US20230227446A9 - Phosphatidylinositol 3-kinase inhibitors - Google Patents

Phosphatidylinositol 3-kinase inhibitors Download PDF

Info

Publication number
US20230227446A9
US20230227446A9 US17/602,206 US202017602206A US2023227446A9 US 20230227446 A9 US20230227446 A9 US 20230227446A9 US 202017602206 A US202017602206 A US 202017602206A US 2023227446 A9 US2023227446 A9 US 2023227446A9
Authority
US
United States
Prior art keywords
compound
mmol
cyclopropylethyl
pharmaceutically acceptable
solvate
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
US17/602,206
Other versions
US20220213096A1 (en
Inventor
Xiaolin Hao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hangzhou Zhengxiang Pharmaceuticals Co Ltd
Nanjing Zhengxiang Pharmaceuticals Co Ltd
Original Assignee
Hangzhou Zhengxiang Pharmaceuticals Co Ltd
Nanjing Zhengxiang Pharmaceuticals Co Ltd
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 Hangzhou Zhengxiang Pharmaceuticals Co Ltd, Nanjing Zhengxiang Pharmaceuticals Co Ltd filed Critical Hangzhou Zhengxiang Pharmaceuticals Co Ltd
Priority to US17/602,206 priority Critical patent/US20230227446A9/en
Assigned to NANJING ZHENGXIANG PHARMACEUTICALS CO., LTD. reassignment NANJING ZHENGXIANG PHARMACEUTICALS CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HANGZHOU ZHENGXIANG PHARMACEUTICALS CO., LTD.
Assigned to HANGZHOU ZHENGXIANG PHARMACEUTICALS CO., LTD. reassignment HANGZHOU ZHENGXIANG PHARMACEUTICALS CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HAO, XIAOLIN
Publication of US20220213096A1 publication Critical patent/US20220213096A1/en
Publication of US20230227446A9 publication Critical patent/US20230227446A9/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed 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
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • 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
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53861,4-Oxazines, e.g. morpholine spiro-condensed or forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • 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
    • C07D498/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65583Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system each of the hetero rings containing nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65586Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system at least one of the hetero rings does not contain nitrogen as ring hetero atom

