, -OH or halogen, or
  • A is phenyl or 5- or 6-membered heteroaryl, each of which is
  • R 17a optionally substituted with R 17a , wherein A and R 17a together are and n is 0, 1, 2, 3, or 4, or
  • (1) and (3) apply. In some embodiments, (1) and (4) apply. In some embodiments, (1) and (5) apply. In some embodiments, (1) and (6) apply. In some embodiments, (2) and (3) apply. In some embodiments, (2) and (4) apply. In some embodiments, (2) and (5) apply. In some embodiments, (2) and (6) apply.
  • salts of compounds referred to herein such as pharmaceutically acceptable salts.
  • the invention 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 salt thereof are provided, such as
  • compositions of substantially pure compounds are 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 is provided wherein the composition contains no more than 25%, 20%, 15%, 10%, or 5% impurity.
  • 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.
  • 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 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 compounds of Table 1.
  • the structure or name is intended to embrace all possible
  • compositions comprising a compound of the invention 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 invention in any ratio, including two or more stereochemical forms, such as in a racemic or non-racemic mixture.
  • the invention 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 compound of Formula (I) 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 invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C 13 N, 15 O, 17 O, 32 P, 35 S, 18 F, 36 Cl.
  • Certain isotope labeled compounds e.g. 3 H and 14 C
  • are 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 invention 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 invention also includes any or all metabolites of any of the compounds described.
  • the metabolites may include any chemical species generated by a
  • 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 invention 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).
  • the symbols when used in the formulae depicted are to be understood to represent those groups described above in relation to the formulae herein.
  • 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 and/or polymorphs of a compound provided herein or a pharmaceutically acceptable salt thereof are also contemplated. Solvates contain either stoichiometric or non- stoichiometric amounts of a solvent, and are often formed during the process of
  • Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and/or solubility. Various factors such as the
  • recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate
  • compounds of Formula (I) may be synthesized according to Schemes 1-3.
  • 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 pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutically acceptable carrier or excipient in one aspect, the
  • 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 salt thereof 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.
  • 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 salt thereof 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.
  • compositions 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. [0086] 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.
  • 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 pharmaceutically acceptable salt thereof 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 of any formula provided herein or a salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or any embodiment, variation or aspect thereof (collectively, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or the present compounds or the compounds detailed or described herein) 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 the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, to the individual.
  • a method of treating cancer in an individual comprising administering an effective amount of the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a
  • 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 TP53 gene in the cancer, or (ii) expression of mutant p53 in the cancer, and administering an effective amount of the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, to the individual.
  • the cancer is assayed for the expression of mutant p53.
  • the TP53 gene of the cancer is sequenced to detect the one or more mutations.
  • the TP53 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
  • provided herein is a method of using a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or any embodiment in the manufacture of a medicament for treatment of cancer.
  • a compound of formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof 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. In some of these
  • 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
  • mesothelioma mesothelioma, Wilms's tumors, gall bladder cancer, trophoblastic neoplasms,
  • 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 the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, to the cell.
  • the G 2 -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 G 2 -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 the compound of Formula (I), (Ia), (Ia- 1), (Ia-2), (Ib-1) or (Ib-2), 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 the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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.
  • a method of inhibiting Wee1 in a cell comprising administering an effective amount of the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, to the cell.
  • Wee1 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.
  • Wee1 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 Wee1 is measured according to a kinase assay.
  • a method of inhibiting Wee1 comprising contacting Wee1 with an effective amount of the compound of Formula (I), (Ia), (Ia-1), (Ia- 2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof.
  • the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof binds to Wee1 with an IC 50 of less than 1 ⁇ M, 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, or less than 10 nM.
  • the compound of Formula (I) or a pharmaceutically acceptable salt thereof binds to Wee1 with an IC50 between 1 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 ⁇ M.
  • 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 the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof.
  • the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is effective in inhibiting the proliferation of the cell with an IC50 of less than 10 ⁇ M, less than 5 ⁇ M, or less than 2 ⁇ M, or less than 1 ⁇ M.
  • the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt is effective in inhibiting the proliferation of the cell with an IC 50 between 1 ⁇ M and 2 ⁇ M, between 2 ⁇ M and 5 ⁇ M, or between 2 ⁇ M and 10 ⁇ M. In some embodiments, the IC 50 is measured according to a cell proliferation assay.
  • the presently disclosed compounds or a salt thereof may activate the immune system, for example by inducing apoptosis or suppressing mitosis of cancer cells. Accordingly, the present compounds or a salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a
  • 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 immunostimulatory 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 platinum- based 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the additional chemotherapeutic agent.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia- 2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 (
  • 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.
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 kinase inhibitor such as bortezomib, erlotinib, gefitinib, imatinib, vemurafenib, or vismodegib.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the kinase inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DNA damaging agent.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the DNA damaging agent.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co- administered with the DNA alkylating agent.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the topoisomerase inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the anthracycline.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the histone deacetylase inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a taxane (such as paclitaxel or docetaxel).
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co- administered with the taxane.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the nucleotide analog or precursor analog.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the platinum-based chemotherapeutic agent.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of pemetrexed.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the pemetrexed.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DDR pathway inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the DDR pathway inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 (Chk1) inhibitors, or combinations thereof.
  • PARP poly(ADP-ribose) polymerase
  • ATM telangiectasia mutated
  • ATR ataxia telangiectasia and Rad3-related
  • Chk1 checkpoint kinase 1
  • a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the PARP inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATM protein inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the ATM protein inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATR protein inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the ATR protein inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 of (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an Chk1 inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the Chk1 inhibitor.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 Chk1 inhibitor.
  • a combination therapy in which a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and one or more immunostimulatory antibodies, such as an anti-PD-1 antibody, an anti-PD-L1 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2),or a salt thereof and an anti-PD-1 antibody.
  • the subject is administered a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and an anti-PD-L1 antibody.
  • the subject is administered a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and an anti-CTLA-4 antibody.
  • the immunostimulatory antibody e.g., anti-PD-1, anti-PD-L1 and/or anti-CTLA-4 antibody
  • the immunostimulatory antibody is a human antibody.
  • the immunostimulatory antibody can be, for example, a chimeric or humanized antibody (e.g., prepared from a mouse anti-PD-1, anti-PD-L1 and/or anti-CTLA-4 antibody).
  • the present disclosure provides a method for treating a proliferative disease (e.g., cancer), comprising administering a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2),or a salt thereof and an anti-PD-1 antibody to a subject.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof 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 invention provides a method for treating a hyperproliferative disease (e.g., cancer), comprising administering a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and an anti-PD-L1 antibody to a subject.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib- 2), or a salt thereof is administered at a subtherapeutic dose
  • the anti-PD-L1 antibody is administered at a subtherapeutic dose
  • both are administered at a subtherapeutic dose.
  • the present invention provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and a subtherapeutic dose of anti-PD-L1 antibody to a subject.
  • the subject is human.
  • the anti-PD-L1 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.
  • the combination of therapeutic agents can be administered sequentially.
  • an anti-CTLA-4 antibody and a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can be administered sequentially, such as anti- CTLA-4 antibody being administered first and a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof second, or a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof being administered first and anti-CTLA-4 antibody second.
  • an anti-PD-1 antibody and a compound of Formula (I), (Ia), (Ia- 1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can be administered sequentially, such as anti-PD- 1 antibody being administered first and a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof second, or a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof being administered first and anti-PD-1 antibody second.
  • an anti-PD-L1 antibody and a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can be administered sequentially, such as anti-PD-L1 antibody being administered first and a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof second, or a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof being administered first and anti-PD-L1 antibody second.
  • the combination of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib- 1) or (Ib-2), or a salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can also be further combined with standard cancer treatments.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can be effectively combined with chemotherapeutic regimes.
  • 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof include radiation, surgery, or hormone deprivation.
  • Angiogenesis inhibitors can also be combined with a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof.
  • a treatment of a hyperproliferative disease can include an anti-cancer antibody in combination with a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and anti-CTLA-4 and/or anti-PD-1 and/or anti-PD-L1 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof.
  • a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can be combined with an anti-CD73 therapy, such as an anti-CD73 antibody.
  • the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof is administered in combination with another Wee1 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 invention 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 salt thereof and a pharmaceutically acceptable excipient.
  • a compound or composition of the invention 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 salt thereof 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 invention 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.
  • kits 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
  • kits for carrying out the methods of the invention 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 pharmaceutically acceptable 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 invention.
  • the instructions included with the kit generally include information as to the components and their administration to an individual.
  • Step-1 Synthesis of 4-hydroxy-2-(methylthio)-N-(2,4,6- trichlorophenyl)pyrimidine-5-carboxamide: To a stirring suspension of 4-hydroxy-2- methylsulfanyl-pyrimidine-5-carboxylic acid (5.0 g, 26.88 mmol, 1.0 eq) and 2,4,6- trichloroaniline (5.81 g, 29.56 mmol, 1.1 eq) in toluene (200 mL) is added PCl3 (30 mL), then the suspension is heated at 100 °C for 12 h. The reaction is monitored by LCMS.
  • 4-hydroxy-2-(methylthio)-N-(2,4,6- trichlorophenyl)pyrimidine-5-carboxamide To a stirring suspension of 4-hydroxy-2- methylsulfanyl-pyrimidine-5-carboxylic acid (5.0 g, 26.88 mmol, 1.0 eq) and 2,4,6- trichlor
  • Step-2 Synthesis of 7-(methylthio)-3-(2,4,6-trichlorophenyl)-2,3-dihydro-4H- pyrimido[5,4-e][1,3]oxazin-4-one: To a stirring suspension of 4-hydroxy-2-(methylthio)-N- (2,4,6-trichlorophenyl)pyrimidine-5-carboxamide (1.23 g, 3.38 mmol, 1.0 eq) in CH 3 CN (30 mL) and DMSO (5 mL) is added cesium carbonate (3.29 g, 10.14 mmol, 3.0eq) and stirred at RT for 5 min.
  • Step-3 Synthesis of tert-butyl 1,1-dimethyl-6-((4-oxo-3-(2,4,6- trichlorophenyl)-3,4-dihydro-2H-pyrimido[5,4-e][1,3]oxazin-7-yl)amino)-3,4- dihydroisoquinoline-2(1H)-carboxylate: To a stirring solution of 7-(methylthio)-3-(2,4,6- trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (160 mg, 0.423 mmol, 1.0 eq) in 5 mL of toluene is added mCPBA (224 mg, 0.84 mmol, 2.0 eq) and stirred at RT for 30 min.
