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 languages
English (en)
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 Table
    • 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 Table
    • 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.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Transplantation (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
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 (fr) 2019-04-10 2020-04-08 Inhibiteur de la phosphatidylinositol 3-kinase
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 (fr) 2019-04-10 2020-04-08 Inhibiteur de la phosphatidylinositol 3-kinase

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 (fr)
EP (1) EP3952870A4 (fr)
JP (1) JP2022526854A (fr)
KR (1) KR20210150491A (fr)
CN (2) CN115677692A (fr)
CA (1) CA3134173A1 (fr)
TW (1) TW202104223A (fr)
WO (1) WO2020210379A1 (fr)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK3119397T3 (da) 2014-03-19 2022-03-28 Infinity Pharmaceuticals Inc Heterocykliske forbindelser til anvendelse i behandling af PI3K-gamma-medierede lidelser
KR20180058741A (ko) 2015-09-14 2018-06-01 인피니티 파마슈티칼스, 인코포레이티드 이소퀴놀리논의 고체형, 그의 제조 방법, 이를 포함하는 조성물 및 이를 사용하는 방법
KR20210150491A (ko) 2019-04-10 2021-12-10 난징 젱시앙 파마슈티칼스 컴퍼니 리미티드 포스파티딜이노시톨 3-키나제 저해제
WO2023165551A1 (fr) * 2022-03-02 2023-09-07 上海海雁医药科技有限公司 Dérivé à cycle aromatique à six chaînons pyrrolidone, composition pharmaceutique de celui-ci et son utilisation
WO2023168378A1 (fr) * 2022-03-04 2023-09-07 Synnovation Therapeutics, Inc. Inhibiteurs de la pi3ka
WO2024046454A1 (fr) * 2022-09-01 2024-03-07 上海海雁医药科技有限公司 Dérivé de pyridopyrrolidone substituté par hétéroaryle, ainsi que composition pharmaceutique et son utilisation
CN117683036A (zh) * 2022-09-09 2024-03-12 上海璎黎药业有限公司 一种含氮杂环化合物、其药物组合物及应用
CN116813608B (zh) * 2023-06-08 2024-03-22 英矽智能科技(上海)有限公司 噻唑类化合物及其应用

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG10201502073PA (en) 2009-12-22 2015-05-28 Vertex Pharma Isoindolinone inhibitors of phosphatidylinositol 3-kinase
US20170119786A1 (en) 2011-03-08 2017-05-04 3-V Biosciences, Inc. Heterocyclic modulators of lipid synthesis
CA2881322A1 (fr) 2012-09-10 2014-03-13 F. Hoffmann-La Roche Ag Heteroaryldihydropyrimidines d'acide 6-amine pour le traitement et la prophylaxie d'une infection par le virus de l'hepatite b
CA2925601C (fr) 2013-09-25 2022-11-01 Vertex Pharmaceuticals Incorporated Inhibiteur selectif de la phosphatidylinositol-3-kinase-gamma
KR20160141855A (ko) * 2014-04-24 2016-12-09 노파르티스 아게 포스파티딜이노시톨 3-키나제 억제제로서의 아미노 피라진 유도체
US20170190689A1 (en) * 2016-01-05 2017-07-06 Incyte Corporation Pyridine and pyridimine compounds as pi3k-gamma inhibitors
JP6893520B2 (ja) * 2016-03-10 2021-06-23 アストラゼネカ・アクチエボラーグAstrazeneca Aktiebolag ホスファチジルイノシトール3−キナーゼガンマの新規の阻害剤
KR20210150491A (ko) 2019-04-10 2021-12-10 난징 젱시앙 파마슈티칼스 컴퍼니 리미티드 포스파티딜이노시톨 3-키나제 저해제

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
PUBCHEM CID 130421534 Synonym SCHEMBL19347132 (2017); 1 p. *

Also Published As

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

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 (zh) 作為hpk1抑制劑之氮雜內醯胺化合物
US9580432B2 (en) Fused pyrimidine compound or salt thereof
KR20190038915A (ko) 피리도피리미딘온 cdk2/4/6 억제제
US11564930B2 (en) RIP1 inhibitory compounds and methods for making and using the same
US11578078B2 (en) Heterocyclic RIP1 inhibitory compounds
US10065946B2 (en) Benzazole compounds and methods for making and using the 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
US20240140931A1 (en) Tricyclic compounds and uses thereof
US20220315597A1 (en) Tricyclic compounds and their use
US20230310454A1 (en) Rip1 inhibitory compounds and methods for making and using the same
KR20210149077A (ko) 포스포이노시타이드 3-키나아제 억제제로서 아이소크로멘 유도체

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

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

Free format text: NON FINAL ACTION MAILED