Definitions

  • the present disclosure relates generally to novel inhibitors of phosphatidylinositol 3-kinase (PI3K). More specifically, the invention further relates to the preparation of the disclosed PI3K gamma inhibitor analogs and their use in pharmaceutical compositions for the treatment of various diseases, conditions and disorders related to PI3K gamma activity.
  • PI3K phosphatidylinositol 3-kinase
  • PIP3 The class I phosphoinositide 3-kinases (PI3Ks) regulate phosphatidylinositol 4,5-bisphosphate (PIP2) phosphorylation.
  • PIP2 phosphatidylinositol 4,5-bisphosphate
  • PIP3 phosphatidylinositol (3,4,5)-trisphosphate
  • PIP3 plays a key regulatory role in cell survival, signal transduction, control of membrane trafficking and other functions.
  • Class I PI3Ks consist of four kinases further delineated into 2 subclasses.
  • Class 1A PI3Ks consist of three closely related kinases, PI3K ⁇ , ⁇ , and ⁇ existing as heterodimers composed of a catalytic subunit (p110 ⁇ , ⁇ or ⁇ ) and one of several regulatory subunits. They generally respond to signaling through receptor tyrosine kinases (RTKs).
  • PI3K ⁇ single class 1B isoform, responds mainly to G-protein coupled receptors (GPCRs), and is composed of a p110 ⁇ catalytic subunit and one of two distinct regulatory subunits.
  • GPCRs G-protein coupled receptors
  • PI3K ⁇ and PI3K ⁇ are ubiquitously expressed throughout a wide variety of tissue and organ types.
  • the expression pattern of PI3K ⁇ is restricted, to spleen, thymus, and peripheral blood leukocytes (Knight, Z. et al. Cell 2006, 125, 733).
  • PI3K ⁇ is found mainly in leukocytes, but also in skeletal muscle, liver, pancreas, and heart (Cantly, C. Science 2002, 1655).
  • PI3k ⁇ serves as a single convergent point promoting tumor inflammation and progression.
  • treatment of mice with selective PI3K ⁇ inhi-biter inhibited myeloid cell p110 ⁇ catalytic activity and adhesion to VCAM-1, due to effect on the tumor microenvironment instead of direct inhibition of tumor cells.
  • PI3K ⁇ controls the TAM switch between immune suppression and immune stimulation. (Kaneda, M. M. et al. Nature 539, 437-442, 2016; De Henau, O. et al. Nature 539, 443-447, 2016).
  • Camps et al. described that treatment with selective PI3K ⁇ inhibitor AS-60485023 suppresses the progression of joint inflammation and damage in two distinct mouse models of rheumatoid arthritis (Camps M, et al., Nat. Med. 2005, 11, 936-943).
  • PI3K ⁇ inhibitors could potentially be used to treat a variety of diseases such as inflammation, metabolic and cancer (Cushing, T. D., et al, J. Med. Chem. 2012, 55, 8559-8581; Ruckle, T. et al, Nat. Rev. Drug Discovery 2006, 5, 903-918; Stark, A. K. et al, Curr. Opin. Pharmacol. 2015, 23, 82-91).
  • PI3K gamma selective inhibitors have been disclosed in recent years. IPI-549 has been in the clinical trials as single agent and combination immuno-oncology therapies with check point agent PD-1 inhibitor Nivolumab (Evans, C. A. et al, ACS Med. Chem. Lett. 2016, 7, 862-867). Pemberton N et al reported that selective PI3K gamma inhibition resulted in a dose dependent inhibition of LPS-induced airway neutrophilia in rats (Pemberton, N. et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441). Modification of a series of azaisoindolinones by Come, J. H.
  • PI3K gamma inhibitors with related structure but different binding mode have been disclosed in recent years.
  • WO2015048318 disclosed (R)-6-(1-(2,2-difluoroethyl)-1H-pyrazol-4-yl)-4,7,7-trimethyl-2-(5-(2,2,2-trifluoro-1-hydroxyethyl)pyhdin3-yl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-5-one as a selective inhibitor of PI3K gamma.
  • WO2011087776 disclosed Isoindolinone inhibitors of phosphatidylinositol 3-kinase.
  • WO2017153527 disclosed novel inhibitors of phosphatidylinositol 3-kinase gamma. Discovery of highly isoform selective orally bioavailable phosphoinositide 3-kinase (PI3K) ⁇ inhibitors, and design and synthesis of a novel series of 3-kinase ⁇ (PI3K ⁇ ) inhibitors were reported (Journal of Medicinal Chemistry 2018, 61, 5435-5441 and Journal of Medicinal Chemistry 2018, 61, 5245-5256).
  • PI3K phosphoinositide 3-kinase
  • compositions including pharmaceutical compositions that include the compounds are also provided, as are methods of using and making the compounds.
  • the compounds provided herein may find use in treating diseases, disorders, or conditions that are mediated by PI3K isoforms, such as PI3K gamma.
  • A is CH 2 , CH(C 1-6 alkyl), O, or S:
  • Y is CH or N
  • Z is CH or N
  • W is N, CH, or CX
  • X is selected from the group consisting of
  • each 3-10 membered heterocyclyl independently contains 1 or 2 heteroatoms, wherein the 1 or 2 heteroatoms are selected from the group consisting of O, N, and S, and wherein each C 3-10 cycloalkyl or 3-10 membered heterocyclyl is independently optionally substituted with one or more G, wherein:
  • Q 1 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with one or more OH or halo, and
  • each G is independently selected from the group consisting of H, D, OH, C 1-6 alkoxy, oxo, NH 2 , SO 2 (C 1-6 alkyl), C(O)—C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclyl, and C 1-6 alkyl, wherein the C 3-10 cycloalkyl or C 1-6 alkyl is independently optionally substituted with one or more D, OH, C 1-6 alkoxy, CN, N(C 1-6 alkyl) 2 , SO 2 (C 1-6 alkyl), or halo, or
  • X is selected from the group consisting of
  • R 1 is C 1-6 alkyl, C 3-10 cycloalkyl, or C 1-6 alkyl-C 3-10 cycloalkyl, wherein the C 1-6 alkyl, C 3-10 cycloalkyl, or C 1-6 alkyl-C 3-10 cycloalkyl is independently optionally substituted with one or more halo; and R 2 is selected from the group consisting of
  • L is H, halo, or C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with one or more halo, C 1-6 alkoxy, or OH, and
  • each Q 2 and Q 3 is independently C 1-6 alkyl, C 3-10 cycloalkyl, or 3-10 membered heterocyclyl, wherein the C 1-6 alkyl, C 3-10 cycloalkyl, or 3-10 membered heterocyclyl is independently optionally substituted with one or more halo.
  • the compound described herein has improved selectivity against the PI3K ⁇ isoform, improved solubility, or improved oral bioavailability in comparison to a known PI3K gamma inhibitor (e.g., IPI-549 (Evans, C. et al, ACS Med. Chem. Lett. 2016, 7, 862-867) or AZ-17 (Pemberton, N, et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441)), or any combination of the foregoing.
  • a known PI3K gamma inhibitor e.g., IPI-549 (Evans, C. et al, ACS Med. Chem. Lett. 2016, 7, 862-867) or AZ-17 (Pemberton, N, et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441)
  • a method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma comprising contacting the PI3K ⁇ with a compound of Formula (I), (I-a), (I-b), (I-c), or (I-d), or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof, and one or more pharmaceutically acceptable carriers.
  • PI3K ⁇ phosphoinositide 3-kinase gamma
  • PI3K ⁇ phosphoinositide 3-kinase gamma
  • the compound has improved selectivity against the PI3K ⁇ isoform, improved solubility, or improved oral bioavailability in comparison to a known PI3K gamma inhibitor (e.g., IPI-549 or AZ-17), or any combination of the foregoing.
  • PI3K gamma PI3K ⁇
  • a method of treating a disorder of uncontrolled cellular proliferation related to related to one or more PI3K isoforms, such as PI3K gamma (PI3K ⁇ ) comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), (I-a), (I-b), (I-c), or (I-d), or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • PI3K ⁇ PI3K gamma
  • provided herein is a method of treating an autoimmune diseases, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), (I-a), (I-b), (I-c), or (I-d), or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • provided herein is a method of treating a cancer, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), (I-a), (I-b), (I-c), or (I-d), or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • FIG. 1 shows the antitumor efficacy of an exemplary compound (compound 34) in 4T1 syngeneic mouse tumor model.
  • FIG. 2 shows that treatment with an exemplary compound (compound 34) at 20 mg/kg and 80 mg/kg BID significantly ameliorated the clinical disability symptom of EAE induced mice.
  • PI3K gamma inhibitors are compounds that function as PI3K gamma inhibitors.
  • A is CH 2 , CH(C 1-6 alkyl), O, or S;
  • Y is CH or N:
  • Z is CH or N
  • W is N, CH, or CX; wherein X is selected from the group consisting of
  • each 3-10 membered heterocyclyl independently contains 1 or 2 heteroatoms, wherein the 1 or 2 heteroatoms are selected from the group consisting of O, N, and S, and wherein each C 3-10 cycloalkyl or 3-10 membered heterocyclyl is independently optionally substituted with one or more G, wherein:
  • Q 1 is C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with one or more OH or halo, and
  • each G is independently selected from the group consisting of H, D, ON, C 1-6 alkoxy, oxo, NH 2 , SO 2 (C 1-6 alkyl), C(O)—C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclyl, and C 1-6 alkyl, wherein the C 3-10 cycloalkyl or C 1-6 alkyl is independently optionally substituted with one or more D, OH, C 1-6 alkoxy, CN, N(C 1-6 alkyl) 2 , SO 2 (C 1-6 alkyl), or halo, or
  • X is selected from the group consisting of
  • R 1 is C 1-6 alkyl, C 3-10 cycloalkyl, or C 1-6 alkyl-C 3-10 cycloalkyl, wherein the C 1-6 alkyl, C 3-10 cycloalkyl, or C 1-6 alkyl-C 3-10 cycloalkyl is independently optionally substituted with one or more halo; and R 2 is selected from the group consisting of
  • L is H, halo, or C 1-6 alkyl, wherein the C 1-6 alkyl is optionally substituted with one or more halo, C 1-6 alkoxy, or OH, and
  • each Q 2 and Q 3 is independently C 1-6 alkyl, C 3-10 cycloalkyl, or 3-10 membered heterocyclyl, wherein the C 1-6 alkyl, C 3-10 cycloalkyl, or 3-10 membered heterocyclyl is independently optionally substituted with one or more halo.
  • At least one of Y, Z and W is N. In some embodiments, Y is N and Z is CH. In some embodiments, Z is N and Y is CH.
  • X is selected from the group consisting of
  • X is selected from the group consisting of NHG, CHG 2 , COOH, OG, SO 2 G, SO 2 NHG, NGSO 2 G, C 1-6 alkyl-NGSO 2 G, NHC(O)G, NHC(O)NG 2 , C(O)NHG, C(O)NG 2 , C 3-10 cycloalkyl, and 3-10 membered heterocyclyl, wherein each 3-10 membered heterocyclyl independently contains 1 or 2 heteroatoms, wherein the 1 or 2 heteroatoms are selected from the group consisting of O, N, and S, and wherein each C 3-10 cycloalkyl or 3-10 membered heterocyclyl is independently optionally substituted with one or more G.
  • Y and Z are each CH, W is CX, and X is selected from the group consisting of
  • Y is N
  • Z is CH
  • W is CX
  • X is selected from the group consisting of NHG, CHG 2 , COOH, OG, SO 2 G, SO 2 NHG, NGSO 2 G, C 1-6 alkyl-NGSO 2 G, NHC(O)G, NHC(O)NG 2 , C(O)NHG, C(O)NG 2 , C 3-10 cycloalkyl, and 3-10 membered heterocyclyl, wherein each 3-10 membered heterocyclyl independently contains 1 or 2 heteroatoms, wherein the 1 or 2 heteroatoms are selected from the group consisting of O, N, and S, and wherein each C 3-10 cycloalkyl or 3-10 membered heterocyclyl is independently optionally substituted with one or more G.
  • R 2 is selected from the group consisting of
  • R 2 is
  • R 2 is selected from the group consisting of
  • Q 2 is methyl, ethyl, isopropyl, cyclopropyl, or difluoromethyl.
  • R 2 is
  • R 1 is selected from the group consisting of
  • R 1 is selected from the group consisting of
  • R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • R 1 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • X is selected from the group consisting of
  • X is selected from the group consisting of
  • X is selected from the group consisting of NHG, OG, NHSO 2 G, and C(O)NG 2 . In still other embodiments, X is selected from the group consisting of OMe, OCD 3 , NHSO 2 Me, NHSO 2 Et, C(O)NH 2 , C(O)NHMe, and C(O)NMe 2 .
  • a compound of the following structures (Table 1), or a pharmaceutically acceptable salt, prodrug, or solvate wherein:
  • compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or pharmaceutically acceptable salts, prodrugs, or solvates thereof.
  • compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or pharmaceutically acceptable salts, stereoisomers, prodrugs, or solvents thereof.
  • Alkyl refers to and includes, unless otherwise stated, a saturated linear (i.e., unbranched) or branched univalent hydrocarbon chain or combination thereof, having the number of carbon atoms designated (i.e., C 1 -C 10 means one to ten carbon atoms).
  • Particular alkyl groups are those having 1 to 20 carbon atoms (a “C 1 -C 20 alkyl”), having 1 to 10 carbon atoms (a “C 1 -C 10 alkyl”), having 6 to 10 carbon atoms (a “C 6 -C 10 alkyl”), having 1 to 6 carbon atoms (a “C 1 -C 6 alkyl”), having 2 to 6 carbon atoms (a “C 2 -C 6 alkyl”), or having 1 to 4 carbon atoms (a “C 1 -C 4 alkyl”).
  • alkyl groups include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, and the like.
  • Cycloalkyl refers to and includes, unless otherwise stated, saturated cyclic univalent hydrocarbon structures, having the number of carbon atoms designated (i.e., C 3 -C 10 means three to ten carbon atoms). Cycloalkyl can consist of one ring, such as cyclohexyl, or multiple rings, such as adamantyl. A cycloalkyl comprising more than one ring may be fused, spiro or bridged, or combinations thereof. Particular cycloalkyl groups are those having from 3 to 12 annular carbon atoms.
  • a preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a “C 3 -C 8 cycloalkyl”), having 3 to 6 carbon atoms (a “C 3 -C 6 cycloalkyl”), or having from 3 to 4 annular carbon atoms (a “C 3 -C 4 cycloalkyl”).
  • Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, and the like.
  • Heteroaryl refers to an unsaturated aromatic cyclic group having from 1 to 14 annular carbon atoms and at least one annular heteroatom, including but not limited to heteroatoms such as nitrogen, oxygen and sulfur.
  • a heteroaryl group may have a single ring (e.g., pyridyl, furyl) or multiple condensed rings (e.g., indolizinyl, benzothienyl) which condensed rings may or may not be aromatic.
  • Particular heteroaryl groups are 5 to 14-membered rings having 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 5 to 10-membered rings having 1 to 8 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, or 5, 6 or 7-membered rings having 1 to 5 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • particular heteroaryl groups are monocyclic aromatic 5-, 6- or 7-membered rings having from 1 to 6 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • particular heteroaryl groups are polycyclic aromatic rings having from 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • a heteroaryl group having more than one ring where at least one ring is non-aromatic may be connected to the parent structure at either an aromatic ring position or at a non-aromatic ring position.
  • a heteroaryl group having more than one ring where at least one ring is non-aromatic is connected to the parent structure at an aromatic ring position.
  • a heteroaryl group may be connected to the parent structure at a ring carbon atom or a ring heteroatom.
  • Heterocycle refers to a saturated or an unsaturated non-aromatic cyclic group having a single ring or multiple condensed rings, and having from 1 to 14 annular carbon atoms and from 1 to 6 annular heteroatoms, such as nitrogen, sulfur or oxygen, and the like.
  • a heterocycle comprising more than one ring may be fused, bridged or spiro, or any combination thereof, but excludes heteroaryl groups.
  • the heterocyclyl group may be optionally substituted independently with one or more substituents described herein.
  • Particular heterocyclyl groups are 3 to 14-membered rings having 1 to 13 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 12-membered rings having 1 to 11 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 10-membered rings having 1 to 9 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 8-membered rings having 1 to 7 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, or 3 to 6-membered rings having 1 to 5 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • heterocyclyl includes monocyclic 3-, 4-, 5-, 6- or 7-membered rings having from 1 to 2, 1 to 3, 1 to 4, 1 to 5, or 1 to 6 annular carbon atoms and 1 to 2, 1 to 3, or 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • heterocyclyl includes polycyclic non-aromatic rings having from 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • Halo or “halogen” refers to elements of the Group 17 series having atomic number 9 to 85.
  • Preferred halo groups include the radicals of fluorine, chlorine, bromine and iodine. 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 halogen; 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 trifluoromethyl (—CF 3 ).
  • Oxo refers to the moiety ⁇ O.
  • 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, 1 to 3, 1 to 4, 1 to 5, 2 to 3, 2 to 4, or 2 to 5 substituents.
  • an optionally substituted group is unsubstituted.
  • “Pharmaceutically-acceptable salt” means a salt prepared by conventional means, and are well known by those skilled in the art.
  • the “pharmacologically acceptable salts” include basic salts of inorganic and organic acids (Berge et al., J. Pharm. Sci. 1977, 66:1).
  • “Pharmaceutically acceptable salts” are those salts which retain at least some of the biological activity of the free (non-salt) compound and which can be administered as drugs or pharmaceuticals to an individual.
  • Such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, oxalic acid, propionic acid, succinic acid, maleic acid, tartaric acid and the like; (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base.
  • Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine and the like.
  • Acceptable inorganic bases which can be used to prepared salts include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.
  • Pharmaceutically acceptable salts can be prepared in situ in the manufacturing process, or by separately reacting a purified compound of the invention in its free acid or base form with a suitable organic or inorganic base or acid, respectively, and isolating the salt thus formed during subsequent purification.
  • “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.
  • excipient means an inert or inactive substance that may be used in the production of a drug or pharmaceutical, such as a tablet containing a compound of the invention as an active ingredient.
  • a drug or pharmaceutical such as a tablet containing a compound of the invention as an active ingredient.
  • Various substances may be embraced by the term excipient, including without limitation any substance used as a binder, disintegrant, coating, compression/encapsulation aid, cream or lotion, lubricant, solutions for parenteral administration, materials for chewable tablets, sweetener or flavoring, suspending/gelling agent, or wet granulation agent.
  • a “solvate” is formed by treating a compound in a solvent. Solvates of salts of the compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d) are also provided. In the case of treating compounds with water, the solvate is hydrates. Hydrates of the compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d) are also provided.
  • a “prodrug” includes any compound that converts into a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), when administered to a subject, e.g., upon metabolic processing of the prodrug.
  • the compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof may be used for treating PI3K mediated diseases or disorders.
  • selected compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, have improved physicochemical and pharmacokinetic properties, for an example, improved solubility in water and oral bioavailability.
  • treatment is an approach for obtaining beneficial or desired results including clinical results.
  • beneficial or desired results include, but are not limited to, one or more of the following: decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of an individual.
  • “treatment” of a disorder does not include prevention of the disorder, and “prevention” is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a disorder or biological manifestation thereof, or to delay the onset of such disorder or biological manifestation thereof.
  • 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 is an amount sufficient to delay development. In some embodiments, an effective amount is an amount sufficient to prevent or delay recurrence.
  • 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 “subject” is a mammal, including humans.
  • a subject includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate. In some embodiments, the subject is human (including adults and children).
  • “Inhibition of PI3K gamma activity” or variants refer to a decrease in PI3K gamma activity as a direct or indirect response to the presence of a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, relative to the activity of PI3K gamma in the absence of the compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
  • PI3K gamma selective inhibitor generally refers to a compound that inhibits the activity of the PI3K gamma isoform more effectively than other isoforms of the PI3K family (e.g., PI3K alpha, beta, or delta).
  • potencies of compounds as inhibitors of an enzyme activity can be established by determining the concentrations at which each compound inhibits the activity to a predefined extent and then comparing the results.
  • concentrations at which each compound inhibits the activity can be determined by the concentration that inhibits 50% or 90% of the activity in a biochemical assay, which can be accomplished using conventional techniques known in the art, including the techniques describes in the Examples below.
  • PI3K gamma is expressed primarily in hematopoietic cells including leukocytes such as T-cells, dendritic cells, neutrophils, mast cells, B-cells, and macrophages. Due to its integral role in immune system function, PI3K gamma is also involved in a number of diseases related to undesirable immune response such as allergic reactions, inflammatory diseases, inflammation mediated angiogenesis, rheumatoid arthritis, auto-immune diseases such as lupus, asthma, emphysema and other respiratory diseases. By inhibiting aberrant proliferation of hematopoietic cells, PI3K gamma inhibitors can ameliorate the symptoms and secondary conditions that result from a primary effect such as excessive system or localized levels of leukocytes or lymphocytes.
  • the invention thus provides a method of treating a disorder mediated by inappropriate PI3-kinase activity comprising administering a safe and effective dose of a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof
  • PI3K mediated diseases or disorders are selected from the group consisting of respiratory diseases (including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF)); allergic diseases (including allergic rhinitis and atopic dermatitis); autoimmune diseases (including SLE, rheumatoid arthritis and multiple sclerosis); inflammatory disorders (including inflammatory bowel disease); hematologic malignancies; solid tumors; neurodegenerative diseases; pancreatitis; kidney diseases; transplantation rejection; graft rejection; lung injuries
  • respiratory diseases including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF)
  • allergic diseases including allergic rhinitis and atopic dermatitis
  • autoimmune diseases including SLE, rheumatoid arthritis and multiple sclerosis
  • inflammatory disorders including inflammatory bowel disease); hematologic malignancies; solid tumors; neurodegenerative diseases; pancreatiti
  • the compounds described herein may be used to treat cancers that are mediated by inappropriate PI3K gamma activity.
  • the disease is a hematologic malignancy.
  • the disease is lymphoma, such as Burkitt lymphoma, diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL), follicular lymphoma, lymphoplasmacytic lymphoma, Waldenstrom macroglobulinemia, and marginal zone lymphoma.
  • the disorder is multiple myeloma, or leukemia, such as acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), myelodysplastic syndrome (MDS), myeloproliferative disease (MPD), chronic myeloid leukemia (CML).
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • MDS myelodysplastic syndrome
  • MPD myeloproliferative disease
  • CML chronic myeloid leukemia
  • the disease is a solid tumor.
  • the indication is to treat solid tumor with abnormal PI3K gamma expression, such as pancreatic ductal adenocarcinoma (PDAC) and hepatocellular carcinoma (HCC), gastrointestinal cancer, prostate cancer, ovarian cancer, medulloblastoma, and breast cancer.
  • PDAC pancreatic ductal adenocarcinoma
  • HCC hepatocellular carcinoma
  • the compounds alone or with combination of other anti-cancer therapies may be used to treat prostate cancer, bladder cancer, colorectal cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, head and neck cancer, melanoma, neuroendocrine cancers, brain tumors, bone cancer, or soft tissue sarcoma.
  • PI3K mediated diseases or disorders are severe autoimmune disease as asthma, type I diabetes, rheumatoid arthritis, multiple sclerosis, chronic obstructive pulmonary disease (COPD), and lupus.
  • COPD chronic obstructive pulmonary disease
  • a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate may be used in combination with one or more additional therapeutic agents to treat cancers or inflammatory disorders.
  • the one or more additional therapeutic agents may be a chemotherapeutic agent, a radiotherapy, a targeted therapy, an immunotherapeutic agent or any current best of care treatment, either as a small molecule or a biologic nature.
  • Targeted therapies include but not limit to an inhibitor to cyclin-dependent kinase (CDK) such as CDK1, CDK2, CDK4/6, CDK7, and CDK9, Janus kinase (JAK) such as JAK1, JAK2 and/or JAK3, spleen tyrosine kinase (SYK), Bruton's tyrosine kinase (BTK), mitogen-activated protein kinase (MEK) such as MEK 1 and MEK2, bromodomain containing protein inhibitor (BRD) such as BRD4, isocitrate dehydrogenase (IDH) such as IDH1, histone deacetylase (HDAC), or any combination thereof.
  • CDK cyclin-dependent kinase
  • JAK1 JAK2 and/or JAK3 Janus kinase
  • SYK spleen tyrosine kinase
  • BTK Bruton's
  • Chemotherapeutic agents may be categorized by their mechanism of action into: alkylating agents, antimetabolites, anti-microtubule agents, topoisomerase inhibitors and cytotoxic agents.
  • a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate may be used in combination with chemotherapeutics to sensitize and improve the efficacy of certain chemotherapeutic agents to treat blood or solid tumors.
  • the immunotherapeutic agents include and are not limited to therapeutic antibodies, small molecules and vaccines suitable for treating patients; such as IDO1 and TDO2 inhibitors, A2A receptor inhibitors, arginase inhibitors, toll-like receptor agonists, chemokine regulators (including CCR and CXCR families), check point blockage antibodies such as antibodies that regulate PD-1, PD-L1, CTLA-4, OX40-OX40 ligand, LAG3, TIM3, or any combination thereof.
  • IDO1 and TDO2 inhibitors such as IDO1 and TDO2 inhibitors, A2A receptor inhibitors, arginase inhibitors, toll-like receptor agonists, chemokine regulators (including CCR and CXCR families), check point blockage antibodies such as antibodies that regulate PD-1, PD-L1, CTLA-4, OX40-OX40 ligand, LAG3, TIM3, or any combination thereof.
  • Radiotherapy is part of cancer treatment to control or kill malignant cells and commonly applied to the cancerous tumor because of its ability to control cell growth.
  • a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate may be used in combination with radiotherapy, to improve the efficacy of radiotherapy to treat blood or solid tumors, or with surgery, chemotherapy, immunotherapy and combination of the four.
  • a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate may be used in combination with one or more additional therapeutic agents to treat patients who are substantially refractory to at least one chemotherapy treatment, or in relapse after treatment with chemotherapy.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate salt thereof and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition can be formulated for particular routes of administration such as oral administration, parenteral administration, and topical administration, etc.
  • compositions intended for oral use are prepared according to any method known in the art for the manufacture of pharmaceutical compositions and can be prepared in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups, and capsules.
  • Oral composition may contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • the tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • Formulations for oral use can be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example, peanut oil, liquid paraffin or olive oil.
  • compositions are aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
  • compositions for transdermal application include an effective amount of a compound of the invention with a suitable carrier.
  • Carriers suitable for transdermal delivery include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • compositions for topical application include aqueous solutions, suspensions, ointments, creams, gels or spray formulations, e. g., for delivery by aerosol or the like.
  • Such topical delivery systems will in particular be appropriate for dermal application, e.g., for the treatment of skin cancer, e.g., for prophylactic use in sun creams, lotions, sprays and the like.
  • a topical application may also pertain to an inhalation or to an intranasal application. They may be conveniently delivered in the form of a dry powder (either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids) from a dry powder inhaler or an aerosol spray presentation from a pressurized container, pump, spray, atomizer or nebulizer, with or without the use of a suitable propellant.
  • a dry powder either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids
  • compositions and dosage forms that comprise one or more agents that reduce the rate by which the compound of the present invention as an active ingredient will decompose.
  • agents which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, etc.
  • compositions may be administered in either single or multiple doses.
  • a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate salt thereof can be formulated so as to provide the desired release schedule of the active ingredient based on the therapeutic treatment purpose.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups, and capsules.
  • these may contain an amount of active ingredient from about 0.1 to 1000 mg, preferably from about 0.1 to 500 mg.
  • a suitable daily dose for a human or other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods.
  • the daily dose can be administered in one to four doses per day.
  • the active compounds of this invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration drops suitable for administration to the eye, ear, or nose.
  • a suitable topical dose of active ingredient of a compound of the invention is 0.1 mg to 150 mg administered one to four, preferably one or two times daily.
  • the active ingredient may comprise from 0.001% to 10% w/w, e.g. from 1% to 2% by weight of the formulation, preferably not more than 5% w/w, and more preferably from 0.1% to 1% of the formulation.
  • the method comprises administering to the subject an initial daily dose of about 0.1 to 500 mg of a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d) and increasing the dose by increments until clinical efficacy is achieved. Increments of about 5, 10, 25, 50, or 100 mg can be used to increase the dose. The dosage can be increased daily, every other day, twice per week, or once per week.
  • the compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d) may be prepared using the methods disclosed herein and routine modifications thereof, which will be apparent given the disclosure herein and methods are well known in the art. Conventional and well-known synthetic methods may be used in addition to the teachings herein. The synthesis of representative compounds described herein may be accomplished as described in the following examples. If available, reagents may be purchased commercially, e.g., from Sigma Aldrich or other chemical suppliers.
  • B is selected from: Me, Et, isopropyl, cyclopropyl, monofluoromethyl and difluoromethyl.
  • each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof.
  • solvent refers to a solvent inert under the conditions of the reaction being described in conjunction therewith (including, for example, benzene, toluene, acetonitrile, tetrahydrofuran (“THF”), dimethylformamide (“DMF”), ethyl acetate (EA or EtOAc), dichloromethane (DCM), diethyl ether, methanol, pyridine, formic acid (FA) and the like.
  • the solvents used in the reactions of the present invention are inert organic solvents, and the reactions are carried out under an inert gas, preferably nitrogen and argon.
  • Step 1 A mixture of 4-bromo-2-methylbenzoic acid (1.0 g, 4.67 mmol), NIS (1.58 g, 7.01 mmol), Pd(OAc) 2 (209 mg, 0.93 mmol) in DMF (20 mL) was stirred at 100° C. for 3 h. The mixture was cooled to rt, diluted with EtOAc (150 mL), washed with H 2 O (80 mL*4), brine (100 mL).
  • Step 2 To a solution of 4-bromo-2-iodo-6-methylbenzoic acid (1 g, 2.94 mmol) in DMF (15 mL) was added K 2 CO 3 (1.22 g, 8.82 mmol) followed by CH 3 I (835 mg, 5.88 mmol). The mixture was stirred at 40° C. for 1 h. The mixture was cooled to rt, diluted with EtOAc (150 mL), washed with H 2 O (80 mL*4), brine (100 mL).
  • Step 3 To a solution of methyl 4-bromo-2-iodo-6-methylbenzoate (970 mg, 2.74 mmol) in CCl 4 (15 mL) was added NBS (1.17 g, 6.54 mmol), AIBN (180 mg, 1.1 mmol). The mixture was stirred at 80° C. for 17 h. The solvent was removed and the residue was purified by purified by silica gel column eluting with ethyl acetate in petroleum ether (0%-5%) to afford methyl 4-bromo-2-(bromomethyl)-6-iodobenzoate as a yellow oil. (920 mg, 82% yield).
  • Step 4 Preparation of (S)-5-bromo-2-(1-cyclopropylethyl)-7-iodoisoindolin-1-one (intermediate 1).
  • MeCN MeCN
  • (S)-1-cyclopropylethan-1-amine 270 mg, 3.18 mmol
  • boric acid 129 mg, 2.12 mmol
  • K 2 CO 3 878 mg, 6.36 mmol
  • Step 5 Synthesis of (S)-5-bromo-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-isoindolin-1-one (intermediate 2).
  • Step 6 (S)—N-(5-(2-(I-cyclopropylethyl)-7-(dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (General Procedure A)
  • Step 1 To a solution of 4-methylthiazol-2-amine (500 mg, 4.38 mmol) in pyridine (10 mL) was added cyclopropanecarbonyl chloride (547 mg, 5.26 mmol) at room temperature. Then the reaction mixture was stirred for 2 h at room temperature. The solvent was evaporated and the residue was diluted EA (30 mL), washed with aqueous HCl (3N, 10 mL) and brine (20 mL), dried over anhydrous Na 2 SO 4 , filtered and the filtrate was concentrated.
  • cyclopropanecarbonyl chloride 547 mg, 5.26 mmol
  • Step 1 To a solution of (S)-5-bromo-2-(1-cyclopropylethyl)-7-iodoisoindolin-1-one (250 mg, 0.62 mmol) in 1,4-dioxane (5 mL) was added diethylphosphine oxide (71 mg, 0.68 mmol), Pd 2 (dba) 3 (56 mg, 0.06 mmol), Xantphose (35 mg, 0.06 mmol), TEA (0.3 mL, 1.15 mmol) under N 2 . The mixture was stirred at rt for 16 h. Water (30 mL) and EA (30 mL) were added. The reaction mixture was extracted with EA (30 mL ⁇ 3).
  • Step 2 A mixture of (S)-5-bromo-2-(1-cyclopropylethyl)-7-(diethylphosphoryl)isoindolin-1-one (60 mg, 0.16 mmol), N-(4-methylthiazol-2-yl)acetamide (36 mg, 0.23 mmol), t-Bu 3 PBF 4 (10 mg, 0.03 mmol), cesium carbonate (102 mg, 0.31 mmol) and palladium (II) acetate (4 mg, 0.016 mmol) in dimethylformamide (2 mL) was stirred at 100° C. for 16 hs under N 2 .
  • Step 1 To a solution of N-(5-bromo-6-methylpyridin-2-yl)acetamide (300.0 mg, 1.31 mol) in dioxane (3 mL) was added 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (399.07 mg, 1.57 mol), potassium acetate (192.79 mg, 1.96 mol) and Pd(dppf)Cl 2 (10 mg). The mixture was stirred at 120° C. for 3 h under the N 2 . LCMS showed the reaction was completed. Water was added and extracted with EtOAc (3*10 mL). The organic layer was dried by NaSO 4 and filtration.
  • Step 2 To a solution of N-(6-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)acetamide (50.0 mg, 181.1 umol) in dioxane/H 2 O (2:0.5 mL), was added (S)-5-bromo-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)isoindolin-1-one (77.39 mg, 0.22 mmol), Na 2 CO 3 (38.38 mg, 0.36 mmol) and Pd(dppf)Cl 2 (5 mg). The mixture was stirred at 85° C. for 8 h under N 2 . LCMS showed the reaction was completed.
  • LC-MS (ESI) [M+H] + 307.9
  • Step 2 A mixture of methyl 5-bromo-3-(bromomethyl)picolinate (150 mg, 0.49 mmol), (S)-1-cyclopropylethan-1-amine hydrochloride (59 mg, 0.49 mmol), boric acid (30 mg, 0.49 mmol) and potassium carbonate (201 mg, 1.46 mmol) in acetonitrile (4 mL) was stirred at rt for 16 hr under the N 2 . Filtered, the filtrate was concentrated in vacuo.
  • Step 3 A mixture of (S)-3-bromo-6-(1-cyclopropylethyl)-5,6-dihydro-7H-pyrrolo[3,4-b]pyridin-7-one (70 mg, 0.25 mmol), N-(4-methylthiazol-2-yl)acetamide (58 mg, 0.37 mmol), t-Bu 3 PBF 4 (14 mg, 0.05 mmol), cesium carbonate (163 mg, 0.5 mmol) and palladium (H) acetate (6 mg, 0.025 mmol) in dimethylformamide (2 mL) was stirred at 100° C. for 16 hr under N 2 .
  • Step 2 A mixture of compound of methyl 6-chloro-2-methylnicotinate (361 mg, 2 mmol), NBS (391 mg, 2.2 mmol) and AIBN (32.8 mg, 0.2 mmol) was added in CCl 4 (5 mL) then stirred under reflux for 4 h. After the reaction was completed, to the mixture was added EA (30 mL), washed with water (20 mL ⁇ 2) and brine, dried over anhydrous Na 2 SO 4 and concentrated. The residue was purified by column chromatography on silica gel to afford methyl 2-(bromomethyl)-6-chloronicotinate as a white solid (300 mg, 52.3%).
  • Step 3 A mixture of compound of methyl 2-(bromomethyl)-6-chloronicotinate (260 mg, 1.1 mmol), (S)-1-cyclopropylethan-1-amine hydrochloride (180 mg, 1.5 mmol), boric acid (61 mg 1.0 mmol), K 2 CO 3 (560 mg, 4 mmol) was added in MeCN (10 mL) and stirred at rt overnight. After the reaction was completed, to the mixture was added EA (30 mL), washed with water (20 mL ⁇ 2) and brine, dried over anhydrous Na 2 SO 4 and concentrated.
  • Step 4 A mixture of compound of (S)-2-chloro-6-(1-cyclopropylethyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-5-one (74 mg, 0.315 mmol), N-(4-methylthiazol-2-yl)acetamide (60 mg, 0.378 mmol).
  • t-Bu 3 PBF 4 (18 mg, 0.063 mmol), cesium carbonate (204.7 mg, 0.63 mmol), and palladium (II) acetate (7 mg, 0.0315 mmol) was added in DMF (4 mL) stirred at 100° C. overnight.
  • Step 1 A mixture of 4-methylthiazol-2-amine (1.0 g, 8.77 mmol), 3-bromooxetane (2.4 g, 17.54 mmol), K 2 CO 3 (4 g, 21.93 mmol) in DMF (15 mL) was stirred at 120° C. for 48 h. The mixture was cooled to rt, diluted with EtOAc (150 mL), washed with H 2 O (80 mL*4), brine (100 mL).
  • Step 2 To a solution of (S)-5-bromo-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)isoindolin-1-one (60 mg, 0.17 mmol) in DMF (3 mL) was added 4-methyl-N-(oxetan-3-yl)thiazol-2-amine (57 mg, 0.34 mmol), t-Bu 3 PBF 4 (10 mg, 0.034 mmol), Cs 2 CO 3 (110 mg, 0.34 mmol) and Pd(OAc) 2 (3.8 mg, 0.017 mmol) under N 2 . The mixture was stirred at 100° C. for 3 h. Water (30 mL) and EA (30 mL) were added.
  • Step 1 To a solution of intermediate 4 (100 mg, 0.250 mmol) in DMF (2.00 mL) was added EtSNa (43.0 mg, 0.510 mmol) under N 2 . The mixture was stirred at 110° C. for 2 h. After being cooled to room temperature, the reaction was diluted with water (30 mL) and extracted with EA (20 mL ⁇ 3).
  • Step 2 To a solution of (S)—N-(5-(2-(1-cyclopropylethyl)-7-(ethylthio)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (80 mg, crude) in DCM (2 mL) was added m-CPBA (33 mg, 0.190 mmol) at ⁇ 40° C. under Ar. The mixture was stirred at ⁇ 40° C. for 1 h. aq.NaHCO 3 (5 mL) was added, and then the mixture was stirred for about 30 min, diluted with water (30 mL) and extracted with DCM (20 mL ⁇ 3).
  • Step 1 To a solution of intermediate 4 (100 mg, 0.256 mmol) in DMF (2 mL) was added NaH (20.5 mg, 0.512 mmol, 60% w/w dispersion in mineral oil) and propane-2-thiol (39.0 mg, 0.512 mmol) at rt. The mixture was stirred at 110° C. for 2 h under N 2 . After being cooled to rt, the reaction mixture was diluted with water (30 mL) and extracted with EA (20 mL ⁇ 3).
  • the mixture was purified by pre-HPLC to give a racemic mixture of N-(5-(2-((S)-1-cyclopropylethyl)-7-((R)—S-methylsulfinimidoyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide and N-(5-(2-((S)-1-cyclopropylethyl)-7-((S)—S-methylsulfinimidoyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (3.22 mg, 15.54%) as a white solid.
  • Step 1 To a solution of 4-bromo-2-ethylbenzoic acid (Prepared according to Buckley, D. et al, U.S. Pat. Appl. Publ., 20170119786) (2 g, 8.7 mmol) in DMF (20 mL) was added Pd(OAc) 2 (194 mg, 0.87 mmol), NIS (2.9 g, 13.1 mmol) under N 2 . The mixture was stirred at 100° C. for 3 h. Water (30 mL) and EtOAc (30 mL) were added. The reaction mixture was extracted with EtOAc (30 mL ⁇ 2). Then it was washed with water (20 mL ⁇ 3), dried over anhydrous Na 2 SO 4 and concentrated. The crude was purified by silica gel column (PE:EA, 1:0-5:1) to give 4-bromo-2-ethyl-6-iodobenzoic acid (3.2 g, crude).
  • Step 2 To a solution of 4-bromo-2-ethyl-6-iodobenzoic acid (3 g, 8.47 mmol) in DMF (50 mL) was added CH 3 I (2.41 g, 16.96 mmol), K 2 CO 3 (3.5 g, 25.4 mmol). The mixture was stirred at 40° C. for 2 h. Water (30 mL) and EtOAc (30 mL) were added. The reaction mixture was extracted with EtOAc (30 mL ⁇ 2). Then it was washed with water (30 mL ⁇ 5), dried over anhydrous Na 2 SO 4 and concentrated.
  • Step 4 To a solution of methyl 4-bromo-2-(1-bromoethyl)-6-iodobenzoate (1 g, 2.25 mmol) in MeCN (30 mL) was added (S)-1-cyclopropylethan-1-amine (286 mg, 3.37 mmol), boric acid (137 mg, 2.25 mmol), K 2 CO 3 (1.3 g, 8.99 mmol). The mixture was stirred at 85° C. for 17 h. Water (30 mL) and EtOAc (50 mL) were added. The reaction mixture was extracted with EtOAc (50 mL ⁇ 2). Then it was washed with brine, dried over anhydrous Na 2 SO 4 and concentrated.
  • Step 5 To a solution of 5-bromo-2-((S)-1-cyclopropylethyl)-7-iodo-3-methylisoindolin-1-one (650 mg, 1.56 mmol) in 1,4-dioxane (25 mL) was added dimethylphosphine oxide (133 mg, 1.71 mmol), Pd 2 (dba) 3 (142 mg, 0.16 mmol), xantphose (90 mg, 0.16 mmol), TEA (0.64 mL, 4.67 mmol) under N 2 . The mixture was stirred at rt for 17 h. Water (50 mL) and EA (50 mL) were added.
  • Step 6 To a solution of 5-bromo-2-((S)-1-cyclopropylethyl)-7-(dimethylphosphoryl)-3-methylisoindolin-1-one (260 mg, 0.71 mmol) in DMF was added N-(4-methylthiazol-2-yl)acetamide (132 mg, 0.85 mmol), t-Bu 3 PBF 4 (41 mg, 0.14 mmol), Cs 2 CO 3 (460 mg, 1.41 mmol) and Pd(OAc) 2 (16 mg, 0.07 mmol) under N 2 . The mixture was stirred at 100° C. for 2 h. Water (30 mL) and EA (30 mL) were added. The reaction mixture was extracted with EA (30 mL ⁇ 3).
  • Step 3 To a solution of S-(6-bromo-2-(tert-butyl)-3-oxoisoindolin-4-yl) ethanethioate (60 mg, 0.175 mmol) in MeOH (4.00 mL) was added Cs 2 CO 3 (85 mg, 0.262 mmol) at room temperature. The mixture was stirred at room temperature under argon atmosphere for 1 hour. MeI (124 mg, 0.877 mmol) was added and the mixture was stirred at room temperature under argon atmosphere for another 16 hours.
  • Step 1 A mixture of 5-bromo-2-((S)-1-cyclopropylethyl)-7-(methylsulfinyl)isoindolin-1-one (260 mg, 0.760 mmol), 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (386 mg, 1.519 mmol), Pd(dppf)Cl 2 (56 mg, 0.076 mmol) and KOAc (224 mg, 2.280 mmol) in dioxane (2.00 mL) was stirred at 80° C. under argon atmosphere for 1 hours.
  • Step 2 A mixture of 2-((S)-1-cyclopropylethyl)-7-(methylsulfinyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoindolin-1-one (50 mg, 0.128 mmol), intermediate 5 (45 mg, 0.141 mmol), Pd(dppf)Cl 2 (10 mg, 0.0130 mmol) and K 2 CO 3 (53 mg, 0.385 mmol) in dioxane/H 2 O (2.00 mL/0.4 mL) was stirred at 90° C. under argon atmosphere for 2 hours.
  • Step a To a solution of 2,6-dichloro-4-methylnicotinic acid (50.0 g, 244 mmol) in DMF (350 mL) was added K 2 CO 3 (101 g, 731 mmol) and CH 3 I (104 g, 731 mmol) at rt. The mixture was stirred at 40° C. for 2 h. After being cooled to rt, the mixture was diluted with water (530 mL) and extracted with EtOAc (250 mL ⁇ 3). The combined organic layers was washed with brine (250 mL ⁇ 4), dried over anhydrous Na 2 SO 4 and concentrated.
  • K 2 CO 3 101 g, 731 mmol
  • CH 3 I 104 g, 731 mmol
  • Step b To a solution of methyl 2,6-dichloro-4-methylnicotinate (50.0 g, 228 mmol) in CCl 4 (450 mL) was added NBS (163 g, 913 mmol), BPO (55.3 g, 159 mmol) at rt. The mixture was stirred at 90° C. and with concomitant irradiation from a tungsten lamp for 48 hours. After being cooled to rt, the mixture was concentrated under reduced pressure.
  • Step c To a solution of 2,6-dichloro-4-(dibromomethyl)nicotinate (85.2 g, 226 mmol) in 260 mL of CH 3 CN, DIPEA (58.5 g, 453 mmol) was added at 0° C. Then a solution of diethyl phosphite (31.3 g, 227 mmol) in 200 mL of CH 3 CN was added drop wise carefully at 0° C. The mixture was stirred at 0° C. for another hour. The mixture was diluted with cold aq. NaHCO 3 (200 mL) and extracted with EA (200 mL ⁇ 4).
  • Step d To a solution of methyl 4-(bromomethyl)-2,6-dichloronicotinate (60.2 g, 201 mmol) in MeCN (350 mL) was added (S)-1-cyclopropylethan-1-amine (29.4 g, 242 mmol), boric acid (14.9 g, 242 mmol) and K 2 C 0 M (83.4 g, 604 mmol) at rt. The mixture was stirred at 60° C. for 2 h. After being cooled to rt, the mixture was diluted with water (450 mL) and EA (350 mL). The reaction mixture was extracted with EA (250 mL ⁇ 3).
  • Step a To a solution of 4-methylthiazol-2-amine (40.0 g, 347 mmol) in THF/H 2 O (800 mL/800 mL) was added NaHCO 3 (175 g, 2.09 mol) and Boc 2 O (454 g, 2.09 mol) at rt. The mixture was stirred at 50° C. for 16 hours. The reaction mixture was cooled to rt. The mixture was concentrated in vacuum to remove the THF. The mixture was diluted with n-hexane, the solid was filtered and dried to give tert-butyl (4-methylthiazol-2-yl)carbamate (42.0 g, yield 56.6%) as a white solid.
  • Step b To a solution of tert-butyl (4-methylthiazol-2-yl)carbamate (42.0 g, 196 mmol) in THF (300 mL) was added LDA (304 mL, 608 mmol, 2.0 M) dropwise at ⁇ 78° C. under argon atmosphere. The mixture was stirred at ⁇ 78° C. for 1 hour under Ar. To the mixture was added a solution of n-Bu 3 SnCl (63.8 g, 196 mmol) in THF (120 mL). And then the mixture was stirred at ⁇ 78° C. for 12 h. The reaction mixture was quenched with ice water (200 mL) and extracted with EA (200 mL ⁇ 3).
  • LDA 304 mL, 608 mmol, 2.0 M
  • Step a To a solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (intermediate 6) (500 mg, 1.80 mmol) in dioxane (10 mL) was added tert-butyl (4-methyl-5-(tributylstannyl)thiazol-2-yl)carbamate (intermediate 7) (1.81 g, 3.60 mmol), Pd(PPh 3 ) 4 (415 mg, 0.360 mmol) at rt. The mixture was stirred at 160° C. for 1 h under argon atmosphere in microwave reactor.
  • Step b To a solution of tert-butyl (S)-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)carbamate (566 mg, 1.26 mmol) in DCM (2 mL) was added TFA (1.5 mL) at rt. The mixture was stirred at rt for 1 h. The mixture was quenched with aq. NaHCO 3 (20 mL) and extracted with DCM/MeOH (20 mL/1 mL ⁇ 3).
  • Step c To a solution of (S)-6-(2-amino-4-methylthiazol-5-yl)-4-chloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (300 mg, 0.86 mmol) in DCM (10 mL) was added TEA (261 mg, 2.58 mmol) and acetyl chloride (134 mg, 1.72 mmol) at 0° C. The mixture was stirred at rt for 1 h under Ar. The reaction mixture was diluted with ice water (50 mL) and extracted with EA (50 mL ⁇ 3).
  • Step a To a solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (intermediate 6) (1.00 g, 3.69 mmol) in DMF (10.0 mL) was added sodium methanethiolate (271 mg, 3.87 mmol) at room temperature. The mixture was stirred at room temperature for 16 h. The mixture was diluted with water (100 mL) and extracted with ethyl acetate (50.0 mL ⁇ 3). The combined organic layer was washed with brine (50 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • Step b (S)-6-(2-amino-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-4-(methylthio)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one was prepared according to general procedure C (200 mg, 40.1%) as a brown solid. Boc group was removed under this reaction condition. LC-MS (ESI) [M+H] + 361.1
  • Step c To a solution of (S)-6-(2-amino-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-4-(methylthio)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (200 mg, 76.3% purity, 0.424 mmol) in dichloromethane (10.0 mL) was added triethylamine (128 mg, 1.27 mmol) and acetyl chloride (66 mg, 0.848 mmol) at 0° C. under Ar. The mixture was stirred at room temperature for 1 hour. The mixture was diluted with water (30.0 mL) and extracted with ethyl acetate (20.0 mL ⁇ 3).
  • Step d To a solution of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(methylthio)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (60 mg, 0.149 mmol) in dichloromethane (5.00 mL) was added 3-chlorobenzoperoxoic acid (76 mg, 0.440 mmol) at 0° C. The mixture was stirred at 0° C. for 4 hours. The mixture was diluted with sodium sulfite solution (20.0 mL) and extracted with ethyl acetate (20.0 mL ⁇ 3).
  • Step a To a solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (intermediate 6) (300 mg, 1.11 mmol) in MeOH (5 mL) was added MeONa (120 mg, 2.22 mmol) slowly at rt. The mixture was stirred at rt overnight. The mixture was quenched with aq. NH 4 Cl (10 mL) and extracted with ethyl acetate (10 mL ⁇ 3). The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • Step b Prepared 22 according to General procedure C (135° C., 1 h) (5.00 mg FA salt, 3.33% yield) as a white solid.
  • 1 H NMR (400 MHz, CDCl 3 ): ⁇ 8.19 (s, 0.29H, FA), 7.21 (s, 1H), 4.56-4.40 (m, 2H), 4.14 (s, 3H), 3.76-3.72 (m, 1H), 2.68 (s, 3H), 2.31 (s, 3H), 1.33 (d, J 6.8 Hz, 3H), 1.00-0.98 (m, 1H), 0.63-0.62 (m, 1H), 0.44-0.37 (m, 3H).
  • Step a A solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (intermediate 6) (2.00 g, 7.38 mmol), bis(2,4-dimethoxybenzyl)amine (7.00 g, 22.1 mmol) and Et 3 N (1.50 g, 14.8 mmol) in dioxane (20 mL) was heated at 160° C. for 5 h in a sealed tube. The mixture was quenched with aq. NH 4 Cl (60 mL) and extracted with ethyl acetate (60 mL ⁇ 3).
  • Step b Prepared (S)—N-(5-(2-(1-cyclopropylethyl)-4-(2,4-dimethoxybenzylamino)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl) according to General procedure C (135° C., 1 h) (1.20 g, 23.0% yield) as a yellow solid.
  • Step c To a suspension of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(2,4-dimethoxybenzylamino)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (1.00 g, 1.92 mmol) in DCM (6 mL) was added TFA (6 mL) at rt. The reaction mixture was stirred at rt overnight. The reaction mixture was concentrated. The residue was diluted with aq. NaHCO 3 (40 mL), extracted with ethyl acetate (30 mL ⁇ 3).
  • Step d To a suspension of (S)—N-(5-(4-amino-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (150 mg, 0.404 mmol) in DCM (3 mL) was added triphosgene (48 mg, 0.161 mmol) slowly at 0° C., followed by the addition of Et 3 N (82 mg, 0.808 mmol) at 0° C. The resulting mixture was stirred at rt for 6 h.
  • Step a To a solution of (S)—N-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (400 mg, 1.02 mmol) in DMF (5.00 mL) was added ethanol (5.00 mL), Cs 2 CO 3 (834 mg, 2.56 mmol), DPPP (42 mg, 0.102 mmol) and Pd(OAc) 2 (23 mg, 0.103 mmol) at room temperature. The mixture was purged with a balloon of CO three times. The mixture was stirred at 70° C.
  • Step b To a solution of (S)-ethyl 6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxylate (100 mg, 0.233 mmol) in methanol (6 mL) was added water (2 mL) and lithium hydroxide monohydrate (49 mg, 1.17 mmol) at room temperature. The mixture was stirred at room temperature for 2 hours. The mixture was diluted with water (20.0 mL) and extracted with ethyl acetate (20.0 mL).
  • Step c To a solution of (S)-6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxylic acid (30) (40 mg, 0.100 mmol) in dichloromethane (5.00 mL) was added oxalyl dichloride (127 mg, 1.00 mmol) at 0° C. The mixture was stirred at room temperature for 1 hour. The mixture was concentrated under reduced pressure. The residue was dissolved in dichloromethane (5.00 mL) and cooled to 0° C.
  • Step a Prepared (S)-6-chloro-2-(1-cyclopropylethyl)-4-morpholino-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one according to the preparation of 28 (160° C. for 1 hours) (200 mg, 56.0% yield) as a green solid.
  • Step b Prepared 34 according to general procedure C (135° C., 1 h) (14.94 mg, 7.26% yield) as a white solid.
  • Step a Prepared 6-chloro-4-morpholino-2-(1,1,1-trifluoropropan-2-yl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one according to the preparation of 28 (160° C. for 1 hours) (230 mg, 0.660 mmol, 43.1% yield) as a light yellow solid.
  • Step b Prepared racemic mixture of 36 and 37 according to general procedure C (135° C., 1 h) (60.6 mg, 19.6% yield, racemic) as a white solid.
  • a mixture of stereoisomers may be separated by any suitable method, including, but not limited to, chiral HPLC.
  • chiral HPLC when a mixture of stereoisomers is separated by HPLC, it is to be appreciated that the resultant individual stereoisomers or mixtures can be assigned sequential labels (e.g., a first enantiomer, and a second enantiomer), the order of which implies the order in which the isomers eluted from the HPLC column.
  • the absolute stereochemistry for a first enantiomer and a second enantiomer may be obtained by known methods.
  • Step a Prepared 38 according to the preparation of 34 (21.2 mg, 10.61%) as a yellow solid.
  • Step a A solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (intermediate 6) (300 mg, 1.11 mmol) and tert-butyl piperazine-1-carboxylate (2.06 g, 11.1 mmol) in dioxane (10.0 mL) was stirred at 160° C. for 1 h in sealed tube.
  • Step b According to the general procedure B (135° C., 1 h), tert-butyl (S)-4-(6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-4-yl)piperazine-1-carboxylate was synthesized (300 mg, 80.5% yield) as a yellow gum. LC-MS (ESI) [M+H] + 541.0.
  • Step c To a suspension of tert-butyl (S)-4-(6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-4-yl)piperazine-1-carboxylate (43) (300 mg, 0.556 mmol) in DCM (3 mL) was added TFA (1 mL) at rt. The reaction mixture was stirred at rt overnight and then concentrated under reduced pressure. The residue was diluted with aq. NaHCO 3 (10 mL) and extracted with ethyl acetate (10 mL ⁇ 3).
  • the title product (41) was synthesized from (S)-6-chloro-2-(1-cyclopropylethyl)-4-morpholino-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one according to the general procedure C (135° C., 1 h) (23.81 mg, 17.9% yield) as a white solid.
  • Step a To methanol-d4 (0.600 mL) was added sodium hydride (80 mg, 2.00 mmol, 60% w/w dispersion in mineral oil) at 0° C. The mixture was stirred at room temperature for 0.5 hour, and then added to a solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (6) (271 mg, 1.00 mmol) in methanol-d 4 (1.40 mL) at 0° C. The resulted mixture was stirred at room temperature for 3 hours.
  • Step b According to general procedure C (135° C., 1 h), the title product (43) was synthesized (50.07 mg, 23.1% yield) as a white solid.
  • 1 H NMR (400 MHz, CDCl 3 ): ⁇ 7.21 (s, 1H), 4.55-4.40 (m, 2H), 3.79-3.71 (m, 1H), 2.69 (s, 3H), 2.30 (s, 3H), 1.33 (d, J 6.8 Hz, 3H), 1.02-0.94 (m, 1H), 0.65-0.60 (m, 1H), 0.46-0.32 (m, 3H).
  • Step a A solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (intermediate 6) (500 mg, 1.11 mmol) and NH 3 H 2 O (3 mL) in THF (10 mL) was heated in a sealed tube at 160° C. for 1 h. After being cooled to rt, the mixture was quenched with aq. NH 4 Cl (20 mL) and extracted with ethyl acetate (50 mL ⁇ 3). The combined organic layers was washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • Step b A solution of (S)-4-amino-6-chloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (130 mg, 0.516 mmol) in Ac 2 O (1.50 mL) was heated at 130° C. for 3 h. The mixture was cooled to rt and quenched with aq. NaHCO 3 (10 mL) and extracted with ethyl acetate (10 mL ⁇ 3). The combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered and concentrated under reduced pressure.
  • Step c Prepared 44 according to general procedure C (135° C., 1 h) (4.31 mg FA salt, 5.10% yield) as a white solid.
  • 1 H NMR (400 MHz, CDCl 3 ): ⁇ 9.82 (s, 1H), 8.16 (s, 0.24H, FA), 7.29 (s, 1H), 4.56-4.50 (m, 2H), 3.72-3.68 (m, 1H), 2.74 (s, 3H), 2.56 (s, 3H), 2.31 (s, 3H), 1.36 (d, J 6.8 Hz, 3H), 1.02-1.01 (m, 1H), 0.69-0.66 (m, 1H), 0.50-0.38 (m, 3H).
  • Step a A mixture of (S)—N-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (280 mg, 0.71 mmol), tributyl(1-ethoxyvinyl)stannane (311 mg, 0.86 mmol) and Pd(PPh 3 ) 2 Cl 2 (25 mg, 0.036 mmol) in dioxane (2 mL) was stirred at 100° C. for 8 h under Ar.
  • Step b A mixture of (S)—N-(5-(4-acetyl-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (50 mg, 0.12 mmol), morpholine (22 mg, 0.25 mmol) and titanium tetraisopropanolate (71 mg, 0.25 mmol) in DCE (1 mL) was stirred at 50° C. for 1 h. The reaction mixture was cooled to room temperature, and then NaBH(AcO) 3 (79 mg, 0.38 mmol) was added.
  • This example compares the biological activity for compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d).
  • Enzymatic activity of different PI3K isoforms was measured to compare the inhibitory potency and selectivity of the compounds provided herein against PI3K isoforms, particularly selectivity against PI3K gamma over delta. Solubility was also measured to access drug-ability of the tested compounds in the aspect of physicochemical properties.
  • Enzymatic activity of PI3K isoforms Enzymatic activity of the class I PI3K isoforms in the presence of the compounds of Table 1 was measured using ADP-Glo luminescent assay against PI3K ⁇ , PI3K ⁇ , PI3K ⁇ and PI3K ⁇ with ATP concentration at 25 ⁇ M. IPI-549 was used as the reference compound. The compounds would be tested from 1 or 10 ⁇ M, 3-fold dilution for 10 doses, in duplicate.
  • Compound preparation Dilute a compound to 100 ⁇ of the final desired highest inhibitor concentration in reaction by 100% DMSO. Transfer 100 ⁇ l of this compound dilution to a well in a 96-well plate and serially dilute it with DMSO for a total of 10 concentrations.
  • Kinase reaction Prepare a solution of PI3K in 1 ⁇ kinase buffer at 2-fold the final concentration of each reagent in the assay. Control wells contain 1 ⁇ kinase buffer without enzyme. Prepare substrate solution of PIP2 substrate and ATP in 1 ⁇ kinase reaction buffer at 2-fold of the final concentration. Add concentration substrate solution to each well to the final concentration. Cover the assay plate, shake to mix, and incubate at room temperature for 1 hour. Add 5 ⁇ l of ADP-Glo reagent to each well to stop the reaction. Mix briefly with a quick spin, shake slowly on the shaker for 120 min. Add 10 ul Kinase Detection Reagent to each well, shake 1 min, equilibrate for 30 min before reading on a luminescence plate reader.
  • Kinetic solubility test The stock solutions of test compounds and control compound progesterone were prepared in DMSO at the concentrations of 10 mM. 15 ⁇ L of stock solution (10 mM) was placed in order into their proper 96-well rack. 485 ⁇ L of PBS pH7.4 was added into each vial of the cap-less Solubility Sample plate. The assay was performed in singlet. Add one stir stick to each vial and seal using a molded PTFE/Silicone plug. Then the solubility sample plate was shaked at 25° C. at 1100 rpm for 2 hours. After completion of the 2 hours, the samples from the Solubility Sample plate were transferred into the filter plate. all the samples were filtered using the Vacuum Manifold.
  • Procedure for sample analysis The plate was placed into the well plate autosampler. The samples were evaluated by LC-MS/MS analysis.
  • the claimed compounds demonstrate remarkable selectivity of PI3K gamma over delta, beta and alpha isoforms.
  • compounds have dramatically improved physicochemical properties such as solubility and pharmacokinetic properties.
  • physicochemical properties such as solubility and pharmacokinetic properties.
  • compounds showed >1000 fold selectivity of PI3K gamma over delta; in another example, compound 20 showed >200 fold improvement over a known literature compound AZ-17 in kinetic solubility test at physiologic pH.
  • This study aims to determine the inhibitory effect of compounds on PI3K gamma and PI3K delta in corresponding cell based assays.
  • PI3K gamma activity was reflected by phosphorylation of AKT in C5a stimulated RAW264.7 cells
  • PI3K delta activity was reflected by phosphorylation of AKT in anti-IgM stimulated Raji cells.
  • Phosphorylation of AKT in cells was determined using AlphaLISA technology from PerkinElmer.
  • Raji cell assay Prepare Passage 11 Raji cell and add 6 ⁇ L of 60K cells per well in 384-well plate. Centrifuge at 500 RPM for 30 s and incubate for 2 hours at 37° C., 5% CO 2 . Add 30 nL compound by Echo and incubate for 30 min at 37° C. Add 2 ⁇ L of IgM (4 ⁇ , 12 ⁇ g/mL) per well, centrifuge at 500 RPM for 30 s and incubate for 10 min at 37° C. Add 2 ⁇ L of 5 ⁇ lysis buffer by Multidrop. Shake 10 min on a plate shaker. Add 5 ⁇ L acceptor mix provided in the kit. Centrifuge at 1000 RPM for 1 min. Add 5 ⁇ L donor mix provided in the kit. Centrifuge at 1000 RPM for 1 min. Then incubate for 2 hours at 25° C., keep the plate in dark. Read AlphaLISA signal on Envision.
  • Raw264.7 cell assay Prepare Passage 15 Raw264.7 cell and add 6 ⁇ L of 30K cells per well in 384-well plate by multidrop. Centrifuge at 500 RPM for 30 s and incubate for 2 hours at 37° C., 5% CO2. Add 30 nL compound by echo and incubate for 30 min at 37° C., 5% CO2. Add 2 ⁇ L of C5 ⁇ (4 ⁇ , 320 ng/mL) per well by multidrop, centrifuge at 500 RPM for 30 s and incubate for 5 min at 37° C., 5% CO2. Add 2 ⁇ L of 5 ⁇ lysis buffer by multidrop, incubate for 10 min on a plate shaker.
  • Dosing solutions were prepared at 1 mg/mL in 5% DMSO/40% PEG400/55% water.
  • a dosing solution was administered to male beagle dogs (Approximately 9-14 kg, 3 dogs each group) via intravenous (IV) bolus at 1 mg/kg and by oral gavage (P0) at 5 mg/kg.
  • the dosing volume was 1 mL/kg for IV administration and 5 mL % kg for oral gavage.
  • Plasma samples ( ⁇ 0.3 mL each time point) were collected into tubes containing potassium ethylenediaminetetraacetic acid (K 2 EDTA) as the anticoagulant at 0.033, 0.083, 0.25, 0.5, 1, 2, 4, 8 and 24 hours post dose for IV administration and 0.083, 0.25, 0.5, 1, 2, 4, 8 and 24 hours post dose for PO administration.
  • K 2 EDTA potassium ethylenediaminetetraacetic acid
  • the blood samples were then centrifuged for 5 minutes in a centrifuge refrigerated at 4° C.
  • the resultant plasma samples were analyzed using LC/MS/MS to determine concentrations of a test compound.
  • Non-compartmental model with WinNonlin (PhoenixTM, version 6.1) software was used to calculate pharmacokinetic (PK) parameters.
  • the PK results are listed in Table 5.
  • the pharmacokinetic profiles of the compounds provided herein including oral bioavailability (F), half-life time (Tin) and volume of distribution at steady state (Vss) are much improved over AZ-17.
  • the oral bioavailability of compound 20 in dog was 107%, in comparison, the oral bioavailability of AZ-17 in dog was reported to be 1.6% (Pemberton, N, et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441).
  • a dosing solution of Compound 34 was prepared at 2 mg/mL in 5% DMSO/40% PEG400/55% water. The dosing solution was administered to male CD-1 mice via oral gavage (PO) at 20 mg/kg. Blood and brain tissue samples were collected at 0.5, 2 and 8 hours post-dose. The blood samples were then centrifuged for 5 minutes in a centrifuge refrigerated at 4° C. The brain samples after blood removal were homogenized in water. The plasma and brain homogenate samples were then analyzed using LC/MS/MS to determine concentrations of Compound 34. The results are listed in Table 6. Compound 34 showed good exposure in the brain tissues of mice, demonstrating good penetration of blood-brain.
  • a dosing solution of Compound 34 was prepared at 2 mg/mL in 5% DMSO/40% PEG400/55% water.
  • BALB/c mice were inoculated subcutaneously at the fourth mammary pad with 4T1 cells for tumor development.
  • the mice were assigned into 2 groups using stratified randomization with 10 mice in each group based upon their body weight and inoculation order.
  • the treatments of the mice via oral gavage with either the vehicle or Compound 34 at 100 mg/kg were started from the day of randomization.
  • the tumor sizes were measured three times per week during the treatment. Tumor volume was calculated by the formula: length width 2 /2.
  • treatment of Compound 34 significantly suppressed tumor growth.
  • EAE Experimental autoimmune encephalomyelitis
  • MOG myelin oligodendrocyte glycoprotein
  • mice in group 3 and 4 were received Compound 34 following the dosing regimen specified.
  • Mice in group 2 were received positive compound FTY-720 as the reference.
  • Mice in group 1 were the vehicle control. The dosing started from disease onset day 10 and continued for 14 days.
  • mice can no longer use hind limbs to maintain rump posture or walk but can still move one or both limbs to some extent.
  • Complete hind limb paralysis total loss of movement in hind limbs; mouse drags itself only on its forelimbs.
  • Data analysis Clinical score at baseline, day 8 and during treatment was analyzed with GraphPad Prism software.

