WO2024069448A1 - Composés de 1-arylaminocarbonyl-1'-hétéroaryle substitués, composés de 1-hétéroarylaminocarbonyl-1'-hétéroaryle substitués et procédés les utilisant - Google Patents

Composés de 1-arylaminocarbonyl-1'-hétéroaryle substitués, composés de 1-hétéroarylaminocarbonyl-1'-hétéroaryle substitués et procédés les utilisant Download PDF

Info

Publication number
WO2024069448A1
WO2024069448A1 PCT/IB2023/059571 IB2023059571W WO2024069448A1 WO 2024069448 A1 WO2024069448 A1 WO 2024069448A1 IB 2023059571 W IB2023059571 W IB 2023059571W WO 2024069448 A1 WO2024069448 A1 WO 2024069448A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
amino
chloro
methoxy
oxopyrrolidin
Prior art date
Application number
PCT/IB2023/059571
Other languages
English (en)
Inventor
Andrew G. Cole
Yi Fan
Gavin D. Heffernan
Seyma OZTURK
Jorge G. Quintero
Original Assignee
Arbutus Biopharma Corporation
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 Arbutus Biopharma Corporation filed Critical Arbutus Biopharma Corporation
Publication of WO2024069448A1 publication Critical patent/WO2024069448A1/fr

Links

Classifications

    • 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
    • A61P31/20Antivirals for DNA viruses
    • 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
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • 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
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • 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
    • C07D405/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/10Spiro-condensed systems
    • C07D491/107Spiro-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring

Definitions

  • HBV Hepatitis B virus
  • HBV infection is one of the world's most prevalent diseases, being listed by National Institute of Allergy and Infectious Diseases (NIAID) as a High Priority Area of Interest. Although most individuals resolve the infection following acute symptoms, approximately 30% of cases become chronic.350-400 million people worldwide are estimated to have chronic hepatitis B, leading to 0.5-1 million deaths per year, due largely to the development of hepatocellular carcinoma, cirrhosis and/or other complications.
  • NIAID National Institute of Allergy and Infectious Diseases
  • a limited number of drugs are currently approved for the management of chronic hepatitis B, including two formulations of alpha-interferon (standard and pegylated) and five nucleoside/nucleotide analogues (lamivudine, adefovir, entecavir, telbivudine, and tenofovir) that inhibit HBV DNA polymerase.
  • the first-line treatment choices are entecavir, tenofovir and/or peg-interferon alfa-2a.
  • peg-interferon alfa-2a achieves desirable serological milestones in only one third of treated patients, and is frequently associated with severe side effects.
  • Entecavir and tenofovir are potent HBV inhibitors, but require long-term or possibly lifetime administration to continuously suppress HBV replication, and may eventually fail due to emergence of drug-resistant viruses. There is thus a pressing need for the introduction of novel, safe and effective therapies for chronic hepatitis B.
  • Hepatitis D virus (HDV) is a small circular enveloped RNA virus that can propagate only in the presence of HBV.
  • HDV requires the HBV surface antigen protein to propagate itself. Infection with both HBV and HDV results in more severe complications compared to infection with HBV alone. These complications include a greater likelihood of experiencing liver failure in acute infections and a rapid progression to liver cirrhosis, with an increased chance of developing liver cancer in chronic infections.
  • hepatitis D In combination with hepatitis B virus, hepatitis D has the highest mortality rate of all the hepatitis infections.
  • the routes of transmission of HDV are similar to those for HBV. Infection is largely restricted to persons at high risk of HBV infection, particularly injecting drug users and persons receiving clotting factor concentrates.
  • novel compounds that can be used to treat, ameliorate, and/or prevent HBV infection in a subject.
  • the novel compounds can be used in patients that are HBV infected, patients who are at risk of becoming HBV infected, and/or patients that are infected with drug-resistant HBV.
  • the HBV-infected subject is further HDV-infected.
  • the present disclosure addresses this need.
  • BRIEF SUMMARY The present disclosure provides certain compounds of formula (I), or a salt, solvate, geometric isomer, isotopologue, stereoisomer, or tautomer thereof, or any mixtures thereof, wherein the substituents in (I) are defined elsewhere herein: (I).
  • the present disclosure further provides pharmaceutical compositions comprising at least one compound of the present disclosure and at least one pharmaceutically acceptable carrier.
  • the pharmaceutical composition further comprises at least one additional agent that treats, ameliorates, and/or prevents hepatitis virus infection.
  • the present disclosure further provides methods of treating, ameliorating, and/or preventing hepatitis virus infection in a subject.
  • the method comprises administering to the subject in need thereof a therapeutically effective amount of at least one compound of the present disclosure and/or at least one pharmaceutical composition of the present disclosure.
  • the subject is infected with hepatitis B virus (HBV).
  • HBV hepatitis D virus
  • the subject is infected with HBV and HDV.
  • the present disclosure further provides methods of treating, ameliorating, and/or preventing cancer in a subject.
  • the method comprises administering to the subject in need thereof a therapeutically effective amount of at least one compound of the present disclosure and/or at least one pharmaceutically acceptable composition of the present disclosure.
  • the cancer is amenable to treatment by inhibiting PD-1, PD-L1, or the PD-1/PD-L1 interaction.
  • the cancer is at least one of pancreatic cancer, bladder cancer, colorectal cancer, breast cancer, prostate cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, gastric cancer, esophageal cancer, head and neck cancer, melanoma, neuroendocrine cancer, CNS cancer, brain cancer, bone cancer, soft tissue sarcoma, non-small cell lung cancer, small-cell lung cancer, or colon cancer.
  • the cancer is at least one of acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), myelodysplastic syndrome (MDS), myeloproliferative disease (MPD), chronic myeloid leukemia (CML), multiple myeloma (MM), non-Hodgkin's lymphoma (NHL), mantle cell lymphoma (MCL), follicular lymphoma, Waldestrom's macroglobulinemia (WM), T-cell lymphoma, B-cell lymphoma and diffuse large B-cell lymphoma (DLBCL).
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • MDS myelodysplastic syndrome
  • MDS myeloproliferative disease
  • the present disclosure relates, in certain aspects, to the discovery of certain substituted 1-arylaminocarbonyl-1'-heteroaryl or substituted 1-heteroarylaminocarbonyl-1'- heteroaryl compounds.
  • the compounds of the present disclosure are useful to treat, ameliorate, and/or prevent hepatitis B virus (HBV) infection and/or hepatitis B virus- hepatitis D virus (HBV-HDV) infection and related conditions in a subject.
  • these compounds are administered along with at least one additional agent useful for treating, ameliorating, and/or preventing the viral infection.
  • the subject is infected with HBV.
  • the HBV-infected subject is further infected with HDV.
  • the compounds of the disclosure are useful to treat, ameliorate and/or prevent cancer and related conditions in a subject.
  • Definitions As used herein, each of the following terms has the meaning associated with it in this section. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Generally, the nomenclature used herein and the laboratory procedures in animal pharmacology, pharmaceutical science, separation science and organic chemistry are those well-known and commonly employed in the art. It should be understood that the order of steps or order for performing certain actions is immaterial, so long as the present teachings remain operable.
  • the articles “a” and “an” refer 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.
  • alkenyl employed alone or in combination with other terms, means, unless otherwise stated, a stable monounsaturated or diunsaturated straight chain or branched chain hydrocarbon group having the stated number of carbon atoms.
  • Examples include vinyl, propenyl (or allyl), crotyl, isopentenyl, butadienyl, 1,3-pentadienyl, 1,4-pentadienyl, and the higher homologs and isomers.
  • alkoxy employed alone or in combination with other terms means, unless otherwise stated, an alkyl group having the designated number of carbon atoms, as defined elsewhere herein, connected to the rest of the molecule via an oxygen atom, such as, for example, methoxy, ethoxy, 1-propoxy, 2-propoxy (or isopropoxy) and the higher homologs and isomers.
  • oxygen atom such as, for example, methoxy, ethoxy, 1-propoxy, 2-propoxy (or isopropoxy) and the higher homologs and isomers.
  • a specific example is (C 1 -C 3 )alkoxy, such as, but not limited to, ethoxy and methoxy.
  • 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., C 1 -C 10 means one to ten carbon atoms) and includes straight, branched chain, or cyclic substituent groups. Examples include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, neopentyl, hexyl, and cyclopropylmethyl.
  • a specific embodiment is (C 1 -C 6 ) alkyl, such as, but not limited to, ethyl, methyl, isopropyl, isobutyl, n-pentyl, n-hexyl and cyclopropylmethyl.
  • alkynyl employed alone or in combination with other terms means, unless otherwise stated, a stable straight chain or branched chain hydrocarbon group with a triple carbon-carbon bond, having the stated number of carbon atoms. Non-limiting examples include ethynyl and propynyl, and the higher homologs and isomers.
  • the term "propargylic” refers to a group exemplified by -CH 2 -C ⁇ CH.
  • homopropargylic refers to a group exemplified by -CH 2 CH 2 -C ⁇ CH.
  • aromatic refers to a carbocycle or heterocycle with one or more polyunsaturated rings and having aromatic character, i.e., having (4n+2) delocalized ⁇ (pi) electrons, where 'n' is an integer.
  • aryl employed alone or in combination with other terms means, unless otherwise stated, a carbocyclic aromatic system containing one or more rings (typically one, two or three rings) wherein such rings may be attached together in a pendent manner, such as a biphenyl, or may be fused, such as naphthalene. Examples include phenyl, anthracyl and naphthyl.
  • Aryl groups also include, for example, phenyl or naphthyl rings fused with one or more saturated or partially saturated carbon rings (e.g., bicyclo[4.2.0]octa-1,3,5- trienyl, or indanyl), which can be substituted at one or more carbon atoms of the aromatic and/or saturated or partially saturated rings.
  • aryl-(C 1 -C 6 )alkyl refers to a functional group wherein a one to six carbon alkylene chain is attached to an aryl group, e.g., -CH 2 CH 2 -phenyl or -CH 2 - phenyl (or benzyl).
  • aryl-CH 2 - and aryl-CH(CH 3 )- are aryl-CH 2 - and aryl-CH(CH 3 )-.
  • substituted aryl-(C 1 -C 6 )alkyl refers to an aryl-(C 1 -C 6 )alkyl functional group in which the aryl group is substituted.
  • substituted aryl(CH 2 )- is substituted.
  • heteroaryl-(C 1 -C 6 )alkyl refers to a functional group wherein a one to three carbon alkylene chain is attached to a heteroaryl group, e.g., -CH 2 CH 2 -pyridyl.
  • a specific example is heteroaryl-(CH 2 )-.
  • substituted heteroaryl-(C 1 -C 6 )alkyl refers to a heteroaryl-(C 1 -C 6 )alkyl functional group in which the heteroaryl group is substituted.
  • a specific example is substituted heteroaryl-(CH 2 )-.
  • co-administered and co-administration as relating to a subject refer to administering to the subject a compound and/or composition of the disclosure along with a compound and/or composition that may also treat or prevent a disease or disorder contemplated herein.
  • the co-administered compounds and/or compositions are administered separately, or in any kind of combination as part of a single therapeutic approach.
  • the co-administered compound and/or composition may be formulated in any kind of combinations as mixtures of solids and liquids under a variety of solid, gel, and liquid formulations, and as a solution.
  • cycloalkyl by itself or as part of another substituent refers to, unless otherwise stated, a cyclic chain hydrocarbon having the number of carbon atoms designated (i.e.,C 3 -C 6 refers to a cyclic group comprising a ring group consisting of three to six carbon atoms) and includes straight, branched chain or cyclic substituent groups.
  • Examples of (C 3 -C 6 )cycloalkyl groups are cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. Cycloalkyl rings can be optionally substituted.
  • Non-limiting examples of cycloalkyl groups include: cyclopropyl, 2-methyl-cyclopropyl, cyclopropenyl, cyclobutyl, 2,3-dihydroxycyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctanyl, decalinyl, 2,5-dimethylcyclopentyl, 3,5- dichlorocyclohexyl, 4-hydroxycyclohexyl, 3,3,5-trimethylcyclohex-1-yl, octahydropentalenyl, oc
  • cycloalkyl also includes bicyclic hydrocarbon rings, non-limiting examples of which include, bicyclo-[2.1.1]hexanyl, bicyclo[2.2.1]heptanyl, bicyclo[3.1.1]heptanyl, 1,3-dimethyl[2.2.1] heptan-2-yl, bicyclo[2.2.2]octanyl, and bicyclo[3.3.3]undecanyl.
  • DCM dichloromethane.
  • a "disease” is a state of health of a subject wherein the subject cannot maintain homeostasis, and wherein if the disease is not ameliorated then the subject's health continues to deteriorate.
  • a disorder in a subject is a state of health in which the subject is able to maintain homeostasis, but in which the subject's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the subject's state of health.
  • halide refers to a halogen atom bearing a negative charge. The halide anions are fluoride (F ⁇ ), chloride (Cl ⁇ ), bromide (Br ⁇ ), and iodide (I ⁇ ).
  • halo or “halogen” alone or as part of another substituent refers to, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom.
  • Hepatitis B virus or HBV refers to a virus species of the genus Orthohepadnavirus, which is a part of the Hepadnaviridae family of viruses, and that is capable of causing liver inflammation in humans.
  • Hepatitis D virus (or HDV) refers to a virus species of the genus Deltaviridae, which is capable of causing liver inflammation in humans.
  • the HDV particle comprises an envelope, which is provided by HBV and surrounds the RNA genome and the HDV antigen.
  • the HDV genome is a single, negative stranded, circular RNA molecule nearly 1.7 kb in length.
  • the genome contains several sense and antisense open reading frames (ORFs), only one of which is functional and conserved.
  • ORFs open reading frames
  • the RNA genome is replicated through an RNA intermediate, the antigenome.
  • the genomic RNA and its complement, the antigenome can function as ribozymes to carry out self-cleavage and self- ligation reactions.
  • a third RNA present in the infected cell also complementary to the genome, but 800 bp long and polyadenylated, is the mRNA for the synthesis of the delta antigen (HDAg).
  • heteroalkenyl by itself or in combination with another term refers to, unless otherwise stated, a stable straight or branched chain monounsaturated or diunsaturated hydrocarbon group consisting of the stated number of carbon atoms and one or two heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. Up to two heteroatoms may be placed consecutively.
  • heteroalkyl by itself or in combination with another term refers to, unless otherwise stated, a stable straight or branched chain alkyl group consisting of the stated number of carbon atoms and one or two heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may be optionally oxidized and the nitrogen heteroatom may be optionally quaternized.
  • the heteroatom(s) may be placed at any position of the heteroalkyl group, including between the rest of the heteroalkyl group and the fragment to which it is attached, as well as attached to the most distal carbon atom in the heteroalkyl group.
  • Up to two heteroatoms may be consecutive, such as, for example, -CH 2 NH-OCH 3 , or -CH 2 CH 2 SSCH 3 .
  • heteroaryl or “heteroaromatic” refers to a heterocycle having aromatic character.
  • a polycyclic heteroaryl may include one or more rings that are partially saturated. Examples include tetrahydroquinoline and 2,3-dihydrobenzofuryl.
  • heterocycle or “heterocyclyl” or “heterocyclic” by itself or as part of another substituent refers to, unless otherwise stated, an unsubstituted or substituted, stable, mono- or multi-cyclic heterocyclic ring system that comprises carbon atoms and at least one heteroatom selected from the group consisting of N, O, and S, and wherein the nitrogen and sulfur heteroatoms may be optionally oxidized, and the nitrogen atom may be optionally quaternized.
  • the heterocyclic system may be attached, unless otherwise stated, at any heteroatom or carbon atom that affords a stable structure.
  • a heterocycle may be aromatic or non-aromatic in nature. In certain embodiments, the heterocycle is a heteroaryl.
  • non-aromatic heterocycles include monocyclic groups such as aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, pyrroline, imidazoline, pyrazolidine, dioxolane, sulfolane, 2,3-dihydrofuran, 2,5-dihydrofuran, tetrahydrofuran, thiophane, piperidine, 1,2,3,6-tetrahydropyridine, 1,4-dihydropyridine, piperazine, morpholine, thiomorpholine, pyran, 2,3-dihydropyran, tetrahydropyran, 1,4-dioxane, 1,3- dioxane, homopiperazine, homopiperidine, 1,3-dioxepane, 4,7-dihydro-1,3-dioxepin and hexamethyleneoxide.
  • heteroaryl groups include pyridyl, pyrazinyl, pyrimidinyl (such as, but not limited to, 2- and 4-pyrimidinyl), pyridazinyl, thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,3,4-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3-oxadiazolyl, 1,3,4-thiadiazolyl and 1,3,4-oxadiazolyl.
  • polycyclic heterocycles include indolyl (such as, but not limited to, 3-, 4- , 5-, 6- and 7-indolyl), indolinyl, quinolyl, tetrahydroquinolyl, isoquinolyl (such as, but not limited to, 1- and 5-isoquinolyl), 1,2,3,4-tetrahydroisoquinolyl, cinnolinyl, quinoxalinyl (such as, but not limited to, 2- and 5-quinoxalinyl), quinazolinyl, phthalazinyl, 1,8-naphthyridinyl, 1,4-benzodioxanyl, coumarin, dihydrocoumarin, 1,5-naphthyridinyl, benzofuryl (such as, but not limited to, 3-, 4-, 5-, 6- and 7-benzofuryl), 2,3-dihydrobenzofuryl, 1,2-benzisoxazolyl, benzothienyl (
  • the term "PD-L1 inhibitor” includes any compound that is capable of inhibiting the expression and/or function of the protein Programmed Death-Ligand 1 (PD-L1) either directly or indirectly.
  • PD-L1 also known as cluster of differentiation 274 (CD274) or B7 homolog 1 (B7-H1), is a type 1 transmembrane protein that plays a major role in suppressing the adaptive arm of immune system during pregnancy, tissue allograft transplants, autoimmune disease, and hepatitis.
  • the PD-L1 binds to its receptor, the inhibitory checkpoint molecule PD-1 (which is found on activated T cells, B cells, and myeloid cells) so as to modulate activation or inhibition of the adaptive arm of immune system.
  • the PD-L1 inhibitor inhibits the expression and/or function of PD-L1 by at least 5%, at least 10%, at least 20%, at least 50%, at least 75%, or at least 90%.
  • pharmaceutical composition or “composition” refers to a mixture of at least one compound useful within the disclosure with a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a subject.
  • the term "pharmaceutically acceptable” refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound useful within the disclosure, and is relatively non-toxic, i.e., the material may be administered to a subject 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 disclosure within or to the subject 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 disclosure within or to the subject 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 disclosure, and not injurious to the subject.
  • materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline
  • 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 disclosure, and are physiologically acceptable to the subject. Supplementary active compounds may also be incorporated into the compositions.
  • the "pharmaceutically acceptable carrier” may further include a pharmaceutically acceptable salt of the compound useful within the disclosure.
  • Other additional ingredients that may be included in the pharmaceutical compositions used in the practice of the disclosure are known in the art and described, for example in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.
  • the language “pharmaceutically acceptable salt” refers to a salt of the administered compound prepared from pharmaceutically acceptable non-toxic acids and/or bases, including inorganic acids, inorganic bases, organic acids, inorganic bases, solvates (including hydrates) and clathrates thereof.
  • a “pharmaceutically effective amount,” “therapeutically effective amount,” or “effective amount” of a compound is that amount of compound that is sufficient to provide a beneficial effect to the subject to which the compound is administered.
  • the term “prevent,” “preventing,” or “prevention” as used herein means avoiding or delaying the onset of symptoms associated with a disease or condition in a subject that has not developed such symptoms at the time the administering of an agent or compound commences.
  • telomeres Disease, condition and disorder are used interchangeably herein.
  • specifically bind or “specifically binds” as used herein is meant that a first molecule preferentially binds to a second molecule (e.g., a particular receptor or enzyme), but does not necessarily bind only to that second molecule.
  • the terms “subject” and “individual” and “patient” can be used interchangeably and may refer to a human or non-human mammal or a bird.
  • Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline and murine mammals. In certain embodiments, the subject is human.
  • substituted refers to that an atom or group of atoms has replaced hydrogen as the substituent attached to another group.
  • substituted alkyls include, but are not limited to, 2,2-difluoropropyl, 2- carboxycyclopentyl and 3-chloropropyl.
  • substituted alkyls include, but are not limited to, 2,2-difluoropropyl, 2- carboxycyclopentyl and 3-chloropropyl.
  • substituted alkyls include, but are not limited to, 2,2-difluoropropyl, 2- carboxycyclopentyl and 3-chloropropyl.
  • substituted as applied to the rings of these groups refers to any level of substitution, namely mono-, di-, tri-, tetra-, or penta-substitution, where such substitution is permitted.
  • the substituents are independently selected, and substitution may be at any chemically accessible position. In certain embodiments, the substituents vary in number between one and four. In other embodiments, the substituents vary in number between one and three.
  • the substituents vary in number between one and two. In yet other embodiments, the substituents are independently selected from the group consisting of C 1 -C 6 alkyl, -OH, C 1 -C 6 alkoxy, halo, amino, acetamido and nitro. As used herein, where a substituent is an alkyl or alkoxy group, the carbon chain may be branched, straight or cyclic.
  • each occurrence of alkyl or cycloalkyl is independently optionally substituted with at least one substituent selected from the group consisting of C 1 - C 6 alkyl, halo, -OR, phenyl (thus yielding, in non-limiting examples, optionally substituted phenyl-(C 1 -C 3 alkyl), such as, but not limited to, benzyl or substituted benzyl) and -N(R)(R), wherein each occurrence of R is independently H, C 1 -C 6 alkyl or C 3 -C 8 cycloalkyl.
  • each occurrence of aryl or heteroaryl is independently optionally substituted with at least one substituent selected from the group consisting of C 1 - C 6 alkyl, C 1 -C 6 haloalkyl, C 1 -C 6 haloalkoxy, halo, -CN, -OR, -N(R)(R), and C 1 -C 6 alkoxycarbonyl, wherein each occurrence of R is independently H, C 1 -C 6 alkyl or C 3 -C 8 cycloalkyl.
  • the ring when two substituents are taken together to form a ring having a specified number of ring atoms (e.g., R 2 and R 3 taken together with the nitrogen to which they are attached to form a ring having from 3 to 7 ring members), the ring can have carbon atoms and optionally one or more (e.g., 1 to 3) additional heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • substituents of compounds are disclosed in groups or in ranges. It is specifically intended that the description include each and every individual subcombination of the members of such groups and ranges.
  • C 1-6 alkyl is specifically intended to individually disclose C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1 -C 6 , C 1 -C 5 , C 1 -C 4 , C 1 -C 3 , C 1 -C 2 ,C 2 -C 6 , C 2 -C 5 , C 2 -C 4 , C 2 - C 3 , C 3 -C 6 , C 3 -C 5 , C 3 -C 4 , C 4 -C 6 , C 4 -C 5 , and C 5 -C 6 alkyl.
  • ranges throughout this disclosure, various aspects of the disclosure can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range.
  • a range such as from 1 to 6 should be considered to have specifically disclosed sub-ranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual and partial numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6. This applies regardless of the breadth of the range.
  • Compounds Programmed death-ligand 1 (PD-L1), which is also known as cluster of differentiation 274 (CD274) or B7 homolog 1 (B7-H1), is a human transmembrane protein that plays a major role in suppressing the immune system as needed.
  • PD-L1 PD-L1 to the receptor programmed cell death protein 1 (PD-1) or B7.1 membrane protein (both of which are found on activated T cells, B cells, and myeloid cells), transmits an inhibitory signal, which reduces proliferation of the CD8+ T cells in the lymph nodes.
  • PD-1 programmed cell death protein 1
  • B7.1 membrane protein both of which are found on activated T cells, B cells, and myeloid cells
  • small-molecule immunomodulators targeting the PD-1/PD- L1 signaling pathway are used to treat, ameliorate, and/or prevent hepatitis B virus (HBV) infection and related conditions in a subject.
  • HBV hepatitis B virus
  • inhibition of PDL-1 enhances the immune response to at least one HBV antigen.
  • the disclosure includes a compound of formula (I), or a salt, solvate, prodrug, stereoisomer (such as, in a non-limiting example, an enantiomer or diastereoisomer, and any mixtures thereof, such as, in a non-limiting example, mixtures in any proportion of enantiomers and/or diastereoisomers thereof), tautomer, and/or geometric isomer, and any mixtures thereof.
  • stereoisomer such as, in a non-limiting example, an enantiomer or diastereoisomer, and any mixtures thereof, such as, in a non-limiting example, mixtures in any proportion of enantiomers and/or diastereoisomers thereof
  • tautomer such as, in a non-limiting example, mixtures in any proportion of enantiomers and/or diastereoisomers thereof
  • geometric isomer and any mixture
  • the present disclosure provides a compound formula (I), or a salt, solvate, geometric isomer, isotopologue, stereoisomer, or tautomer thereof: (I), wherein: X 1 is selected from the group consisting of CR 2a and N; X 2 is selected from the group consisting of CR 2b and N; X 3 is selected from the group consisting of CR 2c and N; X 4 is selected from the group consisting of CR 2d and N; X 5 is selected from the group consisting of CR 2e and N; X 6 is selected from the group consisting of CR 2f and N; wherein one to four selected from the group consisting of X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 are N; R 1a and R 1b are each independently selected from the group consisting of H, halogen, optionally substituted C 1 -C 6 alkyl, optionally substituted C 3 -C 8 cyclo
  • the compound of Formula (I) is not a compound selected from the group consisting of: N-(3-(3-chloro-2-(4-(((2-hydroxyethyl)amino)methyl)-3-methoxyphenyl)pyridin-4-yl)-2- methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)picolinamide; N-(2-chloro-3-(3-chloro-2-(4-(((2-hydroxyethyl)amino)methyl)-3- methoxyphenyl)pyridin-4-yl)phenyl)-5-(((2-hydroxyethyl)amino)methyl)picolinamide; N-(3-(2-(4-(((1-acetylpiperidin-4-yl)amino)methyl)-3-methoxyphenyl)-3-chloropyridin-4- yl)-2-methylphenyl)-5-(((2-hydroxyethoxyeth)
  • the compound of formula (I) is a compound of formula (Ia): (Ia). In certain embodiments, the compound of formula (I) is a compound of formula (Ib): (Ib). In certain embodiments, the compound of formula (I) is a compound of formula (Ic): (Ic). In certain embodiments, the compound of formula (I) is a compound of formula (Id): (Id). In certain embodiments, the compound of formula (I) is a compound of formula (Ie): (Ie). In certain embodiments, the compound of formula (I) is a compound of formula (If): (If). In certain embodiments, the compound of formula (I) is a compound of formula (Ig): (Ig).
  • the compound of formula (I) is a compound of formula (Ih): (Ih). In certain embodiments, the compound of formula (I) is a compound of formula (Ii): (Ii). In certain embodiments, the compound of formula (I) is a compound of formula (Ij): (Ij). In certain embodiments, the compound of formula (I) is a compound of formula (Ik): (Ik). In certain embodiments, the compound of formula (I) is a compound of formula (Il): (Il). In certain embodiments, the compound of formula (I) is a compound of formula (Im): (Im). In certain embodiments, the compound of formula (I) is a compound of formula (In): (In).
  • the compound of formula (I) is a compound of formula (Io): (Io).
  • each of R 2b , R 2c , R 2d , R 2e , and R 2f is independently H.
  • each of R 2a , R 2c , R 2d , R 2e , and R 2f is independently H.
  • each of R 2a , R 2b , R 2d , R 2e , and R 2f is independently H.
  • each of R 2a , R 2b , R 2c , R 2e , and R 2f is independently H.
  • each of R 2a , R 2b , R 2c , R 2d , and R 2f is independently H. In certain embodiments, each of R 2a , R 2b , R 2c , R 2d , and R 2e is independently H. In certain embodiments, each of R 2b , R 2c , R 2e , and R 2f is independently H. In certain embodiments, each of R 2b , R 2c , R 2d , and R 2f is independently H. In certain embodiments, each of R 2b , R 2c , R 2d , and R 2e is independently H. In certain embodiments, each of R 2a , R 2c , R 2e , and R 2f is independently H.
  • each of R 2a , R 2c , R 2d , and R 2f is independently H. In certain embodiments, each of R 2a , R 2c , R 2d , and R 2e is independently H. In certain embodiments, each of R 2a , R 2b , R 2e , and R 2f is independently H. In certain embodiments, each of R 2a , R 2b , R 2d , and R 2f is independently H. In certain embodiments, each of R 2a , R 2b , R 2d , and R 2e is independently H. In certain embodiments, R 1a is Me. In certain embodiments, R 1a is Cl. In certain embodiments, R 1a is H.
  • R 1a is F. In certain embodiments, R 1a is OMe. In certain embodiments, R 1b is Me. In certain embodiments, R 1b is Cl. In certain embodiments, R 1b is H. In certain embodiments, R 1b is F. In certain embodiments, R 1b is OMe. In certain embodiments, R 1a and R 1b are identical. In certain embodiments, Z 1 is N. In certain embodiments, Z 1 is CH. In certain embodiments, Z 2 is N. In certain embodiments, Z 2 is CH. In certain embodiments, R 4a is H. In certain embodiments, R 4a is F. In certain embodiments, R 4a is Me. In certain embodiments, R 4a is Et.
  • R 4a is OMe. In certain embodiments, R 4a is OCHF 2 . In certain embodiments, R 4b is H. In certain embodiments, R 4b is F. In certain embodiments, R 4b is Me. In certain embodiments, R 4b is Et. In certain embodiments, R 4b is OMe. In certain embodiments, R 4b is OCHF 2 . In certain embodiments, R 4c is H. In certain embodiments, R 4c is F. In certain embodiments, R 4c is Me. In certain embodiments, R 4c is Et. In certain embodiments, R 4c is OMe. In certain embodiments, R 4c is OCHF 2 . In certain embodiments, R 4d is H.
  • R 4d is F. In certain embodiments, R 4d is Me. In certain embodiments, R 4d is Et. In certain embodiments, R 4d is OMe. In certain embodiments, R 4d is OCHF 2 . In certain embodiments, R 4e is H. In certain embodiments, R 4e is F. In certain embodiments, R 4e is Me. In certain embodiments, R 4e is Et. In certain embodiments, R 4e is OMe. In certain embodiments, R 4e is OCHF 2 . In certain embodiments, R 3a is -(CH 2 ) 1-3 NH(CH 2 ) 1-3 (optionally substituted 5- oxopyrrolidin-2-yl).
  • R 3a is -(CH 2 )NH(optionally substituted C 1 -C 6 haloalkyl). In certain embodiments, R 3a is -(CH 2 ) 1-3 (optionally substituted 2,6- diazaspiro[3.4]octan-2-yl). In certain embodiments, R 3a is -(CH 2 ) 1-3 (optionally substituted 7- oxa-2-azaspiro[3.5]nonan-2-yl). In certain embodiments, R 3a is -(CH 2 ) 1-3 (optionally substituted pyrrolidinyl).
  • R 3a is -(CH 2 ) 1-3 (optionally substituted 2,5- diazaspiro[3.4]octan-2-yl). In certain embodiments, R 3a is -(CH 2 ) 1-3 (optionally substituted piperidinyl). In certain embodiments, R 3a is -(CH 2 ) 1-3 NH(optionally substituted pyrrolidinyl). In certain embodiments, R 3a is -(CH 2 ) 1-3 N(CH 3 )(optionally substituted piperidin-4-yl). In certain embodiments, R 3a is -(CH 2 ) 1-3 NH(optionally substituted piperidin-3-yl). In certain embodiments, R 3a is .
  • R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R
  • R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R 3a is . In certain embodiments, R
  • At least one of R 5a , R 5b , R 5c , R 5d , R 5e , and R 5f is H. In certain embodiments, at least two of R 5a , R 5b , R 5c , R 5d , R 5e , and R 5f are H. In certain embodiments, at least three of R 5a , R 5b , R 5c , R 5d , R 5e , and R 5f are H. In certain embodiments, at least four of R 5a , R 5b , R 5c , R 5d , R 5e , and R 5f are H.
  • R 5a , R 5b , R 5c , R 5d , R 5e , and R 5f are H.
  • each of R 5a , R 5b , R 5c , R 5d , R 5e , and R 5f are H.
  • R 3b is -(CH 2 ) 1-3 CH(CH 3 )(OH).
  • R 3b is -(CH 2 ) 1-3 (optionally substituted 5-oxopyrrolidin-2-yl).
  • R 3b is .
  • R 3b is .
  • R 3b is .
  • R 3b is .
  • R 3b is .
  • R 3b is .
  • R 3b is . In certain embodiments, R 3b is . In certain embodiments, R 3b is . In certain embodiments, R 1c is , R 1d is , R 1a and R 1b are identical, R 3a and R 3c are not identical, and R 3c is -(CH 2 ) 1-3 N(R 8 )(C 1 -C 6 haloalkyl).
  • R 1c is , R 1d is , R 3a and R 3c are not identical, and R 3c is selected from the group consisting of -(CH 2 ) 1-3 (optionally substituted C 2 -C 9 heterocyclyl), -(CH 2 ) 1-3 N(R 8 )(optionally substituted C 3 -C 8 cycloalkyl), and -(CH 2 ) 1-3 N(R 8 )(optionally substituted C 2 -C 9 heterocyclyl).
  • R 1c is , R 1d is , R 3a and R 3c are not identical, and at least one of R 4a , R 4b , R 4d , and R 4e is selected from the group consisting of C 1 -C 6 alkyl and halogen.
  • R 1c is , R 1d is , R 3a and R 3c are not identical, R 1a and R 1b are identical, and R 3c is optionally substituted 2- azaspiro[3.3]heptanyl.
  • R 1c is , R 1d is , and R 3a is 7- oxa-2-azaspiro[3.5]nonanyl.
  • R 1c is , R 1d is , and R 3c is - (CH 2 ) 1-3 N(R 8 )(optionally substituted C 1 -C 6 alkyl), wherein R 8 is selected from the group consisting of optionally substituted C 1 -C 6 alkyl, optionally substituted C 3 -C 8 cycloalkyl, optionally substituted C 2 -C 8 heterocycloalkyl, optionally substituted C7-C 12 aralkyl, optionally substituted C 6 -C10 aryl, and optionally substituted C 2 -C 12 heteroaryl.
  • R 1c is , R 1d is , and R 3c is - (CH 2 ) 1-3 N(R 8 )(C 1 -C 6 alkoxy). In certain embodiments, R 1c is , R 1d is , and R 3c is - (CH 2 ) 1-3 N(R 8 )(optionally substituted C 3 -C 6 hydroxyalkyl). In certain embodiments, R 1c is , R 1d is , and at least one of R 3a and R 3c is -(CH 2 ) 1-3 (optionally substituted pyrrolidin-1-yl).
  • R 1c is , R 1d is , X 3 is N, and at least one of R 4a , R 4b , R 4d , and R 4e is C 1 -C 6 haloalkoxy.
  • R 1c is , R 1d is , X 3 is N, and R 1a and R 1b are identical.
  • R 1c is , R 1d is , X 3 is N, and R 3c is -(CH 2 ) 1-3 N(R 8 )(C 1 -C 6 hydroxyalkyl), wherein the hydroxyalkyl in R 3c is substituted at any position with at least one C 1 -C 6 alkyl.
  • R 1c is , R 1d is , X 3 is N, and R 3c is -(CH 2 ) 1-3 N(R 8 )(C 1 -C 6 haloalkyl).
  • R 1c is , R 1d is , X 3 is N, and R 3c is -(CH 2 ) 1-3 N(R 8 )(CH 2 ) 1-3 (optionally substituted C 2 -C 9 heterocyclyl).
  • R 1c is , R 1d is , X 3 is N, and R 3c is -(CH 2 ) 1-3 (optionally substituted C 2 -C 9 heterocyclyl).
  • R 1c is , R 1d is , and Z 2 is N. In certain embodiments, R 1c is , R 1d is , and Z 3 is CR 6d . In certain embodiments, R 1c is , R 1d is , Z 3 is N, and at least one of R 6a , R 6c , and R 6e is selected from the group consisting of C 1 -C 6 alkyl, hydroxy, C 1 -C 6 alkoxy, and halogen. In certain embodiments, R 1c is , R 1d is , and Z 4 is N.
  • R 1c is , R 1d is , R 3 is -(CH 2 ) 1- 3 N(R 8 )(CH 2 ) 1-3 (1-methyl-5-oxopyrrolidinyl) or R 3c is -(CH 2 ) 1-3 (optionally substituted pyrrolidinyl).
  • R 1c is , R 1d is , Z 1 is N and R 3c is -(CH 2 ) 1-3 (optionally substituted C 2 -C 9 heterocyclyl).
  • R 1c is , R 1d is , Z 1 is N and R 3a and R 3c are identical.
  • R 1c is , R 1d is , Z 1 is N, R 3a is –(CH 2 ) 1-3 (optionally substituted C 2 -C 9 heterocyclyl), and R 3c is -(CH 2 ) 1-3 N(R 8 )(CH 2 ) 1- 3(optionally substituted C 2 -C 0 heterocyclyl).
  • R 1c is , R 1d is , at least one of R 1a and R 1b is F or OMe.
  • R 1c is , R 1d is , R 1a and R 1b are Cl, and R 3c is CH 2 N(R 8 )(C 1 -C 6 haloalkyl).
  • R 1c is , R 1d is , R 3a and R 3c are not identical, and at least one of R 4a , R 4b , R 4d , and R 4e is selected from the group consisting of Me, Et, F, and Cl.
  • R 1c is , R 1d is , and R 3c is CH 2 N(R 8 )(methoxyethyl).
  • R 1c is , R 1d is , R 3c is CH 2 N(CH 3 )(C 1 -C 6 hydroxyalkyl).
  • R 1c is , R 1d is , and R 3c is CH 2 N(R 8 )(optionally substituted hydroxypropyl).
  • R 1c is , R 1d is , X 3 is N, and at least one of R 4a , R 4b , R 4d , and R 4e is OCHF 2 .
  • R 1c is , R 1d is , X 3 is N, and R 1a and R 1b are Cl.
  • R 1c is , R 1d is , X 3 is N, and R 3c is CH 2 N(R 8 )(C 1 -C 6 hydroxyalkyl), wherein the hydroxyalkyl in R 3c is substituted at any position with at least one selected from the group consisting of methyl and isopropyl.
  • R 1c is , R 1d is , X 3 is N, and R 3c is CH 2 N(R 8 )(C 1 -C 6 fluoroalkyl). In certain embodiments, R 1c is , R 1d is , X 3 is N, and R 3c is CH 2 N(R 8 )CH 2 (optionally substituted C 2 -C 9 heterocyclyl). In certain embodiments, R 1c is , R 1d is , X 3 is N, and R 1a and R 1b are identical.
  • R 1c is , R 1d is , Z 3 is N, and at least one of R 6a , R 6c , and R 6e is selected from the group consisting of Me, OMe, and F.
  • R 1c is , R 1d is , and Z 3 is CF.
  • R 3c is -(CH 2 ) 1-3 NH(C 1 -C 6 alkyl).
  • R 3c is -(CH 2 ) 1-3 NH(optionally substituted C 1 -C 6 haloalkyl). In certain embodiments, R 3c is - (CH 2 ) 1-3 NH(optionally substituted C 1 -C 6 hydroxyalkyl). In certain embodiments, R 3c is - (CH 2 ) 1-3 N(C 1 -C 6 alkyl)(optionally substituted C 1 -C 6 hydroxyalkyl). In certain embodiments, R 3c is -(CH 2 ) 1-3 NH(optionally substituted C 1 -C 6 alkoxy). In certain embodiments, R 3c is - (CH 2 ) 1-3 NHCH 2 C(CH 3 )(OH).
  • R 3c is -(CH 2 ) 1-3 NHCH 2 C(CH 3 ) 2 (OH). In certain embodiments, R 3c is -(CH 2 ) 1-3 NHCH(CH(CH 3 ) 2 )CH 2 OH. In certain embodiments, R 3c is -(CH 2 ) 1-3 (optionally substituted azetidin-1-yl). In certain embodiments, R 3c is -(CH 2 ) 1- 3(optionally substituted pyrrolidin-1-yl). In certain embodiments, R 3c is -(CH 2 ) 1- 3 NH(optionally substituted cyclopropyl).
  • R 3c is -(CH 2 ) 1- 3 NH(optionally substituted 2-azaspiro[3.3]heptan-2-yl). In certain embodiments, R 3c is - (CH 2 ) 1-3 N(optionally substituted piperidin-4-yl). In certain embodiments, R 3c is -(CH 2 ) 1- 3 NH(CH 2 ) 1-3 (optionally substituted oxetan-2-yl). In certain embodiments, R 3c is -(CH 2 ) 1- 3 NH(optionally substituted cyclobutyl).
  • R 3c is -(CH 2 ) 1-3 NH(optionally substituted cyclohexyl). In certain embodiments, R 3c is -(CH 2 ) 1-3 NH(CH 2 ) 1-3 (optionally substituted 5-oxopyrrolidin-2-yl). In certain embodiments, R 3c is -(CH 2 ) 1-3 NH(CH 2 ) 1-3 (optionally substituted bicyclo[2.2.1]heptanyl). In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is .
  • R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c . In certain embodiments, R 3c is . In certain embodiments, R 3c . In certain embodiments, R 3c is . In certain embodiments, R 3c . In certain embodiments, R 3c . In certain embodiments, R 3c . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is .
  • R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R
  • R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R 3c is . In certain embodiments, R
  • R 3d is -(CH 2 ) 1-3 NH(C 1 -C 6 hydroxyalkyl). In certain embodiments, R 3d is -(CH 2 ) 1-3 N(C 1 -C 6 alkyl)(C 1 -C 6 haloalkyl). In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R
  • R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, R 3d is . In certain embodiments, at least one of R 5g , R 5h , R 5i , R 5j , R 5k , and R 5l is H. In certain embodiments, at least two of R 5g , R 5h , R 5i , R 5j , R 5k , and R 5l are H. In certain embodiments, at least three of R 5g , R 5h , R 5i , R 5j , R 5k , and R 5l are H.
  • R 5g , R 5h , R 5i , R 5j , R 5k , and R 5l are H. In certain embodiments, at least five of R 5g , R 5h , R 5i , R 5j , R 5k , and R 5l are H. In certain embodiments, each of R 5g , R 5h , R 5i , R 5j , R 5k , and R 5l are H. In certain embodiments, R 1c is selected from the group consisting of , , , , , , , , , , , , , , ,
  • R 9a is methyl.
  • R 9a is ethyl.
  • R 9a is methoxy.
  • R 9a is fluoro.
  • R 9a is chloro.
  • R 9a is difluoromethoxy.
  • R 9b is methyl. In certain embodiments, R 9b is ethyl. In certain embodiments, R 9b is methoxy. In certain embodiments, R 9b is fluoro. In certain embodiments, R 9b is chloro. In certain embodiments, R 9b is difluoromethoxy.
  • R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R
  • R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R
  • R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R
  • R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R
  • R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1c is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is In certain embodiments, R 1d is In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R
  • R 1d is . In certain embodiments, R 1d is . . . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is .
  • R 11a is H.
  • R 11a is methyl.
  • R 11a is CH 3 .
  • R 11b is H.
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d
  • R 1d is , In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d
  • R 1d is . . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, R
  • R 1d is . In certain embodiments, R 1d is . In certain embodiments, R 1d is . In certain embodiments, each occurrence of optionally substituted alkyl, optionally substituted alkoxy, optionally substituted haloalkyl, optionally substituted haloalkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aralkyl, optionally substituted heterocyclyl, optionally substituted aryl, and optionally substituted heteroaryl is independently optionally substituted with at least one substituent selected from the group consisting of C 1 -C 6 alkyl, C 3 -C 8 cycloalkyl, C 2 -C 12 heterocycloalkyl, C 1 -C 6 hydroxyalkyl, halogen, CN, NO 2 OR’, N(R’)(R”), C 1 -C 6 haloalkoxy, C 3 -C 8 halocycloalkoxy, aryl, heteroary
  • each occurrence of optionally substituted C 3 -C 6 hydroxyalkyl, optionally substituted 5-oxopyrrolidin-2-yl, optionally substituted 2,6- diazaspiro[3.4]octan-2-yl, optionally substituted 7-oxa-2-azaspiro[3.5]nonan-2-yl, optionally substituted pyrrolidinyl, optionally substituted 2-azaspiro[3.3]heptan-6-yl, optionally substituted piperidin-4-yl, optionally substituted 5-oxopyrrolidin-2-yl, optionally substituted pyrrolidin-1-yl, optionally substituted azetidin-1-yl, optionally substituted cyclopropyl, and optionally substituted oxetan-2-yl is independently optionally substituted with at least one substituent selected from the group consisting of C 1 -C 6 alkyl, C 3 -C 8 cycloalkyl, C 2 -C
  • the compound is selected from the group consisting of: N-(3-(3-chloro-2-(3-fluoro-5-methoxy-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide; (S)-N-(3-(3-chloro-2-(3-fluoro-5-methoxy-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide; (R)-N-(3-(3-chloro-2-(3-fluoro-5-methoxy-4-((((5-oxopyrrolidin-2-
  • the compounds of the disclosure may possess one or more stereocenters, and each stereocenter may exist independently in either the (R) or (S) configuration.
  • compounds described herein are present in optically active or racemic forms.
  • the compounds described herein encompass racemic, optically active, regioisomeric and stereoisomeric forms, or combinations thereof that possess the therapeutically useful properties described herein.
  • Preparation of optically active forms is achieved in any suitable manner, including by way of non-limiting example, by resolution of the racemic form with recrystallization techniques, synthesis from optically active starting materials, chiral synthesis, or chromatographic separation using a chiral stationary phase.
  • a compound illustrated herein by the racemic formula further represents either of the two enantiomers or mixtures thereof, or in the case where two or more chiral center are present, all diastereomers or mixtures thereof.
  • the compounds of the disclosure exist as tautomers. All tautomers are included within the scope of the compounds recited herein.
  • 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 Cl, 18 F, 123 I, 125 I, 13 N, 15 N, 15 O, 17 O, 18 O, 32 P, and 35 S. In certain embodiments, substitution with heavier isotopes such as deuterium affords greater chemical stability. 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.
  • the compounds described herein are labeled by other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
  • suitable optional substituents are not intended to limit the scope of the claimed disclosure.
  • the compounds of the disclosure may contain any of the substituents, or combinations of substituents, provided herein.
  • Salts The compounds described herein may form salts with acids or bases, and such salts are included in the present disclosure.
  • salts embraces addition salts of free acids or bases that are useful within the methods of the disclosure.
  • pharmaceutically acceptable salt refers to salts that possess toxicity profiles within a range that affords utility in pharmaceutical applications.
  • the salts are pharmaceutically acceptable salts.
  • Pharmaceutically unacceptable salts may nonetheless possess properties such as high crystallinity, which have utility in the practice of the present disclosure, such as for example utility in process of synthesis, purification or formulation of compounds useful within the methods of the disclosure.
  • Suitable pharmaceutically acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid. Examples of inorganic acids include sulfate, hydrogen sulfate, hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric, and phosphoric acids (including hydrogen phosphate and dihydrogen phosphate).
  • organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (or pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, sulfanilic, 2-hydroxyethanesulfonic, trifluoromethanesulfonic, p-toluenesulfonic, cyclohexylaminosulfonic, stearic, alginic, ⁇ -hydroxybutyric, sal
  • Salts may be comprised of a fraction of one, one or more than one molar equivalent of acid or base with respect to any compound of the disclosure.
  • Suitable pharmaceutically acceptable base addition salts of compounds of the disclosure include, for example, ammonium salts and metallic salts including alkali metal, alkaline earth metal and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts.
  • Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, N,N'-dibenzylethylene- diamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (or N- methylglucamine) and procaine.
  • the compounds of the disclosure are useful within the methods of the disclosure in combination with one or more additional agents useful for treating HBV and/or HDV infections.
  • additional agents may comprise compounds or compositions identified herein, or compounds (e.g., commercially available compounds) known to treat, prevent, or reduce the symptoms of HBV and/or HDV infections.
  • Non-limiting examples of one or more additional agents useful for treating HBV and/or HDV infections include: (a) reverse transcriptase inhibitors; (b) capsid inhibitors; (c) cccDNA formation inhibitors; (d) RNA destabilizers; (e) oligomeric nucleotides targeted against the HBV genome; (f) immunostimulators, such as checkpoint inhibitors (e.g., PD-L1 inhibitors); (g) GalNAc-siRNA conjugates targeted against an HBV gene transcript; and (h) therapeutic vaccines.
  • the reverse transcriptase inhibitor is a reverse-transcriptase inhibitor (NARTI or NRTI).
  • the reverse transcriptase inhibitor is a nucleotide analog reverse-transcriptase inhibitor (NtARTI or NtRTI).
  • Reported reverse transcriptase inhibitors include, but are not limited to, entecavir, clevudine, telbivudine, lamivudine, adefovir, and tenofovir, tenofovir disoproxil, tenofovir alafenamide, adefovir dipovoxil, (1R,2R,3R,5R)-3-(6-amino-9H-9-purinyl)-2-fluoro-5- (hydroxymethyl)-4-methylenecyclopentan-1-ol (described in U.S.
  • Reported reverse transcriptase inhibitors further include, but are not limited to, entecavir, lamivudine, and (1R,2R,3R,5R)-3-(6-amino-9H-9-purinyl)-2-fluoro-5- (hydroxymethyl)-4-methylenecyclopentan-1-ol.
  • Reported reverse transcriptase inhibitors further include, but are not limited to, a covalently bound phosphoramidate or phosphonamidate moiety of the above-mentioned reverse transcriptase inhibitors, or as described in for example U.S. Patent No.8,816,074, US Patent Application Publications No. US 2011/0245484 A1, and US 2008/0286230A1, all of which incorporated herein in their entireties by reference.
  • Reported reverse transcriptase inhibitors further include, but are not limited to, nucleotide analogs that comprise a phosphoramidate moiety, such as, for example, methyl ((((1R,3R,4R,5R)-3-(6-amino-9H-purin-9-yl)-4-fluoro-5-hydroxy-2-methylenecyclopentyl) methoxy)(phenoxy) phosphoryl)-(D or L)-alaninate and methyl (((1R,2R,3R,4R)-3-fluoro-2- hydroxy-5-methylene-4-(6-oxo-1,6-dihydro-9H-purin-9-yl)cyclopentyl)methoxy)(phenoxy) phosphoryl)-(D or L)-alaninate.
  • nucleotide analogs that comprise a phosphoramidate moiety, such as, for example, methyl ((((1R,3R,4R,5R)-3-(6
  • the individual diastereomers thereof include, for example, methyl ((R)-(((1R,3R,4R,5R)-3-(6-amino-9H-purin-9-yl)-4-fluoro-5- hydroxy-2-methylenecyclopentyl)methoxy)(phenoxy)phosphoryl)-(D or L)-alaninate and methyl ((S)-(((1R,3R,4R,5R)-3-(6-amino-9H-purin-9-yl)-4-fluoro-5-hydroxy-2- methylenecyclopentyl) methoxy)(phenoxy)phosphoryl)-(D or L)-alaninate.
  • Reported reverse transcriptase inhibitors further include, but are not limited to, compounds comprising a phosphonamidate moiety, such as, for example, tenofovir alafenamide, as well as those described in U.S. Patent Application Publication No. US 2008/0286230 A1, incorporated herein in its entirety by reference.
  • Methods for preparing stereoselective phosphoramidate or phosphonamidate containing actives are described in, for example, U.S. Patent No.8,816,074, as well as U.S. Patent Application Publications No. US 2011/0245484 A1 and US 2008/0286230 A1, all of which incorporated herein in their entireties by reference.
  • capsid inhibitor includes compounds that are capable of inhibiting the expression and/or function of a capsid protein either directly or indirectly.
  • a capsid inhibitor may include, but is not limited to, any compound that inhibits capsid assembly, induces formation of non-capsid polymers, promotes excess capsid assembly or misdirected capsid assembly, affects capsid stabilization, and/or inhibits encapsidation of RNA (pgRNA).
  • Capsid inhibitors also include any compound that inhibits capsid function in a downstream event(s) within the replication process (e.g., viral DNA synthesis, transport of relaxed circular DNA (rcDNA) into the nucleus, covalently closed circular DNA (cccDNA) formation, virus maturation, budding and/or release, and the like).
  • the inhibitor detectably inhibits the expression level or biological activity of the capsid protein as measured, e.g., using an assay described herein.
  • the inhibitor inhibits the level of rcDNA and downstream products of viral life cycle by at least 5%, at least 10%, at least 20%, at least 50%, at least 75%, or at least 90%.
  • Reported capsid inhibitors include, but are not limited to, compounds described in International Patent Applications Publication Nos WO 2013006394, WO 2014106019, and WO 2 014089296, all of which incorporated herein in their entireties by reference.
  • Reported capsid inhibitors also include, but are not limited to, the following compounds and pharmaceutically acceptable salts and/or solvates thereof: Bay-41-4109 (see Int’l Patent Application Publication No. WO 2013144129), AT-61 (see Int’l Patent Application Publication No. WO 1998033501; and King, et al., 1998, Antimicrob. Agents Chemother.42(12):3179–3186), DVR-01 and DVR-23 (see Int’l Patent Application Publication No.
  • capsid inhibitors include, but are not limited to, those generally and specifically described in U.S. Patent Application Publication Nos. US 2015/0225355, US 2015/0132258, US 2016/0083383, US 2016/0052921, US 2019/0225593, and Int’l Patent Application Publication Nos.
  • cccDNA Formation Inhibitors Covalently closed circular DNA (cccDNA) is generated in the cell nucleus from viral rcDNA and serves as the transcription template for viral mRNAs.
  • cccDNA formation inhibitor includes compounds that are capable of inhibiting the formation and/or stability of cccDNA either directly or indirectly.
  • a cccDNA formation inhibitor may include, but is not limited to, any compound that inhibits capsid disassembly, rcDNA entry into the nucleus, and/or the conversion of rcDNA into cccDNA.
  • the inhibitor detectably inhibits the formation and/or stability of the cccDNA as measured, e.g., using an assay described herein. In certain embodiments, the inhibitor inhibits the formation and/or stability of cccDNA by at least 5%, at least 10%, at least 20%, at least 50%, at least 75%, or at least 90%.
  • Reported cccDNA formation inhibitors include, but are not limited to, compounds described in Int’l Patent Application Publication No. WO 2013130703, and are incorporated herein in their entirety by reference.
  • reported cccDNA formation inhibitors include, but are not limited to, those generally and specifically described in U.S. Patent Application Publication No.
  • RNA destabilizer refers to a molecule, or a salt or solvate thereof, that reduces the total amount of HBV RNA in mammalian cell culture or in a live human subject.
  • an RNA destabilizer reduces the amount of the RNA transcript(s) encoding one or more of the following HBV proteins: surface antigen, core protein, RNA polymerase, and e antigen.
  • the RNA destabilizer reduces the total amount of HBV RNA in mammalian cell culture or in a live human subject by at least 5%, at least 10%, at least 20%, at least 50%, at least 75%, or at least 90%.
  • Reported RNA destabilizers include compounds described in U.S. Patent No. 8,921,381, as well as compounds described in U.S. Patent Application Publication Nos. US 2015/0087659 and US 2013/0303552, all of which are incorporated herein in their entireties by reference.
  • reported RNA destabilizers include, but are not limited to, those generally and specifically described in Int’l Patent Application Publication Nos.
  • Oligomeric Nucleotides Targeted Against the HBV Genome Reported oligomeric nucleotides targeted against the HBV genome include, but are not limited to, Arrowhead-ARC-520 (see U.S.
  • the oligomeric nucleotides can be designed to target one or more genes and/or transcripts of the HBV genome.
  • Oligomeric nucleotide targeted to the HBV genome also include, but are not limited to, isolated, double stranded, siRNA molecules, that each include a sense strand and an antisense strand that is hybridized to the sense strand.
  • the siRNA target one or more genes and/or transcripts of the HBV genome.
  • checkpoint inhibitor includes any compound that is capable of inhibiting immune checkpoint molecules that are regulators of the immune system (e.g., stimulate or inhibit immune system activity). For example, some checkpoint inhibitors block inhibitory checkpoint molecules, thereby stimulating immune system function, such as stimulation of T cell activity against cancer cells.
  • a non-limiting example of a checkpoint inhibitor is a PD-L1 inhibitor.
  • PD-L1 inhibitor includes any compound that is capable of inhibiting the expression and/or function of the protein Programmed Death-Ligand 1 (PD-L1) either directly or indirectly.
  • PD-L1 also known as cluster of differentiation 274 (CD274) or B7 homolog 1 (B7-H1), is a type 1 transmembrane protein that plays a major role in suppressing the adaptive arm of immune system during pregnancy, tissue allograft transplants, autoimmune disease, and hepatitis.
  • PD-L1 binds to its receptor, the inhibitory checkpoint molecule PD-1 (which is found on activated T cells, B cells, and myeloid cells) so as to modulate activation or inhibition of the adaptive arm of immune system.
  • the PD-L1 inhibitor inhibits the expression and/or function of PD-L1 by at least 5%, at least 10%, at least 20%, at least 50%, at least 75%, or at least 90%.
  • Reported PD-L1 inhibitors include, but are not limited to, compounds recited in one of the following patent application publications: US 2018/0057455; US 2018/0057486; WO 2017/106634; WO 2018/026971; WO 2018/045142; WO 2018/118848; WO 2018/119221; WO 2018/119236; WO 2018/119266; WO 2018/119286; WO 2018/121560; WO 2019/076343; WO 2019/087214; and are incorporated herein in their entirety by reference.
  • GalNAc-siRNA Conjugates Targeted Against an HBV Gene Transcript “GalNAc” is the abbreviation for N-acetylgalactosamine, and "siRNA” is the abbreviation for small interfering RNA.
  • An siRNA that targets an HBV gene transcript is covalently bonded to GalNAc in a GalNAc-siRNA conjugate useful in the practice of the present disclosure. While not wishing to be bound by theory, it is believed that GalNAc binds to asialoglycoprotein receptors on hepatocytes thereby facilitating the targeting of the siRNA to the hepatocytes that are infected with HBV.
  • siRNA enter the infected hepatocytes and stimulate destruction of HBV gene transcripts by the phenomenon of RNA interference.
  • Examples of GalNAc-siRNA conjugates useful in the practice of this aspect of the present disclosure are set forth in published international application PCT/CA2017/050447 (PCT Application Publication number WO/2017/177326, published on October 19, 2017) which is hereby incorporated by reference in its entirety.
  • Therapeutic Vaccines In certain embodiments, administration of a therapeutic vaccine is useful in the practice of the present disclosure for the treatment of a viral disease in a subject.
  • the viral disease is a hepatitis virus.
  • the hepatitis virus is at least one selected from the group consisting of hepatitis B virus (HBV) and hepatitis D virus (HDV).
  • the subject is a human.
  • a synergistic effect may be calculated, for example, using suitable methods such as, for example, the Sigmoid-E max equation (Holford & Scheiner, 1981, Clin. Pharmacokinet. 6:429-453), the equation of Loewe additivity (Loewe & Muischnek, 1926, Arch. Exp. Pathol Pharmacol.114: 313-326) and the median-effect equation (Chou & Talalay, 1984, Adv. Enzyme Regul.22:27-55).
  • Each equation referred to elsewhere herein may be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination.
  • the corresponding graphs associated with the equations referred to elsewhere herein are the concentration-effect curve, isobologram curve and combination index curve, respectively.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatography such as high pressure liquid chromatograpy (HPLC), gas chromatography (GC), gel-permeation chromatography (GPC), or thin layer chromatography (TLC).
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatography such as high pressure liquid chromatograpy (HPLC), gas chromatography (GC), gel-permeation chromatography (GPC), or thin layer chromatography (TLC).
  • HPLC high pressure liquid chromatograpy
  • GC gas chromatography
  • GPC gel-permeation chromatography
  • the chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in Organic Synthesis, 2d. Ed. (Wiley & Sons, 1991), the entire disclosure of which is incorporated by reference herein for all purposes.
  • the reactions or the processes described herein can be carried out in suitable solvents that can be readily selected by one skilled in the art of organic synthesis. Suitable solvents typically are substantially nonreactive with the reactants, intermediates, and/or products at the temperatures at which the reactions are carried out, i.e., temperatures that can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • a compound of formula (I) can be prepared, for example, according to the synthetic methods outlined in Schemes 1-12.
  • R b1 and R b2 are each independently selected from the group consisting of H, C 1 -C 3 alkyl, C 3
  • Reductive amination of compound 1-4 with an amine 1-5 provides compound 1-6.
  • a suitable reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride optionally in the presence of acetic acid or sodium acetate
  • the amine can optionally be protected with a tert-butyloxycarbonyl (Boc) protecting group (e.g. by reaction with di-tert-butyl dicarbonate in the presence of a tertiary amine such as triethylamine).
  • Boc tert-butyloxycarbonyl
  • Compound 1-6 is then coupled with a boronic acid or boronic acid pinacol ester compound 1-7 under standard metal-catalyzed coupling conditions (e.g.
  • compound 1-8 is then further coupled with a boronic acid or boronic acid pinacol ester compound 1-9 under standard metal-catalyzed coupling conditions (e.g. using a suitable palladium catalyst and a suitable base) to give compound 1-10.
  • a suitable palladium catalyst and a suitable base e.g. using a suitable palladium catalyst and a suitable base
  • Reaction of compound 1-10 with amine compound 1-11 under reductive amination conditions e.g. using a suitable reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride optionally in the presence of acetic acid or sodium acetate
  • a suitable reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride optionally in the presence of acetic acid or sodium acetate
  • compound 3-3 contains a secondary amine, the amine can optionally be protected with a tert- butyloxycarbonyl (Boc) protecting group (e.g. by reaction with di-tert-butyl dicarbonate in the presence of a tertiary amine such as triethylamine). Borylation of compound 3-3 (e.g.
  • Scheme 3 An alternative synthesis of compounds of formula (I) is shown in Scheme 4.
  • Compound 4-2 is then coupled with a boronic acid or boronic acid pinacol ester compound 4-3 under standard metal-catalyzed coupling conditions (e.g. using a suitable palladium catalyst and a suitable base) to give compound 4-4.
  • Compound 4-4 is then further coupled with a boronic acid or boronic acid pinacol ester compound 4-5 under standard metal-catalyzed coupling conditions (e.g. using a suitable palladium catalyst and a suitable base) to give compound 4-6.
  • compound 6-3 contains a secondary amine, the amine can optionally be protected with a tert- butyloxycarbonyl (Boc) protecting group (e.g. by reaction with di-tert-butyl dicarbonate in the presence of a tertiary amine such as triethylamine).
  • Boc tert- butyloxycarbonyl
  • Compound 6-3 is then coupled with the boronic acid or boronic acid pinacol ester compound 6-4 under standard metal-catalyzed coupling conditions (e.g. using a suitable palladium catalyst and a suitable base) to give compound 6-5.
  • the Boc protecting group can be removed by treatment with trifluoroacetic acid in dichloromethane or hydrochloric acid in 1,4-dioxane.
  • Aromatic amine compound 7-4 is coupled with a carboxylic acid (or corresponding lithium salt) compound 7-5 under amide coupling conditions (e.g. using a suitable coupling agent such as HATU, 1,1'- carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N-diisopropylethylamine). Hydrolysis of the acetal or ketal (e.g. under acidic conditions with aqueous hydrochloric acid or trifluoroacetic acid) provides compound 7-6.
  • a suitable coupling agent such as HATU, 1,1'- carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N-diisopropylethylamine.
  • Hydrolysis of the acetal or ketal e.g. under acid
  • Compound 7-3 is then coupled with the boronic acid or boronic acid pinacol ester compound 7-6 under standard metal-catalyzed coupling conditions (e.g. using a suitable palladium catalyst and a suitable base) to give compound 7-7.
  • Reaction of compound 7-7 with amine compound 7-8 under reductive amination conditions e.g. using a suitable reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride optionally in the presence of acetic acid or sodium acetate
  • a suitable reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride optionally in the presence of acetic acid or sodium acetate
  • the Boc protecting group can be removed by treatment with trifluoroacetic acid in dichloromethane or hydrochloric acid in 1,4-dioxane.
  • Scheme 7 An alternative synthesis of compounds of formula (I) is shown in Scheme 8.
  • Compound 8-1 is coupled with the boronic acid or boronic acid pinacol ester compound 8-2 under standard metal-catalyzed coupling conditions (e.g. using a suitable palladium catalyst and a suitable base) to give compound 8-3.
  • Compound 8-4 is reacted with amine compound 8-5 under reductive amination conditions (e.g. using a suitable reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride optionally in the presence of acetic acid or sodium acetate) to give amine compound 8-6.
  • a suitable reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride optionally in the presence of acetic acid or sodium acetate
  • compound 8-6 contains a secondary amine
  • the amine can optionally be protected with a tert-butyloxycarbonyl (Boc) protecting group (e.g. by reaction with di-tert-butyl dicarbonate in the presence of a tertiary amine such as triethylamine).
  • Hydrolysis e.g. using lithium hydroxide in aqueous alcoholic solvent
  • Aromatic amine compound 8-3 is then coupled with the carboxylic acid (or corresponding lithium salt) compound 8-7 under amide coupling conditions (e.g.
  • a suitable coupling agent such as HATU, 1,1'-carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N-diisopropylethylamine
  • a suitable coupling agent such as HATU, 1,1'-carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N-diisopropylethylamine
  • a suitable coupling agent such as HATU, 1,1'-carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N-diisopropylethylamine
  • a vinyl boronic acid or boronic acid pinacol ester under standard metal-catalyzed
  • carboxylic acid compound 10-4 Coupling of carboxylic acid (or corresponding lithium salt) compound 10-4 with aromatic amine compound 10-5 under amide coupling conditions (e.g. using a suitable coupling agent such as HATU, 1,1'-carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N- diisopropylethylamine) then gives compound 10-6. If intermediates 10-3 or 10-5 were protected as the Boc amine, the Boc protecting group/s can be removed by treatment with trifluoroacetic acid in dichloromethane or hydrochloric acid in 1,4-dioxane.
  • a suitable coupling agent such as HATU, 1,1'-carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N- diisopropyleth
  • Aromatic amine compound 11-1 is coupled with a carboxylic acid (or corresponding lithium salt) compound 11-2 under amide coupling conditions (e.g. using a suitable coupling agent such as HATU, 1,1'-carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N-diisopropylethylamine) to provide compound 11-3.
  • a suitable coupling agent such as HATU, 1,1'-carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N-diisopropylethylamine
  • the alcohol group in compound 11-3 is then oxidized (e.g.
  • Scheme 12 An alternative synthesis of compounds of formula (I) is shown in Scheme 12.
  • Aromatic amine compound 12-1 is coupled with a carboxylic acid (or corresponding lithium salt) compound 12-2 under amide coupling conditions (e.g., using a suitable coupling agent such as HATU, 1,1'-carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N-diisopropylethylamine) to provide compound 12-3.
  • a suitable coupling agent such as HATU, 1,1'-carbonyldiimidazole or 2-chloro-1-methylpyridinium iodide (Mukaiyama reagent) and a suitable base such as N,N-diisopropylethylamine
  • Compound 12-3 is then coupled with a vinyl boronic acid or boronic acid pinacol ester under standard metal-catalyzed coupling conditions (e.g., using a suitable palladium catalyst and a suitable base) to give compound 12-4.
  • Oxidative cleavage e.g., using osmium tetroxide in the presence of sodium periodate or ozone followed by triphenylphosphine
  • alkene in compound 12-4 then gives aldehyde compound 12-5.
  • Reaction of compound 12-5 with amine compound 12-6 under reductive amination conditions e.g., using a suitable reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride optionally in the presence of acetic acid or sodium acetate
  • a suitable reducing agent such as sodium cyanoborohydride or sodium triacetoxyborohydride optionally in the presence of acetic acid or sodium acetate
  • the Boc protecting group can be removed by treatment with trifluoroacetic acid in dichloromethane or hydrochloric acid in 1,4-dioxane.
  • Methods The disclosure provides a method of treating, ameliorating, and/or preventing hepatitis virus infection in a subject.
  • the infection comprises hepatitis B virus (HBV) and/or hepatitis D virus (HDV) infection.
  • HBV hepatitis B virus
  • HDV hepatitis D virus
  • the infection comprises hepatitis B virus (HBV) infection. In yet other embodiments, the infection comprises hepatitis D virus (HDV) infection. In yet other embodiments, the method comprises administering to the subject in need thereof a therapeutically effective amount of at least one compound of the disclosure. In yet other embodiments, the compound of the disclosure is the only antiviral agent administered to the subject. In yet other embodiments, the at least one compound is administered to the subject in a pharmaceutically acceptable composition. In yet other embodiments, the subject is further administered at least one additional agent useful for treating the hepatitis virus infection.
  • the at least one additional agent comprises at least one selected from the group consisting of reverse transcriptase inhibitors; capsid inhibitors; cccDNA formation inhibitors; RNA destabilizers; oligomeric nucleotides targeted against the HBV genome; immunostimulators; GalNAc-siRNA conjugates targeted against an HBV gene transcript; and therapeutic vaccines.
  • the subject is co-administered the at least one compound and the at least one additional agent.
  • the at least one compound and the at least one additional agent are coformulated.
  • the disclosure further provides a method of treating, ameliorating, and/or preventing cancer in a subject.
  • the method comprises administering to the subject in need thereof a therapeutically effective amount of at least one compound of the disclosure.
  • the compound of the disclosure is the only anticancer agent administered to the subject.
  • the at least one compound is administered to the subject in a pharmaceutically acceptable composition.
  • the subject is further administered at least one additional agent or therapy useful for treating, ameliorating, or preventing the cancer.
  • the additional anticancer agent or therapy comprises nivolumab, pembrolizumab, atezolizumab, ipilimumab, chemotherapy, radiation therapy, and/or resection therapy.
  • the additional anticancer agent or therapy comprises rituximab, doxorubicin, gemcitabine, nivolumab, pembrolizumab, and/or ipilimumab.
  • the cancer is amenable to treatment by inhibiting PD-1, PD- L1 or the PD-1/PD-L1 interaction.
  • the cancer is at least one of pancreatic cancer, bladder cancer, colorectal cancer, breast cancer, prostate cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, gastric cancer, esophageal cancer, head and neck cancer, melanoma, neuroendocrine cancer, CNS cancer, brain cancer, bone cancer, soft tissue sarcoma, non-small cell lung cancer, small-cell lung cancer, or colon cancer.
  • the cancer is at least one of lymphoma, multiple myeloma, or leukemia.
  • the cancer is at least one of acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), myelodysplastic syndrome (MDS), myeloproliferative disease (MPD), chronic myeloid leukemia (CML), multiple myeloma (MM), non-Hodgkin's lymphoma (NHL), mantle cell lymphoma (MCL), follicular lymphoma, Waldestrom's macroglobulinemia (WM), T-cell lymphoma, B-cell lymphoma and diffuse large B-cell lymphoma (DLBCL).
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • MDS myelodysplastic syndrome
  • MDS myeloproliferative disease
  • compositions and Formulations The disclosure provides pharmaceutical compositions comprising at least one compound of the disclosure or a salt or solvate thereof, which are useful to practice methods of the disclosure.
  • a pharmaceutical composition may consist of at least one compound of the disclosure or a salt or solvate thereof, in a form suitable for administration to a subject, or the pharmaceutical composition may comprise at least one compound of the disclosure or a salt or solvate thereof, and one or more pharmaceutically acceptable carriers, one or more additional ingredients, or some combination of these.
  • At least one compound of the disclosure may be present in the pharmaceutical composition in the form of a physiologically acceptable salt, such as in combination with a physiologically acceptable cation or anion, as is well known in the art.
  • the pharmaceutical compositions useful for practicing the method of the disclosure may be administered to deliver a dose of between 1 ng/kg/day and 100 mg/kg/day.
  • the pharmaceutical compositions useful for practicing the disclosure may be administered to deliver a dose of between 1 ng/kg/day and 1,000 mg/kg/day.
  • the relative amounts of the active ingredient, the pharmaceutically acceptable carrier, and any additional ingredients in a pharmaceutical composition of the disclosure will vary, depending upon the identity, size, and condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100% (w/w) active ingredient.
  • Pharmaceutical compositions that are useful in the methods of the disclosure may be suitably developed for nasal, inhalational, oral, rectal, vaginal, pleural, peritoneal, parenteral, topical, transdermal, pulmonary, intranasal, buccal, ophthalmic, epidural, intrathecal, intravenous or another route of administration.
  • a composition useful within the methods of the disclosure may be directly administered to the brain, the brainstem, or any other part of the central nervous system of a mammal or bird.
  • compositions of the disclosure are part of a pharmaceutical matrix, which allows for manipulation of insoluble materials and improvement of the bioavailability thereof, development of controlled or sustained release products, and generation of homogeneous compositions.
  • a pharmaceutical matrix may be prepared using hot melt extrusion, solid solutions, solid dispersions, size reduction technologies, molecular complexes (e.g., cyclodextrins, and others), microparticulate, and particle and formulation coating processes.
  • Amorphous or crystalline phases may be used in such processes.
  • the route(s) of administration will be readily apparent to the skilled artisan and will depend upon any number of factors including the type and severity of the disease being treated, the type and age of the veterinary or human patient being treated, and the like.
  • the formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology and pharmaceutics. In general, such preparatory methods include the step of bringing the active ingredient into association with a carrier or one or more other accessory ingredients, and then, if necessary or desirable, shaping or packaging the product into a desired single-dose or multi-dose unit.
  • a "unit dose" is a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient that would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one- third of such a dosage.
  • the unit dosage form may be for a single daily dose or one of multiple daily doses (e.g., about 1 to 4 or more times per day). When multiple daily doses are used, the unit dosage form may be the same or different for each dose.
  • compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and perform such modification with merely ordinary, if any, experimentation.
  • Subjects to which administration of the pharmaceutical compositions of the disclosure is contemplated include, but are not limited to, humans and other primates, mammals including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, and dogs.
  • the compositions of the disclosure are formulated using one or more pharmaceutically acceptable excipients or carriers.
  • the pharmaceutical compositions of the disclosure comprise a therapeutically effective amount of at least one compound of the disclosure and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers include, but are not limited to, glycerol, water, saline, ethanol, recombinant human albumin (e.g., RECOMBUMIN®), solubilized gelatins (e.g., GELOFUSINE®), and other pharmaceutically acceptable salt solutions such as phosphates and salts of organic acids. Examples of these and other pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (1991, Mack Publication Co., New Jersey).
  • the carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), recombinant human albumin, solubilized gelatins, suitable mixtures thereof, and vegetable oils.
  • the proper fluidity may be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms may be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol
  • Prolonged absorption of the injectable compositions may be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate or gelatin.
  • Formulations may be employed in admixtures with conventional excipients, i.e., pharmaceutically acceptable organic or inorganic carrier substances suitable for oral, parenteral, nasal, inhalational, intravenous, subcutaneous, transdermal enteral, or any other suitable mode of administration, known to the art.
  • the pharmaceutical preparations may be sterilized and if desired mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure buffers, coloring, flavoring and/or fragrance-conferring substances and the like. They may also be combined where desired with other active agents, e.g., other analgesic, anxiolytics or hypnotic agents.
  • additional ingredients include, but are not limited to, one or more ingredients that may be used as a pharmaceutical carrier.
  • the composition of the disclosure may comprise a preservative from about 0.005% to 2.0% by total weight of the composition.
  • the preservative is used to prevent spoilage in the case of exposure to contaminants in the environment.
  • preservatives useful in accordance with the disclosure include but are not limited to those selected from the group consisting of benzyl alcohol, sorbic acid, parabens, imidurea and combinations thereof.
  • One such preservative is a combination of about 0.5% to 2.0% benzyl alcohol and 0.05% to 0.5% sorbic acid.
  • the composition may include an antioxidant and a chelating agent which inhibit the degradation of the compound.
  • Antioxidants for some compounds are BHT, BHA, alpha- tocopherol and ascorbic acid in the exemplary range of about 0.01% to 0.3%, or BHT in the range of 0.03% to 0.1% by weight by total weight of the composition.
  • the chelating agent may be present in an amount of from 0.01% to 0.5% by weight by total weight of the composition.
  • Exemplary chelating agents include edetate salts (e.g. disodium edetate) and citric acid in the weight range of about 0.01% to 0.20%, or in the range of 0.02% to 0.10% by weight by total weight of the composition.
  • the chelating agent is useful for chelating metal ions in the composition that may be detrimental to the shelf life of the formulation. While BHT and disodium edetate are exemplary antioxidant and chelating agent, respectively, for some compounds, other suitable and equivalent antioxidants and chelating agents may be substituted therefore as would be known to those skilled in the art.
  • Liquid suspensions may be prepared using conventional methods to achieve suspension of the active ingredient in an aqueous or oily vehicle.
  • Aqueous vehicles include, for example, water, and isotonic saline.
  • Oily vehicles include, for example, almond oil, oily esters, ethyl alcohol, vegetable oils such as arachis, olive, sesame, or coconut oil, fractionated vegetable oils, and mineral oils such as liquid paraffin.
  • Liquid suspensions may further comprise one or more additional ingredients including, but not limited to, suspending agents, dispersing or wetting agents, emulsifying agents, demulcents, preservatives, buffers, salts, flavorings, coloring agents, and sweetening agents.
  • Oily suspensions may further comprise a thickening agent.
  • suspending agents include, but are not limited to, sorbitol syrup, hydrogenated edible fats, sodium alginate, polyvinylpyrrolidone, gum tragacanth, gum acacia, and cellulose derivatives such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl cellulose.
  • Known dispersing or wetting agents include, but are not limited to, naturally-occurring phosphatides such as lecithin, condensation products of an alkylene oxide with a fatty acid, with a long chain aliphatic alcohol, with a partial ester derived from a fatty acid and a hexitol, or with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene stearate, heptadecaethyleneoxycetanol, polyoxyethylene sorbitol monooleate, and polyoxyethylene sorbitan monooleate, respectively).
  • naturally-occurring phosphatides such as lecithin
  • condensation products of an alkylene oxide with a fatty acid with a long chain aliphatic alcohol
  • with a partial ester derived from a fatty acid and a hexitol or with a partial ester derived from a fatty acid and a hexito
  • emulsifying agents include, but are not limited to, lecithin, acacia, and ionic or non ionic surfactants.
  • Known preservatives include, but are not limited to, methyl, ethyl, or n-propyl para-hydroxybenzoates, ascorbic acid, and sorbic acid.
  • Known sweetening agents include, for example, glycerol, propylene glycol, sorbitol, sucrose, and saccharin.
  • Liquid solutions of the active ingredient in aqueous or oily solvents may be prepared in substantially the same manner as liquid suspensions, the primary difference being that the active ingredient is dissolved, rather than suspended in the solvent.
  • an "oily" liquid is one which comprises a carbon-containing liquid molecule and which exhibits a less polar character than water.
  • Liquid solutions of the pharmaceutical composition of the disclosure may comprise each of the components described with regard to liquid suspensions, it being understood that suspending agents will not necessarily aid dissolution of the active ingredient in the solvent.
  • Aqueous solvents include, for example, water, and isotonic saline.
  • Oily solvents include, for example, almond oil, oily esters, ethyl alcohol, vegetable oils such as arachis, olive, sesame, or coconut oil, fractionated vegetable oils, and mineral oils such as liquid paraffin. Powdered and granular formulations of a pharmaceutical preparation of the disclosure may be prepared using known methods.
  • Such formulations may be administered directly to a subject, used, for example, to form tablets, to fill capsules, or to prepare an aqueous or oily suspension or solution by addition of an aqueous or oily vehicle thereto.
  • Each of these formulations may further comprise one or more of dispersing or wetting agent, a suspending agent, ionic and non-ionic surfactants, and a preservative. Additional excipients, such as fillers and sweetening, flavoring, or coloring agents, may also be included in these formulations.
  • a pharmaceutical composition of the disclosure may also be prepared, packaged, or sold in the form of oil-in-water emulsion or a water-in-oil emulsion.
  • the oily phase may be a vegetable oil such as olive or arachis oil, a mineral oil such as liquid paraffin, or a combination of these.
  • Such compositions may further comprise one or more emulsifying agents such as naturally occurring gums such as gum acacia or gum tragacanth, naturally- occurring phosphatides such as soybean or lecithin phosphatide, esters or partial esters derived from combinations of fatty acids and hexitol anhydrides such as sorbitan monooleate, and condensation products of such partial esters with ethylene oxide such as polyoxyethylene sorbitan monooleate.
  • These emulsions may also contain additional ingredients including, for example, sweetening or flavoring agents.
  • Methods for impregnating or coating a material with a chemical composition include, but are not limited to methods of depositing or binding a chemical composition onto a surface, methods of incorporating a chemical composition into the structure of a material during the synthesis of the material (i.e., such as with a physiologically degradable material), and methods of absorbing an aqueous or oily solution or suspension into an absorbent material, with or without subsequent drying.
  • Methods for mixing components include physical milling, the use of pellets in solid and suspension formulations and mixing in a transdermal patch, as known to those skilled in the art.
  • Administration/Dosing The regimen of administration may affect what constitutes an effective amount.
  • the therapeutic formulations may be administered to the patient either prior to or after the onset of a disease or disorder. Further, several divided dosages, as well as staggered dosages may be administered daily or sequentially, or the dose may be continuously infused, or may be a bolus injection. Further, the dosages of the therapeutic formulations may be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation. Administration of the compositions of the present disclosure to a patient, such as a mammal, such as a human, may be carried out using known procedures, at dosages and for periods of time effective to treat a disease or disorder contemplated herein.
  • an effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the activity of the particular compound employed; the time of administration; the rate of excretion of the compound; the duration of the treatment; other drugs, compounds or materials used in combination with the compound; the state of the disease or disorder, age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well-known in the medical arts. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • a non-limiting example of an effective dose range for a therapeutic compound of the disclosure is from about 0.01 mg/kg to 100 mg/kg of body weight/per day.
  • the compound may be administered to an animal as frequently as several times daily, or it may be administered less frequently, such as once a day, once a week, once every two weeks, once a month, or even less frequently, such as once every several months or even once a year or less. It is understood that the amount of compound dosed per day may be administered, in non-limiting examples, every day, every other day, every 2 days, every 3 days, every 4 days, or every 5 days.
  • a 5 mg per day dose may be initiated on Monday with a first subsequent 5 mg per day dose administered on Wednesday, a second subsequent 5 mg per day dose administered on Friday, and so on.
  • the frequency of the dose is readily apparent to the skilled artisan and depends upon a number of factors, such as, but not limited to, type and severity of the disease being treated, and type and age of the animal.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this disclosure may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • a medical doctor e.g., physician or veterinarian, having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the patients 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.
  • compositions of the disclosure are administered to the patient in dosages that range from one to five times per day or more.
  • compositions of the disclosure are administered to the patient in range of dosages that include, but are not limited to, once every day, every two days, every three days to once a week, and once every two weeks.
  • Compounds of the disclosure for administration may be in the range of from about 1 ⁇ g to about 7,500 mg, about 20 ⁇ g to about 7,000 mg, about 40 ⁇ g to about 6,500 mg, about 80 ⁇ g to about 6,000 mg, about 100 ⁇ g to about 5,500 mg, about 200 ⁇ g to about 5,000 mg, about 400 ⁇ g to about 4,000 mg, about 800 ⁇ g to about 3,000 mg, about 1 mg to about 2,500 mg, about 2 mg to about 2,000 mg, about 5 mg to about 1,000 mg, about 10 mg to about 750 mg, about 20 mg to about 600 mg, about 30 mg to about 500 mg, about 40 mg to about 400 mg, about 50 mg to about 300 mg, about 60 mg to about 250 mg, about 70 mg to about 200 mg, about 80 mg to about 150 mg, and any and all whole or partial increments there-in- between.
  • the dose of a compound of the disclosure is from about 0.5 ⁇ g and about 5,000 mg. In some embodiments, a dose of a compound of the disclosure used in compositions described herein is less than about 5,000 mg, or less than about 4,000 mg, or less than about 3,000 mg, or less than about 2,000 mg, or less than about 1,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 200 mg, or less than about 50 mg.
  • a dose of a second compound as described herein is less than about 1,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 400 mg, or less than about 300 mg, or less than about 200 mg, or less than about 100 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 25 mg, or less than about 20 mg, or less than about 15 mg, or less than about 10 mg, or less than about 5 mg, or less than about 2 mg, or less than about 1 mg, or less than about 0.5 mg, and any and all whole or partial increments thereof.
  • the present disclosure is directed to a packaged pharmaceutical composition
  • a packaged pharmaceutical composition comprising a container holding a therapeutically effective amount of a compound of the disclosure, alone or in combination with a second pharmaceutical agent; and instructions for using the compound to treat, prevent, or reduce one or more symptoms of a disease or disorder in a patient.
  • the term "container" includes any receptacle for holding the pharmaceutical composition or for managing stability or water uptake.
  • the container is the packaging that contains the pharmaceutical composition, such as liquid (solution and suspension), semisolid, lyophilized solid, solution and powder or lyophilized formulation present in dual chambers.
  • the container is not the packaging that contains the pharmaceutical composition, i.e., the container is a receptacle, such as a box or vial that contains the packaged pharmaceutical composition or unpackaged pharmaceutical composition and the instructions for use of the pharmaceutical composition.
  • packaging techniques are well known in the art. It should be understood that the instructions for use of the pharmaceutical composition may be contained on the packaging containing the pharmaceutical composition, and as such the instructions form an increased functional relationship to the packaged product. However, it should be understood that the instructions may contain information pertaining to the compound's ability to perform its intended function, e.g., treating, preventing, or reducing a disease or disorder in a patient.
  • compositions of the disclosure include inhalational, oral, nasal, rectal, parenteral, sublingual, 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, epidural, intrapleural, intraperitoneal, subcutaneous, intramuscular, intradermal, intra-arterial, intravenous, intrabronchial, inhalation, and topical administration.
  • inhalational e.g., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and perivaginally), intravesical, intrapulmonary, intraduodenal, intragastrical, intrathecal, epidural, intrapleural, intraperitoneal, subcutaneous, intramuscular
  • compositions and dosage forms include, for example, tablets, capsules, caplets, pills, gel caps, troches, emulsions, 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 intravesical administration and the like. It should be understood that the formulations and compositions that would be useful in the present disclosure are not limited to the particular formulations and compositions that are described herein.
  • compositions intended for oral use may be prepared according to any method known in the art and such compositions may contain one or more agents selected from the group consisting of inert, non-toxic, generally recognized as safe (GRAS) pharmaceutically excipients which are suitable for the manufacture of tablets.
  • GRAS inert, non-toxic, generally recognized as safe
  • excipients include, for example an inert diluent such as lactose; granulating and disintegrating agents such as cornstarch; binding agents such as starch; and lubricating agents such as magnesium stearate.
  • Tablets may be non-coated or they may be coated using known methods to achieve delayed disintegration in the gastrointestinal tract of a subject, thereby providing sustained release and absorption of the active ingredient.
  • a material such as glyceryl monostearate or glyceryl distearate may be used to coat tablets.
  • tablets may be coated using methods described in U.S. Patents Nos.4,256,108; 4,160,452; and 4,265,874 to form osmotically controlled release tablets.
  • Tablets may further comprise a sweetening agent, a flavoring agent, a coloring agent, a preservative, or some combination of these in order to provide for pharmaceutically elegant and palatable preparation.
  • Hard capsules comprising the active ingredient may be made using a physiologically degradable composition, such as gelatin.
  • the capsules comprise the active ingredient, and may further comprise additional ingredients including, for example, an inert solid diluent such as calcium carbonate, calcium phosphate, or kaolin.
  • Hard capsules comprising the active ingredient may be made using a physiologically degradable composition, such as gelatin.
  • Such hard capsules comprise the active ingredient, and may further comprise additional ingredients including, for example, an inert solid diluent such as calcium carbonate, calcium phosphate, or kaolin.
  • Soft gelatin capsules comprising the active ingredient may be made using a physiologically degradable composition, such as gelatin from animal-derived collagen or from a hypromellose, a modified form of cellulose, and manufactured using optional mixtures of gelatin, water and plasticizers such as sorbitol or glycerol.
  • Such soft capsules comprise the active ingredient, which may be mixed with water or an oil medium such as peanut oil, liquid paraffin, or olive oil.
  • the compounds of the disclosure may be in the form of tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents; fillers; lubricants; disintegrates; or wetting agents.
  • the tablets may be coated using suitable methods and coating materials such as OPADRY® film coating systems available from Colorcon, West Point, Pa. (e.g., OPADRY® OY Type, OYC Type, Organic Enteric OY-P Type, Aqueous Enteric OY-A Type, OY-PM Type and OPADRY® White, 32K18400). It is understood that similar type of film coating or polymeric products from other companies may be used.
  • a tablet comprising the active ingredient may, for example, be made by compressing or molding the active ingredient, optionally with one or more additional ingredients.
  • Compressed tablets may be prepared by compressing, in a suitable device, the active ingredient in a free-flowing form such as a powder or granular preparation, optionally mixed with one or more of a binder, a lubricant, an excipient, a surface active agent, and a dispersing agent. Molded tablets may be made by molding, in a suitable device, a mixture of the active ingredient, a pharmaceutically acceptable carrier, and at least sufficient liquid to moisten the mixture.
  • Pharmaceutically acceptable excipients used in the manufacture of tablets include, but are not limited to, inert diluents, granulating and disintegrating agents, binding agents, and lubricating agents.
  • Known dispersing agents include, but are not limited to, potato starch and sodium starch glycolate.
  • Known surface-active agents include, but are not limited to, sodium lauryl sulphate.
  • Known diluents include, but are not limited to, calcium carbonate, sodium carbonate, lactose, microcrystalline cellulose, calcium phosphate, calcium hydrogen phosphate, and sodium phosphate.
  • Known granulating and disintegrating agents include, but are not limited to, corn starch and alginic acid.
  • Known binding agents include, but are not limited to, gelatin, acacia, pre-gelatinized maize starch, polyvinylpyrrolidone, and hydroxypropyl methylcellulose.
  • Known lubricating agents include, but are not limited to, magnesium stearate, stearic acid, silica, and talc.
  • Granulating techniques are well known in the pharmaceutical art for modifying starting powders or other particulate materials of an active ingredient.
  • the powders are typically mixed with a binder material into larger permanent free-flowing agglomerates or granules referred to as a "granulation.”
  • solvent-using "wet" granulation processes are generally characterized in that the powders are combined with a binder material and moistened with water or an organic solvent under conditions resulting in the formation of a wet granulated mass from which the solvent must then be evaporated.
  • Melt granulation generally consists in the use of materials that are solid or semi-solid at room temperature (i.e., having a relatively low softening or melting point range) to promote granulation of powdered or other materials, essentially in the absence of added water or other liquid solvents.
  • the low melting solids when heated to a temperature in the melting point range, liquefy to act as a binder or granulating medium.
  • the liquefied solid spreads itself over the surface of powdered materials with which it is contacted, and on cooling, forms a solid granulated mass in which the initial materials are bound together.
  • the resulting melt granulation may then be provided to a tablet press or be encapsulated for preparing the oral dosage form.
  • melt granulation improves the dissolution rate and bioavailability of an active (i.e., drug) by forming a solid dispersion or solid solution.
  • U.S. Patent No.5,169,645 discloses directly compressible wax-containing granules having improved flow properties. The granules are obtained when waxes are admixed in the melt with certain flow improving additives, followed by cooling and granulation of the admixture. In certain embodiments, only the wax itself melts in the melt combination of the wax(es) and additives(s), and in other cases both the wax(es) and the additives(s) will melt.
  • the present disclosure also includes a multi-layer tablet comprising a layer providing for the delayed release of one or more compounds useful within the methods of the disclosure, and a further layer providing for the immediate release of one or more compounds useful within the methods of the disclosure.
  • a gastric insoluble composition may be obtained in which the active ingredient is entrapped, ensuring its delayed release.
  • Liquid preparation for oral administration may be in the form of solutions, syrups or suspensions.
  • the liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agent (e.g., lecithin or acacia); non- aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and preservatives (e.g., methyl or propyl para-hydroxy benzoates or sorbic acid).
  • suspending agents e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats
  • emulsifying agent e.g., lecithin or acacia
  • non- aqueous vehicles e.g., almond oil, oily esters or ethyl alcohol
  • preservatives e.g., methyl or propyl para-hydroxy benzoates or sorbic acid
  • parenteral administration of a pharmaceutical composition includes any route of administration characterized by physical breaching of a tissue of a subject and administration of the pharmaceutical composition through the breach in the tissue.
  • Parenteral administration thus includes, but is not limited to, administration of a pharmaceutical composition by injection of the composition, by application of the composition through a surgical incision, by application of the composition through a tissue-penetrating non-surgical wound, and the like.
  • parenteral administration is contemplated to include, but is not limited to, subcutaneous, intravenous, intraperitoneal, intramuscular, intrasternal injection, and kidney dialytic infusion techniques.
  • Formulations of a pharmaceutical composition suitable for parenteral administration comprise the active ingredient combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline.
  • a pharmaceutically acceptable carrier such as sterile water or sterile isotonic saline.
  • Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration.
  • Injectable formulations may be prepared, packaged, or sold in unit dosage form, such as in ampules or in multidose containers containing a preservative. Injectable formulations may also be prepared, packaged, or sold in devices such as patient-controlled analgesia (PCA) devices.
  • PCA patient-controlled analgesia
  • Formulations for parenteral administration include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and implantable sustained-release or biodegradable formulations. Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents.
  • the active ingredient is provided in dry (i.e., powder or granular) form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • the pharmaceutical compositions may be prepared, packaged, or sold in the form of a sterile injectable aqueous or oily suspension or solution.
  • This suspension or solution may be formulated according to the known art, and may comprise, in addition to the active ingredient, additional ingredients such as the dispersing agents, wetting agents, or suspending agents described herein.
  • additional ingredients such as the dispersing agents, wetting agents, or suspending agents described herein.
  • Such sterile injectable formulations may be prepared using a non- toxic parenterally acceptable diluent or solvent, such as water or 1,3-butanediol, for example.
  • a non- toxic parenterally acceptable diluent or solvent such as water or 1,3-butanediol, for example.
  • Other acceptable diluents and solvents include, but are not limited to, Ringer's solution, isotonic sodium chloride solution, and fixed oils such as synthetic mono- or di-glycerides.
  • compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
  • the stratum corneum is a highly resistant layer comprised of protein, cholesterol, sphingolipids, free fatty acids and various other lipids, and includes cornified and living cells.
  • One of the factors that limit the penetration rate (flux) of a compound through the stratum corneum is the amount of the active substance that can be loaded or applied onto the skin surface. The greater the amount of active substance which is applied per unit of area of the skin, the greater the concentration gradient between the skin surface and the lower layers of the skin, and in turn the greater the diffusion force of the active substance through the skin.
  • Formulations suitable for topical administration include, but are not limited to, liquid or semi-liquid preparations such as liniments, lotions, oil-in-water or water-in-oil emulsions such as creams, ointments or pastes, and solutions or suspensions.
  • Topically administrable formulations may, for example, comprise from about 1% to about 10% (w/w) active ingredient, although the concentration of the active ingredient may be as high as the solubility limit of the active ingredient in the solvent.
  • Formulations for topical administration may further comprise one or more of the additional ingredients described herein.
  • Enhancers of permeation may be used. These materials increase the rate of penetration of drugs across the skin. Typical enhancers in the art include ethanol, glycerol monolaurate, PGML (polyethylene glycol monolaurate), dimethylsulfoxide, and the like. Other enhancers include oleic acid, oleyl alcohol, ethoxydiglycol, laurocapram, alkanecarboxylic acids, dimethylsulfoxide, polar lipids, or N-methyl-2-pyrrolidone.
  • One acceptable vehicle for topical delivery of some of the compositions of the disclosure may contain liposomes.
  • the composition of the liposomes and their use are known in the art (i.e., U.S. Patent No.6,323,219).
  • the topically active pharmaceutical composition may be optionally combined with other ingredients such as adjuvants, anti-oxidants, chelating agents, surfactants, foaming agents, wetting agents, emulsifying agents, viscosifiers, buffering agents, preservatives, and the like.
  • a permeation or penetration enhancer is included in the composition and is effective in improving the percutaneous penetration of the active ingredient into and through the stratum corneum with respect to a composition lacking the permeation enhancer.
  • compositions may further comprise a hydrotropic agent, which functions to increase disorder in the structure of the stratum corneum, and thus allows increased transport across the stratum corneum.
  • hydrotropic agents such as isopropyl alcohol, propylene glycol, or sodium xylene sulfonate, are known to those of skill in the art.
  • the topically active pharmaceutical composition should be applied in an amount effective to affect desired changes.
  • a pharmaceutical composition of the disclosure may be prepared, packaged, or sold in a formulation suitable for buccal administration.
  • formulations may, for example, be in the form of tablets or lozenges made using conventional methods, and may contain, for example, 0.1 to 20% (w/w) of the active ingredient, the balance comprising an orally dissolvable or degradable composition and, optionally, one or more of the additional ingredients described herein.
  • formulations suitable for buccal administration may comprise a powder or an aerosolized or atomized solution or suspension comprising the active ingredient.
  • Such powdered, aerosolized, or aerosolized formulations, when dispersed may have an average particle or droplet size in the range from about 0.1 to about 200 nanometers, and may further comprise one or more of the additional ingredients described herein.
  • a pharmaceutical composition of the disclosure may be prepared, packaged, or sold in a formulation suitable for rectal administration.
  • a composition may be in the form of, for example, a suppository, a retention enema preparation, and a solution for rectal or colonic irrigation.
  • Suppository formulations may be made by combining the active ingredient with a non-irritating pharmaceutically acceptable excipient which is solid at ordinary room temperature (i.e., about 20 ⁇ C) and which is liquid at the rectal temperature of the subject (i.e., about 37 ⁇ C in a healthy human).
  • Suitable pharmaceutically acceptable excipients include, but are not limited to, cocoa butter, polyethylene glycols, and various glycerides. Suppository formulations may further comprise various additional ingredients including, but not limited to, antioxidants, and preservatives. Retention enema preparations or solutions for rectal or colonic irrigation may be made by combining the active ingredient with a pharmaceutically acceptable liquid carrier. As is well known in the art, enema preparations may be administered using, and may be packaged within, a delivery device adapted to the rectal anatomy of the subject. Enema preparations may further comprise various additional ingredients including, but not limited to, antioxidants, and preservatives. Additional Administration Forms Additional dosage forms of this disclosure include dosage forms as described in U.S.
  • compositions and/or formulations of the present disclosure may be, but are not limited to, short-term, rapid-offset, as well as controlled, for example, sustained release, delayed release and pulsatile release formulations.
  • sustained release is used in its conventional sense to refer to a drug formulation that provides for gradual release of a drug over an extended period of time, and that may, although not necessarily, result in substantially constant blood levels of a drug over an extended time period.
  • the period of time may be as long as a month or more and should be a release which is longer that the same amount of agent administered in bolus form.
  • the compounds may be formulated with a suitable polymer or hydrophobic material which provides sustained release properties to the compounds.
  • the compounds for use the method of the disclosure may be administered in the form of microparticles, for example, by injection or in the form of wafers or discs by implantation.
  • the compounds useful within the disclosure are administered to a subject, alone or in combination with another pharmaceutical agent, using a sustained release formulation.
  • delayed release is used herein in its conventional sense to refer to a drug formulation that provides for an initial release of the drug after some delay following drug administration and that may, although not necessarily, include a delay of from about 10 minutes up to about 12 hours.
  • pulsatile release is used herein in its conventional sense to refer to a drug formulation that provides release of the drug in such a way as to produce pulsed plasma profiles of the drug after drug administration.
  • immediate release is used in its conventional sense to refer to a drug formulation that provides for release of the drug immediately after drug administration.
  • short-term refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes and any or all whole or partial increments thereof after drug administration after drug administration.
  • rapid-offset refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes, and any and all whole or partial increments thereof after drug administration.
  • reaction conditions including but not limited to reaction times, reaction size/volume, and experimental reagents, such as solvents, catalysts, pressures, atmospheric conditions, e.g., nitrogen atmosphere, and reducing/oxidizing agents, are within the scope of the present application.
  • LCMS Method C Shimadzu UFLC system employing an ACE UltraCore Super PhenylHexyl, 2.5 ⁇ m, 50 x 2.1 mm column with an aqueous acetonitrile based solvent gradient of 5-100% CH 3 CN/H 2 O (0.05 % Formic acid) over 5.0 mins.
  • Flow rate 1.0 mL/min.
  • the mixture was combined with another batch at the 1 g scale, the combined mixture was filtered and the filtrate concentrated.
  • the residue was purified by normal phase SiO 2 chromatography (0-15 % ethyl acetate / petroleum ether) to afford the semi-purified product.
  • the semi-purified product was triturated with petroleum ether / ethyl acetate (10:1 v/v, 20 mL) at room temperature for 0.5 hours.
  • the mixture was filtered and the solid product dried under vacuum to afford of 2-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (7 g) as a white solid.
  • Example 2 N-(3-(2-(4-(((1-Acetylpiperidin-4-yl)amino)methyl)-3-methoxyphenyl)-3- chloropyridin-4-yl)-2-methylphenyl)-5-((methylamino)methyl)picolinamide (a) tert-Butyl ((6-((3-bromo-2-methylphenyl)carbamoyl)pyridin-3- yl)methyl)(methyl)carbamate To a mixture of N-(3-bromo-2-methylphenyl)-5-formylpicolinamide (Example 1, step (c)) (1 g, 3.13 mmol) and methylamine hydrochloride salt (317 mg, 4.70 mmol) in methanol/dichloromethane (1/1, 30 mL) was added sodium acetate (771 mg, 9.40 mmol) and the mixture stirred for 12 hours at room temperature.
  • Example 4 (S)-N-(3-(3-Chloro-2-(3-methoxy-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- methoxyethyl)amino)methyl)picolinamide (a) tert-Butyl ((6-((3-bromo-2-methylphenyl)carbamoyl)pyridin-3-yl)methyl)(2- methoxyethyl)carbamate To a mixture of N-(3-bromo-2-methylphenyl)-5-formylpicolinamide (Example 1, step (c)) (0.5 g, 1.57 mmol) and 2-methoxyethan-1-amine (0.29 g, 3.92 mmol) in THF/methanol mixture (3:2, 5 mL) was added sodium acetate (0.26 g, 3.13
  • Example 5 N-(3-(2-(4-((7-Oxa-2-azaspiro[3.5]nonan-2-yl)methyl)-3-methoxyphenyl)-3- chloropyridin-4-yl)-2-methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)picolinamide (a) tert-Butyl ((6-((3-(3-chloro-2-(4-formyl-3-methoxyphenyl)pyridin-4-yl)-2- methylphenyl)carbamoyl)pyridin-3-yl)methyl)(2-hydroxyethyl)carbamate To a mixture of tert-butyl ((6-((3-(2,3-dichloropyridin-4-yl)-2- methylphenyl)carbamoyl)pyridin-3-yl)methyl)(2-hydroxyethyl)carbamate (Example 1, step (e))
  • reaction mixture combined with another batch at 50 mg scale.
  • Aqueous brine solution (15 mL) was added and the mixture extracted with ethyl acetate (100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated.
  • Example 6 N-(3-(3-Chloro-2-(3-methoxy-4-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((R)-3- hydroxypyrrolidin-1-yl)methyl)picolinamide
  • (a) Methyl 5-(dimethoxymethyl)picolinate To a solution of methyl 5-formylpicolinate (10 g, 60.6 mmol) and methanol (24.50 mL, 606 mmol, 10 eq) in toluene (100 mL) was added p-toluenesulfonic acid (1.04 g, 6.06 mmol) and the mixture stirred at 130 °C for 1 h under N 2 using a Dean-Stark apparatus.
  • the mixture was combined with another batch at 150 mg scale.
  • the mixture was concentrated, water (150 mL) and saturated aqueous brine solution (25 mL) added to the residue, and the mixture extracted with an ethyl acetate/THF mixture (1:1 v/v, 150 mL).
  • the organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • the mixture was combined with another batch at 500 mg scale.
  • the mixture was concentrated, water (150 mL) and saturated aqueous brine solution (25 mL) added to the residue, and the mixture extracted with an ethyl acetate/THF mixture (1:1 v/v, 100 mL).
  • the organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • Example 7 (R)-N-(3-(2-(4-(((1-Acetylpiperidin-4-yl)amino)methyl)-3-methoxyphenyl)- 3-chloropyridin-4-yl)-2-methylphenyl)-5-((3-hydroxypyrrolidin-1- yl)methyl)picolinamide (a) (R)-N-(3-(2-(4-(((1-Acetylpiperidin-4-yl)amino)methyl)-3-methoxyphenyl)-3- chloropyridin-4-yl)-2-methylphenyl)-5-((3-hydroxypyrrolidin-1-yl)methyl)picolinamide To a mixture of (R)-N-(3-(3-chloro-2-(4-formyl-3-methoxyphenyl)pyridin-4-yl)-2- methylphenyl)-5-((3-hydroxypyrrolidin-1-yl)methyl)picolinamide (
  • Example 8 N-(3-(3-Chloro-2-(4-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-3- methoxyphenyl)pyridin-4-yl)-2-methylphenyl)-5-(((R)-3-hydroxypyrrolidin-1- yl)methyl)picolinamide N-(3-(3-Chloro-2-(4-(((R)-3-hydroxypyrrolidin-1-yl)methyl)-3- methoxyphenyl)pyridin-4-yl)-2-methylphenyl)-5-(((R)-3-hydroxypyrrolidin-1- yl)methyl)picolinamide was prepared in a similar fashion to Example 7, replacing 1-(4- aminopiperidin-1-yl)ethan-1-one hydrochloride salt with (R)-pyrrolidin-3-ol in step (a).
  • Example 9 (S)-N-(3-(3-Chloro-2-(3-methoxy-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)(methyl)amino)methyl)picolinamide (a) N-(3-Bromo-2-methylphenyl)-5-(((tert- butyldimethylsilyl)oxy)methyl)picolinamide To a mixture of N-(3-bromo-2-methylphenyl)-5-(hydroxymethyl)picolinamide (3 g, 9.34 mmol) (Example 1, step (b)) in DMF (30 mL) was added imidazole (0.76 g, 11.2 mmol) and tert-butyldimethylsilyl chloride (2.82 g, 18.7 mmol) in one portion under N 2 and the mixture stir
  • the mixture was combined with another batch at the same scale.
  • the mixture was concentrated, water (20 mL) and saturated aqueous brine solution (20 mL) added to the residue, and the mixture extracted with an ethyl acetate/THF mixture (1:1 v/v, 80 mL).
  • the organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • the mixture was combined with another batch at 100 mg scale.
  • the mixture was concentrated, water (20 mL) and saturated aqueous brine solution (20 mL) added to the residue, and the mixture extracted with an ethyl acetate/THF mixture (1:1 v/v, 100 mL).
  • the organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • the mixture was combined with another batch at 20 mg scale.
  • the mixture was concentrated, water (20 mL) and saturated aqueous brine solution (20 mL) added to the residue, and the mixture was extracted with an ethyl acetate/THF mixture (3:1 v/v, 50 mL).
  • Example 10 (R)-N-(3-(3-Chloro-2-(3-methoxy-4-((7-oxo-2,6-diazaspiro[3.4]octan-2- yl)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-((3-hydroxypyrrolidin-1- yl)methyl)picolinamide (R)-N-(3-(3-chloro-2-(3-methoxy-4-((7-oxo-2,6-diazaspiro[3.4]octan-2- yl)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-((3-hydroxypyrrolidin-1- yl)methyl)picolinamide was prepared in a similar fashion to Example 7, replacing 1-(4- aminopiperidin-1-yl)ethan-1-one hydrochloride salt with 2,6-diazaspiro[3.4]octan-7-one triflu
  • Example 11 N-(3-(2-(4-((((1-Acetylpiperidin-4-yl)amino)methyl)-3-methoxyphenyl)-3- chloropyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)(methyl)amino)methyl)picolinamide (a) N-(3-(2,3-Dichloropyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)(methyl)amino)methyl)picolinamide To a mixture of N-(3-(2,3-dichloropyridin-4-yl)-2-methylphenyl)-5- formylpicolinamide (0.6 g, 1.55 mmol) (Example 6, step (e)) and 2-(methylamino)ethan-1-ol (0.18 g, 2.33 mmol) in THF/methanol mixture (1:1 v/v, 6 mL) was
  • the mixture was combined with another batch at 100 mg scale.
  • the mixture was concentrated, water (50 mL) and saturated aqueous brine solution (15 mL) added to the residue, and the mixture extracted with an ethyl acetate/THF mixture (1:1 v/v, 50 mL).
  • the organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • Example 12 N-(3-(2-(4-(((2-Acetyl-2-azaspiro[3.3]heptan-6-yl)amino)methyl)-3- methoxyphenyl)-3-chloropyridin-4-yl)-2-chlorophenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide (a) tert-Butyl 6-(((benzyloxy)carbonyl)amino)-2-azaspiro[3.3]heptane-2-carboxylate To a mixture of tert-butyl 6-amino-2-azaspiro[3.3]heptane-2-carboxylate (1 g, 4.71 mmol) and benzyl chloroformate (1.21 g, 7.07 mmol) in dichloromethane (15 mL) was added N,N-diisopropylethylamine (1.8 g, 14.1 mmol) and the mixture stirred at room
  • the mixture was stirred at 110 °C for 3 hours then the mixture was combined with additional batches at 30 mg and 130 mg scale.
  • the combined reaction mixtures were concentrated, water (20 mL) and ethyl acetate (20 mL) added to the residue and the mixture extracted with ethyl acetate (2 x 50 mL).
  • the combined organic phases were washed with saturated aqueous brine solution (2 x 50 mL), dried with anhydrous sodium sulfate, filtered and concentrated.
  • Example 13 (S)-5-(((1-Acetylpiperidin-4-yl)amino)methyl)-N-(2-chloro-3-(3-chloro-2- (3-methoxy-4-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)phenyl)pyridin-4- yl)phenyl)picolinamide (a) tert-Butyl (S)-(4-bromo-2-methoxybenzyl)((5-oxopyrrolidin-2- yl)methyl)carbamate To a mixture of 4-bromo-2-methoxybenzaldehyde (3 g, 14 mmol) and (S)-5- (aminomethyl)pyrrolidin-2-one hydrochloride salt (2.31 g, 15.4 mmol) in dichloromethane / methanol mixture (2:1 v/v, 60 mL) was added sodium acetate (3.43 g,
  • Example 14 (S)-N-(2-Chloro-3-(3-chloro-2-(3-methoxy-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)phenyl)-5-(((3- fluoropropyl)amino)methyl)picolinamide (a) tert-Butyl (S)-(4-(3-chloro-4-(2-chloro-3-(5-formylpicolinamido)phenyl)pyridin- 2-yl)-2-methoxybenzyl)((5-oxopyrrolidin-2-yl)methyl)carbamate To a mixture of tert-butyl (S)-(4-(3-chloro-4-(2-chloro-3-(5- (hydroxymethyl)picolinamido)phenyl)pyridin-2-yl)-2-methoxybenzyl)((5
  • Example 15 (S)-5-(((2-Acetyl-2-azaspiro[3.3]heptan-6-yl)amino)methyl)-N-(2-chloro-3- (3-chloro-2-(3-methoxy-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)phenyl)picolinamide (S)-5-(((2-Acetyl-2-azaspiro[3.3]heptan-6-yl)amino)methyl)-N-(2-chloro-3-(3-chloro- 2-(3-methoxy-4-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)phenyl)pyridin-4-yl)phenyl) was prepared in a similar fashion to Example 14, replacing 3-fluoropropan-1-amine hydrochloride salt with 1-(6-amino-2-azaspiro[3.3
  • Example 16 (S)-N-(3-(3-Chloro-2-(3-methyl-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide (a) tert-Butyl ((6-((3-(3-chloro-2-(4-formyl-3-methylphenyl)pyridin-4-yl)-2- methylphenyl)carbamoyl)pyridin-3-yl)methyl)(2-hydroxyethyl)carbamate To a mixture of tert-butyl ((6-((3-(2,3-dichloropyridin-4-yl)-2- methylphenyl)carbamoyl)pyridin-3-yl)methyl)(2-hydroxyethyl)carbamate (Example 1, step
  • Example 17 (S)-N-(3-(3-Chloro-2-(3-ethyl-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide (S)-N-(3-(3-chloro-2-(3-ethyl-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl) was prepared in a similar fashion to Example 16, replacing 2- methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzaldehyde with 2-ethyl-4-(4,4,5,5- tetramethyl
  • Example 18 (S)-N-(3-(3-Chloro-2-(3-fluoro-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide (S)-N-(3-(3-chloro-2-(3-fluoro-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl) was prepared in a similar fashion to Example 16, replacing 2- methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzaldehyde with 2-fluoro-4- (4,4,5,5-tetramethyl-1,3,2-
  • Example 19 (S)-N-(3-(3-Chloro-2-(3-chloro-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide (S)-N-(3-(3-Chloro-2-(3-chloro-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide was prepared in a similar fashion to Example 16, replacing 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzaldehyde with 2- chloro-4-(4,4,5,5-tetramethyl-1,3,
  • Example 21 (S)-N-(3-(3'-Chloro-6-(difluoromethoxy)-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide
  • (a) Methyl 6-chloro-2-hydroxynicotinate To a mixture 6-chloro-2-hydroxynicotinic acid (3.7 g, 21.3 mmol) in MeOH (37 mL) was added concentrated sulfuric acid (2.77 mL), and the mixture stirred at 60 °C for 4 hours under N 2 .
  • Example 22 (S)-N-(3-(3-Chloro-2-(3-methoxy-4-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)phenyl)pyridin-4-yl)-2-methylphenyl)-2-fluoro-4-(((2- hydroxyethyl)amino)methyl)benzamide (a) Methyl 4-((3-bromo-2-methylphenyl)carbamoyl)-3-fluorobenzoate To a mixture of 2-fluoro-4-(methoxycarbonyl)benzoic acid (1.00 g, 5.05 mmol) in DMF (30 mL) was added 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5- b]pyridinium 3-oxide hexafluorophosphate (HATU) (2.88 g, 7.57 mmol) and N,N- diisopropyleth
  • Example 23 (S)-N-(2-chloro-3-(3'-chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide (a) 2',3'-Dichloro-6-methoxy-[2,4'-bipyridine]-5-carbaldehyde A mixture of 2,3-dichloro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (1.00 g, 3.65 mmol), 6-chloro-2-methoxy-pyridine-3-carbaldehyde (0.69 g, 4.02 mmol), potassium carbonate (1.51 g, 10.95 mmol) and tetrakis(triphenylphosphine)palla
  • Example 24 N-(2-Chloro-3-(3'-chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((S)-3-hydroxypyrrolidin-1- yl)methyl)picolinamide (a) tert-Butyl ((3'-chloro-2'-(2-chloro-3-(5-(((S)-3-hydroxypyrrolidin-1- yl)methyl)picolinamido)phenyl)-6-methoxy-[2,4'-bipyridin]-5-yl)methyl)(((S)-5- oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(2-chloro-3-(5-formylpicolinamid
  • Example 25 (S)-N-(2-Chloro-3-(3'-chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((2- methoxyethyl)amino)methyl)picolinamide (a) tert-Butyl (S)-((3'-chloro-2'-(2-chloro-3-(5-(((2- methoxyethyl)amino)methyl)picolinamido)phenyl)-6-methoxy-[2,4'-bipyridin]-5- yl)methyl)((5-oxopyrrolidin-2-yl)methyl)carbamate To a mixture of tert-butyl (S)-((3'-chloro-2'-(2-chloro-3-(5- formylpicolinamid
  • the mixture was stirred at room temperature for 11.5 hours then sodium cyanoborohydride (0.08 g, 1.28 mmol) added and the mixture was stirred for an additional 0.5 hours.
  • the mixture was combined with another batch at 20 mg scale.
  • the combined mixture was concentrated, water (15 mL) and saturated aqueous brine solution (50 mL) were added to the residue and the mixture extracted with an ethyl acetate/THF mixture (1:1 v/v, 50 mL).
  • the organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • Example 26 (S)-N-(2-Chloro-3-(3'-chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((3- fluoropropyl)amino)methyl)picolinamide (S)-N-(2-Chloro-3-(3'-chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((3- fluoropropyl)amino)methyl)picolinamide was prepared in a similar fashion to Example 25, replacing 2-methoxyethan-1-amine with 3-fluoropropan-1-amine hydrochloride salt in step (a).
  • Example 27 N-(3-(3'-Chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((S)-3- hydroxypyrrolidin-1-yl)methyl)picolinamide (a) 5-(((tert-Butyldimethylsilyl)oxy)methyl)-N-(2-methyl-3-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)phenyl)picolinamide To a mixture of N-(3-bromo-2-methylphenyl)-5-(((tert- butyldimethylsilyl)oxy)methyl)picolinamide (4.6 g, 10.5mmol) (Example 9, step (a)) and bis(pinacolato)diboron (16.1 g, 63.4 mmol)
  • the mixture was combined with another two batches at 0.65 g and 0.13 g scale. To the mixture was added water (20 mL) and the mixture extracted with ethyl acetate (20 mL). The organic phase was washed with saturated aqueous brine solution (20 mL), dried with anhydrous sodium sulfate, filtered and concentrated.
  • Example 28 N-(3-(3'-Chloro-6-methoxy-5-((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((R)-3- hydroxypyrrolidin-1-yl)methyl)picolinamide (a) tert-Butyl ((3'-chloro-2'-(3-(5-(((R)-3-hydroxypyrrolidin-1- yl)methyl)picolinamido)-2-methylphenyl)-6-methoxy-[2,4'-bipyridin]-5-yl)methyl)(((S)- 5-oxopyrrolidin-2-yl)methyl)carbamate To a mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-formylpicolinamido)-2--(
  • Example 29 N-(3-(3'-chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((((S)-2- hydroxypropyl)amino)methyl)picolinamide
  • thionyl chloride 5 mL
  • Example 30 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((3- fluoropropyl)amino)methyl)picolinamide (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)- [2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((3-fluoropropyl)amino)methyl)picolinamide was prepared in a similar fashion to Example 28, replacing (R)-pyrrolidin-3-ol with 3- fluoropropan-1-amine hydrochloride salt in step (a).
  • Example 31 N-(3-(3'-chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((((R)-2- hydroxypropyl)amino)methyl)picolinamide (a) tert-Butyl ((3'-chloro-2'-(3-(5-((((R)-2- hydroxypropyl)amino)methyl)picolinamido)-2-methylphenyl)-6-methoxy-[2,4'- bipyridin]-5-yl)methyl)(((S)-5-oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-formylpicolinamido)-2-methylphenyl
  • Example 32 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-hydroxy-2- methylpropyl)amino)methyl)picolinamide (a) tert-Butyl (S)-((3'-chloro-2'-(3-(5-(((2-hydroxy-2- methylpropyl)amino)methyl)picolinamido)-2-methylphenyl)-6-methoxy-[2,4'- bipyridin]-5-yl)methyl)((5-oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-formylpicolinamido)-2-methylphenyl
  • Example 33 (S)-N-(3-(3'-chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)(methyl)amino)methyl)picolinamide (a) tert-Butyl (S)-((3'-chloro-2'-(3-(5-(((2- hydroxyethyl)(methyl)amino)methyl)picolinamido)-2-methylphenyl)-6-methoxy-[2,4'- bipyridin]-5-yl)methyl)((5-oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-formylpicolinamido)-2-methylphen
  • Example 34 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2- methoxyethyl)amino)methyl)picolinamide (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)- [2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-methoxyethyl)amino)methyl)picolinamide (yellow solid) was prepared in a similar fashion to Example 28, replacing (R)-pyrrolidin-3-ol with 2-methoxyethan-1-amine in step (a).
  • Example 35 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((3-hydroxyazetidin- 1-yl)methyl)picolinamide (a) tert-Butyl (S)-((3'-chloro-2'-(3-(5-((3-hydroxyazetidin-1-yl)methyl)picolinamido)- 2-methylphenyl)-6-methoxy-[2,4'-bipyridin]-5-yl)methyl)((5-oxopyrrolidin-2- yl)methyl)carbamate To a mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-formylpicolinamido)-2- methylpheny
  • Example 36 N-(3-(3'-Chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((oxetan-2- ylmethyl)amino)methyl)picolinamide (a) tert-Butyl ((3'-chloro-6-methoxy-2'-(2-methyl-3-(5-(((oxetan-2- ylmethyl)amino)methyl)picolinamido)phenyl)-[2,4'-bipyridin]-5-yl)methyl)(((S)-5- oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-formylpicolinamido)-2-methyl
  • Example 37 N-(3-(3'-Chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((((1r,3r)-3-hydroxy- 3-methylcyclobutyl)amino)methyl)picolinamide (a) tert-Butyl ((3'-chloro-2'-(3-(5-((((1r,3r)-3-hydroxy-3- methylcyclobutyl)amino)methyl)picolinamido)-2-methylphenyl)-6-methoxy-[2,4'- bipyridin]-5-yl)methyl)(((S)-5-oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(3-(
  • Example 38 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((3- (hydroxymethyl)azetidin-1-yl)methyl)picolinamide (a) tert-Butyl (S)-((3'-chloro-2'-(3-(5-((3-(hydroxymethyl)azetidin-1- yl)methyl)picolinamido)-2-methylphenyl)-6-methoxy-[2,4'-bipyridin]-5-yl)methyl)((5- oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-formylpicolinamido)-2
  • Example 39 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((3- hydroxypropyl)amino)methyl)picolinamide (a) tert-Butyl (S)-((3'-chloro-2'-(3-(5-(((3- hydroxypropyl)amino)methyl)picolinamido)-2-methylphenyl)-6-methoxy-[2,4'- bipyridin]-5-yl)methyl)((5-oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-formylpicolinamido)-2-methylphenyl)- 6-me
  • Example 40 N-(3-(3'-Chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((((S)-1-hydroxy-3- methylbutan-2-yl)amino)methyl)picolinamide (a) tert-Butyl ((3'-chloro-2'-(3-(5-((((S)-1-hydroxy-3-methylbutan-2- yl)amino)methyl)picolinamido)-2-methylphenyl)-6-methoxy-[2,4'-bipyridin]-5- yl)methyl)(((S)-5-oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-form
  • Example 41 N-(3-(3'-Chloro-6-methoxy-5-((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((R)-3- methoxypyrrolidin-1-yl)methyl)picolinamide (a) tert-Butyl ((3'-chloro-6-methoxy-2'-(3-(5-(((R)-3-methoxypyrrolidin-1- yl)methyl)picolinamido)-2-methylphenyl)-[2,4'-bipyridin]-5-yl)methyl)(((S)-5- oxopyrrolidin-2-yl)methyl)carbamate A mixture of tert-butyl (S)-((3'-chloro-2'-(3-(5-formylpicolinamido)-2-
  • Example 42 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)-6-methoxypicolinamide
  • (a) Methyl 5-formyl-6-methoxypicolinate To a mixture of 6-chloro-2-methoxynicotinaldehyde (5 g, 29.1 mmol) and sodium acetate (4.78 g, 58.3 mmol) in methanol/toluene mixture (7:2, 90 mL) was added palladium(II)acetate (0.13 g, 0.58 mmol) and 1,1’-bis(diphenylphosphino)ferrocene (0.48 g, 0.87 mmol) in one portion
  • the mixture was concentrated, water (50 mL) and saturated aqueous brine solution (50 mL) added to the residue, and the mixture extracted with an ethyl acetate/THF mixture (1:1 v/v, 200 mL). The organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • Example 43 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-4-(((2- hydroxyethyl)amino)methyl)thiazole-2-carboxamide (a) tert-Butyl (S)-((2'-(3-(4-bromothiazole-2-carboxamido)-2-methylphenyl)-3'- chloro-6-methoxy-[2,4'-bipyridin]-5-yl)methyl)((5-oxopyrrolidin-2-yl)methyl)carbamate To a mixture of 4-bromothiazole-2-carboxylic acid (0.17 g, 0.82 mmol) in DMF (4 mL) was added 1-[bis(dimethylamino)methylene
  • Example 44 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-4-(((2- hydroxyethyl)amino)methyl)-1-methyl-1H-imidazole-2-carboxamide (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)- [2,4'-bipyridin]-2'-yl)-2-methylphenyl)-4-(((2-hydroxyethyl)amino)methyl)-1-methyl-1H- imidazole-2-carboxamide was prepared in a similar fashion to Example 43, replacing 4- bromothiazole-2-carboxylic acid with 4-bromo-1-methyl-1H
  • Example 45 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)pyrazine-2-carboxamide (a) Methyl 5-(((tert-butoxycarbonyl)(2-((tert- butyldimethylsilyl)oxy)ethyl)amino)methyl)pyrazine-2-carboxylate A mixture of methyl 5-formylpyrazine-2-carboxylate (0.2 g, 1.2 mmol) and 2-((tert- butyldimethylsilyl)oxy)ethan-1-amine (0.22 g, 1.26 mmol) in THF/methanol mixture (1:1 v/v, 4 mL) was stirred
  • Example 46 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)thiazole-2-carboxamide
  • (a) Ethyl 5-formylthiazole-2-carboxylate A mixture of ethyl 2-amino-2-thioxoacetate (4.41 g, 33.1 mmol) and 2- bromomalonaldehyde (5 g, 33.1 mmol) in 1,4-dioxane (200 mL) was degassed and purged with N 2 and then the mixture stirred at 60 °C for 12 hours.
  • Example 48 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)-3-methoxypicolinamide (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)- [2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)-3- methoxypicolinamide was prepared in a similar fashion to Example 43, replacing 4- bromothiazole-2-carboxylic acid with 5-bromo-3-methoxypicolinic acid in step (a) and potassium os
  • Example 50 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-4-(((2- hydroxyethyl)amino)methyl)picolinamide (a) tert-Butyl (S)-((3'-chloro-2'-(3-(4-(hydroxymethyl)picolinamido)-2- methylphenyl)-6-methoxy-[2,4'-bipyridin]-5-yl)methyl)((5-oxopyrrolidin-2- yl)methyl)carbamate To a mixture of tert-butyl (S)-((2'-(3-amino-2-methylphenyl)-3'-chloro-6-methoxy- [2,4'-bipyridin]-5-y
  • the mixture was neutralized with saturated aqueous sodium bicarbonate solution and the mixture purified by reverse phase HPLC to afford the product as a formic salt ( ⁇ 50 mg).
  • the product ( ⁇ 50 mg) was neutralized with saturated aqueous sodium bicarbonate solution, deionized water (15 mL) was added and the mixture extracted with dichloromethane (3 x 5 mL).
  • Example 51 (S)-N-(3-(3-Chloro-2-(2-(2-hydroxypropyl)-8-methoxy-1,2,3,4- tetrahydroisoquinolin-6-yl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide (a) (S)-1-(6-bromo-8-methoxy-3,4-dihydroisoquinolin-2(1H)-yl)propan-2-ol To a mixture of 6-bromo-8-methoxy-1,2,3,4-tetrahydroisoquinoline hydrochloride salt (0.25 g, 0.9 mmol) in ethanol (4 mL) was added N,N-diisopropylethylamine (0.94 mL, 5.38 mmol) and (S)-2-methyloxirane (0.38 mL, 5.38 mmol) in one portion under N 2 atmosphere and
  • Example 52 (S)-N-(3-(3-Chloro-2-(8-methoxy-2-((5-oxopyrrolidin-2-yl)methyl)-1,2,3,4- tetrahydroisoquinolin-6-yl)pyridin-4-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide (a) (S)-5-((6-Bromo-8-methoxy-3,4-dihydroisoquinolin-2(1H)-yl)methyl)pyrrolidin- 2-one To a mixture of 6-bromo-8-methoxy-1,2,3,4-tetrahydroisoquinoline hydrochloride salt (0.5 g, 2 mmol) and (S)-5-(bromomethyl)pyrrolidin-2-one (0.4 g, 1.44 mmol) in acetonitrile (10 mL) was added potassium carbonate (0.6 g, 4.31
  • the mixture was combined with another batch at 50 mg scale.
  • the mixture was concentrated, water (5 mL) and saturated aqueous brine solution (5 mL) added to the residue, and the mixture was extracted with an ethyl acetate/THF mixture (10:1 v/v, 10 mL).
  • the organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • Example 53 (S)-N-(3-(3-Chloro-4-(6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)pyrazin-2-yl)pyridin-2-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)picolinamide
  • (a) 5-Bromo-3-methoxypyrazine-2-carbaldehyde To a mixture of 5-bromo-2-iodo-3-methoxypyrazine (22.5 g, 71.5 mmol) in THF (350 mL) was dropwise added 2.5 M n-Butyllithium solution in n-hexane (29 mL, 72.5 mmol) at - 80 °C under N 2 and the mixture stirred at -80 °C for 1 hours.
  • the mixture was concentrated, water (5 mL) and saturated aqueous brine solution (5 mL) added, and the mixture extracted with an ethyl acetate/THF mixture (10:1 v/v, 50 mL). The organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • the mixture was combined with 3 batches at 20 mg scale.
  • the mixture was concentrated, water (5 mL) and saturated aqueous brine solution (5 mL) added, and the mixture extracted with an ethyl acetate/THF mixture (10:1 v/v, 30 mL).
  • the organic phase was dried with anhydrous sodium sulfate, filtered and concentrated.
  • the residue was purified by normal phase SiO 2 chromatography (0-15% ethyl acetate/petroleum ether) to afford 100 mg of the product.170 mg of the product (70 mg from another batch) was purified by reverse phase HPLC. The eluent was neutralized with saturated aqueous sodium carbonate solution.
  • Example 54 (S)-(5-Oxopyrrolidin-2-yl)methyl ((3'-chloro-2'-(2-chloro-3-(5-(((2- hydroxyethyl)amino)methyl)picolinamido)phenyl)-6-methoxy-[2,4'-bipyridin]-5- yl)methyl)carbamate (a) (S)-(5-Oxopyrrolidin-2-yl)methyl 1H-imidazole-1-carboxylate A flask was charged with (S)-5-(hydroxymethyl)pyrrolidin-2-one (0.15 g, 1.30 mmol, 1 eq) and 1,1’-carbonyl diimidazole (0.27 g, 1.69 mmol, 1.3 eq) and the system was purged with argon.
  • the reaction was diluted with ethyl acetate (50 mL), washed with saturated aqueous brine solution (30 mL), the aqueous layer extracted with ethyl acetate (3 x 30 mL), and the combined organic layers dried over sodium sulfate and concentrated under reduced pressure.
  • Sodium cyanoborohydride (0.01 g, 0.15 mmol, 2.5 eq) was added and the mixture was stirred at room temperature for 1 hour. The reaction was then quenched by addition of water (1 mL), the mixture was diluted with ethyl acetate (50 mL), washed with saturated aqueous brine solution (30 mL), and the aqueous layer extracted with ethyl acetate (3 x 30 mL). The combined organic phases were then dried over sodium sulfate and concentrated under reduced pressure.
  • Example 56 (S)-N-(3-(3-Chloro-4-(6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)pyrazin-2-yl)pyridin-2-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)pyrazine-2-carboxamide (a) tert-Butyl ((5-((3-bromo-2-methylphenyl)carbamoyl)pyrazin-2-yl)methyl)(2- ((tert-butyldimethylsilyl)oxy)ethyl)carbamate To a mixture of lithium 5-(((tert-butoxycarbonyl)(2-((tert- butyldimethylsilyl)oxy)ethyl)amino)methyl)pyrazine-2-carboxylate (Example 45, step (b)) (0.44 g, 1.
  • the mixture was combined with another batch at 5 mg scale.
  • the mixture was purified by reverse phase HPLC.
  • the fraction containing product was lyophilized and neutralized with saturated aqueous sodium bicarbonate solution. Water (3 mL) was added, and the mixture extracted with dichloromethane (2 x 3 mL).
  • Example 58 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)-4-methylpicolinamide (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)- [2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)-4- methylpicolinamide was prepared in a similar fashion to Example 57, replacing methyl 5- bromo-6-methylpicolinate with methyl 5-bromo-4-methylpicolinate in step (a).
  • Example 60 N-(2-Chloro-3-(3'-chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((S)-3-hydroxypyrrolidin-1- yl)methyl)thiazole-2-carboxamide (a) tert-Butyl ((3'-chloro-2'-(2-chloro-3-(5-(((S)-3-hydroxypyrrolidin-1- yl)methyl)thiazole-2-carboxamido)phenyl)-6-methoxy-[2,4'-bipyridin]-5-yl)methyl)(((S)- 5-oxopyrrolidin-2-yl)methyl)carbamate To the mixture of tert-butyl (S)-((3'-chloro-2'-(2-chloro-3-
  • Example 61 (S)-N-(2-Chloro-3-(3'-chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((2- hydroxyethyl)amino)methyl)thiazole-2-carboxamide (S)-N-(2-Chloro-3-(3'-chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((2- hydroxyethyl)amino)methyl)thiazole-2-carboxamide was prepared in a similar fashion to Example 60, replacing (S)-pyrrolidin-3-ol with 2-aminoethanol in step (a).
  • Example 62 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-3-hydroxy-5-(((2- hydroxyethyl)amino)methyl)picolinamide
  • (a) Methyl 5-bromo-3-(methoxymethoxy)picolinate To a mixture of methyl 5-bromo-3-hydroxypicolinate (0.65 g, 2.8 mmol) and N,N- diisopropylethylamine (1.1 mL, 6.16 mmol) in dichloromethane (10 mL) was dropwise added chloro(methoxy)methane (0.65 mL, 8.57 mmol) at 0 °C under N 2 and the mixture stirred at room temperature for 3 hours.
  • Example 64 (S)-5-(((1-Acetylpiperidin-4-yl)amino)methyl)-N-(2-chloro-3-(3'-chloro-6- methoxy-5-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'- yl)phenyl)thiazole-2-carboxamide (S)-5-(((1-Acetylpiperidin-4-yl)amino)methyl)-N-(2-chloro-3-(3'-chloro-6-methoxy- 5-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)thiazole-2- carboxamide was prepared in a similar fashion to Example 60, replacing (S)-pyrrolidin-3-ol with 1-(4-aminopiperidin-1
  • Example 65 N-(2-Chloro-3-(3'-chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((R)-3-hydroxypyrrolidin-1- yl)methyl)thiazole-2-carboxamide N-(2-Chloro-3-(3'-chloro-6-methoxy-5-(((((S)-5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)-5-(((R)-3-hydroxypyrrolidin-1- yl)methyl)thiazole-2-carboxamide was prepared in a similar fashion to Example 60, replacing (S)-pyrrolidin-3-ol with (R)-pyrrolidin-3-ol in step (a).
  • Example 66 (S)-1-((2-((2-Chloro-3-(3'-chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)carbamoyl)thiazol-5- yl)methyl)piperidine-4-carboxylic acid (S)-1-((2-((2-Chloro-3-(3'-chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)phenyl)carbamoyl)thiazol-5- yl)methyl)piperidine-4-carboxylic acid (S)-1-((2-((2-Chloro-3-(3'-chloro-6-methoxy-5-((((5-o
  • the mixture was combined with another batch at 300 mg scale.
  • the mixture was concentrated, water (10 mL) and saturated aqueous brine solution (10 mL) added, and the mixture extracted with ethyl acetate (2 x 10 mL).
  • the combined organic phases were dried with anhydrous sodium sulfate, filtered and concentrated.
  • the residue was purified by normal phase SiO 2 chromatography (0-15% ethyl acetate/petroleum ether) to afford 2'-chloro-6-methoxy-3'-methyl-[2,4'-bipyridine]-5- carbaldehyde (0.5 g, 57% yield) as a white solid.
  • Example 68 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-7-(2-hydroxyethyl)- 5,6,7,8-tetrahydro-2,7-naphthyridine-3-carboxamide (a) 2-(2-((tert-Butyldimethylsilyl)oxy)ethyl)-6-chloro-1,2,3,4-tetrahydro-2,7- naphthyridine To a mixture of 6-chloro-1,2,3,4-tetrahydro-2,7-naphthyridine hydrochloride salt (0.3 g, 1.46 mmol) and (2-bromoethoxy)(tert-butyl)dimethylsilane (1.05 g, 4.39 mmol
  • Example 69 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2-yl)methyl)amino) methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)-1- methyl-1H-imidazole-2-carboxamide
  • (a) Ethyl 5-bromo-1-methyl-1H-imidazole-2-carboxylate To the mixture of ethyl chloroformate (13 g, 124 mmol) and N,N- diisopropylethylamine (16 g, 124 mmol) in acetonitrile (250 mL) was slowly added a solution of 5-bromo-1-methyl-1H-imidazole (10 g, 62.1 mmol) in acetonitrile (100 mL) at 0 °C and
  • Example 70 N-(3-(5-(((1-acetylpiperidin-4-yl)amino)methyl)-3'-chloro-6-methoxy-[2,4'- bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)thiazole-2- carboxamide
  • (a) Ethyl 5-(dimethoxymethyl)thiazole-2-carboxylate To a mixture of ethyl 5-formylthiazole-2-carboxylate (3 g, 16.2 mmol) in methanol (20 mL) was added p-toluenesulfonic acid (279 mg, 1.62 mmol) and trimethoxymethane (1.95 mL, 17.8 mmol), and the mixture stirred at 60 °C for 1 hour under N 2 .
  • Example 71 1-((3'-Chloro-2'-(3-(5-(((2-hydroxyethyl)amino)methyl)thiazole-2- carboxamido)-2-methylphenyl)-6-methoxy-[2,4'-bipyridin]-5-yl)methyl)piperidine-4- carboxylic acid
  • 1-((3'-Chloro-2'-(3-(5-(((2-hydroxyethyl)amino)methyl)thiazole-2-carboxamido)-2- methylphenyl)-6-methoxy-[2,4'-bipyridin]-5-yl)methyl)piperidine-4-carboxylic acid was prepared in a similar fashion to Example 70, replacing 1-(4-aminopiperidin-1-yl)ethan-1-one with tert-butyl piperidine-4-carboxylate in step (g).
  • Example 72 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((3-methoxyazetidin- 1-yl)methyl)picolinamide (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)- [2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((3-methoxyazetidin-1-yl)methyl)picolinamide was prepared in a similar fashion to Example 28, replacing (R)-pyrrolidin-3-ol with 3- methoxyazetidine and sodium acetate with acetic acid in step (a).
  • Example 73 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((3- (methoxymethyl)azetidin-1-yl)methyl)picolinamide (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2-yl)methyl)amino)methyl)- [2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-((3-(methoxymethyl)azetidin-1- yl)methyl)picolinamide was prepared in a similar fashion to Example 28, replacing (R)- pyrrolidin-3-ol with 3-(methoxymethyl)azetidine and sodium acetate with acetic acid in
  • Example 74 (R)-N-(3-(3'-Chloro-5-((3-hydroxypyrrolidin-1-yl)methyl)-6-methoxy-[2,4'- bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)thiazole-2- carboxamide
  • R)-N-(3-(3'-Chloro-5-((3-hydroxypyrrolidin-1-yl)methyl)-6-methoxy-[2,4'- bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)thiazole-2-carboxamide was prepared in a similar fashion to Example 70, replacing 1-(4-aminopiperidin-1-yl)ethan- 1-one with (R)-pyrrolidin-3-ol in step (g).
  • Example 75 (S)-N-(3-(3'-Chloro-5-((3-hydroxypyrrolidin-1-yl)methyl)-6-methoxy-[2,4'- bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)thiazole-2- carboxamide
  • S)-N-(3-(3'-Chloro-5-((3-hydroxypyrrolidin-1-yl)methyl)-6-methoxy-[2,4'- bipyridin]-2'-yl)-2-methylphenyl)-5-(((2-hydroxyethyl)amino)methyl)thiazole-2-carboxamide was prepared in a similar fashion to Example 70, replacing 1-(4-aminopiperidin-1-yl)ethan- 1-one with (S)-pyrrolidin-3-ol in step (g).
  • Example 76 (S)-N-(3-(3'-Chloro-6-methoxy-5-((((5-oxopyrrolidin-2- yl)methyl)amino)methyl)-[2,4'-bipyridin]-2'-yl)-2-methylphenyl)-5-(((2- hydroxyethyl)amino)methyl)-4-methoxypicolinamide
  • (a) Lithium 5-bromo-4-methoxypicolinate To a solution of methyl 5-bromo-4-methoxypicolinate (0.9 g, 3.66 mmol) in THF / water mixture (18:1 v/v, 9.5 mL) was added lithium hydroxide monohydrate (0.31 g, 7.32 mmol) in one portion under N 2 , and the mixture was stirred at room temperature for 3 hours.

Landscapes

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

Abstract

La présente invention concerne des composés dr 1-arylaminocarbonyl -1'-hétéroaryle substitués, des composés de 1-hétéroarylaminocarbonyl-1'-hétéroaryle substitués, des analogues de ceux-ci, et des compositions les comprenant. Selon un aspect, les composés envisagés dans l'invention peuvent être utilisés pour traiter, améliorer et/ou prévenir des infections par le virus de l'hépatite B (VHB) et/ou le virus de l'hépatite D (VHD) chez un sujet. Selon un autre aspect, les composés envisagés dans l'invention peuvent être utilisés pour traiter, faire régresser et/ou prévenir le cancer chez un sujet.
PCT/IB2023/059571 2022-09-28 2023-09-27 Composés de 1-arylaminocarbonyl-1'-hétéroaryle substitués, composés de 1-hétéroarylaminocarbonyl-1'-hétéroaryle substitués et procédés les utilisant WO2024069448A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263410766P 2022-09-28 2022-09-28
US63/410,766 2022-09-28

Publications (1)

Publication Number Publication Date
WO2024069448A1 true WO2024069448A1 (fr) 2024-04-04

Family

ID=90476516

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2023/059571 WO2024069448A1 (fr) 2022-09-28 2023-09-27 Composés de 1-arylaminocarbonyl-1'-hétéroaryle substitués, composés de 1-hétéroarylaminocarbonyl-1'-hétéroaryle substitués et procédés les utilisant

Country Status (1)

Country Link
WO (1) WO2024069448A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3152690A1 (fr) * 2019-09-30 2021-04-08 Lingyun Wu Compose utilise comme inhibiteur de pd-1/pd-l1 a petites molecules et application associee
WO2021147940A1 (fr) * 2020-01-21 2021-07-29 上海华汇拓医药科技有限公司 Inhibiteur de pd-1/pd-l1, son procédé de préparation et son utilisation
WO2022208269A2 (fr) * 2021-03-29 2022-10-06 Arbutus Biopharma Corporation Composés 1-aryl-1'-hétéroaryle substitués, composés 1,1'-bihétéroaryle substitués et leurs méthodes d'utilisation
WO2023108108A1 (fr) * 2021-12-09 2023-06-15 Deciphera Pharmaceuticals, Llc Inhibiteurs de raf kinase et leurs méthodes d'utilisation

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3152690A1 (fr) * 2019-09-30 2021-04-08 Lingyun Wu Compose utilise comme inhibiteur de pd-1/pd-l1 a petites molecules et application associee
WO2021147940A1 (fr) * 2020-01-21 2021-07-29 上海华汇拓医药科技有限公司 Inhibiteur de pd-1/pd-l1, son procédé de préparation et son utilisation
WO2022208269A2 (fr) * 2021-03-29 2022-10-06 Arbutus Biopharma Corporation Composés 1-aryl-1'-hétéroaryle substitués, composés 1,1'-bihétéroaryle substitués et leurs méthodes d'utilisation
WO2023108108A1 (fr) * 2021-12-09 2023-06-15 Deciphera Pharmaceuticals, Llc Inhibiteurs de raf kinase et leurs méthodes d'utilisation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
J. LUKE KONIARCZYK; JACOB W. GREENWOOD; JUAN V. ALEGRE‐REQUENA; ROBERT S. PATON; ANDREW MCNALLY: "A Pyridine–Pyridine Cross‐Coupling Reaction via Dearomatized Radical Intermediates", ANGEWANDTE CHEMIE INTERNATIONAL EDITION, VERLAG CHEMIE, HOBOKEN, USA, vol. 58, no. 42, 9 September 2019 (2019-09-09), Hoboken, USA, pages 14882 - 14886, XP072098475, ISSN: 1433-7851, DOI: 10.1002/anie.201906267 *

Similar Documents

Publication Publication Date Title
US11013726B2 (en) Substituted pyridinone-containing trycyclic compounds, and methods using same
JP2018076287A (ja) キナーゼ阻害剤としてのアミノヘテロアリールベンズアミド
AU2019397481B2 (en) Substituted arylmethylureas and heteroarylmethylureas, analogues thereof, and methods using same
JP2023540548A (ja) 抗腫瘍活性を有する化合物及びその使用
US11566002B2 (en) Substituted tetrahydrocyclopenta[C]pyrroles, substituted dihydropyrrolizines, analogues thereof, and methods using same
KR20240042620A (ko) 시아노피리딘 및 시아노피리미딘 bcl6 분해제
WO2022208269A2 (fr) Composés 1-aryl-1'-hétéroaryle substitués, composés 1,1'-bihétéroaryle substitués et leurs méthodes d'utilisation
WO2019222238A2 (fr) Composés 2,2'-bipyrimidinyl substitués, analogues de ceux-ci, et procédés les utilisant
US20230108906A1 (en) Substituted bicyclic and tricyclic ureas and amides, analogues thereof, and methods using same
WO2021097075A1 (fr) Composés d'isoindolonyle 2,2'-bipyrimidinyle substitués, analogues de ceux-ci, et procédés les utilisant
WO2024069448A1 (fr) Composés de 1-arylaminocarbonyl-1'-hétéroaryle substitués, composés de 1-hétéroarylaminocarbonyl-1'-hétéroaryle substitués et procédés les utilisant
JP2023532027A (ja) 新規な化合物およびこれを含む耐性がんの予防または治療用薬学組成物
CA3178647A1 (fr) Amides tricycliques substitues, analogues de ceux-ci et procedes les mettant en oeuvre
WO2021025976A1 (fr) Composés de cyclopropyl-2,2'-bipyrimidinyl substitués, analogues de ceux-ci, et procédés les utilisant
WO2024134466A1 (fr) Composés arylamine substitués, composés hétéroarylamine substitués et leurs procédés d'utilisation
WO2024018403A1 (fr) Composés d'imidazoamide substitués et leurs procédés d'utilisation
TW202400578A (zh) 新穎的化合物及其用於抑制檢查點激酶2的用途
WO2024028169A1 (fr) Nouveaux composés thiophénoliques à substitution spécifique
TW202415643A (zh) 經取代之芳基甲基脲類及雜芳基甲基脲類、其類似物及其使用方法

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: 23871186

Country of ref document: EP

Kind code of ref document: A1