WO2020051356A1 - Imidazoquinoline compounds and uses thereof - Google Patents

Imidazoquinoline compounds and uses thereof Download PDF

Info

Publication number
WO2020051356A1
WO2020051356A1 PCT/US2019/049784 US2019049784W WO2020051356A1 WO 2020051356 A1 WO2020051356 A1 WO 2020051356A1 US 2019049784 W US2019049784 W US 2019049784W WO 2020051356 A1 WO2020051356 A1 WO 2020051356A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
alkyl
subject
Prior art date
Application number
PCT/US2019/049784
Other languages
French (fr)
Inventor
Lihu Yang
Original Assignee
Birdie Biopharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Birdie Biopharmaceuticals, Inc. filed Critical Birdie Biopharmaceuticals, Inc.
Priority to ES19858183T priority Critical patent/ES2963112T3/en
Priority to CN201980068724.0A priority patent/CN113164460A/en
Priority to CA3111786A priority patent/CA3111786A1/en
Priority to US17/274,136 priority patent/US20210214354A1/en
Priority to JP2021512742A priority patent/JP2022501327A/en
Priority to EP19858183.7A priority patent/EP3846807B1/en
Priority to KR1020217010120A priority patent/KR20210074290A/en
Priority to EP23188546.8A priority patent/EP4306523A3/en
Priority to AU2019335366A priority patent/AU2019335366A1/en
Publication of WO2020051356A1 publication Critical patent/WO2020051356A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents

Definitions

  • TLR7 and TLR8 are toll-like receptors 7 and 8 respectively, and they lie in close proximity to each other on the human X chromosome. Both TLR7 and TLR8 recognize single-stranded RNA of viruses such as HIV and HCV. TLR7 has been shown to play a significant role in the pathogenesis of autoimmune disorders such as Systemic Lupus Erythematosus (SLE) as well as in antiviral immunity regulation. Genetic variants in TLR8 have recently been linked to susceptibility to pulmonary tuberculosis.
  • SLE Systemic Lupus Erythematosus
  • TLR7 is functional both in human and mouse, while TLR8 is only functional in human, but it seems to counteract TLR7 activity.
  • TLR7/8 agonists as immune response enhancers is their simultaneous stimulation of several cell types.
  • TLR7 and TLR8 are expressed mostly on immune cells such as antigen presenting cells, including plasmacytoid dendritic cells (pDC) and myeloid dendritic cells (mDC), as well as natural killer cells, and macrophages.
  • pDC plasmacytoid dendritic cells
  • mDC myeloid dendritic cells
  • TLR7/8 activation on pDCs and mDCs results in induction and release of type I interferons (IFN), tumor necrosis factor alpha (TNFa), and interleukin 12 (IL-12), which is an important step for the initiation of innate and adaptive immunities to kill cancer cells. Accordingly, there is a need to develop small molecule agonists of TLR7 and TLR8 as both antiviral and antitumor compounds.
  • IFN interferons
  • TNFa tumor necrosis factor alpha
  • IL-12 interleukin 12
  • the compounds are imidazoquinoline derivatives, or pharmaceutically acceptable salts thereof.
  • the compounds are agonists of toll-like receptors 7 and 8 (TLR7/8).
  • the compounds may be used to treat viral infection (e.g., hepatitis C (HCV)), cancer (e.g., a HER2 positive cancer), allergic disease, or a combination thereof, in a subject need thereof by administering a therapeutically effective amount of an imidazoquinoline derivative provided herein to the subject.
  • viral infection e.g., hepatitis C (HCV)
  • cancer e.g., a HER2 positive cancer
  • allergic disease e.g., allergic disease, or a combination thereof
  • R 2 is— 0-(C 3 -s cycloalkyl),— 0-(Ci_ 3 alkyl),— 0-(Ci_ 3 haloalkyl),— S-(Ci_ 3 alkyl),— S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl); and n is 0, 1 , 2, 3, or 4.
  • Some embodiments include a pharmaceutical dosage form comprising a compound described herein.
  • Compounds or compositions described herein may be used for activating TLR7/8.
  • compounds or compositions described herein may be used as adjuvants in cancer vaccine or adoptive T cell transfer protocols.
  • Some embodiments include a method of treating a disease or disorder associated with a TLR7/8 agonist comprising administering an effective amount of a compound described herein to a mammal in need thereof.
  • Some embodiments include use of a compound described herein in the manufacture of a medicament for the treatment of a disease or disorder associated with a TLR7/8 agonist.
  • Fig. 1 shows PK profiles in rats as mean concentration versus time curves for TLR7/8 agonists as described in Example 15. DETAILED DESCRIPTION
  • the article“a” or“an” refers to one or to more than one (i.e. to at least one) of the grammatical object of the article.
  • “an element” means one element or more than one element.
  • use of the term“including” as well as other forms, such as“include”,“includes,” and“included,” is not limiting.
  • administering refers to administration of the compounds provided herein to a cell or a subject as needed to achieve the desired effect.
  • alkyl by itself or as part of another substituent means, unless otherwise stated, a straight or branched chain hydrocarbon having the number of carbon atoms designated (i.e., Ci_ 3 means one to three carbon atoms) and includes straight or branched chain substituent groups.
  • composition or“pharmaceutical composition” refers to a mixture of at least one compound— useful as described herein— with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary, rectal, subcutaneous, and topical administration.
  • controlling the disease or disorder is used to mean changing the activity of one or more kinases to affect the disease or disorder.
  • cycloalkyl refers to a mono cyclic non-aromatic radical, wherein each of the atoms forming the ring (i.e., skeletal atoms) is a carbon atom.
  • the cycloalkyl group is saturated or partially unsaturated.
  • Cycloalkyl groups include groups having 3 to 5 ring atoms (C 3 -s cycloalkyl).
  • the term“disease or disorder associated with kinase activity” refers to a disease, condition or disorder treatable, in whole or in part, by inhibition of one or more kinases.
  • the term“effective amount,”“pharmaceutically effective amount” or “therapeutically effective amount” refers to a nontoxic but sufficient dosage amount of an agent (e.g., the compounds or compositions provided herein) to provide the desired biological result, which result may be reduction or alleviation, or both, of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system including influencing, reducing or inhibiting the activity of or preventing activation of a kinase (e.g., modulating kinase activity).
  • An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.
  • these terms as used herein may also refer to an amount effective at bringing about a desired in vivo effect in an animal— where in some embodiments, the animal is a human— including, but not limited to, uveitis, reduction in intraocular pressure, or dry eye.
  • excipient refers to physiologically compatible additives useful in preparation of a pharmaceutical composition.
  • examples of pharmaceutically acceptable carriers and excipients can, for example, be found in Remington Pharmaceutical Science, 16 th Ed.
  • haloalkyl refers to an alkyl group independently substituted with one or more fluorine, chlorine, bromine, or iodine atoms. In some embodiments, the alkyl group is independently substituted with one or more fluorine, chlorine, or bromine atoms. In some embodiments, the alkyl group is independently substituted with one or more fluorine or chlorine atoms.
  • the term“subject,”“patient” or“individual” refers to a human or a non-human mammal.
  • Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline, and murine mammals.
  • the patient, subject, or individual is human.
  • the term“pharmaceutically acceptable” refers to a material that does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e. the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the invention within or to the patient such that it may perform its intended function.
  • a pharmaceutically acceptable material, composition or carrier such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the invention within or to the patient such that it may perform its intended function.
  • Such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be“acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound useful within the invention, and not injurious to the patient.
  • pharmaceutically acceptable carrier also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound useful within the invention, and are physiologically acceptable to the patient.
  • pharmaceutically acceptable carrier may further include a pharmaceutically acceptable salt of the compound useful within the invention.
  • Other additional ingredients that may be included in the pharmaceutical compositions used in the practice of the invention are described, for example, in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, Pa.), which is incorporated herein by reference.
  • the “pharmaceutically acceptable carrier” is useful for the preparation of a pharmaceutical composition that is: generally compatible with the other ingredients of the composition, not deleterious to the recipient, and neither biologically nor otherwise undesirable.
  • a pharmaceutically acceptable carrier includes one or more than one carrier. Embodiments include carriers for topical, ocular, parenteral, intravenous, intraperitoneal intramuscular, sublingual, nasal or oral administration.“Pharmaceutically acceptable carrier” also includes agents for preparation of aqueous dispersions and sterile powders for injection or dispersions.
  • the term“pharmaceutically acceptable salt” refers to derivatives of the compounds provided herein wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the compounds provided herein include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the compounds provided herein can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by combining 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; generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile may be used.
  • nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile may be used.
  • Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17 th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.
  • the term“prevent” or“prevention” refers to no disorder or disease development if none had occurred, or no further disorder or disease development if there had already been development of the disorder or disease. Also considered is the ability of one to prevent some or all of the symptoms associated with the disorder or disease.
  • treatment refers to the application or administration of a therapeutic agent, i.e. a compound provided herein, to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g., for diagnosis or ex vivo applications), who has a disease, a symptom of the disease or the potential to develop the disease, with the purpose to heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of the disease, or the potential to develop the disease.
  • a therapeutic agent i.e. a compound provided herein
  • an isolated tissue or cell line from a patient (e.g., for diagnosis or ex vivo applications)
  • Such treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics.
  • the compound is a compound of Table 1 , or a pharmaceutically acceptable salt thereof.
  • R 1 is, independently, F, Cl, Br, or I;
  • R 2 is— 0-(C 3 -s cycloalkyl),— 0-(Ci_ 3 alkyl),— 0-(Ci_ 3 haloalkyl),— S-(Ci_ 3 alkyl),— S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl); and n is 0, 1 , 2, 3, or 4.
  • the compound of Formula (I) is a compound of Formula (II):
  • the compound of Formula (I) is a compound of Formula (III):
  • R 1 is F, Cl, Br, or I. In some embodiments, R 1 is F. In some embodiments, R 1 is H or F. In some embodiments, R 1 is H (i.e. n is 0). In some embodiments, R 1 is H, F, Cl, Br, or I.
  • R 2 is — 0-(C 3 -s cycloalkyl), — 0-(Ci_ 3 alkyl), — 0-(Ci_ 3 monohaloalkyl),— 0-(Ci_ 3 dihaloalkyl),— 0-(Ci_ 3 trihaloalkyl),— S-(Ci_ 3 alkyl),— S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl).
  • R 2 is— 0-CF 3 , — 0-CH 2 -CF 3 ,— O- cyclopropyl, — S-methyl, — S-ethyl, — S(0)-methyl, — S(0)-ethyl, — S(0 2 )-methyl, or — S(0 2 )-ethyl.