  • mCPBA 224 mg, 0.84 mmol, 2.0 eq
  • Step-4 Synthesis of 7-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6- yl)amino)-3-(2,4,6-trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one: To a stirring solution of tert-butyl 1,1-dimethyl-6-((4-oxo-3-(2,4,6-trichlorophenyl)-3,4- dihydro-2H-pyrimido[5,4-e][1,3]oxazin-7-yl)amino)-3,4-dihydroisoquinoline-2(1H)- carboxylate (133 mg, 0.22 mmol, 1.0 eq) in dioxane (1 mL) is added 4.0 M-HCl (1 mL) and allowed to stir at RT for 2h. After completion of the reaction, the reaction mixture
  • Example S-2 7-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-3-(2,4,6- trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]thiazin-4-one (Compound No.1.2)
  • Step-1 Synthesis of 4-hydroxy-2-(methylthio)pyrimidine-5-carboxylic acid: To a stirring solution of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (5 g, 21.425 mmol, 1 eq) in EtOH (25 mL) is added a solution of NaOH (8.38 g, 214.25 mol, 10 eq) in water (25 mL) and the resultant mixture is stirred at RT for 16 h. The progress of the reaction is monitored by TLC. After completion, the reaction mixture is acidified using 2N-HCl (5 mL) to obtain a precipitate which is filtered, washed with water and dried to afford the title compound.
  • Step-2 Synthesis of 4-chloro-2-(methylthio)pyrimidine-5-carbonyl chloride : To a stirring solution of 4-hydroxy-2-(methylthio)pyrimidine-5-carboxylic acid (2.5 g, 13.44 mmol, 1 eq) in DMF (2.5 mL) is added SOCl2 (10 mL) and the resultant mixture is heated at 100 °C for 2 h. After completion, the mixture is cooled to RT and concentrated to afford the desired compound.
  • Step-3 Synthesis of 4-chloro-2-(methylthio)-N-(2,4,6- trichlorophenyl)pyrimidine-5-carboxamide: To a stirring solution of 2,4,6-trichloroaniline (2.32 g, 11.8 mmol, 1eq) in DCM (20 mL) is added Et3N (4.96 mL, 35.6 mmol, 3 eq) and 4- chloro-2-(methylthio)pyrimidine-5-carbonyl chloride (2.65 g, 11.8 mmol, 1 eq) in DCM (20 mL). The resultant mixture is stirred at RT for 5 h. The progress of the reaction is monitored by TLC.
  • Step-4 Synthesis of 4-mercapto-2-(methylthio)-N-(2,4,6- trichlorophenyl)pyrimidine-5-carboxamide: To a stirring solution of 4-chloro-2- (methylthio)-N-(2,4,6-trichlorophenyl)pyrimidine-5-carboxamide (1.43 g, 3.72 mmol, 1 eq) in DMF (20 mL), water (10 mL) is added Na 2 S (0.582 g, 4.47 mmol, 1.2 eq) at RT and the resultant mixture is stirred at RT for 1 h.
  • Step-5 Synthesis of 7-(methylthio)-3-(2,4,6-trichlorophenyl)-2,3-dihydro-4H- pyrimido[5,4-e][1,3]thiazin-4-one: To a stirring solution of 4-mercapto-2-(methylthio)-N- (2,4,6-trichlorophenyl)pyrimidine-5-carboxamide (0.39 g, 1.011 mmol, 1 eq) in acetonitrile (5 mL) is added Cs2CO3 (1.3 g, 4.043 mmol, 4 eq) at RT and the reaction mixture is stirred for 10 min.
  • Step-6 Synthesis of tert-butyl 1,1-dimethyl-6-((4-oxo-3-(2,4,6- trichlorophenyl)-3,4-dihydro-2H-pyrimido[5,4-e][1,3]thiazin-7-yl)amino)-3,4- dihydroisoquinoline-2(1H)-carboxylate: To a stirring solution of 7-(methylthio)-3-(2,4,6- trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]thiazin-4-one (165 mg, 0.423 mmol, 1.0 eq) in 5 mL of toluene is added mCPBA (224 mg, 0.84 mmol, 2.0 eq) and stirred at RT for 30 min.
  • mCPBA 224 mg, 0.84 mmol, 2.0 eq
  • Step-7 Synthesis of 7-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6- yl)amino)-3-(2,4,6-trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]thiazin-4-one: To a stirring solution of tert-butyl 1,1-dimethyl-6-((4-oxo-3-(2,4,6-trichlorophenyl)-3,4- dihydro-2H-pyrimido[5,4-e][1,3]thiazin-7-yl)amino)-3,4-dihydroisoquinoline-2(1H)- carboxylate (136 mg, 0.22 mmol,
  • Example S-3 Synthesis of 3-(2,6-dichlorophenyl)-7-((2-(dimethylglycyl)-1,1-dimethyl- 1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (Compound N0.1.48)
  • Step-1 Synthesis of 4-hydroxy-2-methylsulfanyl-pyrimidine-5-carboxylic acid: To a stirred solution of ethyl 4-chloro-2-methylsulfanyl-pyrimidine-5-carboxylate (10 g, 43.1 mmol, 1.0 eq) in 150 mL of ethanol was added water (2:1) and NaOH (17.2 g, 431 mmol, 10 eq). The reaction was heated at 110 °C for 2 h. Progress of the reaction was monitored by LCMS. Upon the consumption of the starting material, the solvent was removed under reduced pressure. The residue was diluted with 100 mL of water and the pH of the mixture was adjusted up to 5 with 3N HCl solution.
  • Step-2 Synthesis of N-(2,6-dichlorophenyl)-4-hydroxy-2-methylsulfanyl- pyrimidine-5-carboxamide: A stirred solution of N-(2,6-dichlorophenyl)-4-hydroxy-2- methylsulfanyl-pyrimidine-5-carboxamide (6.0 g, 32.2 mmol, 1.0 eq) and 2,6-dichloroaniline (5.20 g, 32.2 mmol, 1.0 eq) in 200 mL of toluene was purged with nitrogen gas for 15 min.
  • Step-3 Synthesis of 3-(2,6-dichlorophenyl)-7-methylsulfanyl-2H- pyrimido[5,4-e][1,3]oxazin-4-one: To a solution of N-(2,6-dichlorophenyl)-4-hydroxy-2- methylsulfanyl-pyrimidine-5-carboxamide (4.0 g, 12.1 mmol, 1.0 eq) in CH3CN (200 mL) was added CH2I2 (4.90 g, 18.5 mmol, 1.5 eq) and Cs2CO3 (12.0 g, 36.4 mmol, 3.0 eq).
  • the reaction mixture was heated at 80 °C for 12 h. Progress of the reaction was monitored by LCMS. After completion of the reaction, the solvent was removed under reduced pressure, the residue was diluted with 50 mL of water and extracted with ethyl acetate (100 mL ⁇ 3). The combined organic layer was washed with water (50 mL ⁇ 3), dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography to afford the desired compound (400 mg, 10%).
  • Step-4 Synthesis of tert-butyl 6-(3-(2,6-dichlorophenyl)-4-oxo-3,4-dihydro- 2H-pyrimido[5,4-e][1,3]oxazin-7-ylamino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)- carboxylate: To a stirred solution of 3-(2,6-dichlorophenyl)-7-methylsulfanyl-2H- pyrimido[5,4-e][1,3]oxazin-4-one (200 mg, 0.59 mmol, 1.0 eq) in toluene (3 mL) was added mCPBA (250 mg, 0.147 mmol, 2.5 eq) and allowed to stir at rt for 30 min.
  • mCPBA 250 mg, 0.147 mmol, 2.5 eq
  • reaction mixture was stirred at RT for 1 h. Progress of the reaction was monitored by LCMS. After completion of the reaction, the precipitated compound was filtered off, washed with dioxane (3 mL) and dried under reduced pressure to obtain the desired compound (104 mg, 73.22%) as an off white solid.
  • Step-6 Synthesis of 3-(2,6-dichlorophenyl)-7-((2-(dimethylglycyl)-1,1- dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-4H-pyrimido[5,4- e][1,3]oxazin-4-one: To a stirred solution of N,N-Dimethylglycine (26.3 mg, 0.25 mmol, 1.2 eq) in DMF (3 mL), HATU (121 mg, 0.31 mmol, 1.5eq) was added and stirred at RT for 5 min.3-(2,6-dichlorophenyl)-7-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3- dihydro-4H-pyrimido[5,4-e][1,
  • reaction mixture was diluted with ice-cold water (30 mL) and extracted with ethyl acetate (50 mL ⁇ 2). The combined organic layer was washed with brine solution (20 mL ⁇ 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain the crude product which was purified by reversed phase chromatography to afford (34 mg, 28.79%) as a white solid.
  • Example S-4 Synthesis of 3-(2,6-dichlorophenyl)-7-((2-(dimethylglycyl)-1,2,3,4- tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (Compound No.1.44)
  • Step-1 Synthesis of tert-butyl 7-[[3-(2,6-dichlorophenyl)-4-oxo-2H- pyrimido[5,4-e][1,3]oxazin-7-yl]amino]-3,4-dihydro-1H-isoquinoline-2-carboxylate: To a stirred solution of 3-(2,6-dichlorophenyl)-7-methylsulfanyl-2H-pyrimido[5,4-e][1,3]oxazin- 4-one (200 mg, 0.59 mmol, 1.0 eq) in toluene (3 mL) was added m-CPBA (250 mg, 0.147 mmol, 2.5 eq) and allowed to stir at RT for 30 min.
  • m-CPBA 250 mg, 0.147 mmol, 2.5 eq
  • Step-2 Synthesis of 3-(2,6-dichlorophenyl)-7-(1,2,3,4-tetrahydroisoquinolin- 7-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin-4-one hydrochloride: Tert-butyl 7-[[3-(2,6- dichlorophenyl)-4-oxo-2H-pyrimido[5,4-e][1,3]oxazin-7-yl]amino]-3,4-dihydro-1H- isoquinoline-2-carboxylate (150 mg, 0.277 mmol, 1.0 eq) was dissolved in 4M HCl in dioxane (2 mL) at 0 °C.
  • reaction mixture was stirred at RT for 1 h. Progress of the reaction was monitored by LCMS. After completion of reaction, precipitated compound was filtered off, washed with dioxane (3 mL) and dried under reduced pressure to obtain 3-(2,6- dichlorophenyl)-7-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin- 4-one hydrochloride (100 mg, 75.6%) as HCl salt.
  • Step-3 Synthesis of 3-(2,6-dichlorophenyl)-7-((2-(dimethylglycyl)-1,2,3,4- tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one: To a stirred solution of N,N-dimethylglycine (27.9 mg, 0.27 mmol, 1.2 eq) in DMF (5 mL), HATU (128.8 mg, 0.33 mmol, 1.5eq) was added and stirred at RT for 5 min.3-(2,6- dichlorophenyl)-7-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-4H-pyrimido[5,4- e][1,3]oxazin-4-
  • reaction mixture was diluted with ice-cold water (30 mL) and extracted with ethyl acetate (50 mL ⁇ 2). The combined organic layer was washed with brine solution (20 mL ⁇ 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain the crude product which was purified by reversed phase chromatography to afford the title compound (6.03 mg, 5.1%) as an off-white solid.
  • Example S-5 Synthesis of 3-(2,6-dichlorophenyl)-7-(2-(2-(dimethylamino)acetyl)-1,2,3,4- tetrahydroisoquinolin-6-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin-4(3H)-one (Compound N0. 1.37)
  • Step-1 Synthesis of tert-butyl 6-((3-(2,6-dichlorophenyl)-4-oxo-3,4-dihydro- 2H-pyrimido[5,4-e][1,3]oxazin-7-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 3-(2,6-dichlorophenyl)-7-(methylthio)-2,3-dihydro-4H-pyrimido[5,4- e][1,3]oxazin-4-one (250 mg, 0.730 mmol, 1.0 eq) in 4 mL of toluene was added mCPBA (314 mg, 1.827 mmol, 2.5 eq) and allowed to stir at RT for 30 minutes.
  • mCPBA 314 mg, 1.827 mmol, 2.5 eq
  • Step-2 Synthesis of 3-(2,6-dichlorophenyl)-7-((1,2,3,4-tetrahydroisoquinolin- 6-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one: To tert-butyl 6-((3-(2,6- dichlorophenyl)-4-oxo-3,4-dihydro-2H-pyrimido[5,4-e][1,3]oxazin-7-yl)amino)-3,4- dihydroisoquinoline-2(1H)-carboxylate (150 mg, 0.276 mmol, 1 eq) was added 4 N HCl in dioxane (3 mL).
  • Step-3 Synthesis of 3-(2,6-dichlorophenyl)-7-(2-(2-(dimethylamino)acetyl)- 1,2,3,4-tetrahydroisoquinolin-6-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin-4(3H)-one: To a stirred solution of N,N-dimethylglycine (27.9 mg, 0.27 mmol, 1.2 eq) in DMF (5 mL), HATU (128.8 mg, 0.33 mmol, 1.5 eq) was added and stirred at RT for 5 min.3-(2,6- dichlorophenyl)-7-(1,2,3,4-tetrahydroisoquinolin-6-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin- 4(3H)-one were added and the mixture was stirred
  • reaction mixture was diluted with ice-cold water (30 mL) and extracted with ethyl acetate (50 mL ⁇ 2). The combined organic layer was washed with brine solution (20 mL ⁇ 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude product which was purified by reversed phase chromatography to afford the desired compound (6.0 mg, 5.1%) as an off-white solid.
  • Example S-6 Synthesis of 3-(2,6-dichlorophenyl)-7-((2-isopropyl-1,2,3,4- tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (Compound No.1.40)
  • Example S-7 Synthesis of 3-(2,6-dichlorophenyl)-7-((2-isopropyl-1,2,3,4- tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (Compound No.1.43)
  • IC50 values of the compounds against WEE1 kinase enzyme were determined by LanthaScreenTM Terbium Labeled TR-FRET assay.
  • Kinase assays were performed in 1X kinase buffer (#PV6135, Invitrogen, Life Technologies, Grand Island, NY) where total reaction volume was 10 ⁇ L in low-volume 384-well plates (#4511,Corning).
  • Serially diluted compounds (3-fold) were incubated with WEE1 Enzyme (1nM) (#PR7373A, Invitrogen, Life Technologies, Grand Island, NY) for 10 min; a mixture of ATP (10 ⁇ M) (#A1852, Sigma, St. Louis, MO) and fluorescent-PolyGT substrate (200 nM) (#PV3610, Invitrogen, Life
  • Inhibition of PKMYT1 kinase activity by test compounds is measured by the HotSpot Kinase Assay at Reaction Biology Corporation (Malvern, PA). Briefly, Myelin Basic Protein substrate is prepared in Reaction Buffer (20 mM Hepes (pH 7.5), 10 mM MgCl2, 1 mM EGTA, 0.01% Brij35, 0.02 mg/mL BSA, 0.1 mM Na3VO4, 2 mM DTT, 1% DMSO). PKMYT1 kinase is delivered into the substrate solution and gently mixed.
  • Reaction Buffer 20 mM Hepes (pH 7.5), 10 mM MgCl2, 1 mM EGTA, 0.01% Brij35, 0.02 mg/mL BSA, 0.1 mM Na3VO4, 2 mM DTT, 1% DMSO.
  • Test compounds in 100% DMSO are added into the kinase reaction mixture by Acoustic technology (Echo550; nanoliter range) and incubated for 20 min at room temperature.
  • 33 P- ATP is delivered into the reaction mixture to initiate the reaction. Reactions are carried out at 10 ⁇ M ATP. After a 2 hour incubation at room temperature, kinase activity is detected by P81 filter-binding method.
  • Compounds are tested in 10-dose IC 50 mode with a 3-fold serial dilution.
  • a nonlinear regression model with a sigmoidal dose response and variable slope within GraphPad Prism is used to calculate the IC 50 value of individual test compounds.
  • Example B Determination of potency of compounds in cytotoxicity assay in A427 cell line
  • 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 ⁇ L 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 ⁇ L) within the desired testing concentration ranges were added to the culture plate and the cells further incubated in culture conditions for 72 hr.
  • MEM medium
  • 41090101 Gibco
  • the experiment was terminated at the designated incubation time by replacing the medium with 100 ⁇ L 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.
  • Example B4 Determination of potency of compounds in cell proliferation assay in selected cancer cell lines and cellular PD effects.
  • 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 ⁇ L 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 ⁇ M) 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-Glo® as recommended by Promega (Cat. No.: G7572, Promega) typically 72 hrs post- treatment.
  • a desired concentration range e.g.1.5 nM– 10 ⁇ M
  • Test compounds may be studied in the same and/or other cancer cell lines with varying sensitivities to reported Wee1 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 g-H2AX are two clinically relevant biomarkers associated with Wee1 inhibition. CDC2Y15 phosphorylation in cells was reported to be abolished by Wee1 inhibitors (Gavory G et. al., Almac Discovery, AACR poster, 2016). g-H2AX, a DNA double-strand break marker, was upregulated by Wee1 treatment in Wee1 senstive cell lines (Guertin AD et al., Molecular Cancer Therapeutics, 2013). The effects of selected test compounds on pCDC2 and g-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 other suitable cell line) are plated in 6-well plates and cultured for 24 hr to approximately 80-90% confluency. Medium is replaced, and the cells are 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 are rinsed with ice-cold PBS and lysed in 1X cell lysis buffer containing protease inhibitors and phosphatase inhibitors.
  • ELISA enzyme-linked immunosorbent assay
  • the cells are 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 are 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 are estimated by the Bradford method or equivalent.
  • the ELISA is carried out with the
  • PathScan® Phospho-CDC2 (Tyr15) Sandwich ELISA Kit (Cat. #7176, Cell Signaling Technology, Danvers, MA) or similar product according to the manufacturer’s instructions. Changes in the levels of phospho-CDC2 may alternatively or additionally be analyzed by Western blotting of the samples using a primary antibody to phospho-CDC2 such as phospho-CDC2 (Tyr15) (10A11) rabbit mAb (Cat. #4539, Cell Signaling Technology) or rabbit polyclonal anti-CDK1 (phospho Y15) antibody (Cat. #ab47594, Abcam, Cambridge, United Kingdom).
  • phospho-CDC2 Tyr15
  • 10A11 rabbit mAb
  • rabbit polyclonal anti-CDK1 (phospho Y15) antibody Cat. #ab47594, Abcam, 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 temozolomide)
  • 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 CO 2 .
  • 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

