WO2022182395A1 - Small molecule inhibitors of stat3 n-terminal domain and methods of use - Google Patents

Small molecule inhibitors of stat3 n-terminal domain and methods of use Download PDF

Info

Publication number
WO2022182395A1
WO2022182395A1 PCT/US2021/058936 US2021058936W WO2022182395A1 WO 2022182395 A1 WO2022182395 A1 WO 2022182395A1 US 2021058936 W US2021058936 W US 2021058936W WO 2022182395 A1 WO2022182395 A1 WO 2022182395A1
Authority
WO
WIPO (PCT)
Prior art keywords
stnd
formula
pharmaceutically acceptable
group
acceptable salt
Prior art date
Application number
PCT/US2021/058936
Other languages
French (fr)
Inventor
Nadya I. Tarasova
Pedro F. ANDRADE BONILLA
Sourav Sinha
Karen M. STEFANISKO
Marc C. Nicklaus
Original Assignee
The United States Of America, As Represented By The Secretary, Department Of Health And Human Services
Oncolinx Pharmaceuticals, Llc
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 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services, Oncolinx Pharmaceuticals, Llc filed Critical The United States Of America, As Represented By The Secretary, Department Of Health And Human Services
Publication of WO2022182395A1 publication Critical patent/WO2022182395A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/121Ketones acyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/136Amines having aromatic rings, e.g. ketamine, nortriptyline having the amino group directly attached to the aromatic ring, e.g. benzeneamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4021-aryl substituted, e.g. piretanide
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41661,3-Diazoles having oxo groups directly attached to the heterocyclic ring, e.g. phenytoin
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4465Non condensed piperidines, e.g. piperocaine only substituted in position 4
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • 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
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4747Quinolines; Isoquinolines spiro-condensed
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • C07D221/06Ring systems of three rings
    • C07D221/16Ring systems of three rings containing carbocyclic rings other than six-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/72Two oxygen atoms, e.g. hydantoin
    • C07D233/80Two oxygen atoms, e.g. hydantoin with hetero atoms or acyl radicals directly attached to ring nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/06Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2
    • C07D311/08Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring
    • C07D311/18Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring substituted otherwise than in position 3 or 7
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • Signal transducer and activator of transcription 3 (STAT3) activity is a marker for many types of cancer and other diseases, such as diabetic nephropathy, skeletal muscle insulin resistance in type 2 diabetes, endometriosis, depression, asthma, colitis, renal fibrosis, inflammatory bowel disease, systemic lupus erythematosus (SLE), Alzheimer’s disease, Huntington’s disease, and autism.
  • STAT3 Signal transducer and activator of transcription 3
  • Inhibitors of the N-terminal domain of STAT3 produce different cellular and molecular effects compared to other known STAT3 inhibitors.
  • inhibition of STAT3 N-terminal domain is considered a promising drug target for the treatment of cancer and infectious diseases.
  • Peptide inhibitors of the domain have been demonstrated to induce immune responses to and clear Mycobacterium tuberculosis in mice and induce expression of proapoptotic genes in cancer cells.
  • peptides can suffer from several drawbacks, including limited modes of administration, low selectivity, short life in circulation, and poor cell permeability.
  • the invention provides a method of treating a disease associated with increased activity of signal transducer and activator of transcription 3 (STAT3) protein comprising administering to a subject in need thereof a compound as described herein, particularly a phenol group-containing small molecule.
  • STAT3 signal transducer and activator of transcription 3
  • the small molecule compounds have been discovered to be inhibitors of STAT3 N-domain, thereby providing therapeutic treatments for a variety of diseases without specialized modes of administration.
  • FIG. 1 is a reaction scheme of preparing STND-25.
  • DIC is N.N' - diisopropylcarbodiimide.
  • FIG. 2 is a reaction scheme of preparing STND-27.
  • DIEA is N.N' - diisopropylethylamine.
  • FIG. 3 is a reaction scheme of preparing STND-28.
  • FIG. 4 is a reaction scheme of preparing STND-29.
  • FIG. 5 is a reaction scheme of preparing STND-30.
  • FIG. 6 is a reaction scheme of preparing STND-31.
  • FIG. 7 is a reaction scheme of preparing STND-33.
  • FIG. 8 is a reaction scheme of preparing STND-34.
  • TMSC1 is chlorotrimethylsilane.
  • FIG. 9 is a reaction scheme of preparing STND-37.
  • FIG. 10 is a reaction scheme of preparing STND-38.
  • FIG. 11 is a reaction scheme of preparing STND-39.
  • FIG. 12 is a reaction scheme of preparing STND-40.
  • FIG. 13 is a reaction scheme of preparing STND-42.
  • FIG. 14 is a reaction scheme of preparing STND-43.
  • FIG. 15 is a reaction scheme of preparing STND-44.
  • FIG. 16 is a reaction scheme of preparing STND-45.
  • FIG. 17 is a reaction scheme of preparing STND-46.
  • FIG. 18 is a reaction scheme of preparing STND-47.
  • FIG. 19 is bar graph of percent secreted embryonic alkaline phosphatase (SEAP) of compounds of aspects of the invention incubated with HEK-BLUE IL-10 reporter cells relative to a control set at 100%. From left to right, the bars represent STND-1 (black), STND-3 (stripe), STND-4 (white), STND-5 (black), STND-6 (stripe), STND-7 (white), STND-8 (black), STND-10 (stripe), STND-11 (white), STND-12 (black), STND-13 (stripe), STND-14 (white), STND-15 (black), STND-17 (stripe), STND-19 (white), STND-20 (black), and the control (last column).
  • SEAP percent secreted embryonic alkaline phosphatase
  • FIG. 20 is a graph showing the inhibition of IL-10 induced STAT3 signaling in HEK-BLUE IL-10 reporter cells versus concentration of STND-9.
  • the data show the cell number, % from untreated ( ⁇ ) and IL-10-induced STAT3 activity, % of untreated (A).
  • compounds exhibited cell toxicity ( ⁇ ).
  • inhibition of STAT3 activity (A) was more significant and did not appear to be toxicity-mediated.
  • FIG. 21 is a dose response curve of Fnorm (Fi/Fo) versus concentration of STND-9.
  • FIG. 22 is graph of DU145 cell numbers after treatment versus concentration of inhibitor for the following compounds: ST3-H2A2 (positive control, STAT3 ND peptide inhibitor) ( ⁇ ), STND-2 ( ⁇ ), STND-8 (X), and STND-9 ( ⁇ ).
  • ST3-H2A2 positive control, STAT3 ND peptide inhibitor
  • STND-2
  • STND-8 X
  • STND-9
  • FIG. 23 is a graph of DU145 cell (solid line) and normal prostate epithelial cells (RWPE-1) (dashed line) cell numbers after treatment versus concentration of inhibitor for the following compounds: STND-9 ( ⁇ ), STND-25 ( ⁇ ), and STND-34 ( ⁇ ).
  • FIG. 24 is a graph of percent KPC95775 cells after treatment versus concentration of inhibitor for the following compounds: STND-9 ( ⁇ ), STND-25 ( ⁇ ), STND-30 (X), STND-31 (*), and STND-32 (A).
  • the invention provides a method of treating a disease associated with increased activity of signal transducer and activator of transcription 3 (STAT3) protein comprising administering to a subject in need thereof a phenol group-containing compound, a compound that is STND-3, STND-4, or STND-19, or a pharmaceutically acceptable salt thereof.
  • STAT3 signal transducer and activator of transcription 3
  • the phenol group-containing compound is a small molecule of formula (I) in which the remainder of the molecule is para to the OH group on the phenyl ring.
  • the residue of the small molecule is bonded at the para position of the phenyl group through a carbon atom (e.g., that of an alkylene, an alkenyl, or a fused ring), a carbonyl, or an amino moiety.
  • the phenol group-containing compound has a core structural residue selected from the group consisting of formula (la) wherein R 1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, provided that the compound of formula (la) is not formula (lb)
  • each of the core structural residues of formulas (la) through (Ih) is bonded to the remainder of the phenol group-containing compound, or a compound or a pharmaceutically acceptable salt thereof that is selected from the group consisting of
  • the core structural residue of formula (la) does not include the following compounds:
  • R 1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido
  • R 2 is selected from the group consisting of wherein
  • R 3 is heterocyclyl
  • R 4 is aryl, heteroaryl, or alkenyl
  • R 5 is haloalkyl or alkenyl
  • R 6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -SC alkenyl
  • R 7 is H or alkyl
  • R 8 is aryl or heteroaryl, each of which is optionally substituted, R 9 is alkyl or haloalkyl,
  • X 3 is NH or O
  • R 2 is selected from the group consisting of
  • Exemplary compounds of formula (la-1) include , 5, , or a pharmaceutically acceptable salt thereof.
  • the compound of formula (la-1) is STND-9
  • STND-25, or STND-34 more preferably STND-25 or STND-34, or any pharmaceutically acceptable salt thereof.
  • the phenol group-containing compound has a core structural residue of formula (lb) that forms formula (Ib-1)
  • R 10 is selected from the group consisting of
  • the phenol group-containing compound has a core structural residue of formula (Ic) that forms formula (Ic-1)
  • the phenol group-containing compound has a core structural residue of formula (Id) that forms formula (Id-1)
  • the phenol group-containing compound has a core structural residue of formula (Ie) that forms formula (Ie-1)
  • X 1 is CH or N
  • X 2 is a bond, CH2, or C(O),
  • R 14 is H or F
  • R 15 is selected from the group consisting of pharmaceutically acceptable salt thereof.
  • Exemplary compounds of formula (Ie-1) include , -18, or a pharmaceutically acceptable salt thereof.
  • the phenol group-containing compound has a core structural residue of formula (If) that forms formula (If-1)
  • R 17 is H, alk l, R 18 is H or alkyl, n is an integer of 1-3, and R 19 is selected from the group consisting of or a pharmaceutically acceptable salt thereof.
  • Exemplary compounds of formula (If-1) include , or a pharmaceutically acceptable salt thereof.
  • the phenol group-containing compound has a core structural residue of formula (Ig) that forms formula (Ig-1) (Ig-1), wherein
  • R 21 is selected from the group consisting of pharmaceutically acceptable salt thereof.
  • Exemplary compounds of formula (Ig-1) include or a pharmaceutically acceptable salt thereof.
  • the phenol group-containing compound has a core structural residue of formula (Ih) that forms formula (Ih-1) (Ih-1), wherein R 22 is selected from the group consisting of -32, or a pharmaceutically acceptable salt thereof.
  • the compound used in the method is selected from the group consisting of -19, or a pharmaceutically acceptable salt thereof.
  • the present invention further provides exemplary compounds of formula (la-1) wherein
  • R 1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido
  • R 2 is selected from the group consisting of
  • R 3 is heterocyclyl
  • R 4 is aryl, heteroaryl, or alkenyl
  • R 5 is haloalkyl or alkenyl
  • R 6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -S02alkenyl,
  • R 7 is H or alkyl
  • R 8 is aryl or heteroaryl, each of which is optionally substituted
  • R 9 is alkyl or haloalkyl
  • X 3 is NH or O
  • X 4 is CH 2, C(O), or NH, and m is an integer of 1-5, or a pharmaceutically acceptable salt thereof provided that the compound of formula (la-1) is not STND-5, STND-8, STND-9, STND-24, STND-25, STND-35, or STND-36, and in some aspects of the invention, the compound of formula (la-1) also is not STND-43, STND-44, or STND-45: , [0052] In some aspects of the invention, the genus of formula (la-1) does not include one or more of the following compounds:
  • the compound of formula (la-1) preferably is -33, or a pharmaceutically acceptable salt thereof.
  • the compound of formula (la-1) is STND-29, STND-33, STND-34, STND-37, STND-38, STND-39, STND-40, STND-41, STND-42,
  • alkyl implies a straight-chain or branched alkyl substituent containing from, for example, from 1 to about 8 carbon atoms, e.g., from about 1 to about 6 carbon atoms, from 1 to about 4 carbon atoms.
  • alkyl group include methyl, ethyl, «-propyl, isopropyl, «-butyl, vec-butyl. isobutyl, tert-butyl, «-pentyl, isopentyl, «-hexyl, and the like.
  • alkyl occurs as part of a group, such as, e.g., in C3-C6 cycloalkylalkyl, hydroxyalkyl, haloalkyl (e.g., monohaloalkyl, dihaloalkyl, and trihaloalkyl), cyanoalkyl, aminoalkyl, alkylamino, dialkylamino, alkylaminoalkyl, dialkylaminoalkyl, alkylcarbonyl (-C(O)alkyl), alkylcarboxy (-C(O)Oalkyl), arylalkyl, heteroarylalkyl, etc.
  • haloalkyl e.g., monohaloalkyl, dihaloalkyl, and trihaloalkyl
  • cyanoalkyl aminoalkyl, alkylamino, dialkylamino, alkylaminoalkyl, dialkylaminoalkyl, dialkylaminoalkyl,
  • the alkyl can be substituted or unsubstituted, as described herein. Even in instances in which the alkyl is an alkylene chain (e.g., -(CH2)n-), the alkyl group can be substituted or unsubstituted.
  • the alkyl is an alkylene chain (e.g., -(CH2)n-)
  • the alkyl group can be substituted or unsubstituted.
  • alkenyl means a linear alkenyl substituent containing from, for example, 2 to about 8 carbon atoms (branched alkenyls are about 3 to about 8 carbons atoms), e.g., from 2 to about 6 carbon atoms (branch alkenyls are about 3 to about 6 carbon atoms), from about 3 to about 5 carbon atoms (branched alkenyls are about 3 to about 6 carbons atoms).
  • the alkenyl group is a C2-C6 alkenyl or C2-C4 alkenyl.
  • alkenyl group examples include ethenyl, allyl, 2-propenyl, 1-butenyl, 2-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 1- hexenyl, and the like.
  • the alkenyl can be substituted or unsubstituted, as described herein.
  • cycloalkyl means a cyclic alkyl moiety containing from, for example, 3 to about 6 carbon atoms or from 5 to about 6 carbon atoms. Examples of such moieties include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
  • the cycloalkyl can be substituted or unsubstituted, as described herein.
  • aryl refers to a mono, bi, or tricyclic carbocycbc ring system having one, two, or three aromatic rings, for example, phenyl, naphthyl, anthracenyl, or biphenyl.
  • aryl refers to an unsubstituted or substituted aromatic carbocycbc moiety, as commonly understood in the art, and includes monocyclic and polycyclic aromatics such as, for example, phenyl, biphenyl, naphthyl, anthracenyl, pyrenyl, and the like.
  • heterocyclyl encompasses both heteroaryl groups and heterocycloalkyl groups, as described herein.
  • heteroaryl refers to aromatic 5- or 6- membered monocyclic groups, 9- or 10-membered bicycbc groups, and 11- to 14-membered tricyclic groups which have at least one heteroatom (O, S, or N) in at least one of the rings.
  • Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom.
  • the fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated.
  • the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen atoms may optionally be quatemized.
  • Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non-aromatic.
  • the heteroaryl group may be attached at any available nitrogen or carbon atom of any ring.
  • heteroaryl groups are pyridinyl, pyridazinyl, pyrimidyl, pyrazinyl, benzimidazolyl, triazinyl, imidazolyl, (1,2,3)- and (l,2,4)-triazolyl, pyrazinyl, tetrazolyl, furyl, pyrrolyl, thienyl (thiophenyl), isothiazolyl, thiazolyl, isoxazolyl, and oxadiazolyl.
  • the heteroaryl can be substituted or unsubstituted, as described herein.
  • heterocycloalkyl means a stable, saturated, or partially unsaturated monocyclic, bicyclic, and spiro ring system containing 3 to 7 ring members of carbon atoms and other atoms selected from the group consisting of nitrogen, sulfur, and/or oxygen.
  • a heterocycloalkyl is a 5-, 6-, or 7-membered monocyclic ring and contains one, two, or three heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur.
  • the heterocycloalkyl may be attached to the parent structure through a carbon atom or through any heteroatom of the heterocycloalkyl that results in a stable structure.
  • heterocycloalkyl rings examples include isoxazolyl, thiazolinyl, imidazolidinyl, piperazinyl, homopiperazinyl, pyrrolyl, pyrrolinyl, pyrazolyl, pyranyl, piperidyl, oxazolyl, and morpholinyl.
  • the heterocycloalkyl can be substituted or unsubstituted, as described herein.
  • the term “hydroxy” refers to the group -OH.
  • cyano refers to the group -CN
  • thiocyano refers to -SCN
  • alkoxy and cycloalkyloxy embrace linear or branched alkyl and cycloalkyl groups, respectively, that are attached to a divalent oxygen.
  • the alkyl and cycloalkyl groups are the same as described herein.
  • halo refers to a halogen selected from fluorine, chlorine, bromine, and iodine.
  • the term “carboxylato” refers to the group -C(0)OH.
  • the term “amino” refers to the group -NH2.
  • alkylamino refers to -NHR, whereas the term “dialkylamino” refers to -NRR'.