  • the haloalkyl halogen is independently F, Cl, Br, or I. In some embodiments the haloalkyl halogen is F.
  • R 2 is— 0-(C 3-5 cycloalkyl),— 0-(Ci_ 3 alkyl),— S-(Ci_ 3 alkyl), — S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl). In some embodiments, R 2 is— 0-(C 3-5 cycloalkyl), — S-(Ci_ 3 alkyl),— S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl).
  • R 2 is— O- cyclopropyl, — S-methyl, — S-ethyl, — S(0)-methyl, — S(0)-ethyl, — S(0 2 )-methyl, or — S(0 2 )-ethyl.
  • R 2 is— 0-(C 3-5 cycloalkyl). In some embodiments, R 2 is— O- (C 3 -5 cycloalkyl), and R 1 is F, Cl, Br, or I. In some embodiments, R 2 is— 0-(C 3-5 cycloalkyl), and R 1 is H or F. In some embodiments, R 2 is— 0-(C 3-5 cycloalkyl), and R 1 is H. In some embodiments, R 2 is— 0-(C 3-5 cycloalkyl), and R 1 is F.
  • R 2 is — 0-(C 3-5 cycloalkyl), — S-(Ci_ 3 alkyl), — S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl), and R 1 is F, Cl, Br, or I.
  • R 2 is— 0-(C 3-5 cycloalkyl),— S-(Ci_ 3 alkyl),— S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl), and R 1 is H or F.
  • R 2 is— 0-(C 3-5 cycloalkyl),— S-(Ci_ 3 alkyl),— S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl), and R 1 is F.
  • R 2 is— 0-(C 3 -s cycloalkyl),— S-(Ci_ 3 alkyl),— S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl), and R 1 is H.
  • R 2 is— S-(Ci_ 3 alkyl), — S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl), and R 1 is F, Cl, Br, or I.
  • R 2 is— S-(Ci_ 3 alkyl),— S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl), and R 1 is H or F.
  • R 2 is— S-(Ci_ 3 alkyl), — S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl), and R 1 is F.
  • R 2 is— S-(Ci_ 3 alkyl),— S(0)(Ci_ 3 alkyl), or— S(0 2 )(Ci_ 3 alkyl), and R 1 is H.
  • R 2 is— O-cyclopropyl,— S-methyl,— S-ethyl,— S(0)-methyl, — S(0)-ethyl, — S(0 2 )-methyl, or — S(0 2 )-ethyl, and R 1 is F, Cl, Br, or I.
  • R 2 is— O-cyclopropyl,— S-methyl,— S-ethyl,— S(0)-methyl,— S(0)-ethyl,— S(0 2 )-methyl, or— S(0 2 )-ethyl, and R 1 is H or F.
  • R 2 is — O- cyclopropyl, — S-methyl, — S-ethyl, — S(0)-methyl, — S(0)-ethyl, — S(0 2 )-methyl, or — S(0 2 )-ethyl, and R 1 is H.
  • R 2 is— O-cyclopropyl,— S-methyl,— S- ethyl,— S(0)-methyl,— S(0)-ethyl,— S(0 2 )-methyl, or— S(0 2 )-ethyl, and R 1 is F.
  • n is 0. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 1 , 2, 3, or 4. In some embodiments, n is 0 or 1.
  • the compound is:
  • the compound is:
  • the compound is:
  • the compound is:
  • the compound is:
  • the compound is:
  • the compound is: or a pharmaceutically acceptable salt thereof.
  • the compound is:
  • the compound is:
  • the compound is:
  • Compounds described herein also include isotopically-labeled compounds wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes suitable for inclusion in the compounds described herein include and are not limited to 2 H, 3 H, 11 C, 13 C, 14 C, 36 CI, 18 F, 123 l, 125 l, 13 N, 15 N, 15 0, 17 0, 18 0, 32 P, and 35 S.
  • isotopically-labeled compounds are useful in drug and/or substrate tissue distribution studies.
  • substitution with heavier isotopes such as deuterium affords greater metabolic stability (for example, increased in vivo half-life or reduced dosage requirements).
  • substitution with positron emitting isotopes, such as 11 C, 18 F, 15 0 and 13 N is useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
  • Isotopically-labeled compounds are prepared by any suitable method or by processes using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.
  • compounds described herein are labeled by other means, including, but not limited to, use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
  • compositions comprising a compound provided herein (i.e., a compound of Table 1 or a pharmaceutically acceptable salt thereof).
  • compositions comprising a compound provided herein.
  • a cancer in another aspect, comprising administering a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof) to the subject.
  • the cancer is a HER2 positive cancer.
  • the cancer is esophageal, stomach, colon, rectal, pancreatic, lung, breast, cervix uteri, corpus uteri, ovary, bladder, head and neck, endometrial, osteosarcoma, prostate, neuroblastoma, or a combination thereof.
  • kits for treating viral infection, cancer, an allergic disease, or a combination thereof, in a subject in need thereof comprising administering a compound provided herein (i.e. a compound of Table 1 or a
  • kits for treating viral infection comprising administering a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof) to the subject.
  • kits for treating cancer comprising administering a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof) to the subject.
  • a compound provided herein i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof
  • a compound provided herein i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof
  • the viral infection comprises a hepatitis C viral (HCV) infection.
  • HCV hepatitis C viral
  • provided herein is a method of modulating kinase activity in a cell, comprising contacting the cell with an amount effective to modulate kinase activity of a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof).
  • the subject is a human.
  • a compound provided herein i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof
  • a composition provided herein or a pharmaceutical composition provided herein, in the manufacture of a
  • compositions comprising a compound provided herein and a pharmaceutically acceptable carrier.
  • Actual dosage levels of an active ingredient in the pharmaceutical compositions provided herein may be varied so as to obtain an amount of the active ingredient that is effective to achieve a desired therapeutic response for a particular subject, composition, or mode of administration, without being toxic to the subject.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle.
  • the dosage unit forms of the present disclosure are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding/formulating such a therapeutic compound for the treatment of the diseases referred to herein in a subject in need thereof.
  • the compounds or compositions provided herein are formulated using one or more pharmaceutically acceptable excipients or carriers.
  • the pharmaceutical compositions provided herein comprise a therapeutically effective amount of a compound provided herein and a pharmaceutically acceptable carrier.
  • the present disclosure provides packaged pharmaceutical compositions comprising a container holding at least one therapeutically effective amount of a compound provided herein, and instructions for using the compound to treat one or more symptoms of a disease referred to herein in a subject in need thereof.
  • Routes of administration of any of the compositions provided herein include oral, nasal, rectal, intravaginal, parenteral, buccal, sublingual, topical, or ocular.
  • the compounds for use as provided herein may be formulated for administration by any suitable route, such as for ocular, oral or parenteral, for example, transdermal, transmucosal (e.g., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and perivaginally), (intra)nasal and (trans) rectal), intravesical, intrapulmonary, intraduodenal, intragastrical, intrathecal, subcutaneous, intramuscular, intradermal, intra-arterial, intravenous, intrabronchial, inhalation, and topical administration.
  • compositions and dosage forms include, for example, drops, tablets, capsules, caplets, pills, gel caps, troches, dispersions, suspensions, solutions, syrups, granules, beads, transdermal patches, gels, powders, pellets, magmas, lozenges, creams, pastes, plasters, lotions, discs, suppositories, liquid sprays for nasal or oral administration, dry powder or aerosolized formulations for inhalation, compositions and formulations for ocular or intravesical administration and the like. It should be understood that the formulations and compositions that would be useful as provided herein are not limited to the particular formulations and compositions that are described herein.
  • dosage forms suitable for administration to a subject in need thereof comprising a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof).
  • kits comprising a composition including a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof) and instructions for use thereof.
  • the kit further includes one or more of a syringe, a vial, or a dosage form.
  • NMR spectra and 13 C-NMR are recorded on a Varian Mercury-VX400 instrument operating at 400 MHZ.
  • NMR spectra are obtained as CDCI 3 solutions (reported in ppm), using chloroform as the reference standard (7.27 ppm for 1 H and 77.00 ppm for 13 C), CD 3 OD (3.4 and 4.8 ppm for 1 H and 49.3 ppm for 13 C), DMSO-d s (2.49 ppm for 1 H), or internal tetramethylsilane (0.00 ppm for 1 H) when appropriate.
  • Other NMR solvents are used as needed.
  • SPF SD rats were purchased from BioLASCO Taiwan Co. Ltd. Animals were randomly assigned to five groups with 3 males and 3 females in each group before dosing. The body weight variation of animals used were in an interval within ⁇ 20 percent of the mean weight for each gender. The basic design was as shown in Table 2.
  • Blood samples (200-350 pL/time point) were obtained through tail vein at pre-dose and 2 min, 5 min, 15 min, 30 min, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr, 12 hr and 24 hr post dosing.
  • the blood samples were collected into tubes containing K 2 EDTA and maintained on ice no more than 30 minutes, then centrifuged (1500-1750 xg, 15 min, 2-8 °C) for plasma harvest.
  • Resiquimod is a Toll-like receptor (TLR) agonist that activates the immune system.
  • TLR Toll-like receptor
  • Example 16 TLR7/8 Agonist Potency Assay in HEK-Blue hTLR7 Cell
  • HEK-Blue hTLR7 cells were seeded at 3x10 4 cell/well and treated with nine concentrations of positive control (CL264) and three compounds (resiquimod, compound 2, compound 3).
  • HEK-Blue TLR7 cells can serve to measure the bioactivity of TLR7 through the secretion of embryonic alkaline phosphatase (SEAP) upon NF-kB activation following TLR7 stimulation. Water was used as negative control. After about 16 hrs incubation at 37 °C in 5% C0 2 , the SEAP was determined by using a spectrophotometer at 635 nm.
  • SEAP embryonic alkaline phosphatase
  • Example 17 TLR7/8 Agonist Potency Assay in HEK-Blue hTLR8 Cell
  • HEK-Blue hTLR8 cells were seeded at 3x10 4 cell/well and treated with nine concentrations of positive control (ssRNA40) and four test articles (resiquimod, compound 2, compound 3).
  • HEK-Blue hTLR8 cells can serve to measure the bioactivity of TLR8 through the secretion of embryonic alkaline phosphatase (SEAP) upon NF-kB activation following TLR8 stimulation. Water was used as negative control. After about 16 hrs incubation at 37 °C in 5% C0 2 , the SEAP was determined by using a spectrophotometer at 635 nm.
  • SEAP embryonic alkaline phosphatase