Abstract

The present disclosure provides selective phosphoinositide 3-kinase gamma inhibitors of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or pharmaceutically acceptable salts thereof. These compounds are useful for the treatment of conditions mediated by one or more PI3K isoforms, such as PI3K gamma (PI3Kγ). The present disclosure further provides methods of inhibiting phosphoinositide 3-kinase gamma using these compounds for treatment of disorders related to phosphatidylinositol 3-kinase gamma activity.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims prior benefit of U.S. Provisional Patent Application No. 62/832,133, filed Apr. 10, 2019, and of U.S. Provisional Patent Application No. 62/836,659, filed Apr. 20, 2019, the disclosures of each of which are hereby incorporated herein by reference in their entirety.
  • FIELD OF THE INVENTION
  • The present disclosure relates generally to novel inhibitors of phosphatidylinositol 3-kinase (PI3K). More specifically, the invention further relates to the preparation of the disclosed PI3K gamma inhibitor analogs and their use in pharmaceutical compositions for the treatment of various diseases, conditions and disorders related to PI3K gamma activity.
  • BACKGROUND OF THE INVENTION
  • The class I phosphoinositide 3-kinases (PI3Ks) regulate phosphatidylinositol 4,5-bisphosphate (PIP2) phosphorylation. PI3K Converts PIP2 to the scaffolding binding element phosphatidylinositol (3,4,5)-trisphosphate (PIP3). PIP3 plays a key regulatory role in cell survival, signal transduction, control of membrane trafficking and other functions. (Di Paolo, G. et al. Nature 2006, 443, 651; Parker, P. J. et al. Biochem. Soc. Trans. 2004, 32, 893; Hawkins, P. T. et al. Biochem. Soc. Trans. 2006, 34, 647; Schaeffer, E. M. et al. Curr. Opin. Immnunol. 2000, 12, 282). Its dysregulation leads to various disease states such as cancer, inflammatory and auto-immune disorders.
  • The Class I PI3Ks consist of four kinases further delineated into 2 subclasses. Class 1A PI3Ks consist of three closely related kinases, PI3Kα, β, and δ existing as heterodimers composed of a catalytic subunit (p110α, β or δ) and one of several regulatory subunits. They generally respond to signaling through receptor tyrosine kinases (RTKs). PI3Kγ single class 1B isoform, responds mainly to G-protein coupled receptors (GPCRs), and is composed of a p110γ catalytic subunit and one of two distinct regulatory subunits. PI3Kα and PI3Kβ are ubiquitously expressed throughout a wide variety of tissue and organ types. The expression pattern of PI3Kδ is restricted, to spleen, thymus, and peripheral blood leukocytes (Knight, Z. et al. Cell 2006, 125, 733). PI3Kγ is found mainly in leukocytes, but also in skeletal muscle, liver, pancreas, and heart (Cantly, C. Science 2002, 1655).
  • Recently, Schmid showed that p110γ can be activated by receptor tyrosine kinases and TLR/IL1Rs in myeloid cells. PI3kγ serves as a single convergent point promoting tumor inflammation and progression. In an animal model, treatment of mice with selective PI3Kγ inhi-biter inhibited myeloid cell p110γ catalytic activity and adhesion to VCAM-1, due to effect on the tumor microenvironment instead of direct inhibition of tumor cells. (Schmid, M. C. et al. Cancer Cell 19, 715-727, 2011, PLoS ONE 8, e60226, 2013). Furthermore, inhibition of PI3Kγ enhanced pro-inflammatory cytokines and decreased the expression of immune-suppressive factors in tumors and TAMs. Kaneda and De Henau concluded that PI3Kγ controls the TAM switch between immune suppression and immune stimulation. (Kaneda, M. M. et al. Nature 539, 437-442, 2016; De Henau, O. et al. Nature 539, 443-447, 2016).
  • Camps et al. described that treatment with selective PI3Kγ inhibitor AS-60485023 suppresses the progression of joint inflammation and damage in two distinct mouse models of rheumatoid arthritis (Camps M, et al., Nat. Med. 2005, 11, 936-943).
  • Based on the studies in cellular levels and efficacies observed in various disease models, PI3Kγ inhibitors could potentially be used to treat a variety of diseases such as inflammation, metabolic and cancer (Cushing, T. D., et al, J. Med. Chem. 2012, 55, 8559-8581; Ruckle, T. et al, Nat. Rev. Drug Discovery 2006, 5, 903-918; Stark, A. K. et al, Curr. Opin. Pharmacol. 2015, 23, 82-91).
  • PI3K gamma selective inhibitors have been disclosed in recent years. IPI-549 has been in the clinical trials as single agent and combination immuno-oncology therapies with check point agent PD-1 inhibitor Nivolumab (Evans, C. A. et al, ACS Med. Chem. Lett. 2016, 7, 862-867). Pemberton N et al reported that selective PI3K gamma inhibition resulted in a dose dependent inhibition of LPS-induced airway neutrophilia in rats (Pemberton, N. et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441). Modification of a series of azaisoindolinones by Come, J. H. et al provided CNS-penetrant and orally bioavailable PI3K gamma inhibitors that were efficacious in murine EAE model, which demonstrated PI3K gamma inhibition have the potential for the treatment of multiple sclerosis (Come, J. H. et al, Journal of Medicinal Chemistry 2018, 61, 5245-5256).
  • PI3K gamma inhibitors with related structure but different binding mode have been disclosed in recent years. For example, WO2015048318 disclosed (R)-6-(1-(2,2-difluoroethyl)-1H-pyrazol-4-yl)-4,7,7-trimethyl-2-(5-(2,2,2-trifluoro-1-hydroxyethyl)pyhdin3-yl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-5-one as a selective inhibitor of PI3K gamma. WO2011087776 disclosed Isoindolinone inhibitors of phosphatidylinositol 3-kinase. WO2017153527 disclosed novel inhibitors of phosphatidylinositol 3-kinase gamma. Discovery of highly isoform selective orally bioavailable phosphoinositide 3-kinase (PI3K) γ inhibitors, and design and synthesis of a novel series of 3-kinase γ (PI3Kγ) inhibitors were reported (Journal of Medicinal Chemistry 2018, 61, 5435-5441 and Journal of Medicinal Chemistry 2018, 61, 5245-5256).
  • However, these reported PI3K gamma inhibitors have less optimal potency, selectivity and pharmacokinetic properties. Therefore, there are needs in the art to develop novel PI3K gamma inhibitors that have improved potency, selectivity and pharmacokinetic properties.
  • SUMMARY
  • Compounds and pharmaceutically acceptable salts, stereoisomers, prodrugs, or solvates thereof useful for inhibiting PI3K isoforms, such as PI3K gamma, are described herein. Compositions, including pharmaceutical compositions that include the compounds are also provided, as are methods of using and making the compounds. The compounds provided herein may find use in treating diseases, disorders, or conditions that are mediated by PI3K isoforms, such as PI3K gamma.
  • In one aspect, provided herein is a compound of Formula (I):
  • Figure US20230227446A9-20230720-C00001
  • or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein:
    A is CH2, CH(C1-6alkyl), O, or S:
  • Y is CH or N; Z is CH or N; W is N, CH, or CX;
  • wherein X is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00002
  • NHG, CHG2, COOH, OG, SO2G, SO2NHG, NGSO2G, C1-6alkyl-NGSO2G, NHC(O)G, NHC(O)NG2, C(O)NHG, C(O)NG2, C3-10cycloalkyl, and 3-10 membered heterocyclyl, wherein each 3-10 membered heterocyclyl independently contains 1 or 2 heteroatoms, wherein the 1 or 2 heteroatoms are selected from the group consisting of O, N, and S, and wherein each C3-10cycloalkyl or 3-10 membered heterocyclyl is independently optionally substituted with one or more G, wherein:
  • Q1 is C1-6alkyl, wherein the C1-6alkyl is optionally substituted with one or more OH or halo, and
  • each G is independently selected from the group consisting of H, D, OH, C1-6alkoxy, oxo, NH2, SO2(C1-6alkyl), C(O)—C1-6alkyl, C3-10cycloalkyl, 3-10 membered heterocyclyl, and C1-6alkyl, wherein the C3-10cycloalkyl or C1-6alkyl is independently optionally substituted with one or more D, OH, C1-6alkoxy, CN, N(C1-6alkyl)2, SO2(C1-6alkyl), or halo, or
  • two G groups, together with the atoms to which they are attached, form a C3-10cycloalkyl or 3-10 membered heterocyclyl,
  • provided that, when Y and Z are each CH, and W is CX, then X is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00003
  • R1 is C1-6alkyl, C3-10cycloalkyl, or C1-6alkyl-C3-10cycloalkyl, wherein the C1-6alkyl, C3-10cycloalkyl, or C1-6alkyl-C3-10cycloalkyl is independently optionally substituted with one or more halo; and
    R2 is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00004
  • wherein:
  • L is H, halo, or C1-6alkyl, wherein the C1-6alkyl is optionally substituted with one or more halo, C1-6alkoxy, or OH, and
  • each Q2 and Q3 is independently C1-6alkyl, C3-10cycloalkyl, or 3-10 membered heterocyclyl, wherein the C1-6alkyl, C3-10cycloalkyl, or 3-10 membered heterocyclyl is independently optionally substituted with one or more halo.
  • In some embodiments, provided is a compound of Formula (I-a):
  • Figure US20230227446A9-20230720-C00005
  • or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • In some embodiments, provided is a compound of Formula (I-b):
  • Figure US20230227446A9-20230720-C00006
  • or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • In some embodiments, provided is a compound of Formula (I-c):
  • Figure US20230227446A9-20230720-C00007
  • or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • In some embodiments, provided is a compound of Formula (I-d):
  • Figure US20230227446A9-20230720-C00008
  • or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • In some embodiments, the compound described herein has improved selectivity against the PI3Kδ isoform, improved solubility, or improved oral bioavailability in comparison to a known PI3K gamma inhibitor (e.g., IPI-549 (Evans, C. et al, ACS Med. Chem. Lett. 2016, 7, 862-867) or AZ-17 (Pemberton, N, et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441)), or any combination of the foregoing.
  • In one aspect, provided herein is a method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) comprising contacting the PI3Kγ with a compound of Formula (I), (I-a), (I-b), (I-c), or (I-d), or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof, and one or more pharmaceutically acceptable carriers.
  • In some embodiments, provided herein is a method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) of Formula (I), (I-a), (I-b), (I-c), or (I-d), or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof, wherein the compound has improved selectivity against the PI3Kδ isoform, improved solubility, or improved oral bioavailability in comparison to a known PI3K gamma inhibitor (e.g., IPI-549 or AZ-17), or any combination of the foregoing.
  • In yet another aspect, provided herein is a method of treating a disorder of uncontrolled cellular proliferation related to related to one or more PI3K isoforms, such as PI3K gamma (PI3Kγ), comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), (I-a), (I-b), (I-c), or (I-d), or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • In some embodiments, provided herein is a method of treating an autoimmune diseases, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), (I-a), (I-b), (I-c), or (I-d), or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • In some embodiments, provided herein is a method of treating a cancer, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), (I-a), (I-b), (I-c), or (I-d), or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows the antitumor efficacy of an exemplary compound (compound 34) in 4T1 syngeneic mouse tumor model.
  • FIG. 2 shows that treatment with an exemplary compound (compound 34) at 20 mg/kg and 80 mg/kg BID significantly ameliorated the clinical disability symptom of EAE induced mice.
  • DETAILED DESCRIPTION
  • PI3K Gamma Inhibitor Compounds
  • Provided herein are compounds that function as PI3K gamma inhibitors. In some embodiments, provided herein is a compound of Formula (I):
  • Figure US20230227446A9-20230720-C00009
  • or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
    wherein:
    A is CH2, CH(C1-6alkyl), O, or S;
  • Y is CH or N: Z is CH or N;
  • W is N, CH, or CX; wherein X is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00010
  • NHG, CHG2, COOH, OG, SO2G, SO2NHG, NGSO2G, C1-6alkyl-NGSO2G, NHC(O)G, NHC(O)NG2, C(O)NHG, C(O)NG2, C3-10cycloalkyl, and 3-10 membered heterocyclyl, wherein each 3-10 membered heterocyclyl independently contains 1 or 2 heteroatoms, wherein the 1 or 2 heteroatoms are selected from the group consisting of O, N, and S, and wherein each C3-10cycloalkyl or 3-10 membered heterocyclyl is independently optionally substituted with one or more G, wherein:
  • Q1 is C1-6alkyl, wherein the C1-6alkyl is optionally substituted with one or more OH or halo, and
  • each G is independently selected from the group consisting of H, D, ON, C1-6alkoxy, oxo, NH2, SO2(C1-6alkyl), C(O)—C1-6alkyl, C3-10cycloalkyl, 3-10 membered heterocyclyl, and C1-6alkyl, wherein the C3-10cycloalkyl or C1-6alkyl is independently optionally substituted with one or more D, OH, C1-6alkoxy, CN, N(C1-6alkyl)2, SO2(C1-6alkyl), or halo, or
  • two G groups, together with the atoms to which they are attached, form a C3-10cycloalkyl or 3-10 membered heterocyclyl,
  • provided that, when Y and Z are each CH, and W is CX, then X is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00011
  • R1 is C1-6alkyl, C3-10cycloalkyl, or C1-6alkyl-C3-10cycloalkyl, wherein the C1-6alkyl, C3-10cycloalkyl, or C1-6alkyl-C3-10cycloalkyl is independently optionally substituted with one or more halo; and
    R2 is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00012
  • wherein:
  • L is H, halo, or C1-6alkyl, wherein the C1-6alkyl is optionally substituted with one or more halo, C1-6alkoxy, or OH, and
  • each Q2 and Q3 is independently C1-6alkyl, C3-10cycloalkyl, or 3-10 membered heterocyclyl, wherein the C1-6alkyl, C3-10cycloalkyl, or 3-10 membered heterocyclyl is independently optionally substituted with one or more halo.
  • In some embodiments, at least one of Y, Z and W is N. In some embodiments, Y is N and Z is CH. In some embodiments, Z is N and Y is CH.
  • In some embodiments, X is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00013
  • In some embodiments, X is selected from the group consisting of NHG, CHG2, COOH, OG, SO2G, SO2NHG, NGSO2G, C1-6alkyl-NGSO2G, NHC(O)G, NHC(O)NG2, C(O)NHG, C(O)NG2, C3-10cycloalkyl, and 3-10 membered heterocyclyl, wherein each 3-10 membered heterocyclyl independently contains 1 or 2 heteroatoms, wherein the 1 or 2 heteroatoms are selected from the group consisting of O, N, and S, and wherein each C3-10cycloalkyl or 3-10 membered heterocyclyl is independently optionally substituted with one or more G.
  • In some embodiments, Y and Z are each CH, W is CX, and X is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00014
  • In some embodiments, Y is N, Z is CH, W is CX, and X is selected from the group consisting of NHG, CHG2, COOH, OG, SO2G, SO2NHG, NGSO2G, C1-6alkyl-NGSO2G, NHC(O)G, NHC(O)NG2, C(O)NHG, C(O)NG2, C3-10cycloalkyl, and 3-10 membered heterocyclyl, wherein each 3-10 membered heterocyclyl independently contains 1 or 2 heteroatoms, wherein the 1 or 2 heteroatoms are selected from the group consisting of O, N, and S, and wherein each C3-10cycloalkyl or 3-10 membered heterocyclyl is independently optionally substituted with one or more G.
  • It is intended and understood that each and every variation of R1, R2, and A described herein may be combined with each and every variation of W, Y and Z as described, as if each and every combination is individually described.
  • In some embodiments, provided herein is a compound of Formula (I), wherein W is N, A is CH2, Y is CH, and Z is CH, such that the compound has the structure of Formula (I-a):
  • Figure US20230227446A9-20230720-C00015
  • or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • In some embodiments, provided herein is a compound of Formula (I), wherein W is CX, A is CH2, Y is N, and Z is CH, such that the compound of Formula (I) has the structure of Formula (I-b):
  • Figure US20230227446A9-20230720-C00016
  • or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
  • In some embodiments, R2 is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00017
  • In some embodiments, R2 is
  • Figure US20230227446A9-20230720-C00018
  • In certain embodiments, R2 is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00019
  • In some embodiments, Q2 is methyl, ethyl, isopropyl, cyclopropyl, or difluoromethyl.
  • In some embodiments, R2 is
  • Figure US20230227446A9-20230720-C00020
  • wherein L is methyl and Q2 is methyl, such that the compound of Formula (I) has the structure of Formula (I-c):
  • Figure US20230227446A9-20230720-C00021
  • or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
  • In some embodiments, R1 is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00022
  • In some embodiments, R1 is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00023
  • In some embodiments, R1 is
  • Figure US20230227446A9-20230720-C00024
  • In certain embodiments, R1 is
  • Figure US20230227446A9-20230720-C00025
  • and the compound of Formula (I) has the structure of Formula (I-d):
  • Figure US20230227446A9-20230720-C00026
  • or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
  • In some embodiments, X is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00027
  • In other embodiments, X is selected from the group consisting of
  • Figure US20230227446A9-20230720-C00028
  • In still other embodiments, X is selected from the group consisting of NHG, OG, NHSO2G, and C(O)NG2. In still other embodiments, X is selected from the group consisting of OMe, OCD3, NHSO2Me, NHSO2Et, C(O)NH2, C(O)NHMe, and C(O)NMe2.
  • In some embodiments, provided herein is a compound of Formula (I), (I-a), (I-b), (I-c), or (I-d), as the case may be, wherein: wherein the compound is selected from the group consisting of:
  • Figure US20230227446A9-20230720-C00029
    Figure US20230227446A9-20230720-C00030
    Figure US20230227446A9-20230720-C00031
    Figure US20230227446A9-20230720-C00032
    Figure US20230227446A9-20230720-C00033
    Figure US20230227446A9-20230720-C00034
    Figure US20230227446A9-20230720-C00035
    Figure US20230227446A9-20230720-C00036
    Figure US20230227446A9-20230720-C00037
    Figure US20230227446A9-20230720-C00038
  • In one embodiment, provided herein is a compound of Formula (I), wherein Z is N, A is CH2, Y is CH, and W is CH, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
  • In one embodiment, provided is a compound of the following structures (Table 1), or a pharmaceutically acceptable salt, prodrug, or solvate wherein:
  • TABLE 1
    Representative Compounds and their flat names.
    # Structure Flat Name
    1
    Figure US20230227446A9-20230720-C00039
    N-(5-(2-(1-cyclopropylethyl)-7- (dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4- methylthiazol-2-yl)acetamide
    2
    Figure US20230227446A9-20230720-C00040
    N-(5-(2-(1-cyclopropylethyl)-7- (dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4- methylthiazol-2-yl)propionamide
    3
    Figure US20230227446A9-20230720-C00041
    N-(5-(2-(1-cyclopropylethyl)-7- (dimethylphosphoryl)-oxoisoindolin-5-yl)-4- methylthiazol-2-yl)cyclopropanecarboxamide
    4
    Figure US20230227446A9-20230720-C00042
    N-(5-(2-(1-cyclopropylethyl)-7- (dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4- methylthiazol-2-yl)isobutyramide
    5
    Figure US20230227446A9-20230720-C00043
    N-(5-(2-(1-cyclopropylethyl)-7- (diethylphosphoryl)-1-oxoisoindolin-5-yl)-4- methylthiazol-2-yl)acetamide
    6
    Figure US20230227446A9-20230720-C00044
    N-(5-(2-(1-cyclopropylethyl)-7- (dimethylphosphoryl)-1-oxoisoindolin-5- yl)pyridin-2-yl)acetamide
    7
    Figure US20230227446A9-20230720-C00045
    N-(5-(2-(1-cyclopropylethyl)-7- (dimethylphosphoryl)-1-oxoisoindolin-5- yl)pyridin-2-yl)acetamide
    8
    Figure US20230227446A9-20230720-C00046
    N-(5-(6-(1-cyclopropylethyl)-7-oxo-6,7- dihydro-5H-pyrrolo[3,4-b]pyridin-3-yl)-4- methylthiazol-2-yl)acetamide
    9
    Figure US20230227446A9-20230720-C00047
    N-(5-(6-(1-cyclopropylethyl)-5-oxo-6,7- dihydro-5H-pyrrolo[3,4-b]pyridin-2-yl)-4- methylthiazol-2-yl)acetamide
    10
    Figure US20230227446A9-20230720-C00048
    N-(5-(2-(1-cyclopropylethyl)-1-oxoisoindolin- 5-yl)-4-methylthiazol-2-yl)acetamide
    11
    Figure US20230227446A9-20230720-C00049
    2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)- 5-(4-methyl-2-(oxetan-3-ylamino)thiazol-5- yl)isoindolin-1-one
    12
    Figure US20230227446A9-20230720-C00050
    N-(5-(2-(1-cyclopropylethyl)-7- (methylsulfinyl)-1-oxoisoindolin-5-yl)-4- methylthiazol-2-yl)acetamide
    Figure US20230227446A9-20230720-C00051
    13
    Figure US20230227446A9-20230720-C00052
    N-(5-(2-(1-cyclopropylethyl)-7-(ethylsulfinyl)- 1-oxoisoindolin-5-yl)-4-methylthiazol-2- yl)acetamide
    Figure US20230227446A9-20230720-C00053
    14
    Figure US20230227446A9-20230720-C00054
    N-(5-(2-(1-cyclopropylethyl)-7- (isopropylsulfinyl)-1-oxoisoindolin-5-yl)-4- methylthiazol-2-yl)acetamide
    Figure US20230227446A9-20230720-C00055
    15
    Figure US20230227446A9-20230720-C00056
    N-(5-(2-(1-cyclopropylethyl)-7-(S- methylsulfonimidoyl)-1-oxoisoindolin-5-yl)-4- methylthiazol-2-yl)acetamide
    Figure US20230227446A9-20230720-C00057
    16
    Figure US20230227446A9-20230720-C00058
    N-(5-(2-(1-cyclopropylethyl)-7- (dimethylphosphoryl)-3-methyl-1- oxoisoindolin-5-yl)-4-methylthiazol-2- yl)acetamide
    Figure US20230227446A9-20230720-C00059
    17
    Figure US20230227446A9-20230720-C00060
    N-(5-(2-(tert-butyl)-7-(methylsulfinyl)-1- oxoisoindolin-5-yl)4-methylthiazol-2- yl)acetamide
    Figure US20230227446A9-20230720-C00061
    18
    Figure US20230227446A9-20230720-C00062
    N-(5-(2-(1-cyclopropylethyl)-7- (methylsulfinyl)-1-oxoisoindolin-5-yl)-4- (difluoromethyl)thiazol-2-yl)acetamide
    Figure US20230227446A9-20230720-C00063
    19
    Figure US20230227446A9-20230720-C00064
    N-(5-(2-(1-cyclopropylethyl)-4- (methylsulfonyl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    20
    Figure US20230227446A9-20230720-C00065
    N-(5-(2-(1-cyclopropylethyl)-4- (methylsulfonamido)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    21
    Figure US20230227446A9-20230720-C00066
    N-(5-(2-(1-cyclopropylethyl)-4-(ethylsulfonyl)- 3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6- yl)-4-methylthiazol-2-yl)acetamide
    22
    Figure US20230227446A9-20230720-C00067
    N-(5-(2-(1-cyclopropylethyl)-4-methoxy-3-oxo- 2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    23
    Figure US20230227446A9-20230720-C00068
    N-(5-(2-(1-cyclopropylethyl)-4- (ethylsulfonamido)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    24
    Figure US20230227446A9-20230720-C00069
    N-(5-(4-(cyclopropanesulfonamido)-2-(1- cyclopropylethyl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    25
    Figure US20230227446A9-20230720-C00070
    N-(5-(2-(1-cyclopropylethyl)-3-oxo-4-(2- oxopyrrolidin-1-yl)-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    26
    Figure US20230227446A9-20230720-C00071
    N-(6-(2-acetamido-4-methylthiazol-5-yl)-2-(1- cyclopropylethyl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-4- yl)cyclopropanecarboxamide
    27
    Figure US20230227446A9-20230720-C00072
    N-(5-(2-(1-cyclopropylethyl)-4-methyl-3-oxo- 2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    28
    Figure US20230227446A9-20230720-C00073
    N-(5-(2-(1-cyclopropylethyl)-4-(3,3- dimethylureido)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    29
    Figure US20230227446A9-20230720-C00074
    6-(2-acetamido-4-methylthiazol-5-yl)-2-(1- cyclopropylethyl)-N,N-dimethyl-3-oxo-2,3- dihydro-1H-pyrrolo[3,4-c]pyridine-4- carboxamide
    30
    Figure US20230227446A9-20230720-C00075
    6-(2-acetamido-4-methylthiazol-5-yl)-2-(1- cyclopropylethyl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridine-4-carboxylic acid
    31
    Figure US20230227446A9-20230720-C00076
    6-(2-acetamido-4-methylthiazol-5-yl)-2-(1- cyclopropylethyl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridine-4-carboxamide
    32
    Figure US20230227446A9-20230720-C00077
    6-(2-acetamido-4-methylthiazol-5-yl)-2-(1- cyclopropylethyl)-N-methyl-3-oxo-2,3-dihydro- 1H-pyrrolo[3,4-c]pyridine-4-carboxamide
    33
    Figure US20230227446A9-20230720-C00078
    N-(5-(2-(1-cyclopropylethyl)-4-(morpholine-4- carbonyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c] pyridin-6-yl)-4-methylthiazol-2-yl)acetamide
    34
    Figure US20230227446A9-20230720-C00079
    N-(5-(2-(1-cyclopropylethyl)-4-morpholino-3- oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6- yl)-4-methylthiazol-2-yl)acetamide
    35
    Figure US20230227446A9-20230720-C00080
    N-(5-(2-(1-cyclopropylethyl)-4-morpholino-3- oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6- yl)-4-methylthiazol-2-yl)acetamide
    36
    Figure US20230227446A9-20230720-C00081
    N-(1-methyl-5-(4-morpholino-3-oxo-2-(1,1,1- trifluoropropan-2-yl)-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)thiazol-2- yl)acetamide
    37
    Figure US20230227446A9-20230720-C00082
    N-(4-methyl-5-(4-morpholino-3-oxo-2-(1,1,1- trifluoropropan-2-yl)-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)thiazol-2- yl)acetamide
    38
    Figure US20230227446A9-20230720-C00083
    N-(5-(2-(1-cyclopropylethyl)-4-(1,1- dioxidothiomorpholino)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    39
    Figure US20230227446A9-20230720-C00084
    N-(5-(2-(1-cyclopropylethyl)-3-oxo-4- (piperazin-1-yl)-2,3-dihydro-1H-pyrrolo[3,4-c] pyridin-6-yl)-4-methylthiazol-2-yl)acetamide
    40
    Figure US20230227446A9-20230720-C00085
    N-(5-(4-(4-acetylpiperazin-1-yl)-2-(1- cyclopropylethyl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetatnide
    41
    Figure US20230227446A9-20230720-C00086
    N-(5-(2-(1-cyclopropylethyl)-4-morpholino-3- oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6- yl)thiazol-2-yl)acetamide
    42
    Figure US20230227446A9-20230720-C00087
    N-(5-(2-(1-cyclopropylethyl)-4-(2- methoxyethoxy)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    43
    Figure US20230227446A9-20230720-C00088
    N-(5-(2-(1-cyclopropylethyl)-4-(methoxy-d3)- 3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6- yl)-4-methylthiazol-2-yl)acetamide
    44
    Figure US20230227446A9-20230720-C00089
    N-(5-(4-acetamido-2-(1-cyclopropylethyl)-3- oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6- yl)-4-methylthiazol-2-yl)acetatnide
    45
    Figure US20230227446A9-20230720-C00090
    N-(6-(2-acetamido-4-methylthiazol-5-yl)-2-(1- cyclopropylethyl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-4-yl)-2- cyanocyclopropane-1-carboxamide
    46
    Figure US20230227446A9-20230720-C00091
    N-(5-(2-(1-cyclopropylethyl)-4-(4- hydroxypiperidin-1-yl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    47
    Figure US20230227446A9-20230720-C00092
    N-(5-(2-(1-cyclopropylethyl)-4-(3- hydroxypyrrolidin-1-yl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetarnide
    48
    Figure US20230227446A9-20230720-C00093
    N-(5-(2-(1-cyclopropylethyl)-4-(3- hydroxyazetidin-1-yl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    49
    Figure US20230227446A9-20230720-C00094
    N-(5-(2-(1-cyclopropylethyl)-4-(3- (methylsulfonyl)azetidin-1-yl)-3-oxo-2,3- dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    50
    Figure US20230227446A9-20230720-C00095
    N-(5-(2-(1-cyclopropylethyl)-4-(2- hydroxyethoxy)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    51
    Figure US20230227446A9-20230720-C00096
    N-(5-(2-(1-cyclopropylethyl)-3-oxo-4- ((tetrahydrofuran-3-yl)oxy)-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    52
    Figure US20230227446A9-20230720-C00097
    N-(5-(2-(1-cyclopropylethyl)-4-(oxetan-3- yloxy)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c] pyridin-6-yl)-4-methylthiazol-2-yl)acetamide
    53
    Figure US20230227446A9-20230720-C00098
    N-(4-chloro-5-(2-(1-cyclopropylethyl)-4- morpholino-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c] pyridin-6-yl)thiazol-2-yl)acetamide
    54
    Figure US20230227446A9-20230720-C00099
    N-(5-(2-(1-cyclopropylethyl)-4- (morpholinomethyl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    55
    Figure US20230227446A9-20230720-C00100
    N-(5-(2-(1-cyclopropylethyl)-4-(1- morpholinoethyl)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    56
    Figure US20230227446A9-20230720-C00101
    N-(5-(2-(1-cyclopropylethyl)-4-(3- methylmorpholino)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    Figure US20230227446A9-20230720-C00102
    57
    Figure US20230227446A9-20230720-C00103
    N-(5-(2-(1-cyclopropylethyl)-4-(3- (hydroxymethyl)morpholino)-3-oxo-2,3- dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    58
    Figure US20230227446A9-20230720-C00104
    N-(5-(2-(1-cyclopropylethyl)-4-(3- (hydroxymethyl)morpholino)-3-oxo-2,3- dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    59
    Figure US20230227446A9-20230720-C00105
    N-(5-(2-(1-cyclopropylethyl)-4-(2- (hydroxymethyl)morpholino)-3-oxo-2,3- dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    60
    Figure US20230227446A9-20230720-C00106
    N-(5-(2-(1-cyclopropylethyl)-4-(2- (methoxymethyl)morpholino)-3-oxo-2,3- dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    61
    Figure US20230227446A9-20230720-C00107
    N-(5-(2-(1-cyclopropylethyl)-4-((2- (dimethylamino)ethyl)amino)-3-oxo-2,3- dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    62
    Figure US20230227446A9-20230720-C00108
    N-(5-(2-(1-cyclopropylethyl)-4-((2- methoxyethyl)amino)-3-oxo-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    63
    Figure US20230227446A9-20230720-C00109
    N-(5-(2-(1-cyclopropylethyl)-4-(2- (methylsulfonyl)ethyl)amino)-3-oxo-2,3- dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    64
    Figure US20230227446A9-20230720-C00110
    N-(5-(2-(1-cyclopropylethyl)-3-oxo-4- (tetrahydro-2H-pyran-4-yl)-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    65
    Figure US20230227446A9-20230720-C00111
    N-(5-(4-(6-oxa-3-azabicyclo[3.1.1]heptan-3- yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro- 1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazoi-2-yl)acetamide
    66
    Figure US20230227446A9-20230720-C00112
    N-(5-(4-(2-oxa-5-azabicyclo[2.2.1]heptan-5- yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro- 1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    67
    Figure US20230227446A9-20230720-C00113
    N-(5-(2-(1-cyclopropylethyl)-3-oxo-4-(2-oxa-6- azaspiro[3.3]heptan-6-yl)-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
    68
    Figure US20230227446A9-20230720-C00114
    N-(5-(2-(1-cyclopropylethyl)-4-((N- methylmethylsulfonamido)methyl)-3-oxo-2,3- dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4- methylthiazol-2-yl)acetamide
    69
    Figure US20230227446A9-20230720-C00115
    N-(5-(2-(1-cyclopropylethyl)-3-oxo-4-(2-oxa-7- azaspiro[3.5]nonan-7-yl)-2,3-dihydro-1H- pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2- yl)acetamide
  • Provided are also compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or pharmaceutically acceptable salts, prodrugs, or solvates thereof. In certain embodiments, provided herein are also crystalline and amorphous forms of the compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or pharmaceutically acceptable salts, stereoisomers, prodrugs, or solvents thereof.
  • “Alkyl” as used herein refers to and includes, unless otherwise stated, a saturated linear (i.e., unbranched) or branched univalent hydrocarbon chain or combination thereof, having the number of carbon atoms designated (i.e., C1-C10 means one to ten carbon atoms). Particular alkyl groups are those having 1 to 20 carbon atoms (a “C1-C20 alkyl”), having 1 to 10 carbon atoms (a “C1-C10 alkyl”), having 6 to 10 carbon atoms (a “C6-C10 alkyl”), having 1 to 6 carbon atoms (a “C1-C6 alkyl”), having 2 to 6 carbon atoms (a “C2-C6 alkyl”), or having 1 to 4 carbon atoms (a “C1-C4 alkyl”). Examples of alkyl groups include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, n-decyl, and the like.
  • “Cycloalkyl” as used herein refers to and includes, unless otherwise stated, saturated cyclic univalent hydrocarbon structures, having the number of carbon atoms designated (i.e., C3-C10 means three to ten carbon atoms). Cycloalkyl can consist of one ring, such as cyclohexyl, or multiple rings, such as adamantyl. A cycloalkyl comprising more than one ring may be fused, spiro or bridged, or combinations thereof. Particular cycloalkyl groups are those having from 3 to 12 annular carbon atoms. A preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a “C3-C8 cycloalkyl”), having 3 to 6 carbon atoms (a “C3-C6 cycloalkyl”), or having from 3 to 4 annular carbon atoms (a “C3-C4 cycloalkyl”). Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, and the like.
  • “Heteroaryl” as used herein refers to an unsaturated aromatic cyclic group having from 1 to 14 annular carbon atoms and at least one annular heteroatom, including but not limited to heteroatoms such as nitrogen, oxygen and sulfur. A heteroaryl group may have a single ring (e.g., pyridyl, furyl) or multiple condensed rings (e.g., indolizinyl, benzothienyl) which condensed rings may or may not be aromatic. Particular heteroaryl groups are 5 to 14-membered rings having 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 5 to 10-membered rings having 1 to 8 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, or 5, 6 or 7-membered rings having 1 to 5 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In one variation, particular heteroaryl groups are monocyclic aromatic 5-, 6- or 7-membered rings having from 1 to 6 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In another variation, particular heteroaryl groups are polycyclic aromatic rings having from 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. A heteroaryl group having more than one ring where at least one ring is non-aromatic may be connected to the parent structure at either an aromatic ring position or at a non-aromatic ring position. In one variation, a heteroaryl group having more than one ring where at least one ring is non-aromatic is connected to the parent structure at an aromatic ring position. A heteroaryl group may be connected to the parent structure at a ring carbon atom or a ring heteroatom.
  • “Heterocycle”, “heterocyclic”, or “heterocyclyl” as used herein refers to a saturated or an unsaturated non-aromatic cyclic group having a single ring or multiple condensed rings, and having from 1 to 14 annular carbon atoms and from 1 to 6 annular heteroatoms, such as nitrogen, sulfur or oxygen, and the like. A heterocycle comprising more than one ring may be fused, bridged or spiro, or any combination thereof, but excludes heteroaryl groups. The heterocyclyl group may be optionally substituted independently with one or more substituents described herein. Particular heterocyclyl groups are 3 to 14-membered rings having 1 to 13 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 12-membered rings having 1 to 11 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 10-membered rings having 1 to 9 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 8-membered rings having 1 to 7 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, or 3 to 6-membered rings having 1 to 5 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In one variation, heterocyclyl includes monocyclic 3-, 4-, 5-, 6- or 7-membered rings having from 1 to 2, 1 to 3, 1 to 4, 1 to 5, or 1 to 6 annular carbon atoms and 1 to 2, 1 to 3, or 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In another variation, heterocyclyl includes polycyclic non-aromatic rings having from 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur.
  • “Halo” or “halogen” refers to elements of the Group 17 series having atomic number 9 to 85. Preferred halo groups include the radicals of fluorine, chlorine, bromine and iodine. 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 halogen; 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 trifluoromethyl (—CF3).
  • “Oxo” refers to the moiety ═O.
  • “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, 1 to 3, 1 to 4, 1 to 5, 2 to 3, 2 to 4, or 2 to 5 substituents. In one embodiment, an optionally substituted group is unsubstituted.
  • “Pharmaceutically-acceptable salt” means a salt prepared by conventional means, and are well known by those skilled in the art. The “pharmacologically acceptable salts” include basic salts of inorganic and organic acids (Berge et al., J. Pharm. Sci. 1977, 66:1). “Pharmaceutically acceptable salts” are those salts which retain at least some of the biological activity of the free (non-salt) compound and which can be administered as drugs or pharmaceuticals to an individual. Such salts, for example, include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, oxalic acid, propionic acid, succinic acid, maleic acid, tartaric acid and the like; (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base. Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine and the like. Acceptable inorganic bases which can be used to prepared salts include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. Pharmaceutically acceptable salts can be prepared in situ in the manufacturing process, or by separately reacting a purified compound of the invention in its free acid or base form with a suitable organic or inorganic base or acid, respectively, and isolating the salt thus formed during subsequent purification. “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.
  • The term “excipient” as used herein means an inert or inactive substance that may be used in the production of a drug or pharmaceutical, such as a tablet containing a compound of the invention as an active ingredient. Various substances may be embraced by the term excipient, including without limitation any substance used as a binder, disintegrant, coating, compression/encapsulation aid, cream or lotion, lubricant, solutions for parenteral administration, materials for chewable tablets, sweetener or flavoring, suspending/gelling agent, or wet granulation agent. Binders include, e.g., carbomers, povidone, xanthan gum, etc.; coatings include, e.g., cellulose acetate phthalate, ethylcellulose, gellan gum, maltodextrin, enteric coatings, etc.; compression/encapsulation aids include, e.g., calcium carbonate, dextrose, fructose de (dc=“directly compressible”), honey dc, lactose (anhydrate or monohydrate; optionally in combination with aspartame, cellulose, or microcrystalline cellulose), starch dc, sucrose, etc.; disintegrants include, e.g., croscarmellose sodium, gellan gum, sodium starch glycolate, etc.; creams or lotions include, e.g., maltodextrin, carrageenans, etc.; lubricants include, e.g., magnesium stearate, stearic acid, sodium stearyl fumarate, etc.; materials for chewable tablets include, e.g., dextrose, fructose dc, lactose (monohydrate, optionally in combination with aspartame or cellulose), etc.; suspending/gelling agents include, e.g., carrageenan, sodium starch glycolate, xanthan gum, etc.; sweeteners include, e.g., aspartame, dextrose, fructose dc, sorbitol, sucrose dc, etc.; and wet granulation agents include, e.g., calcium carbonate, maltodextrin, microcrystalline cellulose, etc.