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biomedical Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Radiology & Medical Imaging (AREA)
  • Pathology (AREA)
  • Molecular Biology (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 APPLICATION
[0001] This application claims priority to U.S. Provisional Application No.62/831,677, filed on April 9, 2019, the content of which is incorporated herein by reference in its entirety. FIELD OF THE INVENTION
[0002] This disclosure relates generally to therapeutics engaged in inhibition of the DNA damage checkpoint kinase, Wee1, which potentiates genotoxic chemotherapies by abrogating cell-cycle arrest and proper DNA repair. The invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of diseases associated with this pathway. BACKGROUND OF THE INVENTION
[0003] Wee1 is a tyrosine kinase that phosphorylates and inactivates Cdc2 and is involved in G checkpoint signaling. More particularly, Wee1 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 G1-S checkpoint, p53-deficient tumors rely only on the G2-M checkpoint after DNA damage. Hence, such tumors are selectively sensitized to DNA-dam ging agents by Wee1 inhibition.
[0004] Wee1 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. Wee1 was first identified in fission yeast, where Wee1 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 G1-S, intra-S, and G2-M checkpoints. Wee1 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 Chk1-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 Wee1 and Chk1/Cdc25. Wee1 inhibition is thus expected to abrogate G2-M arrest and propel cells into premature mitosis, a hypothesis confirmed by studies documenting that Wee1 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 Wee1 inhibitors have been disclosed. Among them is a selective inhibitor, AZD-1775 (1, 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((4- (4methylpiperazin-1-yl)phenyl)amino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one). AZD-1775 exhibited antitumor activity in various preclinical studies in potentiating chemo- and radiotherapy and is currently in phase I/II clinical trials.
[0007] Wee1 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 Wee1 inhibitors in clinical development. There is scope to improve Wee1 inhibitor selectivity and the properties of the inhibitors to permit targeting of specific cancer types. BRIEF SUMMARY OF THE INVENTION
[0009] In one aspect, provided is a compound of Formula (I):
(I),
or a salt thereof, wherein U, W, X, Y, Z, R3a, R3b, and R4 are as detailed herein.
[0010] In some embodiments, the compound of Formula (I) or a salt thereof, is of Formula (Ia), (Ia-1), (Ia-2), (I-b), (Ib-1) or (Ib-2), as detailed herein.
[0011] 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 detailed herein, such as a compound of Formula (I), or a pharmaceutically acceptable salt thereof. Also provided is a method of inhibiting Wee1 in a cell, comprising administering a compound detailed herein, or a salt thereof, to the cell.
[0012] In another aspect, provided are pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier or excipient. Kits comprising a compound detailed herein or a pharmaceutically acceptable salt thereof are also provided. Compounds as detailed herein or a pharmaceutically acceptable salt thereof are also provided for the manufacture of a medicament for the treatment of cancer. DETAILED DESCRIPTION OF THE INVENTION Definitions
[0013] “Alkyl” refers to and includes saturated linear and branched univalent hydrocarbon structures and combination thereof, having the number of carbon atoms designated (i.e., C1-C10 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“C1-C8 alkyl”), 3 to 8 carbon atoms (a“C3-C8 alkyl”), 1 to 6 carbon atoms (a “C1-C6 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.
[0014] “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 (i.e., having at least one moiety of the formula C=C) and having the number of carbon atoms designated (i.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-1-enyl, prop-2-enyl (or allyl), 2-methylprop-1-enyl, but-1-enyl, but-2-enyl, but-3-enyl, buta-1,3-dienyl, 2-methylbuta-1,3-dienyl, homologs and isomers thereof, and the like.
[0015] “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“C1-C6 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.
[0016] “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 (i.e., having at least one moiety of the formula C^C) and having the number of carbon atoms designated (i.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-1-ynyl, prop-2-ynyl (or propargyl), but-1-ynyl, but-2-ynyl, but-3-ynyl, homologs and isomers thereof, and the like.
[0017] “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.
[0018] “Carbonyl” refers to the group C=O.
[0019] “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.
[0020] “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).
[0021] “Heteroaryl” 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 quaternized. 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.
[0022] “Heterocycle” or“heterocyclyl” refers to a saturated or an unsaturated non- aromatic 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 quaternized. 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 systems, 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-1(2H)-yl, and the like.
[0023] “Oxo” refers to the moiety =O.
[0024] “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.
[0025] 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.
[0026] 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.
[0027] 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.
[0028] 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.
[0029] 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).
[0030] 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”.
[0031] It is understood that aspects and variations described herein also include “consisting” and/or“consisting essentially of” aspects and variations. Compounds
[0032] In one aspect, provided is a compound of Formula (I):
Figure imgf000009_0001
or a salt thereof, wherein:
U is O or S;
W is A or AB, wherein A and B are fused together;
A is phenyl or 5- or 6-membered heteroaryl, each of which is optionally substituted
with R17a, wherein A and R17a together are and n is 0, 1, 2, 3, or 4;
Figure imgf000009_0002
B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl, each of which is optionally substituted with R17b, wherein A, B, R17a, and R17b
together are
Figure imgf000009_0003
and m and n are independently 0, 1, 2, 3, or 4;
X is hydrogen or C1-C6 alkyl;
Y is N or CR1; Z is N or CR2;
R1 and R2 are independently hydrogen or R17a;
R3a and R3b are independently hydrogen or R17a, or R3a and R3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl;
each R17b is independently oxo or R17a, or
any two R17b groups, when bound to the same carbon atom or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R10;
each R17a is independently C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -CN, -OR10, -SR10, -NR11R12, -C(O)R10, -C(O)OR10, -Si(C1-C6 alkyl)3, -C(O)NR11R12,
-OC(O)NR11R12, -NR10C(O)R11, -NR10C(O)NR11R12, -S(O)2R10, -NR10S(O)2R11,
-S(O)2NR11R12, C3-C6 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14 aryl, -(C1-C3 alkylene)CN, -(C1-C3 alkylene)OR10, -(C1-C3 alkylene)SR10, -(C1-C3 alkylene)NR11R12, -(C1-C3 alkylene)CF3, -(C1-C3 alkylene)C(O)R10, -(C1- C3 alkylene)C(O)NR11R12, -(C1-C3 alkylene)NR10C(O)R11, -(C1- C3 alkylene)NR10C(O)NR11R12, -(C1-C3 alkylene)S(O)2R10, -(C1-C3 alkylene)NR10S(O)2R11, -(C1-C3 alkylene)S(O)2NR11R12, -(C1-C3 alkylene)(C3-C6 cycloalkyl), -(C1-C3 alkylene)(3- to 12-membered heterocyclyl), -(C1-C3 alkylene)(5- to 10-membered heteroaryl) or
-(C1-C3 alkylene)(C6-C14 aryl), wherein each R17a is independently optionally substituted by halogen, oxo, -CN, -OR13, -NR13R14, -C(O)R13, -C(O)NR13R14, -NR13C(O)R14, -S(O)2R13, -NR13S(O)2R14, -S(O)2NR13R14, -(C1-C3 alkylene)C(O)NR13R14, -(C1- C3 alkylene)NR13C(O)R14, -(C1-C3 alkylene)S(O)2R13, -(C1-C3 alkylene)NR13S(O)2R14, -(C1- C3 alkylene)S(O)2NR13R14, -(C1-C3 alkylene)(C3-C6 cycloalkyl), -(C1-C3 alkylene)(3- to 12- membered heterocyclyl), -Si(C1-C6 alkyl)3, -CN, -(C1-C3 alkylene)OR13, -(C1- C3 alkylene)NR13R14, -(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, or C1-C6 alkyl optionally substituted by oxo, -OH or halogen;
R4 is 5- to 10-membered heteroaryl or phenyl, wherein the 5- to 10-membered heteroaryl and phenyl of R4 are each independently optionally substituted by halogen, oxo, -OR13, -NR13R14, - C(O)NR13R14, -C(O)R13, -CN, C3-C8 cycloalkyl, or C1-C6 alkyl optionally substituted by oxo, -CN, -OH or halogen; R10 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6- membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -OR15, -NR15R16, or C1-C6 alkyl optionally substituted by halogen, -OH or oxo;
R11 and R12 are each independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl of R11 and R12 are independently optionally substituted by halogen, oxo, -CN, -OR15, -NR15R16 or C1-C6 alkyl optionally substituted by halogen, -OH, or oxo,
or R11 and R12 are taken together with the atom to which they attached to form a 3- to 6- membered heterocyclyl optionally substituted by halogen, oxo, or C1-C6 alkyl optionally substituted by halogen;
R13 and R14 are each independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C3-C6 cycloalkyl, or 3- to 6-membered heterocyclyl of R13 and R14 are optionally substituted by halogen, -CN, -OR15, -NR15R16, or oxo,
or R13 and R14 are taken together with the atom to which they attached to form a 3- to 6- membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by halogen or oxo; and
R15 and R16 are each independently hydrogen, C1-C6 alkyl optionally substituted by halogen or oxo, C2-C6 alkenyl optionally substituted by halogen or oxo, or C2-C6 alkynyl optionally substituted by halogen or oxo,
or R15 and R16 are taken together with the atom to which they attached to form a 3- to 6- membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by oxo or halogen.
[0033] In some embodiments of a compound of Formula (I), U is S. In some
embodiments, U is O.
[0034] In some embodiments of a compound of Formula (I), Y is N. In some
embodiments, Y is CR1. In some embodiments, Y is CR1 and R1 is hydrogen. [0035] In some embodiments of a compound of Formula (I), Z is N. In some
embodiments, Z is CR2. In some embodiments, Y is CR2 and R2 is hydrogen.
[0036] In some embodiments of a compound of Formula (I), at least one of Y and Z is N. In some embodiments, Y is N and Z is N. In some embodiments, Y is CR1 and Z is N. In some embodiments, Y is CH and Z is N. In some embodiments, Y is N and Z is CR2. In some embodiments, Y is N and Z is CH.
[0037] In some embodiments, provided is a compound of Formula (Ia):
Figure imgf000012_0001
or a salt thereof, wherein W, X, Z, R3a, R3b, and R4 are as detailed herein.
[0038] In some embodiments, provided is a compound of Formula (Ia-1):
Figure imgf000012_0002
or a salt thereof, wherein W, X, R3a, R3b, and R4 are as detailed herein.
[0039] In some embodiments, provided is a compound of Formula (Ia-2):
Figure imgf000012_0003
( )
or a salt thereof, wherein W, and R4 are as detailed herein.
[0040] In some embodiments, provided is a compound of Formula (Ib):
Figure imgf000013_0001
or a salt thereof, wherein W, X, Z, R3a, R3b, and R4 are as detailed herein.
[0041] In some embodiments, the compound of Formula (Ib-1):
Figure imgf000013_0002
or a salt thereof, wherein W, X, R3a, R3b, and R4 are as detailed herein.
[0042] In some embodiments, the compound of Formula (Ib-2):
Figure imgf000013_0003
or a salt thereof, wherein W, and R4 are as detailed herein,
[0043] In some embodiments of a compound of Formula (I), X is hydrogen. In some embodiments, X is C1-C6 alkyl. In some embodiments, X is methyl.
[0044] In some embodiments of a compound of Formula (I), R3a and R3b are
independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -CN, -OR10, -SR10, -NR11R12, -C(O)R10, -C(O)NR11R12; or R3a and R3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl. In some embodiments of R3a and R3b are independently hydrogen or C1-C6 alkyl; or R3a and R3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl. In some embodiments, R3a is hydrogen and R3b is C1-C6 alkyl. In some embodiments, R3a is hydrogen and R3b is hydrogen. In some embodiments, R3a is C1-C6 alkyl and R3b is C1-C6 alkyl. In some embodiments, R3a is methyl and R3b is hydrogen. In some embodiments, R3a is methyl and R3b is methyl. In some embodiments, R3a and R3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl.
[0045] In some embodiments of a compound of Formula (I), X is hydrogen; R3a and R3b are independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -CN, -OR10, -SR10, -NR11R12, -C(O)R10, -C(O)NR11R12; or R3a and R3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl. In some embodiments, X is C1- C6 alkyl; R3a and R3b are independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -CN, -OR10, -SR10, -NR11R12, -C(O)R10, -C(O)NR11R12; or R3a and R3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl. In some embodiments, X is hydrogen; R3a and R3b are independently hydrogen, C1-C6 alkyl, C2- C6 alkenyl, C2-C6 alkynyl, halogen, -CN, -OR10, -SR10, -NR11R12, -C(O)R10, -C(O)NR11R12; or R3a and R3b are taken together with the carbon to which they are attached to form a C3- C6 cycloalkyl.
[0046] In some embodiments of a compound of Formula (I), R4 is phenyl optionally substituted by halogen, oxo, -OR13, -NR13R14, -C(O)NR13R14, -C(O)R13, -CN, C3-C8 cycloalkyl, or C1-C6 alkyl optionally substituted by oxo, -CN, -OH or halogen. In some
embodiments, R4 is
Figure imgf000014_0001
, , , . In some embodiments,
R4 is
Figure imgf000014_0002
. In some embodiments, R4 is
Figure imgf000014_0003
. In some embodiments, R4 is
Figure imgf000014_0004
. In some embodiments, R4 is
Figure imgf000014_0005
.
[0047] In some embodiments of a compound of Formula (I), W is A, wherein A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted with R17a. In some embodiments, W is AB, wherein A and B are fused together; A is phenyl or 5- to 6- membered heteroaryl, each of which is optionally substituted with R17a; and B is C3- C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl, each of which is optionally substituted with R17b. In some embodiments, W is A, wherein A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted with R17a, and wherein A and R17a together are
Figure imgf000015_0001
and n is 0, 1, 2, 3, or 4. In some embodiments, W is AB, wherein A and B are fused together; A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted with R17a; B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl, each of which is optionally substituted
with R17b; and wherein A, B, R17a, and R17b together are and m and n
Figure imgf000015_0002
are independently 0, 1, 2, 3, or 4.
[0048] In some embodiments of a compound of Formula (I), W is A, wherein A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted with R17a. In some embodiments, W is A, wherein A is phenyl or 5- to 6-membered heteroaryl, each of which is
optionally substituted with R17a, wherein A and R17a together are and n is 0, 1, 2, 3,
Figure imgf000015_0003