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Pulmonology (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

Provided herein are imidazoquinoline derivatives, or pharmaceutically acceptable salts thereof, that are agonists of toll-like receptors 7 and 8 (TLR7/8). Also provided herein are compositions and pharmaceutical compositions comprising the imidazoquinoline derivatives, or pharmaceutically acceptable salts thereof, provided herein. Also provided herein are methods of use of the imidazoquinoline derivatives, or pharmaceutically acceptable salts thereof, provided herein to treat various diseases, such as viral, cancer, and allergic diseases, in a subject in need thereof by administering a therapeutically effective amount of the imidazoquinoline derivative, or pharmaceutically acceptable salt thereof, to the subject.

Description

IMIDAZOQUINOLINE COMPOUNDS AND USES THEREOF
RELATED APPLICATIONS
[1] This application claims the benefit of U.S. Provisional Patent Application No.
62/728,556, filed September 7, 2018, the entire content of which is incorporated herein by reference.
BACKGROUND
[2] Toll-like receptors (TLRs) play a fundamental role in pathogen recognition and activation of innate immunity. TLR7 and TLR8 are toll-like receptors 7 and 8 respectively, and they lie in close proximity to each other on the human X chromosome. Both TLR7 and TLR8 recognize single-stranded RNA of viruses such as HIV and HCV. TLR7 has been shown to play a significant role in the pathogenesis of autoimmune disorders such as Systemic Lupus Erythematosus (SLE) as well as in antiviral immunity regulation. Genetic variants in TLR8 have recently been linked to susceptibility to pulmonary tuberculosis. TLR7 is functional both in human and mouse, while TLR8 is only functional in human, but it seems to counteract TLR7 activity. One benefit of TLR7/8 agonists as immune response enhancers is their simultaneous stimulation of several cell types. TLR7 and TLR8 are expressed mostly on immune cells such as antigen presenting cells, including plasmacytoid dendritic cells (pDC) and myeloid dendritic cells (mDC), as well as natural killer cells, and macrophages. TLR7/8 activation on pDCs and mDCs results in induction and release of type I interferons (IFN), tumor necrosis factor alpha (TNFa), and interleukin 12 (IL-12), which is an important step for the initiation of innate and adaptive immunities to kill cancer cells. Accordingly, there is a need to develop small molecule agonists of TLR7 and TLR8 as both antiviral and antitumor compounds.
SUMMARY
[3] Provided herein are compounds that are imidazoquinoline derivatives, or pharmaceutically acceptable salts thereof. The compounds are agonists of toll-like receptors 7 and 8 (TLR7/8). The compounds may be used to treat viral infection (e.g., hepatitis C (HCV)), cancer (e.g., a HER2 positive cancer), allergic disease, or a combination thereof, in a subject need thereof by administering a therapeutically effective amount of an imidazoquinoline derivative provided herein to the subject.
[4] In one aspect, provided herein are compounds of Formula (I):
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof, wherein each occurrence of R1 is, independently, F, Cl, Br, or I;
R2 is— 0-(C3-s cycloalkyl),— 0-(Ci_3 alkyl),— 0-(Ci_3 haloalkyl),— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl); and n is 0, 1 , 2, 3, or 4.
[5] Some embodiments include a pharmaceutical dosage form comprising a compound described herein. Compounds or compositions described herein may be used for activating TLR7/8. In addition to use as a stand-alone immunotherapeutic agent, such as an antiviral and anticancer agent, compounds or compositions described herein may be used as adjuvants in cancer vaccine or adoptive T cell transfer protocols.
[6] Some embodiments include a method of treating a disease or disorder associated with a TLR7/8 agonist comprising administering an effective amount of a compound described herein to a mammal in need thereof.
[7] Some embodiments include use of a compound described herein in the manufacture of a medicament for the treatment of a disease or disorder associated with a TLR7/8 agonist.
BRIEF DESCRIPTION OF THE DRAWINGS
[8] Fig. 1 shows PK profiles in rats as mean concentration versus time curves for TLR7/8 agonists as described in Example 15. DETAILED DESCRIPTION
[9] Listed below are definitions of various terms used in the present disclosure. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group.
[10] As used herein, the article“a” or“an” refers to one or to more than one (i.e. to at least one) of the grammatical object of the article. By way of example,“an element” means one element or more than one element. Furthermore, use of the term“including” as well as other forms, such as“include”,“includes,” and“included,” is not limiting.
[11] As used herein, the term“administering” refers to administration of the compounds provided herein to a cell or a subject as needed to achieve the desired effect.
[12] As used herein, the term“alkyl,” by itself or as part of another substituent means, unless otherwise stated, a straight or branched chain hydrocarbon having the number of carbon atoms designated (i.e., Ci_3 means one to three carbon atoms) and includes straight or branched chain substituent groups.
[13] As used herein, the term“composition” or“pharmaceutical composition” refers to a mixture of at least one compound— useful as described herein— with a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary, rectal, subcutaneous, and topical administration.
[14] As used herein, the term “controlling the disease or disorder” is used to mean changing the activity of one or more kinases to affect the disease or disorder.
[15] As used herein, the term“cycloalkyl” refers to a mono cyclic non-aromatic radical, wherein each of the atoms forming the ring (i.e., skeletal atoms) is a carbon atom. In one embodiment, the cycloalkyl group is saturated or partially unsaturated. Cycloalkyl groups include groups having 3 to 5 ring atoms (C3-s cycloalkyl).
[16] As used herein, the term“disease or disorder associated with kinase activity” refers to a disease, condition or disorder treatable, in whole or in part, by inhibition of one or more kinases.
[17] As used herein, the term“effective amount,”“pharmaceutically effective amount” or “therapeutically effective amount” refers to a nontoxic but sufficient dosage amount of an agent (e.g., the compounds or compositions provided herein) to provide the desired biological result, which result may be reduction or alleviation, or both, of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system including influencing, reducing or inhibiting the activity of or preventing activation of a kinase (e.g., modulating kinase activity). An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation. These terms as used herein may also refer to an amount effective at bringing about a desired in vivo effect in an animal— where in some embodiments, the animal is a human— including, but not limited to, uveitis, reduction in intraocular pressure, or dry eye.
[18] As used herein, the term “excipient” refers to physiologically compatible additives useful in preparation of a pharmaceutical composition. Examples of pharmaceutically acceptable carriers and excipients can, for example, be found in Remington Pharmaceutical Science, 16th Ed.
[19] As used herein, the term “haloalkyl” refers to an alkyl group independently substituted with one or more fluorine, chlorine, bromine, or iodine atoms. In some embodiments, the alkyl group is independently substituted with one or more fluorine, chlorine, or bromine atoms. In some embodiments, the alkyl group is independently substituted with one or more fluorine or chlorine atoms.
[20] As used herein, the term“subject,”“patient” or“individual” refers to a human or a non-human mammal. Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline, and murine mammals. In some embodiments, the patient, subject, or individual is human.
[21] As used herein, the term“pharmaceutically acceptable” refers to a material that does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e. the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
[22] As used herein, the term “pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound useful within the invention within or to the patient such that it may perform its intended function. Typically, such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be“acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound useful within the invention, and not injurious to the patient. As used herein, “pharmaceutically acceptable carrier” also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound useful within the invention, and are physiologically acceptable to the patient. The term “pharmaceutically acceptable carrier” may further include a pharmaceutically acceptable salt of the compound useful within the invention. Other additional ingredients that may be included in the pharmaceutical compositions used in the practice of the invention are described, for example, in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, Pa.), which is incorporated herein by reference. The “pharmaceutically acceptable carrier” is useful for the preparation of a pharmaceutical composition that is: generally compatible with the other ingredients of the composition, not deleterious to the recipient, and neither biologically nor otherwise undesirable. “A pharmaceutically acceptable carrier” includes one or more than one carrier. Embodiments include carriers for topical, ocular, parenteral, intravenous, intraperitoneal intramuscular, sublingual, nasal or oral administration.“Pharmaceutically acceptable carrier” also includes agents for preparation of aqueous dispersions and sterile powders for injection or dispersions.
[23] As used herein, the term“pharmaceutically acceptable salt” refers to derivatives of the compounds provided herein wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the compounds provided herein include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the compounds provided herein can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by combining 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; generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile may be used. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety. [24] As used herein, the term“prevent” or“prevention” refers to no disorder or disease development if none had occurred, or no further disorder or disease development if there had already been development of the disorder or disease. Also considered is the ability of one to prevent some or all of the symptoms associated with the disorder or disease.