  • A “solvate” is formed by treating a compound in a solvent. Solvates of salts of the compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d) are also provided. In the case of treating compounds with water, the solvate is hydrates. Hydrates of the compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d) are also provided.
  • A “prodrug” includes any compound that converts into a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), when administered to a subject, e.g., upon metabolic processing of the prodrug.
  • Therapeutic Uses of the Compounds
  • The compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof may be used for treating PI3K mediated diseases or disorders. In one embodiment, provided are methods for inhibiting PI3K gamma activity using a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
  • In addition to the therapeutic uses described herein, selected compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, have improved physicochemical and pharmacokinetic properties, for an example, improved solubility in water and oral bioavailability.
  • 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 an individual. In some embodiments, “treatment” of a disorder does not include prevention of the disorder, and “prevention” is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a disorder or biological manifestation thereof, or to delay the onset of such disorder or biological manifestation thereof.
  • 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 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 recurrence. 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.
  • As used herein, the term “subject” is a mammal, including humans. A subject includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate. In some embodiments, the subject is human (including adults and children).
  • “Inhibition of PI3K gamma activity” or variants refer to a decrease in PI3K gamma activity as a direct or indirect response to the presence of a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, relative to the activity of PI3K gamma in the absence of the compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
  • The term “PI3K gamma selective inhibitor” generally refers to a compound that inhibits the activity of the PI3K gamma isoform more effectively than other isoforms of the PI3K family (e.g., PI3K alpha, beta, or delta).
  • The potencies of compounds as inhibitors of an enzyme activity (or other biological activity) can be established by determining the concentrations at which each compound inhibits the activity to a predefined extent and then comparing the results. “IC50” or “IC90” of an inhibitor can be determined by the concentration that inhibits 50% or 90% of the activity in a biochemical assay, which can be accomplished using conventional techniques known in the art, including the techniques describes in the Examples below.
  • PI3K gamma is expressed primarily in hematopoietic cells including leukocytes such as T-cells, dendritic cells, neutrophils, mast cells, B-cells, and macrophages. Due to its integral role in immune system function, PI3K gamma is also involved in a number of diseases related to undesirable immune response such as allergic reactions, inflammatory diseases, inflammation mediated angiogenesis, rheumatoid arthritis, auto-immune diseases such as lupus, asthma, emphysema and other respiratory diseases. By inhibiting aberrant proliferation of hematopoietic cells, PI3K gamma inhibitors can ameliorate the symptoms and secondary conditions that result from a primary effect such as excessive system or localized levels of leukocytes or lymphocytes.
  • In one aspect, the invention thus provides a method of treating a disorder mediated by inappropriate PI3-kinase activity comprising administering a safe and effective dose of a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof
  • In one embodiment, PI3K mediated diseases or disorders are selected from the group consisting of respiratory diseases (including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF)); allergic diseases (including allergic rhinitis and atopic dermatitis); autoimmune diseases (including SLE, rheumatoid arthritis and multiple sclerosis); inflammatory disorders (including inflammatory bowel disease); hematologic malignancies; solid tumors; neurodegenerative diseases; pancreatitis; kidney diseases; transplantation rejection; graft rejection; lung injuries
  • In one embodiment, the compounds described herein may be used to treat cancers that are mediated by inappropriate PI3K gamma activity. In certain embodiments, the disease is a hematologic malignancy. In certain embodiments, the disease is lymphoma, such as Burkitt lymphoma, diffuse large B-cell lymphoma (DLBCL) and mantle cell lymphoma (MCL), follicular lymphoma, lymphoplasmacytic lymphoma, Waldenstrom macroglobulinemia, and marginal zone lymphoma. In one embodiment, the disorder is multiple myeloma, or leukemia, such as acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), myelodysplastic syndrome (MDS), myeloproliferative disease (MPD), chronic myeloid leukemia (CML).
  • In other embodiments, the disease is a solid tumor. In particular embodiments, the indication is to treat solid tumor with abnormal PI3K gamma expression, such as pancreatic ductal adenocarcinoma (PDAC) and hepatocellular carcinoma (HCC), gastrointestinal cancer, prostate cancer, ovarian cancer, medulloblastoma, and breast cancer. In some embodiment, the compounds alone or with combination of other anti-cancer therapies may be used to treat prostate cancer, bladder cancer, colorectal cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, head and neck cancer, melanoma, neuroendocrine cancers, brain tumors, bone cancer, or soft tissue sarcoma.
  • In some embodiments, PI3K mediated diseases or disorders are severe autoimmune disease as asthma, type I diabetes, rheumatoid arthritis, multiple sclerosis, chronic obstructive pulmonary disease (COPD), and lupus.
  • Combination Therapies
  • In one embodiment, a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate may be used in combination with one or more additional therapeutic agents to treat cancers or inflammatory disorders. The one or more additional therapeutic agents may be a chemotherapeutic agent, a radiotherapy, a targeted therapy, an immunotherapeutic agent or any current best of care treatment, either as a small molecule or a biologic nature.
  • Targeted therapies include but not limit to an inhibitor to cyclin-dependent kinase (CDK) such as CDK1, CDK2, CDK4/6, CDK7, and CDK9, Janus kinase (JAK) such as JAK1, JAK2 and/or JAK3, spleen tyrosine kinase (SYK), Bruton's tyrosine kinase (BTK), mitogen-activated protein kinase (MEK) such as MEK 1 and MEK2, bromodomain containing protein inhibitor (BRD) such as BRD4, isocitrate dehydrogenase (IDH) such as IDH1, histone deacetylase (HDAC), or any combination thereof.
  • Chemotherapeutic agents may be categorized by their mechanism of action into: alkylating agents, antimetabolites, anti-microtubule agents, topoisomerase inhibitors and cytotoxic agents. A compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate may be used in combination with chemotherapeutics to sensitize and improve the efficacy of certain chemotherapeutic agents to treat blood or solid tumors.
  • The immunotherapeutic agents include and are not limited to therapeutic antibodies, small molecules and vaccines suitable for treating patients; such as IDO1 and TDO2 inhibitors, A2A receptor inhibitors, arginase inhibitors, toll-like receptor agonists, chemokine regulators (including CCR and CXCR families), check point blockage antibodies such as antibodies that regulate PD-1, PD-L1, CTLA-4, OX40-OX40 ligand, LAG3, TIM3, or any combination thereof.
  • Radiotherapy is part of cancer treatment to control or kill malignant cells and commonly applied to the cancerous tumor because of its ability to control cell growth. A compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate may be used in combination with radiotherapy, to improve the efficacy of radiotherapy to treat blood or solid tumors, or with surgery, chemotherapy, immunotherapy and combination of the four.
  • In certain embodiments, a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate may be used in combination with one or more additional therapeutic agents to treat patients who are substantially refractory to at least one chemotherapy treatment, or in relapse after treatment with chemotherapy.
  • Pharmaceutical Compositions
  • In another aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate salt thereof and a pharmaceutically acceptable carrier or excipient. The pharmaceutical composition can be formulated for particular routes of administration such as oral administration, parenteral administration, and topical administration, etc.
  • Compositions intended for oral use are prepared according to any method known in the art for the manufacture of pharmaceutical compositions and can be prepared in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups, and capsules. Oral composition may contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. The tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. Formulations for oral use can be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • Certain injectable compositions are aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
  • Suitable compositions for transdermal application include an effective amount of a compound of the invention with a suitable carrier. Carriers suitable for transdermal delivery include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • Suitable compositions for topical application, e.g., to the skin and eyes, include aqueous solutions, suspensions, ointments, creams, gels or spray formulations, e. g., for delivery by aerosol or the like. Such topical delivery systems will in particular be appropriate for dermal application, e.g., for the treatment of skin cancer, e.g., for prophylactic use in sun creams, lotions, sprays and the like.
  • As used herein a topical application may also pertain to an inhalation or to an intranasal application. They may be conveniently delivered in the form of a dry powder (either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids) from a dry powder inhaler or an aerosol spray presentation from a pressurized container, pump, spray, atomizer or nebulizer, with or without the use of a suitable propellant.
  • The invention further provides pharmaceutical compositions and dosage forms that comprise one or more agents that reduce the rate by which the compound of the present invention as an active ingredient will decompose. Such agents, which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, etc.
  • Modes of Administration and Dosing
  • The pharmaceutical compositions may be administered in either single or multiple doses. A compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d), or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate salt thereof can be formulated so as to provide the desired release schedule of the active ingredient based on the therapeutic treatment purpose.
  • The pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups, and capsules. For example, these may contain an amount of active ingredient from about 0.1 to 1000 mg, preferably from about 0.1 to 500 mg. A suitable daily dose for a human or other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods. The daily dose can be administered in one to four doses per day. For therapeutic purposes, the active compounds of this invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration drops suitable for administration to the eye, ear, or nose. A suitable topical dose of active ingredient of a compound of the invention is 0.1 mg to 150 mg administered one to four, preferably one or two times daily. For topical administration, the active ingredient may comprise from 0.001% to 10% w/w, e.g. from 1% to 2% by weight of the formulation, preferably not more than 5% w/w, and more preferably from 0.1% to 1% of the formulation.
  • In a particular embodiment, the method comprises administering to the subject an initial daily dose of about 0.1 to 500 mg of a compound of Formula (I) including (I-a), (I-b), (I-c), and (I-d) and increasing the dose by increments until clinical efficacy is achieved. Increments of about 5, 10, 25, 50, or 100 mg can be used to increase the dose. The dosage can be increased daily, every other day, twice per week, or once per week.
  • Synthesis of the Compounds
  • The compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d) may be prepared using the methods disclosed herein and routine modifications thereof, which will be apparent given the disclosure herein and methods are well known in the art. Conventional and well-known synthetic methods may be used in addition to the teachings herein. The synthesis of representative compounds described herein may be accomplished as described in the following examples. If available, reagents may be purchased commercially, e.g., from Sigma Aldrich or other chemical suppliers.
  • Enumerated Embodiments
      • 1. A selective phosphoinositide 3-kinase gamma (PI3K) inhibitor compound having a structure of formula (A), or a pharmaceutically acceptable salt, or solvate thereof. In one embodiment, provided is a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate wherein:
  • Figure US20230227446A9-20230720-C00116
        • A is selected from CH2, CH(Me), O, and S;
        • X is selected from N, CH, and CR;
        • When Y=Z=CH, X is selected from N, CH and CR,
        • R is selected from:
  • Figure US20230227446A9-20230720-C00117
        • G is selected from C1-4 alkyl, optionally substituted with OH and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl.
        • Y is CH or N
        • Z is CH or N
        • When either Z or Y is N and X═CH and CR, R is selected from the following groups:
        • NHSO2G, SO2G, SO2NHG, OG, H, Me and
  • Figure US20230227446A9-20230720-C00118
        • G is selected from C1-4 alkyl, optionally substituted with OH and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl.
        • R1 is branched alkane, including substituted cyclopropanes, cyclobutanes and alkyl fluorides, selected from:
  • Figure US20230227446A9-20230720-C00119
        • In one embodiment of formula (A), R2 is selected from:
  • Figure US20230227446A9-20230720-C00120
        • In one embodiment, B is selected from: Me, Et, isopropyl, cyclopropyl, monofluoromethyl and difluoromethyl.
      • 2. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound of embodiment 1, wherein A is independently selected for the group consisting of O, S, CH2, and CH(Me), or a pharmaceutically acceptable salt, or solvate thereof.
      • 3. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound of embodiment 1, wherein X is N, each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof.
      • 4. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound of embodiment 1, wherein X is CH, each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof.
      • 5. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound of embodiment 1, wherein X is CR, each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof, R is selected from:
  • Figure US20230227446A9-20230720-C00121
        • G is selected from C1-4 alkyl, optionally substituted with OH and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl.
      • 6. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound of embodiment 1, wherein X is CR, Y is N and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof, R is selected from NHSO2G, SO2G, SO2NHG, OG, H, Me, and
  • Figure US20230227446A9-20230720-C00122
        • G is selected from C1-4 alkyl, optionally substituted with OH and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl.
      • 7. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound of embodiment 1, wherein A is O or S, each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof.
      • 8. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound of embodiment 1, wherein A is CH2, each of Y and Z is CH, X is CR, R is
  • Figure US20230227446A9-20230720-C00123
  • or a pharmaceutically acceptable salt, or solvate thereof.
      • 9. A method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) comprising: contacting kinase cells with an effective amount of a phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound having a structure of formula (A), or a pharmaceutically acceptable salt, or solvate thereof:
  • Figure US20230227446A9-20230720-C00124
        • A is selected from CH2, CH(Me), 0, and S;
        • X is selected from N, CH, and CR;
        • When Y=Z=CH, X is selected from N, CH and CR,
        • R is selected from:
  • Figure US20230227446A9-20230720-C00125
        • G is selected from C1-4 alkyl, optionally substituted with OH and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl.
        • Y is CH or N
        • Z is CH or N
        • When either Z or Y is N and X═CH and CR, R is selected from the following groups:
        • NHSO2G, SO2G, SO2NHG, OG, H, Me and
  • Figure US20230227446A9-20230720-C00126
        • G is selected from C1-4 alkyl, optionally substituted with OH and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl.
        • R1 is branched alkane, including substituted cyclopropanes, cyclobutanes and alkyl fluorides, selected from:
  • Figure US20230227446A9-20230720-C00127
        • In some embodiments of formula (A), R2 is selected from:
  • Figure US20230227446A9-20230720-C00128
  • In some embodiments, B is selected from: Me, Et, isopropyl, cyclopropyl, monofluoromethyl and difluoromethyl.
      • 10. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein A is independently selected for the group consisting of CH2, O and S, or a pharmaceutically acceptable salt, or solvate thereof.
      • 11. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein X is N, each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof.
      • 12. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein A is independently selected for the group consisting of O, S, CH2, and CH(Me), or a pharmaceutically acceptable salt, or solvate thereof.
      • 13. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein X is N, each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof.
      • 14. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein X is CH, each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof.
      • 15. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein X is CR, each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof, R is selected from:
  • Figure US20230227446A9-20230720-C00129
        • G is selected from C1-4 alkyl, optionally substituted with OH and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl.
      • 16. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein X is CR, Y is N and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof, R is selected from NHSO2G, SO2G, SO2NHG, OG, H, Me, and
  • Figure US20230227446A9-20230720-C00130
        • G is selected from C1-4 alkyl, optionally substituted with OH and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl.
      • 17. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein A is O or S, each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof.
      • 18. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein A is CH2, each of Y and Z is CH, X is CR, R is
  • Figure US20230227446A9-20230720-C00131
  • or a pharmaceutically acceptable salt, or solvate thereof.
      • 19. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 9, wherein the compound has optimized selectivity over PI3Kδ over a known PI3K gamma inhibitor.
      • 20. A selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor having a structure of formula (A), or a pharmaceutically acceptable salt, prodrug or solvate thereof:
        • provided is a compound of formula (A), or a pharmaceutically acceptable salt, prodrug, or solvate wherein:
  • Figure US20230227446A9-20230720-C00132
        • X=NHG, CH2G, OG, SO2G, SO2NHG, NHC(O)G, NHSO2G, C(O)NH2, C(O)NHG, C(O)NG1G2, cyclic amines or amides with 0-2 of O, N, or S atom and substituted with group G, and functional group with examples shown below.
  • Figure US20230227446A9-20230720-C00133
        • G is selected from H, D, OH, OMe, NH, SO2Me, C1-4 alkyl optionally substituted with H, OH, OMe, CN and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl
        • R1 is branched alkane, including substituted cyclopropanes, cyclobutanes and alkyl fluorides, selected from:
  • Figure US20230227446A9-20230720-C00134
        • In formula (A), R2 is selected from:
  • Figure US20230227446A9-20230720-C00135
        • A is selected from H, Me, Cl and difluoromethyl, B is selected from Me, Et, isopropyl, cyclopropyl, and difluoromethyl.
      • 21. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor of embodiment 20 wherein R1 is
  • Figure US20230227446A9-20230720-C00136
        • and R2 is
  • Figure US20230227446A9-20230720-C00137
          • X=NHG, CH2G, OG, SO2G, SO2NHG, NHSO2G, NHC(O)G, C(O)NH2, C(O)NHG, C(O)NG1G2, cyclic amines or amides with 0-2 of O, N, or S atom; G is selected from H, D, OH, OMe, NH2, C1-4 alkyl optionally substituted with H, OH, OMe, CN and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
      • 22. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor of embodiment 20, wherein R1 is
  • Figure US20230227446A9-20230720-C00138
        • and R2 is
  • Figure US20230227446A9-20230720-C00139
        • X═
  • Figure US20230227446A9-20230720-C00140
        • G is selected from H, D, OH, OMe, NH2, SO2Me, C1-4alkyl optionally substituted with H, OH, OMe, CN and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
      • 23. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor of embodiment 20, wherein
        • R1 is
  • Figure US20230227446A9-20230720-C00141
        • and R2 is
  • Figure US20230227446A9-20230720-C00142
          • X=NHG, OG, OCD3, NHSO2G, C(O)NH2, C(O)NHG, C(O)NG1G2; G is selected from C1-4 alkyl optionally substituted with H, OH, OMe, CN and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
      • 24. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor of embodiment 20, wherein R1 is
  • Figure US20230227446A9-20230720-C00143
        • and R2 is
  • Figure US20230227446A9-20230720-C00144
        • X═
  • Figure US20230227446A9-20230720-C00145
        • G is selected from H, D, OH, OMe, NH2, SO2Me, C1-4 alkyl optionally substituted with H, OH, OMe, CN and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
      • 25. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor of embodiment 20, wherein R is
  • Figure US20230227446A9-20230720-C00146
        • and R2 is
  • Figure US20230227446A9-20230720-C00147
        • X═OMe, OCD3, NHSO2Me, NHSO2Et, C(O)NH2, C(O)NHMe, C(O)NMe2, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
      • 26. The selective phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound of embodiment 20, wherein have the following structures, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
  • Figure US20230227446A9-20230720-C00148
    Figure US20230227446A9-20230720-C00149
    Figure US20230227446A9-20230720-C00150
    Figure US20230227446A9-20230720-C00151
    Figure US20230227446A9-20230720-C00152
    Figure US20230227446A9-20230720-C00153
    Figure US20230227446A9-20230720-C00154
      • 27. A method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) comprising: contacting kinase cells with an effective amount of a phosphoinositide 3-kinase gamma (PI3Kγ) inhibitor compound having a structure of formula (A), or a pharmaceutically acceptable salt, prodrug or solvate thereof:
  • Figure US20230227446A9-20230720-C00155
        • X=NHG, CH2G, OG, SO2G, SO2NHG, NHC(O)G, NHSO2G, C(O)NH2. C(O)NHG, C(O)NG1G2, cyclic amines or amides with 0-2 of O, N, or S atom and substituted with group G, and functional group with examples shown below.
  • Figure US20230227446A9-20230720-C00156
        • G is selected from H, D, OH, OMe, NH2, SO2Me, C1-4alkyl optionally substituted with H, OH, OMe, CN and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl
        • R1 is branched alkane, including substituted cyclopropanes, cyclobutanes and alkyl fluorides, selected from:
  • Figure US20230227446A9-20230720-C00157
        • In one embodiment of formula (A), R2 is selected from:
  • Figure US20230227446A9-20230720-C00158
        • In one embodiment, A is selected from H, Me, Cl and difluoromethyl, B is selected from Me, Et, isopropyl, cyclopropyl, and difluoromethyl.
      • 28. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 27, wherein
        • R1 is
  • Figure US20230227446A9-20230720-C00159
        • and R2 is
  • Figure US20230227446A9-20230720-C00160
  • or a pharmaceutically acceptable salt, prodrug or solvate thereof.
      • 29. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 27, wherein, X=NHG, CH2G, OG, SO2G, SO2NHG, NHC(O)G, NHSO2G, C(O)NH2, C(O)NHG, C(O)NG1G2, cyclic amines or amides with 0-2 of O, N, or S atom; G is selected from H, D, OH, OMe, NH2, SO2Me, C1-4 alkyl optionally substituted with H, OH, OMe, CN and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
      • 30. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 27, wherein X is selected from the following groups:
  • Figure US20230227446A9-20230720-C00161
        • G is selected from H, D, OH, OMe, NH2, SO2Me, C1-4 alkyl optionally substituted with H, OH, OMe, CN and F, such as methyl, ethyl, isopropyl, cyclopropyl, trifluoroethyl, 2-hydroxyethyl, 2-methoxyethyl, 3-hydroxypropyl, 3-methoxypropyl, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
      • 31. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 27, wherein X is,
  • Figure US20230227446A9-20230720-C00162
  • each of Y and Z is CH, or a pharmaceutically acceptable salt, or solvate thereof.
      • 32. The method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) according to embodiment 27, wherein the compound has optimized PI3Kδ selectivity and solubility compared with a known PI3K gamma inhibitor.
    EXAMPLES
  • Synthesis
  • Reagents and solvents used below can be obtained from commercial sources. 1H-NMR spectra were recorded on a Bruker 400 MHZ NMR spectrometer. Significant peaks are tabulated in the order: multiplicity (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br s, broad singlet), coupling constant(s) in Hertz (Hz) and number of protons. Mass spectrometry results are reported as the ratio of mass over charge, followed by the relative abundance of each ion (in parentheses Electrospray ionization (ESI) mass spectrometry analysis was conducted on an Shimadzu LC/MSD electrospray mass spectrometer.
  • The terms “solvent”, “inert organic solvent”, or “inert solvent” refer to a solvent inert under the conditions of the reaction being described in conjunction therewith (including, for example, benzene, toluene, acetonitrile, tetrahydrofuran (“THF”), dimethylformamide (“DMF”), ethyl acetate (EA or EtOAc), dichloromethane (DCM), diethyl ether, methanol, pyridine, formic acid (FA) and the like. Unless specified to the contrary, the solvents used in the reactions of the present invention are inert organic solvents, and the reactions are carried out under an inert gas, preferably nitrogen and argon.
  • (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-axoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (1)
  • Figure US20230227446A9-20230720-C00163
  • Step 1. A mixture of 4-bromo-2-methylbenzoic acid (1.0 g, 4.67 mmol), NIS (1.58 g, 7.01 mmol), Pd(OAc)2 (209 mg, 0.93 mmol) in DMF (20 mL) was stirred at 100° C. for 3 h. The mixture was cooled to rt, diluted with EtOAc (150 mL), washed with H2O (80 mL*4), brine (100 mL). The organic layer was dried, filtered, concentrated and purified by silica gel column eluting with ethyl acetate in petroleum ether (10%-25%) to afford 4-bromo-2-iodo-6-methylbenzoic acid as a light yellow solid (1.05 g, 66% yield). LC-MS (ESI) [M−H] 339.1.
  • Step 2. To a solution of 4-bromo-2-iodo-6-methylbenzoic acid (1 g, 2.94 mmol) in DMF (15 mL) was added K2CO3 (1.22 g, 8.82 mmol) followed by CH3I (835 mg, 5.88 mmol). The mixture was stirred at 40° C. for 1 h. The mixture was cooled to rt, diluted with EtOAc (150 mL), washed with H2O (80 mL*4), brine (100 mL). The organic layer was dried, filtered, concentrated and purified by silica gel column eluting with ethyl acetate in petroleum ether (0%-5%) to afford methyl 4-bromo-2-iodo-6-methylbenzoate as a yellow oil (970 mg, 93% yield).
  • Step 3. To a solution of methyl 4-bromo-2-iodo-6-methylbenzoate (970 mg, 2.74 mmol) in CCl4 (15 mL) was added NBS (1.17 g, 6.54 mmol), AIBN (180 mg, 1.1 mmol). The mixture was stirred at 80° C. for 17 h. The solvent was removed and the residue was purified by purified by silica gel column eluting with ethyl acetate in petroleum ether (0%-5%) to afford methyl 4-bromo-2-(bromomethyl)-6-iodobenzoate as a yellow oil. (920 mg, 82% yield).
  • Step 4. Preparation of (S)-5-bromo-2-(1-cyclopropylethyl)-7-iodoisoindolin-1-one (intermediate 1). To a solution of methyl 4-bromo-2-(bromomethyl)-6-iodobenzoate (920 mg, 2.12 mmol) in MeCN (15 mL) was added (S)-1-cyclopropylethan-1-amine (270 mg, 3.18 mmol), boric acid (129 mg, 2.12 mmol), K2CO3 (878 mg, 6.36 mmol). The mixture was stirred at rt for 17 h. Water (30 mL) and EtOAc (50 mL) were added. The reaction mixture was extracted with EtOAc (50 mL×2). The organic layer was dried, filtered, concentrated and purified by silica gel column eluting with ethyl acetate in petroleum ether (10%-80%) to afford (S)-5-bromo-2-(1-cyclopropylethyl)-7-iodoisoindolin-1-one as a yellow solid (470 mg, 55% yield). LC-MS (ESI) [M−H]+ 408.0.
  • Step 5. Synthesis of (S)-5-bromo-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-isoindolin-1-one (intermediate 2).
  • To a solution of (S)-5-bromo-2-(1-cyclopropylethyl)-7-iodoisoindolin-1-one (340 mg, 0.84 mmol) in 1,4-dioxane (12 mL) was added dimethylphosphine oxide (72 mg, 0.92 mmol), Pd2(dba)3 (77 mg, 0.084 mmol), xantphose (49 mg, 0.084 mmol), TEA (254 mg, 2.52 mmol) under N2. The mixture was stirred at rt for 6 h. Water (50 mL) and EA (50 mL) were added. The reaction mixture was extracted with EA (50 mL×2). The organic layer was dried, filtered, concentrated and purified by silica gel column eluting with ethyl acetate in petroleum ether (50%-100%) to afford (S)-5-bromo-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)isoindolin-1-one as a yellow solid (160 mg, 54% yield). LC-MS (ESI) [M−H]+ 356.1.
  • Step 6: (S)—N-(5-(2-(I-cyclopropylethyl)-7-(dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (General Procedure A)
  • To a solution of (S)-5-bromo-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-isoindolin-1-one (60 mg, 0.17 mmol) in DMF (3 mL) was added N-(4-methylthiazol-2-yl)acetamide (32 mg, 0.20 mmol), t-Bu3PBF4 (10 mg, 0.034 mmol), Cs2CO3 (110 mg, 0.34 mmol) and Pd(OAc)2 (4 mg, 0.017 mmol) under N2. The mixture was stirred at 100° C. for 5 h. Water (20 mL) and EA (20 mL) were added. The reaction mixture was extracted with EA (30 mL×3). The organic layer was dried, filtered, concentrated and purified by HPLC, condition: (SunFire C18 5 um 4.6×150 mm 25 C 1.000 mL/min 16 min; Mobile Phase B: 0.03% HCl in MeCN, Mobile Phase A: 0.03% HCl in H2O; rt: 8.86 min; 254 nm: 99.59%, 214 nm: 96.54%). LC-MS (ESI) [M+H]+ 432.1. 1H NMR (400 MHz, DMSO) δ 12.25 (s, 1H), 8.10 (dd, J=12.3, 1.5 Hz, 1H), 7.91 (s, 1H), 4.66 (s, 2H), 3.72-3.53 (m, 1H), 2.43 (s, 3H), 2.17 (s, 3H), 1.88 (d, J=14.4 Hz, 6H), 1.30 (d, J=6.8 Hz, 3H), 1.22-1.03 (m, 1H), 0.67-0.51 (m, 1H), 0.48-0.32 (m, 2H), 0.31-0.17 (m, 1H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-axoisoindolin-5-yl)-4-methylthiazol-2-yl)propionamide (2)
  • Figure US20230227446A9-20230720-C00164
  • A mixture of (S)-5-bromo-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)isoindolin-1-one (174 mg, 0.49 mmol), N-(4-methylthiazol-2-yl)propanamide (100 mg, 0.588 mmol), t-Bu3PBF4 (29 mg, 0.1 mmol), cesium carbonate (325 mg, 1.0 mmol) and palladium (II) acetate (11 mg, 0.05 mmol) in dimethylformamide (2 mL) was stirred at 100° C. for 16 h under N2. Cooled to rt, added EA (30 mL), washed with water (20 mL×2) and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by Pre-HPLC to afford (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)propionamide. (6.28 mg, 2.7%). LC-MS (ESI) [M+H]+=446.2. 1 H NMR (400 MHz, MeOH) δ 8.20 (d, J=12.8 Hz, 1H, 7.90 (s, 1H), 4.80-4.60 (m, 2H), 3.65 (dd, J=9.3, 6.9 Hz), 2.51 (q, J=7.6 Hz, 3H), 2.45 (s, 3H), 2.03 (dd, J=14.5, 2.5 Hz, 6H), 1.49-1.34 (m, 4H), 1.25-1.11 (m, 4H), 0.76-0.61 (m, 1H), 0.55-0.23 (m, 3H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)cyclopropanecarboxamide (3)
  • Step 1. To a solution of 4-methylthiazol-2-amine (500 mg, 4.38 mmol) in pyridine (10 mL) was added cyclopropanecarbonyl chloride (547 mg, 5.26 mmol) at room temperature. Then the reaction mixture was stirred for 2 h at room temperature. The solvent was evaporated and the residue was diluted EA (30 mL), washed with aqueous HCl (3N, 10 mL) and brine (20 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated. The residue was purified by silica gel column eluting with ethyl acetate/petroleum (0-20%) to afford N-(4-methylthiazol-2-yl)cyclopropanecarboxamide as a white solid (800 mg, 100% yield). LC-MS (ESI) [M+H]+ 183.2.
  • Step 2. (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)cyclopropanecarboxamide was prepared according to the general procedure A as an off-white solid (2.65 mg, 4.1% yield). LC-MS (ESI) [M+H]+ 458.2. 1H NMR (400 MHz, MeOD-d4) δ 8.60-8.43 (m, 1H), 8.19 (d, J=12.8 Hz, 1H), 7.90 (s, 1H), 4.77-4.65 (m, 2H), 3.70-3.62 (m, 1H), 2.48 (s, 3H), 2.04 (dd, J=14.5, 2.4 Hz, 6H), 1.94-1.88 (m, 1H), 1.40 (d, J=6.8 Hz, 3H), 1.21-1.13 (m, 1H), 1.08-1.02 (m, 2H), 1.01-0.95 (m, 2H), 0.72-0.65 (m, 1H), 0.54-0.48 (m, 1H), 0.46-0.40 (m, 2H), 0.30-0.18 (m, 1H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)isobutyramide (4)
  • (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)isobutyramide was prepared according to the general procedure A as a yellow solid (14.92 mg, 7.2%). LC-MS (ESI) [M+H]+=460.2, 1H NMR (400 MHz, DMSO) δ 8.10 (dd, J=12.4 Hz, 1H), 7.86 (s, 1H), 4.65 (s, 2H), 3.62 (dd, J=8.9, 6.8 Hz, 1H), 2.69 (dd, J=13.7, 6.8 Hz, 1H), 2.41 (s, 3H), 1.87 (d, J=14.1, 6H), 1.29 (d, J=6.8 Hz, 3H), 1.13 (t, J=8.4 Hz, 7H), 0.64-0.50 (m, 1H), 0.48-0.31 (m, 2H), 0.30-0.18 (m, 1H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-7-(diethylphosphoryl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (5)
  • Figure US20230227446A9-20230720-C00165
  • Step 1. To a solution of (S)-5-bromo-2-(1-cyclopropylethyl)-7-iodoisoindolin-1-one (250 mg, 0.62 mmol) in 1,4-dioxane (5 mL) was added diethylphosphine oxide (71 mg, 0.68 mmol), Pd2(dba)3 (56 mg, 0.06 mmol), Xantphose (35 mg, 0.06 mmol), TEA (0.3 mL, 1.15 mmol) under N2. The mixture was stirred at rt for 16 h. Water (30 mL) and EA (30 mL) were added. The reaction mixture was extracted with EA (30 mL×3). Then it was washed with brine, dried over anhydrous Na2SO4 and concentrated. The crude was purified by silica gel chromatography (PE:EA=1:0-1:1-0:1) the (DCM:MeOH=10:1) to give (S)-5-bromo-2-(1-cyclopropylethyl)-7-(diethylphosphoryl)isoindolin-1-one (100 mg, 42.1%) as a yellow oil. LC-MS (ESI) [M+H]+ 384.1.
  • Step 2. A mixture of (S)-5-bromo-2-(1-cyclopropylethyl)-7-(diethylphosphoryl)isoindolin-1-one (60 mg, 0.16 mmol), N-(4-methylthiazol-2-yl)acetamide (36 mg, 0.23 mmol), t-Bu3PBF4 (10 mg, 0.03 mmol), cesium carbonate (102 mg, 0.31 mmol) and palladium (II) acetate (4 mg, 0.016 mmol) in dimethylformamide (2 mL) was stirred at 100° C. for 16 hs under N2. Cooled to rt, added EA (30 mL), washed with water (20 mL×2) and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by Pre-HPLC to afford (S)—N-(5-(2-(1-cyclopropylethyl)-7-(diethylphosphoryl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (20 mg, 28.3%) as a white solid. LC-MS (ESI) [M+H]+=460.1; 1H NMR (400 MHz, DMSO) δ: 12.22 (s, 1H), 8.11 (m, 1H), 7.90 (s, 1H), 4.67 (s, 2H), 3.65 (m, 1H), 2.34 (s, 3H), 2.28 (m, 4H), 2.19 (s, 3H), 1.30 (d, 3H), 1.15 (m, 1H), 0.97 (m, 6H), 0.63 (m, 1H), 0.45 (m, 2H), 0.24 (m, 1H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-axoisoindolin-5-yl)pyridin-2-yl)acetamide (6)
  • Figure US20230227446A9-20230720-C00166
  • Step 1. To a solution of N-(5-bromo-6-methylpyridin-2-yl)acetamide (300.0 mg, 1.31 mol) in dioxane (3 mL) was added 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (399.07 mg, 1.57 mol), potassium acetate (192.79 mg, 1.96 mol) and Pd(dppf)Cl2 (10 mg). The mixture was stirred at 120° C. for 3 h under the N2. LCMS showed the reaction was completed. Water was added and extracted with EtOAc (3*10 mL). The organic layer was dried by NaSO4 and filtration. The filtrate was concentrated to afford crude product. The crude product was purification by silica gel chromatography PE:EtOAc (5:1) to afford N-(5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)acetamide (300 mg, 82.96%) as a white solid.
  • Step 2. To a solution of N-(6-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)acetamide (50.0 mg, 181.1 umol) in dioxane/H2O (2:0.5 mL), was added (S)-5-bromo-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)isoindolin-1-one (77.39 mg, 0.22 mmol), Na2CO3 (38.38 mg, 0.36 mmol) and Pd(dppf)Cl2 (5 mg). The mixture was stirred at 85° C. for 8 h under N2. LCMS showed the reaction was completed. Water was added and exacted with EtOAc (3*10 mL). The organic layer was dried by Na2SO4 and filtration. The filtrate was concentrated to afford crude product. The crude product was purification by HPLC (AcOH in water, ACN) to afford (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-oxoisoindolin-5-yl)pyridin-2-yl)acetamide (19.08 mg 26%) as white solid. LC-MS (ESI) [M+H]+ 426.3, 1H NMR (400 MHz, DMSO) δ 10.61 (s, 1H), 8.13 (s, 1H), 8.04 (d, J=8.4 Hz, 1H), 7.99 (d, J=12.1 Hz, 1H), 7.85 (s, 1H), 7.67 (d, J=8.5 Hz, 1H), 4.67 (s, 2H), 3.63 (s, 1H), 2.40 (s, 3H), 2.11 (s, 3H), 1.88 (d, J=14.3 Hz, 6H), 1.31 (d, J=6.8 Hz, 3H), 1.18-1.10 (m, 1H), 0.59 (s, 1H), 0.41 (dd, J=11.4, 5.4 Hz, 2H), 0.26 (d, J=5.4 Hz, 1H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-axoisoindolin-5-yl)pyridin-2-yl)acetamide (7)