or 4. In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, R17a is independently 3- to 12- membered heterocyclyl, -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen or -OH, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10, wherein the 3- to 12-membered heterocyclyl of R17a is optionally substituted with C1-C6 alkyl optionally substituted by halogen or - OH, -C(O)R13, -(C1-C3 alkylene)OR13, -S(O)2R13, C3-C8 cycloalkyl, oxo, halogen, or -OR13, wherein the 3- to 12-membered heterocyclyl of R17a is optionally fused with 5- to 10- membered heteroaryl or C6-C14 aryl. In some embodiments, R17a is independently C1-C6 alkyl optionally substituted by halogen or -OH, halogen, -CN, or 3- to 12-membered heterocyclyl optionally substituted with C1-C6 alkyl optionally substituted by halogen or -OH. In some embodiments, W is A, wherein A is phenyl optionally substituted with R17a, wherein R17a is independently 3- to 12-membered heterocyclyl, -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen or -OH, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10, wherein the 3- to 12-membered heterocyclyl of R17a is optionally substituted with C1-C6 alkyl optionally substituted by halogen or - OH, -C(O)R13, -(C1-C3 alkylene)OR13, -S(O)2R13, C3-C8 cycloalkyl, oxo, halogen, or -OR13, wherein the 3- to 12-membered heterocyclyl of R17a is optionally fused with 5- to 10- membered heteroaryl or C6-C14 aryl. In some embodiments, W is A wherein A is phenyl optionally substituted with R17a, wherein R17a is independently C1-C6 alkyl optionally substituted by halogen or -OH, halogen, -CN, or 3- to 12-membered heterocyclyl optionally substituted with C1-C6 alkyl optionally substituted by halogen or -OH.
[0049] In some embodiments of a compound of Formula (I), W is A, wherein A is pyridinyl optionally substituted with R17a. In some embodiments, W is A, wherein A is pyridinyl optionally substituted with R17a, wherein R17a is independently 3- to 12-membered heterocyclyl, -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen or - OH, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10, wherein the 3- to 12-membered heterocyclyl of R17a is optionally substituted with C1-C6 alkyl optionally substituted by halogen or - OH, -C(O)R13, -(C1-C3 alkylene)OR13, -S(O)2R13, C3-C8 cycloalkyl, oxo, halogen, or -OR13, wherein the 3- to 12-membered heterocyclyl of R17a is optionally fused with 5- to 10- membered heteroaryl or C6-C14 aryl. In some embodiments,, W is A, wherein A is pyridinyl optionally substituted with R17a, wherein R17a is independently C1-C6 alkyl optionally substituted by halogen or -OH, halogen, -CN, or 3- to 12-membered heterocyclyl optionally substituted with C1-C6 alkyl optionally substituted by halogen or -OH.
[0050] In some embodiments of a compound of Formula (I), W is selected from the group consisting of:
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
, , , , , a d
Figure imgf000022_0002
wherein the wavy lines denote attachment points to the parent molecule. In some embodiments, W is selected from the group consisting of:
Figure imgf000022_0003
Figure imgf000023_0001
Figure imgf000023_0002
, a d , wherein the wavy lines denote attachment points to the parent molecule.
[0051] In some embodiments of a compound of Formula (I), W is AB, wherein A and B are fused together; A is phenyl or 5- or 6-membered heteroaryl, each of which is optionally substituted with R17a; B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7- membered heteroaryl, or C6 aryl, each of which is optionally substituted with R17b; and
wherein A, B, R17a, and R17b together are
Figure imgf000023_0003
and m and n are independently 0, 1, 2, 3, or 4. In some embodiments, W is AB wherein A and B are fused together; A is phenyl or 6-membered heteroaryl, each of which is optionally substituted with
R17a, wherein A and R17a together are
Figure imgf000023_0004
and n is 0, 1, 2, 3, or 4; B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl, each of which is optionally substituted with R17b, wherein A, B, R17a, and R17b together are
Figure imgf000023_0005
and m and n are independently 0, 1, 2, 3, or 4.
[0052] In some embodiments of a compound of Formula (I), A, B, R17a and R17b together
are
Figure imgf000023_0006
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl and m and n are independently 0, 1, 2, or 3. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments of a compound of Formula (I), m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments of a compound of Formula (I), n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, A, B, R17a and R17b together are
Figure imgf000024_0001
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl; each R17a is independently -(C1-C3 alkylene)OR10, C1- C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; each R17b is independently oxo, -(C1- C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1- C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; or any two groups R17b, when bound to the same carbon atom or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R10; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a and R17b together are
Figure imgf000024_0002
; wherein B is 3- to 7-membered heterocyclyl; each R17a is
independently -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by
halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3- C6 cycloalkyl, or -OR10 ; each R17b is independently oxo, -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, - NR11R12, C3-C6 cycloalkyl, or -OR10, or any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3- C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a
and R17b together are
Figure imgf000024_0003
; wherein B is 3- to 7-membered heterocyclyl; each R17a is independently halogen or C1-C6 alkyl optionally substituted by halogen; each R17b is independently C1-C6 alkyl optionally substituted by halogen, or any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
[0053] In some embodiments of a compound of Formula (I), A, B, R17a and R17b together
are
Figure imgf000025_0001
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl and m and n are independently 0, 1, 2, or 3. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments of a compound of Formula (I), m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments of a compound of Formula (I), n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, A, B, R17a and R17b together are
Figure imgf000025_0002
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl; each R17a is independently -(C1-C3 alkylene)OR10, C1- C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; each R17b is independently oxo, -(C1- C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1- C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; or any two groups R17b, when bound to the same carbon atom or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R10; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a and R17b together are
Figure imgf000025_0003
; wherein B is 3- to 7-membered heterocyclyl; each R17a is independently -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by
halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3- C6 cycloalkyl, or -OR10 ; each R17b is independently oxo, -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, - NR11R12, C3-C6 cycloalkyl, or -OR10, or any two groups R17b, when bound to the same carbon atom are taken together with the carbon to which they are attached to form a C3- C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a
and R17b together are
Figure imgf000026_0001
; wherein B is 3- to 7-membered heterocyclyl; each R17a is independently halogen or C1-C6 alkyl optionally substituted by halogen; each R17b is independently C1-C6 alkyl optionally substituted by halogen, or any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
[0054] In some embodiments of a compound of Formula (I), A, B, R17a and R17b together
are
Figure imgf000026_0002
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl and m and n are independently 0, 1, 2, or 3. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments of a compound of Formula (I), m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments of a compound of Formula (I), n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, A, B, R17a and R17b together are
Figure imgf000026_0003
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl; each R17a is independently -(C1-C3 alkylene)OR10, C1- C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; each R17b is independently oxo, -(C1- C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1- C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; or any two groups R17b, when bound to the same carbon atom or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R10; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a and R17b together are
Figure imgf000027_0001
; wherein B is 3- to 7-membered heterocyclyl; each R17a is independently -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by
halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3- C6 cycloalkyl, or -OR10 ; each R17b is independently oxo, -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, - NR11R12, C3-C6 cycloalkyl, or -OR10, or any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3- C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a
and R17b together are
Figure imgf000027_0002
; wherein B is 3- to 7-membered heterocyclyl; each R17a is independently halogen or C1-C6 alkyl optionally substituted by halogen; each R17b is independently C1-C6 alkyl optionally substituted by halogen, or any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
[0055] In some embodiments of a compound of Formula (I), A, B, R17a and R17b together
are
Figure imgf000027_0003
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl and m and n are independently 0, 1, 2, or 3. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments of a compound of Formula (I), m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments of a compound of Formula (I), n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I), n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, A, B, R17a and R17b together are
Figure imgf000027_0004
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl; each R17a is independently -(C1-C3 alkylene)OR10, C1- C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; each R17b is independently oxo, -(C1- C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1- C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; or any two groups R17b, when bound to the same carbon atom or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R10; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a and R17b together are
Figure imgf000028_0001
; wherein B is 3- to 7-membered heterocyclyl; each R17a is independently -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by
halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3- C6 cycloalkyl, or -OR10 ; each R17b is independently oxo, -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, - NR11R12, C3-C6 cycloalkyl, or -OR10, or any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3- C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a
and R17b together are
Figure imgf000028_0002
; wherein B is 3- to 7-membered heterocyclyl; each R17a is independently halogen or C1-C6 alkyl optionally substituted by halogen; each R17b is independently C1-C6 alkyl optionally substituted by halogen, or any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
[0056] In some embodiments of a compound of Formula (I), A, B, R17a and R17b together
are
Figure imgf000028_0003
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl and m and n are independently 0, 1, 2, or 3. In some embodiments, m is 0. In some embodiments, m is 1. In some embodiments of a compound of Formula (I), m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments of a compound of Formula (I), n is 0. In some embodiments, n is 1. In some embodiments of a compound of Formula (I) n is 2 In some embodiments n is 3 In some embodiments, n is 4. In some embodiments, A, B, R17a and R17b together are
Figure imgf000029_0001
; wherein B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl; each R17a is independently -(C1-C3 alkylene)OR10, C1- C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; each R17b is independently oxo, -(C1- C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1- C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10; or any two groups R17b, when bound to the same carbon atom or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R10; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a and R17b together are
Figure imgf000029_0002
; wherein B is 3- to 7-membered heterocyclyl; each R17a is independently -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by
halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3- C6 cycloalkyl, or -OR10 ; each R17b is independently oxo, -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, - NR11R12, C3-C6 cycloalkyl, or -OR10, or any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3- C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3. In some embodiments, A, B, R17a
and R17b together are
Figure imgf000029_0003
; wherein B is 3- to 7-membered heterocyclyl; each R17a is independently halogen or C1-C6 alkyl optionally substituted by halogen; each R17b is independently C1-C6 alkyl optionally substituted by halogen, or any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl; and m and n are independently 0, 1, 2, or 3.
[0057] In some embodiments of a compound of Formula (I), W is AB ,wherein A and B are fused together and AB is selected from the group consisting of:
Figure imgf000030_0001
, , , , , ,
Figure imgf000030_0002
, wherein the wavy lines denote attachment points to the parent molecule and each is optionally substituted by R17a and R17b. In some embodiments, AB is
Figure imgf000030_0003
or
Figure imgf000030_0004
wherein the wavy line denotes attachment point to the parent molecule and AB is optionally substituted by R17a and R17b. In some embodiments, AB is
Figure imgf000030_0005
, wherein the wavy line denotes attachment points to the parent molecule and AB is substituted by C1-C6 alkyl. In some embodiments, AB is
Figure imgf000030_0006
, wherein the wavy line denotes attachment points to the parent molecule and AB is substituted by C1-C6 alkyl.
[0058] In some embodiments, W is selected from the group consisting of:
Figure imgf000030_0007
Figure imgf000031_0001
, wherein the wavy lines denote attachment
Figure imgf000031_0002
points to the parent molecule.
[0059] In some embodiments of a compound of Formula (I), the compound has one or more of the following features:
(I) R4 is
(1) R4 is phenyl optionally substituted by halogen, oxo, -OR13, -NR13R14, - C(O)NR13R14, -C(O)R13, -CN, C3-C8 cycloalkyl, or C1-C6 alkyl optionally substituted by oxo, -CN, -OH or halogen, or
Figure imgf000031_0003
(II) W is
(3) AB wherein A and B are fused together, wherein A, B, R17a and R17b together are
Figure imgf000032_0001
Figure imgf000033_0001
(5) A, wherein A is phenyl or 5- or 6-membered heteroaryl, each of which is
optionally substituted with R17a, wherein A and R17a together are
Figure imgf000033_0002
and n is 0, 1, 2, 3, or 4, or
Figure imgf000033_0003
In some embodiments, (1) and (3) apply. In some embodiments, (1) and (4) apply. In some embodiments, (1) and (5) apply. In some embodiments, (1) and (6) apply. In some embodiments, (2) and (3) apply. In some embodiments, (2) and (4) apply. In some embodiments, (2) and (5) apply. In some embodiments, (2) and (6) apply.