[25] As used herein, the term “treatment” or “treating” refers to the application or administration of a therapeutic agent, i.e. a compound provided herein, to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g., for diagnosis or ex vivo applications), who has a disease, a symptom of the disease or the potential to develop the disease, with the purpose to heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of the disease, or the potential to develop the disease. Such treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics.
COMPOUNDS
[26] In one aspect, provided herein are compounds that are imidazoquinoline derivatives. In one embodiment, the compound is a compound of Table 1 , or a pharmaceutically acceptable salt thereof.
Table 1.
Figure imgf000008_0001
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0003
[27] In another aspect, provided herein are compounds of Formula (I):
Figure imgf000011_0001
or a pharmaceutically acceptable salt thereof, wherein each occurrence of R1 is, independently, F, Cl, Br, or I;
R2 is— 0-(C3-s cycloalkyl),— 0-(Ci_3 alkyl),— 0-(Ci_3 haloalkyl),— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl); and n is 0, 1 , 2, 3, or 4.
[28] In some embodiments, the compound of Formula (I) is a compound of Formula (II):
Figure imgf000011_0002
(II) or a pharmaceutically acceptable salt thereof.
[29] In some embodiments, the compound of Formula (I) is a compound of Formula (III):
Figure imgf000012_0001
or a pharmaceutically acceptable salt thereof.
[30] In some embodiments, R1 is F, Cl, Br, or I. In some embodiments, R1 is F. In some embodiments, R1 is H or F. In some embodiments, R1 is H (i.e. n is 0). In some embodiments, R1 is H, F, Cl, Br, or I.
[31] In some embodiments, R2 is — 0-(C3-s cycloalkyl), — 0-(Ci_3 alkyl), — 0-(Ci_3 monohaloalkyl),— 0-(Ci_3 dihaloalkyl),— 0-(Ci_3 trihaloalkyl),— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl). In some embodiments, R2 is— 0-CF3, — 0-CH2-CF3,— O- cyclopropyl, — S-methyl, — S-ethyl, — S(0)-methyl, — S(0)-ethyl, — S(02)-methyl, or — S(02)-ethyl. In some embodiments the haloalkyl halogen is independently F, Cl, Br, or I. In some embodiments the haloalkyl halogen is F.
[32] In some embodiments, R2 is— 0-(C3-5 cycloalkyl),— 0-(Ci_3 alkyl),— S-(Ci_3 alkyl), — S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl). In some embodiments, R2 is— 0-(C3-5 cycloalkyl), — S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl). In some embodiments, R2 is— O- cyclopropyl, — S-methyl, — S-ethyl, — S(0)-methyl, — S(0)-ethyl, — S(02)-methyl, or — S(02)-ethyl.
[33] In some embodiments, R2 is— 0-(C3-5 cycloalkyl). In some embodiments, R2 is— O- (C3-5 cycloalkyl), and R1 is F, Cl, Br, or I. In some embodiments, R2 is— 0-(C3-5 cycloalkyl), and R1 is H or F. In some embodiments, R2 is— 0-(C3-5 cycloalkyl), and R1 is H. In some embodiments, R2 is— 0-(C3-5 cycloalkyl), and R1 is F.
[34] In some embodiments, R2 is — 0-(C3-5 cycloalkyl), — S-(Ci_3 alkyl), — S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl), and R1 is F, Cl, Br, or I. In some embodiments, R2 is— 0-(C3-5 cycloalkyl),— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl), and R1 is H or F. In some embodiments, R2 is— 0-(C3-5 cycloalkyl),— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl), and R1 is F. In some embodiments, R2 is— 0-(C3-s cycloalkyl),— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl), and R1 is H.
[35] In some embodiments, R2 is— S-(Ci_3 alkyl), — S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl), and R1 is F, Cl, Br, or I. In some embodiments, R2 is— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl), and R1 is H or F. In some embodiments, R2 is— S-(Ci_3 alkyl), — S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl), and R1 is F. In some embodiments, R2 is— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl), and R1 is H.
[36] In some embodiments, R2 is— O-cyclopropyl,— S-methyl,— S-ethyl,— S(0)-methyl, — S(0)-ethyl, — S(02)-methyl, or — S(02)-ethyl, and R1 is F, Cl, Br, or I. In some embodiments, R2 is— O-cyclopropyl,— S-methyl,— S-ethyl,— S(0)-methyl,— S(0)-ethyl,— S(02)-methyl, or— S(02)-ethyl, and R1 is H or F. In some embodiments, R2 is — O- cyclopropyl, — S-methyl, — S-ethyl, — S(0)-methyl, — S(0)-ethyl, — S(02)-methyl, or — S(02)-ethyl, and R1 is H. In some embodiments, R2 is— O-cyclopropyl,— S-methyl,— S- ethyl,— S(0)-methyl,— S(0)-ethyl,— S(02)-methyl, or— S(02)-ethyl, and R1 is F.
[37] In some embodiments, n is 0. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 1 , 2, 3, or 4. In some embodiments, n is 0 or 1.
[38] In some embodiments, the compound is:
Figure imgf000013_0001
or a pharmaceutically acceptable salt thereof.
[39] In some embodiments, the compound is:
Figure imgf000013_0002
or a pharmaceutically acceptable salt thereof.
[40] In some embodiments, the compound is:
Figure imgf000014_0001
or a pharmaceutically acceptable salt thereof.
[41] In some embodiments, the compound is:
Figure imgf000014_0002
or a pharmaceutically acceptable salt thereof.
[42] In some embodiments, the compound is:
Figure imgf000014_0003
or a pharmaceutically acceptable salt thereof.
[43] In some embodiments, the compound is:
Figure imgf000014_0004
or a pharmaceutically acceptable salt thereof.
[44] In some embodiments, the compound is:
Figure imgf000015_0001
or a pharmaceutically acceptable salt thereof.
[45] In some embodiments, the compound is:
Figure imgf000015_0002
or a pharmaceutically acceptable salt thereof.
[46] In some embodiments, the compound is:
Figure imgf000015_0003
or a pharmaceutically acceptable salt thereof.
[47] In some embodiments, the compound is:
Figure imgf000015_0004
or a pharmaceutically acceptable salt thereof.
[48] Compounds described herein also include isotopically-labeled compounds wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds described herein include and are not limited to 2H, 3H, 11C, 13C, 14C, 36CI, 18F, 123l, 125l, 13N, 15N, 150, 170, 180, 32P, and 35S. In one embodiment, isotopically-labeled compounds are useful in drug and/or substrate tissue distribution studies. In another embodiment, substitution with heavier isotopes such as deuterium affords greater metabolic stability (for example, increased in vivo half-life or reduced dosage requirements). In yet another embodiment, substitution with positron emitting isotopes, such as 11C, 18F, 150 and 13N, is useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds are prepared by any suitable method or by processes using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.
[49] In one embodiment, compounds described herein are labeled by other means, including, but not limited to, use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
[50] The compounds described herein are synthesized using techniques and materials described herein and as described, for example, in Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplements (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989), March, Advanced Organic Chemistry 4th Ed., (Wiley 1992); Carey and Sundberg, Advanced Organic Chemistry 4th Ed., Vols. A and B (Plenum 2000, 2001), and Greene and Wuts, Protective Groups in Organic Synthesis 3rd Ed., (Wiley 1999) (all of which are incorporated by reference for such disclosure). General methods for the preparation of compound as described herein are modified by the use of appropriate reagents and conditions, for the introduction of the various moieties found in the formula as provided herein.
[51] Also provided herein are compositions comprising a compound provided herein (i.e., a compound of Table 1 or a pharmaceutically acceptable salt thereof).
[52] Also provided herein are pharmaceutical compositions, comprising a compound provided herein.
METHODS
[53] In another aspect, provided herein are methods of treating a cancer in a subject in need thereof, comprising administering a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof) to the subject. In some embodiments, the cancer is a HER2 positive cancer. [54] In some embodiments, the cancer is esophageal, stomach, colon, rectal, pancreatic, lung, breast, cervix uteri, corpus uteri, ovary, bladder, head and neck, endometrial, osteosarcoma, prostate, neuroblastoma, or a combination thereof.
[55] In another aspect, provided herein are methods of treating viral infection, cancer, an allergic disease, or a combination thereof, in a subject in need thereof, comprising administering a compound provided herein (i.e. a compound of Table 1 or a
pharmaceutically acceptable salt thereof) to the subject.
[56] In another aspect, provided herein are methods of treating viral infection, in a subject in need thereof, comprising administering a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof) to the subject.
[57] In another aspect, provided herein are methods of treating cancer, in a subject in need thereof, comprising administering a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof) to the subject.
[58] In another aspect, provided herein are methods of treating an allergic disease, in a subject in need thereof, comprising administering a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof) to the subject.
[59] In some embodiments, the viral infection comprises a hepatitis C viral (HCV) infection.
[60] In some embodiments, provided herein is a method of modulating kinase activity in a cell, comprising contacting the cell with an amount effective to modulate kinase activity of a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof).
[61] In some embodiments of these methods, the subject is a human.
[62] In another aspect, provided herein are uses of a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof), a composition provided herein, or a pharmaceutical composition provided herein, in the manufacture of a
medicament for the treatment of a viral infection, a cancer, or an allergic disease.
[63] In another aspect, provided herein are pharmaceutical compositions comprising a compound provided herein and a pharmaceutically acceptable carrier. [64] Actual dosage levels of an active ingredient in the pharmaceutical compositions provided herein may be varied so as to obtain an amount of the active ingredient that is effective to achieve a desired therapeutic response for a particular subject, composition, or mode of administration, without being toxic to the subject.
[65] In some embodiments, it is especially advantageous to formulate the compound in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle. The dosage unit forms of the present disclosure are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding/formulating such a therapeutic compound for the treatment of the diseases referred to herein in a subject in need thereof.
[66] In one embodiment, the compounds or compositions provided herein are formulated using one or more pharmaceutically acceptable excipients or carriers. In one embodiment, the pharmaceutical compositions provided herein comprise a therapeutically effective amount of a compound provided herein and a pharmaceutically acceptable carrier.
[67] In one embodiment, the present disclosure provides packaged pharmaceutical compositions comprising a container holding at least one therapeutically effective amount of a compound provided herein, and instructions for using the compound to treat one or more symptoms of a disease referred to herein in a subject in need thereof.
[68] Routes of administration of any of the compositions provided herein include oral, nasal, rectal, intravaginal, parenteral, buccal, sublingual, topical, or ocular. The compounds for use as provided herein may be formulated for administration by any suitable route, such as for ocular, oral or parenteral, for example, transdermal, transmucosal (e.g., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and perivaginally), (intra)nasal and (trans) rectal), intravesical, intrapulmonary, intraduodenal, intragastrical, intrathecal, subcutaneous, intramuscular, intradermal, intra-arterial, intravenous, intrabronchial, inhalation, and topical administration.
[69] Suitable compositions and dosage forms include, for example, drops, tablets, capsules, caplets, pills, gel caps, troches, dispersions, suspensions, solutions, syrups, granules, beads, transdermal patches, gels, powders, pellets, magmas, lozenges, creams, pastes, plasters, lotions, discs, suppositories, liquid sprays for nasal or oral administration, dry powder or aerosolized formulations for inhalation, compositions and formulations for ocular or intravesical administration and the like. It should be understood that the formulations and compositions that would be useful as provided herein are not limited to the particular formulations and compositions that are described herein.
[70] In another aspect, provided herein are dosage forms suitable for administration to a subject in need thereof, comprising a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof).
[71] In another aspect, provided herein are kits, comprising a composition including a compound provided herein (i.e. a compound of Table 1 or a pharmaceutically acceptable salt thereof) and instructions for use thereof. In some embodiments, the kit further includes one or more of a syringe, a vial, or a dosage form.
[72] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific procedures, embodiments, claims, and examples described herein. Such equivalents were considered to be within the scope of this disclosure and covered by the claims appended hereto. For example, it should be understood, that modifications in reaction conditions, including but not limited to reaction times, reaction size or volume, and experimental reagents, such as solvents, catalysts, pressures, atmospheric conditions, e.g., nitrogen atmosphere, and reducing or oxidizing agents, with art-recognized alternatives and using no more than routine experimentation, are within the scope of the present application.
[73] It is to be understood that wherever values and ranges are provided herein, all values and ranges encompassed by these values and ranges, are meant to be encompassed within the scope of the present disclosure. Moreover, all values that fall within these ranges, as well as the upper or lower limits of a range of values, are also contemplated by the present application.
[74] The following examples further illustrate aspects of the present disclosure. However, they are in no way a limitation of the teachings or present disclosure as set forth herein.
EXAMPLES
[75] In the synthetic schemes shown below, unless otherwise indicated all temperatures are set forth in degrees Celsius and all parts and percentages are by weight. Reagents and solvents are purchased from commercial suppliers and are used without further purification unless otherwise indicated. Anhydrous tetrahydrofuran (THF) and N,N-dimethylforamide (DMF) are purchased from commercial sources and used as received.
[76] The reactions set forth below are done generally under a positive pressure of argon or nitrogen at an ambient temperature (unless otherwise stated) in anhydrous solvents. Glassware is oven dried or heat dried. The reactions are assayed by TLC or analyzed by LC-MS and terminated as judged by the consumption of starting material. Analytical thin layer chromatography (TLC) is performed on glass plates pre-coated with silica gel 60 F254 0.25 mm plates (EM Science), and visualized with UV light (254 nm) or heating with commercial ethanolic phosphomolybdic acid. Preparative thin layer chromatography (TLC) is performed on glass-plates pre-coated with silica gel 60 F254 0.5 mm plates (20 x 20 cm, from commercial sources) and visualized with UV light (254 nm).
[77] Work-up of the reactions is typically done by doubling the reaction volume with the reaction solvent or extraction solvent and then washing with the indicated aqueous solutions using 25 % by volume of the extraction volume unless otherwise indicated. Product solutions are dried over anhydrous Na2S04 or Mg2S04 prior to filtration and evaporation of the solvents under reduced pressure on a rotary evaporator and noted as solvents removed in vacuo. Column chromatography is completed under positive pressure using 230-400 mesh silica gel.
[78] 1H -NMR spectra and 13C-NMR are recorded on a Varian Mercury-VX400 instrument operating at 400 MHZ. NMR spectra are obtained as CDCI3 solutions (reported in ppm), using chloroform as the reference standard (7.27 ppm for 1H and 77.00 ppm for 13C), CD3OD (3.4 and 4.8 ppm for 1H and 49.3 ppm for 13C), DMSO-ds (2.49 ppm for 1H), or internal tetramethylsilane (0.00 ppm for 1H) when appropriate. Other NMR solvents are used as needed.
Example 1 : Synthesis of common intermediate A.
Figure imgf000020_0001
common intermediate A
Scheme 1.
Example 2: Synthesis of common intermediate B.
Figure imgf000021_0001
common intermediate B
Scheme 2.
Example 3: Synthesis of Compound 1.
Figure imgf000021_0002
Scheme 3.
Example 4: Synthesis of Compound 2.
Figure imgf000021_0003
Scheme 4.
Example 5: Synthesis of Compound 3.
Figure imgf000021_0004
Scheme 5.
Example 6: Synthesis of Compound 4.
Figure imgf000022_0001
Scheme 6.
Example 7: Synthesis of Compound 5.
Figure imgf000022_0002
Scheme 7.
Example 8: Synthesis of Compound 6.
Figure imgf000022_0003
Scheme 8.
Example 9: Synthesis of Compound 10.
Figure imgf000022_0004
Scheme 9. Example 10: Synthesis of Compound 1 1.
Figure imgf000023_0001
Scheme 10.
Example 11 : Synthesis of Compound 12.
Figure imgf000023_0002
Scheme 1 1.
Example 12: Synthesis of Compound 13.
Figure imgf000023_0003
Scheme 12.
Example 13: Synthesis of Compound 14.
Figure imgf000024_0001
Scheme 13.
Example 14: Synthesis of Compound 15.
Figure imgf000024_0002
Scheme 14.
Example 15: TLR7/8 Agonist PK Study
[79] SPF SD rats were purchased from BioLASCO Taiwan Co. Ltd. Animals were randomly assigned to five groups with 3 males and 3 females in each group before dosing. The body weight variation of animals used were in an interval within ± 20 percent of the mean weight for each gender. The basic design was as shown in Table 2.
Table 2. TLR7/8 agonist PK Study Design
Figure imgf000024_0003
Figure imgf000025_0001
[80] Blood samples (200-350 pL/time point) were obtained through tail vein at pre-dose and 2 min, 5 min, 15 min, 30 min, 1 hr, 2 hr, 4 hr, 6 hr, 8 hr, 12 hr and 24 hr post dosing. The blood samples were collected into tubes containing K2EDTA and maintained on ice no more than 30 minutes, then centrifuged (1500-1750 xg, 15 min, 2-8 °C) for plasma harvest.
[81] The study samples were prepared and analyzed in parallel using liquid chromatography-tandem mass spectrometry. In pharmacokinetics evaluation following the determined concentrations in rat plasma, pharmacokinetic parameters were calculated using WinNonlin (Pharsight Corporation, version 6.3) with non-compartmental methods.
[82] Results are shown in Fig. 1 , and below in Table 3 (italicized values showed significant difference (P<0.05) when compared with the resiquimod group). Resiquimod is a Toll-like receptor (TLR) agonist that activates the immune system.
Table 3. Pharmacokinetic Parameters of TLR7/8 Agonists
Figure imgf000025_0002
Figure imgf000026_0001
Example 16: TLR7/8 Agonist Potency Assay in HEK-Blue hTLR7 Cell
[83] HEK-Blue hTLR7 cells were seeded at 3x104 cell/well and treated with nine concentrations of positive control (CL264) and three compounds (resiquimod, compound 2, compound 3). HEK-Blue TLR7 cells can serve to measure the bioactivity of TLR7 through the secretion of embryonic alkaline phosphatase (SEAP) upon NF-kB activation following TLR7 stimulation. Water was used as negative control. After about 16 hrs incubation at 37 °C in 5% C02, the SEAP was determined by using a spectrophotometer at 635 nm.
[84] The dose-response curves were fit with four parameter logistic curve (SigmaPlot, version 9.0). The calculated ECso-values for positive control (CL264) and the tested compounds are shown below in Table 4 (italicized values showed significant difference (P<0.05) when compared with the resiquimod group).
Table 4. TLR7 Agonistic EC5o of TLR7/8 Agonists
Figure imgf000026_0002
Example 17: TLR7/8 Agonist Potency Assay in HEK-Blue hTLR8 Cell [85] HEK-Blue hTLR8 cells were seeded at 3x104 cell/well and treated with nine concentrations of positive control (ssRNA40) and four test articles (resiquimod, compound 2, compound 3). HEK-Blue hTLR8 cells can serve to measure the bioactivity of TLR8 through the secretion of embryonic alkaline phosphatase (SEAP) upon NF-kB activation following TLR8 stimulation. Water was used as negative control. After about 16 hrs incubation at 37 °C in 5% C02, the SEAP was determined by using a spectrophotometer at 635 nm.
[86] The dose-response curves were fit with four parameter logistic curve (SigmaPlot, version 9.0). The calculated ECso-values for positive control (ssRNA40) and the tested compounds were shown below in Table 5 (italicized values showed significant difference (P<0.05) when compared with the resiquimod group).
Table 5. TLR8 Agonistic EC5o of TLR7/8 Agonists
Figure imgf000027_0001

Claims

CLAIMS What is claimed is:
1. A compound of Formula (I):
Figure imgf000028_0001
or a pharmaceutically acceptable salt thereof, wherein each occurrence of R1 is, independently, F, Cl, Br, or I;
R2 is— 0-(C3-s cycloalkyl),— 0-(Ci_3 haloalkyl),— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl); and n is 0, 1 , 2, 3, or 4.
2. The compound of claim 1 , wherein the compound of Formula (I) is a compound of Formula (II):
Figure imgf000028_0002
or a pharmaceutically acceptable salt thereof.
3. The compound of claim 1 , wherein R1 is F, Cl, Br, or I.
4. The compound of claim 1 , wherein R1 is F.
5. The compound of claim 1 , wherein R2 is — 0-(C3-s cycloalkyl), — 0-(Ci_3 monohaloalkyl),— 0-(Ci_3 dihaloalkyl),— 0-(Ci_3 trihaloalkyl),— S-(Ci_3 alkyl),— S(0)(Ci_3 alkyl), or— S(02)(Ci_3 alkyl).
6. The compound of claim 1 , wherein R2 is— 0-CF3,— 0-CH2-CF3,— O-cyclopropyl,— S-methyl,— S-ethyl,— S(0)-methyl,— S(0)-ethyl,— S(02)-methyl, or— S(02)-ethyl.
7. The compound of claim 1 , wherein n is 0.
8. The compound of claim 1 , wherein n is 1.
9. The compound of claim 1 , wherein n is 2.
10. The compound of claim 1 , wherein the compound is:
Figure imgf000029_0001
or a pharmaceutically acceptable salt thereof.
11. The compound of claim 1 , wherein the compound is:
Figure imgf000029_0002
or a pharmaceutically acceptable salt thereof.
12. The compound of claim 1 , wherein the compound is:
Figure imgf000029_0003
or a pharmaceutically acceptable salt thereof.
13. The compound of claim 1 , wherein the compound is
Figure imgf000030_0001
or a pharmaceutically acceptable salt thereof.
14. The compound of claim 1 , wherein the compound is:
Figure imgf000030_0002
or a pharmaceutically acceptable salt thereof.
15. The compound of claim 1 , wherein the compound is:
Figure imgf000030_0003
or a pharmaceutically acceptable salt thereof.
16. The compound of claim 1 , wherein the compound is:
Figure imgf000030_0004
or a pharmaceutically acceptable salt thereof.
17. The compound of claim 1 , wherein the compound is:
Figure imgf000031_0001
or a pharmaceutically acceptable salt thereof.
18. The compound of claim 1 , wherein the compound is:
Figure imgf000031_0002
or a pharmaceutically acceptable salt thereof.
19. The compound of claim 1 , wherein the compound is:
Figure imgf000031_0003
or a pharmaceutically acceptable salt thereof.
20. A composition comprising the compound of one of claims 1-19.
21. A pharmaceutical composition comprising the compound of one of claims 1-19, or a pharmaceutically acceptable salt thereof.
22. A method of treating viral infection in a subject in need thereof, comprising administering the compound of one of claims 1-19 to the subject.
23. The method of claim 22, wherein the viral infection comprises a hepatitis C viral (HCV) infection.
24. A method of treating cancer in a subject in need thereof, comprising administering the compound of one of claims 1-19 to the subject.
25. A method of treating a human epidermal growth factor receptor 2 (HER2) positive cancer in a subject in need thereof, comprising administering the compound of one of claims 1-19 to the subject.
26. The method of claim 24 or claim 25, wherein the cancer is esophageal, stomach, colon, rectal, pancreatic, lung, breast, cervix uteri, corpus uteri, ovary, bladder, head and neck, endometrial, osteosarcoma, prostate, or neuroblastoma.
27. A method of treating an allergic disease in a subject in need thereof, comprising administering the compound of one of claims 1-19 to the subject.
28. Use of the compound of one of claims 1-19, the composition of claim 20, or the pharmaceutical composition of claim 21 , in the manufacture of a medicament for the treatment of a viral infection, a cancer, or an allergic disease.
29. A dosage form suitable for administration to a subject in need thereof, comprising the compound of one of claims 1-19.
30. A kit, comprising the compound of one of claims 1-19 and instructions for use thereof.
PCT/US2019/049784 2018-09-07 2019-09-05 Imidazoquinoline compounds and uses thereof WO2020051356A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
ES19858183T ES2963112T3 (en) 2018-09-07 2019-09-05 Imidazoquinoline compounds and uses thereof
CN201980068724.0A CN113164460A (en) 2018-09-07 2019-09-05 Imidazole quinoline compound and application thereof
CA3111786A CA3111786A1 (en) 2018-09-07 2019-09-05 Imidazoquinoline compounds and uses thereof
US17/274,136 US20210214354A1 (en) 2018-09-07 2019-09-05 Imidazoquinoline compounds and uses thereof
JP2021512742A JP2022501327A (en) 2018-09-07 2019-09-05 Imidazoquinoline compounds and their use
EP19858183.7A EP3846807B1 (en) 2018-09-07 2019-09-05 Imidazoquinoline compounds and uses thereof
KR1020217010120A KR20210074290A (en) 2018-09-07 2019-09-05 Imidazoquinoline compounds and uses thereof
EP23188546.8A EP4306523A3 (en) 2018-09-07 2019-09-05 Imidazoquinoline compounds and uses thereof
AU2019335366A AU2019335366A1 (en) 2018-09-07 2019-09-05 Imidazoquinoline compounds and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862728556P 2018-09-07 2018-09-07
US62/728,556 2018-09-07

Publications (1)

Publication Number Publication Date
WO2020051356A1 true WO2020051356A1 (en) 2020-03-12

Family

ID=69722888

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/049784 WO2020051356A1 (en) 2018-09-07 2019-09-05 Imidazoquinoline compounds and uses thereof

Country Status (9)

Country Link
US (1) US20210214354A1 (en)
EP (2) EP4306523A3 (en)
JP (1) JP2022501327A (en)
KR (1) KR20210074290A (en)
CN (1) CN113164460A (en)
AU (1) AU2019335366A1 (en)
CA (1) CA3111786A1 (en)
ES (1) ES2963112T3 (en)
WO (1) WO2020051356A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10780180B2 (en) 2014-01-10 2020-09-22 Birdie Biopharmaceuticals, Inc. Compounds and compositions for immunotherapy
US11046781B2 (en) 2016-01-07 2021-06-29 Birdie Biopharmaceuticals, Inc. Anti-HER2 combinations for treating tumors
US11053240B2 (en) 2017-04-27 2021-07-06 Birdie Biopharmaceuticals, Inc. 2-amino-quinoline derivatives
US11110178B2 (en) 2016-07-07 2021-09-07 The Board Of Trustees Of The Leland Standford Junior University Antibody adjuvant conjugates
US11130812B2 (en) 2014-09-01 2021-09-28 Birdie Biopharmaceuticals, Inc. Anti PD-L1 conjugates for treating tumors
US11136397B2 (en) 2016-01-07 2021-10-05 Birdie Pharmaceuticals, Inc. Anti-EGFR combinations for treating tumors
US11220552B2 (en) 2016-01-07 2022-01-11 Birdie Biopharmaceuticals, Inc. Anti-CD20 combinations for treating tumors
US11279761B2 (en) 2014-07-09 2022-03-22 Birdie Biopharmaceuticals, Inc. Anti-PD-L1 combinations for treating tumors
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2
US11517567B2 (en) 2017-06-23 2022-12-06 Birdie Biopharmaceuticals, Inc. Pharmaceutical compositions
EP4178574A4 (en) * 2020-07-08 2024-04-10 Purdue Research Foundation Compounds, compositions, and methods for the treatment of fibrotic diseases and cancer