  • R and R' are the same or different and each is a substituted or unsubstituted alkyl group, as described herein.
  • the term “amido” refers to the group -C(0)NRR', which R and R' are the same or different and each is hydrogen or a substituted or unsubstituted alkyl group, as described herein.
  • any substituent that is not hydrogen can be an optionally substituted moiety.
  • the substituted moiety typically comprises at least one substituent (e.g., 1, 2, 3, 4, 5, 6, etc.) in any suitable position (e.g., 1-, 2-, 3-, 4-, 5-, or 6-position, etc.).
  • aryl group When an aryl group is substituted with a substituent, e.g., halo, amino, alkyl, OH, alkoxy, and others, the aromatic ring hydrogen is replaced with the substituent and this can take place in any of the available hydrogens, e.g.,
  • substituents for any non-hydrogen substituent include, e.g., halo, alkyl, alkenyl, hydroxy, nitro, cyano, thiocyano, amino, alkylamino, alkoxy, aryloxy, aralkoxy, carboxyl, carboxyalkyl, carboxyalkyloxy, amido, alkylamido, haloalkylamido, aryl, heteroaryl, and heterocycloalkyl, each of which is described herein.
  • the substituent is at least one (e.g., 1 or 2) alkyl, halo, and/or haloalkyl.
  • any of the aspects above whenever a range of the number of atoms in a structure is indicated (e.g., a Ci-12, Ci-8, Ci-6, C1-4, etc.), it is specifically contemplated that any sub-range or individual number of carbon atoms falling within the indicated range also can be used. Thus, for instance, the recitation of a range of 1-8 carbon atoms (e.g., Ci-Cx).
  • 1- 6 carbon atoms e.g., C1-C6), 1-4 carbon atoms (e.g., C1-C4), 1-3 carbon atoms (e.g., C1-C3), or 2-8 carbon atoms (e.g., C2-C8) as used with respect to any chemical group (e.g., alkyl, cycloalkyl, etc.) referenced herein encompasses and specifically describes 1, 2, 3, 4, 5, 6, 7, and/or 8 carbon atoms, as appropriate, as well as any sub-range thereof (e.g., 1-2 carbon atoms, 1-3 carbon atoms, 1-4 carbon atoms, 1-5 carbon atoms, 1-6 carbon atoms, 1-7 carbon atoms, 1-8 carbon atoms, 2-3 carbon atoms, 2-4 carbon atoms, 2-5 carbon atoms, 2-6 carbon atoms, 2-7 carbon atoms, 2-8 carbon atoms, 3-4 carbon atoms, 3-5 carbon atoms, 3-6 carbon atoms, 3
  • n represents the number of methylene repeat units in formula (If).
  • n is an integer of either 1, 2, or 3.
  • the subscript “m” represents the number of methylene repeat units in formula (Ia- 1).
  • the subscript m is an integer from 1-5 (i.e., 1, 2, 3, 4, or 5).
  • salts or “pharmaceutically acceptable salt” is intended to include nontoxic salts synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two.
  • an inorganic acid e.g., hydrochloric acid, sulfuric acid, phosphoric acid, or hydrobromic acid
  • an organic acid e.g., formic acid, oxalic acid, malonic acid, citric acid, fumaric acid, lactic acid, malic acid, succinic acid, tartaric acid, acetic acid, trifluoroacetic acid, gluconic acid, ascorbic acid, methylsulfonic acid, or benzylsulfonic acid
  • an inorganic base e.g., sodium hydroxide, potassium hydroxide, calcium hydroxide, magnesium hydroxide, or ammonium hydroxide
  • an organic base e.g., methylamine, diethylamine, triethylamine, triethanolamine, ethylenediamine, tris(hydroxymethyl)methylamine, guanidine, choline, or cinchonine
  • an amino acid e.g., lysine, arginine, or alanine
  • nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are typical.
  • suitable salts are found in Remington ’s Pharmaceutical Sciences , 18th ed., Mack Publishing Company, Easton, PA, 1990, p. 1445, and Journal of Pharmaceutical Science, 66, 2-19 (1977).
  • they can be a salt of an alkali metal (e.g., sodium or potassium), alkaline earth metal (e.g., calcium), or ammonium of salt.
  • co-administered or “co-administration” used herein refer to simultaneous or sequential administration.
  • the Enamine library of in-stock compounds containing almost 2 million entries was used for initial screens that evaluated the binding pockets and identified early hits (e.g., STND-1 through STND-7). Additional hits came from screening larger Enamine diversity libraries containing 5 and 15 million compounds (e.g., STND-15 through STND-18). The highest scoring hits have been used for identification of potential binders in similarity searches. Search engine GIGA (www.molsoft.com/giga-search.html) was used for screening of the REAL database of 1.2 billion synthesizable compounds from Enamine (enamine.net/bbrary-synthesis/real-compounds/real-database).
  • Quantitative Neighborhoods of Atoms (QNA) methodology was used for screening SAVI (Synthetically Accessible Virtual Inventory).
  • SAVI has been recently generated using CHMTRN (CHeMistry TRaNslator) adaptation for a knowledge-based forward synthesis computer program (Judson et al., J Chem Inf Model, 2020, 60(1), 3336-3341).
  • CHMTRN CHeMistry TRaNslator
  • the latest version of SAVI used in the screens contains 1.75 billion entries and first became publicly available November 11, 2020 (Patel et al., Scientific Data, November 11, 2020, 7(384), 1-14).
  • a pharmaceutical composition comprises at least one STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable excipients described herein for example, vehicles, adjuvants, carriers or diluents, are well-known to those who are skilled in the art and are readily available.
  • the pharmaceutically acceptable carrier is one that is chemically inert to the active compounds and one that has no detrimental side effects or toxicity under the conditions of use.
  • the pharmaceutical compositions can be administered as oral, sublingual, transdermal, subcutaneous, topical, absorption through epithelial or mucocutaneous linings, intravenous, intranasal, intraarterial, intraperitoneal, intramuscular, intratumoral, peritumoral, intraperitoneal, intrathecal, rectal, vaginal, or aerosol formulations.
  • the pharmaceutical composition is administered orally or intravenously.
  • the STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered orally to a subject in need thereof.
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice and include an additive, such as cyclodextrin (e.g., a-, b-, or g-cyclodextrin, hydroxypropyl cyclodextrin) or polyethylene glycol (e.g., PEG400); (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions and gels.
  • diluents such as water, saline
  • Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent.
  • Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and cornstarch.
  • Tablet forms can include one or more of lactose, sucrose, mannitol, com starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers.
  • Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
  • a flavor usually sucrose and acacia or tragacanth
  • pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
  • Formulations suitable for parenteral administration include aqueous and non- aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), or a salt thereof can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, glycerol ketals, such as 2, 2-dimethyl- 1,3- dioxolane-4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers
  • Oils which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, com, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene-polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (3) mixtures thereof.
  • the parenteral formulations typically contain from about 0.5 to about 25% by weight of the inhibitors in solution. Suitable preservatives and buffers can be used in such formulations. In order to minimize or eliminate irritation at the site of injection, such compositions can contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5 to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol.
  • HLB hydrophile-lipophile balance
  • parenteral formulations can be presented in unit-dose or multi dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, for injections, immediately prior to use.
  • sterile liquid carrier for example, water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • the inhibitors can be made into injectable formulations.
  • the requirements for effective pharmaceutical carriers for injectable compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, ⁇ . B. Lippincott Co., Philadelphia, Pa., Banker and Chalmers, eds., pages 238-250 (1982), and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 (1986).
  • Topically applied compositions are generally in the form of liquids (e.g., mouthwash), creams, pastes, lotions, gels, and transdermal patches.
  • Topical administration includes application to the skin and oral mucosa, which includes the oral cavity, oral epithelium, palate, gingival, and the nasal mucosa.
  • the composition contains at least one active component and a suitable vehicle or carrier. It can also contain other components, such as an anti-irritant.
  • the carrier can be a liquid, solid or semi-solid.
  • the composition is an aqueous solution, such as a mouthwash.
  • the composition can be a dispersion, emulsion, gel, lotion, or cream vehicle for the various components.
  • the primary vehicle is water or a biocompatible solvent that is substantially neutral or that has been rendered substantially neutral.
  • the liquid vehicle can include other materials, such as buffers, alcohols, glycols, and mineral oils with various emulsifiers or dispersing agents as known in the art to obtain the desired pH, consistency and viscosity. It is possible that the compositions can be produced as solids, such as powders or granules. The solids can be applied directly or dissolved in water or a biocompatible solvent prior to use to form a solution that is substantially neutral or that has been rendered substantially neutral and that can then be applied to the target site.
  • the vehicle for topical application to the skin can include water, buffered solutions, various alcohols, glycols such as glycerin, lipid materials such as fatty acids, mineral oils, phosphoglycerides, collagen, gelatin, and silicone based materials.
  • the STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation.
  • aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer.
  • the dose administered to the subject, particularly human and other mammals, in accordance with the present invention should be sufficient to affect the desired response.
  • dosage will depend upon a variety of factors, including the age, condition or disease state, predisposition to disease, genetic defect or defects, and body weight of the mammal.
  • the size of the dose will also be determined by the route, timing and frequency of administration as well as the existence, nature, and extent of any adverse side-effects that might accompany the administration of a particular inhibitor and the desired effect. It will be appreciated by one of skill in the art that various conditions or disease states may require prolonged treatment involving multiple administrations.
  • inventive methods comprise administering an effective amount of a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof.
  • An “effective amount” means an amount sufficient to show a meaningful benefit in an individual, cell, or tissue to be treated.
  • a meaningful benefit includes, for example, detectably treating, relieving, healing, preventing, delaying the onset of, ameliorating, or lessening one or more symptoms of a disease mediated by overexpression of STAT3 (e.g., inflammation, fluid accumulation), inhibiting, arresting development, preventing, or halting further development of a viral infection or disease, promoting at least one aspect of tumor cell cytotoxicity (e.g., inhibition of growth, inhibiting survival of a cancer cell, reducing proliferation, reducing size and/or mass of a tumor (e.g., solid tumor)).
  • the meaningful benefit observed in the subject can be to any suitable degree (10, 20, 30, 40, 50, 60, 70, 80, 90% or more or anything in between each of these percentages).
  • one or more symptoms of the disease are treated, prevented, reduced, halted, or eliminated subsequent to administration of a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof, thereby effectively treating the disease (e.g., disease caused by a bacterium or virus, cancer) to at least some degree.
  • a STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof, thereby effectively treating the disease (e.g., disease caused by a bacterium or virus, cancer) to at least some degree.
  • Effective amounts can vary depending upon the biological effect desired in the individual, condition to be treated, and/or the specific characteristics of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof, and the individual.
  • any suitable dose of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered to the subject (e.g., human), according to the type of disease (e.g., disease caused by a bacterium or virus, cancer) to be treated.
  • the dose of the STAT3 inhibitor compound desirably comprises about 0.01 mg per kilogram (kg) of the body weight of the subject (mg/kg) or more (e.g., about 0.05 mg/kg or more, 0.1 mg/kg or more, 0.5 mg/kg or more, 1 mg/kg or more, 2 mg/kg or more, 5 mg/kg or more, 10 mg/kg or more, 15 mg/kg or more, 20 mg/kg or more, 30 mg/kg or more, 40 mg/kg or more, 50 mg/kg or more, 75 mg/kg or more, 100 mg/kg or more, 125 mg/kg or more, 150 mg/kg or more, 175 mg/kg or more, 200 mg/kg or more, 225 mg/kg or more, 250 mg/kg or more, 275 mg/kg or more, 300 mg/kg or more, 325 mg/kg or more, 350 mg/kg or more, 375 mg
  • the dose will be about 500 mg/kg or less (e.g., about 475 mg/kg or less, about 450 mg/kg or less, about 425 mg/kg or less, about 400 mg/kg or less, about 375 mg/kg or less, about 350 mg/kg or less, about 325 mg/kg or less, about 300 mg/kg or less, about 275 mg/kg or less, about 250 mg/kg or less, about 225 mg/kg or less, about 200 mg/kg or less, about 175 mg/kg or less, about 150 mg/kg or less, about 125 mg/kg or less, about 100 mg/kg or less, about 75 mg/kg or less, about 50 mg/kg or less, about 40 mg/kg or less, about 30 mg/kg or less, about 20 mg/kg or less, about 15 mg/kg or less, about 10 mg/kg or less, about 5 mg/kg or less, about 2 mg/kg or less, about 1 mg/kg or less, about 0.5 mg/kg or less, or about 0.1 mg/kg or less, about
  • the term “subject” preferably is directed to a mammal.
  • Mammals include, but are not limited to, the order Rodentia, such as mice, and the order Lagomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order Artiodactyla, including Bovines (cows) and Swines (pigs) or of the order Perissodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Cebids, or Simioids (monkeys) or of the order Anthropoids (humans and apes).
  • STAT3 has been long considered a promising drug target due to its involvement in proliferation of tumor cells, inflammation, and immune responses. See, e.g., Bharadwaj et al., Pharmacol Rev 2020, 72 (2), 486-526; Huynh et al., Nat Rev Cancer 2019, 19 (2), 82-96; Gkouveris et al., Journal of Cancer Therapy , 2015, 6, 709-726; WO 2012/159107, and WO 2010/062681. An aberrant STAT3 pathway has also been shown to play a critical role in the pathogenesis of SARS-CoV-2 (Matsuyama et al., Cell Death Differ, October 9, 2020,
  • N-terminal domain of STAT3 has emerged as a very different target (Hu et al ,Mol Cell Biol, 2015, 35(19), 3284-300) and inhibitors of the domain produce significantly different cellular and molecular effects compared to other STAT3 inhibitors (Timofeeva et al., ACS Chem Biol, 2007, 2(12), 799-809; and Timofeeva et al., Proc Natl Acad Sci USA, 2013, 110(A), 1267-1272).
  • a functional aspect is that inhibitors of STAT3 N-domain impact not only tumor cells proliferation, but also immune responses to tumors and bacterial and viral pathogens.
  • a STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), inhibits and/or reduces STAT3 activity, preferably STAT3 N-domain activity, in a cell, such as a cancer cell.
  • the method includes contacting a cell with a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof, whereby the activity of STAT3, particularly STAT3 N-domain, in the cell is inhibited.
  • the STAT3 activity can be measured by any method, including the assays described herein.
  • STAT3 has been described in the art as a viable treatment of diseases that are characterized as overexpressing STAT3.
  • peptide STAT3 N-domain inhibitors have been shown to up-regulate expression of several proapoptotic proteins, in particular c-Fos (Timofeeva et al., Proc Natl Acad Sci USA, 2013, 110 (4), 1267-1272).
  • Western blot analysis of c-Fos expression has shown that the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), effectively increase c-Fos expression in prostate cancer cells.
  • the STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), can be administered to a subject in need thereof as part of a treatment method for diseases that overexpress STAT3 (e.g., a disease caused by a bacterium or virus, cancer).
  • diseases that overexpress STAT3 e.g., a disease caused by a bacterium or virus, cancer.
  • the disease associated with increased activity of STAT3 protein can be, for example, a bacterial infection, a viral infection, an inflammatory disease, or cancer.
  • the disease can be a bacterial infection (including a disease caused by a bacterial infection) or a viral infection (including a disease caused by a viral infection).