  • (S)—N-(5-(2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-1-oxoisoindolin-5-yl)pyridin-2-yl)acetamide was prepared in the similar manner as the above analog (2.98 mg 4.75%) as white solid. LC-MS (ESI) [M+H]+ 412.31H NMR (400 MHz, MeOD) δ 8.71 (s, 1H), 8.50 (s, 1H), 8.35 (d, J=12.8 Hz, 1H), 8.25 (d, J=8.5 Hz, 1H), 8.20-8.15 (m, 1H), 8.11 (s, 1H), 4.58 (s, 2H), 3.72-3.59 (m, 1H), 2.21 (s, 3H), 2.04 (dd, J=14.4, 2.1 Hz, 6H), 1.40 (d, J=6.8 Hz, 3H), 1.18 (d, J=5.8 Hz, 1H), 0.69 (d, J=4.5 Hz, 1H), 0.55-0.37 (m, 2H), 0.36-0.26 (m, 1H).
  • (S)—N-(5-(6-(1-cyclopropylethyl)-7-oxo-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-3-yl)-4-methylthiazol-2-yl)acetamide (8)
  • Figure US20230227446A9-20230720-C00167
  • Step 1. A mixture of methyl 5-bromo-3-methylpicolinate (1.0 g, 4.35 mmol), N-bromosuccinimide (851 mg, 4.81 mmol), 2,2′-azobis(2-methylpropionitrile) (72 mg, 0.44 mmol) in carbon tetrachloride (20 mL) was stirred for 16 hr at 80° C. Removed solvent in vacuo, the residue was purified by silica gel chromatography (PE/EtOAc=1/1) to afford methyl 5-bromo-3-(bromomethyl)picolinate (800 mg, 59.7%) as white solid. LC-MS (ESI) [M+H]+=307.9
  • Step 2. A mixture of methyl 5-bromo-3-(bromomethyl)picolinate (150 mg, 0.49 mmol), (S)-1-cyclopropylethan-1-amine hydrochloride (59 mg, 0.49 mmol), boric acid (30 mg, 0.49 mmol) and potassium carbonate (201 mg, 1.46 mmol) in acetonitrile (4 mL) was stirred at rt for 16 hr under the N2. Filtered, the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (MeOH/DCM=1/10) to afford (S)-3-bromo-6-(1-cyclopropylethyl)-5,6-dihydro-7H-pyrrolo[3,4-b]pyridin-7-one (70 mg, 51.2% yield). LC-MS (ESI) [M+H]+=281.0.
  • Step 3. A mixture of (S)-3-bromo-6-(1-cyclopropylethyl)-5,6-dihydro-7H-pyrrolo[3,4-b]pyridin-7-one (70 mg, 0.25 mmol), N-(4-methylthiazol-2-yl)acetamide (58 mg, 0.37 mmol), t-Bu3PBF4 (14 mg, 0.05 mmol), cesium carbonate (163 mg, 0.5 mmol) and palladium (H) acetate (6 mg, 0.025 mmol) in dimethylformamide (2 mL) was stirred at 100° C. for 16 hr under N2. Cooled to rt, added EA (30 mL), washed with water (20 mL×2) and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by Pre-HPLC to afford (S)—N-(5-(6-(1-cyclopropylethyl)-7-oxo-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-3-yl)-4-methylthiazol-2-yl)acetamide (11.21 mg, 12.6%) as a white solid. LC-MS (ESI) [M+H]+=357.2, 1H NMR (400 MHz, DMSO) δ H: 12.27 (s, 1H), 8.79 (S, 1H), 8.15 (s, 1H), 4.61 (s, 2H), 7.67 (q, 1H), 2.41 (s, 3H), 2.17 (s, 3H), 1.32 (d, 3H), 1.28 (m, 1H), 0.58 (m, 1H), 0.43 (m, 2H), 0.24 (m, 2H).
  • (S)—N-(5-(6-(1-cyclopropylethyl)-5-oxo-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-2-yl)-4-methylthiazol-2-yl)acetamide (9)
  • Figure US20230227446A9-20230720-C00168
  • Step 1. A mixture of compound of methyl 6-hydroxy-2-methylnicotinate (500 mg, 2.99 mmol), POCl3 (2 mL) was added in DMF (5 mL) stirred at 120° C. under N2 for 2 h. After the reaction completed, the mixture was added Na2CO3 aqueous solution to adjust to pH=8. EA (30 mL) was added, washed with water (20 mL×2) and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel to afford methyl 6-chloro-2-methylnicotinate as a white solid (450 mg, 81.3%).
  • Step 2. A mixture of compound of methyl 6-chloro-2-methylnicotinate (361 mg, 2 mmol), NBS (391 mg, 2.2 mmol) and AIBN (32.8 mg, 0.2 mmol) was added in CCl4 (5 mL) then stirred under reflux for 4 h. After the reaction was completed, to the mixture was added EA (30 mL), washed with water (20 mL×2) and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel to afford methyl 2-(bromomethyl)-6-chloronicotinate as a white solid (300 mg, 52.3%).
  • Step 3. A mixture of compound of methyl 2-(bromomethyl)-6-chloronicotinate (260 mg, 1.1 mmol), (S)-1-cyclopropylethan-1-amine hydrochloride (180 mg, 1.5 mmol), boric acid (61 mg 1.0 mmol), K2CO3 (560 mg, 4 mmol) was added in MeCN (10 mL) and stirred at rt overnight. After the reaction was completed, to the mixture was added EA (30 mL), washed with water (20 mL×2) and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel to afford (S)-2-chloro-6-(1-cyclopropylethyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-5-one as white solid (74 mg, 31.3%).
  • Step 4. A mixture of compound of (S)-2-chloro-6-(1-cyclopropylethyl)-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-5-one (74 mg, 0.315 mmol), N-(4-methylthiazol-2-yl)acetamide (60 mg, 0.378 mmol). t-Bu3PBF4 (18 mg, 0.063 mmol), cesium carbonate (204.7 mg, 0.63 mmol), and palladium (II) acetate (7 mg, 0.0315 mmol) was added in DMF (4 mL) stirred at 100° C. overnight. After the reaction was completed, to the mixture was added EA (30 mL), washed with water (20 mL×2) and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by Pre-HPLC to afford (S)—N-(5-(6-(1-cyclopropylethyl)-5-oxo-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-2-yl)-4-methylthiazol-2-yl)acetamide as white solid (8.64 mg, 7.1%). 1H NMR (400 MHz, DMSO) δ H: 8.07-8.05 (d, J=8.0 Hz, 1H), 7.74-7.72 (d, J=8 Hz, 1H), 4.62 (s, 2H), 3.65-3.61 (m, 1H), 2.60 (s, 3H), 2.15 (s, 3H), 1.31-1.29 (d, J=8 Hz, 3H), 1.18-1.11 (m, 1H), 0.59-0.54 (m, 1H), 0.43-0.37 (m, 2H), 0.27-0.20 (m, 1H).
  • (S)-5-bromo-7-chloro-2-(1-cyclopropylethyl)isoindolin-1-one (intermediate 3) and (S)—N-(5-(7-chloro-2-(1-cyclopropylethyl)-1-axoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (Intermediate 4)
  • Figure US20230227446A9-20230720-C00169
  • (S)-5-bromo-7-chloro-2-(1-cyclopropylethyl)isoindolin-1-one (intermediate 3) and (S)—N-(5-(7-chloro-2-(1-cyclopropylethyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (intermediate 4) were prepared according to a known procedure (Pemberton, N., et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441)
  • (S)—N-(5-(2-(1-cyclopropylethyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (10)
  • Figure US20230227446A9-20230720-C00170
  • To a solution of (S)-5-bromo-7-chloro-2-(1-cyclopropylethyl)isoindolin-1-one containing 20% of (S)-5-bromo-2-(1-cyclopropylethyl)isoindolin-1-one (700 mg, 2.23 mmol) in DMF (10 mL) was added N-(4-methylthiazol-2-yl)acetamide (417 mg, 2.68 mmol), Cs2CO3 (1.4 g, 4.46 mmol), Pd(OAc)2 (50 mg, 0.22 mmol), t-Bu3PBF4 (129 mg, 0.46 mmol) under N2. The mixture was stirred at 100° C. for 3 h. Water (20 mL) and EtOAc (30 mL) were added. The reaction mixture was extracted with EtOAc (50 mL×3). Then it was washed with water (20 mL×4), dried over anhydrous Na2SO4 and concentrated to give the crude (700 mg). The crude (100 mg) was purified by pre-HPLC to give (S)—N-(5-(2-(1-cyclopropylethyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (7.82 mg) as a white solid. LCMS (ESI)[M+H]+356.2, 1H NMR (400 MHz, DMSO) δ 12.18 (s, 1H), 7.75-7.65 (m, 2H), 7.56 (d, J=7.9 Hz, 1H), 4.59 (s, 2H), 3.63-3.53 (m, 1H), 2.41 (d, J=9.0 Hz, 3H), 2.15 (d, J=4.6 Hz, 3H), 1.29 (d, J=6.8 Hz, 3H), 1.17-1.08 (m, 1H), 0.62-0.53 (m, 1H), 0.44-0.34 (m, 2H), 0.27-0.18 (m, 1H).
  • (S)-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-5-(4-methyl-2-(oxetan-3-ylamino)thiazol-5-yl)isoindolin-1-one (11)
  • Figure US20230227446A9-20230720-C00171
  • Step 1. A mixture of 4-methylthiazol-2-amine (1.0 g, 8.77 mmol), 3-bromooxetane (2.4 g, 17.54 mmol), K2CO3 (4 g, 21.93 mmol) in DMF (15 mL) was stirred at 120° C. for 48 h. The mixture was cooled to rt, diluted with EtOAc (150 mL), washed with H2O (80 mL*4), brine (100 mL). The organic layer was dried, filtered, concentrated and purified by silica gel column eluting with ethyl acetate in petroleum ether (0%-75%) to 4-methyl-N-(oxetan-3-yl)thiazol-2-amine as a light yellow solid. (320 mg, 21%). LC-MS (ESI) [M−H] 171.2.
  • Step 2. To a solution of (S)-5-bromo-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)isoindolin-1-one (60 mg, 0.17 mmol) in DMF (3 mL) was added 4-methyl-N-(oxetan-3-yl)thiazol-2-amine (57 mg, 0.34 mmol), t-Bu3PBF4 (10 mg, 0.034 mmol), Cs2CO3 (110 mg, 0.34 mmol) and Pd(OAc)2 (3.8 mg, 0.017 mmol) under N2. The mixture was stirred at 100° C. for 3 h. Water (30 mL) and EA (30 mL) were added. The reaction mixture was extracted with EA (30 mL×3). The organic layer was dried, filtered, concentrated and purified by Prep-HPLC to afford (S)-2-(1-cyclopropylethyl)-7-(dimethylphosphoryl)-5-(4-methyl-2-(oxetan-3-ylamino)thiazol-5-yl)isoindolin-1-one as a white solid (5.42 mg, 7% yield). HPLC condition: (SunFire C18 5 um 4.6×150 mm 25 C 1.000 mL/min 16 min; Mobile Phase B: 0.03% HCl in MeCN, Mobile Phase A: 0.03% HCl in H2O; rt: 8.86 min; 254 nm: 89.52%, 214 nm: 92.87%). LC-MS (ESI) [M+H]+ 446.2 1H NMR (400 MHz, MeOD) δ 8.11 (d, J=12.5 Hz, 1H), 7.95 (s, 1H), 4.81-4.66 (m, 2H), 4.14 (s, 1H), 3.98-3.75 (m, 4H), 3.73-3.60 (m, 1H), 2.43 (s, 3H), 2.05 (d, 6H), 1.40 (d, J=6.8 Hz, 3H), 1.23-1.11 (m, 1H), 0.76-0.65 (m, 1H), 0.55-0.39 (m, 2H), 0.37-0.28 (m, 1H), 0.36-0.30 (m, 1H).
  • Synthesis of (N-(5-(2-((S)-1-cyclopropylethyl)-7-(methylsulfinyl)-1-axoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (12)
  • Figure US20230227446A9-20230720-C00172
  • To a solution of (S)—N-(5-(2-(1-cyclopropylethyl)-7-(methylthio)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (prepared according to a known procedure (Pemberton, N., et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441) (230 mg, 057 mmol) in DCM (4 mL) was added mCPBA (99 mg, 0.57 mmol) at −40° C. The mixture was stirred at −40° C. for 1 h. Then aq.NaHCO3 (5 mL) was added, and stirring was continued for about 30 min. Water (30 mL) and DCM (20 mL) were added. The reaction mixture was extracted with DCM (20 mL×3). Then it was washed with brine, dried over anhydrous Na2SO4 and concentrated to give the crude product (135 mg). The crude product (50 mg) was purified by pre-HPLC to give racemic mixture of N-(5-(2-((S)-1-cyclopropylethyl)-7-((R)-methylsulfinyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide and N-(5-(2-((S)-1-cyclopropylethyl)-7-((S)-methylsulfinyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (12.54 mg, 24.12%) as a white solid. LC-MS (ESI) [M+H]+ 418.2. 1 H NMR (400 MHz, DMSO) δ 12.23 (s, 1H), 7.89 (s, 1H), 7.85 (s, 1H), 4.71 (s, 2H), 3.55 (t, J=6.9 Hz, 1H), 2.90 (d, J=3.8 Hz, 3H), 2.44 (s, 3H), 2.17 (s, 3H), 1.31-1.26 (m, 3H), 1.13 (d, J=7.6 Hz, 1H), 0.61-0.54 (m, 1H), 0.45-0.35 (m, 2H), 0.25 (d, J=4.9 Hz, 1H).
  • Preparation of N-(5-(2-((S)-1-cyclopropylethyl)-7-(ethylsulfinyl)-1-axoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (13)
  • Figure US20230227446A9-20230720-C00173
  • Step 1. To a solution of intermediate 4 (100 mg, 0.250 mmol) in DMF (2.00 mL) was added EtSNa (43.0 mg, 0.510 mmol) under N2. The mixture was stirred at 110° C. for 2 h. After being cooled to room temperature, the reaction was diluted with water (30 mL) and extracted with EA (20 mL×3). The combined organic layers was washed with brine (20 mL×3), dried over anhydrous Na2SO4, filtered and concentrated to give (S)—N-(5-(2-(1-cyclopropylethyl)-7-(ethylthio)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (80.0 mg, 0.193 mmol, 75.0% yield) as an oil. LC-MS (ESI) [M+H]+416.2.
  • Step 2. To a solution of (S)—N-(5-(2-(1-cyclopropylethyl)-7-(ethylthio)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (80 mg, crude) in DCM (2 mL) was added m-CPBA (33 mg, 0.190 mmol) at −40° C. under Ar. The mixture was stirred at −40° C. for 1 h. aq.NaHCO3 (5 mL) was added, and then the mixture was stirred for about 30 min, diluted with water (30 mL) and extracted with DCM (20 mL×3). The combined organic layers was dried over anhydrous Na2SO4, filtered, concentrated and purified by prep-HPLC to give a racemic mixture of N-(5-(2-((S)-1-cyclopropylethyl)-7-((R)-ethylsulfinyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide and N-(5-(2-((S)-1-cyclopropylethyl)-7-((S)-ethylsulfinyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (18.1 mg, 0.0421 mmol, 22.5% yield) as a white solid. LC-MS (ESI) [M+H]+432.1. 1H NMR (400 MHz, DMSO-d6): δ 12.2 (s, 1H), 7.83-7.82 (m, 2H), 4.71 (s, 2H), 3.56-3.52 (m, 1H), 3.30-3.25 (m, 1H), 2.94-2.88 (m, 1H), 2.43 (s, 3H), 2.17 (s, 3H), 1.29 (t, J=6.6 Hz, 3H), 1.16-1.12 (m, 1H), 1.10-1.05 (m, 3H) 0.61-0.55 (m, 1H), 0.44-0.36 (m, 2H), 0.27-0.24 (m, 1H).
  • Preparation of N-(5-(2-((S)-1-cyclopropylethyl)-7-(isopropylsulfinyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (14)
  • Figure US20230227446A9-20230720-C00174
  • Step 1. To a solution of intermediate 4 (100 mg, 0.256 mmol) in DMF (2 mL) was added NaH (20.5 mg, 0.512 mmol, 60% w/w dispersion in mineral oil) and propane-2-thiol (39.0 mg, 0.512 mmol) at rt. The mixture was stirred at 110° C. for 2 h under N2. After being cooled to rt, the reaction mixture was diluted with water (30 mL) and extracted with EA (20 mL×3). The combined organic layers was washed with brine (20 mL×3) and dried over anhydrous Na2SO4, filtered and concentrated to give (S)—N-(5-(2-(1-cyclopropylethyl)-7-(isopropylthio)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (40 mg, 0.0932 mmol, 36.3% yield). LC-MS (ESI) [M+H]+430.1.
  • Step 2. Compound 14 was prepared according to a similar procedure as the preparation of 13 (14.1 mg, 0.0317 mmol, 28.9% yield) as a white solid. LC-MS (EST) [M+H]+446.1. 1H NMR (400 MHz, DMSO-d6): δ 7.82 (s, 1H), 7.75 (s, 1H), 4.70 (d, J=2.4 Hz, 2H), 3.55-3.53 (m, 1H), 3.36-3.32 (m, 1H), 2.42 (s, 3H), 2.15 (s, 3H), 1.39 (d, J=6.8 Hz, 3H), 1.29 (t, J=8.0 Hz, 3H), 1.19-1.07 (m, 1H), 0.82 (t, J=7.6 Hz, 3H), 0.62-0.20 (m, 4H).
  • Synthesis of N-(5-(2-((S)-1-cyclopropylethyl)-7-(S-methylsulfonimidoyl)-1-axoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (15)
  • Figure US20230227446A9-20230720-C00175
  • To a solution of N-(5-(2-((S)-1-cyclopropylethyl)-7-(methylsulfinyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (20 mg, 0.048 mmol) in MeOH (3 mL) was added NH2COONH4 (16 mg, 0.192 mmol), PhI(OAc)2 (46 mg, 0.144 mmol). The mixture was stirred at 45° C. for 2 h. The mixture was purified by pre-HPLC to give a racemic mixture of N-(5-(2-((S)-1-cyclopropylethyl)-7-((R)—S-methylsulfinimidoyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide and N-(5-(2-((S)-1-cyclopropylethyl)-7-((S)—S-methylsulfinimidoyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (3.22 mg, 15.54%) as a white solid. LC-MS (ESI) [M+H]+433.2, 1H NMR (400 MHz, DMSO) δ 12.26 (s, 1H), 8.01 (s, 1H), 7.94 (s, 1H), 4.69 (s, 2H), 4.60 (s, 1H), 3.67-3.58 (m, 1H), 3.46 (s, 3H), 2.44 (s, 3H), 2.17 (s, 3H), 1.31 (d, J=5.2 Hz, 3H), 1.20-1.12 (m, 1H), 0.64-0.55 (m, 1H), 0.47-0.37 (m, 2H), 0.31-0.23 (m, 1H).
  • N-(5-((R)-2-((S)-1-cyclopropylethyl)-7-(dimethylphosphoryl)-3-methyl-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide and N-(5-((S)-2-((S)-1-cyclopropylethyl)-7-(dimethylphosphoryl)-3-methyl-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (16)
  • Figure US20230227446A9-20230720-C00176
  • Step 1. To a solution of 4-bromo-2-ethylbenzoic acid (Prepared according to Buckley, D. et al, U.S. Pat. Appl. Publ., 20170119786) (2 g, 8.7 mmol) in DMF (20 mL) was added Pd(OAc)2 (194 mg, 0.87 mmol), NIS (2.9 g, 13.1 mmol) under N2. The mixture was stirred at 100° C. for 3 h. Water (30 mL) and EtOAc (30 mL) were added. The reaction mixture was extracted with EtOAc (30 mL×2). Then it was washed with water (20 mL×3), dried over anhydrous Na2SO4 and concentrated. The crude was purified by silica gel column (PE:EA, 1:0-5:1) to give 4-bromo-2-ethyl-6-iodobenzoic acid (3.2 g, crude).
  • Step 2. To a solution of 4-bromo-2-ethyl-6-iodobenzoic acid (3 g, 8.47 mmol) in DMF (50 mL) was added CH3I (2.41 g, 16.96 mmol), K2CO3 (3.5 g, 25.4 mmol). The mixture was stirred at 40° C. for 2 h. Water (30 mL) and EtOAc (30 mL) were added. The reaction mixture was extracted with EtOAc (30 mL×2). Then it was washed with water (30 mL×5), dried over anhydrous Na2SO4 and concentrated. The crude was purified by silica gel column (PE:EA 100:0-100:15) to give methyl 4-bromo-2-ethyl-6-iodobenzoate (2.2 g, 71%) as a yellow oil. 1H NMR (400 MHz, DMSO) δ 7.95 (d, J=1.8 Hz, 1H), 7.60 (d, J=1.8 Hz, 1H), 3.87 (s, 3H), 2.49 (s, 2H), 1.11 (t, J=7.5 Hz, 3H).
  • Step 3. To a solution of methyl 4-bromo-2-ethyl-6-iodobenzoate (1 g, 2.82 mmol) in CCl4 (10 mL) was added NBS (528 mg, 2.97 mmol), AIBN (93 mg, 0.56 mmol). The mixture was stirred at 80° C. for 3 h. After being concentrated, the crude was purified by silica gel column (PE:EA=20:1) to give methyl 4-bromo-2-(1-bromoethyl)-6-iodobenzoate (1.1 g, 90%) as a yellow solid. 1H NMR (400 MHz, DMSO) δ 8.12 (d, J=1.7 Hz, 1H), 7.98 (d, J=1.7 Hz, 1H), 5.19 (d, J=6.8 Hz, 1H), 3.91 (s, 3H), 1.95 (d, J=6.8 Hz, 3H).
  • Step 4. To a solution of methyl 4-bromo-2-(1-bromoethyl)-6-iodobenzoate (1 g, 2.25 mmol) in MeCN (30 mL) was added (S)-1-cyclopropylethan-1-amine (286 mg, 3.37 mmol), boric acid (137 mg, 2.25 mmol), K2CO3 (1.3 g, 8.99 mmol). The mixture was stirred at 85° C. for 17 h. Water (30 mL) and EtOAc (50 mL) were added. The reaction mixture was extracted with EtOAc (50 mL×2). Then it was washed with brine, dried over anhydrous Na2SO4 and concentrated. The crude was purified by silica gel column (PE:EA, 1:0-20:1-10:1-5:1) to give 5-bromo-2-((S)-1-cyclopropylethyl)-7-iodo-3-methylisoindolin-1-one (650 mg, 69.31%) as a yellow oil. LC-MS (ESI) [M+H]+ 420.0.
  • Step 5. To a solution of 5-bromo-2-((S)-1-cyclopropylethyl)-7-iodo-3-methylisoindolin-1-one (650 mg, 1.56 mmol) in 1,4-dioxane (25 mL) was added dimethylphosphine oxide (133 mg, 1.71 mmol), Pd2(dba)3 (142 mg, 0.16 mmol), xantphose (90 mg, 0.16 mmol), TEA (0.64 mL, 4.67 mmol) under N2. The mixture was stirred at rt for 17 h. Water (50 mL) and EA (50 mL) were added. The reaction mixture was extracted with EA (50 mL×2). Then it was washed with brine, dried over anhydrous Na2SO4 and concentrated. The crude was purified by silica gel column (PE:EA=1:0-10:1-0:1; DCM:MeOH=10:1) to give 5-bromo-2-((S)-1-cyclopropylethyl)-7-(dimethylphosphoryl)-3-methylisoindolin-1-one (300 mg, 52.37%) as a yellow oil. LC-MS (ESI) [M+H]+ 372.0.
  • Step 6. To a solution of 5-bromo-2-((S)-1-cyclopropylethyl)-7-(dimethylphosphoryl)-3-methylisoindolin-1-one (260 mg, 0.71 mmol) in DMF was added N-(4-methylthiazol-2-yl)acetamide (132 mg, 0.85 mmol), t-Bu3PBF4 (41 mg, 0.14 mmol), Cs2CO3 (460 mg, 1.41 mmol) and Pd(OAc)2 (16 mg, 0.07 mmol) under N2. The mixture was stirred at 100° C. for 2 h. Water (30 mL) and EA (30 mL) were added. The reaction mixture was extracted with EA (30 mL×3). Then it was washed with brine, dried over anhydrous Na2SO4 and concentrated. The crude was purified by pre-HPLC to give P1 (6.42 mg) and P2 (5.84 mg). LC-MS (ESI) [M+H]+ 446.3. P1: 1H NMR (400 MHz, DMSO) δ 8.10 (d, J=12.6 Hz, 1H), 7.92 (s, 1H), 4.94 (d, J=6.5 Hz, 1H), 2.43 (s, 3H), 2.17 (s, 3H), 1.92-1.80 (m, 6H), 1.59 (d, J=6.5 Hz, 3H), 1.38 (t, J=6.7 Hz, 4H), 0.62 (d, J=3.6 Hz, 1H), 0.49 (s, 1H), 0.37 (d, J=4.6 Hz, 1H), 0.13 (d, J=4.4 Hz, 1H); P2: 1H NMR (400 MHz, DMSO) δ 8.10 (dd, J=12.3, 1.3 Hz, 1H), 7.90 (s, 1H), 4.84 (d, J=6.6 Hz, 1H), 2.42 (s, 3H), 2.17 (s, 3H), 1.88 (t, J=13.9 Hz, 6H), 1.60 (d, J=6.6 Hz, 3H), 1.43 (d, J=7.0 Hz, 3H), 1.37-1.28 (m, 1H), 0.55 (dd, J=8.5, 5.0 Hz, 1H), 0.40 (dd, J=9.1, 5.0 Hz, 2H), 0.31 (dd, J=9.2, 4.5 Hz, 1H).
  • Preparation of N-(5-(2-(tert-butyl)-7-(methylsulfinyl)-1-axoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide (17)
  • Figure US20230227446A9-20230720-C00177
  • Step 1. 5-bromo-2-(tert-butyl)-7-iodoisoindolin-1-one was synthesized based on the similar procedure of preparation of intermediate 1 (100 mg, yield 10.9%) as colorless oil. LC-MS (ESI) [M+H]˜ 393.9
  • Step 2. To a mixture of 5-bromo-2-(tert-butyl)-7-iodoisoindolin-1-one (90 mg, 0.219 mmol) in Tol/acetone (3 mL/1.5 mL) was added potassium ethanethioate (21 mg, 0.276 mmol), Pd2(dba)3 (19.8 mg, 0.0319 mmol) and XantPhos (25.2 mg, 0.0438 mmol) at room temperature. The mixture was stirred at 45° C. under argon atmosphere for 0.5 hours. The reaction mixture was cooled to room temperature and concentrated in vacuum to give the residue, which was purified by prep-TLC (PE:EA=3:1) to give S-(6-bromo-2-(tert-butyl)-3-oxoisoindolin-4-yl) ethanethioate (90 mg, crude) as a white solid. LC-MS (ESI) [M+H]+ 342.0
  • Step 3. To a solution of S-(6-bromo-2-(tert-butyl)-3-oxoisoindolin-4-yl) ethanethioate (60 mg, 0.175 mmol) in MeOH (4.00 mL) was added Cs2CO3 (85 mg, 0.262 mmol) at room temperature. The mixture was stirred at room temperature under argon atmosphere for 1 hour. MeI (124 mg, 0.877 mmol) was added and the mixture was stirred at room temperature under argon atmosphere for another 16 hours. The reaction mixture was concentrated in vacuum to give the residue, which was purified by prep-TLC (PE:EA=3:1, v/v) to give 5-bromo-2-(tert-butyl)-7-(methylthio)isoindolin-1-one (60 mg, over 100% yield) as a white solid. LC-MS (ESI) [M+H]+ 316.2
  • Step 4 and 5. Racemic mixture of (R)—N-(5-(2-(tert-butyl)-7-(methylsulfinyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide and (S)—N-(5-(2-(tert-butyl)-7-(methylsulfinyl)-1-oxoisoindolin-5-yl)-4-methylthiazol-2-yl)acetamide was synthesized in a similar manner as the preparation of 13 (5 mg, 13.0% yield) as a white solid. LC-MS (ESI) [M+H]+406.0. 1H NMR (400 MHz, MeOD-d4): δ 8.00 (s, 1H), 7.77 (s, 1H), 4.76 (s, 2H), 2.98 (s, 3H), 2.45 (s, 3H), 2.23 (s, 3H), 1.56 (s, 9H).
  • N-(4-(difluoromethyl)-5-iodothiazol-2-yl)acetamide (intermediate 5)
  • Figure US20230227446A9-20230720-C00178
  • Step 1. A solution of 4-(difluoromethyl)thiazol-2-amine hydrochloride (Prepared according to Guo, L. et al, PCT Int. Appl., 2014037480) (700 mg, crude) in acetic anyhydride (12.0 mL) was stirred at 140° C. for 2 hours. The reaction mixture was cooled to rt and concentrated in vacuum to give the residue, which was purified by chromatography on silica gel (PE:EA=10:1 to 3:1) to give the title compound N-(4-(difluoromethyl)thiazol-2-yl)acetamide (350 mg, 39.0% yield over 2 steps) as a white solid. LC-MS (ESI) [M+H]+=193.1
  • Step 2. A solution of N-(4-(difluoromethyl)thiazol-2-yl)acetamide (120 mg, 0.624 mmol) and NIS (168 mg, 0.748 mmol) in MeCN (5.00 mL) was stirred at 90° C. for 16 hours under Ar. The reaction mixture was cooled to rt and concentrated in vacuum to give the residue, which was purified by chromatography on silica gel (PE:EA=10:1 to 3:1) to give intermediate 5 N-(4-(difluoromethyl)-5-iodothiazol-2-yl)acetamide (130 mg, 65.5% yield) as a yellow solid. LC-MS (ESI) [M+H]+318.9
  • Preparation of N-(5-(2-((S)-1-cyclopropylethyl)-7-(methylsulfinyl)-1-axoisoindolin-5-yl)-4-(difluoromethyl)thiazol-2-yl)acetamide (18)
  • Figure US20230227446A9-20230720-C00179
  • Step 1. A mixture of 5-bromo-2-((S)-1-cyclopropylethyl)-7-(methylsulfinyl)isoindolin-1-one (260 mg, 0.760 mmol), 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (386 mg, 1.519 mmol), Pd(dppf)Cl2 (56 mg, 0.076 mmol) and KOAc (224 mg, 2.280 mmol) in dioxane (2.00 mL) was stirred at 80° C. under argon atmosphere for 1 hours. The reaction mixture was cooled to rt and concentrated in vacuum to give the residue, which was purified by chromatography on silica gel (PE:EA=10:1 to 3:1) to give the title compound 2-((S)-1-cyclopropylethyl)-7-(methylsulfinyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoindolin-1-one (160 mg, yield 54.1%) as a white solid. LC-MS (ESI) [M+H]+ 390.1
  • Step 2. A mixture of 2-((S)-1-cyclopropylethyl)-7-(methylsulfinyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)isoindolin-1-one (50 mg, 0.128 mmol), intermediate 5 (45 mg, 0.141 mmol), Pd(dppf)Cl2 (10 mg, 0.0130 mmol) and K2CO3 (53 mg, 0.385 mmol) in dioxane/H2O (2.00 mL/0.4 mL) was stirred at 90° C. under argon atmosphere for 2 hours. The reaction mixture was cooled to rt and concentrated in vacuum to give the residue, which was purified by prep-HPLC (base, NH3H2O) to give a racemic mixture of N-(5-(2-((S)-1-cyclopropylethyl)-7-((R)-methylsulfinyl)-1-oxoisoindolin-5-yl)-4-(difluoromethyl)thiazol-2-yl)acetamide and N-(5-(2-((S)-1-cyclopropylethyl)-7-((S)-methylsulfinyl)-1-oxoisoindolin-5-yl)-4-(difluoromethyl)-thiazol-2-yl)acetamide (5 mg, yield 8.60%) as a white solid. LC-MS (ESI) [M+H]+ 454.1. 1H NMR (400 MHz, CDCl3) δ: 9.80 (s, 1H), 8.10 (s, 1H), 7.57 (s, 1H), 6.50 (t, J=54.0 Hz, 1H), 4.65-4.49 (m, 2H), 3.65-3.61 (m, 3H), 2.97 (s, 3H), 1.32-1.29 (m, 3H), 0.96-0.81 (m, 1H), 0.80-0.79 (m, 1H), 0.43-0.33 (m, 3H).
  • General Procedure B:
  • To a solution of intermediate 8 (0.128 mmol) in 1,4-dioxane (2.00 mL) was added amide or other nucleophilic reagents (0.512 mmol), Cs2CO3 (125 mg, 0.384 mmol), X-phos G3 (8.80 mg, 0.0128 mmol) and Pd(OAc)2 (8.32 mg, 0.0256 mmol) at room temperature. The mixture was heated to 110° C. for 1 hour-8 hours under Ar. The reaction mixture was cooled to room temperature. The mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC to give the desired product.
  • General Procedure C:
  • A mixture of halides (0.375 mmol), thiazole (0.375 mmol), Pd(OAc)2 (17 mg, 0.0750 mmol), t-Bu3P.BF4 (22 mg, 0.0750 mmol) and Cs2CO3 (366 mg, 1.13 mmol) in DMA (2 mL) was stirred at 100° C.-135° C. for 1 h-4 h under Ar. The mixture was cooled to rt and diluted with water (10 mL), extracted with ethyl acetate (10 mL×3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC to give the title product.
  • Preparation of(S)-4,6-dichloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (intermediate 6)
  • Figure US20230227446A9-20230720-C00180
  • Step a. To a solution of 2,6-dichloro-4-methylnicotinic acid (50.0 g, 244 mmol) in DMF (350 mL) was added K2CO3 (101 g, 731 mmol) and CH3I (104 g, 731 mmol) at rt. The mixture was stirred at 40° C. for 2 h. After being cooled to rt, the mixture was diluted with water (530 mL) and extracted with EtOAc (250 mL×3). The combined organic layers was washed with brine (250 mL×4), dried over anhydrous Na2SO4 and concentrated. The residue was purified by silica gel chromatography (PE:EA=1:0-20:1, v/v) to give 2,6-dichloro-4-methylnicotinate (50.2 g, 93.93% yield, contained DMF) as a white solid. 1H NMR (400 MHz, CDCl3): δ 7.14 (s, 1H), 3.95 (s, 3H), 2.32 (s, 3H).
  • Step b. To a solution of methyl 2,6-dichloro-4-methylnicotinate (50.0 g, 228 mmol) in CCl4 (450 mL) was added NBS (163 g, 913 mmol), BPO (55.3 g, 159 mmol) at rt. The mixture was stirred at 90° C. and with concomitant irradiation from a tungsten lamp for 48 hours. After being cooled to rt, the mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EA=1:0-20:1, v/v) to give methyl 2,6-dichloro-4-(dibromomethyl)nicotinate (85.2 g, 99.25% yield, contained benzoic acid) as a yellow oil. 1H NMR (400 MHz, CDCl3): δ 7.87 (s, 1H), 6.69 (s, 1H), 4.03 (s, 3H).
  • Step c. To a solution of 2,6-dichloro-4-(dibromomethyl)nicotinate (85.2 g, 226 mmol) in 260 mL of CH3CN, DIPEA (58.5 g, 453 mmol) was added at 0° C. Then a solution of diethyl phosphite (31.3 g, 227 mmol) in 200 mL of CH3CN was added drop wise carefully at 0° C. The mixture was stirred at 0° C. for another hour. The mixture was diluted with cold aq. NaHCO3 (200 mL) and extracted with EA (200 mL×4). The combined organic layers was washed with brine (200 mL) and dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE:EA=1:0-20:1, v/v) to give methyl 4-(bromomethyl)-2,6-dichloronicotinate (60.2 g, 88.9% yield) as a yellow oil. 1H NMR (400 MHz, CDCl3): δ 7.37 (s, 1H), 4.40 (s, 2H), 4.01 (s, 3H).
  • Step d. To a solution of methyl 4-(bromomethyl)-2,6-dichloronicotinate (60.2 g, 201 mmol) in MeCN (350 mL) was added (S)-1-cyclopropylethan-1-amine (29.4 g, 242 mmol), boric acid (14.9 g, 242 mmol) and K2C0M (83.4 g, 604 mmol) at rt. The mixture was stirred at 60° C. for 2 h. After being cooled to rt, the mixture was diluted with water (450 mL) and EA (350 mL). The reaction mixture was extracted with EA (250 mL×3). The combined organic layers was washed with brine (200 mL), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by silica gel column chromatography (PE:EA=1:0-20:1, v/v) to give (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (intermediate 6) (30.2 g, 55.6% yield) as a white solid. LC-MS (ESI) [M+H]+ 271.1. 1H NMR (400 MHz, DMSO-d6): δ 7.89 (s, 1H), 4.62 (s, 2H), 3.57-3.53 (m, 1H), 1.27 (d, J=6.8 Hz, 3H), 1.12-1.09 (m, 1H), 0.59-0.54 (m, 1H), 0.43-0.21 (m, 3H).
  • Preparation of tert-butyl (4-methyl-5-(tributylstannyl)thiazol-2-yl)carbamate (intermediate 7)
  • Figure US20230227446A9-20230720-C00181
  • Step a. To a solution of 4-methylthiazol-2-amine (40.0 g, 347 mmol) in THF/H2O (800 mL/800 mL) was added NaHCO3 (175 g, 2.09 mol) and Boc2O (454 g, 2.09 mol) at rt. The mixture was stirred at 50° C. for 16 hours. The reaction mixture was cooled to rt. The mixture was concentrated in vacuum to remove the THF. The mixture was diluted with n-hexane, the solid was filtered and dried to give tert-butyl (4-methylthiazol-2-yl)carbamate (42.0 g, yield 56.6%) as a white solid. LC-MS (ESI): [M+H]+ 215.1
  • Step b. To a solution of tert-butyl (4-methylthiazol-2-yl)carbamate (42.0 g, 196 mmol) in THF (300 mL) was added LDA (304 mL, 608 mmol, 2.0 M) dropwise at −78° C. under argon atmosphere. The mixture was stirred at −78° C. for 1 hour under Ar. To the mixture was added a solution of n-Bu3SnCl (63.8 g, 196 mmol) in THF (120 mL). And then the mixture was stirred at −78° C. for 12 h. The reaction mixture was quenched with ice water (200 mL) and extracted with EA (200 mL×3). The combined organic phase was concentrated in vacuum to give the residue, which was purified by chromatography on silica gel (PE:EA=200:1 to 10:1) to give intermediate 7 tert-butyl (4-methyl-5-(tributylstannyl)thiazol-2-yl)carbamate (32.0 g, yield 32.4%) as a colorless oil. 1H NMR (400 MHz, DMSO-d6): δ: 11.27 (s, 1H), 2.22 (s, 3H), 1.55-1.41 (m, 15H), 1.33-1.26 (m, 6H), 1.19-1.10 (m, 5H), 0.99-0.78 (m, 10H).
  • Preparation of (S)—N-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (Intermediate 8)
  • Figure US20230227446A9-20230720-C00182
  • Step a. To a solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (intermediate 6) (500 mg, 1.80 mmol) in dioxane (10 mL) was added tert-butyl (4-methyl-5-(tributylstannyl)thiazol-2-yl)carbamate (intermediate 7) (1.81 g, 3.60 mmol), Pd(PPh3)4 (415 mg, 0.360 mmol) at rt. The mixture was stirred at 160° C. for 1 h under argon atmosphere in microwave reactor. The reaction mixture was cooled to rt, diluted with ice water (50 mL) and extracted with EA (50 mL×3). The combined organic phase was washed with brine and dried over Na2SO4, filtered and concentrated. The residue was purified by chromatography on silica gel (DCM:MeOH=200:1 to 20:1, v/v) to give the title compound tert-butyl (S)-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)carbamate (566 mg, 35.0% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6): 5:11.67 (s, 1H), 7.86 (s, 1H), 4.63 (s, 2H), 3.61-3.55 (m, 1H), 2.57 (s, 3H), 1.47 (s, 9H), 1.31-1.23 (m, 3H), 1.14-1.09 (m, 1H), 0.59-0.57 (m, 1H), 0.42-0.37 (m, 2H), 0.33-0.26 (m, 1H).
  • Step b. To a solution of tert-butyl (S)-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)carbamate (566 mg, 1.26 mmol) in DCM (2 mL) was added TFA (1.5 mL) at rt. The mixture was stirred at rt for 1 h. The mixture was quenched with aq. NaHCO3 (20 mL) and extracted with DCM/MeOH (20 mL/1 mL×3). The combined organic phase was washed with brine and dried over Na2SO4, filtered and concentrated. The residue was purified by chromatography on silica gel (DCM:MeOH=200:1 to 20:1) to give the title compound (S)-6-(2-amino-4-methylthiazol-5-yl)-4-chloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (300 mg, 68.3% yield) as a yellow solid. LC-MS (ESI): [M+H]+349.1
  • Step c. To a solution of (S)-6-(2-amino-4-methylthiazol-5-yl)-4-chloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (300 mg, 0.86 mmol) in DCM (10 mL) was added TEA (261 mg, 2.58 mmol) and acetyl chloride (134 mg, 1.72 mmol) at 0° C. The mixture was stirred at rt for 1 h under Ar. The reaction mixture was diluted with ice water (50 mL) and extracted with EA (50 mL×3). The combined organic phase was washed with brine and dried over Na2SO4, filtered and concentrated. The residue was purified by chromatography on silica gel (DCM:MeOH=200:1 to 20:1, v/v) to give the title compound (S)—N-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (intermediate 8) (56 mg, yield 16.7%) as a yellow solid. LC-MS (ESI): [M+H]+ 391.0. 1H NMR (400 MHz, DMSO-d6): δ: 12.30 (s, 1H) 7.90 (s, 1H), 4.64 (s, 2H), 3.59-3.55 (m, 1H), 2.61 (s, 3H), 2.17 (s, 3H), 1.27 (d, J=6.8 Hz, 3H), 1.15-1.10 (m, 1H), 0.59-0.57 (m, 1H), 0.43-0.41 (m, 2H), 0.33-0.25 (m, 1H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(methylsulfonyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (19)
  • Figure US20230227446A9-20230720-C00183
  • Step a. To a solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (intermediate 6) (1.00 g, 3.69 mmol) in DMF (10.0 mL) was added sodium methanethiolate (271 mg, 3.87 mmol) at room temperature. The mixture was stirred at room temperature for 16 h. The mixture was diluted with water (100 mL) and extracted with ethyl acetate (50.0 mL×3). The combined organic layer was washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EA=5/1, v/v) to give (S)-6-chloro-2-(1-cyclopropylethyl)-4-(methylthio)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (300 mg, 28.8% yield) as a yellow solid. LC-MS (ESI) [M+H]+ 283.0
  • Step b. (S)-6-(2-amino-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-4-(methylthio)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one was prepared according to general procedure C (200 mg, 40.1%) as a brown solid. Boc group was removed under this reaction condition. LC-MS (ESI) [M+H]+ 361.1
  • Step c. To a solution of (S)-6-(2-amino-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-4-(methylthio)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (200 mg, 76.3% purity, 0.424 mmol) in dichloromethane (10.0 mL) was added triethylamine (128 mg, 1.27 mmol) and acetyl chloride (66 mg, 0.848 mmol) at 0° C. under Ar. The mixture was stirred at room temperature for 1 hour. The mixture was diluted with water (30.0 mL) and extracted with ethyl acetate (20.0 mL×3). The combined organic layer was washed with brine (30.0 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane/methanol=10/1, v/v) to give (S)—N-(5-(2-(1-cyclopropylethyl)-4-(methylthio)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (60 mg, 35.1% yield) as a slight yellow solid. LC-MS (ESI) [M+H]+ 403.2. 1H NMR (400 MHz, MeOD-d4): δ 7.51 (s, 1H), 4.69-4.57 (m, 2H), 3.62-3.57 (m, 1H), 2.63 (d, J=8.4 Hz, 6H), 2.23 (s, 3H), 1.37 (d, J=6.8 Hz, 3H), 1.14-1.11 (m, 1H), 0.68-0.64 (m, 1H), 0.51-0.48 (m, 1H), 0.42-0.38 (m, 1H), 0.35-0.30 (m, 1H).
  • Step d. To a solution of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(methylthio)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (60 mg, 0.149 mmol) in dichloromethane (5.00 mL) was added 3-chlorobenzoperoxoic acid (76 mg, 0.440 mmol) at 0° C. The mixture was stirred at 0° C. for 4 hours. The mixture was diluted with sodium sulfite solution (20.0 mL) and extracted with ethyl acetate (20.0 mL×3). The combined organic layer was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC to give (S)—N-(5-(2-(1-cyclopropylethyl)-4-(methylsulfonyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (19) (37.91 mg, 58.5% yield) as a white solid. LC-MS (ESI) [M+H]+ 435.1. 1H NMR (400 MHz, MeOD-d4): δ 8.08 (s, 1H), 4.86-4.73 (m, 2H), 3.70-3.65 (m, 1H), 3.52 (s, 3H), 2.71 (s, 3H), 2.24 (s, 3H), 1.39 (d, J=7.2 Hz, 3H), 1.16-1.15 (m, 1H), 0.69-0.67 (m, 1H), 0.52-0.34 (m, 3H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(methylsulfonamido)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (20)
  • Figure US20230227446A9-20230720-C00184
  • Prepared 20 according to the general procedure B (110° C. for 1 hours) (4.47 mg, 7.78% yield) as a light yellow solid. LC-MS (ESI) [M+H]+450.1. 1H NMR (400 MHz, MeOD-d4): δ 7.36 (s, 1H), 4.68-4.60 (m, 2H), 3.58-3.50 (m, 1H), 3.46 (s, 3H), 3.31 (m, 3H), 2.22 (s, 3H), 1.36 (d, J=6.8 Hz, 3H), 1.05-0.96 (m, 1H), 0.64-0.58 (m, 1H), 0.43-0.26 (m, 3H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(ethylsulfonyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (21)
  • Prepared 21 according to the similar procedure as that of 19 (1.86 mg, 6.70% yield) as a light yellow solid. LC-MS (ESI) [M+H]+449.3. 1H NMR (400 MHz, MeOD-d4): δ 8.08 (s, 1H), 4.86-4.75 (m, 2H), 3.82-3.76 (m, 2H), 3.70-3.59 (m, 1H), 2.70 (s, 3H), 2.24 (s, 3H), 1.44-1.38 (m, 6H), 1.20-1.13 (m, 1H), 0.71-0.66 (m, 1H), 0.54-0.44 (m, 2H), 0.42-0.32 (m, 1H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-methoxy-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (22)
  • Figure US20230227446A9-20230720-C00185
  • Step a. To a solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (intermediate 6) (300 mg, 1.11 mmol) in MeOH (5 mL) was added MeONa (120 mg, 2.22 mmol) slowly at rt. The mixture was stirred at rt overnight. The mixture was quenched with aq. NH4Cl (10 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography eluted with petroleum ether:ethyl acetate (100:1-9:1) to give (S)-6-chloro-2-(1-cyclopropylethyl)-4-methoxy-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (130 mg, 43.9% yield) as a white solid. LC-MS (ESI) [M+H]+267.1.
  • Step b. Prepared 22 according to General procedure C (135° C., 1 h) (5.00 mg FA salt, 3.33% yield) as a white solid. LC-MS (ESI) [M+H]+387.0. 1H NMR (400 MHz, CDCl3): δ 8.19 (s, 0.29H, FA), 7.21 (s, 1H), 4.56-4.40 (m, 2H), 4.14 (s, 3H), 3.76-3.72 (m, 1H), 2.68 (s, 3H), 2.31 (s, 3H), 1.33 (d, J=6.8 Hz, 3H), 1.00-0.98 (m, 1H), 0.63-0.62 (m, 1H), 0.44-0.37 (m, 3H).
  • Preparation of (R)-4,6-dichloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (Intermediate 9)
  • Figure US20230227446A9-20230720-C00186
  • Prepared 9 according to a similar procedure as preparation of intermediate 6 (60° C., 2 hours) (2.50 g, 9.26 mmol, 69.1% yield) as a white solid. LC-MS (ESI) [M+H]+270.9. 1H NMR (400 MHz, DMSO-d6): δ 7.88 (s, 1H), 4.62 (s, 2H), 3.55-3.52 (m, 1H), 1.27 (d, J=6.8 Hz, 3H), 1.15-1.02 (m, 1H), 0.64-0.51 (m, 1H), 0.47-0.33 (m, 2H), 0.30-0.16 (m, 1H).
  • Preparation of 4,6-dichloro-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (Intermediate 10)
  • Figure US20230227446A9-20230720-C00187
  • Prepared intermediate 10 according to a similar procedure as the preparation of intermediate 6 (1.16 g, 3.89 mmol, 14.5% yield) as a white solid. LC-MS (ESI) [M+H]+298.9. 1H NMR (400 MHz, DMSO-do): δ 7.90 (s, 1H), 5.03-5.01 (m, 1H), 4.73-4.48 (m, 2H), 1.47 (d, J=7.2 Hz, 3H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(ethylsulfonamido)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (23)