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100256169A1 (en) * 2003-09-05 2010-10-07 Anadys Pharmaceuticals, Inc. Administration of tlr7 ligands and prodrugs thereof for treatment of infection by hepatitis c virus
US20170056391A1 (en) * 2014-07-09 2017-03-02 Birdie Biopharmaceuticals, Inc. Combination therapy compositions and methods for treating cancers
US20180134701A1 (en) * 2011-05-18 2018-05-17 The University Of Kansas Toll-like receptor-7 and -8 modulatory 1h imidazoquinoline derived compounds
US20180148452A1 (en) * 2014-12-29 2018-05-31 Medshine Discovery Inc. Toll-like receptor-7 agonist

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US689338A (en) * 1901-03-29 1901-12-17 Nat Faucet Company Tapping-bung.
US5389640A (en) * 1991-03-01 1995-02-14 Minnesota Mining And Manufacturing Company 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
CA2580343A1 (en) * 2004-09-14 2006-03-23 Novartis Vaccines And Diagnostics, Inc. Imidazoquinoline compounds
WO2013033345A1 (en) * 2011-08-30 2013-03-07 Regents Of The University Of Minnesota Immunomodulators and immunomodulator conjugates
RS60430B1 (en) * 2014-04-22 2020-07-31 Hoffmann La Roche 4-amino-imidazoquinoline compounds
CA2954446A1 (en) * 2014-07-09 2016-01-14 Shanghai Birdie Biotech, Inc. Anti-pd-l1 combinations for treating tumors
CN115350279A (en) * 2016-01-07 2022-11-18 博笛生物科技有限公司 anti-HER 2 combinations for the treatment of tumors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100256169A1 (en) * 2003-09-05 2010-10-07 Anadys Pharmaceuticals, Inc. Administration of tlr7 ligands and prodrugs thereof for treatment of infection by hepatitis c virus
US20180134701A1 (en) * 2011-05-18 2018-05-17 The University Of Kansas Toll-like receptor-7 and -8 modulatory 1h imidazoquinoline derived compounds
US20170056391A1 (en) * 2014-07-09 2017-03-02 Birdie Biopharmaceuticals, Inc. Combination therapy compositions and methods for treating cancers
US20180148452A1 (en) * 2014-12-29 2018-05-31 Medshine Discovery Inc. Toll-like receptor-7 agonist

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
"Fieser and Fieser's Reagents for Organic Synthesis", vol. 1-40, 1991, JOHN WILEY AND SONS
"Larock's Comprehensive Organic Transformations", vol. 1-5, 1989, ELSEVIER SCIENCE PUBLISHERS
"March, Advanced Organic Chemistry", 1992, WILEY
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
CAREYSUNDBERG: "Advanced Organic Chemistry", 2000
GREENEWUTS: "Protective Groups in Organic Synthesis", 1999, WILEY
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2
See also references of EP3846807A4

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11786604B2 (en) 2014-01-10 2023-10-17 Birdie Biopharmaceuticals, Inc. Compounds and compositions for treating HER2 positive tumors
US11633494B2 (en) 2014-01-10 2023-04-25 Birdie Biopharmaceuticals, Inc. Compounds and compositions for immunotherapy
US11633495B2 (en) 2014-01-10 2023-04-25 Birdie Biopharmaceuticals, Inc. Compounds and compositions for immunotherapy
US10780180B2 (en) 2014-01-10 2020-09-22 Birdie Biopharmaceuticals, Inc. Compounds and compositions for immunotherapy
US11279761B2 (en) 2014-07-09 2022-03-22 Birdie Biopharmaceuticals, Inc. Anti-PD-L1 combinations for treating tumors
US11130812B2 (en) 2014-09-01 2021-09-28 Birdie Biopharmaceuticals, Inc. Anti PD-L1 conjugates for treating tumors
US11220552B2 (en) 2016-01-07 2022-01-11 Birdie Biopharmaceuticals, Inc. Anti-CD20 combinations for treating tumors
US11136397B2 (en) 2016-01-07 2021-10-05 Birdie Pharmaceuticals, Inc. Anti-EGFR combinations for treating tumors
US11702476B2 (en) 2016-01-07 2023-07-18 Birdie Biopharmaceuticals, Inc. Anti-EGFR combinations for treating tumors
US11046781B2 (en) 2016-01-07 2021-06-29 Birdie Biopharmaceuticals, Inc. Anti-HER2 combinations for treating tumors
US11547761B1 (en) 2016-07-07 2023-01-10 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
US11110178B2 (en) 2016-07-07 2021-09-07 The Board Of Trustees Of The Leland Standford Junior University Antibody adjuvant conjugates
US11053240B2 (en) 2017-04-27 2021-07-06 Birdie Biopharmaceuticals, Inc. 2-amino-quinoline derivatives
US11834448B2 (en) 2017-04-27 2023-12-05 Birdie Biopharmaceuticals, Inc. 2-amino-quinoline derivatives
US11517567B2 (en) 2017-06-23 2022-12-06 Birdie Biopharmaceuticals, Inc. Pharmaceutical compositions
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2
EP4178574A4 (en) * 2020-07-08 2024-04-10 Purdue Research Foundation Compounds, compositions, and methods for the treatment of fibrotic diseases and cancer

Also Published As

Publication number Publication date
JP2022501327A (en) 2022-01-06
EP3846807B1 (en) 2023-08-02
KR20210074290A (en) 2021-06-21
CN113164460A (en) 2021-07-23
AU2019335366A1 (en) 2021-03-25
EP4306523A3 (en) 2024-01-24
EP3846807A4 (en) 2022-05-18
US20210214354A1 (en) 2021-07-15
ES2963112T3 (en) 2024-03-25
EP4306523A2 (en) 2024-01-17
EP3846807A1 (en) 2021-07-14
CA3111786A1 (en) 2020-03-12

Similar Documents

Publication Publication Date Title
EP3846807B1 (en) Imidazoquinoline compounds and uses thereof
US9994546B2 (en) Cyclopropylamines as LSD1 inhibitors
TWI652269B (en) Azapyridone compound and use thereof
JP2022105091A (en) cGAS ANTAGONIST COMPOUNDS
US9695167B2 (en) Substituted triazolo[1,5-a]pyridines and triazolo[1,5-a]pyrazines as LSD1 inhibitors
KR102083857B1 (en) New substituted indazoles, methods for the production thereof, pharmaceutical preparations that contain said new substituted indazoles, and use of said new substituted indazoles to produce drugs
US10280180B2 (en) Macrocyclic purines for the treatment of viral infections
EP3802534B1 (en) Tricyclic heterocyclic compounds as sting activators
US9790191B2 (en) Substituted acylaminopyrimidines as inducers of alpha interferon production
EA024359B1 (en) Purine derivatives and their use as modulators of toll-like receptor 7
US20210363165A1 (en) Nitroxoline prodrug and use thereof
CN111686095A (en) Use of cannabidiol in preparation of medicament for treating coronavirus infection
US20200268730A1 (en) Amide derivatives useful in the treatment of hbv infection or hbv-induced diseases
JP4262092B2 (en) Thieno [2,3-d] pyrimidine derivatives having both LH and FSH agonist activity
EP3067351B1 (en) Compound having higher inhibition of protein kinase g activity and preparation method therefor
TW202102502A (en) Fused polycyclic pyridone compounds as influenza virus replication inhibitors
CN110590898A (en) Phosphorus (phosphine) amide ester compound and preparation method and application thereof
CN117003771B (en) Anti-influenza virus derivative and application thereof
US20240229037A1 (en) SIRNA TARGETING 17Beta-HYDROXYSTEROID DEHYDROGENASE TYPE 13 AND SIRNA CONJUGATE
WO2023168844A1 (en) Deuterated lactam compound, preparation method therefor, composition and use thereof
CN114008029A (en) Quinazoline prodrugs for the treatment of viral infections and additional diseases
CN117683042A (en) Fused tetracyclic compound, preparation method thereof and application thereof in medicines
JPWO2018194172A1 (en) Compound or salt thereof, antiviral agent, pharmaceutical composition

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: 19858183

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3111786

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021512742

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019335366

Country of ref document: AU

Date of ref document: 20190905

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019858183

Country of ref document: EP

Effective date: 20210407