  • the bacterial infection can be caused by, for example, Staphylococcus aureus, gram positive methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus saprophyticus, Pseudomonas aeruginosa, Listeria monocytogenes, Klebsiella pneumoniae, Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus agalactiae, Haemophilus influenzae, Helicobacter pylori, Salmonella, Shigella, Clostridium, Enterobacter aerogenes, gram negative Escherichia coli, Clostridium difficile, or a combination thereof.
  • Diseases caused by a bacterial infection include, e.g., tuberculosis, gastritis, meningitis, gonorrhea, pneumonia, bacterial food poisoning, strep throat, cellulitis, wound infections, and toxic shock syndrome.
  • the bacterial infection is tuberculosis.
  • the viral infection can be, e.g., influenza virus (e.g., H1N1, H1N2, and H5N1), gastroenteritis, hepatitis virus, herpes simplex virus, West Nile virus, Ebola, human papillomavirus, coronavirus, chickenpox virus, flavivirus, togavirus, levivirus, norovirus, rotavirus, adenovirus, parvovirus, poliovirus, or a combination thereof.
  • the coronavirus can be, for example, SARS-CoV, SARS-CoV-2, or MERS-CoV.
  • the disease associated with increased activity of STAT3 protein is SARS-CoV-2.
  • Diseases caused by a viral infection include, e.g., coronavirus disease (COVID-19), severe acute respiratory syndrome (SARS) virus, Middle East respiratory syndrome (MERS), a respiratory disease (e.g., pneumonia, bronchitis, pleural effusion), an inflammatory disease (e.g., inflammation, COVID-19-induced inflammation, pediatric multi-system inflammatory syndrome (PMIS)), reproductive and respiratory syndrome virus (PRRSV), equine arteritis virus (EAV), or gastroenteritis.
  • coronavirus disease COVID-19
  • SARS severe acute respiratory syndrome
  • MERS Middle East respiratory syndrome
  • a respiratory disease e.g., pneumonia, bronchitis, pleural effusion
  • an inflammatory disease e.g., inflammation, COVID-19-induced inflammation, pediatric multi-system inflammatory syndrome (PMIS)
  • PRRSV reproductive and respiratory syndrome virus
  • EAV equine arteritis virus
  • the inhibitor compound described herein is used in conjunction with a known therapy (e.g., an antibiotic, an antiviral agent, etc.).
  • a known therapy e.g., an antibiotic, an antiviral agent, etc.
  • An antibiotic can be from any suitable class, such as a penicillin, a tetracycline, a cephalosporin, a quinolone (also known as a fluoroquinolone), a lincomycin, a macrolide, a sulfonamide, a gly copeptide, an aminoglycoside, and a carbapenem.
  • antibiotics include, for example, amoxicillin, ampicillin, dicloxacilbn, oxacillin, doxycycbne, demeclocycline, ervacycbne, minocycline, omadacycline, tetracycline, cefotaxime, ceftazidime, cefuroxime, cephalexin, ciprofloxacin, moxifloxacin, clindamycin, lincomycin, metronidazole, azithromycin, clarithromycin, erythromycin, sulfamethoxazole and trimethoprim, amoxicillin and clavulanate, levofloxacin, and a combination thereof.
  • An antiviral agent can be from any suitable class, such as a chemokine receptor antagonist, a reverse transcriptase inhibitor, an integrase inhibitor, a protease inhibitor, and a fusion inhibitor.
  • antiviral agents include, for example, acyclovir, oseltamivir, zanamivir, peramvir, baloxavir, lopinavir, ritonavir, ivermectin, remdesivir, valacyclovir, cidofovir, foscamet, ganciclovir, valganciclovir, penciclovir, famciclovir, idoxuridine, trifluorothymidine, vidarabine, fomivirsen, amantadine, rimantadine, ribavirin, lamivudine, adefovir dipivoxil, entecavir, telbivudine, clevudine
  • the STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be co-administered with an antibiotic or antiviral agent.
  • a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered before, concurrently with, or after administration of an antibiotic or antiviral agent.
  • One or more than one, e.g., two, three, or more antibiotic or antiviral agents can be administered.
  • the present invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a combination of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof and at least one antibiotic or antiviral agent.
  • the disease to be treated is an inflammatory disease.
  • an inflammatory disease is that which causes inflammation in at least one tissue.
  • Inflamed tissue includes, e.g., tissue from the gastrointestinal system, pulmonary system, skin, musculature, joints, nervous system, and a combination thereof.
  • Typical inflammatory diseases include, for example, arthritis (including rheumatoid arthritis, osteoarthritis, and psoriatic arthritis), gut inflammation, chronic obstructive pulmonary disease (COPD), asthma, bronchitis, inflammatory bowel disease (IBD), Alzheimer’s disease, Parkinson’s disease, and bursitis.
  • the STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be co-administered with an anti-inflammatory agent.
  • the anti-inflammatory agent can be from any suitable class, such as a nonsteroidal anti-inflammatory drug (NSAID) (e.g., a nonselective COX inhibitor, a selective COX2 inhibitor).
  • NSAID nonsteroidal anti-inflammatory drug
  • anti inflammatory agents include, for example, celecoxib, etoricoxib, valdecoxib, aspirin, diclofenac, ibuprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, diflunisal, naproxen, phenylbutazone, piroxicam, sulindac, mefenamic acid, etodolac, meloxicam, nambumetone, oxaprozin, tolmetin, acetaminophen, duloxetine, and a combination thereof.
  • a STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered before, concurrently with, or after administration of an anti-inflammatory agent.
  • One or more than one, e.g., two, three, or more anti-inflammatory agents can be administered.
  • the present invention is directed to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a combination of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof and at least one anti-inflammatory agent.
  • the disease associated with increased activity of STAT3 protein is cancer.
  • the type of cancer to be treated or prevented is not particularly limited, but in certain aspects, the cancer is characterized as having increased STAT3 activity relative to normal tissue of the same type. See, for example, Garcia et ak, Cell Growth Differ, 1997, 8 (12), 1267-1276; Watson et ak, Br J Cancer, 1995, 71(4), 840-844; Huang et ak, Gynecol Oncol, 2000, 79(1), 67-73; Dhir et ak, Prostate, 2002, 51(4), 241-246; Mora et ak, Cancer Res, 2002, 62(22), 6659-6666; Corvinus et ak, Neoplasia, 2005, 7(6), 545-555; Guo et ak,
  • STAT3 is constitutively active in over 40% of all breast cancers, particularly in triple-negative breast cancers which lack the expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2/Neu) (Banerjee et ak, Int J Cancer, 2016, 138(11), 2570-2578).
  • ER estrogen receptor
  • PR progesterone receptor
  • HER2/Neu human epidermal growth factor receptor 2
  • cancers including cancerous cells and tissue, of the head and neck, eye, skin, mouth, throat, esophagus, chest, bone, lung, colon, sigmoid, rectum, stomach, prostate, breast, ovaries, kidney, liver, pancreas, brain, intestine, heart, or adrenals.
  • cancers include solid tumor, sarcoma, carcinomas, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendothelio sarcoma, synovioma, mesothelioma, Ewing’s tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma,
  • the cancer is breast cancer, prostate cancer, pancreatic cancer, or leukemia.
  • Anti-cancer activity can be measured by any suitable method, including the assays described herein.
  • the STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be co-administered with an anti-cancer agent (e.g., a chemotherapeutic agent) and/or radiation therapy.
  • the method comprises administering an amount of a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof that is effective to sensitize the cancer cells to one or more therapeutic regimens (e.g., chemotherapy or radiation therapy).
  • a STAT3 inhibitor compound including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered before, concurrently with, or after administration of another anti-cancer agent (e.g., a chemotherapeutic agent).
  • another anti-cancer agent e.g., a chemotherapeutic agent
  • One or more than one, e.g., two, three, or more anti-cancer agents can be administered.
  • the present invention is directed to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a combination of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof and at least one anti-cancer agent (e.g., chemotherapeutic agent).
  • anti-cancer agents include platinum compounds (e.g., cisplatin, carboplatin, oxaliplatin), alkylating agents (e.g., cyclophosphamide, ifosfamide, chlorambucil, nitrogen mustard, thiotepa, melphalan, busulfan, procarbazine, streptozocin, temozolomide, dacarbazine, bendamustine), antitumor antibiotics (e.g., daunorubicin, doxorubicin, idarubicin, epirubicin, mitoxantrone, bleomycin, mitomycin C, plicamycin, dactinomycin), taxanes (e.g., pacbtaxel and docetaxel), antimetabolites (e.g., 5-fluorouracil, cytarabine, pemetrexed, thioguanine, floxuridine, capecitabine,
  • a method of treating a disease associated with increased activity of STAT3 protein comprising administering to a subject in need thereof a phenol group- containing compound or a pharmaceutically acceptable salt thereof, wherein the phenol group-containing compound has a core structural residue selected from the group consisting of formula (la)
  • Aspect (2) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (la) that forms formula (la-1) wherein
  • R 1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido
  • R 2 is selected from the group consisting of wherein
  • R 3 is heterocyclyl
  • R 4 is aryl, heteroaryl, or alkenyl
  • R 5 is haloalkyl or alkenyl
  • R 6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -S02alkenyl
  • R 7 is H or alkyl
  • R 8 is aryl or heteroaryl, each of which is optionally substituted
  • R 9 is alkyl or haloalkyl
  • X 3 is NH or O
  • X 4 is CH 2, C(O), or NH, and m is an integer of 1-5, or a pharmaceutically acceptable salt thereof.
  • Aspect (3) The method of aspect (2), wherein R 2 is selected from the group consisting of
  • Aspect (4) The method of aspect (3), wherein the compound of formula (la-1) is selected from the group consisting of STND-5, STND-8, STND-9, STND-24, STND-25, STND-29, STND-33, STND-34, STND-35, STND-36, STND-37, STND-38, STND-39, STND-40, STND-41, STND-42, STND-43, STND-44, STND-45, STND-46, and STND-47, or a pharmaceutically acceptable salt thereof.
  • Aspect (5) The method of aspect (4), wherein the compound of formula (la-1) is selected from the group consisting of STND-9, STND-25, and STND-34, or a pharmaceutically acceptable salt thereof.
  • Aspect (11) The method of aspect (10), wherein the compound of formula (Ie-1) is selected from the group consisting of STND-7, STND-12, STND-16, STND-17, and STND- 18, or a pharmaceutically acceptable salt thereof.
  • Aspect (12) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (If) that forms formula (If-1) wherein each instance of R 17 is H, alkyl, r alkyl, n is 1-3, and R 19 is selected from the group consisting of pharmaceutically acceptable salt thereof.
  • Aspect (13) The method of aspect (12), wherein the compound of formula (If-1) is selected from the group consisting of STND-6, STND-14, STND-15, and STND-20, or a pharmaceutically acceptable salt thereof.
  • Aspect (17) The method of aspect (1), wherein the compound to be administered is selected from the group consisting of STND-3, STND-4, and STND-19, or a pharmaceutically acceptable salt thereof.
  • Aspect (18) The method of any one of aspects (l)-(l 7), wherein the disease associated with increased activity of STAT3 protein is cancer.
  • Aspect (19) The method of aspect (18), wherein the cancer is breast cancer, prostate cancer, pancreatic cancer, or leukemia.
  • Aspect (20) The method of any one of aspects (l)-(l 7), wherein the disease associated with increased activity of STAT3 protein is a bacterial or viral infection.
  • Aspect (21) The method of aspect (20), wherein the bacterial infection is tuberculosis.
  • Aspect (22) The method of aspect (20), wherein the viral infection is by a coronavirus.
  • Aspect (23) The method of any one of aspects (l)-(l 7), wherein the disease associated with increased activity of STAT3 protein is an inflammatory disease.
  • Aspect (24) The method of aspect (23), wherein the inflammatory disease causes inflammation in at least one tissue selected from the group consisting of a gastrointestinal system, a pulmonary system, skin, musculature, joints, a nervous system, and a combination thereof.
  • R 1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido
  • R 2 is selected from the group consisting of
  • R 3 is heterocyclyl
  • R 4 is aryl, heteroaryl, or alkenyl
  • R 5 is haloalkyl or alkenyl
  • R 6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -S02alkenyl,
  • R 7 is H or alkyl
  • R 8 is aryl or heteroaryl, each of which is optionally substituted
  • R 9 is alkyl or haloalkyl
  • X 3 is NH or O
  • X 4 is CH 2, C(O), or NH, and m is an integer of 1-5, or a pharmaceutically acceptable salt thereof, provided that the compound of formula (la-1) is not STND-5, STND-8, STND-9, STND-24, STND-25, STND-35, or STND-36.
  • Aspect (26) The compound of aspect (25) that is selected from the group consisting of STND-29, STND-33, STND-34, STND-37, STND-38, STND-39, STND-40, STND-41, STND-42, STND-43, STND-44, STND-45, STND-46, and STND-47, or a pharmaceutically acceptable salt thereof, preferably STND-29, STND-33, STND-34, STND- 37, STND-38, STND-39, STND-40, STND-41, STND-42, STND-46, or STND-47.
  • Aspect (27) A compound selected from the group consisting of a compound of formula (Ib-1) a compound of formula (Ic-1)
  • R 11 is OH (STND-30) or
  • a pharmaceutical composition comprising a compound of any one of aspects (25)-(27) and a pharmaceutically acceptable carrier.
  • Aspect (29) A method of treating a disease associated with increased activity of signal transducer and activator of transcription 3 (STAT3) protein comprising administering to a subject in need thereof a compound of any one of aspects (25)-(27) or a pharmaceutically acceptable salt thereof.
  • STAT3 signal transducer and activator of transcription 3
  • Exemplary STAT3 N-domain inhibitor compounds can be prepared in accordance with the following reaction schemes.
  • FIG. 1 is a reaction scheme of preparing STND-25.
  • DIC is N,N' - diisopropylcarbodiimide.
  • FIG. 2 is a reaction scheme of preparing STND-27.
  • DIEA is N.N'- diisopropylethylamine.
  • FIG. 3 is a reaction scheme of preparing STND-28.
  • FIG. 4 is a reaction scheme of preparing STND-29.
  • FIG. 5 is a reaction scheme of preparing STND-30.
  • FIG. 6 is a reaction scheme of preparing STND-31.
  • FIG. 7 is a reaction scheme of preparing STND-33.
  • FIG. 8 is a reaction scheme of preparing STND-34.
  • TMSC1 is chlorotrimethylsilane.
  • FIG. 9 is a reaction scheme of preparing STND-37.
  • FIG. 10 is a reaction scheme of preparing STND-38.
  • FIG. 11 is a reaction scheme of preparing STND-39.
  • FIG. 12 is a reaction scheme of preparing STND-40.
  • FIG. 13 is a reaction scheme of preparing STND-42.
  • FIG. 14 is a reaction scheme of preparing STND-43.
  • FIG. 15 is a reaction scheme of preparing STND-44.
  • FIG. 16 is a reaction scheme of preparing STND-45.
  • FIG. 17 is a reaction scheme of preparing STND-46.
  • FIG. 18 is a reaction scheme of preparing STND-47.
  • IC50 was determined in the prostate cancer cell line DU145 after 48 hours of exposure to compounds. Binding to the STAT3 N-domain protein (KD) was determined using microscale thermophoresis (MST). The results are set forth in Table 1. The last column of Table 1 identifies the source of each compound, either from a library or designed. Compounds in the SAVI database were first designed by the inventors prior to uploading to the database.
  • DU145 cells were treated with compounds STND-2 (3 mM), STND-9 (3 mM), and
  • prostate cancer cells (DU145 cell line) were exposed to STND-2, STND-8, and STND-9 for 48 hours, and the number of surviving cells was quantitated using an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) cell viability assay (Sigma- Aldrich, St. Louis, MO).
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • MTT 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • the N-domain peptide inhibitor ST3H2-A2 (Timofeeva et al., Proc Natl Acad Sci USA, 2013, 110(4), 1267-1272) was used as a positive control.
  • the effect of STAT3 N-domain inhibitors of the invention on the growth of prostate cancer is shown in FIG. 22.
  • FIG. 23 shows that the compounds of the invention are selectively toxic to prostate tumor cells (DU145) and show very little toxicity to normal prostate epithelial cells (RWPE-1).
  • STAT3 N-domain inhibitors STND-9, STND-25, STND-30, STND- 31, and STND-32 are shown in FIG. 24. Cells were exposed to compounds for 48 hours, and the number of surviving cells was quantitated using an MTT assay.
  • the cytotoxicity studies demonstrate that the STAT3 N-domain inhibitors of the invention are more than an order of magnitude more potent than previously described peptide inhibitors.
  • the inhibitors of the invention show very similar in vitro effects on STAT3 signaling, and consequently can be used for both tumor growth inhibition and modulation of immune responses to tumors and infectious agents.