  • Prepared 23 according to General procedure B (110° C. for 6 hours) (10.39 mg, 21.9% yield) as a slightly yellow solid. LC-MS (ESI) [M+H]+ 464.3. 1H NMR (400 MHz, CDCl3): δ 9.04 (s, 1H), 7.32 (s, 1H), 4.61-4.45 (m, 2H), 3.76-3.65 (m, 3H), 2.70 (s, 3H), 2.34 (s, 3H), 1.31 (t, J=7.2 Hz, 3H), 1.36 (d, J=6.8 Hz, 3H), 1.02-0.99 (m, 1H), 0.69-0.66 (m, 1H), 0.54-0.26 (m, 3H).
  • Preparation of (S)—N-(5-(4-(cyclopropanesulfonamido)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (24)
  • Prepared 24 according to General procedure B (110° C. for 8 hours) (5.90 mg, 12.1% yield) as a slight yellow solid. LC-MS (ESI) [M+H]+476.3. 1H NMR (400 MHz, CDCl3): δ 9.20 (s, 1H), 7.32 (s, 1H), 4.61-4.45 (m, 2H), 3.76-3.65 (m, 1H), 3.29-3.26 (m, 1H), 2.72 (s, 3H), 2.55 (s, 3H), 1.49-1.44 (m, 2H), 1.36 (d, J=6.8 Hz, 3H), 1.14-1.09 (m, 2H), 1.05-0.96 (m, 1H), 0.71-0.64 (m, 1H), 0.54-0.33 (m, 3H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-3-oxo-4-(2-oxopyrrolidin-1-yl)-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (25)
  • Prepared 25 according to General procedure B (110° C. for 6 hours) (5.73 mg, 12.8% yield) as a white solid. LC-MS (ESI) [M+H]+440.4. 1H NMR (400 MHz, MeOD-d4): δ 7.76 (s, 1H), 4.75-4.61 (m, 2H), 4.10-4.07 (m, 2H), 3.69-3.55 (m, 1H), 2.65 (s, 3H), 2.62 (d, J=8.2 Hz, 2H), 2.34-2.25 (m, 2H), 2.15 (s, 1H), 1.37 (d, J=6.8 Hz, 3H), 1.18-1.10 (m, 1H), 0.72-0.61 (m, 1H), 0.53-0.27 (m, 3H).
  • Preparation of (S)—N-(6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-4-yl)cyclopropanecarboxamide (26)
  • Prepared 26 according to General procedure B (110° C. for 2 hours) (2.11 mg, 4.69% yield) as a yellow solid. LC-MS (ESI) [M+H]+ 440.3. 1H NMR (400 MHz, CDCl3): δ 9.98 (s, 1H), 7.26 (s, 1H), 4.59-4.43 (m, 2H), 3.71-3.67 (m, 1H), 2.76 (s, 3H), 2.67 (m, 3H), 1.37 (d, J=6.8 Hz, 3H), 1.22-1.18 (m, 2H), 1.03-0.96 (m, 4H), 0.71-0.67 (m, 1H), 0.53-0.33 (m, 3H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-methyl-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (27)
  • Figure US20230227446A9-20230720-C00188
  • To a solution of (S)—N-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (40.0 mg, 0.102 mmol) in dioxane (4.00 mL) was added 2,4,6-trimethyl-1,3,5,2,4,6-trioxatriborinane (154 mg, 1.22 mmol), Pd(PPh3)4 (12.0 mg, 0.0102 mmol) and K2CO3 (56.0 mg, 0.409 mmol) at rt. The mixture was stirred at 90° C. under argon atmosphere for 2 hours. The reaction mixture was cooled to rt. The mixture was filtered and the filtrate was concentrated in vacuum to give the residue, which was purified by prep-HPLC (base, NH3H2O) to give the title compound (S)—N-(5-(2-(1-cyclopropylethyl)-4-methyl-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (27) (1.50 mg, yield 3.97%) as a white solid. LC-MS (ESI) [M+H]+=371.0. 1H NMR (400 MHz, MeOD-d4): δ 7.68 (s, 1H), 4.66-4.61 (m, 1H), 3.66-3.62 (m, 1H), 2.82 (s, 3H), 2.61 (s, 3H), 2.23 (s, 3H), 1.37 (d, J=7.8, 3H), 1.15-1.13 (m, 1H), 0.67-0.66 (m, 1H), 0.49-0.42 (m, 2H), 0.41-0.37 (m, 1H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(3,3-dimethylureido)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (28)
  • Figure US20230227446A9-20230720-C00189
  • Step a. A solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (intermediate 6) (2.00 g, 7.38 mmol), bis(2,4-dimethoxybenzyl)amine (7.00 g, 22.1 mmol) and Et3N (1.50 g, 14.8 mmol) in dioxane (20 mL) was heated at 160° C. for 5 h in a sealed tube. The mixture was quenched with aq. NH4Cl (60 mL) and extracted with ethyl acetate (60 mL×3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography eluted with petroleum ether:ethyl acetate (100:1-8:1) to give (S)-6-chloro-2-(1-cyclopropylethyl)-4-(2,4-dimethoxybenzylamino)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (2.00 g, 67.4% yield) as a green solid (one of DMB was removed under reaction condition). 1H NMR (400 MHz, CDCl3): δ 7.30-7.26 (m, 1H), 7.23-7.20 (m, 1H), 6.55 (s, 1H), 6.46-6.40 (m, 2H), 4.64 (d, J=5.6 Hz, 2H), 4.37-4.22 (m, 2H), 3.86 (s, 3H), 3.80 (s, 3H), 3.61-3.57 (m, 1H), 1.28 (d, J=6.8 Hz, 3H), 0.95-0.93 (m, 1H), 0.63-0.59 (m, 1H), 0.45-0.35 (m, 3H).
  • Step b. Prepared (S)—N-(5-(2-(1-cyclopropylethyl)-4-(2,4-dimethoxybenzylamino)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl) according to General procedure C (135° C., 1 h) (1.20 g, 23.0% yield) as a yellow solid. LC-MS (ESI) [M+H]+522.1.
  • Step c. To a suspension of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(2,4-dimethoxybenzylamino)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (1.00 g, 1.92 mmol) in DCM (6 mL) was added TFA (6 mL) at rt. The reaction mixture was stirred at rt overnight. The reaction mixture was concentrated. The residue was diluted with aq. NaHCO3 (40 mL), extracted with ethyl acetate (30 mL×3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give (S)—N-(5-(4-amino-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (600 mg, 84.1% yield) as a yellow solid. LC-MS (ESI) [M+H]+ 372.3.
  • Step d. To a suspension of (S)—N-(5-(4-amino-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (150 mg, 0.404 mmol) in DCM (3 mL) was added triphosgene (48 mg, 0.161 mmol) slowly at 0° C., followed by the addition of Et3N (82 mg, 0.808 mmol) at 0° C. The resulting mixture was stirred at rt for 6 h. After that, to the above mixture was added dimethylamine (0.6 mL, 1.21 mmol, 2.0 M in THF) slowly at 0° C. The reaction mixture was stirred at rt overnight. The mixture was quenched with aq. NH4Cl (10 mL) and extracted with dichloromethane (10 mL×3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC to give (S)—N-(5-(2-(1-cyclopropylethyl)-4-(3,3-dimethylureido)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (28) (7.48 mg FA salt, 4.20% yield) as a white solid. LC-MS (ESI) [M+H]+443.2. 1H NMR (400 MHz, DMSO-d6): δ 12.18 (br s, 1H), 9.10 (s, 1H), 8.45 (s, 0.61H, FA), 7.41 (s, 1H), 4.61 (s, 2H), 3.54-3.50 (m, 1H), 2.98 (s, 6H), 2.64 (s, 3H), 2.15 (s, 3H), 1.28 (d, J=6.8 Hz, 3H), 1.13-1.11 (m, 1H), 0.59-0.58 (m, 1H), 0.43-0.30 (m, 3H).
  • (S)-6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-N,N-dimethyl-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxamide (29), (S)-6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxylic acid (30) and (S)-6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylmethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxamide (31)
  • Figure US20230227446A9-20230720-C00190
  • Step a. To a solution of (S)—N-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (400 mg, 1.02 mmol) in DMF (5.00 mL) was added ethanol (5.00 mL), Cs2CO3 (834 mg, 2.56 mmol), DPPP (42 mg, 0.102 mmol) and Pd(OAc)2 (23 mg, 0.103 mmol) at room temperature. The mixture was purged with a balloon of CO three times. The mixture was stirred at 70° C. for 3 hours under CO (1 atm). The mixture was cooled to rt and diluted with water (100 mL), extracted with ethyl acetate (50.0 mL×3). The combined organic layer was washed with brine, dried over Na2S04, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (DCM/MeOH=20/1) to give (S)-ethyl 6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxylate (100 mg, 22.7% yield) as a yellow solid, LC-MS (ESI) [M+H]+ 429.0, and amide byproduct 29 (30 mg), which was purified with prep-HPLC to give (S)-6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-N,N-dimethyl-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxamide (29) (17.20 mg, 3.92% yield) as a white solid. LC-MS (ESI) [M+H]+ 427.9. 1H NMR (400 MHz, DMSO-d6): δ 12.22 (br s, 1H), 7.93 (s, 1H), 4.68 (s, 2H), 3.56-3.52 (m, 1H), 3.03 (s, 3H), 2.74 (s, 3H), 2.60 (s, 3H), 2.17 (s, 3H), 1.28 (d, J=6.8 Hz, 3H), 1.16-1.09 (m, 1H), 0.60-0.55 (m, 1H), 0.43-0.33 (m, 2H), 0.27-0.21 (m, 1H).
  • Step b. To a solution of (S)-ethyl 6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxylate (100 mg, 0.233 mmol) in methanol (6 mL) was added water (2 mL) and lithium hydroxide monohydrate (49 mg, 1.17 mmol) at room temperature. The mixture was stirred at room temperature for 2 hours. The mixture was diluted with water (20.0 mL) and extracted with ethyl acetate (20.0 mL). The aqueous phase was adjusted pH to 2 with IN hydrochloric acid aqueous solution and extracted with ethyl acetate (20.0 mL×3). The combined organic layers was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give (S)-6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxylic acid (30) (80 mg, 86.0% yield) as a white solid. LC-MS (ESI) [M+H]+401.0
  • Step c. To a solution of (S)-6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxylic acid (30) (40 mg, 0.100 mmol) in dichloromethane (5.00 mL) was added oxalyl dichloride (127 mg, 1.00 mmol) at 0° C. The mixture was stirred at room temperature for 1 hour. The mixture was concentrated under reduced pressure. The residue was dissolved in dichloromethane (5.00 mL) and cooled to 0° C. A solution of NH3/THF (0.100 mL, 0.200 mmol) in dichloromethane (1.00 mL) was added at 0° C. The mixture was stirred at room temperature for 2 hours. The mixture was diluted with water (30.0 mL) and extracted with DCM (20.0 mL×3). The combined organic layers was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (DCM/MeOH=20/1, v/v) to give (S)-6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxamide (31) (10.40 mg, 26.1% yield) as a slight yellow solid. LC-MS (ESI) [M+H]+ 400.1. 1H NMR (400 MHz, DMSO-d6): δ 12.23 (s, 1H), 8.85 (s, 1H), 7.95 (s, 1H), 7.74 (s, 1H), 4.69 (s, 2H), 3.60-3.56 (m, 1H), 2.63 (s, 3H), 2.17 (s, 3H), 1.29 (d, J=6.8 Hz, 3H), 1.16-1.12 (m, 1H), 0.60-0.58 (m, 1H), 0.45-0.38 (m, 2H), 0.26-0.21 (m, 1H).
  • (S)-6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-N-methyl-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridine-4-carboxamide (32)
  • In a similar manner as the preparation of 31 (methylamine tetrahydrofuran solution was used), the title product (32) was synthesized (13.50 mg, 32.7/a yield) as a pale yellow solid. LC-MS (ESI) [M+H]+ 414.1. 1H NMR (400 MHz, CDCl3): δ 10.95 (br s, 1H), 7.65 (s, 1H), 4.70-4.53 (s, 2H), 3.77-3.75 (m, 1H), 3.10 (d, J=4.4 Hz, 3H), 2.81 (s, 3H), 2.34 (s, 3H), 1.40 (d, J=6.8 Hz, 3H), 1.07-1.04 (m, 1H), 0.73-0.70 (m, 1H), 0.54-0.37 (m, 3H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(morpholine-4-carbonyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (33)
  • Figure US20230227446A9-20230720-C00191
  • A mixture of (S)—N-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (100 mg, 0.256 mmol), morpholine (224 mg, 2.56 mmol), Pd(OAc)2 (6 mg, 0.0256 mmol), DPPP (11 mg, 0.0256 mmol) and Cs2CO3 (210 mg, 0.643 mmol) in DMA (2 mL) under a balloon of CO was stirred at 70° C. for 3 h. The mixture was cooled to rt, quenched with water (5 mL) and extracted with EtOAc (8 mL×3). The organic layer was dried over anhydrous Na2S04, filtered and concentrated. The crude was purified by prep-HPLC to give (S)—N-(5-(2-(1-cyclopropylethyl)-4-(morpholine-4-carbonyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (1.9 mg FA salt, 2% yield) as a white solid. LC-MS (ESI) [M+H]+ 470.1. 1H NMR (400 MHz, DMSO-d6): δ 12.25 (s, 1H), 8.36 (s, 0.92H, FA salt), 7.94 (s, 1H), 4.69 (s, 2H), 3.69 (s, 4H), 3.58-3.49 (m, 1H), 3.14 (s, 4H), 2.60 (s, 3H), 2.17 (s, 3H), 1.28 (d, J=6.8 Hz, 3H), 1.19-1.05 (m, 1H), 0.60-0.52 (m, 1H), 0.45-0.18 (m, 3H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-morpholino-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (34)
  • Figure US20230227446A9-20230720-C00192
  • Step a. Prepared (S)-6-chloro-2-(1-cyclopropylethyl)-4-morpholino-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one according to the preparation of 28 (160° C. for 1 hours) (200 mg, 56.0% yield) as a green solid. LC-MS (ESI) [M+H]+ 322.3.
  • Step b. Prepared 34 according to general procedure C (135° C., 1 h) (14.94 mg, 7.26% yield) as a white solid. LC-MS (ESI) [M+H]+442.0. 1H NMR (400 MHz, CDCl3): δ 7.07 (s, 1H), 4.53-4.37 (m, 2H), 3.90 (t, J=4.0 Hz, 4H), 3.81 (t, J=4.0 Hz, 4H), 3.71-3.67 (m, 1H), 2.67 (s, 3H), 2.31 (s, 3H), 1.32 (d, J=6.8 Hz, 3H), 1.00-0.98 (m, 1H), 0.66-0.64 (m, 1H), 0.47-0.35 (m, 3H).
  • Preparation of (R)—N-(5-(2-(1-cyclopropylethyl)-4-morpholino-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (35)
  • Prepared 35 according to general procedure C from intermediate 9 (135° C., 1 h) (20.2 mg, 12.3% yield) as a white solid. LC-MS (ESI) [M+H]+ 442.1. 1H NMR (400 MHz, DMSO-d6): δ 12.10 (s, 1H), 7.26 (s, 1H), 4.53 (s, 2H), 3.77-3.66 (m, 8H), 3.58-3.48 (m, 1H), 2.59 (s, 3H), 2.15 (s, 3H), 1.23 (t, J=8.0 Hz, 3H), 1.15-1.02 (m, 1H), 0.63-0.50 (m, 1H), 0.38-0.33 (m, 2H), 0.22-0.20 (m, 1H).
  • Preparation of (R)—N-(4-methyl-5-(4-morpholino-3-oxo-2-(1,1,1-trifluoropropan-2-yl)-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)thiazol-2-yl)acetamide (36).& (S)—N-(4-methyl-5-(4-morpholino-3-oxo-2-(1,1,1-trifluoropropan-2-yl)-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)thiazol-2-yl)acetamide (37)
  • Figure US20230227446A9-20230720-C00193
  • Step a. Prepared 6-chloro-4-morpholino-2-(1,1,1-trifluoropropan-2-yl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one according to the preparation of 28 (160° C. for 1 hours) (230 mg, 0.660 mmol, 43.1% yield) as a light yellow solid. LC-MS (ESI) [M+H]+349.9. 1H NMR (400 MHz, DMSO-d6): δ 7.08 (s, 1H), 4.99-4.97 (m, 1H), 4.61-4.34 (m, 2H), 3.78-3.67 (m, 8H), 1.43 (d, J=7.2 Hz, 3H).
  • Step b. Prepared racemic mixture of 36 and 37 according to general procedure C (135° C., 1 h) (60.6 mg, 19.6% yield, racemic) as a white solid. The racemic solid was separated with SFC [Separation condition: Instrument: Waters-SFC80; Column: OZ (2.5*25 cm, 10 um); Mobile phase A: Supercritical CO2, Mobile phase B: MeOH (0.1% NH3); A:B=70/30; at 70 ml/min; Detector wavelength: 214 nm; column temperature: 25 centigrade; back pressure: 100 bar] to give First Enantiomer (11.80 mg, 0.0252 mmol, 3.81% yield, first eluent) as a white solid and Second Enantiomer (10.14 mg, 0.0216 mmol, 3.28% yield, second eluent) as a light yellow solid. First Enantiomer: LC-MS (ESI) [M+H]+ 470.0. 1H NMR (400 MHz, DMSO-d6): δ 12.2 (s, 1H), 7.28 (s, 1H), 5.00-4.95 (m, 1H), 4.65-4.39 (m, 2H), 3.74-3.67 (m, 8H), 2.59 (s, 3H), 2.15 (s, 3H), 1.45 (d, J=7.2 Hz, 3H); Second Enantiomer: LC-MS (ESI) [M+H]+ 470.0. 1H NMR (400 MHz, DMSO-d6): h 12.2 (s, 1H), 7.28 (s, 1H), 5.00-4.95 (m, 1H), 4.65-4.39 (m, 2H), 3.74-3.67 (m, 8H), 2.59 (s, 3H), 2.15 (s, 3H), 1.45 (d, J=7.2 Hz, 3H)
  • A mixture of stereoisomers (including, for example, a pair of enantiomers or a mixture of diastereomers) may be separated by any suitable method, including, but not limited to, chiral HPLC. In some embodiments, when a mixture of stereoisomers is separated by HPLC, it is to be appreciated that the resultant individual stereoisomers or mixtures can be assigned sequential labels (e.g., a first enantiomer, and a second enantiomer), the order of which implies the order in which the isomers eluted from the HPLC column. The absolute stereochemistry for a first enantiomer and a second enantiomer may be obtained by known methods.
  • Preparation of (S)—N-(5-(2-(1-cycopropylethyl)-4-(1,1-dioxidothiomorpholino)-3-(xo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (38)
  • Step a. Prepared 38 according to the preparation of 34 (21.2 mg, 10.61%) as a yellow solid. LC-MS (ESI) [M+H]+ 490.1. 1H NMR (400 MHz, CDCl3): δ 7.16 (s, 1H), 4.51-4.44 (m, 2H), 4.38-4.35 (m, 4H), 3.73-3.62 (m, 1H), 3.24 (t, J=4.8 Hz, 4H), 2.63 (s, 3H), 2.28 (s, 3H), 1.33 (d, J=6.8 Hz, 4H), 1.03-0.93 (m, 1H), 0.69-0.58 (m, 1H), 0.51-0.27 (m, 3H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-3-oxo-4-(piperazin-1-yl)-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (39)
  • Figure US20230227446A9-20230720-C00194
  • Step a. A solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (intermediate 6) (300 mg, 1.11 mmol) and tert-butyl piperazine-1-carboxylate (2.06 g, 11.1 mmol) in dioxane (10.0 mL) was stirred at 160° C. for 1 h in sealed tube. The mixture was cooled to rt and concentrated under reduced pressure to give the residue, which was purified by silica gel column chromatography (PE:EA=50:1-1:1, v/v) to give tert-butyl (S)-4-(6-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-4-yl)piperazine-1-carboxylate (370 mg, 79.19% yield) as a green solid. LC-MS (ESI) [M+H]+ 421.0.
  • Step b. According to the general procedure B (135° C., 1 h), tert-butyl (S)-4-(6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-4-yl)piperazine-1-carboxylate was synthesized (300 mg, 80.5% yield) as a yellow gum. LC-MS (ESI) [M+H]+541.0.
  • Step c. To a suspension of tert-butyl (S)-4-(6-(2-acetamido-4-methylthiazol-5-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-4-yl)piperazine-1-carboxylate (43) (300 mg, 0.556 mmol) in DCM (3 mL) was added TFA (1 mL) at rt. The reaction mixture was stirred at rt overnight and then concentrated under reduced pressure. The residue was diluted with aq. NaHCO3 (10 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layers was washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by prep-TLC (DCM:MeOH=10:1, v/v) to give (S)—N-(5-(2-(1-cyclopropylethyl)-3-oxo-4-(piperazin-1-yl)-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (70 mg, 23.4% yield). 10 mg of this material was re-purified by Prep-HPLC to give (S)—N-(5-(2-(1-cyclopropylethyl)-3-oxo-4-(piperazin-1-yl)-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (39) (4.81 mg, free base) as a yellow solid. LC-MS (ESI) [M+H]+ 441.0. 1H NMR (400 MHz, DMSO-d6): δ 7.22 (s, 1H), 4.51 (s, 2H), 3.61 (br s, 4H), 3.55-3.53 (m, 1H), 2.86 (br s, 4H), 2.58 (s, 3H), 2.15 (s, 3H), 1.24 (d, J=6.8 Hz, 3H), 1.08-1.06 (m, 1H), 0.56-0.55 (m, 1H), 0.39-0.21 (m, 3H).
  • (S)—N-(5-(4-(4-acetylpiperazin-1-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (40)
  • Figure US20230227446A9-20230720-C00195
  • To a suspension of (S)—N-(5-(2-(1-cyclopropylethyl)-3-oxo-4-(piperazin-1-yl)-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (39) (20 mg, 0.0455 mmol) and Et3N (12 mg, 0.114 mmol) in DCM (1 mL) was added acetyl chloride (5 mg, 0.0591 mmol) slowly at 0° C. The reaction mixture was stirred at rt for 2 h. The mixture was quenched with methanol (3 mL) and concentrated under reduced pressure. The residue was purified by prep-HPLC to give (S)—N-(5-(4-(4-acetylpiperazin-1-yl)-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (40) (12.69 mg, 57.8% yield) as a yellow solid. LC-MS (ESI) [M+H]+483.2. 1H NMR (400 MHz, DMSO-do): δ 12.11 (s, 1H), 7.27 (s, 1H), 4.53 (s, 2H), 3.69-3.53 (m, 9H), 2.59 (s, 3H), 2.15 (s, 3H), 2.05 (s, 3H), 1.25 (d, J=6.8 Hz, 3H), 1.09-1.07 (m, 1H), 0.58-0.56 (m, 1H), 0.41-0.22 (m, 3H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-4-morpholino-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)thiazol-2-yl)acetamide (41)
  • Figure US20230227446A9-20230720-C00196
  • The title product (41) was synthesized from (S)-6-chloro-2-(1-cyclopropylethyl)-4-morpholino-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one according to the general procedure C (135° C., 1 h) (23.81 mg, 17.9% yield) as a white solid. LC-MS (ESI) [M+H]+ 428.2. 1H NMR (400 MHz, DMSO-d6): δ 8.18 (s, 1H), 7.49 (s, 1H), 4.51 (s, 2H), 3.75-3.68 (m, 8H), 3.52-3.50 (m, 1H), 2.16 (s, 3H), 1.24 (d, J=6.8 Hz, 3H), 1.09-1.07 (m, 1H), 0.56-0.54 (m, 1H), 0.38-0.21 (m, 3H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-4-(2-methoxyethoxy)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (42)
  • According to the general procedure B (110° C., 2 h, 2-methoxyethan-1-ol was utilized), the title product (42) was synthesized (6.00 mg, 11.1% yield, 0.1 FA salt) as a white solid. LC-MS (ESI) [M+H]+ 431.0. 1H NMR (400 MHz, CDCl3): δ 8.21 (s, 0.1H, FA salt), 7.19 (s, 1H), 4.69-4.66 (m, 2H), 4.54-4.38 (m, 2H), 3.88 (t, J=4.8 Hz, 2H), 3.87-3.70 (m, 1H), 3.49 (s, 3H), 2.66 (s, 3H), 2.29 (s, 3H), 1.32 (d, J=7.2 Hz, 3H), 0.99-0.97 (m, 1H), 0.63-0.62 (m, 1H), 0.46-0.38 (m, 3H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-4-(methoxy-d3)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (43)
  • Figure US20230227446A9-20230720-C00197
  • Step a. To methanol-d4 (0.600 mL) was added sodium hydride (80 mg, 2.00 mmol, 60% w/w dispersion in mineral oil) at 0° C. The mixture was stirred at room temperature for 0.5 hour, and then added to a solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (6) (271 mg, 1.00 mmol) in methanol-d4 (1.40 mL) at 0° C. The resulted mixture was stirred at room temperature for 3 hours. The mixture was diluted with water (10.0 mL) and extracted with ethyl acetate (10.0 mL×3). The combined organic layers was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EA=2/1) to give (S)-6-chloro-2-(1-cyclopropylethyl)-4-(methoxy-d3)-1,2-dihydro-3H-pyrrolo[3,4-c]pyridin-3-one (170 mg, 63.0% yield) as a white solid. LC-MS (ESI) [M+H]+270.0. 1H NMR (400 MHz, CDCl3): δ 7.04 (s, 1H), 4.50-4.35 (m, 2H), 3.75-3.67 (m, 1H), 1.31 (d, J=6.8 Hz, 3H), 1.01-0.92 (m, 1H), 0.66-0.60 (m, 1H), 0.46-0.33 (m, 3H).
  • Step b. According to general procedure C (135° C., 1 h), the title product (43) was synthesized (50.07 mg, 23.1% yield) as a white solid. LC-MS (ESI) [M+H]+ 390.1. 1H NMR (400 MHz, CDCl3): δ 7.21 (s, 1H), 4.55-4.40 (m, 2H), 3.79-3.71 (m, 1H), 2.69 (s, 3H), 2.30 (s, 3H), 1.33 (d, J=6.8 Hz, 3H), 1.02-0.94 (m, 1H), 0.65-0.60 (m, 1H), 0.46-0.32 (m, 3H).
  • (S)—N-(5-(4-acetamido-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (44)
  • Figure US20230227446A9-20230720-C00198
  • Step a. A solution of (S)-4,6-dichloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (intermediate 6) (500 mg, 1.11 mmol) and NH3H2O (3 mL) in THF (10 mL) was heated in a sealed tube at 160° C. for 1 h. After being cooled to rt, the mixture was quenched with aq. NH4Cl (20 mL) and extracted with ethyl acetate (50 mL×3). The combined organic layers was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluted with petroleum ether:ethyl acetate (100:1˜ 5:1, v/v) to give (S)-4-amino-6-chloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (356 mg, 1.42 mmol, 76.6% yield) as a pink solid. LC-MS (ESI) [M+H]+ 252.1.
  • Step b. A solution of (S)-4-amino-6-chloro-2-(1-cyclopropylethyl)-1H-pyrrolo[3,4-c]pyridin-3 (2H)-one (130 mg, 0.516 mmol) in Ac2O (1.50 mL) was heated at 130° C. for 3 h. The mixture was cooled to rt and quenched with aq. NaHCO3 (10 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-TLC (petroleum ether:ethyl acetate=2:1, v/v) to give (S)—N-(6-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-4-yl)acetamide (70 mg, 46.2% yield) as a yellow solid. LC-MS (ESI) [M+H]+294.1.
  • Step c. Prepared 44 according to general procedure C (135° C., 1 h) (4.31 mg FA salt, 5.10% yield) as a white solid. LC-MS (ESI) [M+H]+414.4. 1H NMR (400 MHz, CDCl3): δ 9.82 (s, 1H), 8.16 (s, 0.24H, FA), 7.29 (s, 1H), 4.56-4.50 (m, 2H), 3.72-3.68 (m, 1H), 2.74 (s, 3H), 2.56 (s, 3H), 2.31 (s, 3H), 1.36 (d, J=6.8 Hz, 3H), 1.02-1.01 (m, 1H), 0.69-0.66 (m, 1H), 0.50-0.38 (m, 3H).
  • N-(6-(2-acetamido-4-methythiazole-5-yl)-2-((S)-1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-4-yl)-2-cyanocyclopropane-1-carboxamide (45)
  • Figure US20230227446A9-20230720-C00199
  • According to the general procedure B (110° C., 2 h), the title product (45) was synthesized (10.9 mg, 0.0235 mmol, 26.2% yield) as a white solid. LC-MS (ESI) [M+H]+465.1. 1H NMR (400 MHz, DMSO-d6): δ 12.2 (br s, 1H), 10.4 (br s, 1H), 7.67 (s, 1H), 4.65 (s, 2H), 3.56-3.44 (m, 1H), 2.62 (s, 3H), 2.23-2.18 (m, 1H), 2.16 (s, 3H), 1.64-1.61 (m, 1H), 1.50-1.44 (m, 1H), 1.28 (d, J=6.8 Hz, 3H), 1.24-1.21 (m, 1H), 1.17-1.06 (m, 1H), 0.62-0.53 (m, 1H), 0.47-0.34 (m, 2H), 0.26-0.19 (m, 1H).
  • Compounds 46-49 were Prepared According to a Similar Procedure as the Preparation of Compound 34.
  • (S)—N-(5-(2-(1-cyclopropylethyl)-4-(4-hydroxypiperidin-1-yl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (46)
  • LC-MS (ESI) [M+H]+=456.1. 1H NMR (400 MHz, CD3OD-d4): δ 7.20 (s, 1H), 4.56-4.54 (m, 2H), 4.27-4.23 (m, 2H), 3.86-3.77 (m, 1H), 3.61-3.51 (m, 1H), 3.27-3.20 (m, 2H), 2.62 (s, 3H), 2.22 (s, 3H), 1.98-1.96 (m, 2H), 1.67-1.65 (m, 2H), 1.35-1.33 (m, 3H), 1.11-1.10 (m, 1H), 0.66-0.65 (m, 1H), 0.49-0.28 (m, 3H).
  • N-(5-(2-((S)-1-cyclopropylmethyl)-4-(3-hydroxypyrrolidin-1-yl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (47)
  • LC-MS (ESI) [M+H]+=442.1. 1H NMR (400 MHz, CD3OD-d4): δ 8.50 (s, 0.5H, FA), 7.06 (s, 1H), 4.59-4.46 (m, 3H), 4.02-4.01 (m, 2H), 3.85-3.84 (m, 1H), 3.74-3.69 (m, 1H), 3.68-3.60 (m, 1H), 2.62 (s, 3H), 2.22 (s, 3H), 2.08-1.96 (m, 2H), 1.37-1.31 (m, 3H), 1.11-1.10 (m, 1H), 0.66-0.65 (m, 1H), 0.47-0.28 (m, 3H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-4-(3-hydroxyazetidine-1-yl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (48)
  • LC-MS (ESI) [M+H]+=428.1. 1H NMR (400 MHz, CD3OD-d4): δ 7.08 (s, 1H), 4.66-4.53 (m, 5H), 4.10-4.05 (m, 2H), 3.57-3.47 (m, 1H), 2.62 (s, 3H), 2.22 (s, 3H), 1.42-1.30 (m, 3H), 1.11-1.10 (m, 1H), 0.66-0.65 (m, 1H), 0.47-0.28 (m, 3H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-4-(3-(methylsulfonyl)azetidin-1-yl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (49)
  • LC-MS (ESI) [M+H]+490.1. 1H NMR (400 MHz, DMSO-d6): δ12.1 (br s, 1H), 7.18 (s, 1H), 4.54-4.50 (m, 4H), 4.42-4.41 (m, 3H), 3.53-3.48 (m, 1H), 3.05 (s, 3H), 2.59 (s, 3H), 2.15 (s, 3H), 1.24 (d, J=6.8 Hz, 3H), 1.14-1.03 (m, 1H), 0.61-0.51 (m, 1H), 0.44-0.30 (m, 2H), 0.26-0.17 (m, 1H).
  • Compounds 50-52 were Prepared According to a Similar Procedure as the Preparation of Compound 42.
  • (S)—N-(5-(2-(1-cyclopropylethyl)-4-(2-hydroxyethoxy)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (50)
  • LC-MS (ESI) [M+H]+450.1. 1H NMR (400 MHz, MeOD-d4): δ 7.36 (s, 1H), 4.68-4.60 (m, 2H), 3.58-3.50 (m, 1H), 3.46 (s, 3H), 3.31 (m, 3H), 2.22 (s, 3H), 1.36 (d, J=6.8 Hz, 3H), 1.05-0.96 (m, 1H), 0.64-0.58 (m, 1H), 0.43-0.26 (m, 3H).
  • N-(5-(2-((S)-1-cyclopropylethyl)-3-oxo-4-((tetrahydrofuran-3-yl)oxy)-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (51)
  • LC-MS (ESI) [M+H]+ 443.1. 1H NMR (400 MHz, DMSO-d6): δ 7.46 (s, 1H), 5.65-5.54 (m, 1H), 4.56 (s, 2H), 4.08-3.99 (m, 1H), 3.93-3.73 (m, 3H), 3.54-3.43 (m, 1H), 2.60 (s, 3H), 2.39-2.34 (m, 1H), 2.16 (s, 3H), 2.13-1.99 (m, 1H), 1.25 (d, J=6.8 Hz, 3H), 1.08-0.99 (m, 1H), 0.62-0.50 (m, 1H), 0.45-0.31 (m, 2H), 0.28-0.21 (m, 1H).
  • (S)—N-(5-(2-(1-cyclopropylethyl)-4-(oxetan-3-yloxy)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (52)
  • LC-MS (ESI) [M+H]+ 429.0. 1H NMR (400 MHz, DMSO-d6): δ 12.2 (br s, 1H), 7.50 (s, 1H), 5.71-5.64 (m, 1H), 4.95 (t, J=6.8 Hz, 2H), 4.68-4.61 (m, 2H), 4.58 (s, 2H), 3.57-3.47 (m, 1H), 2.58 (s, 3H), 2.16 (s, 3H), 1.26 (d, J=6.8 Hz, 3H), 1.15-1.10 (m, 1H), 0.60-0.50 (m, 1H), 0.44-0.32 (m, 2H), 0.27-0.18 (m, 1H).
  • (S)—N-(4-chloro-5-(2-(1-cyclopropylethyl)-4-morpholino-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)thiazol-2-yl)acetamide (53)
  • Compound 53 was prepared according to general procedure C from N-(4-chlorothiazol-2-yl)acetamide as a yellow solid. LC-MS (ESI) [M+H]+ 462.0. 1H NMR (400 MHz, CDCl3): δ 9.05 (s, 1H), 7.74 (s, 1H), 4.55-4.39 (m, 2H), 3.98-3.68 (m, 8H), 3.67-3.60 (m, 1H), 2.31 (s, 3H), 1.24 (d, J=6.8 Hz, 3H), 0.98-0.96 (m, 1H), 0.70-0.60 (m, 1H), 0.49-0.25 (m, 3H).
  • Preparation of (S)—N-(5-(2-(1-cyclopropylethyl)-4-(morpholinohexyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (54)
  • Figure US20230227446A9-20230720-C00200
  • A mixture of (S)—N-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (100 mg, 0.256 mmol), potassium trifluoro(morpholinomethyl)borate (159 mg, 0.767 mmol), Xphos-G3 (22 mg, 0.0256 mmol) and Cs2CO3 (167 mg, 0.512 mmol) in dioxane (6 mL) under Ar was stirred at 110° C. for 1 h. The mixture was quenched with water (5 mL) and extracted with EtOAc (8 mL×3). The organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The crude was purified by prep-HPLC to give (S)—N-(5-(2-(1-cyclopropylethyl)-4-(morpholinomethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (45 mg, 38.1% yield) as a yellow solid. LC-MS (ESI) [M+H]+456.2. 1H NMR (400 MHz, CDCl3): δ 9.74 (s, 1H), 7.53 (s, 1H), 4.58-4.42 (m, 2H), 4.32-4.21 (m, 2H), 3.77-3.72 (m, 5H), 2.83 (s, 4H), 2.66 (s, 3H), 2.28 (s, 3H), 1.25 (d, J=6.8 Hz, 3H), 1.03-0.98 (m, 1H), 0.69-0.65 (m, 1H), 0.50-0.28 (m, 3H).
  • Preparation of N-(5-(2-((S)-1-cyclopropylethyl)-4-(1-morpholinoethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (55)
  • Figure US20230227446A9-20230720-C00201
  • Step a. A mixture of (S)—N-(5-(4-chloro-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (280 mg, 0.71 mmol), tributyl(1-ethoxyvinyl)stannane (311 mg, 0.86 mmol) and Pd(PPh3)2Cl2 (25 mg, 0.036 mmol) in dioxane (2 mL) was stirred at 100° C. for 8 h under Ar. The reaction mixture was cooled to room temperature, diluted with water (20 mL), extracted with ethyl acetate (20 mL×3). The combined organic layers was washed with brine, dried over Na2S04, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC to give the title compound (55 mg, 19% yield) as an off-white solid. LC-MS (ESI) [M+H]+ 399.1.
  • Step b. A mixture of (S)—N-(5-(4-acetyl-2-(1-cyclopropylethyl)-3-oxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)-4-methylthiazol-2-yl)acetamide (50 mg, 0.12 mmol), morpholine (22 mg, 0.25 mmol) and titanium tetraisopropanolate (71 mg, 0.25 mmol) in DCE (1 mL) was stirred at 50° C. for 1 h. The reaction mixture was cooled to room temperature, and then NaBH(AcO)3 (79 mg, 0.38 mmol) was added. The resulting mixture was stirred at room temperature for 4 h. The mixture was quenched with aq NaHCO3 (10 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layers was washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC to give 55 (8 mg, 14% yield) as an off-white solid. LC-MS (ESI) [M+H]+ 470.2. 1H NMR (400 MHz, DMSO-d6): δ 12.18 (br s, 1H), 8.32 (br s, 1H), 7.76 (s, 1H), 5.18-5.08 (m, 1H), 4.50-4.59 (m, 2H), 3.62-3.53 (m, 5H), 2.66-2.60 (m, 7H), 2.13 (s, 3H), 1.38-1.37 (m, 3H), 1.28-1.26 (m, 3H), 1.11-1.08 (m, 1H), 0.52-0.50 (m, 1H), 0.38-0.36 (m, 3H), 0.23-0.21 (m, 1H).
  • Biological Examples
  • Activity testing was conducted in the Examples below using methods described herein and those well known in the art.
  • Characterization of Compounds
  • This example compares the biological activity for compounds of Formula (I) including (I-a), (I-b), (I-c), and (I-d).
  • Enzymatic activity of different PI3K isoforms was measured to compare the inhibitory potency and selectivity of the compounds provided herein against PI3K isoforms, particularly selectivity against PI3K gamma over delta. Solubility was also measured to access drug-ability of the tested compounds in the aspect of physicochemical properties.
  • Each of these biological experiments are described below.
  • Enzymatic activity of PI3K isoforms: Enzymatic activity of the class I PI3K isoforms in the presence of the compounds of Table 1 was measured using ADP-Glo luminescent assay against PI3Kα, PI3Kα, PI3Kδ and PI3Kγ with ATP concentration at 25 μM. IPI-549 was used as the reference compound. The compounds would be tested from 1 or 10 μM, 3-fold dilution for 10 doses, in duplicate.
  • Compound preparation: Dilute a compound to 100× of the final desired highest inhibitor concentration in reaction by 100% DMSO. Transfer 100 μl of this compound dilution to a well in a 96-well plate and serially dilute it with DMSO for a total of 10 concentrations.
  • Kinase reaction: Prepare a solution of PI3K in 1× kinase buffer at 2-fold the final concentration of each reagent in the assay. Control wells contain 1× kinase buffer without enzyme. Prepare substrate solution of PIP2 substrate and ATP in 1× kinase reaction buffer at 2-fold of the final concentration. Add concentration substrate solution to each well to the final concentration. Cover the assay plate, shake to mix, and incubate at room temperature for 1 hour. Add 5 μl of ADP-Glo reagent to each well to stop the reaction. Mix briefly with a quick spin, shake slowly on the shaker for 120 min. Add 10 ul Kinase Detection Reagent to each well, shake 1 min, equilibrate for 30 min before reading on a luminescence plate reader.
  • Data Analysis: Convert RLU values to percent inhibition values as Percent inhibition=(max−sample RLU)/(max−min)*100. “min”—no enzyme control; “max”—DMSO control. Fit the data in XLFit excel add-in version 5.4.0.8 to obtain IC50 values. Y=Bottom+(Top−Bottom)/(1+(IC50/X){circumflex over ( )}HillSlope). Compounds of the present disclosure, as exemplified in Examples, showed the IC50 values in table 2 and 3.
  • Kinetic solubility test: The stock solutions of test compounds and control compound progesterone were prepared in DMSO at the concentrations of 10 mM. 15 μL of stock solution (10 mM) was placed in order into their proper 96-well rack. 485 μL of PBS pH7.4 was added into each vial of the cap-less Solubility Sample plate. The assay was performed in singlet. Add one stir stick to each vial and seal using a molded PTFE/Silicone plug. Then the solubility sample plate was shaked at 25° C. at 1100 rpm for 2 hours. After completion of the 2 hours, the samples from the Solubility Sample plate were transferred into the filter plate. all the samples were filtered using the Vacuum Manifold. Aliquot of 5 μL was taken from the filtrate followed by addition of 495 μL of a mixture of H2O and acetonitrile containing internal standard (1:1). A certain proportion of ultrapure water was used to dilute the diluent according to the peak shape. The dilution factor was changed according to the solubility values and the LC-MS signal response.
  • Preparation of 3 μM standards (STD): From the 10 mM DMSO STD plate, 6 μL was transferred into the remaining empty plate, and then 194 μL of DMSO was added to that plate to have a STD concentration of 300 μM. From the 300 μM DMSO STD plate, 5 μL was transferred into the remaining empty plate, and then 495 μL of a mixture of H2O and acetonitrile containing internal standard (1:1) was added to that plate to have a final STD concentration of 3 μM. A certain proportion of ultrapure water was used to dilute the diluent according to the peak shape. The concentrations of the standard samples were changed according to the LC-MS signal response.
  • Procedure for sample analysis: The plate was placed into the well plate autosampler. The samples were evaluated by LC-MS/MS analysis.
  • Data analysis: All calculations were carried out using Microsoft Excel.
  • The filtrate was analyzed and quantified against a standard of known concentration using LC coupled with mass spectral peak identification and quantitation. Solubility values of the test compound and control compound were calculated as follows:
  • [ Sample ] = Area ratio sample × INJ VOL STD × DF sample × [ STD ] Area ratio STD × INJ VOL sample
      • Any value of the compounds that was not within the specified limits was rejected and the experiment was repeated. The solubility value is shown in table 2.
  • The claimed compounds demonstrate remarkable selectivity of PI3K gamma over delta, beta and alpha isoforms. In some embodiments, compounds have dramatically improved physicochemical properties such as solubility and pharmacokinetic properties. For an example, several compounds showed >1000 fold selectivity of PI3K gamma over delta; in another example, compound 20 showed >200 fold improvement over a known literature compound AZ-17 in kinetic solubility test at physiologic pH.
  • TABLE 2
    Inhibitory activities (IC50) of compounds 1-55 against P13K
    gamma and delta isoformand solubility in PBS.
    Solubility in PBS
    # PI3K gamma (nM) PI3K delta (nM) at pH 7.4 (μM)
    IPI-549 1.4 65 6.4
    AZ-17** 0.63* 251* <1*
     1 1.4 >1000
     2 3.7 >1000
     3 7.7
     4 4.5
     5 5.9
     6
     7
     8 34 361
     9 22
    10 2.5 359
    11
    12 1.7 2080 211
    13 3.1
    14 4.2
    15 1.9 784 245
    16
    17 4.5
    18 16
    19 1.6 >1000
    20 1.3 1168 240
    21
    22 1.7 >1000 3.
    23 1.0 1209 202
    24 1.5 794 83
    25 3.8
    26 1.4 >1000
    27 29
    28 1.5
    29 2.5
    30 56
    31 2.3
    32 2.2
    33 6.5
    34 1.5 >1000 9.6
    35 1.0 1326 23
    36 2.4 1254 32
    37 9.0
    38 1.9 1552 2.3
    39 3.7
    40 2.0 25
    41 2.3 3.5
    42 1.8 2.6
    43 2.6
    44 0.87 >1000 1.2
    45 0.80
    46 0.70 6.1
    47 0.61
    48 0.84
    49 1.0
    50 1.0
    51 0.65
    52 1.1
    53 1.8
    54 4.6
    55 1.7
    IPI-549 is a PI3Kγ selective inhibitor in phase II clinical trial (Evans, C. et al ACS Med. Chem. Lett, 2016, 7, 862-867).
    **Compound AZ-17 is a known compound for comparison purpose
    (*reported data: Pemberton, N, et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441)
  • TABLE 3
    Inhibitory activities (IC50) of representative compounds
    20, 23, 34, 35 and 38 against PI3K alpha and beta isoforms.
    # PI3K alpha (nM) PI3K beta (nM) PI3K gamma (nM)
    20 3887 >10000 1.3
    73 3333 >10000 1.0
    34 3333 >10000 1.5
    35 4144 >10000 1.0
    38 8421 >10000 1.9
  • Cellular Activity Test
  • This study aims to determine the inhibitory effect of compounds on PI3K gamma and PI3K delta in corresponding cell based assays. PI3K gamma activity was reflected by phosphorylation of AKT in C5a stimulated RAW264.7 cells, and PI3K delta activity was reflected by phosphorylation of AKT in anti-IgM stimulated Raji cells. Phosphorylation of AKT in cells was determined using AlphaLISA technology from PerkinElmer.
  • Raji cell assay: Prepare Passage 11 Raji cell and add 6 μL of 60K cells per well in 384-well plate. Centrifuge at 500 RPM for 30 s and incubate for 2 hours at 37° C., 5% CO2. Add 30 nL compound by Echo and incubate for 30 min at 37° C. Add 2 μL of IgM (4×, 12 μg/mL) per well, centrifuge at 500 RPM for 30 s and incubate for 10 min at 37° C. Add 2 μL of 5× lysis buffer by Multidrop. Shake 10 min on a plate shaker. Add 5 μL acceptor mix provided in the kit. Centrifuge at 1000 RPM for 1 min. Add 5 μL donor mix provided in the kit. Centrifuge at 1000 RPM for 1 min. Then incubate for 2 hours at 25° C., keep the plate in dark. Read AlphaLISA signal on Envision.
  • Raw264.7 cell assay: Prepare Passage 15 Raw264.7 cell and add 6 μL of 30K cells per well in 384-well plate by multidrop. Centrifuge at 500 RPM for 30 s and incubate for 2 hours at 37° C., 5% CO2. Add 30 nL compound by echo and incubate for 30 min at 37° C., 5% CO2. Add 2 μL of C5α (4×, 320 ng/mL) per well by multidrop, centrifuge at 500 RPM for 30 s and incubate for 5 min at 37° C., 5% CO2. Add 2 μL of 5× lysis buffer by multidrop, incubate for 10 min on a plate shaker. Add 5 μL acceptor mix provided in the kit, centrifuge at 1000 RPM for 1 min. Add 5 μL donor mix provided in the kit, centrifuge at 1000 RPM for 1 min and incubate in dark for 2 hours at 25° C. Read AlphaLISA signal on Envision.
  • Data analysis: Fit the cpd IC50 from non-linear regression equation.