[0060] In the descriptions herein, it is understood that every description, variation, embodiment or aspect of a moiety may be combined with every description, variation, embodiment or aspect of other moieties the same as if each and every combination of descriptions is specifically and individually listed. For example, every description, variation, embodiment or aspect provided herein with respect to W of Formula (I) may be combined with every description, variation, embodiment or aspect of X, R1, R2, R3a, R3b, and R4, the same as if each and every combination were specifically and individually listed. It is also understood that all descriptions, variations, embodiments or aspects of Formula (I), where applicable, apply equally to other formulae detailed herein, and are equally described, the same as if each and every description, variation, embodiment or aspect were separately and individually listed for all formulae. For example, all descriptions, variations, embodiments or aspects of formula (I), where applicable, apply equally to any of formulae as detailed herein, such as Formula (Ia), (Ia-1), (Ia-2), (Ib), (Ib-1) and (Ib-2) and are equally described, the same as if each and every description, variation, embodiment or aspect were separately and individually listed for all formulae.
[0061] Also provided are salts of compounds referred to herein, such as pharmaceutically acceptable salts. The invention 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.
[0062] 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 salt thereof 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.
[0063] Representative compounds are listed in Table 1.
Table 1
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
[0064] 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 salt thereof.
[0065] The embodiments and variations described herein are suitable for compounds of any formulae detailed herein, where applicable.
[0066] 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.
[0067] 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.
[0068] 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.
[0069] The present disclosure also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms of the compounds described, such as 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 invention, such as crystalline or non-crystalline forms of the compounds. Compositions comprising a compound of the invention 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 invention in any ratio, including two or more stereochemical forms, such as in a racemic or non-racemic mixture.
[0070] The invention 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 compound of Formula (I) 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 invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, chlorine, such as 2H, 3H, 11C, 13C, 14C 13N, 15O, 17O, 32P, 35S, 18F, 36Cl. Certain isotope labeled compounds (e.g.3H and 14C) are 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. [0071] Isotopically-labeled compounds of the present invention 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.
[0072] The invention 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.
[0073] 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.
[0074] Preferably, the compounds detailed herein are orally bioavailable. However, the compounds may also be formulated for parenteral (e.g., intravenous) administration.
[0075] 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
[0076] The compounds of the invention 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). In the following process descriptions, the symbols when used in the formulae depicted are to be understood to represent those groups described above in relation to the formulae herein.
[0077] 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.
[0078] 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.
[0079] Solvates and/or polymorphs of a compound provided herein or a pharmaceutically acceptable 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. Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and/or solubility. Various factors such as the
recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate
[0080] In some embodiments, compounds of Formula (I) may be synthesized according to Schemes 1-3.
Scheme 1
Figure imgf000044_0001
Scheme 2
Figure imgf000045_0001
wherein W, Y, Z, R3a, R3b, and R4 are as defined for Formula (I). Particular examples are provided in the Example Section below. Here L is a leaving group like Br, Cl or I etc. Pharmaceutical Compositions and Formulations
[0081] 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 pharmaceutically acceptable salt thereof 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.
[0082] 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 salt thereof 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.
[0083] 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.
[0084] 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.
[0085] One or several compounds described herein or a salt thereof 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. [0086] 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.
[0087] 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 pharmaceutically acceptable salt thereof can be formulated as a 10 mg tablet.
[0088] 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
[0089] Compounds and compositions detailed herein, such as a pharmaceutical composition containing a compound of any formula provided herein or a salt thereof 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.
[0090] Provided herein is a method of treating a disease in an individual comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or any embodiment, variation or aspect thereof (collectively, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or the present compounds or the compounds detailed or described herein) 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 the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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.
[0091] 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 TP53 gene in the cancer, or (ii) expression of mutant p53 in the cancer, and administering an effective amount of the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, to the individual. In some embodiments, the cancer is assayed for the expression of mutant p53. In some embodiments, the TP53 gene of the cancer is sequenced to detect the one or more mutations. In some embodiments, the TP53 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.
[0092] In some embodiments, provided herein is a method of using a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or any embodiment in the manufacture of a medicament for treatment of cancer.
[0093] In some embodiments, a compound of formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof 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.
[0094] 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.
[0095] 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 the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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.
[0096] In some embodiments, provided herein is a method of inducing premature mitosis in a cell comprising administering an effective amount of the compound of Formula (I), (Ia), (Ia- 1), (Ia-2), (Ib-1) or (Ib-2), 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.
[0097] In some embodiments, provided herein is a method of inducing apoptosis in a cell comprising administering an effective amount of the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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.
[0098] In some embodiments, provided herein is a method of inhibiting Wee1 in a cell comprising administering an effective amount of the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, to the cell. In some embodiments, Wee1 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, Wee1 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 Wee1 is measured according to a kinase assay.
[0099] In some embodiments, provided herein is a method of inhibiting Wee1 comprising contacting Wee1 with an effective amount of the compound of Formula (I), (Ia), (Ia-1), (Ia- 2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof binds to Wee1 with an IC50 of less than 1 µM, 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, or less than 10 nM. In some embodiments, the compound of Formula (I) or a pharmaceutically acceptable salt thereof binds to Wee1 with an IC50 between 1 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 µM. 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.
[0100] In some embodiments, provided herein is a method of inhibiting the proliferation of a cell, comprising contacting the cell with an effective amount of the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof. In some embodiments, the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is effective in inhibiting the proliferation of the cell with an IC50 of less than 10 µM, less than 5 µM, or less than 2 µM, or less than 1 µM. In some embodiments, the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt is effective in inhibiting the proliferation of the cell with an IC50 between 1 µM and 2 µM, between 2 µM and 5 µM, or between 2 µM and 10 µM. In some embodiments, the IC50 is measured according to a cell proliferation assay. Combination Therapy
[0101] As provided herein, the presently disclosed compounds or a salt thereof may activate the immune system, for example by inducing apoptosis or suppressing mitosis of cancer cells. Accordingly, the present compounds or a salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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.
[0102] 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 immunostimulatory 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 platinum- based 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.
[0103] 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, in combination with a radiation therapy.
[0104] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the additional chemotherapeutic agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia- 2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0105] Examples of chemotherapeutic agents that can be used in combination with a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0106] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the kinase inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0107] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DNA damaging agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the DNA damaging agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0108] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co- administered with the DNA alkylating agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0109] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the topoisomerase inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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. [0110] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the anthracycline. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0111] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the histone deacetylase inhibitor. In some
embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0112] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co- administered with the taxane. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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. [0113] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the nucleotide analog or precursor analog. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0114] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the platinum-based chemotherapeutic agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0115] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of pemetrexed. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the pemetrexed. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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. [0116] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DDR pathway inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the DDR pathway inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 (Chk1) inhibitors, or combinations thereof.
[0117] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the PARP inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0118] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATM protein inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the ATM protein inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0119] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATR protein inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the ATR protein inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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.
[0120] In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an Chk1 inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the Chk1 inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a pharmaceutically acceptable salt thereof 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 Chk1 inhibitor.
[0121] In another aspect, provided herein is a combination therapy in which a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and one or more immunostimulatory antibodies, such as an anti-PD-1 antibody, an anti-PD-L1 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2),or a salt thereof and an anti-PD-1 antibody. In another embodiment, the subject is administered a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and an anti-PD-L1 antibody. In yet another embodiment, the subject is administered a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and an anti-CTLA-4 antibody. In another embodiment, the immunostimulatory antibody (e.g., anti-PD-1, anti-PD-L1 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-L1 and/or anti-CTLA-4 antibody).
[0122] In one embodiment, the present disclosure provides a method for treating a proliferative disease (e.g., cancer), comprising administering a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2),or a salt thereof and an anti-PD-1 antibody to a subject. In further embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof 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.
[0123] In one embodiment, the present invention provides a method for treating a hyperproliferative disease (e.g., cancer), comprising administering a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and an anti-PD-L1 antibody to a subject. In further embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib- 2), or a salt thereof is administered at a subtherapeutic dose, the anti-PD-L1 antibody is administered at a subtherapeutic dose, or both are administered at a subtherapeutic dose. In another embodiment, the present invention provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and a subtherapeutic dose of anti-PD-L1 antibody to a subject. In certain embodiments, the subject is human. In certain embodiments, the anti-PD-L1 antibody is a human sequence monoclonal antibody. [0124] 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can be administered sequentially, such as anti- CTLA-4 antibody being administered first and a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof second, or a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof being administered first and anti-CTLA-4 antibody second. Additionally or alternatively, an anti-PD-1 antibody and a compound of Formula (I), (Ia), (Ia- 1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can be administered sequentially, such as anti-PD- 1 antibody being administered first and a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof second, or a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof being administered first and anti-PD-1 antibody second. Additionally or alternatively, an anti-PD-L1 antibody and a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can be administered sequentially, such as anti-PD-L1 antibody being administered first and a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof second, or a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof being administered first and anti-PD-L1 antibody second.
[0125] 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.
[0126] Optionally, the combination of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib- 1) or (Ib-2), or a salt thereof 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.
[0127] A compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can also be further combined with standard cancer treatments. For example, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof include radiation, surgery, or hormone deprivation. Angiogenesis inhibitors can also be combined with a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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.
[0128] In another example, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof and anti-CTLA-4 and/or anti-PD-1 and/or anti-PD-L1 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 of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof.
[0129] In some embodiments, a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof can be combined with an anti-CD73 therapy, such as an anti-CD73 antibody.
[0130] In yet further embodiments, the compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ib-1) or (Ib-2), or a salt thereof is administered in combination with another Wee1 inhibitor.
Dosing and Method of Administration
[0131] 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.
[0132] 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 invention 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.
[0133] 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 salt thereof and a pharmaceutically acceptable excipient.
[0134] A compound or composition of the invention 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.
[0135] The compounds provided herein or a salt thereof 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.
[0136] In one aspect, the invention 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.
[0137] The invention 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.
[0138] 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
[0139] The present disclosure further provides kits for carrying out the methods of the invention, 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 pharmaceutically acceptable 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.
[0140] 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.
[0141] 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).
[0142] 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 invention. The instructions included with the kit generally include information as to the components and their administration to an individual.
[0143] The invention 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
Synthesis Examples Example S-1: Synthesis of 7-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-3- (2,4,6-trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (Compound No. 1.1)
Figure imgf000064_0001
[0144] Step-1: Synthesis of 4-hydroxy-2-(methylthio)-N-(2,4,6- trichlorophenyl)pyrimidine-5-carboxamide: To a stirring suspension of 4-hydroxy-2- methylsulfanyl-pyrimidine-5-carboxylic acid (5.0 g, 26.88 mmol, 1.0 eq) and 2,4,6- trichloroaniline (5.81 g, 29.56 mmol, 1.1 eq) in toluene (200 mL) is added PCl3 (30 mL), then the suspension is heated at 100 °C for 12 h. The reaction is monitored by LCMS. After completion of reaction, the solvent is removed under reduced pressure; the residue is cooled to 0 °C and basified using saturated NaHCO3 solution. Ethyl acetate (50 mL) is added into it. The product is insoluble in this biphasic system, which is filtered and dried to afford the title compound. [0145] Step-2: Synthesis of 7-(methylthio)-3-(2,4,6-trichlorophenyl)-2,3-dihydro-4H- pyrimido[5,4-e][1,3]oxazin-4-one: To a stirring suspension of 4-hydroxy-2-(methylthio)-N- (2,4,6-trichlorophenyl)pyrimidine-5-carboxamide (1.23 g, 3.38 mmol, 1.0 eq) in CH3CN (30 mL) and DMSO (5 mL) is added cesium carbonate (3.29 g, 10.14 mmol, 3.0eq) and stirred at RT for 5 min. CH2I2 (1.36 g, 5.08 mmol, 1.5 eq) is added and heated at 80 °C for 12 h. The reaction is monitored by LCMS. After completion of the reaction, the solvent is removed under reduced pressure; the residue is diluted with ice-cold water and extracted with ethyl acetate (100 mL × 2). The combined organic layer is washed with brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to give the crude product as a white solid.
[0146] Step-3: Synthesis of tert-butyl 1,1-dimethyl-6-((4-oxo-3-(2,4,6- trichlorophenyl)-3,4-dihydro-2H-pyrimido[5,4-e][1,3]oxazin-7-yl)amino)-3,4- dihydroisoquinoline-2(1H)-carboxylate: To a stirring solution of 7-(methylthio)-3-(2,4,6- trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (160 mg, 0.423 mmol, 1.0 eq) in 5 mL of toluene is added mCPBA (224 mg, 0.84 mmol, 2.0 eq) and stirred at RT for 30 min. Tert-butyl 6-amino-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate ( 117 mg, 0.423 mmol, 1.0 eq) and DIPEA (163 mg, 1.27 mmol, 3.0 eq) are added and allowed to stir at RT for 12 h. The reaction is monitored by LCMS. After completion of the reaction, the solvent is removed under reduced pressure. The residue is diluted with saturated NaHCO3 solution and extracted with CH2Cl2 (100 mL × 2). The combined organic layer is washed with brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to give the crude product which is purified by flash chromatography to give the title compound.
[0147] Step-4: Synthesis of 7-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6- yl)amino)-3-(2,4,6-trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one: To a stirring solution of tert-butyl 1,1-dimethyl-6-((4-oxo-3-(2,4,6-trichlorophenyl)-3,4- dihydro-2H-pyrimido[5,4-e][1,3]oxazin-7-yl)amino)-3,4-dihydroisoquinoline-2(1H)- carboxylate (133 mg, 0.22 mmol, 1.0 eq) in dioxane (1 mL) is added 4.0 M-HCl (1 mL) and allowed to stir at RT for 2h. After completion of the reaction, the reaction mixture is filtered and dried under reduced pressure to afford the desired compound.
Example S-2: 7-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-3-(2,4,6- trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]thiazin-4-one (Compound No.1.2)
Figure imgf000066_0001
[0148] Step-1: Synthesis of 4-hydroxy-2-(methylthio)pyrimidine-5-carboxylic acid: To a stirring solution of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (5 g, 21.425 mmol, 1 eq) in EtOH (25 mL) is added a solution of NaOH (8.38 g, 214.25 mol, 10 eq) in water (25 mL) and the resultant mixture is stirred at RT for 16 h. The progress of the reaction is monitored by TLC. After completion, the reaction mixture is acidified using 2N-HCl (5 mL) to obtain a precipitate which is filtered, washed with water and dried to afford the title compound.
[0149] Step-2: Synthesis of 4-chloro-2-(methylthio)pyrimidine-5-carbonyl chloride : To a stirring solution of 4-hydroxy-2-(methylthio)pyrimidine-5-carboxylic acid (2.5 g, 13.44 mmol, 1 eq) in DMF (2.5 mL) is added SOCl2 (10 mL) and the resultant mixture is heated at 100 °C for 2 h. After completion, the mixture is cooled to RT and concentrated to afford the desired compound.
[0150] Step-3: Synthesis of 4-chloro-2-(methylthio)-N-(2,4,6- trichlorophenyl)pyrimidine-5-carboxamide: To a stirring solution of 2,4,6-trichloroaniline (2.32 g, 11.8 mmol, 1eq) in DCM (20 mL) is added Et3N (4.96 mL, 35.6 mmol, 3 eq) and 4- chloro-2-(methylthio)pyrimidine-5-carbonyl chloride (2.65 g, 11.8 mmol, 1 eq) in DCM (20 mL). The resultant mixture is stirred at RT for 5 h. The progress of the reaction is monitored by TLC. After completion, the reaction mixture is diluted with water (50 mL) and extracted with DCM (50 mL × 3). The combined organic layers are dried over Na2SO4, filtered and concentrated to afford to afford the desired compound [0151] Step-4: Synthesis of 4-mercapto-2-(methylthio)-N-(2,4,6- trichlorophenyl)pyrimidine-5-carboxamide: To a stirring solution of 4-chloro-2- (methylthio)-N-(2,4,6-trichlorophenyl)pyrimidine-5-carboxamide (1.43 g, 3.72 mmol, 1 eq) in DMF (20 mL), water (10 mL) is added Na2S (0.582 g, 4.47 mmol, 1.2 eq) at RT and the resultant mixture is stirred at RT for 1 h. The progress of the reaction is monitored by TLC. After completion, the mixture is diluted with water (50 mL) and extracted with DCM (100 mL x 2). The combined organic layers are dried over Na2SO4, filtered and concentrated to obtain a crude residue which is purified by column chromatography to afford the desired compound.
[0152] Step-5: Synthesis of 7-(methylthio)-3-(2,4,6-trichlorophenyl)-2,3-dihydro-4H- pyrimido[5,4-e][1,3]thiazin-4-one: To a stirring solution of 4-mercapto-2-(methylthio)-N- (2,4,6-trichlorophenyl)pyrimidine-5-carboxamide (0.39 g, 1.011 mmol, 1 eq) in acetonitrile (5 mL) is added Cs2CO3 (1.3 g, 4.043 mmol, 4 eq) at RT and the reaction mixture is stirred for 10 min. Dibromomethane (0.263 g, 1.516 mmol, 3 eq) is added and the mixture is heated at 50 °C for 3 h. The progress of the reaction is monitored by TLC. After completion, the mixture is diluted with water (20 mL) and then extracted with EtOAc (20 mL × 3). The combined organic layers are washed with brine (25 mL), dried over Na2SO4, filtered and concentrated to afford the title compound.
[0153] Step-6: Synthesis of tert-butyl 1,1-dimethyl-6-((4-oxo-3-(2,4,6- trichlorophenyl)-3,4-dihydro-2H-pyrimido[5,4-e][1,3]thiazin-7-yl)amino)-3,4- dihydroisoquinoline-2(1H)-carboxylate: To a stirring solution of 7-(methylthio)-3-(2,4,6- trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]thiazin-4-one (165 mg, 0.423 mmol, 1.0 eq) in 5 mL of toluene is added mCPBA (224 mg, 0.84 mmol, 2.0 eq) and stirred at RT for 30 min. Tert-butyl 6-amino-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate ( 117 mg, 0.423 mmol, 1.0 eq) and DIPEA (163 mg, 1.27 mmol, 3.0 eq) are added and the mixture is allowed to stir at RT for 12 h. The reaction is monitored by LCMS. After completion of the reaction, the solvent is removed under reduced pressure. The residue is diluted with saturated NaHCO3 solution and extracted with CH2Cl2 (100 mL × 2). The combined organic layer is washed with brine solution (50 mL), dried over anhydrous Na2SO4 and concentrate under reduced pressure to give the crude product which is purified by flash chromatography to afford the title compound. [0154] Step-7: Synthesis of 7-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6- yl)amino)-3-(2,4,6-trichlorophenyl)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]thiazin-4-one: To a stirring solution of tert-butyl 1,1-dimethyl-6-((4-oxo-3-(2,4,6-trichlorophenyl)-3,4- dihydro-2H-pyrimido[5,4-e][1,3]thiazin-7-yl)amino)-3,4-dihydroisoquinoline-2(1H)- carboxylate (136 mg, 0.22 mmol, 1.0 eq) in dioxane (1 mL) is added 4.0 M-HCl (1 mL) and allow to stir at RT for 1h. After completion of reaction, the reaction mixture is filtered and dried under reduced pressure to afford the desired compound.
Example S-3: Synthesis of 3-(2,6-dichlorophenyl)-7-((2-(dimethylglycyl)-1,1-dimethyl- 1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (Compound N0.1.48)
Figure imgf000068_0001
[0155] Step-1: Synthesis of 4-hydroxy-2-methylsulfanyl-pyrimidine-5-carboxylic acid: To a stirred solution of ethyl 4-chloro-2-methylsulfanyl-pyrimidine-5-carboxylate (10 g, 43.1 mmol, 1.0 eq) in 150 mL of ethanolwas added water (2:1) and NaOH (17.2 g, 431 mmol, 10 eq). The reaction was heated at 110 °C for 2 h. Progress of the reaction was monitored by LCMS. Upon the consumption of the starting material, the solvent was removed under reduced pressure. The residue was diluted with 100 mL of water and the pH of the mixture was adjusted up to 5 with 3N HCl solution. The precipitated compound was filtered off, washed with water (50 mL) and dried under vacuum to obtain the desired product (6.0 g, 75%). LCMS: 187 [M+1]+ [0156] Step-2: Synthesis of N-(2,6-dichlorophenyl)-4-hydroxy-2-methylsulfanyl- pyrimidine-5-carboxamide: A stirred solution of N-(2,6-dichlorophenyl)-4-hydroxy-2- methylsulfanyl-pyrimidine-5-carboxamide (6.0 g, 32.2 mmol, 1.0 eq) and 2,6-dichloroaniline (5.20 g, 32.2 mmol, 1.0 eq) in 200 mL of toluene was purged with nitrogen gas for 15 min. To the above solution, PCl3 (30 mL) was added. The reaction mixture was heated at 100 °C for 48 h. Progress of the reaction was monitored by LCMS. After completion of reaction, the solvent was removed under reduced pressure, the residue was diluted with 100 mL of mixture of diethyl ether:MeOH (10:1), stirred for 15 min then filtered off. The resulting solid was suspended in MeOH (20 mL), stirred for 5 min, filtered off and washed with MeOH (10 mL), and then dried under vacuum to obtain the title compound (6.0 g, 57%). LCMS: 330 [M+1]+ [0157] Step-3: Synthesis of 3-(2,6-dichlorophenyl)-7-methylsulfanyl-2H- pyrimido[5,4-e][1,3]oxazin-4-one: To a solution of N-(2,6-dichlorophenyl)-4-hydroxy-2- methylsulfanyl-pyrimidine-5-carboxamide (4.0 g, 12.1 mmol, 1.0 eq) in CH3CN (200 mL) was added CH2I2 (4.90 g, 18.5 mmol, 1.5 eq) and Cs2CO3 (12.0 g, 36.4 mmol, 3.0 eq). The reaction mixture was heated at 80 °C for 12 h. Progress of the reaction was monitored by LCMS. After completion of the reaction, the solvent was removed under reduced pressure, the residue was diluted with 50 mL of water and extracted with ethyl acetate (100 mL × 3). The combined organic layer was washed with water (50 mL × 3), dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography to afford the desired compound (400 mg, 10%). LCMS: 342 [M+1]+ [0158] Step-4: Synthesis of tert-butyl 6-(3-(2,6-dichlorophenyl)-4-oxo-3,4-dihydro- 2H-pyrimido[5,4-e][1,3]oxazin-7-ylamino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)- carboxylate: To a stirred solution of 3-(2,6-dichlorophenyl)-7-methylsulfanyl-2H- pyrimido[5,4-e][1,3]oxazin-4-one (200 mg, 0.59 mmol, 1.0 eq) in toluene (3 mL) was added mCPBA (250 mg, 0.147 mmol, 2.5 eq) and allowed to stir at rt for 30 min. Tert-butyl 6- amino-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (162 mg, 0.590 mmol, 1.0 eq) and DIPEA (300 mg, 2.36 mmol, 4.0 eq) were added and allowed to stir at RT for 1 h. Progress of the reaction was monitored by LCMS. After completion of the reaction, the solvent was removed under reduced pressure, the residue was diluted with water (20 mL) and extracted with ethyl acetate (50 mL × 3). The combined organic layer was washed with water (20 mL × 3), dried over anhydrous Na2SO4 and concentrated under reduced pressure. Crude residue was purified by reversed phase chromatography to obtain the desired compound (160 mg, 47.3%). LCMS: 572 [M+1]+ [0159] Step-5: 3-(2,6-dichlorophenyl)-7-(1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin- 6-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin-4(3H)-one: Tert-butyl 6-(3-(2,6- dichlorophenyl)-4-oxo-3,4-dihydro-2H-pyrimido[5,4-e][1,3]oxazin-7-ylamino)-1,1-dimethyl- 3,4-dihydroisoquinoline-2(1H)-carboxylate (160 mg, 0.28 mmol, 1.0 eq) was dissolved in dioxane (2 mL) and 4M HCl in dioxane (2 mL) was added at 0 °C. The reaction mixture was stirred at RT for 1 h. Progress of the reaction was monitored by LCMS. After completion of the reaction, the precipitated compound was filtered off, washed with dioxane (3 mL) and dried under reduced pressure to obtain the desired compound (104 mg, 73.22%) as an off white solid. LCMS: 472 [M+1]+ [0160] Step-6: Synthesis of 3-(2,6-dichlorophenyl)-7-((2-(dimethylglycyl)-1,1- dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-4H-pyrimido[5,4- e][1,3]oxazin-4-one: To a stirred solution of N,N-Dimethylglycine (26.3 mg, 0.25 mmol, 1.2 eq) in DMF (3 mL), HATU (121 mg, 0.31 mmol, 1.5eq) was added and stirred at RT for 5 min.3-(2,6-dichlorophenyl)-7-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3- dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (100 mg, 0.21 mmol, 1.0 eq) and DIPEA (0.14 mL, 0.84 mmol, 4.0 eq) were added and stirred at RT for 16h. After completion of the reaction, the reaction mixture was diluted with ice-cold water (30 mL) and extracted with ethyl acetate (50 mL × 2). The combined organic layer was washed with brine solution (20 mL × 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain the crude product which was purified by reversed phase chromatography to afford (34 mg, 28.79%) as a white solid. LCMS: 555.46 [M+1]+; 1H NMR (400 MHz, DMSO-d6): d ppm 8.84 (s, 1H), 7.66 (d, J = 8.3 Hz, 2H), 7.41 - 7.61 (m, 3H), 7.35 (d, J = 8.7 Hz, 1H) 5.74 (s, 2H), 3.61 (br s, 2H), 3.10 (s, 2H), 2.77 (br s, 2H), 2.19 (s, 6H), 1.82 (s, 3H), 1.67 - 1.79 (m, 3H).
Example S-4: Synthesis of 3-(2,6-dichlorophenyl)-7-((2-(dimethylglycyl)-1,2,3,4- tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (Compound No.1.44)
Figure imgf000070_0001
[0161] Step-1: Synthesis of tert-butyl 7-[[3-(2,6-dichlorophenyl)-4-oxo-2H- pyrimido[5,4-e][1,3]oxazin-7-yl]amino]-3,4-dihydro-1H-isoquinoline-2-carboxylate: To a stirred solution of 3-(2,6-dichlorophenyl)-7-methylsulfanyl-2H-pyrimido[5,4-e][1,3]oxazin- 4-one (200 mg, 0.59 mmol, 1.0 eq) in toluene (3 mL) was added m-CPBA (250 mg, 0.147 mmol, 2.5 eq) and allowed to stir at RT for 30 min. Tert-butyl 7-amino-3,4-dihydro-1H- isoquinoline-2-carboxylate (150 mg, 0.590 mmol, 1.0 eq) and DIPEA (300 mg, 2.36 mmol, 4.0 eq) were added and the mixture was allowed to stir at RT for 1 h. Progress of the reaction was monitored by LCMS. After completion of the reaction, the solvent was removed under reduced pressure, the residue was diluted with water (20 mL) and extracted with ethyl acetate (50 mL × 3). The combined organic layer was washed with water (20 mL × 3), dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude residue was purified by reversed phase chromatography to obtain desired compound (150 mg, 45.6%) as off white solid. LCMS: 542 [M+1]+ [0162] Step-2: Synthesis of 3-(2,6-dichlorophenyl)-7-(1,2,3,4-tetrahydroisoquinolin- 7-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin-4-one hydrochloride: Tert-butyl 7-[[3-(2,6- dichlorophenyl)-4-oxo-2H-pyrimido[5,4-e][1,3]oxazin-7-yl]amino]-3,4-dihydro-1H- isoquinoline-2-carboxylate (150 mg, 0.277 mmol, 1.0 eq) was dissolved in 4M HCl in dioxane (2 mL) at 0 °C. The reaction mixture was stirred at RT for 1 h. Progress of the reaction was monitored by LCMS. After completion of reaction, precipitated compound was filtered off, washed with dioxane (3 mL) and dried under reduced pressure to obtain 3-(2,6- dichlorophenyl)-7-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin- 4-one hydrochloride (100 mg, 75.6%) as HCl salt. LCMS: 442 [M+1]+ [0163] Step-3: Synthesis of 3-(2,6-dichlorophenyl)-7-((2-(dimethylglycyl)-1,2,3,4- tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one: To a stirred solution of N,N-dimethylglycine (27.9 mg, 0.27 mmol, 1.2 eq) in DMF (5 mL), HATU (128.8 mg, 0.33 mmol, 1.5eq) was added and stirred at RT for 5 min.3-(2,6- dichlorophenyl)-7-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-4H-pyrimido[5,4- e][1,3]oxazin-4-one (100 mg, 0.22 mmol, 1.0 eq) and DIPEA (0.15 mL, 0.90 mmol, 4.0 eq) were added and the mixture was stirred at RT for 16 h . After completion of the reaction, the reaction mixture was diluted with ice-cold water (30 mL) and extracted with ethyl acetate (50 mL × 2). The combined organic layer was washed with brine solution (20 mL × 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain the crude product which was purified by reversed phase chromatography to afford the title compound (6.03 mg, 5.1%) as an off-white solid. LCMS: 527.41 [M+1]+;1H NMR (400 MHz, DMSO- d6): d 10.39 (br s, 1H) 8.85 (s, 1H) 7.66 (d, J = 7.9 Hz, 2H) 7.43 - 7.61 (m, 3H) 7.14 (d, J = 8.3 Hz, 1H) 5.74 (s, 2H) 4.75 (s, 1H) 4.58 (s, 1H) 3.74 (d, J = 5.3 Hz, 2H) 3.12 - 3.20 (m, 2H) 2.82 (br s, 2H) 2.19 (d, J = 3.9 Hz, 6H)
Example S-5: Synthesis of 3-(2,6-dichlorophenyl)-7-(2-(2-(dimethylamino)acetyl)-1,2,3,4- tetrahydroisoquinolin-6-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin-4(3H)-one (Compound N0. 1.37)
Figure imgf000072_0001
[0164] Step-1: Synthesis of tert-butyl 6-((3-(2,6-dichlorophenyl)-4-oxo-3,4-dihydro- 2H-pyrimido[5,4-e][1,3]oxazin-7-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 3-(2,6-dichlorophenyl)-7-(methylthio)-2,3-dihydro-4H-pyrimido[5,4- e][1,3]oxazin-4-one (250 mg, 0.730 mmol, 1.0 eq) in 4 mL of toluene was added mCPBA (314 mg, 1.827 mmol, 2.5 eq) and allowed to stir at RT for 30 minutes. Tert-butyl 6-amino- 3,4-dihydroisoquinoline-2(1H)-carboxylate (217 mg, 0.877 mmol, 1.2 eq) and DIPEA (377 mg, 2.923 mmol, 4.0 eq) were added and stirred at RT for 1h. Progress of the reaction was monitored by LCMS. After consumption of the starting material, the solvent was removed under reduced pressure. The residue was diluted with 20 mL of water and extracted with ethyl acetate (200 mL × 2). The combined organic layer was washed with water (50 mL × 3), dried over anhydrous Na2SO4 and concentrated under reduced pressure. The crude product was purified by flash chromatography using MeOH:CH2Cl2 as eluent to obtain 180 mg (37.9%) of tert-butyl 6-((3-(2,6-dichlorophenyl)-4-oxo-3,4-dihydro-2H-pyrimido[5,4- e][1,3]oxazin-7-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate. LCMS: 543 [M+1]+ [0165] Step-2: Synthesis of 3-(2,6-dichlorophenyl)-7-((1,2,3,4-tetrahydroisoquinolin- 6-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one: To tert-butyl 6-((3-(2,6- dichlorophenyl)-4-oxo-3,4-dihydro-2H-pyrimido[5,4-e][1,3]oxazin-7-yl)amino)-3,4- dihydroisoquinoline-2(1H)-carboxylate (150 mg, 0.276 mmol, 1 eq) was added 4 N HCl in dioxane (3 mL). Reaction mixture was stirred at RT for 1h. Progress of the reaction was monitored by LCMS. After consumption of the starting material, the precipitated compound was filtered off and washed with diethyl ether and dried under vacuum to obtain (103 mg, 77.3%) of 3-(2,6-dichlorophenyl)-7-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro- 4H-pyrimido[5,4-e][1,3]oxazin-4-one. LCMS: 443 [M+1]+ [0166] Step-3: Synthesis of 3-(2,6-dichlorophenyl)-7-(2-(2-(dimethylamino)acetyl)- 1,2,3,4-tetrahydroisoquinolin-6-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin-4(3H)-one: To a stirred solution of N,N-dimethylglycine (27.9 mg, 0.27 mmol, 1.2 eq) in DMF (5 mL), HATU (128.8 mg, 0.33 mmol, 1.5 eq) was added and stirred at RT for 5 min.3-(2,6- dichlorophenyl)-7-(1,2,3,4-tetrahydroisoquinolin-6-ylamino)-2H-pyrimido[5,4-e][1,3]oxazin- 4(3H)-one were added and the mixture was stirred at RT for 16 h . After completion of the reaction, the reaction mixture was diluted with ice-cold water (30 mL) and extracted with ethyl acetate (50 mL × 2). The combined organic layer was washed with brine solution (20 mL × 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude product which was purified by reversed phase chromatography to afford the desired compound (6.0 mg, 5.1%) as an off-white solid. LCMS: 527.41 [M+1]+; 1H NMR (400 MHz, DMSO-d6): d 1H NMR (400 MHz, DMSO-d6) d ppm 10.40 (br s, 1H), 8.84 (s, 1H), 7.66 (d, J = 7.89 Hz, 2H), 7.43 - 7.61 (m, 3H), 7.15 (br s, 1H), 5.74 (s, 2H), 4.71 (br s, 1H), 4.55 (br s, 1H), 3.74- 3.65 (m, 2H), 3.15 (s, 2H), 2.85-2.32 (m, 2H), 2.19 (s, 6H).
Example S-6: Synthesis of 3-(2,6-dichlorophenyl)-7-((2-isopropyl-1,2,3,4- tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (Compound No.1.40)
Figure imgf000073_0001
[0167] 3-(2,6-dichlorophenyl)-7-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3- dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (125 mg, 0.28 mmol, 1.0 eq) was dissolved in IPA (3 mL). To this solution were added acetone (0.31 mL, 0.42 mmol, 1.5 eq), acetic Acid (0.1 mL) and NaBH3CN (70.8 mg, 1.12 mmol, 4.0 eq), and reaction mixture was heated at 70 oC for 16 h. After completion the reaction mixture was diluted with ice-cold water (30 mL) and extracted with ethyl acetate (50 mL × 2). Combined organic layer was washed with brine solution (20 mL× 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude product which was purified by reversed phase chromatography to afford the desired product (12 mg 88%) as an off white solid LCMS: 4844 (M+1)+; 1H NMR (400 MHz, DMSO-d6): d 10.55 (br s, 1H), 9.82 (br s, 1H), 8.86 (s, 1H), 7.67 (d, J = 8.3 Hz, 2H), 7.59 (br s, 1H), 7.36 - 7.56 (m, 1H), 7.24 (d, J = 9.2 Hz, 1H), 5.76 (s, 2H), 4.40 (m, 3H), 3.65 (br s, 2H), 3.14 (br s, 2H), 3.04 (br s, 2H), 1.35 (br s, 6H).
Example S-7: Synthesis of 3-(2,6-dichlorophenyl)-7-((2-isopropyl-1,2,3,4- tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (Compound No.1.43)
Figure imgf000074_0001
[0168] 3-(2,6-dichlorophenyl)-7-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3- dihydro-4H-pyrimido[5,4-e][1,3]oxazin-4-one (100 mg, 0.22 mmol, 1.0 eq) was dissolved in Acetone (0.25 mL, 0.33 mmol, 1.5 eq) , IPA (2 mL), NaCNBH3 (57 mg, 0.90 mmol, 4.0 eq), acetic Acid (0.1 mL) was added and heated for 48 h. After completion the reaction mixture was diluted with ice-cold water (30 mL) and extracted with ethyl acetate (50 mL × 2).
Combined organic layer was washed with brine solution (20 mL × 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude product which was purified by reversed phase chromatography to afford the desire compound (10 mg, 8.3%) as an off white solid. LCMS: 484.4 [M+1]+; 1H NMR (400 MHz, DMSO-d6): d 10.33 (br s, 1H), 8.83 (s, 1H), 8.17 (s, 1H), 7.66 (d, J = 8.3 Hz, 2H), 7.36 - 7.55 (m, 3H), 7.03 (d, J = 8.7 Hz, 1H), 5.73 (s, 1H), 3.62 (s, 2H), 2.84 - 2.94 (m, 1H), 2.78 (br s, 2H), 2.65 - 2.75 (m, 2H), 1.06 (d, J = 6.5 Hz, 6H).
[0169] Compounds disclosed herein are prepared according to the experimental details exemplified in Examples S-1 to S-7 and Schemes 1 to 3, using the appropriate starting materials and reagents. Biological Examples
Example B1. WEE1 IC50 Determination
[0170] IC50 values of the compounds against WEE1 kinase enzyme were determined by LanthaScreen™ Terbium Labeled TR-FRET assay. Kinase assays were performed in 1X kinase buffer (#PV6135, Invitrogen, Life Technologies, Grand Island, NY) where total reaction volume was 10 µL in low-volume 384-well plates (#4511,Corning). Serially diluted compounds (3-fold) were incubated with WEE1 Enzyme (1nM) (#PR7373A, Invitrogen, Life Technologies, Grand Island, NY) for 10 min; a mixture of ATP (10 µM) (#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 µL 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) were 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.
[0171] 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.
Figure imgf000075_0001
Example B2. PKMYT1 IC50 Determination
[0172] Inhibition of PKMYT1 kinase activity by test compounds is measured by the HotSpot Kinase Assay at Reaction Biology Corporation (Malvern, PA). Briefly, Myelin Basic Protein substrate is prepared in Reaction Buffer (20 mM Hepes (pH 7.5), 10 mM MgCl2, 1 mM EGTA, 0.01% Brij35, 0.02 mg/mL BSA, 0.1 mM Na3VO4, 2 mM DTT, 1% DMSO). PKMYT1 kinase is delivered into the substrate solution and gently mixed. Test compounds in 100% DMSO are added into the kinase reaction mixture by Acoustic technology (Echo550; nanoliter range) and incubated for 20 min at room temperature.33P- ATP is delivered into the reaction mixture to initiate the reaction. Reactions are carried out at 10 ^M ATP. After a 2 hour incubation at room temperature, kinase activity is detected by P81 filter-binding method. Compounds are 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) is used to calculate the IC50 value of individual test compounds.
Example B3. Determination of potency of compounds in cytotoxicity assay in A427 cell line
[0173] 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 ^L 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 ^L) within the desired testing concentration ranges were added to the culture plate and the cells further incubated in culture conditions for 72 hr. The experiment was terminated at the designated incubation time by replacing the medium with 100 ^L 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. The results are shown in Table 3. Potency of compounds in other cell lines (such as A549, As-Pc-1, Panc-10.05, MDA-MB- 231, A172, U-87MG) may be determined in an analogous manner. The results are shown in Table 3.
Figure imgf000076_0001
Example B4. Determination of potency of compounds in cell proliferation assay in selected cancer cell lines and cellular PD effects. [0174] 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 ^L 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.
[0175] Cells are treated with test compounds at 9 concentrations within a desired concentration range (e.g.1.5 nM– 10 µM) 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-Glo® as recommended by Promega (Cat. No.: G7572, Promega) typically 72 hrs post- treatment.
[0176] 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.
[0177] Test compounds may be studied in the same and/or other cancer cell lines with varying sensitivities to reported Wee1 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.
[0178] pCDC2 and g-H2AX are two clinically relevant biomarkers associated with Wee1 inhibition. CDC2Y15 phosphorylation in cells was reported to be abolished by Wee1 inhibitors (Gavory G et. al., Almac Discovery, AACR poster, 2016). g-H2AX, a DNA double-strand break marker, was upregulated by Wee1 treatment in Wee1 senstive cell lines (Guertin AD et al., Molecular Cancer Therapeutics, 2013). The effects of selected test compounds on pCDC2 and g-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).
[0179] Changes in the levels of phospho-CDC2 following treatment of cells with test compounds are assessed by enzyme-linked immunosorbent assay (ELISA) or Western blotting. A427 cells (or other suitable cell line) are plated in 6-well plates and cultured for 24 hr to approximately 80-90% confluency. Medium is replaced, and the cells are 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 are rinsed with ice-cold PBS and lysed in 1X cell lysis buffer containing protease inhibitors and phosphatase inhibitors. The cells are 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 are 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 are estimated by the Bradford method or equivalent. The ELISA is carried out with the
PathScan® Phospho-CDC2 (Tyr15) Sandwich ELISA Kit (Cat. #7176, Cell Signaling Technology, Danvers, MA) or similar product according to the manufacturer’s instructions. Changes in the levels of phospho-CDC2 may alternatively or additionally be analyzed by Western blotting of the samples using a primary antibody to phospho-CDC2 such as phospho-CDC2 (Tyr15) (10A11) rabbit mAb (Cat. #4539, Cell Signaling Technology) or rabbit polyclonal anti-CDK1 (phospho Y15) antibody (Cat. #ab47594, Abcam, Cambridge, United Kingdom).
Example B6. Evaluation of test compound in mouse xenograft models
[0180] 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 temozolomide), 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.
[0181] 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.
[0182] Although the foregoing invention 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 invention.

Claims

What is claimed is: 1. A compound of Formula (I):
Figure imgf000080_0001
or a salt thereof, wherein:
U is O or S;
W is A or AB, wherein A and B are fused together;
A is phenyl or 5- or 6-membered heteroaryl, each of which is optionally substituted
with R17a, wherein A and R17a together are
Figure imgf000080_0002
and n is 0, 1, 2, 3, or 4;
B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl, each of which is optionally substituted with R17b, wherein A, B, R17a, and R17b
together are and m and n are independently 0, 1, 2, 3, or 4;
Figure imgf000080_0003
X is hydrogen or C1-C6 alkyl;
Y is N or CR1;
Z is N or CR2;
R1 and R2 are independently hydrogen or R17a;
R3a and R3b are independently hydrogen or R17a, or R3a and R3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl;
each R17b is independently oxo or R17a, or any two R17b groups, when bound to the same carbon atom or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R10;
each R17a is independently C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -CN, -OR10, -SR10, -NR11R12, -C(O)R10, -C(O)OR10, -Si(C1-C6 alkyl)3, -C(O)NR11R12,
-OC(O)NR11R12, -NR10C(O)R11, -NR10C(O)NR11R12, -S(O)2R10, -NR10S(O)2R11,
-S(O)2NR11R12, C3-C6 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14 aryl, -(C1-C3 alkylene)CN, -(C1-C3 alkylene)OR10, -(C1-C3 alkylene)SR10, -(C1-C3 alkylene)NR11R12, -(C1-C3 alkylene)CF3, -(C1-C3 alkylene)C(O)R10, -(C1- C3 alkylene)C(O)NR11R12, -(C1-C3 alkylene)NR10C(O)R11, -(C1- C3 alkylene)NR10C(O)NR11R12, -(C1-C3 alkylene)S(O)2R10, -(C1-C3 alkylene)NR10S(O)2R11, -(C1-C3 alkylene)S(O)2NR11R12, -(C1-C3 alkylene)(C3-C6 cycloalkyl), -(C1-C3 alkylene)(3- to 12-membered heterocyclyl), -(C1-C3 alkylene)(5- to 10-membered heteroaryl) or
-(C1-C3 alkylene)(C6-C14 aryl), wherein each R17a is independently optionally substituted by halogen, oxo, -CN, -OR13, -NR13R14, -C(O)R13, -C(O)NR13R14, -NR13C(O)R14, -S(O)2R13, -NR13S(O)2R14, -S(O)2NR13R14, -(C1-C3 alkylene)C(O)NR13R14, -(C1- C3 alkylene)NR13C(O)R14, -(C1-C3 alkylene)S(O)2R13, -(C1-C3 alkylene)NR13S(O)2R14, -(C1- C3 alkylene)S(O)2NR13R14, -(C1-C3 alkylene)(C3-C6 cycloalkyl), -(C1-C3 alkylene)(3- to 12- membered heterocyclyl), -Si(C1-C6 alkyl)3, -CN, -(C1-C3 alkylene)OR13, -(C1- C3 alkylene)NR13R14, -(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, or C1-C6 alkyl optionally substituted by oxo, -OH or halogen;
R4 is 5- to 10-membered heteroaryl or phenyl, wherein the 5- to 10-membered heteroaryl and phenyl of R4 are each independently optionally substituted by halogen, oxo, -OR13, -NR13R14, - C(O)NR13R14, -C(O)R13, -CN, C3-C8 cycloalkyl, or C1-C6 alkyl optionally substituted by oxo, -CN, -OH or halogen;
R10 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6- membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, -CN, -OR15, -NR15R16, or C1-C6 alkyl optionally substituted by halogen, -OH or oxo; R11 and R12 are each independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl of R11 and R12 are independently optionally substituted by halogen, oxo, -CN, -OR15, -NR15R16 or C1-C6 alkyl optionally substituted by halogen, -OH, or oxo,
or R11 and R12 are taken together with the atom to which they attached to form a 3- to 6- membered heterocyclyl optionally substituted by halogen, oxo, or C1-C6 alkyl optionally substituted by halogen;
R13 and R14 are each independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C3-C6 cycloalkyl, or 3- to 6-membered heterocyclyl of R13 and R14 are optionally substituted by halogen, -CN, -OR15, -NR15R16, or oxo,
or R13 and R14 are taken together with the atom to which they attached to form a 3- to 6- membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by halogen or oxo; and
R15 and R16 are each independently hydrogen, C1-C6 alkyl optionally substituted by halogen or oxo, C2-C6 alkenyl optionally substituted by halogen or oxo, or C2-C6 alkynyl optionally substituted by halogen or oxo,
or R15 and R16 are taken together with the atom to which they attached to form a 3- to 6- membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by oxo or halogen.
2. The compound of claim 1, or a salt thereof, wherein Y is N.
3. The compound of claim 1 or 2, or a salt thereof, wherein Y is CR1 and R1 is hydrogen.
4. The compound of any one of claims 1-3, or a salt thereof, wherein Z is N.
5. The compound of any one of claims 1-4, or a salt thereof, wherein Z is CR2 and R2 is hydrogen.
6. The compound of any one of claims 1-5, or a salt thereof, wherein at least one of Y and Z is N.
7. The compound of claim 1, or a salt thereof, wherein the compound is of Formula (Ia- 1):
Figure imgf000083_0001
8. The compound of claim 1, or a salt thereof, wherein the compound is of Formula (Ib- 1):
Figure imgf000083_0002
9. The compound of any one of claims 1-8, or a salt thereof, wherein X is hydrogen.
10. The compound of any one of claims 1-9, or a salt thereof, wherein R3a and R3b are independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, -CN, -OR10, -SR10, -NR11R12, -C(O)R10, or -C(O)NR11R12; or R3a and R3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl.
11. The compound of any one of claims 1-10, or a salt thereof, wherein R3a and R3b are independently hydrogen or C1-C6 alkyl; or R3a and R3b are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl.
12. The compound of any one of claims 1-11, or a salt thereof, wherein R3a and R3b are both hydrogen.
13. The compound of any one of claims 1-12, or a salt thereof, wherein R4 is phenyl optionally substituted by halogen, -OR13, -NR13R14, - C(O)NR13R14, -C(O)R13, -CN, C3-C8 cycloalkyl, or C1-C6 alkyl optionally substituted by oxo, -CN, -OH or halogen.
14. The compound of any one of claims 1-13, or a salt thereof, wherein R4 is
Figure imgf000083_0003
Figure imgf000083_0004
15. The compound of any one of claims 1-14, or a salt thereof, wherein W is AB wherein A and B are fused together;
A is phenyl or 6-membered heteroaryl, each of which is optionally substituted with
R17a, wherein A and R17a together are
Figure imgf000084_0001
and n is 0, 1, 2, 3, or 4;
B is C3-C6 cycloalkyl, 3- to 7-membered heterocyclyl, 5- to 7-membered heteroaryl, or C6 aryl, each of which is optionally substituted with R17b, wherein A, B, R17a, and R17b
together are
Figure imgf000084_0002
and m and n are independently 0, 1, 2, 3, or 4.
16. The compound of claim 15, or a salt thereof, wherein A, B, R17a and R17b together are
Figure imgf000084_0003
17. The compound of claim 15 or 16, or a salt thereof, wherein B is 3- to 7-membered heterocyclyl;
each R17a is independently -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3- C6 cycloalkyl, or -OR10 ;
each R17b is independently oxo, -(C1-C3 alkylene)OR10, C1-C6 alkyl optionally substituted by halogen, -C(O)NR11R12, -(C1-C3 alkylene)NR11R12, -CN, halogen, -NR11R12, C3-C6 cycloalkyl, or -OR10, or
any two groups R17b, when bound to the same carbon atom, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl;
and m and n are independently 0, 1, 2, or 3.
18. The compound of any one of claims 15-17, or a salt thereof, wherein m is 0.
19. The compound of any one of claims 15-17, or a salt thereof, wherein m is 1.
20. The compound of any one of claims 15-19, or a salt thereof, wherein n is 0.
21. The compound of any one of claims 15-19, or a salt thereof, wherein n is 1.
22. The compound of any one of claims 15-19, or a salt thereof, wherein n is 2.
23. The compound of any one of claims 1-14, or a salt thereof, wherein W is selected from the group consisting of:
Figure imgf000085_0001
, and , wherein the wavy lines denote attachment
Figure imgf000086_0001
Figure imgf000086_0002
points to the parent molecule.
24. The compound of any one of claims 1-14, or a salt thereof, wherein W is A, A is phenyl or 5- or 6-membered heteroaryl, each of which is optionally substituted with R17a,
wherein A and R17a together are
Figure imgf000086_0003
and n is 0, 1, 2, 3, or 4.
25. The compound of claim 24, wherein W is selected from the group consisting of:
Figure imgf000086_0004
Figure imgf000086_0005
, wherein the wavy lines denote attachment points to the parent molecule.
26. A compound selected from the group consisting of the compounds in Table 1, or a salt thereof.
27. The compound of claim 26, wherein the compound is a pharmaceutically acceptable salt of a compound in Table 1.
28. A pharmaceutical composition comprising a compound of any one of claims 1-26, or a salt thereof, and a pharmaceutically acceptable carrier.
29. A method of treating a cancer in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound of any one of claims 1-26, or a salt thereof.
30. The method of claim 29, further comprising administering a radiation therapy to the individual.
31. The method of claim 29 or 30, further comprising administering to the individual a therapeutically effective amount of an additional therapeutic agent.
32. The method of claim 31, wherein the additional therapeutic agent is a cancer immunotherapy agent or a chemotherapeutic agent.
33. The method of claim 31 or 32, 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.
34. The method of any one of claims 29-33, wherein the cancer comprises a mutant TP53 gene.
35. The method of any one of claims 29-34, 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.
36. A method of suppressing a G2-M checkpoint in a cell, comprising administering a compound of any one of claims 1-26, or a salt thereof, to the cell.
37. A method of inducing premature mitosis in a cell, comprising administering a compound of any one of claims 1-26, or a salt thereof, to the cell.
38. A method of inducing apoptosis in a cell, comprising administering a compound of any one of claims 1-26, or a salt thereof, to the cell.
39. A method of inhibiting Wee1 in a cell, comprising administering a compound of any one of claims 1-26, or a salt thereof, to the cell.
40. A method of inhibiting Wee1, comprising contacting Wee1 with a compound of any one of claims 1-26, or a salt thereof.
41. The method of claim 40, wherein the inhibitor binds to Wee1 with an IC50 of less than 1 µM according to a kinase assay.
42. Use of a compound of any one of claims 1-26, or a salt thereof, in the manufacture of a medicament for treatment of cancer.
43. A kit comprising a compound of any one of claims 1-26, or a salt thereof.
PCT/US2020/027308 2019-04-09 2020-04-08 Heterocyclic compounds and uses thereof WO2020210383A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/594,299 US20220168313A1 (en) 2019-04-09 2020-04-08 Heterocyclic compounds and uses thereof
EP20787952.9A EP3952878A4 (en) 2019-04-09 2020-04-08 Heterocyclic compounds and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962831677P 2019-04-09 2019-04-09
US62/831,677 2019-04-09

Publications (1)

Publication Number Publication Date
WO2020210383A1 true WO2020210383A1 (en) 2020-10-15

Family

ID=72750567

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/027308 WO2020210383A1 (en) 2019-04-09 2020-04-08 Heterocyclic compounds and uses thereof

Country Status (3)

Country Link
US (1) US20220168313A1 (en)
EP (1) EP3952878A4 (en)
WO (1) WO2020210383A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11332473B2 (en) 2019-04-09 2022-05-17 Nuvation Bio Inc. Substituted pyrazolo[3,4-d]pyrimidines as Wee1 inhibitors

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6995153B2 (en) * 2002-01-18 2006-02-07 Kyorin Pharmaceutical. Co., Ltd. Fused bicyclic pyrimidine derivatives
US20060069093A1 (en) * 2004-09-29 2006-03-30 Portola Pharmaceuticals, Inc. Substituted 2H-1,3-benzoxazin-4(3H)-ones
US9655899B2 (en) * 2012-11-28 2017-05-23 Merck Sharp & Dohme Corp. Compositions and methods for treating cancer
WO2018090939A1 (en) * 2016-11-16 2018-05-24 上海瑛派药业有限公司 8,9-dihydroimidazole[1,2-a]pyrimido[5,4-e]pyrimidine-5(6h)-ketone compound
WO2018162932A1 (en) * 2017-03-10 2018-09-13 Almac Discovery Limited Pyrimidopyrimidinones useful as wee-1 kinase inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2168966B1 (en) * 2007-06-15 2016-09-28 Msd K.K. Bicycloaniline derivative
GB201322602D0 (en) * 2013-12-19 2014-02-05 Almac Discovery Ltd Pharmaceutical compounds
CA3078945A1 (en) * 2017-10-09 2019-04-18 Nuvation Bo Inc. Heterocyclic compounds and uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6995153B2 (en) * 2002-01-18 2006-02-07 Kyorin Pharmaceutical. Co., Ltd. Fused bicyclic pyrimidine derivatives
US20060069093A1 (en) * 2004-09-29 2006-03-30 Portola Pharmaceuticals, Inc. Substituted 2H-1,3-benzoxazin-4(3H)-ones
US9655899B2 (en) * 2012-11-28 2017-05-23 Merck Sharp & Dohme Corp. Compositions and methods for treating cancer
WO2018090939A1 (en) * 2016-11-16 2018-05-24 上海瑛派药业有限公司 8,9-dihydroimidazole[1,2-a]pyrimido[5,4-e]pyrimidine-5(6h)-ketone compound
WO2018162932A1 (en) * 2017-03-10 2018-09-13 Almac Discovery Limited Pyrimidopyrimidinones useful as wee-1 kinase inhibitors

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11332473B2 (en) 2019-04-09 2022-05-17 Nuvation Bio Inc. Substituted pyrazolo[3,4-d]pyrimidines as Wee1 inhibitors

Also Published As

Publication number Publication date
US20220168313A1 (en) 2022-06-02
EP3952878A1 (en) 2022-02-16
EP3952878A4 (en) 2023-01-04

Similar Documents

Publication Publication Date Title
US11299493B2 (en) Heterocyclic compounds and uses thereof
US10807994B2 (en) Heterocyclic compounds and uses thereof
JP2021098721A (en) Substituted quinazoline compounds and methods for use thereof
AU2020329288A1 (en) Heterocyclic compounds as kinase inhibitors
WO2020210380A1 (en) Heterocyclic compounds and uses thereof
US11332473B2 (en) Substituted pyrazolo[3,4-d]pyrimidines as Wee1 inhibitors
EP3952876A1 (en) Heterocyclic compounds and uses thereof
WO2021003314A1 (en) Heterocyclic compounds as kinase inhibitors
WO2022082174A1 (en) Heterocyclic compounds and uses thereof
WO2020210383A1 (en) Heterocyclic compounds and uses thereof
EP3952879A1 (en) Heterocyclic compounds and uses thereof
WO2022236256A1 (en) Heterocyclic compounds as kinase inhibitors
WO2021030620A1 (en) Heterocyclic compounds as kinase inhibitors
WO2022236253A1 (en) Heterocyclic compounds as kinase inhibitors
WO2022236255A2 (en) Heterocyclic compounds as kinase inhibitors
WO2022236257A1 (en) Heterocyclic compounds as kinase inhibitors
WO2023226658A1 (en) Nitrogen-containing five-membered heterocyclic derivatives as checkpoint kinase 1 inhibitor and uses thereof

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: 20787952

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020787952

Country of ref document: EP

Effective date: 20211109