Abstract

Disclosed is a method of treating a disease associated with increased activity of signal transducer and activator of transcription 3 (STAT3) protein, such as cancer, comprising administering to a subject in need thereof a STAT3 N-domain inhibitor, such as a phenol group-containing compound as described herein. Also disclosed are compounds of formulas (Ia-1), (Ib-1), (Ic-1), (Id-1), and (Ih-1), as described herein.

Description

SMALL MOLECULE INHIBITORS OF STAT3 N-TERMINAL DOMAIN
AND METHODS OF USE
CROSS-REFERENCE TO A RELATED APPLICATION
[0001] This patent application claims the benefit of U.S. Provisional Patent Application No. 63/154,440, filed February 26, 2021, which is incorporated by reference for all purposes.
STATEMENT REGARDING
FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] This invention was made with Government support under project number Z01BC011306-04 by the National Institutes of Health, National Cancer Institute. The Government has certain rights in the invention.
BACKGROUND OF THE INVENTION
[0003] Signal transducer and activator of transcription 3 (STAT3) activity is a marker for many types of cancer and other diseases, such as diabetic nephropathy, skeletal muscle insulin resistance in type 2 diabetes, endometriosis, depression, asthma, colitis, renal fibrosis, inflammatory bowel disease, systemic lupus erythematosus (SLE), Alzheimer’s disease, Huntington’s disease, and autism. See, e.g., Gkouveris et ak, Journal of Cancer Therapy , 2015; 6: 709-726, WO 2012/159107, and WO 2010/062681. With respect to cancerous cells, aberrant STAT3 is believed to promote tumor cell invasion and metastasis. See, e.g., Yue et ak, Expert Opinion Investig Drugs , 2009; 18(1): 45-56.
[0004] Inhibitors of the N-terminal domain of STAT3 produce different cellular and molecular effects compared to other known STAT3 inhibitors. As a result, inhibition of STAT3 N-terminal domain is considered a promising drug target for the treatment of cancer and infectious diseases. Peptide inhibitors of the domain have been demonstrated to induce immune responses to and clear Mycobacterium tuberculosis in mice and induce expression of proapoptotic genes in cancer cells. However, peptides can suffer from several drawbacks, including limited modes of administration, low selectivity, short life in circulation, and poor cell permeability. [0005] Thus, there remains an unmet need to provide small molecule compounds that inhibit STAT3 N-terminal domain to be used in a method of treating diseases associated with an overexpression of STAT3.
BRIEF SUMMARY OF THE INVENTION
[0006] The invention provides a method of treating a disease associated with increased activity of signal transducer and activator of transcription 3 (STAT3) protein comprising administering to a subject in need thereof a compound as described herein, particularly a phenol group-containing small molecule.
[0007] Also provided are novel compounds of formulas (la-1), (Ib-1), (Ic-1), (Id-1), and (Ih-1), as described herein.
[0008] The small molecule compounds have been discovered to be inhibitors of STAT3 N-domain, thereby providing therapeutic treatments for a variety of diseases without specialized modes of administration.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
[0009] FIG. 1 is a reaction scheme of preparing STND-25. DIC is N.N' - diisopropylcarbodiimide.
[0010] FIG. 2 is a reaction scheme of preparing STND-27. DIEA is N.N' - diisopropylethylamine.
[0011] FIG. 3 is a reaction scheme of preparing STND-28.
[0012] FIG. 4 is a reaction scheme of preparing STND-29.
[0013] FIG. 5 is a reaction scheme of preparing STND-30.
[0014] FIG. 6 is a reaction scheme of preparing STND-31.
[0015] FIG. 7 is a reaction scheme of preparing STND-33.
[0016] FIG. 8 is a reaction scheme of preparing STND-34. TMSC1 is chlorotrimethylsilane.
[0017] FIG. 9 is a reaction scheme of preparing STND-37.
[0018] FIG. 10 is a reaction scheme of preparing STND-38.
[0019] FIG. 11 is a reaction scheme of preparing STND-39.
[0020] FIG. 12 is a reaction scheme of preparing STND-40.
[0021] FIG. 13 is a reaction scheme of preparing STND-42. [0022] FIG. 14 is a reaction scheme of preparing STND-43.
[0023] FIG. 15 is a reaction scheme of preparing STND-44.
[0024] FIG. 16 is a reaction scheme of preparing STND-45.
[0025] FIG. 17 is a reaction scheme of preparing STND-46.
[0026] FIG. 18 is a reaction scheme of preparing STND-47.
[0027] FIG. 19 is bar graph of percent secreted embryonic alkaline phosphatase (SEAP) of compounds of aspects of the invention incubated with HEK-BLUE IL-10 reporter cells relative to a control set at 100%. From left to right, the bars represent STND-1 (black), STND-3 (stripe), STND-4 (white), STND-5 (black), STND-6 (stripe), STND-7 (white), STND-8 (black), STND-10 (stripe), STND-11 (white), STND-12 (black), STND-13 (stripe), STND-14 (white), STND-15 (black), STND-17 (stripe), STND-19 (white), STND-20 (black), and the control (last column).
[0028] FIG. 20 is a graph showing the inhibition of IL-10 induced STAT3 signaling in HEK-BLUE IL-10 reporter cells versus concentration of STND-9. The data show the cell number, % from untreated (¨) and IL-10-induced STAT3 activity, % of untreated (A). As expected, compounds exhibited cell toxicity (¨). However, inhibition of STAT3 activity (A) was more significant and did not appear to be toxicity-mediated.
[0029] FIG. 21 is a dose response curve of Fnorm (Fi/Fo) versus concentration of STND-9.
[0030] FIG. 22 is graph of DU145 cell numbers after treatment versus concentration of inhibitor for the following compounds: ST3-H2A2 (positive control, STAT3 ND peptide inhibitor) (¨), STND-2 (■), STND-8 (X), and STND-9 (·).
[0031] FIG. 23 is a graph of DU145 cell (solid line) and normal prostate epithelial cells (RWPE-1) (dashed line) cell numbers after treatment versus concentration of inhibitor for the following compounds: STND-9 (¨), STND-25 (■), and STND-34 (·).
[0032] FIG. 24 is a graph of percent KPC95775 cells after treatment versus concentration of inhibitor for the following compounds: STND-9 (¨), STND-25 (■), STND-30 (X), STND-31 (*), and STND-32 (A).
DETAILED DESCRIPTION OF THE INVENTION
[0033] In an aspect, the invention provides a method of treating a disease associated with increased activity of signal transducer and activator of transcription 3 (STAT3) protein comprising administering to a subject in need thereof a phenol group-containing compound, a compound that is STND-3, STND-4, or STND-19, or a pharmaceutically acceptable salt thereof. In particular, the phenol group-containing compound is a small molecule of formula (I) in which the remainder of the molecule is para to the OH group on the phenyl ring. The residue of the small molecule is bonded at the para position of the phenyl group through a carbon atom (e.g., that of an alkylene, an alkenyl, or a fused ring), a carbonyl, or an amino moiety. In particular, the phenol group-containing compound has a core structural residue selected from the group consisting of formula (la)
Figure imgf000005_0001
wherein R1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, provided that the compound of formula (la) is not
Figure imgf000005_0002
formula (lb)
(Ie), wherein X1 is CH or N, X2 is a bond, CH2, or C(O), and R14 is H or F, formula (If) wherein each instance of R17 is H, alkyl,
Figure imgf000007_0001
r alkyl, and n is an integer of 1-3, formula (Ig)
Figure imgf000007_0002
(Ig), wherein R20 is CH2 or CH=CH, and formula (Ih)
Figure imgf000007_0003
(Ih), wherein each of the core structural residues of formulas (la) through (Ih) is bonded to the remainder of the phenol group-containing compound, or a compound or a pharmaceutically acceptable salt thereof that is selected from the group consisting of
[0034] In some aspects of the method, the core structural residue of formula (la) does not include the following compounds:
structural residue of formula (la) that forms formula (la-1)
Figure imgf000009_0001
wherein
R1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, and R2 is selected from the group consisting of
Figure imgf000010_0001
wherein
R3 is heterocyclyl,
R4 is aryl, heteroaryl, or alkenyl,
R5 is haloalkyl or alkenyl,
R6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -SC alkenyl, R7 is H or alkyl,
R8 is aryl or heteroaryl, each of which is optionally substituted, R9 is alkyl or haloalkyl,
X3 is NH or O,
X4 is CH 2, C(O), or NH, and m is an integer of 1-5, or a pharmaceutically acceptable salt thereof. [0036] In some aspects of (la-1), R2 is selected from the group consisting of
Figure imgf000011_0001
[0037] Exemplary compounds of formula (la-1) include ,
Figure imgf000014_0001
5, , or a pharmaceutically acceptable salt thereof.
[0038] In a preferred aspect of the method, the compound of formula (la-1) is STND-9,
STND-25, or STND-34, more preferably STND-25 or STND-34, or any pharmaceutically acceptable salt thereof.
[0039] In some aspects of the method, the phenol group-containing compound has a core structural residue of formula (lb) that forms formula (Ib-1)
Figure imgf000017_0001
(Ib-1),
wherein R10 is selected from the group consisting of
Figure imgf000018_0002
(STND-28), or a pharmaceutically acceptable salt thereof.
[0040] In some aspects of the method, the phenol group-containing compound has a core structural residue of formula (Ic) that forms formula (Ic-1)
Figure imgf000018_0001
(Ic-1), wherein R11 is OH (STND-30) or
Figure imgf000019_0001
31) or a pharmaceutically acceptable salt thereof.
[0041] In some aspects of the method, the phenol group-containing compound has a core structural residue of formula (Id) that forms formula (Id-1)
Figure imgf000019_0002
(Id-1), wherein
Figure imgf000019_0003
Figure imgf000019_0004
wherein R13 is alkyl, or a pharmaceutically acceptable salt thereof. [0042] In some aspects of the method, methyl (STND-1) or ethyl (STND-22), or a pharmaceutically acceptable salt thereof.
[0043] In some aspects of the method, the phenol group-containing compound has a core structural residue of formula (Ie) that forms formula (Ie-1)
Figure imgf000020_0001
(Ie-1), wherein
X1 is CH or N,
X2 is a bond, CH2, or C(O),
R14 is H or F, and
R15 is selected from the group consisting of
Figure imgf000020_0002
pharmaceutically acceptable salt thereof.
[0044] Exemplary compounds of formula (Ie-1) include ,
Figure imgf000022_0001
-18, or a pharmaceutically acceptable salt thereof.
[0045] In some aspects of the method, the phenol group-containing compound has a core structural residue of formula (If) that forms formula (If-1)
Figure imgf000022_0002
(if-i), wherein each instance of R17 is H, alk l,
Figure imgf000022_0003
R18 is H or alkyl, n is an integer of 1-3, and R19 is selected from the group consisting of
Figure imgf000022_0004
or a pharmaceutically acceptable salt thereof.
[0046] Exemplary compounds of formula (If-1) include , or a pharmaceutically acceptable salt thereof.
[0047] In some aspects of the method, the phenol group-containing compound has a core structural residue of formula (Ig) that forms formula (Ig-1)
Figure imgf000023_0001
(Ig-1), wherein
R20 is CH 2 or CH=CH, and
R21 is selected from the group consisting of
Figure imgf000024_0001
pharmaceutically acceptable salt thereof.
[0048] Exemplary compounds of formula (Ig-1) include
Figure imgf000024_0002
or a pharmaceutically acceptable salt thereof.
[0049] In some aspects of the method, the phenol group-containing compound has a core structural residue of formula (Ih) that forms formula (Ih-1) (Ih-1), wherein R22 is selected from the group consisting of
Figure imgf000025_0001
-32, or a pharmaceutically acceptable salt thereof.
[0050] In some aspects, the compound used in the method is selected from the group consisting of
Figure imgf000025_0002
-19, or a pharmaceutically acceptable salt thereof.
[0051] In an aspect, the present invention further provides exemplary compounds of formula (la-1)
Figure imgf000026_0001
wherein
R1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, and R2 is selected from the group consisting of
wherein
R3 is heterocyclyl,
R4 is aryl, heteroaryl, or alkenyl,
R5 is haloalkyl or alkenyl,
R6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -S02alkenyl,
R7 is H or alkyl,
R8 is aryl or heteroaryl, each of which is optionally substituted,
R9 is alkyl or haloalkyl,
X3 is NH or O,
X4 is CH 2, C(O), or NH, and m is an integer of 1-5, or a pharmaceutically acceptable salt thereof provided that the compound of formula (la-1) is not STND-5, STND-8, STND-9, STND-24, STND-25, STND-35, or STND-36, and in some aspects of the invention, the compound of formula (la-1) also is not STND-43, STND-44, or STND-45: , [0052] In some aspects of the invention, the genus of formula (la-1) does not include one or more of the following compounds:
Figure imgf000030_0001
Figure imgf000031_0001
[0053] In an aspect, the compound of formula (la-1) preferably is
Figure imgf000032_0001
-33,
Figure imgf000033_0001
or a pharmaceutically acceptable salt thereof.
[0054] In some aspects of the invention, the compound of formula (la-1) is STND-29, STND-33, STND-34, STND-37, STND-38, STND-39, STND-40, STND-41, STND-42,
STND-46, or STND-47, or any pharmaceutically acceptable salt thereof.
[0055] Further provided is a compound selected from the group consisting of a compound of formula (Ib-1)
Figure imgf000035_0001
(Ib-1),
a compound of formula (Ic-1)
Figure imgf000036_0001
(STND-31), a compound of formula (Id-1) (Id-1), wherein
Figure imgf000037_0001
a compound of formula (Ih-1)
Figure imgf000037_0002
wherein
Figure imgf000037_0003
or a pharmaceutically acceptable salt thereof.
[0056] In any of the aspects above, the term “alkyl” implies a straight-chain or branched alkyl substituent containing from, for example, from 1 to about 8 carbon atoms, e.g., from about 1 to about 6 carbon atoms, from 1 to about 4 carbon atoms. Examples of alkyl group include methyl, ethyl, «-propyl, isopropyl, «-butyl, vec-butyl. isobutyl, tert-butyl, «-pentyl, isopentyl, «-hexyl, and the like. This definition also applies wherever “alkyl” occurs as part of a group, such as, e.g., in C3-C6 cycloalkylalkyl, hydroxyalkyl, haloalkyl (e.g., monohaloalkyl, dihaloalkyl, and trihaloalkyl), cyanoalkyl, aminoalkyl, alkylamino, dialkylamino, alkylaminoalkyl, dialkylaminoalkyl, alkylcarbonyl (-C(O)alkyl), alkylcarboxy (-C(O)Oalkyl), arylalkyl, heteroarylalkyl, etc. The alkyl can be substituted or unsubstituted, as described herein. Even in instances in which the alkyl is an alkylene chain (e.g., -(CH2)n-), the alkyl group can be substituted or unsubstituted.
[0057] In any of the aspects above, the term “alkenyl,” as used herein, means a linear alkenyl substituent containing from, for example, 2 to about 8 carbon atoms (branched alkenyls are about 3 to about 8 carbons atoms), e.g., from 2 to about 6 carbon atoms (branch alkenyls are about 3 to about 6 carbon atoms), from about 3 to about 5 carbon atoms (branched alkenyls are about 3 to about 6 carbons atoms). In accordance with an aspect, the alkenyl group is a C2-C6 alkenyl or C2-C4 alkenyl. Examples of alkenyl group include ethenyl, allyl, 2-propenyl, 1-butenyl, 2-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 1- hexenyl, and the like. The alkenyl can be substituted or unsubstituted, as described herein. [0058] In any of the aspects above, the term “cycloalkyl,” as used herein, means a cyclic alkyl moiety containing from, for example, 3 to about 6 carbon atoms or from 5 to about 6 carbon atoms. Examples of such moieties include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. The cycloalkyl can be substituted or unsubstituted, as described herein.
[0059] In any of the aspects above, the term “aryl” refers to a mono, bi, or tricyclic carbocycbc ring system having one, two, or three aromatic rings, for example, phenyl, naphthyl, anthracenyl, or biphenyl. The term “aryl” refers to an unsubstituted or substituted aromatic carbocycbc moiety, as commonly understood in the art, and includes monocyclic and polycyclic aromatics such as, for example, phenyl, biphenyl, naphthyl, anthracenyl, pyrenyl, and the like. An aryl moiety generally contains from, for example, 6 to about 30 carbon atoms, from 6 to about 18 carbon atoms, from 6 to about 14 carbon atoms, or from 6 to about 10 carbon atoms. It is understood that the term aryl includes carbocycbc moieties that are planar and comprise 4n+2 p electrons, according to HiickeTs Rule, wherein n = 1, 2, or 3. This definition also applies wherever “aryl” occurs as part of a group, such as, e.g., in haloaryl (e.g., monohaloaryl, dihaloaryl, and trihaloaryl), arylalkyl, etc. The aryl can be substituted or unsubstituted, as described herein.
[0060] In any of the aspects above, the term “heterocyclyl” encompasses both heteroaryl groups and heterocycloalkyl groups, as described herein.
[0061] In any of the aspects above, the term “heteroaryl” refers to aromatic 5- or 6- membered monocyclic groups, 9- or 10-membered bicycbc groups, and 11- to 14-membered tricyclic groups which have at least one heteroatom (O, S, or N) in at least one of the rings. Each ring of the heteroaryl group containing a heteroatom can contain one or two oxygen or sulfur atoms and/or from one to four nitrogen atoms provided that the total number of heteroatoms in each ring is four or less and each ring has at least one carbon atom. The fused rings completing the bicyclic and tricyclic groups may contain only carbon atoms and may be saturated, partially saturated, or unsaturated. The nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen atoms may optionally be quatemized. Heteroaryl groups which are bicyclic or tricyclic must include at least one fully aromatic ring but the other fused ring or rings may be aromatic or non-aromatic. The heteroaryl group may be attached at any available nitrogen or carbon atom of any ring. Illustrative examples of heteroaryl groups are pyridinyl, pyridazinyl, pyrimidyl, pyrazinyl, benzimidazolyl, triazinyl, imidazolyl, (1,2,3)- and (l,2,4)-triazolyl, pyrazinyl, tetrazolyl, furyl, pyrrolyl, thienyl (thiophenyl), isothiazolyl, thiazolyl, isoxazolyl, and oxadiazolyl. The heteroaryl can be substituted or unsubstituted, as described herein.
[0062] The term “heterocycloalkyl” means a stable, saturated, or partially unsaturated monocyclic, bicyclic, and spiro ring system containing 3 to 7 ring members of carbon atoms and other atoms selected from the group consisting of nitrogen, sulfur, and/or oxygen. In an aspect, a heterocycloalkyl is a 5-, 6-, or 7-membered monocyclic ring and contains one, two, or three heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur. The heterocycloalkyl may be attached to the parent structure through a carbon atom or through any heteroatom of the heterocycloalkyl that results in a stable structure. Examples of such heterocycloalkyl rings are isoxazolyl, thiazolinyl, imidazolidinyl, piperazinyl, homopiperazinyl, pyrrolyl, pyrrolinyl, pyrazolyl, pyranyl, piperidyl, oxazolyl, and morpholinyl. The heterocycloalkyl can be substituted or unsubstituted, as described herein. [0063] In any of the aspects above, the term “hydroxy” refers to the group -OH.
[0064] In any of the aspects above, the term “cyano” refers to the group -CN, whereas the term “thiocyano” refers to -SCN.
[0065] In any of the aspects above, the terms “alkoxy” and “cycloalkyloxy” embrace linear or branched alkyl and cycloalkyl groups, respectively, that are attached to a divalent oxygen. The alkyl and cycloalkyl groups are the same as described herein.
[0066] In any of the aspects above, the term “halo” refers to a halogen selected from fluorine, chlorine, bromine, and iodine.
[0067] In any of the aspects above, the term “carboxylato” refers to the group -C(0)OH. [0068] In any of the aspects above, the term “amino” refers to the group -NH2. The term “alkylamino” refers to -NHR, whereas the term “dialkylamino” refers to -NRR'. R and R' are the same or different and each is a substituted or unsubstituted alkyl group, as described herein.
[0069] In any of the aspects above, the term “amido” refers to the group -C(0)NRR', which R and R' are the same or different and each is hydrogen or a substituted or unsubstituted alkyl group, as described herein.
[0070] In other aspects, any substituent that is not hydrogen (e.g., alkyl, alkenyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, or heterocycloalkylalkyl) can be an optionally substituted moiety. The substituted moiety typically comprises at least one substituent (e.g., 1, 2, 3, 4, 5, 6, etc.) in any suitable position (e.g., 1-, 2-, 3-, 4-, 5-, or 6-position, etc.). When an aryl group is substituted with a substituent, e.g., halo, amino, alkyl, OH, alkoxy, and others, the aromatic ring hydrogen is replaced with the substituent and this can take place in any of the available hydrogens, e.g.,
2-, 3-, 4-, 5-, and/or 6-position wherein the 1-position is the point of attachment of the aryl group in the compound of the present invention. Suitable substituents for any non-hydrogen substituent include, e.g., halo, alkyl, alkenyl, hydroxy, nitro, cyano, thiocyano, amino, alkylamino, alkoxy, aryloxy, aralkoxy, carboxyl, carboxyalkyl, carboxyalkyloxy, amido, alkylamido, haloalkylamido, aryl, heteroaryl, and heterocycloalkyl, each of which is described herein. In some instances, the substituent is at least one (e.g., 1 or 2) alkyl, halo, and/or haloalkyl.
[0071] In any of the aspects above, whenever a range of the number of atoms in a structure is indicated (e.g., a Ci-12, Ci-8, Ci-6, C1-4, etc.), it is specifically contemplated that any sub-range or individual number of carbon atoms falling within the indicated range also can be used. Thus, for instance, the recitation of a range of 1-8 carbon atoms (e.g., Ci-Cx). 1- 6 carbon atoms (e.g., C1-C6), 1-4 carbon atoms (e.g., C1-C4), 1-3 carbon atoms (e.g., C1-C3), or 2-8 carbon atoms (e.g., C2-C8) as used with respect to any chemical group (e.g., alkyl, cycloalkyl, etc.) referenced herein encompasses and specifically describes 1, 2, 3, 4, 5, 6, 7, and/or 8 carbon atoms, as appropriate, as well as any sub-range thereof (e.g., 1-2 carbon atoms, 1-3 carbon atoms, 1-4 carbon atoms, 1-5 carbon atoms, 1-6 carbon atoms, 1-7 carbon atoms, 1-8 carbon atoms, 2-3 carbon atoms, 2-4 carbon atoms, 2-5 carbon atoms, 2-6 carbon atoms, 2-7 carbon atoms, 2-8 carbon atoms, 3-4 carbon atoms, 3-5 carbon atoms, 3-6 carbon atoms, 3-7 carbon atoms, 3-8 carbon atoms, 4-5 carbon atoms, 4-6 carbon atoms, 4-7 carbon atoms, 4-8 carbon atoms, etc., as appropriate).
[0072] The subscript “n” represents the number of methylene repeat units in formula (If). The subscript n is an integer of either 1, 2, or 3.
[0073] The subscript “m” represents the number of methylene repeat units in formula (Ia- 1). The subscript m is an integer from 1-5 (i.e., 1, 2, 3, 4, or 5).
[0074] In any of the aspects above, the phrase “salt” or “pharmaceutically acceptable salt” is intended to include nontoxic salts synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. For example, an inorganic acid (e.g., hydrochloric acid, sulfuric acid, phosphoric acid, or hydrobromic acid), an organic acid (e.g., formic acid, oxalic acid, malonic acid, citric acid, fumaric acid, lactic acid, malic acid, succinic acid, tartaric acid, acetic acid, trifluoroacetic acid, gluconic acid, ascorbic acid, methylsulfonic acid, or benzylsulfonic acid), an inorganic base (e.g., sodium hydroxide, potassium hydroxide, calcium hydroxide, magnesium hydroxide, or ammonium hydroxide), an organic base (e.g., methylamine, diethylamine, triethylamine, triethanolamine, ethylenediamine, tris(hydroxymethyl)methylamine, guanidine, choline, or cinchonine), or an amino acid (e.g., lysine, arginine, or alanine) can be used. Generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are typical. Lists of suitable salts are found in Remington ’s Pharmaceutical Sciences , 18th ed., Mack Publishing Company, Easton, PA, 1990, p. 1445, and Journal of Pharmaceutical Science, 66, 2-19 (1977). For example, they can be a salt of an alkali metal (e.g., sodium or potassium), alkaline earth metal (e.g., calcium), or ammonium of salt.
[0075] The terms “co-administered” or “co-administration” used herein refer to simultaneous or sequential administration.
[0076] The Enamine library of in-stock compounds containing almost 2 million entries was used for initial screens that evaluated the binding pockets and identified early hits (e.g., STND-1 through STND-7). Additional hits came from screening larger Enamine diversity libraries containing 5 and 15 million compounds (e.g., STND-15 through STND-18). The highest scoring hits have been used for identification of potential binders in similarity searches. Search engine GIGA (www.molsoft.com/giga-search.html) was used for screening of the REAL database of 1.2 billion synthesizable compounds from Enamine (enamine.net/bbrary-synthesis/real-compounds/real-database).
[0077] The Quantitative Neighborhoods of Atoms (QNA) methodology (Filimonov et al., SAR QSAR Environ Res, 2009, 20(7-8), 679-709) was used for screening SAVI (Synthetically Accessible Virtual Inventory). SAVI has been recently generated using CHMTRN (CHeMistry TRaNslator) adaptation for a knowledge-based forward synthesis computer program (Judson et al., J Chem Inf Model, 2020, 60(1), 3336-3341). The latest version of SAVI used in the screens contains 1.75 billion entries and first became publicly available November 11, 2020 (Patel et al., Scientific Data, November 11, 2020, 7(384), 1-14).
Libraries of potential hits from REAL and SAVI were subjected to a three-step docking screen procedure that has proven to reliably identify effective binders. Promising compounds from these screens and related analogs were synthesized using one-step reactions and tested for protein binding and STAT3-dependent tumor cell grows inhibition. It was discovered that the para-phenolic group contributes significantly to the binding. Phenol fits well in the binding pocket and forms hydrogen bonds with the amide carbonyl of Glul 11 and the guanido group of Argll4, thus adding significantly to the interaction energy.
[0078] Compounds described herein can be purchased commercially or synthetically prepared. General methods for preparing compounds of the invention are described herein. [0079] The methods described herein comprise administering, to a subject in need thereof, a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof in the form of a pharmaceutical composition. In particular, a pharmaceutical composition comprises at least one STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier. The pharmaceutically acceptable excipients described herein, for example, vehicles, adjuvants, carriers or diluents, are well-known to those who are skilled in the art and are readily available. Typically, the pharmaceutically acceptable carrier is one that is chemically inert to the active compounds and one that has no detrimental side effects or toxicity under the conditions of use.
[0080] The pharmaceutical compositions can be administered as oral, sublingual, transdermal, subcutaneous, topical, absorption through epithelial or mucocutaneous linings, intravenous, intranasal, intraarterial, intraperitoneal, intramuscular, intratumoral, peritumoral, intraperitoneal, intrathecal, rectal, vaginal, or aerosol formulations. In some aspects, the pharmaceutical composition is administered orally or intravenously.
[0081] In accordance with any of the aspects, the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered orally to a subject in need thereof. Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice and include an additive, such as cyclodextrin (e.g., a-, b-, or g-cyclodextrin, hydroxypropyl cyclodextrin) or polyethylene glycol (e.g., PEG400); (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions and gels.
Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent. Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and cornstarch. Tablet forms can include one or more of lactose, sucrose, mannitol, com starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art.
[0082] Formulations suitable for parenteral administration include aqueous and non- aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a salt thereof can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, glycerol ketals, such as 2, 2-dimethyl- 1,3- dioxolane-4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other pharmaceutical adjuvants.
[0083] Oils, which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, com, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters. Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts, and suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene-polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (3) mixtures thereof.
[0084] The parenteral formulations typically contain from about 0.5 to about 25% by weight of the inhibitors in solution. Suitable preservatives and buffers can be used in such formulations. In order to minimize or eliminate irritation at the site of injection, such compositions can contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5 to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. The parenteral formulations can be presented in unit-dose or multi dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
[0085] The inhibitors can be made into injectable formulations. The requirements for effective pharmaceutical carriers for injectable compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, }. B. Lippincott Co., Philadelphia, Pa., Banker and Chalmers, eds., pages 238-250 (1982), and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 (1986).
[0086] Topically applied compositions are generally in the form of liquids (e.g., mouthwash), creams, pastes, lotions, gels, and transdermal patches. Topical administration includes application to the skin and oral mucosa, which includes the oral cavity, oral epithelium, palate, gingival, and the nasal mucosa. In some aspects, the composition contains at least one active component and a suitable vehicle or carrier. It can also contain other components, such as an anti-irritant. The carrier can be a liquid, solid or semi-solid. In aspects, the composition is an aqueous solution, such as a mouthwash. Alternatively, the composition can be a dispersion, emulsion, gel, lotion, or cream vehicle for the various components. In one aspect, the primary vehicle is water or a biocompatible solvent that is substantially neutral or that has been rendered substantially neutral. The liquid vehicle can include other materials, such as buffers, alcohols, glycols, and mineral oils with various emulsifiers or dispersing agents as known in the art to obtain the desired pH, consistency and viscosity. It is possible that the compositions can be produced as solids, such as powders or granules. The solids can be applied directly or dissolved in water or a biocompatible solvent prior to use to form a solution that is substantially neutral or that has been rendered substantially neutral and that can then be applied to the target site. In aspects of the invention, the vehicle for topical application to the skin can include water, buffered solutions, various alcohols, glycols such as glycerin, lipid materials such as fatty acids, mineral oils, phosphoglycerides, collagen, gelatin, and silicone based materials.
[0087] The STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. These aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. [0088] The dose administered to the subject, particularly human and other mammals, in accordance with the present invention should be sufficient to affect the desired response.
One skilled in the art will recognize that dosage will depend upon a variety of factors, including the age, condition or disease state, predisposition to disease, genetic defect or defects, and body weight of the mammal. The size of the dose will also be determined by the route, timing and frequency of administration as well as the existence, nature, and extent of any adverse side-effects that might accompany the administration of a particular inhibitor and the desired effect. It will be appreciated by one of skill in the art that various conditions or disease states may require prolonged treatment involving multiple administrations.
[0089] The inventive methods comprise administering an effective amount of a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof. An “effective amount” means an amount sufficient to show a meaningful benefit in an individual, cell, or tissue to be treated. A meaningful benefit includes, for example, detectably treating, relieving, healing, preventing, delaying the onset of, ameliorating, or lessening one or more symptoms of a disease mediated by overexpression of STAT3 (e.g., inflammation, fluid accumulation), inhibiting, arresting development, preventing, or halting further development of a viral infection or disease, promoting at least one aspect of tumor cell cytotoxicity (e.g., inhibition of growth, inhibiting survival of a cancer cell, reducing proliferation, reducing size and/or mass of a tumor (e.g., solid tumor)). The meaningful benefit observed in the subject can be to any suitable degree (10, 20, 30, 40, 50, 60, 70, 80, 90% or more or anything in between each of these percentages). In some aspects, one or more symptoms of the disease (e.g., disease caused by a bacterium or virus, cancer) are treated, prevented, reduced, halted, or eliminated subsequent to administration of a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof, thereby effectively treating the disease (e.g., disease caused by a bacterium or virus, cancer) to at least some degree.
[0090] Effective amounts can vary depending upon the biological effect desired in the individual, condition to be treated, and/or the specific characteristics of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof, and the individual. In this respect, any suitable dose of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered to the subject (e.g., human), according to the type of disease (e.g., disease caused by a bacterium or virus, cancer) to be treated. Various general considerations taken into account in determining the “effective amount” are known to those of skill in the art and are described, e.g., in Gilman et ak, eds., Goodman and Gilman’s: The Pharmacological Bases of Therapeutics, 8th ed., Pergamon Press, 1990; and Remington’s Pharmaceutical Sciences, 17th Ed., Mack Publishing Co., Easton, Pa., 1990, each of which is herein incorporated by reference. The dose of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof desirably comprises about 0.01 mg per kilogram (kg) of the body weight of the subject (mg/kg) or more (e.g., about 0.05 mg/kg or more, 0.1 mg/kg or more, 0.5 mg/kg or more, 1 mg/kg or more, 2 mg/kg or more, 5 mg/kg or more, 10 mg/kg or more, 15 mg/kg or more, 20 mg/kg or more, 30 mg/kg or more, 40 mg/kg or more, 50 mg/kg or more, 75 mg/kg or more, 100 mg/kg or more, 125 mg/kg or more, 150 mg/kg or more, 175 mg/kg or more, 200 mg/kg or more, 225 mg/kg or more, 250 mg/kg or more, 275 mg/kg or more, 300 mg/kg or more, 325 mg/kg or more, 350 mg/kg or more, 375 mg/kg or more, 400 mg/kg or more, 425 mg/kg or more, 450 mg/kg or more, or 475 mg/kg or more) per day. Typically, the dose will be about 500 mg/kg or less (e.g., about 475 mg/kg or less, about 450 mg/kg or less, about 425 mg/kg or less, about 400 mg/kg or less, about 375 mg/kg or less, about 350 mg/kg or less, about 325 mg/kg or less, about 300 mg/kg or less, about 275 mg/kg or less, about 250 mg/kg or less, about 225 mg/kg or less, about 200 mg/kg or less, about 175 mg/kg or less, about 150 mg/kg or less, about 125 mg/kg or less, about 100 mg/kg or less, about 75 mg/kg or less, about 50 mg/kg or less, about 40 mg/kg or less, about 30 mg/kg or less, about 20 mg/kg or less, about 15 mg/kg or less, about 10 mg/kg or less, about 5 mg/kg or less, about 2 mg/kg or less, about 1 mg/kg or less, about 0.5 mg/kg or less, or about 0.1 mg/kg or less). Any two of the foregoing endpoints can be used to define a close- ended range, or a single endpoint can be used to define an open-ended range.
[0091] For purposes of the present invention, the term “subject” preferably is directed to a mammal. Mammals include, but are not limited to, the order Rodentia, such as mice, and the order Lagomorpha, such as rabbits. It is preferred that the mammals are from the order Carnivora, including Felines (cats) and Canines (dogs). It is more preferred that the mammals are from the order Artiodactyla, including Bovines (cows) and Swines (pigs) or of the order Perissodactyla, including Equines (horses). It is most preferred that the mammals are of the order Primates, Cebids, or Simioids (monkeys) or of the order Anthropoids (humans and apes). An especially preferred mammal is a human. [0092] STAT3 has been long considered a promising drug target due to its involvement in proliferation of tumor cells, inflammation, and immune responses. See, e.g., Bharadwaj et al., Pharmacol Rev 2020, 72 (2), 486-526; Huynh et al., Nat Rev Cancer 2019, 19 (2), 82-96; Gkouveris et al., Journal of Cancer Therapy , 2015, 6, 709-726; WO 2012/159107, and WO 2010/062681. An aberrant STAT3 pathway has also been shown to play a critical role in the pathogenesis of SARS-CoV-2 (Matsuyama et al., Cell Death Differ, October 9, 2020,
27(12), 3209-3225). However, the N-terminal domain of STAT3 has emerged as a very different target (Hu et al ,Mol Cell Biol, 2015, 35(19), 3284-300) and inhibitors of the domain produce significantly different cellular and molecular effects compared to other STAT3 inhibitors (Timofeeva et al., ACS Chem Biol, 2007, 2(12), 799-809; and Timofeeva et al., Proc Natl Acad Sci USA, 2013, 110(A), 1267-1272). A functional aspect is that inhibitors of STAT3 N-domain impact not only tumor cells proliferation, but also immune responses to tumors and bacterial and viral pathogens. Thus, in an aspect, a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), inhibits and/or reduces STAT3 activity, preferably STAT3 N-domain activity, in a cell, such as a cancer cell. The method includes contacting a cell with a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof, whereby the activity of STAT3, particularly STAT3 N-domain, in the cell is inhibited. The STAT3 activity can be measured by any method, including the assays described herein.
[0093] Inhibition of STAT3 has been described in the art as a viable treatment of diseases that are characterized as overexpressing STAT3. In particular, peptide STAT3 N-domain inhibitors have been shown to up-regulate expression of several proapoptotic proteins, in particular c-Fos (Timofeeva et al., Proc Natl Acad Sci USA, 2013, 110 (4), 1267-1272). Western blot analysis of c-Fos expression has shown that the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), effectively increase c-Fos expression in prostate cancer cells. Thus, the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), can be administered to a subject in need thereof as part of a treatment method for diseases that overexpress STAT3 (e.g., a disease caused by a bacterium or virus, cancer). In particular, the disease associated with increased activity of STAT3 protein can be, for example, a bacterial infection, a viral infection, an inflammatory disease, or cancer.
[0094] In an aspect, the disease can be a bacterial infection (including a disease caused by a bacterial infection) or a viral infection (including a disease caused by a viral infection). [0095] The bacterial infection can be caused by, for example, Staphylococcus aureus, gram positive methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus saprophyticus, Pseudomonas aeruginosa, Listeria monocytogenes, Klebsiella pneumoniae, Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus agalactiae, Haemophilus influenzae, Helicobacter pylori, Salmonella, Shigella, Clostridium, Enterobacter aerogenes, gram negative Escherichia coli, Clostridium difficile, or a combination thereof. Diseases caused by a bacterial infection include, e.g., tuberculosis, gastritis, meningitis, gonorrhea, pneumonia, bacterial food poisoning, strep throat, cellulitis, wound infections, and toxic shock syndrome. In some aspects of the method, the bacterial infection is tuberculosis. [0096] The viral infection can be, e.g., influenza virus (e.g., H1N1, H1N2, and H5N1), gastroenteritis, hepatitis virus, herpes simplex virus, West Nile virus, Ebola, human papillomavirus, coronavirus, chickenpox virus, flavivirus, togavirus, levivirus, norovirus, rotavirus, adenovirus, parvovirus, poliovirus, or a combination thereof. The coronavirus can be, for example, SARS-CoV, SARS-CoV-2, or MERS-CoV. Preferably, the disease associated with increased activity of STAT3 protein is SARS-CoV-2. Diseases caused by a viral infection include, e.g., coronavirus disease (COVID-19), severe acute respiratory syndrome (SARS) virus, Middle East respiratory syndrome (MERS), a respiratory disease (e.g., pneumonia, bronchitis, pleural effusion), an inflammatory disease (e.g., inflammation, COVID-19-induced inflammation, pediatric multi-system inflammatory syndrome (PMIS)), reproductive and respiratory syndrome virus (PRRSV), equine arteritis virus (EAV), or gastroenteritis.
[0097] In some aspects when the disease associated with increased activity of STAT3 protein is a bacterial infection (including a disease caused by a bacterial infection) or a viral infection (including a disease caused by a viral infection), the inhibitor compound described herein is used in conjunction with a known therapy (e.g., an antibiotic, an antiviral agent, etc.). An antibiotic can be from any suitable class, such as a penicillin, a tetracycline, a cephalosporin, a quinolone (also known as a fluoroquinolone), a lincomycin, a macrolide, a sulfonamide, a gly copeptide, an aminoglycoside, and a carbapenem. Examples of antibiotics include, for example, amoxicillin, ampicillin, dicloxacilbn, oxacillin, doxycycbne, demeclocycline, ervacycbne, minocycline, omadacycline, tetracycline, cefotaxime, ceftazidime, cefuroxime, cephalexin, ciprofloxacin, moxifloxacin, clindamycin, lincomycin, metronidazole, azithromycin, clarithromycin, erythromycin, sulfamethoxazole and trimethoprim, amoxicillin and clavulanate, levofloxacin, and a combination thereof. An antiviral agent can be from any suitable class, such as a chemokine receptor antagonist, a reverse transcriptase inhibitor, an integrase inhibitor, a protease inhibitor, and a fusion inhibitor. Examples of antiviral agents include, for example, acyclovir, oseltamivir, zanamivir, peramvir, baloxavir, lopinavir, ritonavir, ivermectin, remdesivir, valacyclovir, cidofovir, foscamet, ganciclovir, valganciclovir, penciclovir, famciclovir, idoxuridine, trifluorothymidine, vidarabine, fomivirsen, amantadine, rimantadine, ribavirin, lamivudine, adefovir dipivoxil, entecavir, telbivudine, clevudine, interferons, imiquimod, pleconaril, maraviroc, enfuvirtide, zidovudine, didanosine, zalcitabine, stavudine, abacavir, emtricitabine, tenofovir disoproxil fumarate, nevirapine, delavirdine, efavirenz, raltegravir, saquinavir, indinavir, ritonavir, nelfmavir, amprenavir, fosamprenavir, lopinavir, atazanavir, tipranavir, darunavir, and a combination thereof.
[0098] In certain aspects of the method, the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be co-administered with an antibiotic or antiviral agent. A STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered before, concurrently with, or after administration of an antibiotic or antiviral agent. One or more than one, e.g., two, three, or more antibiotic or antiviral agents can be administered. Accordingly, the present invention is directed to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a combination of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof and at least one antibiotic or antiviral agent.
[0099] In an aspect, the disease to be treated is an inflammatory disease. Generally, an inflammatory disease is that which causes inflammation in at least one tissue. Inflamed tissue includes, e.g., tissue from the gastrointestinal system, pulmonary system, skin, musculature, joints, nervous system, and a combination thereof. Typical inflammatory diseases include, for example, arthritis (including rheumatoid arthritis, osteoarthritis, and psoriatic arthritis), gut inflammation, chronic obstructive pulmonary disease (COPD), asthma, bronchitis, inflammatory bowel disease (IBD), Alzheimer’s disease, Parkinson’s disease, and bursitis.
[0100] In certain aspects of the method, the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be co-administered with an anti-inflammatory agent. The anti-inflammatory agent can be from any suitable class, such as a nonsteroidal anti-inflammatory drug (NSAID) (e.g., a nonselective COX inhibitor, a selective COX2 inhibitor). Examples of anti inflammatory agents include, for example, celecoxib, etoricoxib, valdecoxib, aspirin, diclofenac, ibuprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, diflunisal, naproxen, phenylbutazone, piroxicam, sulindac, mefenamic acid, etodolac, meloxicam, nambumetone, oxaprozin, tolmetin, acetaminophen, duloxetine, and a combination thereof. [0101] A STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered before, concurrently with, or after administration of an anti-inflammatory agent. One or more than one, e.g., two, three, or more anti-inflammatory agents can be administered. Accordingly, the present invention is directed to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a combination of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof and at least one anti-inflammatory agent.
[0102] In some aspects, the disease associated with increased activity of STAT3 protein is cancer. The type of cancer to be treated or prevented is not particularly limited, but in certain aspects, the cancer is characterized as having increased STAT3 activity relative to normal tissue of the same type. See, for example, Garcia et ak, Cell Growth Differ, 1997, 8 (12), 1267-1276; Watson et ak, Br J Cancer, 1995, 71(4), 840-844; Huang et ak, Gynecol Oncol, 2000, 79(1), 67-73; Dhir et ak, Prostate, 2002, 51(4), 241-246; Mora et ak, Cancer Res, 2002, 62(22), 6659-6666; Corvinus et ak, Neoplasia, 2005, 7(6), 545-555; Guo et ak,
Am J Transl Res , 2009, 1(3), 283-290; Schaefer et ak, Oncogene, 2002, 21(13), 2058-2065; and Wei et ak, Oncogene, 2003, 22(3), 319-329. For example, STAT3 is constitutively active in over 40% of all breast cancers, particularly in triple-negative breast cancers which lack the expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2/Neu) (Banerjee et ak, Int J Cancer, 2016, 138(11), 2570-2578).
[0103] Examples of cancer treatable with the inventive method include cancers, including cancerous cells and tissue, of the head and neck, eye, skin, mouth, throat, esophagus, chest, bone, lung, colon, sigmoid, rectum, stomach, prostate, breast, ovaries, kidney, liver, pancreas, brain, intestine, heart, or adrenals. More particularly, cancers include solid tumor, sarcoma, carcinomas, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendothelio sarcoma, synovioma, mesothelioma, Ewing’s tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms’ tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, Kaposi’s sarcoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, retinoblastoma, a blood-borne tumor, acute lymphoblastic leukemia, acute lymphoblastic B-cell leukemia, acute lymphoblastic T-cell leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute monoblastic leukemia, acute erythroleukemic leukemia, acute megakaryoblastic leukemia, acute myelomonocytic leukemia, acute nonlymphocytic leukemia, acute undifferentiated leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia, hairy cell leukemia, or multiple myeloma. See, e.g., Harrison ’s Principles of Internal Medicine, Eugene Braunwald et ak, eds., pp. 491-762 (15th Ed. 2001). In some aspects, the cancer is breast cancer, prostate cancer, pancreatic cancer, or leukemia. Anti-cancer activity can be measured by any suitable method, including the assays described herein.
[0104] In certain aspects of this method, the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be co-administered with an anti-cancer agent (e.g., a chemotherapeutic agent) and/or radiation therapy. In an aspect, the method comprises administering an amount of a STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof that is effective to sensitize the cancer cells to one or more therapeutic regimens (e.g., chemotherapy or radiation therapy). A STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof can be administered before, concurrently with, or after administration of another anti-cancer agent (e.g., a chemotherapeutic agent).
[0105] One or more than one, e.g., two, three, or more anti-cancer agents can be administered. In this regard, the present invention is directed to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a combination of the STAT3 inhibitor compound, including formulas (la) through (Ih) and (la-1) through (Ih-1), or a pharmaceutically acceptable salt thereof and at least one anti-cancer agent (e.g., chemotherapeutic agent).
[0106] Examples of anti-cancer agents include platinum compounds (e.g., cisplatin, carboplatin, oxaliplatin), alkylating agents (e.g., cyclophosphamide, ifosfamide, chlorambucil, nitrogen mustard, thiotepa, melphalan, busulfan, procarbazine, streptozocin, temozolomide, dacarbazine, bendamustine), antitumor antibiotics (e.g., daunorubicin, doxorubicin, idarubicin, epirubicin, mitoxantrone, bleomycin, mitomycin C, plicamycin, dactinomycin), taxanes (e.g., pacbtaxel and docetaxel), antimetabolites (e.g., 5-fluorouracil, cytarabine, pemetrexed, thioguanine, floxuridine, capecitabine, and methotrexate), nucleoside analogues (e.g., fludarabine, clofarabine, cladribine, pentostatin, nelarabine), topoisomerase inhibitors (e.g., topotecan and irinotecan), hypomethylating agents (e.g., azacitidine and decitabine), proteosome inhibitors (e.g., bortezomib), epipodophyllotoxins (e.g., etoposide and teniposide), DNA synthesis inhibitors (e.g., hydroxyurea), vinca alkaloids (e.g., vincristine, vindesine, vinorelbine, and vinblastine), tyrosine kinase inhibitors (e.g., imatinib, dasatinib, nilotinib, sorafenib, sunitinib), monoclonal antibodies (e.g., rituximab, cetuximab, panitumumab, tositumomab, trastuzumab, alemtuzumab, gemtuzumab ozogamicin, bevacizumab), nitrosoureas (e.g., carmustine, fotemustine, and lomustine), enzymes (e.g., L- Asparaginase), biological agents (e.g., interferons and interleukins), hexamethylmelamine, mitotane, angiogenesis inhibitors (e.g., thalidomide, lenalidomide), steroids (e.g., prednisone, dexamethasone, and prednisolone), a CDK4/6 inhibitor (e.g., abemaciclib, palbocicbb, ribocicbb), anti-cancer hormonal agents (e.g., tamoxifen, fulvestrant, raloxifene, leuprobde, bicalutamide, granisetron, flutamide, goserebn), aromatase inhibitors (e.g., exemestane, letrozole, and anastrozole), arsenic trioxide, tretinoin, nonselective cyclooxygenase inhibitors (e.g., nonsteroidal anti-inflammatory agents, salicylates, aspirin, piroxicam, ibuprofen, indomethacin, naprosyn, diclofenac, tolmetin, ketoprofen, nabumetone, oxaprozin), selective cyclooxygenase-2 (COX-2) inhibitors, immune checkpoint inhibitors (e.g., anti-PDl, anti- CTLA4, and anti-PD-Ll), cellular immunotherapy (e.g., chimeric antigen receptor T cell therapy, tumor-infiltrating lymphocyte therapy), or any combination thereof.
[0107] The invention is further illustrated by the following aspects.
[0108] Aspect (1) A method of treating a disease associated with increased activity of STAT3 protein comprising administering to a subject in need thereof a phenol group- containing compound or a pharmaceutically acceptable salt thereof, wherein the phenol group-containing compound has a core structural residue selected from the group consisting of formula (la)
Figure imgf000054_0001
(la), wherein R1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, formula (lb)
Figure imgf000054_0002
(lb). formula (Ic)
Figure imgf000054_0003
(Ic), formula (Id)
Figure imgf000055_0001
(Id), formula (Ie)
Figure imgf000055_0005
(Ie), wherein X1 is CH or N, X2 is a bond, CH2, or C(O), and R14 is H or F formula (If)
Figure imgf000055_0002
wherein each instance of R17 is H, alkyl,
Figure imgf000055_0003
r alkyl, and n is an integer of 1-3, formula (Ig)
Figure imgf000055_0004
(Ig), wherein R20 is CH2 or CH=CH, and formula (Ih) (Ih), wherein each of the core structural residues of formulas (la) through (Ih) is bonded to the remainder of the phenol group-containing compound, or a compound or a pharmaceutically acceptable salt thereof selected from the group consisting of
Figure imgf000056_0001
[0109] Aspect (2) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (la) that forms formula (la-1) wherein
R1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, and R2 is selected from the group consisting of
Figure imgf000057_0001
wherein
R3 is heterocyclyl,
R4 is aryl, heteroaryl, or alkenyl, R5 is haloalkyl or alkenyl, R6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -S02alkenyl,
R7 is H or alkyl,
R8 is aryl or heteroaryl, each of which is optionally substituted,
R9 is alkyl or haloalkyl,
X3 is NH or O,
X4 is CH 2, C(O), or NH, and m is an integer of 1-5, or a pharmaceutically acceptable salt thereof.
[0110] Aspect (3) The method of aspect (2), wherein R2 is selected from the group consisting of
Figure imgf000058_0001
Figure imgf000059_0001
or a pharmaceutically acceptable salt thereof.
[0111] Aspect (4) The method of aspect (3), wherein the compound of formula (la-1) is selected from the group consisting of STND-5, STND-8, STND-9, STND-24, STND-25, STND-29, STND-33, STND-34, STND-35, STND-36, STND-37, STND-38, STND-39, STND-40, STND-41, STND-42, STND-43, STND-44, STND-45, STND-46, and STND-47, or a pharmaceutically acceptable salt thereof.
[0112] Aspect (5) The method of aspect (4), wherein the compound of formula (la-1) is selected from the group consisting of STND-9, STND-25, and STND-34, or a pharmaceutically acceptable salt thereof.
[0113] Aspect (6) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (lb) that forms formula (Ib-1)
Figure imgf000060_0001
(Ib-1), wherein R10 is selected from the group consisting of
Figure imgf000061_0001
pharmaceutically acceptable salt thereof. [0114] Aspect (7) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (Ic) that forms formula (Ic-1)
Figure imgf000061_0002
(Ic-1), wherein R11 is OH (STND-30) or
Figure imgf000062_0001
(STND-31) or a pharmaceutically acceptable salt thereof.
[0115] Aspect (8) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (Id) that forms formula (Id-1)
Figure imgf000062_0002
(Id-1), wherein
Figure imgf000062_0003
Figure imgf000062_0004
wherein R13 is alkyl, or a pharmaceutically acceptable salt thereof. [0116] Aspect (9) The method of aspect (8), wherein is methyl (STND-1) or ethyl (STND-22), or a pharmaceutically acceptable salt thereof. [0117] Aspect (10) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (Ie) that forms formula (Ie-1)
Figure imgf000063_0001
(Ie-1), wherein X1 is CH or N, X2 is a bond, CH2, or C(O), R14 is H or F, and R15 is selected from the group consisting of
Figure imgf000063_0002
pharmaceutically acceptable salt thereof.
[0118] Aspect (11) The method of aspect (10), wherein the compound of formula (Ie-1) is selected from the group consisting of STND-7, STND-12, STND-16, STND-17, and STND- 18, or a pharmaceutically acceptable salt thereof.
[0119] Aspect (12) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (If) that forms formula (If-1) wherein each instance of R17 is H, alkyl,
Figure imgf000064_0001
r alkyl, n is 1-3, and R19 is selected from the group consisting of
Figure imgf000064_0002
pharmaceutically acceptable salt thereof.
[0120] Aspect (13) The method of aspect (12), wherein the compound of formula (If-1) is selected from the group consisting of STND-6, STND-14, STND-15, and STND-20, or a pharmaceutically acceptable salt thereof.
[0121] Aspect (14) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (Ig) that forms formula (Ig-1)
Figure imgf000064_0003
(Ig-1), wherein R20 is CTk or CH=CH, and R21 is selected from the group consisting of
Figure imgf000064_0004
pharmaceutically acceptable salt thereof. [0122] Aspect (15) The method of aspect (14), wherein the compound of formula (Ig-1) is selected from the group consisting of STND-10 and STND-13 or a pharmaceutically acceptable salt thereof.
[0123] Aspect (16) The method of aspect (1), wherein the phenol group-containing compound has a core structural residue of formula (Ih) that forms formula (Ih-1)
Figure imgf000065_0001
(Ih-1), wherein R22 is selected from the group consisting of
Figure imgf000065_0002
pharmaceutically acceptable salt thereof.
[0124] Aspect (17) The method of aspect (1), wherein the compound to be administered is selected from the group consisting of STND-3, STND-4, and STND-19, or a pharmaceutically acceptable salt thereof.
[0125] Aspect (18) The method of any one of aspects (l)-(l 7), wherein the disease associated with increased activity of STAT3 protein is cancer.
[0126] Aspect (19) The method of aspect (18), wherein the cancer is breast cancer, prostate cancer, pancreatic cancer, or leukemia.
[0127] Aspect (20) The method of any one of aspects (l)-(l 7), wherein the disease associated with increased activity of STAT3 protein is a bacterial or viral infection.
[0128] Aspect (21) The method of aspect (20), wherein the bacterial infection is tuberculosis.
[0129] Aspect (22) The method of aspect (20), wherein the viral infection is by a coronavirus. [0130] Aspect (23) The method of any one of aspects (l)-(l 7), wherein the disease associated with increased activity of STAT3 protein is an inflammatory disease.
[0131] Aspect (24) The method of aspect (23), wherein the inflammatory disease causes inflammation in at least one tissue selected from the group consisting of a gastrointestinal system, a pulmonary system, skin, musculature, joints, a nervous system, and a combination thereof.
[0132] Aspect (25) A compound of formula (la-1)
Figure imgf000066_0001
(la-1), wherein
R1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, and R2 is selected from the group consisting of
wherein
R3 is heterocyclyl,
R4 is aryl, heteroaryl, or alkenyl,
R5 is haloalkyl or alkenyl,
R6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -S02alkenyl,
R7 is H or alkyl,
R8 is aryl or heteroaryl, each of which is optionally substituted,
R9 is alkyl or haloalkyl,
X3 is NH or O,
X4 is CH 2, C(O), or NH, and m is an integer of 1-5, or a pharmaceutically acceptable salt thereof, provided that the compound of formula (la-1) is not STND-5, STND-8, STND-9, STND-24, STND-25, STND-35, or STND-36.
[0133] Aspect (26) The compound of aspect (25) that is selected from the group consisting of STND-29, STND-33, STND-34, STND-37, STND-38, STND-39, STND-40, STND-41, STND-42, STND-43, STND-44, STND-45, STND-46, and STND-47, or a pharmaceutically acceptable salt thereof, preferably STND-29, STND-33, STND-34, STND- 37, STND-38, STND-39, STND-40, STND-41, STND-42, STND-46, or STND-47. [0134] Aspect (27) A compound selected from the group consisting of a compound of formula (Ib-1)
Figure imgf000068_0002
a compound of formula (Ic-1)
Figure imgf000068_0001
(Ic-1),
wherein R11 is OH (STND-30) or
Figure imgf000069_0001
(Id-1), wherein
Figure imgf000069_0002
a compound of formula (Ih-1)
Figure imgf000069_0003
(Ih-1), wherein pharmaceutically acceptable salt thereof.
[0135] Aspect (28) A pharmaceutical composition comprising a compound of any one of aspects (25)-(27) and a pharmaceutically acceptable carrier.
[0136] Aspect (29) A method of treating a disease associated with increased activity of signal transducer and activator of transcription 3 (STAT3) protein comprising administering to a subject in need thereof a compound of any one of aspects (25)-(27) or a pharmaceutically acceptable salt thereof.
[0137] The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope.
EXAMPLE 1
[0138] Exemplary STAT3 N-domain inhibitor compounds can be prepared in accordance with the following reaction schemes.
[0139] FIG. 1 is a reaction scheme of preparing STND-25. DIC is N,N' - diisopropylcarbodiimide.
[0140] FIG. 2 is a reaction scheme of preparing STND-27. DIEA is N.N'- diisopropylethylamine.
[0141] FIG. 3 is a reaction scheme of preparing STND-28.
[0142] FIG. 4 is a reaction scheme of preparing STND-29.
[0143] FIG. 5 is a reaction scheme of preparing STND-30.
[0144] FIG. 6 is a reaction scheme of preparing STND-31.
[0145] FIG. 7 is a reaction scheme of preparing STND-33.
[0146] FIG. 8 is a reaction scheme of preparing STND-34. TMSC1 is chlorotrimethylsilane.
[0147] FIG. 9 is a reaction scheme of preparing STND-37.
[0148] FIG. 10 is a reaction scheme of preparing STND-38.
[0149] FIG. 11 is a reaction scheme of preparing STND-39. [0150] FIG. 12 is a reaction scheme of preparing STND-40.
[0151] FIG. 13 is a reaction scheme of preparing STND-42.
[0152] FIG. 14 is a reaction scheme of preparing STND-43.
[0153] FIG. 15 is a reaction scheme of preparing STND-44.
[0154] FIG. 16 is a reaction scheme of preparing STND-45.
[0155] FIG. 17 is a reaction scheme of preparing STND-46.
[0156] FIG. 18 is a reaction scheme of preparing STND-47.
EXAMPLE 2
[0157] This example demonstrates the STAT3 N-domain specificity and cytotoxicity of exemplary compounds in an aspect of the invention.
[0158] IC50 was determined in the prostate cancer cell line DU145 after 48 hours of exposure to compounds. Binding to the STAT3 N-domain protein (KD) was determined using microscale thermophoresis (MST). The results are set forth in Table 1. The last column of Table 1 identifies the source of each compound, either from a library or designed. Compounds in the SAVI database were first designed by the inventors prior to uploading to the database.
Table 1.
Figure imgf000071_0001
Figure imgf000072_0001
EXAMPLE 3
[0159] This example demonstrates that exemplary STAT3 N-domain binders of the invention inhibit STAT3 signaling in HEK-BLUE IL-10 reporter cells.
[0160] Inhibition of 11-10- induced secretion of embryonic alkaline phosphatase in the reporter cell line, HEK-Blue IL10 cells (Invivogen, www.invivogen.com/hek-blue-il 10) was measured. Cells (-70.000 cells/mL) were incubated with IL-10 (0.1 ng/ml) without an inhibitor (control) or with 500 mM of compound by measuring the secreted embryonic alkaline phosphatase (SEAP) levels induced after 24 hours incubation. As seen in FIG. 19, compounds of the invention not only effectively interfered with STAT3 activity, but the degree of inhibition by different compounds correlated well with the binding score and compound’s toxicity in STAT3-dependent cancer cells. The effects on STAT3 signaling were concentration dependent (FIG. 20). Although the compounds showed some toxicity on HEK cells, the toxicity was less pronounced than the effects on the signaling, strongly suggesting that the observed reduction in transcription of the marker gene was not a result of cell loss.
EXAMPLE 4
[0161] This example demonstrates the dose response of STAT3 N-domain bonding for an exemplary compound of the invention.
[0162] Recombinant STAT3 N-terminal domain titration of STND-9 was monitored by microscale thermophoresis (MST). Hexahistidine-tagged (His-tag) STAT3 N-domain was labeled with the non-covalent fluorescent dye RED-tris-NTA (RED-tris-nitrilotriacetic acid). The protein concentration was 50 nM. Fnorm was calculated as Fi/Fo, in which Fi is the fluorescence value after the laser is turned on, and Fo is the fluorescence value before the laser was turned on. As seen in FIG. 21, the apparent KD was 7.3 mM.
EXAMPLE 5
[0163] This example demonstrates the cytotoxicity of exemplary compounds of the invention.
[0164] DU145 cells were treated with compounds STND-2 (3 mM), STND-9 (3 mM), and
STND-9 (10 mM) for 3 hours (with DMSO treatment as a negative control), and nuclear fraction was isolated and used for Western blot analysis as described in Timofeeva et al.
(Proc Natl Acad Sci USA, 2013, 110(4), 1267-1272). Anti-c-Fos antibody was used at 1 : 1000 dilution. The blot was stripped and re-probed with D-Actin antibody at 1 : 1000 to demonstrate equal loading of the wells. Western blot analysis of c-Fos expression showed that the small molecule domain inhibitors of the invention effectively increase STAT3 N- domain inhibition marker c-Fos expression in prostate cancer cells.
[0165] Next, prostate cancer cells (DU145 cell line) were exposed to STND-2, STND-8, and STND-9 for 48 hours, and the number of surviving cells was quantitated using an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) cell viability assay (Sigma- Aldrich, St. Louis, MO). In this colorimetric assay, yellow crystals of MTT were reduced to purple crystals of formazan by metabolically active cells. Insoluble formazan crystals were dissolved using a solubilization solution, and the resulting colored solution was quantified by measuring the absorbance at 500-600 nm. The N-domain peptide inhibitor ST3H2-A2 (Timofeeva et al., Proc Natl Acad Sci USA, 2013, 110(4), 1267-1272) was used as a positive control. The effect of STAT3 N-domain inhibitors of the invention on the growth of prostate cancer is shown in FIG. 22. FIG. 23 shows that the compounds of the invention are selectively toxic to prostate tumor cells (DU145) and show very little toxicity to normal prostate epithelial cells (RWPE-1).
EXAMPLE 6
[0166] This example demonstrates the cytotoxicity of exemplary compounds of the invention.
[0167] The effect of STAT3 N-domain inhibitors STND-9, STND-25, STND-30, STND- 31, and STND-32 on the growth of pancreatic cancer cells (KPC95775 cell line) is shown in FIG. 24. Cells were exposed to compounds for 48 hours, and the number of surviving cells was quantitated using an MTT assay.
[0168] The cytotoxicity studies demonstrate that the STAT3 N-domain inhibitors of the invention are more than an order of magnitude more potent than previously described peptide inhibitors. The inhibitors of the invention show very similar in vitro effects on STAT3 signaling, and consequently can be used for both tumor growth inhibition and modulation of immune responses to tumors and infectious agents.
[0169] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
[0170] The use of the terms “a” and “an” and “the” and “at least one” and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The use of the term “at least one” followed by a list of one or more items (for example, “at least one of A and B”) is to be construed to mean one item selected from the listed items (A or B) or any combination of two or more of the listed items (A and B), unless otherwise indicated herein or clearly contradicted by context. The terms “comprising,” “having,” “including,” and “containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to,”) unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
[0171] Preferred aspects of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred aspects may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims

CLAIM(S)
1. A signal transducer and activator of transcription 3 (STAT3) protein inhibitor compound or a pharmaceutically acceptable salt thereof for use in treating a disease associated with increased activity of STAT3 protein, wherein the inhibitor compound is a phenol group-containing compound with a core structural residue selected from the group consisting of formula (la)
Figure imgf000076_0001
wherein R1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, provided that the compound of formula (la) is not
Figure imgf000076_0002
formula (lb) (lb), formula (Ic)
Figure imgf000077_0001
Ic , formula (Id)
Figure imgf000077_0002
(Id), formula (Ie)
Figure imgf000077_0003
(Ie), wherein X1 is CH or N, X2 is a bond, CH2, or C(O), and R14 is H or F formula (If) wherein each instance of R17 is H, alkyl,
Figure imgf000078_0001
r alkyl, and n is an integer of 1-3, formula (Ig)
Figure imgf000078_0002
(Ig), wherein R20 is CH2 or CH=CH, and formula (Ih)
Figure imgf000078_0003
Oh), wherein each of the core structural residues of formulas (la) through (Ih) is bonded to the remainder of the phenol group-containing compound, or an inhibitor compound selected from the group consisting of
Figure imgf000078_0004
or a pharmaceutically acceptable salt thereof.
2. The STAT3 protein inhibitor compound or a pharmaceutically acceptable salt thereof for use according to claim 1, wherein the phenol group-containing compound has a core structural residue selected from the group consisting of:
(i) formula (la-1)
Figure imgf000079_0001
(la-1), wherein
R1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, and R2 is selected from the group consisting of
Figure imgf000080_0001
wherein
R3 is heterocyclyl,
R4 is aryl, heteroaryl, or alkenyl,
R5 is haloalkyl or alkenyl,
R6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -SC alkenyl, R7 is H or alkyl,
R8 is aryl or heteroaryl, each of which is optionally substituted, R9 is alkyl or haloalkyl,
X3 is NH or O,
X4 is CH 2, C(O), or NH, and m is an integer of 1-5,
(ii) formula (Ib-1) (Ib-1), wherein R10 is selected from the group consisting of
Figure imgf000081_0001
(STND-28),
(iii) formula (Ic-1)
Figure imgf000082_0001
wherein R13 is alkyl,
(v) formula (Ie-1) (Ie-1), wherein
X1 is CH orN,
X2 is a bond, CH2, or C(O),
R14 is H or F, and
R15 is selected from the group consisting of
Figure imgf000083_0001
wherein R16 is alkyl or cycloalkyl, (vi) formula
Figure imgf000083_0003
(If-1), wherein each instance of R17 is H, alkyl,
Figure imgf000083_0002
R18 is H or alkyl, n is 1-3, and R19 is selected from the group consisting
Figure imgf000084_0001
Figure imgf000084_0002
(Ig-1), wherein
R20 is CH 2 or CH=CH, and
R21 is selected from the group consisting
Figure imgf000084_0003
Figure imgf000084_0004
and
(viii) formula (Ih-1) (Ih-1), wherein R22 is selected from the group consisting of
Figure imgf000085_0001
3. The STAT3 protein inhibitor compound or a pharmaceutically acceptable salt thereof for use according to claim 2, wherein R2 of formula (la-1) is selected from the group
Figure imgf000085_0002
Figure imgf000086_0001
or a pharmaceutically acceptable salt thereof.
4. The STAT3 protein inhibitor compound or a pharmaceutically acceptable salt thereof for use according to claim 3, wherein the compound of formula (la-1) is selected from the group consisting of ,
Figure imgf000089_0001
5, ,
5. The STAT3 protein inhibitor compound or a pharmaceutically acceptable salt thereof for use according to claim 4, wherein the compound of formula (la-1) is selected from the group consisting of
Figure imgf000092_0001
or a pharmaceutically acceptable salt thereof.
6. The STAT3 protein inhibitor compound or a pharmaceutically acceptable salt thereof for use according to claim 1, wherein the inhibitor compound is selected from the group consisting of 7. The STAT3 protein inhibitor compound or a pharmaceutically acceptable salt thereof for use according to any one of claims 1-6, wherein the disease associated with increased activity of STAT3 protein is cancer.
8. The STAT3 protein inhibitor compound or a pharmaceutically acceptable salt thereof for use according to claim 7, wherein the cancer is breast cancer, prostate cancer, pancreatic cancer, or leukemia.
9. The STAT3 protein inhibitor compound or a pharmaceutically acceptable salt thereof for use according to any one of claims 1-6, wherein the disease associated with increased activity of STAT3 protein is a bacterial infection or a viral infection.
10. The STAT3 protein inhibitor compound or a pharmaceutically acceptable salt thereof for use according to any one of claims 1-6, wherein the disease associated with increased activity of STAT3 protein is an inflammatory disease.
11. A compound of formula (la-1)
Figure imgf000094_0001
wherein
R1 is H, alkylcarbonyl, alkylcarboxy, amido, alkylamido, or dialkylamido, and
R2 is selected from the group consisting
Figure imgf000094_0002
Figure imgf000094_0003
Figure imgf000095_0001
wherein
R3 is heterocyclyl,
R4 is aryl, heteroaryl, or alkenyl,
R5 is haloalkyl or alkenyl,
R6 is aralkyl, heteroarylalkyl, CH2C(0)alkenyl, or -S02alkenyl,
R7 is H or alkyl,
R8 is aryl or heteroaryl, each of which is optionally substituted,
R9 is alkyl or haloalkyl,
X3 is NH or O,
X4 is CH 2, C(O), or NH, and m is an integer of 1-5, or a pharmaceutically acceptable salt thereof, provided that the compound of formula (la-1) is not STND-5, STND-8, STND-9, STND-24, STND-25, STND-35, STND-36, STND-43, STND-44, or STND-45: ,
Figure imgf000097_0001
12. The compound according to claim 11 that is selected from the group consisting of
Figure imgf000098_0001
-38,
, or a pharmaceutically acceptable salt thereof.
13. A pharmaceutical composition comprising a compound according to claim 11 or 12 and a pharmaceutically acceptable carrier.
14. A compound or a pharmaceutically acceptable salt thereof according to claim 11 or 12 for use in treating a disease associated with increased activity of signal transducer and activator of transcription 3 (STAT3) protein.
15. The compound or a pharmaceutically acceptable salt thereof for use according to claim 14, wherein the disease associated with increased activity of STAT3 protein is selected from the group consisting of cancer, a bacterial infection, a viral infection, and an inflammatory disease.
PCT/US2021/058936 2021-02-26 2021-11-11 Small molecule inhibitors of stat3 n-terminal domain and methods of use WO2022182395A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163154440P 2021-02-26 2021-02-26
US63/154,440 2021-02-26

Publications (1)

Publication Number Publication Date
WO2022182395A1 true WO2022182395A1 (en) 2022-09-01

Family

ID=78829805

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/058936 WO2022182395A1 (en) 2021-02-26 2021-11-11 Small molecule inhibitors of stat3 n-terminal domain and methods of use

Country Status (1)

Country Link
WO (1) WO2022182395A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023196958A3 (en) * 2022-04-07 2023-11-23 Oncolinx Pharmaceuticals Tumor and cancer targeting compounds

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008151037A1 (en) * 2007-05-30 2008-12-11 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Peptide-based stat inhibitor
WO2010062681A2 (en) 2008-10-30 2010-06-03 University Of South Florida Luteolin and diosmin/diosmetin as novel stat3 inhibitors for treating autism
WO2012159107A1 (en) 2011-05-19 2012-11-22 Rhode Island Hospital Inhibition of renal fibrosis
WO2012166891A2 (en) * 2011-05-31 2012-12-06 Algynomics Inc. Mu-opioid receptor binding compounds

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008151037A1 (en) * 2007-05-30 2008-12-11 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Peptide-based stat inhibitor
WO2010062681A2 (en) 2008-10-30 2010-06-03 University Of South Florida Luteolin and diosmin/diosmetin as novel stat3 inhibitors for treating autism
WO2012159107A1 (en) 2011-05-19 2012-11-22 Rhode Island Hospital Inhibition of renal fibrosis
WO2012166891A2 (en) * 2011-05-31 2012-12-06 Algynomics Inc. Mu-opioid receptor binding compounds

Non-Patent Citations (42)

* Cited by examiner, † Cited by third party
Title
"ASHP Handbook on Injectable Drugs,", 1986, pages: 622 - 630
"Gilman's: The Pharmacological Bases of Therapeutics,", 1990, MACK PUBLISHING COMPANY, pages: 1445
"Harrison's Principles of Internal Medicine", 2001, pages: 491 - 762
"Pharmaceutics and Pharmacy Practice", 1982, J. B. LIPPINCOTT CO., pages: 238 - 250
BANERJEE ET AL., INT J CANCER,, vol. 138, no. 11, 2016, pages 2570 - 2578
BHARADWAJ ET AL., PHARMACOL REV, vol. 72, no. 2, 2020, pages 486 - 526
CORVINUS ET AL., NEOPLASIA, vol. 7, no. 6, 2005, pages 545 - 555
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 11 September 2011 (2011-09-11), XP002805642, Database accession no. 1331421-06-7 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 March 2008 (2008-03-12), XP002805646, Database accession no. 1007541-92-5 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 20 June 2007 (2007-06-20), XP002805647, Database accession no. 937973-42-7 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 20 June 2007 (2007-06-20), XP002805648, Database accession no. 937930-85-3 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 21 March 2010 (2010-03-21), XP002805643, Database accession no. 1212795-69-1 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 28 March 2008 (2008-03-28), XP002805645, Database accession no. 1010555-66-4 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 4 April 2008 (2008-04-04), XP002805644, Database accession no. 1012024-62-2 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002805649, Database accession no. 937911-75-6 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002805650, Database accession no. 937901-20-7 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002805651, Database accession no. 937845-10-8 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002805652, Database accession no. 1331726-49-8 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002805653, Database accession no. 937911-66-5 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002805654, Database accession no. 849040-14-8 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; XP002805655, Database accession no. 849040-13-7 *
DHIR ET AL., PROSTATE, vol. 51, no. 4, 2002, pages 241 - 246
FILIMONOV ET AL., SAR QSAR ENVIRON RES, vol. 20, no. 7-8, 2009, pages 679 - 709
GARCIA ET AL., CELL GROWTH DIFFER, vol. 8, no. 12, 1997, pages 1267 - 1276
GKOUVERIS ET AL., JOURNAL OF CANCER THERAPY, vol. 6, 2015, pages 709 - 726
GKOUVERIS ET AL., JOURNAL OF CANCER THERAPY,, vol. 6, 2015, pages 709 - 726
GUO ET AL., AM J TRANSL RES, vol. 1, no. 3, 2009, pages 283 - 290
HU ET AL., MOL CELL BIOL, vol. 35, no. 19, 2015, pages 3284 - 300
HU WANTING: "Progress of JAK/STAT 3 and its inhibitors in the treatment of cancer", AIP CONFERENCE PROCEEDINGS AIP CONFERENCE PROCEEDINGS, 19 February 2020 (2020-02-19), pages 20045 - 1, XP055890177, Retrieved from the Internet <URL:https://aip.scitation.org/doi/pdf/10.1063/5.0000440?casa_token=IlXRJMItsoQAAAAA:PNkP49H9aVBbFJW0Syp6fka2ZnyXwrv7zn2DrP4Es0IPuMM2MLlLRe11l1B98bKc5zWPkoTARMZX> [retrieved on 20220210] *
HUANG ET AL., GYNECOL ONCOL, vol. 79, no. 1, 2000, pages 67 - 73
HUYNH ET AL., NAT REV CANCER, vol. 19, no. 2, 2019, pages 82 - 96
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2 - 19
JUDSON ET AL., J CHEM INFMODEL, vol. 60, no. 7, 2020, pages 3336 - 3341
MATSUYAMA ET AL., CELL DEATH DIFFER, vol. 27, no. 12, 9 October 2020 (2020-10-09), pages 3209 - 3225
MORA ET AL., CANCER RES, vol. 62, no. 22, 2002, pages 6659 - 6666
PATEL ET AL., SCIENTIFIC DATA, vol. 7, no. 384, 11 November 2020 (2020-11-11), pages 1 - 14
SCHAEFER ET AL., ONCOGENE, vol. 21, no. 13, 2002, pages 2058 - 2065
TIMOFEEVA ET AL., ACS CHEM BIOL, vol. 2, no. 12, 2007, pages 799 - 809
TIMOFEEVA ET AL., PROC NATL ACAD SCI USA, vol. 110, no. 4, 2013, pages 1267 - 1272
WATSON ET AL., BR J CANCER, vol. 71, no. 4, 1995, pages 840 - 844
WEI ET AL., ONCOGENE, vol. 22, no. 3, 2003, pages 319 - 329
YUE ET AL., EXPERT OPINION INVESTIG DRUGS,, vol. 18, no. 1, 2009, pages 45 - 56

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023196958A3 (en) * 2022-04-07 2023-11-23 Oncolinx Pharmaceuticals Tumor and cancer targeting compounds

Similar Documents

Publication Publication Date Title
TW202014408A (en) Compounds and compositions for treating conditions associated with sting activity
CN112823151A (en) Compounds and compositions for treating diseases associated with STING activity
SA518400704B1 (en) Crystalline forms of a triazolopyrimidine compound
SA518391985B1 (en) Modulators of 5&#39;-Nucleotidase, ECTO and The Use Thereof
CA3011528A1 (en) Cyclic dinucleotides for treating conditions associated with sting activity such as cancer
TW202043198A (en) Compounds and compositions for treating conditions associated with sting activity
ES2824026T3 (en) Synthetic Antibody Mimetic Compounds (SyAM) Targeting Cancer, Especially Prostate Cancer
KR20160125361A (en) Var2csa-drug conjugates
CN113905766A (en) Programmable polymeric pharmaceuticals
WO2020106736A1 (en) Compounds and compositions for treating conditions associated with sting activity
KR20140000275A (en) Bifunctional molecules with antibody-recruiting and entry inhibitory activity against the human immunodeficiency virus
WO2020106741A1 (en) Compounds and compositions for treating conditions associated with sting activity
WO2010013011A1 (en) P53 tumor suppressor protein activating
BR112021000395A2 (en) DIMMERIC IMMUNOMODULATING COMPOUNDS AGAINST CEREBLON-BASED MECHANISMS
US11767298B2 (en) Substituted benzimidazoles as inhibitors of transforming growth factor-β kinase
WO2019122202A1 (en) Sting inhibitors
WO2022182395A1 (en) Small molecule inhibitors of stat3 n-terminal domain and methods of use
JP2024502474A (en) Urea-bearing heterobicyclic compounds or analogs and compositions thereof for treating conditions associated with STING activity
JP2021533183A (en) Compounds, compositions, methods and uses for the treatment of cancer and immune disorders
US10000469B2 (en) Heat shock protein 70 (hsp-70) receptor ligands
JP2020529988A (en) Compounds and their use for the prevention and treatment of medical disorders
US10538500B2 (en) Tricyclic prodrugs
TW202334084A (en) Compounds and compositions for treating conditions associated with sting activity
US20240083879A1 (en) Oxalamide compounds and compositions for treating conditions associated with sting activity
WO2021163467A1 (en) Linking amino acid sequences, manufacturing method thereof, and use thereof

Legal Events

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

Ref document number: 21823700

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21823700

Country of ref document: EP

Kind code of ref document: A1