  • Y=Bottom+(Top−Bottom)/(1+10{circumflex over ( )}((Log IC50−X)*HillSlope))
  • X: Log of cpd concentration
  • Y: Inhibition
  • TABLE 4
    Cellular Activity for compounds 1-55 (nM)
    PI3K gamma PI3K delta
    # Raw264.7 cell (nM) Raji cell (nM)
    IPI-549 2.1 133
    AZ-47* 4.0 1190
     1 315 >1000
     4 1853
     5 307
     8 115
    10 290
    12 30
    13 22
    14 27
    15 130
    17 118
    19 28
    20 4.3 2163
    22 2.5 2282
    23 2.0 1509
    24 2.2 1423
    25 446
    27 29
    28 65
    29 3243
    32 208
    34 1.6 1964
    35 3.2
    36 3.3
    38 9.3
    39 271
    40 6.3
    44 5.1
    46 3.8
    48 14
    49 38
    50 20
    52 12
    54 17
    55 18
  • Figure US20230227446A9-20230720-C00202
  • Pharmacokinetics Studies: Pharmacokinetics in Dogs
  • Dosing solutions were prepared at 1 mg/mL in 5% DMSO/40% PEG400/55% water. A dosing solution was administered to male beagle dogs (Approximately 9-14 kg, 3 dogs each group) via intravenous (IV) bolus at 1 mg/kg and by oral gavage (P0) at 5 mg/kg. The dosing volume was 1 mL/kg for IV administration and 5 mL % kg for oral gavage. Blood samples (˜0.3 mL each time point) were collected into tubes containing potassium ethylenediaminetetraacetic acid (K2EDTA) as the anticoagulant at 0.033, 0.083, 0.25, 0.5, 1, 2, 4, 8 and 24 hours post dose for IV administration and 0.083, 0.25, 0.5, 1, 2, 4, 8 and 24 hours post dose for PO administration. The blood samples were then centrifuged for 5 minutes in a centrifuge refrigerated at 4° C. The resultant plasma samples were analyzed using LC/MS/MS to determine concentrations of a test compound. Non-compartmental model with WinNonlin (Phoenix™, version 6.1) software was used to calculate pharmacokinetic (PK) parameters. The PK results are listed in Table 5. The pharmacokinetic profiles of the compounds provided herein including oral bioavailability (F), half-life time (Tin) and volume of distribution at steady state (Vss) are much improved over AZ-17. In particular, the oral bioavailability of compound 20 in dog was 107%, in comparison, the oral bioavailability of AZ-17 in dog was reported to be 1.6% (Pemberton, N, et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441).
  • TABLE 5
    PK parameters for Compound 20, Compound 34
    and AZ-17 in dogs#
    Cl (mL/min/kg) T1/2 (h) Vss obs (L/kg) F (%)
    Compound 34* 4,5 9.1 2.7 >68
    Compound 20* 6.1 7.2 2.1 108
    AZ-17** 6.2 1.3 0.7 1.6
    *IV bolus at 1 mg/kg and PO at 5 mg/kg in beagle dogs
    **Reported by Pemberton., N, et al, Journal of Medicinal Chemistry 2018, 61, 5435-5441
  • Pharmacokinetics in Mice
  • A dosing solution of Compound 34 was prepared at 2 mg/mL in 5% DMSO/40% PEG400/55% water. The dosing solution was administered to male CD-1 mice via oral gavage (PO) at 20 mg/kg. Blood and brain tissue samples were collected at 0.5, 2 and 8 hours post-dose. The blood samples were then centrifuged for 5 minutes in a centrifuge refrigerated at 4° C. The brain samples after blood removal were homogenized in water. The plasma and brain homogenate samples were then analyzed using LC/MS/MS to determine concentrations of Compound 34. The results are listed in Table 6. Compound 34 showed good exposure in the brain tissues of mice, demonstrating good penetration of blood-brain.
  • TABLE 6
    Concentrations of Compound 34 in brain tissues and
    plasma following oral gavage Administration of
    Compound 34 in Male CD1 Mouse at 20 mg/kg
    Time (h) Plasma (ng/mL) Brain (ng/g) Ratio (Brain/Plasma )
    0.5 2803 2494  0.890
    2 1205 473 0.392
    8 1367 454 0.332
  • Anti-Tumor Efficacy of the Compounds Described Herein in Syngeneic Mouse Tumor Models
  • A dosing solution of Compound 34 was prepared at 2 mg/mL in 5% DMSO/40% PEG400/55% water. BALB/c mice were inoculated subcutaneously at the fourth mammary pad with 4T1 cells for tumor development. On the next day after tumor cell inoculation, the mice were assigned into 2 groups using stratified randomization with 10 mice in each group based upon their body weight and inoculation order. The treatments of the mice via oral gavage with either the vehicle or Compound 34 at 100 mg/kg were started from the day of randomization. The tumor sizes were measured three times per week during the treatment. Tumor volume was calculated by the formula: length width2/2. As shown in FIG. 1 , treatment of Compound 34 significantly suppressed tumor growth.
  • Efficacy Study of compound 34 on MOG35-55 induced EAE in C57BL6 mice
  • Objective: Experimental autoimmune encephalomyelitis (EAE) is the most common animal model of human Multiple Sclerosis for evaluating the efficacy of treatment strategies. The objective of this study was to evaluate the efficacy of compound 34 on myelin oligodendrocyte glycoprotein (MOG) induced EAE mouse model.
  • Experiment Procedure: Animal model: Mix 10 mL of Complete Freund's adjuvant (CFA) with 50 mg of Mycobacterium tuberculosis H37Ra into 6 mg/mL of final concentration. Synthetic peptide MOG35-55 was dissolved in saline to 4 mg/ml of final concentration. Emulsify MOG35-55 solution in equal volume of the mixed CFA on ice for 1 hour, using a high-speed homogenizer (30000 rpm, IKA Equipment Shanghai Co., Ltd).
  • Forty female C57BL/6 mice at age 6-8 weeks were randomly allocated into 4 groups as below.
  • TABLE 7
    Group n Dosage (mg/kg) Route
    1. Vehicle 10 N/A PO,QD
    2. Reference FTY-720 10  1 PO,QD
    3, Compound 34-20 NEM, 10 20 PO,BID
    4. Compound 34-80 IMPR 10 80 PO,BID
  • Inject 0.1 mL of MOG35-55 emulsion subcutaneously on the shaved back of the mice at three sites: one along the midline of the back between the shoulders, and one on each side of the midline on the lower back. Bordetella pertussis toxin (PTX) (400 ng in 200 μL of PBS) was injected intra-peritoneally on day 0 and 48 hours after immunization.
  • Treatment: Mice in group 3 and 4 were received Compound 34 following the dosing regimen specified. Mice in group 2 were received positive compound FTY-720 as the reference. Mice in group 1 were the vehicle control. The dosing started from disease onset day 10 and continued for 14 days.
  • Assessment: Body weight was measured before immunization. From day 8, body weight was measured daily till the end of the study. EAE clinical score was recorded before immunization. From day 8, score was recorded daily still the end of the study. Scoring system for clinical assessment of EAE:
  • Score Clinical signs
    0 Normal mouse; no overt signs of disease
    1 Limp taila or hind limb weaknessb but not both
    2 Limp taila and hind limb weakness b
    3 Partial hind limb paralysisc
    4 Complete hind limb paralysis d
    5 Moribund state; death by EAE: sacrifice for humane reasons
    aLimp tail: complete flaccidity of the tail, and absence of curling at the tip of the tail when mouse is picked up.
    bHind limb weakness: observed as a waddling gait, the objective sign being that, in walking, mouse's hind limbs fall through the wire cage tops.
    cPartial hind limb paralysis: mouse can no longer use hind limbs to maintain rump posture or walk but can still move one or both limbs to some extent.
    dComplete hind limb paralysis: total loss of movement in hind limbs; mouse drags itself only on its forelimbs.
    Data analysis: Clinical score at baseline, day 8 and during treatment was analyzed with GraphPad Prism software.
  • Result
  • As shown in FIG. 2 , compound 34 treatment at 20 mg/kg and 80 mg/kg BID significantly ameliorated the clinical disability symptom of EAE induced mice with efficacy comparable to 1 mg/kg of fingolimod (FTY-720).

Claims (25)

1. A compound of Formula (I):
Figure US20230227446A9-20230720-C00203
or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein:
A is CH2, CH(C1-6alkyl), O, or S;
Y is CH or N;
Z is CH or N;
W is N, CH, or CX; wherein X is selected from the group consisting of
Figure US20230227446A9-20230720-C00204
NHG, CHG2, COOH, OG, SO2G, SO2NHG, NGSO2G, C1-6alkyl-NGSO2G, NHC(O)G, NHC(O)NG2, C(O)NHG, C(O)NG2, C3-10cycloalkyl, and 3-10 membered heterocyclyl, wherein each 3-10 membered heterocyclyl independently contains 1 or 2 heteroatoms, wherein the 1 or 2 heteroatoms are selected from the group consisting of 0, N, and S, and wherein each C3-10cycloalkyl or 3-10 membered heterocyclyl is independently optionally substituted with one or more G, wherein:
Q1 is C1-6alkyl, wherein the C1-6alkyl is optionally substituted with one or more OH or halo, and
each G is independently selected from the group consisting of H, D, OH, C1-6alkoxy, oxo, NH2, SO2(C1-6alkyl), C(O)—C1-6alkyl, C3-10cycloalkyl, 3-10 membered heterocyclyl, and C1-6alkyl, wherein the C3-10cycloalkyl or C1-6alkyl is independently optionally substituted with one or more D, OH, C1-6alkoxy, CN, N(C1-6alkyl)2, SO2(C1-6alkyl), or halo, or
two G groups, together with the atoms to which they are attached, form a C3-10cycloalkyl or 3-10 membered heterocyclyl,
provided that, when Y and Z are each CH, and W is CX, then X is selected from the group consisting of
Figure US20230227446A9-20230720-C00205
R1 is C1-6alkyl, C3-10cycloalkyl, or C1-6alkyl-C3-10cycloalkyl, wherein the C1-6alkyl, C3-10cycloalkyl, or C1-6alkyl-C3-10cycloalkyl is independently optionally substituted with one or more halo; and
R2 is selected from the group consisting of
Figure US20230227446A9-20230720-C00206
wherein:
L is H, halo, or C1-6alkyl, wherein the C1-6alkyl is optionally substituted with one or more halo, C1-6alkoxy, or OH, and
each Q2 and Q3 is independently C1-6alkyl, C3-10cycloalkyl, or 3-10 membered heterocyclyl, wherein the C1-6alkyl, C3-10cycloalkyl, or 3-10 membered heterocyclyl is independently optionally substituted with one or more halo.
2. The compound of claim 1, wherein at least one of Y, Z and W is N.
3. The compound of claim 1, wherein the compound has the structure of Formula (I-b):
Figure US20230227446A9-20230720-C00207
or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
4. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein R2 is
Figure US20230227446A9-20230720-C00208
5. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein R2 is selected from the group consisting of
Figure US20230227446A9-20230720-C00209
6. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein Q2 is methyl, ethyl, isopropyl, cyclopropyl, or difluoromethyl.
7. The compound of claim 1, wherein the compound has the structure of Formula (I-c):
Figure US20230227446A9-20230720-C00210
or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
8. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein R1 is selected from the group consisting of
Figure US20230227446A9-20230720-C00211
9. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein R1 is selected from the group consisting of
Figure US20230227446A9-20230720-C00212
10. The compound of claim 1, wherein the compound has the structure of Formula (I-d):
Figure US20230227446A9-20230720-C00213
or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
11. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein X is selected from the group consisting of
Figure US20230227446A9-20230720-C00214
Figure US20230227446A9-20230720-C00215
12. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein X is selected from the group consisting of
Figure US20230227446A9-20230720-C00216
13. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein X is selected from the group consisting of NHG, OG, NHSO2G, and C(O)NG2.
14. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein X is selected from the group consisting of OMe, OCD3, NHSO2Me, NHSO2Et, C(O)NH2, C(O)NHMe, and C(O)NMe2.
15. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof, wherein the compound is selected from the group consisting of:
Figure US20230227446A9-20230720-C00217
Figure US20230227446A9-20230720-C00218
Figure US20230227446A9-20230720-C00219
Figure US20230227446A9-20230720-C00220
Figure US20230227446A9-20230720-C00221
Figure US20230227446A9-20230720-C00222
Figure US20230227446A9-20230720-C00223
Figure US20230227446A9-20230720-C00224
Figure US20230227446A9-20230720-C00225
Figure US20230227446A9-20230720-C00226
16. The compound of claim 1, wherein Z is N, A is CH2, Y is CH, and W is CH, or a pharmaceutically acceptable salt, stereoisomer, prodrug, or solvate thereof.
17. The compound of claim 1, wherein the compound has the structure of Formula (I-a):
Figure US20230227446A9-20230720-C00227
or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
18. The compound of claim 1, wherein the compound is selected from Table 1, or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
19. A pharmaceutical composition comprising a compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof, and a pharmaceutically acceptable carrier.
20. A method of selectively inhibiting a growth or a proliferation phosphoinositide 3-kinase gamma (PI3Kγ) comprising contacting the PI3Kγ with a compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
21. A method of treating a disorder of uncontrolled cellular proliferation related to one or more PI3K isoforms, such as PI3K gamma (PI3Kγ), comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
22. A method of treating an autoimmune disease, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
23. A method of treating a cancer, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, solvate, or prodrug thereof.
24. (canceled)
25. (canceled)
US17/602,206 2019-04-10 2020-04-08 Phosphatidylinositol 3-kinase inhibitors Pending US20230227446A9 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/602,206 US20230227446A9 (en) 2019-04-10 2020-04-08 Phosphatidylinositol 3-kinase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201962832133P 2019-04-10 2019-04-10
US201962836659P 2019-04-20 2019-04-20
PCT/US2020/027303 WO2020210379A1 (en) 2019-04-10 2020-04-08 Phosphatidylinositol 3-kinase inhibitors
US17/602,206 US20230227446A9 (en) 2019-04-10 2020-04-08 Phosphatidylinositol 3-kinase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/027303 A-371-Of-International WO2020210379A1 (en) 2019-04-10 2020-04-08 Phosphatidylinositol 3-kinase inhibitors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/702,703 Continuation US11542262B2 (en) 2019-04-10 2022-03-23 Phosphatidylinositol 3-kinase inhibitors

Publications (2)

Publication Number Publication Date
US20220213096A1 US20220213096A1 (en) 2022-07-07
US20230227446A9 true US20230227446A9 (en) 2023-07-20

Family

ID=72751422

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/602,206 Pending US20230227446A9 (en) 2019-04-10 2020-04-08 Phosphatidylinositol 3-kinase inhibitors
US17/702,703 Active US11542262B2 (en) 2019-04-10 2022-03-23 Phosphatidylinositol 3-kinase inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/702,703 Active US11542262B2 (en) 2019-04-10 2022-03-23 Phosphatidylinositol 3-kinase inhibitors

Country Status (8)

Country Link
US (2) US20230227446A9 (en)
EP (1) EP3952870A4 (en)
JP (1) JP2022526854A (en)
KR (1) KR20210150491A (en)
CN (2) CN115677692A (en)
CA (1) CA3134173A1 (en)
TW (1) TW202104223A (en)
WO (1) WO2020210379A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2015231413B2 (en) 2014-03-19 2020-04-23 Infinity Pharmaceuticals, Inc. Heterocyclic compounds for use in the treatment of PI3K-gamma mediated disorders
MX2018003058A (en) 2015-09-14 2018-08-01 Infinity Pharmaceuticals Inc Solid forms of isoquinolinone derivatives, process of making, compositions comprising, and methods of using the same.
CN115677692A (en) 2019-04-10 2023-02-03 南京征祥医药有限公司 Phosphatidylinositol 3-kinase inhibitors
WO2023165551A1 (en) * 2022-03-02 2023-09-07 上海海雁医药科技有限公司 Six-membered aromatic ring-pyrrolidone derivative, and pharmaceutical composition thereof and use thereof
WO2023168378A1 (en) * 2022-03-04 2023-09-07 Synnovation Therapeutics, Inc. Pi3ka inhibitors
WO2024046454A1 (en) * 2022-09-01 2024-03-07 上海海雁医药科技有限公司 Heteroaryl-substituted pyridopyrrolidone derivative, and pharmaceutical composition and use thereof
CN117683036A (en) * 2022-09-09 2024-03-12 上海璎黎药业有限公司 Nitrogen-containing heterocyclic compound, pharmaceutical composition and application thereof
CN116813608B (en) * 2023-06-08 2024-03-22 英矽智能科技(上海)有限公司 Thiazole compound and application thereof

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2010341573B2 (en) 2009-12-22 2016-10-13 Vertex Pharmaceuticals Incorporated Isoindolinone inhibitors of phosphatidylinositol 3-kinase
US20170119786A1 (en) 2011-03-08 2017-05-04 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
WO2014037480A1 (en) 2012-09-10 2014-03-13 F. Hoffmann-La Roche Ag 6-amino acid heteroaryldihydropyrimidines for the treatment and prophylaxis of hepatitis b virus infection
EP3049415B1 (en) * 2013-09-25 2018-07-04 Vertex Pharmaceuticals Incorporated A selective inhibitor of phosphatidylinositol 3-kinase-gamma
BR112016024533A8 (en) * 2014-04-24 2021-03-30 Novartis Ag amino pyrazine derivatives as phosphatidylinositol 3-kinase or salt inhibitors, their use, and pharmaceutical composition and combination
EP3400221B1 (en) * 2016-01-05 2020-08-26 Incyte Corporation Pyrazol / imidazol substituted pyridines as pi3k-gamma inhibitors
US10858355B2 (en) * 2016-03-10 2020-12-08 Astrazeneca Ab Inhibitors of phosphatidylinositol 3-kinase gamma
CN115677692A (en) 2019-04-10 2023-02-03 南京征祥医药有限公司 Phosphatidylinositol 3-kinase inhibitors

Also Published As

Publication number Publication date
US20220220113A1 (en) 2022-07-14
CN114025756A (en) 2022-02-08
US11542262B2 (en) 2023-01-03
EP3952870A4 (en) 2023-03-08
CN114025756B (en) 2022-09-27
CA3134173A1 (en) 2020-10-15
WO2020210379A1 (en) 2020-10-15
KR20210150491A (en) 2021-12-10
JP2022526854A (en) 2022-05-26
EP3952870A1 (en) 2022-02-16
US20220213096A1 (en) 2022-07-07
CN115677692A (en) 2023-02-03
TW202104223A (en) 2021-02-01

Similar Documents

Publication Publication Date Title
US11542262B2 (en) Phosphatidylinositol 3-kinase inhibitors
US11479543B2 (en) Heterocyclic RIP1 kinase inhibitors
US11370764B2 (en) RIP1 inhibitory compounds and methods for making and using the same
US20210069188A1 (en) Pyrazolo[3,4-b]pyrazine shp2 phosphatase inhibitors and methods of use thereof
US11919890B2 (en) RIP1 inhibitory compounds and methods for making and using the same
TWI718758B (en) Azalactam compounds as hpk1 inhibitors
US9580432B2 (en) Fused pyrimidine compound or salt thereof
KR20190038915A (en) Pyridopyrimidinone CDK2 / 4/6 inhibitor
US11564930B2 (en) RIP1 inhibitory compounds and methods for making and using the same
US10065946B2 (en) Benzazole compounds and methods for making and using the compounds
US11578078B2 (en) Heterocyclic RIP1 inhibitory compounds
US20130072481A1 (en) mTOR SELECTIVE KINASE INHIBITORS
US11446301B2 (en) Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
US20230066011A1 (en) Pyrrolopyrimidine derivative, and pharmaceutical composition for preventing or treating protein kinase-related disease comprising same as active ingredient
US11492349B2 (en) IRAK inhibitors and method for making and using
US20220315597A1 (en) Tricyclic compounds and their use
US20230310454A1 (en) Rip1 inhibitory compounds and methods for making and using the same
US20240140931A1 (en) Tricyclic compounds and uses thereof
KR20210149077A (en) Isochromene derivatives as phosphoinositide 3-kinase inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: NANJING ZHENGXIANG PHARMACEUTICALS CO., LTD., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HANGZHOU ZHENGXIANG PHARMACEUTICALS CO., LTD.;REEL/FRAME:057770/0880

Effective date: 20210414

Owner name: HANGZHOU ZHENGXIANG PHARMACEUTICALS CO., LTD., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HAO, XIAOLIN;REEL/FRAME:057770/0842

Effective date: 20210922

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION