US20230140047A1 - 1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR - Google Patents

1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR Download PDF

Info

Publication number
US20230140047A1
US20230140047A1 US18/081,558 US202218081558A US2023140047A1 US 20230140047 A1 US20230140047 A1 US 20230140047A1 US 202218081558 A US202218081558 A US 202218081558A US 2023140047 A1 US2023140047 A1 US 2023140047A1
Authority
US
United States
Prior art keywords
compound
mmol
cancer
alkyl
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/081,558
Inventor
Yam B. POUDEL
Sanjeev Gangwar
Liqi He
Prasanna Sivaprakasam
Matthias Broekema
Matthew Cox
Christine M. Tarby
Qian Zhang
Naidu S. Chowdari
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
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 Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Priority to US18/081,558 priority Critical patent/US20230140047A1/en
Assigned to BRISTOL-MYERS SQUIBB COMPANY reassignment BRISTOL-MYERS SQUIBB COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SIVAPRAKASAM, Prasanna, CHOWDARI, NAIDU S., COX, MATTHEW, GANGWAR, SANJEEV, HE, LIQI, POUDEL, YAM B., ZHANG, QIAN, TARBY, CHRISTINE M., BROEKEMA, Matthias
Publication of US20230140047A1 publication Critical patent/US20230140047A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • TLR7 Toll-like receptor 7
  • a TLR7 agonist can be conjugated to a partner molecule, which can be, for example, a phospholipid, a poly(ethylene glycol) (“PEG”), an antibody, or another TLR (commonly TLR2).
  • a partner molecule can be, for example, a phospholipid, a poly(ethylene glycol) (“PEG”), an antibody, or another TLR (commonly TLR2).
  • PEG poly(ethylene glycol)
  • exemplary disclosures include: Carson et al. 2013, 2015, and 2016, Chan et al. 2009 and 2011, Cortez et al. 2017, Gadd et al. 2015, Lioux et al. 2016, Maj et al. 2015, Vernejoul et al. 2014, and Zurawski et al. 2012.
  • a frequent conjugation site is at the R′′ group of formula (A).
  • TLR7 agonists have activity as TLR7 agonists and some can be conjugated to an antibody for targeted delivery to a target tissue or organ of intended action. They can also be PEGylated, to modulate their pharmaceutical properties.
  • Compounds disclosed herein, or their conjugates or their PEGylated derivatives can be used in the treatment of a subject suffering from a condition amenable to treatment by activation of the immune system, by administering to such subject a therapeutically effective amount of such a compound or a conjugate thereof or a PEGylated derivative thereof, especially in combination with a vaccine or a cancer immunotherapy agent.
  • Antigen binding fragment and “antigen binding portion” of an antibody mean one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody, such as (i) a Fab fragment, a monovalent fragment consisting of the V L , V H , C L and CHI domains; (ii) a F(ab′) 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fab′ fragment, which is essentially an Fab with part of the hinge region (see, for example, Abbas et al., Cellular and Molecular Immunology, 6th Ed., Saunders Elsevier 2007); (iv) a Fd fragment consisting of the V H and C H1 domains; (v) a Fv fragment consisting of the V L
  • Preferred antigen binding fragments are Fab, F(ab′) 2 , Fab′, Fv, and Fd fragments.
  • V L and V H the two domains of the Fv fragment, V L and V H , are encoded by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the V L and V H regions pair to form monovalent molecules (known as single chain Fv, or scFv); see, e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • Such single chain antibodies are also encompassed within the term “antigen-binding portion” of an antibody.
  • “Monoclonal antibody” or “monoclonal antibody composition” means a preparation of antibody molecules of single molecular composition, which displays a single binding specificity and affinity for a particular epitope.
  • heteroaryl moieties include pyrrolyl, furanyl, thiophenyl (thienyl), imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, tetrazolyl, pyridyl, N-oxopyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, quinolinyl, isoquinolynyl, quinazolinyl, cinnolinyl, quinozalinyl, naphthyridinyl, benzofuranyl, indolyl, benzothiophenyl, oxadiazolyl, thiadiazolyl, phenothiazolyl, benzimidazolyl, benzotriazolyl, dibenzofuranyl, carbazolyl, dibenzothiophenyl,
  • substituents are halo, hydroxyl, cyano, nitro, alkoxy, —O(aryl), ⁇ O, ⁇ NOH, ⁇ NO(alkyl), —OC( ⁇ O)(alkyl), —OC( ⁇ O)O(alkyl), —OC( ⁇ O)NH 2 , —OC( ⁇ O)NH(alkyl), —OC( ⁇ O)N(alkyl) 2 , azido, —NH 2 , —NH(alkyl), —N(alkyl) 2 , —NH(aryl), —NHC( ⁇ O)(alkyl), —NHC( ⁇ O)H, —NHC( ⁇ O)NH 2 , —NHC( ⁇ O)NH(alkyl), —NHC( ⁇ O)N(alkyl) 2 , and —NHC( ⁇ NH)NH 2 .
  • substituents are alkyl, alkenyl, alkynyl, halo, haloalkyl, hydroxyl, hydroxyalkyl, cyano, nitro, alkoxy, —O(hydroxyalkyl), —O(haloalkyl), —O(aryl), —O(cycloalkyl), —O(heterocycloalkyl), alkylthio, arylthio, —C( ⁇ O)(alkyl), —C( ⁇ O)H, —CO 2 H, —C( ⁇ O)NHOH, —C( ⁇ O)O(alkyl), —C( ⁇ O)O(hydroxyalkyl), —C( ⁇ O)NH 2 , —C( ⁇ O)NH(alkyl), —C( ⁇ O)N(alkyl) 2 , —OC
  • treat in the context of treating a disease or disorder, are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or to slowing the progression, spread or worsening of a disease, disorder or condition or of one or more symptoms thereof.
  • the “treatment of cancer”, refers to one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, (i) slowing down and (ii) complete growth arrest; (2) reduction in the number of tumor cells; (3) maintaining tumor size; (4) reduction in tumor size; (5) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of tumor cell infiltration into peripheral organs; (6) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of metastasis; (7) enhancement of anti-tumor immune response, which may result in (i) maintaining tumor size, (ii) reducing tumor size, (iii) slowing the growth of a tumor, (iv) reducing, slowing or preventing invasion and/or (8) relief, to some extent, of the severity or number of one or more symptoms associated with the disorder.
  • Suitable groups R 1 include:
  • R 3 examples include Cl, OH,
  • TLR7 agonists disclosed herein can be delivered to the site of intended action by localized administration or by targeted delivery in a conjugate with a targeting moiety.
  • the targeting moiety is an antibody or antigen binding portion thereof and its antigen is found at the locality of intended action, for example a tumor associated antigen if the intended site of action is at a tumor (cancer).
  • the tumor associated antigen is uniquely expressed or overexpressed by the cancer cell, compared to a normal cell.
  • the tumor associated antigen can be located on the surface of the cancer cell or secreted by the cancer cell into its environs.
  • Z is a targeting moiety
  • D is an agonist of this invention
  • —(X D ) a C(X Z ) b — are collectively referred to as a “linker moiety” or “linker” because they link Z and D.
  • C is a cleavable group designed to be cleaved at or near the site of intended biological action of D
  • X D and X Z are spacer moieties (or “spacers”) that space apart D and C and C and Z, respectively
  • subscripts a, b, and c are independently 0 or 1 (that is, the presence of X D , X Z and C are optional).
  • Subscript m is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 (preferably 1, 2, 3, or 4).
  • D, X D , C, X Z and Z are more fully described hereinbelow.
  • targeting moiety Z is an antibody.
  • Z and its conjugates are written in the context of its being an antibody, but those skilled in the art will understand that other types of Z can be conjugated, mutatis mutandis.
  • conjugates with folic acid as the targeting moiety can target cells having the folate receptor on their surfaces (Leamon et al., Cancer Res. 2008, 68 (23), 9839).
  • Antibodies that can be used in conjugates of this invention include those recognizing the following antigens: mesothelin, prostate specific membrane antigen (PSMA), CD19, CD22, CD30, CD70, B7H3, B7H4 (also known as 08E), protein tyrosine kinase 7 (PTK7), glypican-3, RG1, fucosyl-GM1, CTLA-4, and CD44.
  • the antibody can be animal (e.g., murine), chimeric, humanized, or, preferably, human.
  • the antibody preferably is monoclonal, especially a monoclonal human antibody. The preparation of human monoclonal antibodies against some of the aforementioned antigens is disclosed in Korman et al., U.S. Pat.
  • No. 8,609,816 B2 (2013; B7H4, also known as 08E; in particular antibodies 2A7, 1G11, and 2F9); Rao-Naik et al., 8,097,703 B2 (2012; CD19; in particular antibodies 5G7, 13F1, 46E8, 21D4, 21D4a, 47G4, 27F3, and 3C10); King et al., U.S. Pat. No. 8,481,683 B2 (2013; CD22; in particular antibodies 12C5, 19A3, 16F7, and 23C6); Keler et al., U.S. Pat. No.
  • the linker comprises up to three elements: a cleavable group C and optional spacers X Z and X D .
  • C contains an amino acid sequence that is a cleavage recognition sequence for a protease.
  • cleavage recognition sequences are known in the art. See, e.g., Matayoshi et al. Science 247: 954 (1990); Dunn et al. Meth. Enzymol. 241: 254 (1994); Seidah et al. Meth. Enzymol. 244: 175 (1994); Thomberry, Meth. Enzymol. 244: 615 (1994); Weber et al. Meth. Enzymol. 244: 595 (1994); Smith et al. Meth. Enzymol. 244: 412 (1994); and Bouvier et al. Meth. Enzymol. 248: 614 (1995); the disclosures of which are incorporated herein by reference.
  • Group C can be bonded directly to Z or D; i.e. spacers X Z or X D , as the case may be, can be absent.
  • Spacers X Z and/or X D where present, preferably provide a linear separation of from 4 to 25 atoms, more preferably from 4 to 20 atoms, between Z and C or D and C, respectively.
  • Conjugates of TLR7 agonists disclosed herein preferably are made by first preparing a compound comprising D and linker (X D ) a (C) c (X Z ) n (where X D , C, X Z , a, b, and c are as defined for formula (II)) to form drug-linker compound represented by formula (V):
  • R 31 is a functional group suitable for reacting with a complementary functional group on Z to form the conjugate.
  • suitable groups R 31 include amino, azide, thiol, cyclooctyne,
  • R 32 is Cl, Br, F, mesylate, or tosylate and R 33 is Cl, Br, I, F, OH, —O—N-succinimidyl, —O-(4-nitrophenyl), —O-pentafluorophenyl, or —O-tetrafluorophenyl.
  • Scheme generally usable for the preparation of suitable moieties D-(X D ) a C(X Z ) b —R 31 is disclosed in Ng et al., U.S. Pat. No. 7,087,600 B2 (2006); Ng et al., U.S. Pat. No. 6,989,452 B2 (2006); Ng et al., U.S. Pat.
  • reactive functional group —R 31 is —NH 2 , —OH, —CO 2 H, —SH, maleimido, cyclooctyne, azido (—N 3 ), hydroxylamino (—ONH 2 ) or N-hydroxysuccinimido.
  • Especially preferred functional groups —R 31 are:
  • An —OH group can be esterified with a carboxy group on the antibody, for example, on an aspartic or glutamic acid side chain.
  • a —CO 2 H group can be esterified with a —OH group or amidated with an amino group (for example on a lysine side chain) on the antibody.
  • N-hydroxysuccinimide group is functionally an activated carboxyl group and can conveniently be amidated by reaction with an amino group (e.g., from lysine).
  • an ⁇ -amino group in the side chain of a lysine residue can be reacted with 2-iminothiolane or N-succinimidyl-3-(2-pyridyldithio)propionate (“SPDP”) to introduce a free thiol (—SH) group-creating a cysteine surrogate, as it were.
  • SPDP N-succinimidyl-3-(2-pyridyldithio)propionate
  • the thiol group can react with a maleimide or other nucleophile acceptor group to effect conjugation. The mechanism if illustrated below with 2-iminothiolane.
  • An alternative conjugation method employs copper-free “click chemistry,” in which an azide group adds across a strained cyclooctyne to form an 1,2,3-triazole ring. See, e.g., Agard et al., J. Amer. Chem. Soc. 2004, 126, 15046; Best, Biochemistry 2009, 48, 6571, the disclosures of which are incorporated herein by reference.
  • the azide can be located on the antibody and the cyclooctyne on the drug-linker moiety, or vice-versa.
  • a preferred cyclooctyne group is dibenzocyclooctyne (DIBO).
  • DIBO dibenzocyclooctyne
  • Various reagents having a DIBO group are available from Invitrogen/Molecular Probes, Eugene, Oreg. The reaction below illustrates click chemistry conjugation for the instance in which the DIBO group is attached to the antibody (Ab):
  • an antibody that is used for making a conjugate has one or more amino acids replaced by a non-natural amino acid, which preferably isp-acetylphenylalanine orp-azidophenylalanine, more preferably p-acetylphenylalanine.
  • the positioning of a glutamine residue on a polypeptide chain has a large effect on its susceptibility to BTG mediated transamidation. None of the glutamine residues on an antibody are normally BTG substrates. However, if the antibody is deglycosylated—the glycosylation site being asparagine 297 (N297; numbering per EU index as set forth in Kabat et al., “Sequences of proteins of immunological interest,” 5th ed., Pub. No. 91-3242, U.S. Dept. Health & Human Services, NIH, Bethesda, Md., 1991; hereinafter “Kabat”) of the heavy chain—nearby glutamine 295 (Q295) is rendered BTG susceptible.
  • N297 numbering per EU index as set forth in Kabat et al., “Sequences of proteins of immunological interest,” 5th ed., Pub. No. 91-3242, U.S. Dept. Health & Human Services, NIH, Beth
  • an antibody can be deglycosylated enzymatically by treatment with PNGase F (Peptide-N-Glycosidase F).
  • PNGase F Peptide-N-Glycosidase F
  • an antibody can be synthesized glycoside free by introducing an N297A mutation in the constant region, to eliminate the N297 glycosylation site. Further, it has been shown that an N297Q substitution not only eliminates glycosylation, but also introduces a second glutamine residue (at position 297) that too is an amine acceptor. Thus, in one embodiment, the antibody is deglycosylated. In another embodiment, the antibody has an N297Q substitution.
  • An antibody can also be rendered susceptible to BTG-mediated conjugation by introducing into it a glutamine containing peptide, or “tag,” as taught, for example, in Pons et al., US 2013/0230543 A1 (2013) and Rao-Naik et al., WO 2016/144608 A1.
  • the substrate specificity of BTG can be altered by varying its amino acid sequence, such that it becomes capable of reacting with glutamine 295 in an unmodified antibody, as taught in Rao-Naik et al., WO 2017/059158 A1 (2017).
  • transglutaminase from S. mobaraensis While the most commonly available bacterial transglutaminase is that from S. mobaraensis , transglutaminase from other bacteria, having somewhat different substrate specificities, can be considered, such as transglutaminase from Streptoverticillium ladakanum (Hu et al., US 2009/0318349 A1 (2009), US 2010/0099610 A1 (2010), and US 2010/0087371 A1 (2010)).
  • TLR7 agonists of this disclosure having a primary or secondary alkyl amine are particularly suitable for use in conjugates, as the secondary amine provides a functional group for attachment of the linker.
  • An example of such a TLR7 agonist-linker compound is compound 41, which contains an enzymatically cleavable linker.
  • FIG. 15 shows a scheme according to which compound 41 can be prepared.
  • TLR7 agonist conjugates such as the ones shown below can be prepared:
  • PEGylation Attachment of a poly(ethylene glycol) (PEG) chain to a drug (“PEGylation”) can improve the latter's pharmacokinetic properties.
  • the circulation half-life of the drug is increased, sometimes by over an order of magnitude, concomitantly reducing the dosage needed to achieve a desired therapeutic effect.
  • PEGylation can also decrease metabolic degradation of a drug and reduce its immunogenicity.
  • a TLR7 agonist disclosed herein can be PEGylated via an enzymatically cleavable linker comprising a self-immolating moiety, to allow release of the un-PEGylated agonist in a designed manner.
  • PEGylation can be combined with conjugation to a protein such as an antibody, if the PEG-containing molecule has a suitable functional group such as an amine for attachment to the protein.
  • the protein can provide an additional therapeutic function or, if an antibody, can provide a targeting function.
  • the valine-citrulline (Val-Cit) dipeptide is cleavable by the enzyme cathepsin B, with a p-aminobenzyl oxycarbonyl (PABC) group serving as a self-immolating spacer.
  • the functional group for conjugation is an amine group, which is temporarily protected by an Fmoc group.
  • Conjugation is effected by the enzyme transglutaminase, with a glutamine (Gln) side chain acting as the acyl acceptor.
  • the subscript x denoting the number of PEG repeat units, can vary widely, depending on the purpose of the PEGylation, as discussed below. For some purposes, x can be relatively small, such as 2, 4, 8, 12, or 24. For other purposes, x is large, for example between about 45 and about 910.
  • the compound lacks aliphatic hydroxyl or aliphatic primary or secondary amine, it still can be PEGylated at the aromatic amine on the pyrimidine ring.
  • a method for PEGylating at this position is disclosed by Zarraga, US 2017/0166384 A1 (2007), the disclosure of which is incorporated by reference.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated and the particular mode of administration and will generally be that amount of the composition which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.01 percent to about ninety-nine percent of active ingredient, preferably from about 0.1 percent to about 70 percent, most preferably from about 1 percent to about 30 percent of active ingredient in combination with a pharmaceutically acceptable carrier.
  • this specification provides a method of treating a cancer, comprising administering to a patient suffering from such cancer a therapeutically effective combination of an anti-cancer immunotherapy agent and a TLR7 agonist as disclosed herein.
  • the timing of administration can be simultaneous, sequential, or alternating.
  • the mode of administration can systemic or local.
  • the TLR7 agonist can be delivered in a targeted manner, via a conjugate.
  • the anti-cancer immunotherapy agent is an antagonistic anti-CTLA-4 antibody, preferably ipilimumab.
  • TLR7 agonists disclosed herein also are useful as vaccine adjuvants.
  • TLR7 agonists The biological activity of compounds disclosed herein as TLR7 agonists can be assayed by the procedures following.
  • the frozen samples were thawed and mRNA was extracted using the Invitrogen mRNA Catcher Plus kit (Cat #K1570-02) according to the manufacturer's instructions. Half yield of mRNA from RNA extraction were used to synthesize cDNA in 20 ⁇ L reverse transcriptase reactions using Invitrogen SuperScript IV VILO Master Mix (Cat #11756500).
  • TaqMan® real-time PCR was performed using QuantStudio Real-Time PCR system from ThermoFisher (Applied Biosystems). All real-time PCR reactions were run in duplicate using commercial predesigned TaqMan assays for mouse IFIT1, IFIT3, MX1 and PPIA gene expression and TaqMan Master Mix. PPIA was utilized as the housekeeping gene. The recommendations from the manufacturer were followed. All raw data (Ct) were normalized by average housekeeping gene (Ct) and then the comparative Ct ( ⁇ Ct) method were utilized to quantify relative gene expression (RQ) for experimental analysis.
  • NMR spectra were taken in either 400 Mz or 500 Mhz Bruker instrument using either DMSO-d6 or CDCl 3 as solvent and internal standard.
  • the crude NMR data was analyzed by using either ACD Spectrus version 2015-01 by ADC Labs or MestReNova software.
  • the procedures disclosed herein produce a mixture of regioisomers, alkylated at the 1H or 2H position of the pyrazolopyrimidine ring system (which are also referred to as N1 and N2 regioisomers, respectively, alluding to the nitrogen that is alkylated).
  • N1 and N2 regioisomers are also referred to as N1 and N2 regioisomers, respectively, alluding to the nitrogen that is alkylated.
  • the N2 regioisomers are not shown for convenience, but it is to be understood that they are present in the initial product mixture and separated at a later time, for example by preparative HPLC.
  • the mixture of regioisomers can be separated at an early stage of the synthesis and the remaining synthetic steps carried out with the 1H regioisomer or, alternatively, the synthesis can be progressed carrying the mixture of regioisomers and separation effected at a later stage, as desired.
  • FIG. 1 illustrate a method for making compounds disclosed herein, using compound 800 as an exemplar.
  • FIGS. 3 A- 3 B illustrate another method for making compounds disclosed herein, using compounds 844, 817, and 861 as exemplars.
  • the deprotected intermediate product from above was dissolved in dioxane (0.5 mL), treated with sodium hydroxide (0.220 mL, 0.220 mmol) and heated at 80° C. for 2 h.
  • LCMS showed completion of the reaction.
  • the reaction mixture was neutralized to pH 7 with aqueous HCl and the solvent was evaporated.
  • Compound 54 can be used to make compounds such as 846 and 847, generally following the Procedure 3 and FIGS. 3 A- 3 B , using compound 16.
  • FIGS. 6 A- 6 B illustrate another method of making compounds disclosed herein, using compound 857 as an exemplar.
  • This Procedure 6 and prior Procedure 4 can be used to make similar compounds.
  • alkylation of 3-bromo compound 55 yields a higher proportion of the 1H regioisomer (2H regioisomer not shown in FIG. 6 A ) than the alkylation of counterpart compound 36 in Procedure 4.
  • this Procedure 6 may be the preferable one, even though it entails the extra bromination and de-bromination steps.
  • Table B lists some compounds that were made by either Procedure 4 or 6.
  • 3-bromo compound 55 can be an intermediate in other synthetic pathways, as shown in Procedures hereinbelow.
  • FIG. 11 illustrate another method for making compounds disclosed herein, using compound 855 as an exemplar.
  • the crude material was purified via preparative HPLC with the following conditions: Column: XBridge C18, 200 mm ⁇ 19 mm, 5- ⁇ m particles; Mobile Phase A: 5:95 acetonitrile:water with 10-mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10-mM ammonium acetate; Gradient: a 0-minute hold at 8% B, 8-48% B over 20 minutes, then a 0-minute hold at 100% B; Flow rate: 20 mL/min; Column Temperature: 25 C. Fraction collection was triggered by MS and UV signals.
  • FIG. 12 illustrate another method for making compounds disclosed herein, using compound 867 as an exemplar.
  • This procedure relates to the preparation of compounds 829 and its regiosiomer. These compounds were prepared by reacting compounds 36 and 37 with MeMgCl Grignard reagent.
  • the mixture was purified via preparative HPLC/MS with the following conditions: Column: XBridge C18, 200 mm ⁇ 19 mm, 5- ⁇ m particles; Mobile Phase A: 5:95 acetonitrile:water with 0.1% TFA; Mobile Phase B: 95:5 acetonitrile:water with 0.1% TFA; Gradient: a 0-minute hold at 10% B, 10-45% B over 20 minutes, then a 4-minute hold at 100% B; Flow Rate: 20 mL/min; Column Temperature: 25° C. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation.
  • Analytical data for compounds of this disclosure are provided in following Table B, along with their human TLR7 (hTLR7) agonism data.
  • the reported activity agonism value is the average of plural assays.
  • the association of a particular compound with a particular Procedure is illustrative, and not exhaustive. A compound could be made by one of the other Procedures and, conversely, the same Procedure could be used to make other compounds of this disclosure.
  • the RT (retention time) column refers to the retention time per LCMS procedure B/C/D/E above, as applicable.

Abstract

Compounds according to formula I are useful as agonists of Toll-like receptor 7 (TLR7).Such compounds can be used in cancer treatment, especially in combination with an anti-cancer immunotherapy agent, or as a vaccine adjuvant.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation application of U.S. patent application Ser. No. 16/527,802, filed Jul. 31, 2019, which claims priority to U.S. Provisional Patent Application No. 62/714,238, filed Aug. 3, 2018, the entire contents of which are incorporated herein by reference.
  • BACKGROUND OF THE DISCLOSURE
  • This disclosure relates to Toll-like receptor 7 (“TLR7”) agonists and conjugates thereof, and methods for the preparation and use of such agonists and their conjugates.
  • Toll-like receptors (“TLRs”) are receptors that recognize pathogen-associated molecular patterns (“PAMPs”), which are small molecular motifs conserved in certain classes of pathogens. TLRs can be located either on a cell's surface or intracellularly. Activation of a TLR by the binding of its cognate PAMP signals the presence of the associated pathogen inside the host—i.e., an infection—and stimulates the host's immune system to fight the infection. Humans have 10 TLRs, named TLR1, TLR2, TLR3, and so on.
  • The activation of a TLR—with TLR7 being the most studied—by an agonist can have a positive effect on the action of vaccines and immunotherapy agents in treating a variety of conditions other than actual pathogen infection, by stimulating the immune response overall. Thus, there is considerable interest in the use of TLR7 agonists as vaccine adjuvants or as enhancers in cancer immunotherapy. See, for example, Vasilakos and Tomai 2013, Sato-Kaneko et al. 2017, Smits et al. 2008, and Ota et al. 2019.
  • TLR7, an intracellular receptor located on the membrane of endosomes, recognizes PAMPs associated with single-stranded RNA viruses. Its activation induces secretion of Type I interferons such as IFNα and IFNβ (Lund et al. 2004). TLR7 has two binding sites, one for single stranded RNA ligands (Berghofer et al. 2007) and one for small molecules such as guanosine (Zhang et al. 2016).
  • TLR7 can bind to, and be activated by, guanosine-like synthetic agonists such as imiquimod, resiquimod, and gardiquimod, which are based on a 1H-imidazo[4,5-c]quinoline scaffold. For a review of small-molecule TLR7 agonists, see Cortez and Va 2018.
  • Figure US20230140047A1-20230504-C00002
  • Synthetic TLR7 agonists based on a pteridinone molecular scaffold are also known, as exemplified by vesatolimod (Desai et al. 2015).
  • Figure US20230140047A1-20230504-C00003
  • Other synthetic TLR7 agonists based on a purine-like scaffold have been disclosed, frequently according to the general formula (A):
  • Figure US20230140047A1-20230504-C00004
  • where R, R′, and R″ are structural variables, with R″ typically containing an unsubstituted or substituted aromatic or heteroaromatic ring.
  • Disclosures of bioactive molecules having a purine-like scaffold and their uses in treating conditions such as fibrosis, inflammatory disorders, cancer, or pathogenic infections include: Akinbobuyi et al. 2015 and 2016; Barberis et al. 2012; Carson et al. 2014; Ding et al. 2016, 2017a, and 2017b; Graupe et al. 2015; Hashimoto et al. 2009; He et al. 2019a and 2019b; Holldack et al. 2012; Isobe et al. 2009a and 2012; Poudel et al. 2019a and 2019b; Pryde 2010; and Young et al. 2019.
  • The group R″ can be pyridyl: Bonfanti et al. 2015a and 2015b; Halcomb et al. 2015; Hirota et al. 2000; Isobe et al. 2002, 2004, 2006, 2009a, 2009b, 2011, and 2012; Kasibhatla et al. 2007; Koga-Yamakawa et al. 2013; Musmuca et al. 2009; Nakamura 2012; Ogita et al. 2007; and Yu et al. 2013.
  • There are disclosures of related molecules in which the 6,5-fused ring system of formula (A)—a pyrimidine six member ring fused to an imidazole five member ring—is modified. (a) Dellaria et al. 2007, Jones et al. 2010 and 2012, and Pilatte et al. 2017 disclose compounds in which the pyrimidine ring is replaced by a pyridine ring. (b) Chen et al. 2011, Coe et al. 2017, and Zhang et al. 2018 disclose compounds in which the imidazole ring is replaced by a pyrazole ring. (c) Cortez et al. 2017 and 2018; Li et al. 2018; and McGowan et al. 2016a, 2016b, and 2017 disclose compounds in which the imidazole ring is replaced by a pyrrole ring.
  • Bonfanti et al. 2015b and 2016 and Purandare et al. 2019 disclose TLR7 modulators in which the two rings of a purine moiety are spanned by a macrocycle:
  • A TLR7 agonist can be conjugated to a partner molecule, which can be, for example, a phospholipid, a poly(ethylene glycol) (“PEG”), an antibody, or another TLR (commonly TLR2). Exemplary disclosures include: Carson et al. 2013, 2015, and 2016, Chan et al. 2009 and 2011, Cortez et al. 2017, Gadd et al. 2015, Lioux et al. 2016, Maj et al. 2015, Vernejoul et al. 2014, and Zurawski et al. 2012. A frequent conjugation site is at the R″ group of formula (A).
  • Jensen et al. 2015 discloses the use of cationic lipid vehicles for the delivery of TLR7 agonists.
  • Some TLR7 agonists, including resiquimod are dual TLR7/TLR8 agonists. See, for example, Beesu et al. 2017, Embrechts et al. 2018, Lioux et al. 2016, and Vernejoul et al. 2014.
  • Full citations for the documents cited herein by first author or inventor and year are listed at the end of this specification.
  • BRIEF SUMMARY OF THE DISCLOSURE
  • This specification relates to compounds having a 1H-pyrazolo[4,3d]pyrimidine aromatic system, having activity as TLR7 agonists.
  • Figure US20230140047A1-20230504-C00005
  • In one aspect, there is provided a compound with a structure according to formula I
  • Figure US20230140047A1-20230504-C00006
    • wherein
    • each X1 is independently N or CR2;
    • X2 is O, CH2, NH, S, or N(C1-C3 alkyl);
    • R1 is H, CH3(CH2)1-3, CH3(CH2)0-1O(CH2)2-3, CH3(CH2)0-3C(═O), CH3(CH2)0-1O(CH2)2-3C(═O),
  • Figure US20230140047A1-20230504-C00007
    • R2 is H, O(C1-C3 alkyl), C1-C3 alkyl, Cl, F, or CN;
    • R3 is H, halo, OH, CN, NH2, NH(C1-C5 alkyl), N(C1-C5 alkyl)2, NH(CH2)0-1(C3-C6 cycloalkyl), NH(C4-C8 bicycloalkyl), NH(C6-C10 spirocycloalkyl), N(C3-C6 cycloalkyl)2, NH(CH2)1-3(aryl), N((CH2)1-3(aryl))2, a cyclic amine moiety having the structure
  • Figure US20230140047A1-20230504-C00008
      •  a 6-membered aromatic or heteroaromatic moiety or a 5-membered heteroaromatic moiety;
      • wherein
      • an alkyl, cycloalkyl, bicycloalkyl, spirocycloalkyl, cyclic amine, 6-membered aromatic or heteroaromatic, or 5-membered heteroaromatic moiety is optionally substituted with one or more substituents selected from OH, halo, CN, (C1-C3 alkyl), O(C1-C3 alkyl), C(═O)(Me), SO2(C1-C3 alkyl), C(═O)(Et), NH2, NH(Me), N(Me)2, NH(Et), N(Et)2, and N(C1-C3 alkyl), (CH2)1-2OH, (CH2)1-2OMe; and a cycloalkyl, bicycloalkyl, spirocycloalkyl, or cyclic amine moiety may have a CH2 group replaced by O, S, NH, N(C1-C3 alkyl), or N(Boc);
      • m is 0 or 1;
      • and
      • n is 1, 2, or 3;
      • or a pharmaceutically acceptable salt thereof.
  • Compounds disclosed herein have activity as TLR7 agonists and some can be conjugated to an antibody for targeted delivery to a target tissue or organ of intended action. They can also be PEGylated, to modulate their pharmaceutical properties.
  • Compounds disclosed herein, or their conjugates or their PEGylated derivatives, can be used in the treatment of a subject suffering from a condition amenable to treatment by activation of the immune system, by administering to such subject a therapeutically effective amount of such a compound or a conjugate thereof or a PEGylated derivative thereof, especially in combination with a vaccine or a cancer immunotherapy agent.
  • BRIEF DESCRIPTION OF THE DRAWING(S)
  • FIGS. 1, 2A, 2B, 3A, 3B, 4A, 4B, 5, 6A, 6B, 7, 8, 9, 10, 11, 12, 13 and 14 show reaction schemes for preparing compounds disclosed herein.
  • FIGS. 15 and 16 show schemes for the attachment of linkers to compounds of this disclosure, rendering them suitable for conjugation.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • “Antibody” means whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion”) or single chain variants thereof. A whole, or full length, antibody is a protein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain comprises a heavy chain variable region (VH) and a heavy chain constant region comprising three domains, CH1, CH2 and CH3. Each light chain comprises a light chain variable region (VL or Vk) and a light chain constant region comprising one single domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with more conserved framework regions (FRs). Each VH and VL comprises three CDRs and four FRs, arranged from amino- to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. The variable regions contain a binding domain that interacts with an antigen. The constant regions may mediate the binding of the antibody to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system. An antibody is said to “specifically bind” to an antigen X if the antibody binds to antigen X with a KD of 5×10−8 M or less, more preferably 1×10−8 M or less, more preferably 6×10−9 M or less, more preferably 3×10−9 M or less, even more preferably 2×10−9 M or less. The antibody can be chimeric, humanized, or, preferably, human. The heavy chain constant region can be engineered to affect glycosylation type or extent, to extend antibody half-life, to enhance or reduce interactions with effector cells or the complement system, or to modulate some other property. The engineering can be accomplished by replacement, addition, or deletion of one or more amino acids or by replacement of a domain with a domain from another immunoglobulin type, or a combination of the foregoing.
  • “Antigen binding fragment” and “antigen binding portion” of an antibody (or simply “antibody portion” or “antibody fragment”) mean one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody, such as (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fab′ fragment, which is essentially an Fab with part of the hinge region (see, for example, Abbas et al., Cellular and Molecular Immunology, 6th Ed., Saunders Elsevier 2007); (iv) a Fd fragment consisting of the VH and CH1 domains; (v) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (vi) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; (vii) an isolated complementarity determining region (CDR); and (viii) a nanobody, a heavy chain variable region containing a single variable domain and two constant domains. Preferred antigen binding fragments are Fab, F(ab′)2, Fab′, Fv, and Fd fragments. Furthermore, although the two domains of the Fv fragment, VL and VH, are encoded by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv, or scFv); see, e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also encompassed within the term “antigen-binding portion” of an antibody.
  • Unless indicated otherwise—for example by reference to the linear numbering in a SEQ ID NO: listing—references to the numbering of amino acid positions in an antibody heavy or light chain variable region (VH or VL) are according to the Kabat system (Kabat et al., “Sequences of proteins of immunological interest, 5th ed., Pub. No. 91-3242, U.S. Dept. Health & Human Services, NIH, Bethesda, Md., 1991, hereinafter “Kabat”) and references to the numbering of amino acid positions in an antibody heavy or light chain constant region (CH1, CH2, CH3, or CL) are according to the EU index as set forth in Kabat. See Lazar et al., US 2008/0248028 A1, the disclosure of which is incorporated herein by reference, for examples of such usage. Further, the ImMunoGeneTics Information System (IMGT) provides at its website a table entitled “IMGT Scientific Chart: Correspondence between C Numberings” showing the correspondence between its numbering system, EU numbering, and Kabat numbering for the heavy chain constant region.
  • An “isolated antibody” means an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds antigen X is substantially free of antibodies that specifically bind antigens other than antigen X). An isolated antibody that specifically binds antigen X may, however, have cross-reactivity to other antigens, such as antigen X molecules from other species. In certain embodiments, an isolated antibody specifically binds to human antigen X and does not cross-react with other (non-human) antigen X antigens. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • “Monoclonal antibody” or “monoclonal antibody composition” means a preparation of antibody molecules of single molecular composition, which displays a single binding specificity and affinity for a particular epitope.
  • “Human antibody” means an antibody having variable regions in which both the framework and CDR regions (and the constant region, if present) are derived from human germ-line immunoglobulin sequences. Human antibodies may include later modifications, including natural or synthetic modifications. Human antibodies may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). However, “human anti-body” does not include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • “Human monoclonal antibody” means an antibody displaying a single binding specificity, which has variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. In one embodiment, human monoclonal antibodies are produced by a hybridoma that includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.
  • “Aliphatic” means a straight- or branched-chain, saturated or unsaturated, non-aromatic hydrocarbon moiety having the specified number of carbon atoms (e.g., as in “C3 aliphatic,” “C1-5 aliphatic,” “C1-C5 aliphatic,” or “C1 to C5 aliphatic,” the latter three phrases being synonymous for an aliphatic moiety having from 1 to 5 carbon atoms) or, where the number of carbon atoms is not explicitly specified, from 1 to 4 carbon atoms (2 to 4 carbons in the instance of unsaturated aliphatic moieties). A similar understanding is applied to the number of carbons in other types, as in C2-4 alkene, C4-C7 cycloaliphatic, etc. In a similar vein, a term such as “(CH2)1-3” is to be understand as shorthand for the subscript being 1, 2, or 3, so that such term represents CH2, CH2CH2, and CH2CH2CH2.
  • “Alkyl” means a saturated aliphatic moiety, with the same convention for designating the number of carbon atoms being applicable. By way of illustration, C1-C4 alkyl moieties include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl, t-butyl, 1-butyl, 2-butyl, and the like. “Alkylene” means a divalent counterpart of an alkyl group, such as CH2CH2, CH2CH2CH2, and CH2CH2CH2CH2.
  • “Alkenyl” means an aliphatic moiety having at least one carbon-carbon double bond, with the same convention for designating the number of carbon atoms being applicable. By way of illustration, C2-C4 alkenyl moieties include, but are not limited to, ethenyl (vinyl), 2-propenyl (allyl or prop-2-enyl), cis-1-propenyl, trans-1-propenyl, E- (or Z-) 2-butenyl, 3-butenyl, 1,3-butadienyl (but-1,3-dienyl) and the like.
  • “Alkynyl” means an aliphatic moiety having at least one carbon-carbon triple bond, with the same convention for designating the number of carbon atoms being applicable. By way of illustration, C2-C4 alkynyl groups include ethynyl (acetylenyl), propargyl (prop-2-ynyl), 1-propynyl, but-2-ynyl, and the like.
  • “Cycloaliphatic” means a saturated or unsaturated, non-aromatic hydrocarbon moiety having from 1 to 3 rings, each ring having from 3 to 8 (preferably from 3 to 6) carbon atoms. “Cycloalkyl” means a cycloaliphatic moiety in which each ring is saturated. “Cycloalkenyl” means a cycloaliphatic moiety in which at least one ring has at least one carbon-carbon double bond. “Cycloalkynyl” means a cycloaliphatic moiety in which at least one ring has at least one carbon-carbon triple bond. By way of illustration, cycloaliphatic moieties include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cyclooctyl, and adamantyl. Preferred cycloaliphatic moieties are cycloalkyl ones, especially cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. “Cycloalkylene” means a divalent counterpart of a cycloalkyl group. Similarly, “bicycloalkylene” and “spirocycloalkylene” (or “spiroalkylene”) refer to divalent counterparts of a bicycloalkyl and spirocycloalkyl/spiroalkyl group.
  • “Heterocycloaliphatic” means a cycloaliphatic moiety wherein, in at least one ring thereof, up to three (preferably 1 to 2) carbons have been replaced with a heteroatom independently selected from N, O, or S, where the N and S optionally may be oxidized and the N optionally may be quaternized. Preferred cycloaliphatic moieties consist of one ring, 5- to 6-membered in size. Similarly, “heterocycloalkyl,” “heterocycloalkenyl,” and “heterocycloalkynyl” means a cycloalkyl, cycloalkenyl, or cycloalkynyl moiety, respectively, in which at least one ring thereof has been so modified. Exemplary heterocycloaliphatic moieties include aziridinyl, azetidinyl, 1,3-dioxanyl, oxetanyl, tetrahydrofuryl, pyrrolidinyl, piperidinyl, piperazinyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydrothiopyranyl sulfone, morpholinyl, thiomorpholinyl, thiomorpholinyl sulfoxide, thiomorpholinyl sulfone, 1,3-dioxolanyl, tetrahydro-1,1-dioxothienyl, 1,4-dioxanyl, thietanyl, and the like. “Heterocycloalkylene” means a divalent counterpart of a heterocycloalkyl group.
  • “Alkoxy,” “aryloxy,” “alkylthio,” and “arylthio” mean —O(alkyl), —O(aryl), —S(alkyl), and —S(aryl), respectively. Examples are methoxy, phenoxy, methylthio, and phenylthio, respectively.
  • “Halogen” or “halo” means fluorine, chlorine, bromine or iodine, unless a narrower meaning is indicated.
  • “Aryl” means a hydrocarbon moiety having a mono-, bi-, or tricyclic ring system (preferably monocyclic) wherein each ring has from 3 to 7 carbon atoms and at least one ring is aromatic. The rings in the ring system may be fused to each other (as in naphthyl) or bonded to each other (as in biphenyl) and may be fused or bonded to non-aromatic rings (as in indanyl or cyclohexylphenyl). By way of further illustration, aryl moieties include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthracenyl, and acenaphthyl. “Arylene” means a divalent counterpart of an aryl group, for example 1,2-phenylene, 1,3-phenylene, or 1,4-phenylene.
  • “Heteroaryl” means a moiety having a mono-, bi-, or tricyclic ring system (preferably 5- to 7-membered monocyclic) wherein each ring has from 3 to 7 carbon atoms and at least one ring is an aromatic ring containing from 1 to 4 heteroatoms independently selected from from N, O, or S, where the N and S optionally may be oxidized and the N optionally may be quatemized. Such at least one heteroatom containing aromatic ring may be fused to other types of rings (as in benzofuranyl or tetrahydroisoquinolyl) or directly bonded to other types of rings (as in phenylpyridyl or 2-cyclopentylpyridyl). By way of further illustration, heteroaryl moieties include pyrrolyl, furanyl, thiophenyl (thienyl), imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, tetrazolyl, pyridyl, N-oxopyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, quinolinyl, isoquinolynyl, quinazolinyl, cinnolinyl, quinozalinyl, naphthyridinyl, benzofuranyl, indolyl, benzothiophenyl, oxadiazolyl, thiadiazolyl, phenothiazolyl, benzimidazolyl, benzotriazolyl, dibenzofuranyl, carbazolyl, dibenzothiophenyl, acridinyl, and the like. “Heteroarylene” means a divalent counterpart of a heteroaryl group.
  • Where it is indicated that a moiety may be substituted, such as by use of “unsubstituted or substituted” or “optionally substituted” phrasing as in “unsubstituted or substituted C1-C5 alkyl” or “optionally substituted heteroaryl,” such moiety may have one or more independently selected substituents, preferably one to five in number, more preferably one or two in number. Substituents and substitution patterns can be selected by one of ordinary skill in the art, having regard for the moiety to which the substituent is attached, to provide compounds that are chemically stable and that can be synthesized by techniques known in the art as well as the methods set forth herein. Where a moiety is identified as being “unsubstituted or substituted” or “optionally substituted,” in a preferred embodiment such moiety is unsubstituted.
  • “Arylalkyl,” (heterocycloaliphatic)alkyl,” “arylalkenyl,” “arylalkynyl,” “biarylalkyl,” and the like mean an alkyl, alkenyl, or alkynyl moiety, as the case may be, substituted with an aryl, heterocycloaliphatic, biaryl, etc., moiety, as the case may be, with the open (unsatisfied) valence at the alkyl, alkenyl, or alkynyl moiety, for example as in benzyl, phenethyl, N-imidazoylethyl, N-morpholinoethyl, and the like. Conversely, “alkylaryl,” “alkenylcycloalkyl,” and the like mean an aryl, cycloalkyl, etc., moiety, as the case may be, substituted with an alkyl, alkenyl, etc., moiety, as the case may be, for example as in methylphenyl (tolyl) or allylcyclohexyl. “Hydroxyalkyl,” “haloalkyl,” “alkylaryl,” “cyanoaryl,” and the like mean an alkyl, aryl, etc., moiety, as the case may be, substituted with one or more of the identified substituent (hydroxyl, halo, etc., as the case may be).
  • For example, permissible substituents include, but are not limited to, alkyl (especially methyl or ethyl), alkenyl (especially allyl), alkynyl, aryl, heteroaryl, cycloaliphatic, heterocyclo-aliphatic, halo (especially fluoro), haloalkyl (especially trifluoromethyl), hydroxyl, hydroxyalkyl (especially hydroxyethyl), cyano, nitro, alkoxy, —O(hydroxyalkyl), —O(haloalkyl) (especially —OCF3), —O(cycloalkyl), —O(heterocycloalkyl), —O(aryl), alkylthio, arylthio, ═O, ═NH, ═N(alkyl), ═NOH, ═NO(alkyl), —C(═O)(alkyl), —C(═O)H, —CO2H, —C(═O)NHOH, —C(═O)O(alkyl), —C(═O)O(hydroxyalkyl), —C(═O)NH2, —C(═O)NH(alkyl), —C(═O)N(alkyl)2, —OC(═O)(alkyl), —OC(═O)(hydroxyalkyl), —OC(═O)O(alkyl), —OC(═O)O(hydroxyalkyl), —OC(═O)NH2, —OC(═O)NH(alkyl), —OC(═O)N(alkyl)2, azido, —NH2, —NH(alkyl), —N(alkyl)2, —NH(aryl), —NH(hydroxyalkyl), —NHC(═O)(alkyl), —NHC(═O)H, —NHC(═O)NH2, —NHC(═O)NH(alkyl), —NHC(═O)N(alkyl)2, —NHC(═NH)NH2, —OSO2(alkyl), —SH, —S(alkyl), —S(aryl), —S(cycloalkyl), —S(═O)alkyl, —SO2(alkyl), —SO2NH2, —SO2NH(alkyl), —SO2N(alkyl)2, and the like.
  • Where the moiety being substituted is an aliphatic moiety, preferred substituents are aryl, heteroaryl, cycloaliphatic, heterocycloaliphatic, halo, hydroxyl, cyano, nitro, alkoxy, —O(hydroxyalkyl), —O(haloalkyl), —O(cycloalkyl), —O(heterocycloalkyl), —O(aryl), alkylthio, arylthio, ═O, ═NH, ═N(alkyl), ═NOH, ═NO(alkyl), —CO2H, —C(═O)NHOH, —C(═O)O(alkyl), —C(═O)O(hydroxyalkyl), —C(═O)NH2, —C(═O)NH(alkyl), —C(═O)N(alkyl)2, —OC(═O)(alkyl), —OC(═O)(hydroxyalkyl), —OC(═O)O(alkyl), —OC(═O)O(hydroxyalkyl), —OC(═O)NH2, —OC(═O)NH(alkyl), —OC(═O)N(alkyl)2, azido, —NH2, —NH(alkyl), —N(alkyl)2, —NH(aryl), —NH(hydroxyalkyl), —NHC(═O)(alkyl), —NHC(═O)H, —NHC(═O)NH2, —NHC(═O)NH(alkyl), —NHC(═O)N(alkyl)2, —NHC(═NH)NH2, —OSO2(alkyl), —SH, —S(alkyl), —S(aryl), —S(═O)alkyl, —S(cycloalkyl), —SO2(alkyl), —SO2NH2, —SO2NH(alkyl), and —SO2N(alkyl)2. More preferred substituents are halo, hydroxyl, cyano, nitro, alkoxy, —O(aryl), ═O, ═NOH, ═NO(alkyl), —OC(═O)(alkyl), —OC(═O)O(alkyl), —OC(═O)NH2, —OC(═O)NH(alkyl), —OC(═O)N(alkyl)2, azido, —NH2, —NH(alkyl), —N(alkyl)2, —NH(aryl), —NHC(═O)(alkyl), —NHC(═O)H, —NHC(═O)NH2, —NHC(═O)NH(alkyl), —NHC(═O)N(alkyl)2, and —NHC(═NH)NH2. Especially preferred are phenyl, cyano, halo, hydroxyl, nitro, C1-C4alkyoxy, O(C2-C4 alkylene)OH, and O(C2-C4 alkylene)halo.
  • Where the moiety being substituted is a cycloaliphatic, heterocycloaliphatic, aryl, or heteroaryl moiety, preferred substituents are alkyl, alkenyl, alkynyl, halo, haloalkyl, hydroxyl, hydroxyalkyl, cyano, nitro, alkoxy, —O(hydroxyalkyl), —O(haloalkyl), —O(aryl), —O(cycloalkyl), —O(heterocycloalkyl), alkylthio, arylthio, —C(═O)(alkyl), —C(═O)H, —CO2H, —C(═O)NHOH, —C(═O)O(alkyl), —C(═O)O(hydroxyalkyl), —C(═O)NH2, —C(═O)NH(alkyl), —C(═O)N(alkyl)2, —OC(═O)(alkyl), —OC(═O)(hydroxyalkyl), —OC(═O)O(alkyl), —OC(═O)O(hydroxyalkyl), —OC(═O)NH2, —OC(═O)NH(alkyl), —OC(═O)N(alkyl)2, azido, —NH2, —NH(alkyl), —N(alkyl)2, —NH(aryl), —NH(hydroxyalkyl), —NHC(═O)(alkyl), —NHC(═O)H, —NHC(═O)NH2, —NHC(═O)NH(alkyl), —NHC(═O)N(alkyl)2, —NHC(═NH)NH2, —OSO2(alkyl), —SH, —S(alkyl), —S(aryl), —S(cycloalkyl), —S(═O)alkyl, —SO2(alkyl), —SO2NH2, —SO2NH(alkyl), and —SO2N(alkyl)2. More preferred substituents are alkyl, alkenyl, halo, haloalkyl, hydroxyl, hydroxyalkyl, cyano, nitro, alkoxy, —O(hydroxyalkyl), —C(═O)(alkyl), —C(═O)H, —CO2H, —C(═O)NHOH, —C(═O)O(alkyl), —C(═O)O(hydroxyalkyl), —C(═O)NH2, —C(═O)NH(alkyl), —C(═O)N(alkyl)2, —OC(═O)(alkyl), —OC(═O)(hydroxyalkyl), —OC(═O)O(alkyl), —OC(═O)O(hydroxyalkyl), —OC(═O)NH2, —OC(═O)NH(alkyl), —OC(═O)N(alkyl)2, —NH2, —NH(alkyl), —N(alkyl)2, —NH(aryl), —NHC(═O)(alkyl), —NHC(═O)H, —NHC(═O)NH2, —NHC(═O)NH(alkyl), —NHC(═O)N(alkyl)2, and —NHC(═NH)NH2. Especially preferred are C1-C4 alkyl, cyano, nitro, halo, and C1-C4alkoxy.
  • Where a range is stated, as in “C1-C5 alkyl” or “5 to 10%,” such range includes the end points of the range, as in C1 and C5 in the first instance and 5% and 10% in the second instance.
  • Unless particular stereoisomers are specifically indicated (e.g., by a bolded or dashed bond at a relevant stereocenter in a structural formula, by depiction of a double bond as having E or Z configuration in a structural formula, or by use stereochemistry-designating nomenclature or symbols), all stereoisomers are included within the scope of the invention, as pure compounds as well as mixtures thereof. Unless otherwise indicated, racemates, individual enantiomers (whether optically pure or partially resolved), diastereomers, geometrical isomers, and combinations and mixtures thereof are all encompassed by this invention.
  • Those skilled in the art will appreciate that compounds may have tautomeric forms (e.g., keto and enol forms), resonance forms, and zwitterionic forms that are equivalent to those depicted in the structural formulae used herein and that the structural formulae encompass such tautomeric, resonance, or zwitterionic forms.
  • “Pharmaceutically acceptable ester” means an ester that hydrolyzes in vivo (for example in the human body) to produce the parent compound or a salt thereof or has per se activity similar to that of the parent compound. Suitable esters include C1-C5 alkyl, C2-C5 alkenyl or C2-C5 alkynyl esters, especially methyl, ethyl or n-propyl.
  • “Pharmaceutically acceptable salt” means a salt of a compound suitable for pharmaceutical formulation. Where a compound has one or more basic groups, the salt can be an acid addition salt, such as a sulfate, hydrobromide, tartrate, mesylate, maleate, citrate, phosphate, acetate, pamoate (embonate), hydroiodide, nitrate, hydrochloride, lactate, methylsulfate, fumarate, benzoate, succinate, mesylate, lactobionate, suberate, tosylate, and the like. Where a compound has one or more acidic groups, the salt can be a salt such as a calcium salt, potassium salt, magnesium salt, meglumine salt, ammonium salt, zinc salt, piperazine salt, tromethamine salt, lithium salt, choline salt, diethylamine salt, 4-phenylcyclohexylamine salt, benzathine salt, sodium salt, tetramethylammonium salt, and the like. Polymorphic crystalline forms and solvates are also encompassed within the scope of this invention.
  • “Subject” refers to an animal, including, but not limited to, a primate (e.g, human), monkey, cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms “subject” and “patient” are used interchangeably herein in reference, for example, to a mammalian subject, such as a human.
  • The terms “treat,” “treating,” and “treatment,” in the context of treating a disease or disorder, are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or to slowing the progression, spread or worsening of a disease, disorder or condition or of one or more symptoms thereof. The “treatment of cancer”, refers to one or more of the following effects: (1) inhibition, to some extent, of tumor growth, including, (i) slowing down and (ii) complete growth arrest; (2) reduction in the number of tumor cells; (3) maintaining tumor size; (4) reduction in tumor size; (5) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of tumor cell infiltration into peripheral organs; (6) inhibition, including (i) reduction, (ii) slowing down or (iii) complete prevention, of metastasis; (7) enhancement of anti-tumor immune response, which may result in (i) maintaining tumor size, (ii) reducing tumor size, (iii) slowing the growth of a tumor, (iv) reducing, slowing or preventing invasion and/or (8) relief, to some extent, of the severity or number of one or more symptoms associated with the disorder.
    Figure US20230140047A1-20230504-P00001
  • In the formulae of this specification, a wavy line (
    Figure US20230140047A1-20230504-P00001
    ) transverse to a bond or an asterisk (*) at the end of the bond denotes a covalent attachment site. For instance, a statement that R is
  • Figure US20230140047A1-20230504-C00009
  • or that R is
  • Figure US20230140047A1-20230504-C00010
  • in the formula
  • Figure US20230140047A1-20230504-C00011
  • means
  • Figure US20230140047A1-20230504-C00012
  • In the formulae of this specification, a bond traversing an aromatic ring between two carbons thereof means that the group attached to the bond may be located at any of the positions of the aromatic ring made available by removal of the hydrogen that is implicitly there. By way of illustration, the formula
  • Figure US20230140047A1-20230504-C00013
  • represents
  • Figure US20230140047A1-20230504-C00014
  • In other illustrations,
  • Figure US20230140047A1-20230504-C00015
  • represents
  • Figure US20230140047A1-20230504-C00016
  • and represents
  • Figure US20230140047A1-20230504-C00017
  • Those skilled in the art will appreciate that certain structures can be drawn in one tautomeric form or another—for example, keto versus enol—and that the two forms are equivalent.
  • Compounds
  • In one embodiment, either each X1 is CR2 or not more than two X1's are N in the moiety
  • Figure US20230140047A1-20230504-C00018
  • Examples of such are
  • Figure US20230140047A1-20230504-C00019
  • A preferred example is
  • Figure US20230140047A1-20230504-C00020
  • equals
  • Figure US20230140047A1-20230504-C00021
  • Suitable groups R1 include:
  • Figure US20230140047A1-20230504-C00022
  • Preferably the group R1 is
  • Figure US20230140047A1-20230504-C00023
  • Examples of suitable groups R3 include Cl, OH,
  • Figure US20230140047A1-20230504-C00024
    Figure US20230140047A1-20230504-C00025
    Figure US20230140047A1-20230504-C00026
  • Examples of where the group R3 has the structure
  • Figure US20230140047A1-20230504-C00027
  • (including instances with one or more methylene (CH2) groups optionally replaced by one or more of O, S, SO2, NH, C(═O), N(C1-C3 alkyl), NC(═O)(C1-C3 alkyl), or N(Boc), or has another ring fused thereto, as disclosed hereinabove) are:
  • Figure US20230140047A1-20230504-C00028
  • In another embodiment, R3 is selected from the group consisting of
  • Figure US20230140047A1-20230504-C00029
  • In one embodiment of formula I, m is 0, in which case the formula simplifies to formula I:
  • Figure US20230140047A1-20230504-C00030
  • Another embodiment of compounds according to formula I is represented by formula Ia:
  • Figure US20230140047A1-20230504-C00031
  • where R1 and R3 are as defined in respect of formulae I hereinabove.
  • An embodiment of compounds according to formula I wherein m is 1 is represented by formula Ib, wherein R1, R3 and X1 are as defined in respect of formula I hereinabove.
  • Figure US20230140047A1-20230504-C00032
  • Preferably, in formula Ib
  • Figure US20230140047A1-20230504-C00033
  • more preferably
  • Figure US20230140047A1-20230504-C00034
  • Specific examples of compounds per in this specification are listed following. Methods for their preparation and their properties are provided in the experimental sections hereinbelow.
  • No. Structure
    800
    Figure US20230140047A1-20230504-C00035
    801
    Figure US20230140047A1-20230504-C00036
    802
    Figure US20230140047A1-20230504-C00037
    803
    Figure US20230140047A1-20230504-C00038
    804
    Figure US20230140047A1-20230504-C00039
    805
    Figure US20230140047A1-20230504-C00040
    806
    Figure US20230140047A1-20230504-C00041
    807
    Figure US20230140047A1-20230504-C00042
    808
    Figure US20230140047A1-20230504-C00043
    809
    Figure US20230140047A1-20230504-C00044
    810
    Figure US20230140047A1-20230504-C00045
    811
    Figure US20230140047A1-20230504-C00046
    812
    Figure US20230140047A1-20230504-C00047
    813
    Figure US20230140047A1-20230504-C00048
    814
    Figure US20230140047A1-20230504-C00049
    815
    Figure US20230140047A1-20230504-C00050
    816
    Figure US20230140047A1-20230504-C00051
    817
    Figure US20230140047A1-20230504-C00052
    818
    Figure US20230140047A1-20230504-C00053
    819
    Figure US20230140047A1-20230504-C00054
    820
    Figure US20230140047A1-20230504-C00055
    821
    Figure US20230140047A1-20230504-C00056
    822
    Figure US20230140047A1-20230504-C00057
    823
    Figure US20230140047A1-20230504-C00058
    824
    Figure US20230140047A1-20230504-C00059
    825
    Figure US20230140047A1-20230504-C00060
    826
    Figure US20230140047A1-20230504-C00061
    827
    Figure US20230140047A1-20230504-C00062
    828
    Figure US20230140047A1-20230504-C00063
    829
    Figure US20230140047A1-20230504-C00064
    830
    Figure US20230140047A1-20230504-C00065
    831
    Figure US20230140047A1-20230504-C00066
    832
    Figure US20230140047A1-20230504-C00067
    833
    Figure US20230140047A1-20230504-C00068
    834
    Figure US20230140047A1-20230504-C00069
    835
    Figure US20230140047A1-20230504-C00070
    836
    Figure US20230140047A1-20230504-C00071
    837
    Figure US20230140047A1-20230504-C00072
    838
    Figure US20230140047A1-20230504-C00073
    839
    Figure US20230140047A1-20230504-C00074
    840
    Figure US20230140047A1-20230504-C00075
    841
    Figure US20230140047A1-20230504-C00076
    842
    Figure US20230140047A1-20230504-C00077
    843
    Figure US20230140047A1-20230504-C00078
    844
    Figure US20230140047A1-20230504-C00079
    845
    Figure US20230140047A1-20230504-C00080
    846
    Figure US20230140047A1-20230504-C00081
    847
    Figure US20230140047A1-20230504-C00082
    848
    Figure US20230140047A1-20230504-C00083
    849
    Figure US20230140047A1-20230504-C00084
    850
    Figure US20230140047A1-20230504-C00085
    851
    Figure US20230140047A1-20230504-C00086
    852
    Figure US20230140047A1-20230504-C00087
    853
    Figure US20230140047A1-20230504-C00088
    854
    Figure US20230140047A1-20230504-C00089
    855
    Figure US20230140047A1-20230504-C00090
    856
    Figure US20230140047A1-20230504-C00091
    857
    Figure US20230140047A1-20230504-C00092
    858
    Figure US20230140047A1-20230504-C00093
    859
    Figure US20230140047A1-20230504-C00094
    860
    Figure US20230140047A1-20230504-C00095
    861
    Figure US20230140047A1-20230504-C00096
    862
    Figure US20230140047A1-20230504-C00097
    863
    Figure US20230140047A1-20230504-C00098
    864
    Figure US20230140047A1-20230504-C00099
    865
    Figure US20230140047A1-20230504-C00100
    866
    Figure US20230140047A1-20230504-C00101
    867
    Figure US20230140047A1-20230504-C00102
    868
    Figure US20230140047A1-20230504-C00103
    869
    Figure US20230140047A1-20230504-C00104
    870
    Figure US20230140047A1-20230504-C00105
    871
    Figure US20230140047A1-20230504-C00106
    872
    Figure US20230140047A1-20230504-C00107
    873
    Figure US20230140047A1-20230504-C00108
    874
    Figure US20230140047A1-20230504-C00109
    875
    Figure US20230140047A1-20230504-C00110
    876
    Figure US20230140047A1-20230504-C00111
    877
    Figure US20230140047A1-20230504-C00112
    878
    Figure US20230140047A1-20230504-C00113
    879
    Figure US20230140047A1-20230504-C00114
  • CONJUGATES General
  • TLR7 agonists disclosed herein can be delivered to the site of intended action by localized administration or by targeted delivery in a conjugate with a targeting moiety. Preferably, the targeting moiety is an antibody or antigen binding portion thereof and its antigen is found at the locality of intended action, for example a tumor associated antigen if the intended site of action is at a tumor (cancer). Preferably, the tumor associated antigen is uniquely expressed or overexpressed by the cancer cell, compared to a normal cell. The tumor associated antigen can be located on the surface of the cancer cell or secreted by the cancer cell into its environs.
  • In one aspect, there is provided a conjugate comprising compound of this invention and a ligand, represented by formula (IV)

  • [D(X D)a(C)c(X Z)b]m Z  (IV)
  • where Z is a targeting moiety, D is an agonist of this invention, and —(XD)aC(XZ)b— are collectively referred to as a “linker moiety” or “linker” because they link Z and D. Within the linker, C is a cleavable group designed to be cleaved at or near the site of intended biological action of D; XD and XZ are spacer moieties (or “spacers”) that space apart D and C and C and Z, respectively; subscripts a, b, and c are independently 0 or 1 (that is, the presence of XD, XZ and C are optional). Subscript m is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 (preferably 1, 2, 3, or 4). D, XD, C, XZ and Z are more fully described hereinbelow.
  • By binding to a target tissue or cell where its antigen or receptor is located, Z directs the conjugate there. Cleavage of group C at the target tissue or cell releases D to exert its effect locally. In this manner, precise delivery of D is achieved at the site of intended action, reducing the dosage needed. Also, D is normally biologically inactive (or significantly less active) in its conjugated state, thereby reducing off-target effects.
  • As reflected by the subscript m, each Z can conjugate with more than one D, depending on the number of sites Z has available for conjugation and the experimental conditions employed. Those skilled in the art will appreciate that, while each individual Z is conjugated to an integer number of Ds, a preparation of the conjugate may analyze for a non-integer ratio of D to Z, reflecting a statistical average. This ratio is referred to as the substitution ratio (“SR”) or the drug-antibody ratio (“DAR”).
  • Targeting Moiety Z
  • Preferably, targeting moiety Z is an antibody. For convenience and brevity and not by way of limitation, the detailed discussion in this specification about Z and its conjugates is written in the context of its being an antibody, but those skilled in the art will understand that other types of Z can be conjugated, mutatis mutandis. For example, conjugates with folic acid as the targeting moiety can target cells having the folate receptor on their surfaces (Leamon et al., Cancer Res. 2008, 68 (23), 9839). For the same reasons, the detailed discussion in this specification is primarily written in terms of a 1:1 ratio of Z to D (m=1).
  • Antibodies that can be used in conjugates of this invention include those recognizing the following antigens: mesothelin, prostate specific membrane antigen (PSMA), CD19, CD22, CD30, CD70, B7H3, B7H4 (also known as 08E), protein tyrosine kinase 7 (PTK7), glypican-3, RG1, fucosyl-GM1, CTLA-4, and CD44. The antibody can be animal (e.g., murine), chimeric, humanized, or, preferably, human. The antibody preferably is monoclonal, especially a monoclonal human antibody. The preparation of human monoclonal antibodies against some of the aforementioned antigens is disclosed in Korman et al., U.S. Pat. No. 8,609,816 B2 (2013; B7H4, also known as 08E; in particular antibodies 2A7, 1G11, and 2F9); Rao-Naik et al., 8,097,703 B2 (2012; CD19; in particular antibodies 5G7, 13F1, 46E8, 21D4, 21D4a, 47G4, 27F3, and 3C10); King et al., U.S. Pat. No. 8,481,683 B2 (2013; CD22; in particular antibodies 12C5, 19A3, 16F7, and 23C6); Keler et al., U.S. Pat. No. 7,387,776 B2 (2008; CD30; in particular antibodies 5F11, 2H9, and 17G1); Terrett et al., U.S. Pat. No. 8,124,738 B2 (2012; CD70; in particular antibodies 2H5, 10B4, 8B5, 18E7, and 69A7); Korman et al., U.S. Pat. No. 6,984,720 B1 (2006; CTLA-4; in particular antibodies 10D1, 4B6, and 1E2); Korman et al., U.S. Pat. No. 8,008,449 B2 (2011; PD-1; in particular antibodies 17D8, 2D3, 4H1, 5C4, 4A11, 7D3, and 5F4); Huang et al., US 2009/0297438 A1 (2009; PSMA. in particular antibodies 1C3, 2A10, 2F5, 2C6); Cardarelli et al., U.S. Pat. No. 7,875,278 B2 (2011; PSMA; in particular antibodies 4A3, 7F12, 8C12, 8A11, 16F9, 2A10, 2C6, 2F5, and 1C3); Terrett et al., U.S. Pat. No. 8,222,375 B2 (2012; PTK7; in particular antibodies 3G8, 4D5, 12C6, 12C6a, and 7C8); Harkins et al., U.S. Pat. No. 7,335,748 B2(2008; RG1; in particular antibodies A, B, C, and D); Terrett et al., U.S. Pat. No. 8,268,970 B2 (2012; mesothelin; in particular antibodies 3C10, 6A4, and 7B1); Xu et al., US 2010/0092484 A1 (2010; CD44; in particular antibodies 14G9.B8.B4, 2D1.A3.D12, and 1A9.A6.B9); Deshpande et al., U.S. Pat. No. 8,258,266 B2 (2012; IP10; in particular antibodies 1D4, 1E1, 2G1, 3C4, 6A5, 6A8, 7C10, 8F6, 10A12, 10A12S, and 13C4); Kuhne et al., U.S. Pat. No. 8,450,464 B2 (2013; CXCR4; in particular antibodies F7, F9, D1, and E2); and Korman et al., U.S. Pat. No. 7,943,743 B2 (2011; PD-L1; in particular antibodies 3G10, 12A4, 10A5, 5F8, 10H10, 1B12, 7H1, 11E6, 12B7, and 13G4); the disclosures of which are incorporated herein by reference. Preferably, the antibody is an anti-mesothelin antibody.
  • In addition to being an antibody, Z can also be an antibody fragment (such as Fab, Fab′, F(ab′)2, Fd, or Fv) or antibody mimetic, such as an affibody, a domain antibody (dAb), a nanobody, a unibody, a DARPin, an anticalin, a versabody, a duocalin, a lipocalin, or an avimer.
  • Any one of several different reactive groups on Z can be a conjugation site, including ε-amino groups in lysine residues, pendant carbohydrate moieties, carboxylic acid groups on aspartic or glutamic acid side chains, cysteine-cysteine disulfide groups, and cysteine thiol groups. For reviews on antibody reactive groups suitable for conjugation, see, e.g., Garnett, Adv. Drug Delivery Rev. 2001, 53, 171-216 and Dubowchik and Walker, Pharmacology & Therapeutics 1999, 83, 67-123, the disclosures of which are incorporated herein by reference.
  • Most antibodies have multiple lysine residues, which can be conjugated via their 8-amino groups via amide, urea, thiourea, or carbamate bonds.
  • A thiol (—SH) group in the side chain of a cysteine can be used to form a conjugate by several methods. It can be used to form a disulfide bond between it and a thiol group on the linker. Another method is via its Michael addition to a maleimide group on the linker.
  • Typically, although antibodies have cysteine residues, they lack free thiol groups because all their cysteines are engaged in intra- or inter-chain disulfide bonds. To generate a free thiol group, a native disulfide group can be reduced. See, e.g., Packard et al., Biochemistry 1986, 25, 3548; King et al., Cancer Res. 1994, 54, 6176; and Doronina et al., Nature Biotechnol. 2003, 21, 778. Alternatively, a cysteine having a free —SH group can be introduced by mutating the antibody, substituting a cysteine for another amino acid or inserting one into the polypeptide chain. See, for example, Eigenbrot et al., U.S. Pat. No. 7,521,541 B2 (2009); Chilkoti et al., Bioconjugate Chem. 1994, 5, 504; Urnovitz et al., U.S. Pat. No. 4,698,420 (1987); Stimmel et al., J. Biol. Chem. 2000, 275, 30445; Bam et al., U.S. Pat. No. 7,311,902 B2 (2007); Kuan et al., J. Biol. Chem. 1994, 269, 7610; Poon et al., J. Biol. Chem. 1995, 270, 8571; Junutula et al., Nature Biotechnology 2008, 26, 925 and Rajpal et al., U.S. Provisional Application No. 62/270,245, filed Dec. 21, 2015. In yet another approach, a cysteine is added to the C-terminus of the heavy of light chain. See, e.g., Liu et al., U.S. Pat. No. 8,865,875 B2 (2014); Cumber et al., J. Immunol. 1992, 149, 120; King et al, Cancer Res. 1994, 54, 6176; Li et al., Bioconjugate Chem. 2002, 13, 985; Yang et al., Protein Engineering 2003, 16, 761; and Olafson et al., Protein Engineering Design & Selection 2004, 17, 21. The disclosures of the documents cited in this paragraph are incorporated herein by reference.
  • Linkers and Their Components
  • As noted above, the linker comprises up to three elements: a cleavable group C and optional spacers XZ and XD.
  • Group C is cleavable under physiological conditions. Preferably it is relatively stable while the conjugate is in circulation in the blood, but is readily cleaved once the conjugate reaches its site of intended action.
  • A preferred group C is a peptide that is cleaved selectively by a protease inside the target cell, as opposed to by a protease in the serum. Typically, the peptide comprises from 1 to 20 amino acids, preferably from 1 to 6 amino acids, more preferably from 2 to 3 amino acids. The amino acid(s) can be natural and/or non-natural α-amino acids. Natural amino acids are those encoded by the genetic code, as well as amino acids derived therefrom, e.g., hydroxyproline, γ-carboxyglutamate, citrulline, and O-phosphoserine. In this specification, the term “amino acid” also includes amino acid analogs and mimetics. Analogs are compounds having the same general H2N(R)CHCO2H structure of a natural amino acid, except that the R group is not one found among the natural amino acids. Examples of analogs include homoserine, norleucine, methionine-sulfoxide, and methionine methyl sulfonium. An amino acid mimetic is a compound that has a structure different from the general chemical structure of an α-amino acid but functions in a manner similar to one. The amino acid can be of the “L” stereochemistry of the genetically encoded amino acids, as well as of the enantiomeric “D” stereochemistry.
  • Preferably, C contains an amino acid sequence that is a cleavage recognition sequence for a protease. Many cleavage recognition sequences are known in the art. See, e.g., Matayoshi et al. Science 247: 954 (1990); Dunn et al. Meth. Enzymol. 241: 254 (1994); Seidah et al. Meth. Enzymol. 244: 175 (1994); Thomberry, Meth. Enzymol. 244: 615 (1994); Weber et al. Meth. Enzymol. 244: 595 (1994); Smith et al. Meth. Enzymol. 244: 412 (1994); and Bouvier et al. Meth. Enzymol. 248: 614 (1995); the disclosures of which are incorporated herein by reference.
  • A group C can be chosen such that it is cleaved by a protease present in the extracellular matrix in the vicinity of a cancer, e.g., a protease released by nearby dying cancer cells or a tumor-associated protease secreted by cancer cells. Exemplary extracellular tumor-associated proteases are plasmin, matrix metalloproteases (MMP), thimet oligopeptidase (TOP) and CD10. See, e.g., Trouet et al., U.S. Pat. No. 7,402,556 B2 (2008); Dubois et al., U.S. Pat. No. 7,425,541 B2 (2008); and Bebbington et al., U.S. Pat. No. 6,897,034 B2 (2005). Cathepsin D, normally lysosomal enzyme found inside cells, is sometimes found in the environs of a tumor, possibly released by dying cancer cells.
  • For conjugates designed to be by an enzyme, C preferably comprises an amino acid sequence selected for cleavage by proteases such cathepsins B, C, D, H, L and S, especially cathepsin B. Exemplary cathepsin B cleavable peptides include Val-Ala, Val-Cit, Val-Lys, Lys-Val-Ala, Asp-Val-Ala, Val-Ala, Lys-Val-Cit, Ala-Val-Cit, Val-Gly, Val-Gln, and Asp-Val-Cit. (Herein, amino acid sequences are written in the N-to-C direction, as in H2N-AA2-AA1-CO2H, unless the context clearly indicates otherwise.) See Dubowchik et al., Biorg. Med. Chem. Lett. 1998, 8, 3341; Dubowchik et al., Bioorg. Med. Chem. Lett. 1998, 8, 3347; and Dubowchik et al., Bioconjugate Chem. 2002, 13, 855; the disclosures of which are incorporated by reference.
  • Another enzyme that can be utilized for cleaving peptidyl linkers is legumain, a lysosomal cysteine protease that preferentially cleaves at Ala-Ala-Asn.
  • In one embodiment, Group C is a peptide comprising a two-amino acid sequence -AA2-AA1- wherein AA1 is lysine, arginine, or citrulline and AA2 is phenylalanine, valine, alanine, leucine or isoleucine. In another embodiment, C consists of a sequence of one to three amino acids, selected from the group consisting of Val-Cit, Ala-Val, Val-Ala-Val, Lys-Lys, Ala-Asn-Val, Val-Leu-Lys, Cit-Cit, Val-Lys, Ala-Ala-Asn, Lys, Cit, Ser, and Glu. More preferably, it is a two to three amino acid peptide from the foregoing group.
  • The preparation and design of cleavable groups C consisting of a single amino acid is disclosed in Chen et al., U.S. Pat. No. 8,664,407 B2 (2014), the disclosure of which is incorporated herein by reference.
  • Group C can be bonded directly to Z or D; i.e. spacers XZ or XD, as the case may be, can be absent.
  • When present, spacer XZ provides spatial separation between C and Z, least the former sterically interfere with antigen binding by latter or the latter sterically interfere with cleavage of the former. Further, spacer XZ can be used to confer increased solubility or decreased aggregation properties to conjugates. A spacer XZ can comprise one or more modular segments, which can be assembled in any number of combinations. Examples of suitable segments for a spacer XZ are:
  • Figure US20230140047A1-20230504-C00115
  • and combinations thereof, where the subscript g is 0 or 1 and the subscript h is 1 to 24, preferably 2 to 4. These segments can be combined, such as illustrated below:
  • Figure US20230140047A1-20230504-C00116
  • Spacer XD, if present, provides spatial separation between C and D, least the latter interfere sterically or electronically with cleavage of the former. Spacer XD also can serve to introduce additional molecular mass and chemical functionality into a conjugate. Generally, the additional mass and functionality will affect the serum half-life and other properties of the conjugate. Thus, through judicious selection of spacer groups, the serum half-live of a conjugate can be modulated. Spacer XD also can be assembled from modular segments, analogously to the description above for spacer XZ.
  • Spacers XZ and/or XD, where present, preferably provide a linear separation of from 4 to 25 atoms, more preferably from 4 to 20 atoms, between Z and C or D and C, respectively.
  • The linker can perform other functions in addition to covalently linking the antibody and the drug. For instance, the linker can contain a poly(ethylene glycol) (“PEG”) group. Since the conjugation step typically involves coupling a drug-linker to an antibody in an aqueous medium, a PEG group many enhance the aqueous solubility of the drug-linker. Also, a PEG group may enhance the solubility or reduce aggregation in the resulting ADC. Where a PEG group is present, it may be incorporated into either spacer XZ of XD, or both. The number of repeat units in a PEG group can be from 2 to 20, preferably between 4 and 10.
  • Either spacer XZ or XD, or both, can comprise a self-immolating moiety. A self-immolating moiety is a moiety that (1) is bonded to C and either Z or D and (2) has a structure such that cleavage from group C initiates a reaction sequence resulting in the self-immolating moiety disbonding itself from Z or D, as the case may be. In other words, reaction at a site distal from Z or D (cleavage from group C) causes the XZ—Z or the XD-D bond to rupture as well. The presence of a self-immolating moiety is desirable in the case of spacer XD because, if, after cleavage of the conjugate, spacer XD or a portion thereof were to remain attached to D, the biological activity of D may be impaired. The use of a self-immolating moiety is especially desirable where cleavable group C is a polypeptide, in which instance the self-immolating moiety typically is located adjacent thereto, in order to prevent D from sterically or electronically interfering with peptide cleavage.
  • Exemplary self-immolating moieties (i)-(v) bonded to a hydroxyl or amino group of D are shown below:
  • Figure US20230140047A1-20230504-C00117
    Figure US20230140047A1-20230504-C00118
  • The self-immolating moiety is the structure between dotted lines a and b (or dotted lines b and c), with adjacent structural features shown to provide context. Self-immolating moieties (i) and (v) are bonded to a D-NH2 (i.e., conjugation is via an amino group), while self-immolating moieties (ii), (iii), and (iv) are bonded to a D-OH (i.e., conjugation is via a hydroxyl or carboxyl group). Cleavage of the bond at dotted line b by an enzyme—a peptidase in the instance of structures (i)-(v) and a β-glucuronidase in the instance of structure (vi)—initiates a self-immolating reaction sequence that results in the cleavage of the bond at dotted line a and the consequent release of D-OH or D-NH2, as the case may be. By way of illustration, self-immolating mechanisms for structures (i) and (iv) are shown below:
  • Figure US20230140047A1-20230504-C00119
  • In other words, cleavage of a first chemical bond at one part of a self-immolating group initiates a sequence of steps that results in the cleavage of a second chemical bond—the one connecting the self-immolating group to the drug—at a different part of the self-immolating group, thereby releasing the drug.
  • In some instances, self-immolating groups can be used in tandem, as shown by structure (vii). In such case, cleavage at dotted line c triggers self-immolation of the moiety between dotted lines b and c by a 1,6-elimination reaction, followed by self-immolation of the moiety between dotted lines a and b by a cyclization-elimination reaction. For additional disclosures regarding self-immolating moieties, see Carl et al., J. Med. Chem. 1981, 24, 479; Carl et al., WO 81/01145 (1981); Dubowchik et al., Pharmacology & Therapeutics 1999, 83, 67; Firestone et al., U.S. Pat. No. 6,214,345 B1 (2001); Toki et al., J. Org. Chem. 2002, 67, 1866; Doronina et al., Nature Biotechnology 2003, 21, 778 (erratum, p. 941); Boyd et al., U.S. Pat. No. 7,691,962 B2; Boyd et al., US 2008/0279868 A1; Sufi et al., WO 2008/083312 A2; Feng, U.S. Pat. No. 7,375,078 B2; Jeffrey et al., U.S. Pat. No. 8,039,273; and Senter et al., US 2003/0096743 A1; the disclosures of which are incorporated by reference.
  • In another embodiment, Z and D are linked by a non-cleavable linker, i.e., C is absent. Metabolism of D eventually reduces the linker to a small appended moiety that does not interfere with the biological activity of D.
  • Conjugation Techniques
  • Conjugates of TLR7 agonists disclosed herein preferably are made by first preparing a compound comprising D and linker (XD)a(C)c(XZ)n (where XD, C, XZ, a, b, and c are as defined for formula (II)) to form drug-linker compound represented by formula (V):

  • D-(X D)a(C)c(X Z)b —R 31  (V)
  • where R31 is a functional group suitable for reacting with a complementary functional group on Z to form the conjugate. Examples of suitable groups R31 include amino, azide, thiol, cyclooctyne,
  • Figure US20230140047A1-20230504-C00120
  • where R32 is Cl, Br, F, mesylate, or tosylate and R33 is Cl, Br, I, F, OH, —O—N-succinimidyl, —O-(4-nitrophenyl), —O-pentafluorophenyl, or —O-tetrafluorophenyl. Chemistry generally usable for the preparation of suitable moieties D-(XD)aC(XZ)b—R31 is disclosed in Ng et al., U.S. Pat. No. 7,087,600 B2 (2006); Ng et al., U.S. Pat. No. 6,989,452 B2 (2006); Ng et al., U.S. Pat. No. 7,129,261 B2 (2006); Ng et al., WO 02/096910 A1; Boyd et al., U.S. Pat. No. 7,691,962 B2; Chen et al., U.S. Pat. No. 7,517,903 B2 (2009); Gangwar et al., U.S. Pat. No. 7,714,016 B2 (2010); Boyd et al., US 2008/0279868 A1; Gangwar et al., U.S. Pat. No. 7,847,105 B2 (2010); Gangwar et al., U.S. Pat. No. 7,968,586 B2 (2011); Sufi et al., U.S. Pat. No. 8,461,117 B2 (2013); and Chen et al., U.S. Pat. No. 8,664,407 B2 (2014); the disclosures of which are incorporated herein by reference.
  • Preferably reactive functional group —R31 is —NH2, —OH, —CO2H, —SH, maleimido, cyclooctyne, azido (—N3), hydroxylamino (—ONH2) or N-hydroxysuccinimido. Especially preferred functional groups —R31 are:
  • Figure US20230140047A1-20230504-C00121
  • An —OH group can be esterified with a carboxy group on the antibody, for example, on an aspartic or glutamic acid side chain.
  • A —CO2H group can be esterified with a —OH group or amidated with an amino group (for example on a lysine side chain) on the antibody.
  • An N-hydroxysuccinimide group is functionally an activated carboxyl group and can conveniently be amidated by reaction with an amino group (e.g., from lysine).
  • A maleimide group can be conjugated with an —SH group on the antibody (e.g., from cysteine or from the chemical modification of the antibody to introduce a sulfhydryl functionality), in a Michael addition reaction.
  • Where an antibody does not have a cysteine —SH available for conjugation, an ε-amino group in the side chain of a lysine residue can be reacted with 2-iminothiolane or N-succinimidyl-3-(2-pyridyldithio)propionate (“SPDP”) to introduce a free thiol (—SH) group-creating a cysteine surrogate, as it were. The thiol group can react with a maleimide or other nucleophile acceptor group to effect conjugation. The mechanism if illustrated below with 2-iminothiolane.
  • Figure US20230140047A1-20230504-C00122
  • Typically, a thiolation level of two to three thiols per antibody is achieved. For a representative procedure, see Cong et al., U.S. Pat. No. 8,980,824 B2 (2015), the disclosure of which is incorporated herein by reference.
  • In a reversed arrangement, an antibody Z can be modified with N-succinimidyl 4-(maleimidomethyl)-cyclohexanecarboxylate (“SMCC”) or its sulfonated variant sulfo-SMCC, both of which are available from Sigma-Aldrich, to introduce a maleimide group thereto. Then, conjugation can be effected with a drug-linker compound having an —SH group on the linker.
  • An alternative conjugation method employs copper-free “click chemistry,” in which an azide group adds across a strained cyclooctyne to form an 1,2,3-triazole ring. See, e.g., Agard et al., J. Amer. Chem. Soc. 2004, 126, 15046; Best, Biochemistry 2009, 48, 6571, the disclosures of which are incorporated herein by reference. The azide can be located on the antibody and the cyclooctyne on the drug-linker moiety, or vice-versa. A preferred cyclooctyne group is dibenzocyclooctyne (DIBO). Various reagents having a DIBO group are available from Invitrogen/Molecular Probes, Eugene, Oreg. The reaction below illustrates click chemistry conjugation for the instance in which the DIBO group is attached to the antibody (Ab):
  • Figure US20230140047A1-20230504-C00123
  • Yet another conjugation technique involves introducing a non-natural amino acid into an antibody, with the non-natural amino acid providing a functionality for conjugation with a reactive functional group in the drug moiety. For instance, the non-natural amino acid p-acetylphenylalanine can be incorporated into an antibody or other polypeptide, as taught in Tian et al., WO 2008/030612 A2 (2008). The ketone group in p-acetylphenyalanine can be a conjugation site via the formation of an oxime with a hydroxylamino group on the linker-drug moiety. Alternatively, the non-natural amino acid p-azidophenylalanine can be incorporated into an antibody to provide an azide functional group for conjugation via click chemistry, as discussed above. Non-natural amino acids can also be incorporated into an antibody or other polypeptide using cell-free methods, as taught in Goerke et al., US 2010/0093024 A1 (2010) and Goerke et al., Biotechnol. Bioeng. 2009, 102 (2), 400-416. The foregoing disclosures are incorporated herein by reference. Thus, in one embodiment, an antibody that is used for making a conjugate has one or more amino acids replaced by a non-natural amino acid, which preferably isp-acetylphenylalanine orp-azidophenylalanine, more preferably p-acetylphenylalanine.
  • Still another conjugation technique uses the enzyme transglutaminase (preferably bacterial transglutaminase from Streptomyces mobaraensis or BTG), per Jeger et al., Angew. Chem. Int. Ed. 2010, 49, 9995. BTG forms an amide bond between the side chain carboxamide of a glutamine (the amine acceptor) and an alkyleneamino group (the amine donor), which can be, for example, the 8-amino group of a lysine or a 5-amino-n-pentyl group. In a typical conjugation reaction, the glutamine residue is located on the antibody, while the alkyleneamino group is located on the linker-drug moiety, as shown below:
  • Figure US20230140047A1-20230504-C00124
  • The positioning of a glutamine residue on a polypeptide chain has a large effect on its susceptibility to BTG mediated transamidation. None of the glutamine residues on an antibody are normally BTG substrates. However, if the antibody is deglycosylated—the glycosylation site being asparagine 297 (N297; numbering per EU index as set forth in Kabat et al., “Sequences of proteins of immunological interest,” 5th ed., Pub. No. 91-3242, U.S. Dept. Health & Human Services, NIH, Bethesda, Md., 1991; hereinafter “Kabat”) of the heavy chain—nearby glutamine 295 (Q295) is rendered BTG susceptible. An antibody can be deglycosylated enzymatically by treatment with PNGase F (Peptide-N-Glycosidase F). Alternatively, an antibody can be synthesized glycoside free by introducing an N297A mutation in the constant region, to eliminate the N297 glycosylation site. Further, it has been shown that an N297Q substitution not only eliminates glycosylation, but also introduces a second glutamine residue (at position 297) that too is an amine acceptor. Thus, in one embodiment, the antibody is deglycosylated. In another embodiment, the antibody has an N297Q substitution. Those skilled in the art will appreciate that deglycosylation by post-synthesis modification or by introducing an N297A mutation generates two BTG-reactive glutamine residues per antibody (one per heavy chain, at position 295), while an antibody with an N297Q substitution will have four BTG-reactive glutamine residues (two per heavy chain, at positions 295 and 297).
  • An antibody can also be rendered susceptible to BTG-mediated conjugation by introducing into it a glutamine containing peptide, or “tag,” as taught, for example, in Pons et al., US 2013/0230543 A1 (2013) and Rao-Naik et al., WO 2016/144608 A1.
  • In a complementary approach, the substrate specificity of BTG can be altered by varying its amino acid sequence, such that it becomes capable of reacting with glutamine 295 in an unmodified antibody, as taught in Rao-Naik et al., WO 2017/059158 A1 (2017).
  • While the most commonly available bacterial transglutaminase is that from S. mobaraensis, transglutaminase from other bacteria, having somewhat different substrate specificities, can be considered, such as transglutaminase from Streptoverticillium ladakanum (Hu et al., US 2009/0318349 A1 (2009), US 2010/0099610 A1 (2010), and US 2010/0087371 A1 (2010)).
  • TLR7 agonists of this disclosure having a primary or secondary alkyl amine are particularly suitable for use in conjugates, as the secondary amine provides a functional group for attachment of the linker. An example of such a TLR7 agonist-linker compound is compound 41, which contains an enzymatically cleavable linker. FIG. 15 shows a scheme according to which compound 41 can be prepared.
  • Figure US20230140047A1-20230504-C00125
  • An example of a TLR7 agonist-linker compound that contains a non-enzymatically cleavable linker is compound 43. FIG. 16 shows a pathway for synthesizing compound 43.
  • Figure US20230140047A1-20230504-C00126
  • Both compounds 41 and 43 contain a primary alkylamino group, rendering them amenable to conjugation with transglutaminase. A suitable conjugation procedure is described in the Examples hereinbelow.
  • Conjugation can also be effected using the enzyme Sortase A, as taught in Levary et al., PLoS One 2011, 6(4), e18342; Proft, Biotechnol. Lett. 2010, 32, 1-10; Ploegh et al., WO 2010/087994 A2 (2010); and Mao et al., WO 2005/051976 A2 (2005). The Sortase A recognition motif (typically LPXTG, where X is any natural amino acid) may be located on the ligand Z and the nucleophilic acceptor motif (typically GGG) may be the group R31 in formula (III), or vice-versa.
  • TLR7 Agonist Conjugates
  • Applying the fore-described techniques, TLR7 agonist conjugates such as the ones shown below can be prepared:
  • Figure US20230140047A1-20230504-C00127
  • where m is 1, 2, 3, or 4 and Ab is an antibody.
  • Pegylation
  • Attachment of a poly(ethylene glycol) (PEG) chain to a drug (“PEGylation”) can improve the latter's pharmacokinetic properties. The circulation half-life of the drug is increased, sometimes by over an order of magnitude, concomitantly reducing the dosage needed to achieve a desired therapeutic effect. PEGylation can also decrease metabolic degradation of a drug and reduce its immunogenicity. For a review, see Kolate et al., J. Controlled Release 2014, 192, 167.
  • Initially, PEGylation was applied to biologic drugs. As of 2016, over ten PEGylated biologics had been approved. Turecek et al., J. Pharmaceutical Sci. 2016, 105, 460. More recently, stimulated by the successful application of the concept to biologics, attention has turned towards its application to small molecule drugs. In addition to the aforementioned benefits, PEGylated small molecule drugs may have increased solubility and cause fewer toxic effects. Li et al. Prog. Polymer Sci. 2013, 38, 421.
  • The compounds disclosed herein can be PEGylated. Where a compound has an aliphatic primary or secondary amine or an aliphatic hydroxyl, such as the case of the compounds shown below (arrows), it can be PEGylated via an ester, amide, carbonate, or carbamate group with a carboxy-containing PEG molecule utilizing conventional techniques such as dicyclohexylcarbodiimide, HATU, N-hydroxysuccinimide esters, and the like. Various other methods for PEGylating pharmaceutical molecules are disclosed in Alconcel et al., Polymer Chem. 2011, 2, 1442, the disclosure of which is incorporated herein by reference.
  • Figure US20230140047A1-20230504-C00128
  • If desired, a TLR7 agonist disclosed herein can be PEGylated via an enzymatically cleavable linker comprising a self-immolating moiety, to allow release of the un-PEGylated agonist in a designed manner. Further, PEGylation can be combined with conjugation to a protein such as an antibody, if the PEG-containing molecule has a suitable functional group such as an amine for attachment to the protein. The protein can provide an additional therapeutic function or, if an antibody, can provide a targeting function. These concepts are illustrated in the following reaction sequence, where TLR7-NH-R generically represents a TLR7 agonist:
  • Figure US20230140047A1-20230504-C00129
  • In the above reaction sequence, the valine-citrulline (Val-Cit) dipeptide is cleavable by the enzyme cathepsin B, with a p-aminobenzyl oxycarbonyl (PABC) group serving as a self-immolating spacer. The functional group for conjugation is an amine group, which is temporarily protected by an Fmoc group. Conjugation is effected by the enzyme transglutaminase, with a glutamine (Gln) side chain acting as the acyl acceptor. The subscript x, denoting the number of PEG repeat units, can vary widely, depending on the purpose of the PEGylation, as discussed below. For some purposes, x can be relatively small, such as 2, 4, 8, 12, or 24. For other purposes, x is large, for example between about 45 and about 910.
  • Those skilled in the art will understand that the sequence is illustrative and that other elements—peptide, self-immolating group, conjugation method, PEG length, etc.—may be employed, as is well known in the art. They will also understand that, while the above sequence combines PEGylation and conjugation, PEGylation does not require conjugation, and vice-versa.
  • Where the compound lacks aliphatic hydroxyl or aliphatic primary or secondary amine, it still can be PEGylated at the aromatic amine on the pyrimidine ring. A method for PEGylating at this position is disclosed by Zarraga, US 2017/0166384 A1 (2007), the disclosure of which is incorporated by reference.
  • In some embodiments, it may be desirable to have multiple PEGylated agonists linked in a single molecule. For instance, four PEGylated arms can be constructed on pentaerythritol (C(CH2OH)4) and a TLR7 agonist can be attached to each PEGylated arm. See Gao et al., US 2013/0028857 A1 (2013), the disclosure of which is incorporated by reference.
  • For modulating pharmacokinetics, it is generally preferred that the PEG moiety have a formula weight of between about 2 kDa (corresponding to about 45 —(CH2CH2O)— repeating units) and between about 40 kDa (corresponding to about 910 —(CH2CH2O)— repeating units), more preferably between about 5 kDa and about 20 kDa. That is, the range of the subscript x in the above formulae is from about 45 to about 910. It is to be understood that PEG compositions are not 100% homogeneous but, rather, exhibit a distribution of molecular weights. Thus, a reference to, for example, “20 kDa PEG” means PEG having an average molecular weight of 20 kDa.
  • PEGylation can also be used for improving the solubility of an agonist. In such instances a shorter PEG chain can be used, for example comprising 2, 4, 8, 12, or 24 repeating units.
  • Pharmaceutical Compositions and Administration
  • In another aspect, there is provided a pharmaceutical composition comprising a compound of as disclosed herein, or of a conjugate thereof, formulated together with a pharmaceutically acceptable carrier or excipient. It may optionally contain one or more additional pharmaceutically active ingredients, such as a biologic or a small molecule drug. The pharmaceutical compositions can be administered in a combination therapy with another therapeutic agent, especially an anti-cancer agent.
  • The pharmaceutical composition may comprise one or more excipients. Excipients that may be used include carriers, surface active agents, thickening or emulsifying agents, solid binders, dispersion or suspension aids, solubilizers, colorants, flavoring agents, coatings, disintegrating agents, lubricants, sweeteners, preservatives, isotonic agents, and combinations thereof. The selection and use of suitable excipients is taught in Gennaro, ed., Remington: The Science and Practice of Pharmacy, 20th Ed. (Lippincott Williams & Wilkins 2003).
  • Preferably, a pharmaceutical composition is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion). Depending on the route of administration, the active compound may be coated in a material to protect it from the action of acids and other natural conditions that may inactivate it. The phrase “parenteral administration” means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. Alternatively, the pharmaceutical composition can be administered via a non-parenteral route, such as a topical, epidermal or mucosal route of administration, for example, intranasally, orally, vaginally, rectally, sublingually or topically.
  • Pharmaceutical compositions can be in the form of sterile aqueous solutions or dispersions. They can also be formulated in a microemulsion, liposome, or other ordered structure suitable to achieve high drug concentration. The compositions can also be provided in the form of lyophilates, for reconstitution in water prior to administration.
  • The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated and the particular mode of administration and will generally be that amount of the composition which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.01 percent to about ninety-nine percent of active ingredient, preferably from about 0.1 percent to about 70 percent, most preferably from about 1 percent to about 30 percent of active ingredient in combination with a pharmaceutically acceptable carrier.
  • Dosage regimens are adjusted to provide a therapeutic response. For example, a single bolus may be administered, several divided doses may be administered over time, or the dose may be proportionally reduced or increased as indicated by the exigencies of the situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. “Dosage unit form” refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic response, in association with the required pharmaceutical carrier.
  • The dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight. For example dosages can be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg, or alternatively 0.1 to 5 mg/kg. Exemplary treatment regimens are administration once per week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 months, or once every three to 6 months. Preferred dosage regimens include 1 mg/kg body weight or 3 mg/kg body weight via intravenous administration, using one of the following dosing schedules: (i) every four weeks for six dosages, then every three months; (ii) every three weeks; (iii) 3 mg/kg body weight once followed by 1 mg/kg body weight every three weeks. In some methods, dosage is adjusted to achieve a plasma antibody concentration of about 1-1000 μg/mL and in some methods about 25-300 μg/mL.
  • A “therapeutically effective amount” of a compound of the invention preferably results in a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction. For example, for the treatment of tumor-bearing subjects, a “therapeutically effective amount” preferably inhibits tumor growth by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects. A therapeutically effective amount of a therapeutic compound can decrease tumor size, or otherwise ameliorate symptoms in a subject, which is typically a human but can be another mammal. Where two or more therapeutic agents are administered in a combination treatment, “therapeutically effective amount” refers to the efficacy of the combination as a whole, and not each agent individually.
  • The pharmaceutical composition can be a controlled or sustained release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, poly-glycolic acid, collagen, polyorthoesters, and polylactic acid. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • Therapeutic compositions can be administered via medical devices such as (1) needleless hypodermic injection devices; (2) micro-infusion pumps; (3) transdermal devices; (4) infusion devices; and (5) osmotic devices.
  • In certain embodiments, the pharmaceutical composition can be formulated to ensure proper distribution in vivo. For example, to ensure that the therapeutic compounds of the invention cross the blood-brain barrier, they can be formulated in liposomes, which may additionally comprise targeting moieties to enhance selective transport to specific cells or organs.
  • INDUSTRIAL APPLICABILITY
  • TLR7 agonist compounds disclosed herein can be used for the treatment of a disease or condition that can be ameliorated by activation of TLR7.
  • In one embodiment, the TLR7 agonist is used in combination with an anti-cancer immunotherapy agent—also known as an immuno-oncology agent. An anti-cancer immunotherapy agent works by stimulating a body's immune system to attack and destroy cancer cells, especially through the activation of T cells. The immune system has numerous checkpoint (regulatory) molecules, to help maintain a balance between its attacking legitimate target cells and preventing it from attacking healthy, normal cells. Some are stimulators (up-regulators), meaning that their engagement promotes T cell activation and enhances the immune response. Others are inhibitors (down-regulators or brakes), meaning that their engagement inhibits T cell activation and abates the immune response. Binding of an agonistic immunotherapy agent to a stimulatory checkpoint molecule can lead to the latter's activation and an enhanced immune response against cancer cells. Reciprocally, binding of an antagonistic immunotherapy agent to an inhibitory checkpoint molecule can prevent down-regulation of the immune system by the latter and help maintain a vigorous response against cancer cells. Examples of stimulatory checkpoint molecules are B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, CD40, ICOS-L, OX40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H. Examples of inhibitory checkpoint molecules are CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, CD96 and TIM-4.
  • Whichever the mode of action of an anti-cancer immunotherapy agent, its effectiveness can be increased by a general up-regulation of the immune system, such as by the activation of TLR7. Thus, in one embodiment, this specification provides a method of treating a cancer, comprising administering to a patient suffering from such cancer a therapeutically effective combination of an anti-cancer immunotherapy agent and a TLR7 agonist as disclosed herein. The timing of administration can be simultaneous, sequential, or alternating. The mode of administration can systemic or local. The TLR7 agonist can be delivered in a targeted manner, via a conjugate.
  • Cancers that could be treated by a combination treatment as described above include acute myeloid leukemia, adrenocortical carcinoma, Kaposi sarcoma, lymphoma, anal cancer, appendix cancer, teratoid/rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain cancer, breast cancer, bronchial tumor, carcinoid tumor, cardiac tumor, cervical cancer, chordoma, chronic lymphocytic leukemia, chronic myeloproliferative neoplasm, colon cancer, colorectal cancer, craniopharyngioma, bile duct cancer, endometrial cancer, ependymoma, esophageal cancer, esthesioneuroblastoma, Ewing sarcoma, eye cancer, fallopian tube cancer, gallbladder cancer, gastrointestinal carcinoid tumor, gastrointestinal stromal tumor, germ cell tumor, hairy cell leukemia, head and neck cancer, heart cancer, liver cancer, hypopharngeal cancer, pancreatic cancer, kidney cancer, laryngeal cancer, chronic myelogenous leukemia, lip and oral cavity cancer, lung cancer, melanoma, Merkel cell carcinoma, mesothelioma, mouth cancer, oral cancer, osteosarcoma, ovarian cancer, penile cancer, pharyngeal cancer, prostate cancer, rectal cancer, salivary gland cancer, skin cancer, small intestine cancer, soft tissue sarcoma, testicular cancer, throat cancer, thyroid cancer, urethral cancer, uterine cancer, vaginal cancer, and vulvar cancer.
  • Anti-cancer immunotherapy agents that can be used in combination therapies as disclosed herein include: AMG 557, AMP-224, atezolizumab, avelumab, BMS 936559, cemiplimab, CP-870893, dacetuzumab, durvalumab, enoblituzumab, galiximab, IMP321, ipilimumab, lucatumumab, MEDI-570, MEDI-6383, MEDI-6469, muromonab-CD3, nivolumab, pembrolizumab, pidilizumab, spartalizumab, tremelimumab, urelumab, utomilumab, varlilumab, vonlerolizumab. Table A below lists their alternative name(s) (brand name, former name, research code, or synonym) and the respective target checkpoint molecule.
  • TABLE A
    Immunotherapy Agent Alternative Name (s) Target
    AMG 557 B7RP-1 (ICOSL)
    AMP-224 PD-1
    Atezolizumab MPDL3280A, RO5541267, PD-L1
    TECENTRIQ ®
    Avelumab BAVENCIO ® PD-L1
    BMS 936559 PD-L1
    Cemiplimab LIBTAYO ® PD-1
    CP-870893 CD40
    Dacetuzumab CD40
    Durvalumab IMFINZI ® PD-L1
    Enoblituzumab MGA271 B7-H3
    Galiximab B7-1 (CD80)
    IMP321 LAG-3
    Ipilimumab YERVOY ® CTLA-4
    Lucatumumab CD40
    MEDI-570 ICOS (CD278)
    MEDI-6383 OX40
    MEDI-6469 OX40
    Muromonab-CD3 CD3
    Nivolumab OPDIVO ® PD-1
    Pembrolizumab KEYTRUDA ® PD-1
    Pidilizumab MDV9300 PD-1
    Spartalizumab PDR001 PD-1
    Tremelimumab Ticilimumab, CP-675, CTLA-4
    CP-675,206
    Urelumab BMS-663513 CD137
    Utomilumab PF-05082566 CD137
    Varlilumab CDX 1127 CD27
    Vonlerolizumab RG7888, MOXR0916, OX40
    pogalizumab
  • In one embodiment of a combination treatment with a TLR7 agonist, the anti-cancer immunotherapy agent is an antagonistic anti-CTLA-4, anti-PD-1, or anti-PD-L1 antibody. The cancer can be lung cancer (including non-small cell lung cancer), pancreatic cancer, kidney cancer, head and neck cancer, lymphoma (including Hodgkin's lymphoma), skin cancer (including melanoma and Merkel skin cancer), urothelial cancer (including bladder cancer), gastric cancer, hepatocellular cancer, or colorectal cancer.
  • In another embodiment of a combination treatment with a TLR7 agonist, the anti-cancer immunotherapy agent is an antagonistic anti-CTLA-4 antibody, preferably ipilimumab.
  • In another embodiment of a combination treatment with a TLR7 agonist, the anti-cancer immunotherapy agent is an antagonistic anti-PD-1 antibody, preferably nivolumab or pembrolizumab.
  • The TLR7 agonists disclosed herein also are useful as vaccine adjuvants.
  • BIOLOGICAL ACTIVITY
  • The biological activity of compounds disclosed herein as TLR7 agonists can be assayed by the procedures following.
  • Human TLR7 Agonist Activity Assay
  • This procedure describes a method for assaying human TLR7 (hTLR7) agonist activity of the compounds disclosed in this specification.
  • Engineered human embryonic kidney blue cells (HEK-Blue™ TLR cells; Invivogen) possessing a human TLR7-secreted embryonic alkaline phosphatase (SEAP) reporter transgene were suspended in a non-selective, culture medium (DMEM high-glucose (Invitrogen), supplemented with 10% fetal bovine serum (Sigma)). HEK-Blue™ TLR7 cells were added to each well of a 384-well tissue-culture plate (15,000 cells per well) and incubated 16-18 h at 37° C., 5% CO2. Compounds (100 nl) were dispensed into wells containing the HEK-Blue™ TLR cells and the treated cells were incubated at 37° C., 5% CO2. After 18 h treatment ten microliters of freshly-prepared Quanti-Blue™ reagent (Invivogen) was added to each well, incubated for 30 min (37° C., 5% CO2) and SEAP levels measured using an Envision plate reader (OD=620 nm). The half maximal effective concentration values (EC50; compound concentration which induced a response halfway between the assay baseline and maximum) were calculated.
  • Induction of Type I Interferon Genes (MX-1) and CD69 in Human Blood
  • The induction of Type I interferon (IFN) MX-1 genes and the B-cell activation marker CD69 are downstream events that occur upon activation of the TLR7 pathway. The following is a human whole blood assay that measures their induction in response to a TLR7 agonist.
  • Heparinized human whole blood was harvested from human subjects and treated with test TLR7 agonist compounds at 1 mM. The blood was diluted with RPMI 1640 media and Echo was used to predot 10 nL per well giving a final concentration of 1 uM (10 nL in 10 uL of blood). After mixing on a shaker for 30 sec, the plates were covered and placed in a 37° C. chamber for o/n=17 hrs. Fixing/lysis buffer was prepared (5×→1× in H2O, warm at 37° C.; Cat #BD 558049) and kept the perm buffer (on ice) for later use.
  • For surface markers staining (CD69): prepared surface Abs: 0.045 ul hCD14-FITC (ThermoFisher Cat #MHCD1401)+0.6 ul hCD19-ef450 (ThermoFisher Cat #48-0198-42)+1.5 ul hCD69-PE (cat #BD555531)+0.855 ul FACS buffer. Added 3 ul/well, spin1000 rpm for 1 min and mixed on shaker for 30 sec, put on ice for 30 mins. Stop stimulation after 30 minutes with 70 uL of prewarmed 1× fix/lysis buffer and use Feliex mate to resuspend (15 times, change tips for each plate) and incubate at 37 C for 10 minutes.
  • Centrifuge at 2000 rpm for 5 minutes aspirate with HCS plate washer, mix on shaker for 30 sec and then wash with 70 uL in dPBS and pelleted 2×s (2000 rpm for 5 min) and 50 ul wash in FACS buffer pelleted 1xs(2000 rpm for 5 min). Mix on shaker for 30 sec. For Intracellular markers staining (MX-1): Add 50 ul of BD Perm buffer III and mix on shaker for 30 sec. Incubate on ice for 30 minutes (in the dark). Wash with 50 uL of FACS buffer 2× (spin @2300 rpm×5 min after perm) followed by mixing on shaker for 30 sec. Resuspended in 20 ul of FACS buffer containing MX1 antibody ( )(4812)-Alexa 647: Novus Biologicals #NBP2-43704AF647) 20 ul FACS bf+0.8 ul hIgG+0.04 ul MX-1. Spin 1000 rpm for 1 min, mix on shaker for 30se and the samples were incubated at RT in the dark for 45 minutes followed by washing 2×FACS buffer (spin @2300 rpm×5 min after perm). Resuspend 20 ul (35 uL total per well) of FACS buffer and cover with foil and place in 4° C. to read the following day. Plates were read on iQuePlus. The results were loaded into toolset and IC50 curves are generated in curve master. The y-axis 100% is set to 1 uM of resiquimod.
  • Induction of TNF-Alpha and Type I IFN Response Genes in Mouse Blood
  • The induction of TNF-alpha and Type I IFN response genes are downstream events that occur upon activation of the TLR7 pathway. The following is an assay that measures their induction in whole mouse blood in response to a TLR7 agonist.
  • Heparinized mouse whole blood was diluted with RPMI 1640 media with Pen-Strep in the ratio of 5:4 (50 uL whole blood and 40 uL of media). A volume of 90 uL of the diluted blood was transferred to wells of Falcon flat bottom 96-well tissue culture plates, and the plates were incubated at 4° C. for 1 h. Test compounds in 100% DMSO stocks were diluted 20-fold in the same media for concentration response assays, and then 10 uL of the diluted test compounds were added to the wells, so that the final DMSO concentration was 0.5%. Control wells received 10 uL media containing 5% DMSO. The plates were then incubated at 37° C. in a 5% CO2 incubator for 17 h. Following the incubation, 100 uL of the culture medium as added to each well. The plates were centrifuged and 130 uL of supernatant was removed for use in assays of TNFa production by ELISA (Invitrogen, Catalog Number 88-7324 by Thermo-Fisher Scientific). A 70 uL volume of mRNA catcher lysis buffer (1×) with DTT from the Invitrogen mRNA Catcher Plus kit (Cat #K1570-02) was added to the remaining 70 uL sample in the well, and was mixed by pipetting up and down 5 times. The plate was then shaken at room temperature for 5-10 min, followed by addition of 2 uL of proteinase K (20 mg/mL) to each well. Plates were then shaken for 15-20 min at RT. The plates were then stored at −80° C. until further processing.
  • The frozen samples were thawed and mRNA was extracted using the Invitrogen mRNA Catcher Plus kit (Cat #K1570-02) according to the manufacturer's instructions. Half yield of mRNA from RNA extraction were used to synthesize cDNA in 20 μL reverse transcriptase reactions using Invitrogen SuperScript IV VILO Master Mix (Cat #11756500). TaqMan® real-time PCR was performed using QuantStudio Real-Time PCR system from ThermoFisher (Applied Biosystems). All real-time PCR reactions were run in duplicate using commercial predesigned TaqMan assays for mouse IFIT1, IFIT3, MX1 and PPIA gene expression and TaqMan Master Mix. PPIA was utilized as the housekeeping gene. The recommendations from the manufacturer were followed. All raw data (Ct) were normalized by average housekeeping gene (Ct) and then the comparative Ct (ΔΔCt) method were utilized to quantify relative gene expression (RQ) for experimental analysis.
  • General Procedures
  • The following general procedures were used for liquid chromatography (preparative or analytical) and nuclear magnetic resonance.
  • Liquid Chromatography
  • Unless noted otherwise, the following general conditions were used for high pressure liquid chromatography (HPLC) purification or for liquid chromatography-mass spectrometry (LC-MS):
      • Preparative HPLC/MS conditions A-1: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 0.05% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile:water with 0.05% trifluoroacetic acid; Gradient: a 0-minute hold at 0% B, 0-40% B over 20 minutes, then a 0-minute hold at 100% B; Flow Rate: 20 mL/min; Column Temperature: 25° C.
      • Preparative HPLC/MS conditions A-2: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 10 mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate; Gradient: a 0-minute hold at 7% B, 7-47% B over 20 minutes, then a 0-minute hold at 100% B; Flow Rate: 20 mL/min; Column Temperature: 25° C.
      • Preparative HPLC/MS conditions A-3: Column: XBridge Shield RP18 OBD Column, 19×150 mm, 5 μm; Mobile phase A: water with 0.05% TFA; Mobile phase B acetonitrile with 0.05% TFA; Flow rate: 18.9 mL/min; Gradient: 0% B for 2 min, 0-35% B for 20 min, 100% B for 3 min; Detector, UV 254/210 nm; Column Temperature: 25° C.
      • LC/MS conditions B: Column: Aquity UPLC BEH C18, 2.1 mm×50 mm, 1.7 μm particles; Mobile Phase A: 100% water with 0.05% TFA; Mobile Phase B: 100% acetonitrile with 0.05% TFA; Gradient: 2% B to 98% B over 1 min, then a 0.50 min hold at 98% B; Flow: 0.8 mL/min.
      • LC/MS conditions C: Column: Waters XBridge C18, 2.1 mm×50 mm, 1.7 μm particles; Mobile Phase A: 5:95 acetonitrile:water with 0.1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile:water with 0.1% trifluoroacetic acid; Temperature: 50° C.; Gradient: 0% B to 100% B over 3 min, then a 0.50 min hold at 100% B; Flow: 1 mL/min; Detection: MS and UV (220 nm).
      • LC/MS conditions D: LC/MS conditions: Column: Aquity UPLC BEH C18, 2.1 mm×50 mm, 1.7 μm particles; Mobile Phase A: 95:5 water:acetonitrile with 10 mM NH4OH; Mobile Phase B: 95:5 acetonitrile:water with 10 mM NH4OH; Gradient: 5% B to 95% B over 1 min, then a 0.50 min hold at 95% B; Flow: 0.8 mL/min to 95% B over 1 min, then a 0.50 min hold at 95% B; Flow: 0.8 mL/min.
      • LC/MS conditions E: LC/MS conditions: Column: Waters XBridge C18, 2.1 mm×50 mm, 1.7 μm particles; Mobile Phase A: water with 0.1% TFA; Mobile Phase B: acetonitrile with 0.1% TFA; Temperature: 40° C.; Gradient: 5% B to 95% B over 2.6 min, then a 0.20 min hold at 95% B; Flow rate: 0.6 mL/min;
    NMR
  • The following conditions were used for obtaining proton nuclear magnetic resonance (NMR) spectra: NMR spectra were taken in either 400 Mz or 500 Mhz Bruker instrument using either DMSO-d6 or CDCl3 as solvent and internal standard. The crude NMR data was analyzed by using either ACD Spectrus version 2015-01 by ADC Labs or MestReNova software.
  • Chemical shifts are reported in parts per million (ppm) downfield from internal tetramethylsilane (TMS) or from the position of TMS inferred by the deuterated NMR solvent. Apparent multiplicities are reported as: singlet-s, doublet-d, triplet-t, quartet-q, or multiplet-m. Peaks that exhibit broadening are further denoted as br. Integrations are approximate. It should be noted that integration intensities, peak shapes, chemical shifts and coupling constants can be dependent on solvent, concentration, temperature, pH, and other factors. Further, peaks that overlap with or exchange with water or solvent peaks in the NMR spectrum may not provide reliable integration intensities. In some cases, NMR spectra may be obtained using water peak suppression, which may result in overlapping peaks not being visible or having altered shape and/or integration.
  • Synthesis
  • The practice of this invention can be further understood by reference to the following examples, which are provided by way of illustration and not of limitation.
  • Generally, the procedures disclosed herein produce a mixture of regioisomers, alkylated at the 1H or 2H position of the pyrazolopyrimidine ring system (which are also referred to as N1 and N2 regioisomers, respectively, alluding to the nitrogen that is alkylated). In the figures, sometimes the N2 regioisomers are not shown for convenience, but it is to be understood that they are present in the initial product mixture and separated at a later time, for example by preparative HPLC.
  • Figure US20230140047A1-20230504-C00130
  • The mixture of regioisomers can be separated at an early stage of the synthesis and the remaining synthetic steps carried out with the 1H regioisomer or, alternatively, the synthesis can be progressed carrying the mixture of regioisomers and separation effected at a later stage, as desired.
  • Procedure 1—Compounds Per FIG. 1
  • This procedure and companion FIG. 1 illustrate a method for making compounds disclosed herein, using compound 800 as an exemplar.
  • Compound 3. Methyl 4-amino-1H-pyrazole-5-carboxylate 2 (4 g, 28.3 mmol) and methyl (Z)-4-(2,3-bis(methoxycarbonyl)guanidino)-1H-pyrazole-5-carboxylate 1 in methanol (50 mL) was treated with acetic acid (8.11 mL, 142 mmol) at which time a precipitate formed. The reaction mixture was stirred overnight. Sodium methoxide (64.8 mL, 283 mmol) was added and stirring was continued overnight. LCMS showed completion of the reaction. The pH was adjusted to 5 by the slow addition of acetic acid, whereby a precipitate formed that was washed with water and then acetonitrile and dried to provide 5.2 g of compound 3 as an off white solid. LCMS ESI: calculated for C7H7N5O3=210.16 (M+H+), found 210.0 (M+H+).
  • Compound 4. Compound 3 (2 g, 9.56 mmol), butan-1-amine (1.8 mL, 9 mmol), and DBU (1.6 mL, 10 mmol) in DMSO (10 mL) was slowly treated with BOP (5 g, 11 mmol). The reaction mixture was heated at 60° C. for 2 h at which time LCMS showed completion of the reaction. The reaction was directly purified on reverse phase COMBIFLASH™ apparatus using 80 g C-18 column eluting with 0-100% acetonitrile/water (0.1% formic acid) to yield compound 4 as a white solid. LCMS ESI: calculated for C11H16N6O2=265.28 (M+H+), found 265.2 (M+H+). 1H NMR (400 MHz, dmso-d6) δ 8.02 (s, 1H), 3.97 (s, 3H), 1.74-1.66 (m, 2H), 1.49-1.38 (m, 2H), 1.25 (s, 1H), 0.95 (t, J=7.4 Hz, 3H).
  • Compound 6. A mixture of methyl 4-(bromomethyl)-2-methoxybenzoate 5 (1 g, 3.86 mmol) and cyclobutanamine 5a (0.659 mL, 7.72 mmol) in DMF (2 mL) was heated at 70° C. over 30 min at which point LCMS showed the formation of an amine product. The excess base was evaporated and Hunig's Base (1.348 mL, 7.72 mmol) was added, followed by addition of Boc-anhydride (0.896 mL, 3.86 mmol). LCMS showed the completion of reaction. The solvent was evaporated and the crude product was purified by COMBIFLASH™ apparatus using EtOAc/hexanes to provide 0.82 g desired product 6 as a colorless oil. LCMS ESI: calculated for C19H27NO5=350.42 (M+H+), found 350.1 (M+H+).
  • Compound 7. A solution of compound 6 (0.82 g, 2.347 mmol) in THF (5 mL) at 0° C. was treated slowly with LiAlH4 (2 M in THF, 1.173 mL, 2.347 mmol) and stirred for 30 min, at which point LCMS showed completion of the reaction. The reaction was quenched by the slow addition of methanol and stirred with Rochelle salt solution for 2 h. The organic layers were separated and the crude product 7 was purified on a COMBIFLASH™ apparatus using EtOAc/hexanes, silica gel column. LCMS ESI: calculated for C18H27NO4=322.41 (M+H+), found 322.1 (M+H+).
  • Compounds 8 and 9. A mixture of compound 4 (100 mg, 0.378 mmol), compound 7 (182 mg, 0.568 mmol) and triphenylphosphine (248 mg, 0.946 mmol) in THF (3 mL) was slowly treated with DIAD (0.110 mL, 0.568 mmol) over 5 min and stirred at RT for 30 min under N2 at which point LCMS showed the completion of the reaction. The solvent was evaporated and the crude product was purified on reverse phase COMBIFLASH™ apparatus using 80 g C-18 column eluting with 0-100% acetonitrile/water (1 mM TEAA) to provide a mixture of compounds 8 and 9 as a white solid. LCMS ESI: calculated for C29H41N7O5=566.69 (M−H+), found 566.3 (M−H+).
  • The isomers were separated by chiral supercritical fluid chromatography using Column: Kromasil 5-CelluCoat, 21×250 mm, 5 micron, Mobile Phase: 15% MeOH-DEA/85%% CO2, Flow Conditions: 45 mL/min, 150 Bar, 40° C., Detector Wavelength: 230 nm, Injection Details: 0.5 mL of −25 mg/mL in MeOH to provide 17 mg of compound 8 and 25 mg of compound 9.
      • Analytical data for compound 8: 1H NMR (400 MHz, DMSO-d6) δ 9.61 (s, 2H), 7.86 (s, 2H), 6.94 (s, 2H), 6.83 (s, 2H), 6.63 (d, J=7.9 Hz, 2H), 6.51 (d, J=7.5 Hz, 2H), 5.69 (s, 4H), 4.39 (s, 4H), 3.79 (s, 6H), 3.63 (s, 6H), 3.48 (d, J=6.2 Hz, 4H), 3.33 (s, 20H), 3.18 (d, J=5.3 Hz, 1H), 2.05-1.95 (m, 8H), 1.53 (t, J=7.5 Hz, 7H), 1.35 (s, 11H), 1.23 (q, J=7.2 Hz, 6H), 0.86 (t, J=7.4 Hz, 6H).
      • Analytical data for compound 9: 1H NMR (400 MHz, DMSO-d6) δ 9.37 (s, 1H), 8.08 (s, 1H), 6.90 (s, 1H), 6.84 (s, 1H), 6.71 (d, J=7.7 Hz, 1H), 5.48 (s, 2H), 4.42 (s, 3H), 3.78 (d, J=18.0 Hz, 4H), 3.69 (s, 1H), 3.61 (s, 3H), 3.51-3.42 (m, 3H), 2.06-1.97 (m, 6H), 1.61-1.47 (m, 6H), 1.36 (s, 8H), 1.34-1.26 (m, 6H), 0.99 (t, J=7.1 Hz, 3H), 0.94-0.85 (m, 4H).
  • Compound 800. A solution of compound 8 (13 mg, 0.023 mmol) was dissolved in THF (0.5 mL) and was treated with TFA (0.018 mL, 0.229 mmol). LCMS in 30 min showed Boc deprotection. The TFA was evaporated and this mixture was treated with sodium hydroxide (9.16 mg, 0.229 mmol) and heated at 60° C. for 2 h, at which point LCMS showed completion of the reaction. The base was neutralized by the slow addition of 6M HCl and worked up with EtOAc/water. The crude material was purified via preparative HPLC under HPLC/MS conditions A-2.
  • Other compounds of this disclosure can be analogously made, mutatis mutandis, by using another amine instead of cyclobutanamine 5a.
  • Procedure 2—Compounds Per FIGS. 2A-2B
  • This procedure and companion FIGS. 2A-2B illustrate another method for making compounds disclosed herein, using compounds 801 and 818 as exemplars.
  • Compounds 12 and 13. A solution of methyl 4-nitro-1H-pyrazole-5-carboxylate 10 (3.27 g, 19.11 mmol) in DMF (20 mL) was treated with K2CO3 (2.90 g, 21.02 mmol) and methyl 4-(bromomethyl)-3-methoxybenzoate 11 (5 g, 19.30 mmol). The reaction was started at 0° C. and allowed to proceed for 1 h, at which point LCMS showed completion of the reaction with ˜1:5 mixture of products. The base was filtered and the reaction was diluted with EtOAc and washed with water 2 times. The solvent was evaporated and the crude product was taken to next step as-is. LCMS ESI: calculated for C15H15N3O7=350.2 (M−H+), found 350.0 (M−H+).
  • For characterization purpose, a small amount of the mixture of products was separated using silica gel column chromatography using 0-50% EtOAc/hexanes.
      • Analytical data for compound 12: 1H NMR (400 MHz, DMSO-d6) δ 8.40 (s, 1H), 7.57 (dd, J=7.8, 1.5 Hz, 1H), 7.50 (d, J=1.6 Hz, 1H), 7.27 (d, J=7.9 Hz, 1H), 5.53 (s, 2H), 3.96 (s, 3H), 3.84 (d, J=16.2 Hz, 6H).
      • Analytical data for compound 13: 1H NMR (400 MHz, DMSO-d6) δ 9.05 (s, 1H), 7.62-7.51 (m, 2H), 7.28 (d, J=7.9 Hz, 1H), 5.47 (s, 2H), 3.87 (s, 8H), 3.31 (s, 1H).
  • Compounds 14 and 15. A solution of compounds 12 and 13 (2 g, 5.73 mmol), zinc and ammonium formate was stirred at RT for 2 h, after which LCMS showed completion of the reaction. Filtration and concentration yielded a crude mixture of compounds 14 and 15. LCMS ESI: calculated for C15H17N3O5=320.3 (M+H+), found 320.2 (M+H+).
  • Compounds 16 and 17 A mixture of compounds 14 and 15 (1.830 g, 5.73 mmol) and compound 1 in MeOH (20 mL) was treated with acetic acid (1.640 mL, 28.7 mmol) and stirred overnight. The solution was treated with sodium methoxide (13.11 mL, 57.3 mmol) and stirred overnight. LCMS showed conversion to the product. The pH was adjusted to 5 and the resulting precipitate was washed with water. The residue was dried to afford a mixture of compounds 16 and 17. LCMS ESI: calculated for C17H17N5O6=388.3 (M+H+), found 388.1 (M+H+).
  • Compounds 18 and 19. A mixture of compounds 16 and 17 (1 g, 2.58 mmol) in DMSO (10 mL) was treated with butan-1-amine (0.510 mL, 5.16 mmol), DBU (0.428 mL, 2.84 mmol) followed slowly by BOP (1.370 g, 3.10 mmol). The reaction was heated at 70° C. for 2 h, at which point LCMS showed completion of the reaction. The reaction was diluted with water and extracted with EtOAc. The combined organic phases were dried over Na2SO4 and taken as-is to the next step. LCMS ESI: calculated for C21H26N6O5=443.4 (M+H+), found 443.2 (M+H+).
  • 1H NMR (400 MHz, DMSO-d6) δ 9.41 (s, 1H), 8.20 (s, 1H), 8.08 (s, 1H), 7.57-7.42 (m, 3H), 6.93 (d, J=8.1 Hz, 1H), 5.80 (s, 1H), 5.60 (s, 2H), 4.04 (q, J=7.1 Hz, 1H), 3.95-3.82 (m, 10H), 3.62 (d, J=6.1 Hz, 4H), 3.45 (q, J=7.0 Hz, 3H), 2.68 (d, J=9.9 Hz, 1H), 2.57-2.50 (m, 6H), 2.00 (s, 1H), 1.59 (p, J=7.3 Hz, 3H), 1.54-1.48 (m, 1H), 1.39-1.26 (m, 3H), 1.18 (t, J=7.1 Hz, 2H), 0.86 (dt, J=29.5, 7.3 Hz, 5H).
  • Compound 818. A solution of compounds 18 and 19 (1.142 g, 2.58 mmol) in THF (2.58 mL, 5.16 mmol) at 0° C. was treated with LiAlH4 (THF, 2.58 mL, 5.16 mmol) and stirred for 1 h, after which LCMS showed completion of the reaction. The reaction was quenched with MeOH and stirred with Rochelle salt solution overnight. The product was extracted with EtOAc and taken to next step as a mixture of crude compounds reduced intermediate. LCMS ESI: calculated for C20H26N6O4=415.4 (M+H+), found 415.2 (M+H+).
  • A mixture of the reduced intermediates (1069 mg, 2.58 mmol) in 1,4-dioxane (10 mL) was treated with aqueous sodium hydroxide (2.58 mL, 25.8 mmol) and heated at 80° C. for 5 h, after which LCMS showed formation of product. The base was neutralized with 6M HCl and the solvent was evaporated. The residue was taken up in 5 mL DMF and syringe filtered. The solvent was evaporated to afford a 3:1 mixture of compounds 818 and its regioisomer 19a.
  • Compound 801 and 19b. A solution of compounds 818 and 19a (420 mg, 1.178 mmol) in THF (1 mL) was treated with thionyl chloride (0.172 mL, 2.357 mmol) and stirred for 30 min, after which LCMS showed completion of the reaction. The solvent was evaporated and the crude benzyl chloride intermediate was taken to next step as-is. LCMS ESI: calculated for C18H23ClN6O=375.8 (M+H+), found 375.2 (M+H+).
  • A mixture of the preceding crude product mixture (20 mg, 0.053 mmol) and tetrahydro-2H-pyran-4-amine 22 (5.40 mg, 0.053 mmol) in DMF (1 mL) was heated at 70° C. for 1 h, after which LCMS showed completion of the reaction. The reaction was syringe filtered and the crude products were purified and separated via preparative LC/MS with the following conditions: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 0.1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile:water with 0.10% trifluoroacetic acid; Gradient: a 2-minute hold at 6% B, 6-27% B over 25 minutes, then a 2-minute hold at 100% B; Flow Rate: 20 mL/min; Column Temperature: 25 C. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation.
  • If desired, the separation of 1H and 2H regioisomers can be effected earlier in the synthetic process, for example at the mixture of compounds 12 and 13 and synthetic steps carried out only with the 1H regioisomer.
  • Other compounds of this disclosure can be analogously prepared, mutatis mutandis, for example by using another amine instead of tetrahydro-2H-pyran-4-amine 22.
  • Procedure 3—Compounds Per FIGS. 3A-3B
  • This procedure and companion FIGS. 3A-3B illustrate another method for making compounds disclosed herein, using compounds 844, 817, and 861 as exemplars.
  • Compound 24. To a mixture of compound 16 (400 mg, 1.033 mmol) and (S)-3-aminohexan-1-ol 23 (242 mg, 2.065 mmol) in DMSO (5 mL) was added DBU (0.463 mL, 3.10 mmol) followed by the slow addition of BOP (502 mg, 1.136 mmol). The reaction mixture was heated at 70° C. for 2 h, at which point LCMS showed completion of the reaction. The reaction mixture was diluted with EtOAc and washed with water. The organic layer was concentrated to leave a crude product, which was used in the next step.
  • The crude product was treated with tert-butylchlorodiphenylsilane (0.295 mL, 1.136 mmol) and imidazole (141 mg, 2.065 mmol) in DMF (5 mL). The reaction mixture was stirred overnight. The reaction mixture was diluted with water and worked up with EtOAc. Purification was performed with a COMBIFLASH™ apparatus using EtOAc/hexane to afford compound 24. LCMS ESI: calculated for C38H48N6O6Si=725.9 (M+H+), found 725.3 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 9.53 (s, 1H), 7.94 (s, 1H), 7.57-7.50 (m, 2H), 7.50-7.30 (m, 7H), 7.25-7.14 (m, 3H), 6.31 (d, J=7.9 Hz, 1H), 6.24 (d, J=8.4 Hz, 1H), 5.78 (d, J=17.2 Hz, 2H), 4.56 (s, 1H), 3.92-3.82 (m, 4H), 3.78 (s, 3H), 3.59 (s, 3H), 3.55-3.43 (m, 2H), 3.31 (s, 2H), 1.86-1.68 (m, 2H), 1.47 (q, J=8.7, 7.9 Hz, 2H), 1.18 (t, J=7.1 Hz, 5H), 0.91 (s, 9H).
  • Compound 25. A solution of compound 24 (408 mg, 0.563 mmol) in THF (5 mL) was treated slowly with LiAlH4 (0.563 mL, 1.126 mmol) and the reaction was stirred at 0° C. for 1 h, at which point LCMS showed completion of the reaction. The reaction was quenched with MeOH and stirred with Rochelle salt solution overnight. Extraction with EtOAc and purification with a COMBIFLASH™ apparatus, 0-50% MeOH in DCM gradient afforded compound 25. LCMS ESI: calculated for C38H48N6O5Si: 697.9 (M+H+), found 679.4 (M+H+).
  • Compound 26. A solution of compound 25 (150 mg, 0.215 mmol) in THF (0.5 mL) was treated slowly with thionyl chloride (0.031 mL, 0.430 mmol) and stirred for 1 h. LCMS showed completion of the reaction. The solvent was evaporated and the crude product 26 was taken to next step. LCMS ESI: calculated for C38H47ClN6O4Si=715.3 (M+H+), found 715.4 (M+H+).
  • Compound 844. Compound 26 (15 mg) was dissolved in DMF (0.5 mL) and treated with 1,2,3,4-tetrahydro-2,6-naphthyridine 27 (5.78 mg, 0.043 mmol) and heated at 70° C. for 2 h after which LCMS showed completion of the reaction. A solution of the intermediate product (0.018 g, 0.022 mmol) was dissolved in dioxane (0.5 mL) and treated with triethylamine trihydrofluoride (0.018 mL, 0.110 mmol). After stirring at RT overnight, LCMS showed removal of the TBDPS protecting group. Neutralization to pH 7, evaporation of solvent and purification on a reverse phase ISCO using TEAA as modifier afforded the deprotected intermediate product.
  • The deprotected intermediate product from above was dissolved in dioxane (0.5 mL), treated with sodium hydroxide (0.220 mL, 0.220 mmol) and heated at 80° C. for 2 h. LCMS showed completion of the reaction. The reaction mixture was neutralized to pH 7 with aqueous HCl and the solvent was evaporated. The residue was dissolved in DMF and syringe-filtered and purified by preparative LC/MS using following conditions: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 10-mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate; Gradient: a 0-minute hold at 15% B, 15-55% B over 20 minutes, then a 4-minute hold at 100% B; Flow Rate: 20 mL/min; Column Temperature: 25° C. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to yield compound 844 (4.7 mg, 40% yield).
  • Compound 817 Compound 25 was treated with trimethylamine trihydrofluoride and then sodium hydroxide, generally following the above procedure, to afford compound 817.
  • FIG. 3B shows a variation of the scheme of FIG. 3A, in which the steps subsequent to the preparation of compound 26 are varied. The scheme of FIG. 3B is illustrated by particular reference to compound 861.
  • Compound 26b. Compound 26 (22 mg, 0.031 mmol) was dissolved in DMF (0.6 mL) at RT and was treated with DIEA (27 μL, 0.15 mmol) and 3-methoxyazetidine (26a) as its hydrochloride (11 mg, 0.093 mmol). After stirring for 2 h at 70° C., the reaction mixture was concentrated in vacuo to give compound 26b, which was used without further purification. LC/MS conditions B: LC RT: 0.88 min. LCMS (M+H)=766.9.
  • Compound 861. A solution of compound 26a (24 mg, 0.031 mmol)) in dioxane (0.3 mL) at RT was treated with NaOH (10M aq. Solution, 0.1 mL, 1.0 mmol) and the reaction was heated at 70° C. for 60 min, before the addition of another portion of NaOH (10M aq. solution, 0.1 mL, 1.0 mmol). After 2 h, the reaction mixture concentrated in vacuo. The residue was treated with MeOH (0.3 mL) and HCl (37% aq. solution, 0.3 mL, 3.7 mmol), stirred at RT for 30 min and concentrated. The crude product was dissolved in DMF, filtered through a PTFE frit, and purified via preparative HPLC conditions A-1. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to give compound 861 as its TFA salt (8.4 mg, 46%). LC/MS conditions C: LC RT: 0.82 min. LCMS (M+H)=470.3.
  • Other compounds of this disclosure can be analogously prepared, mutatis mutandis, for example by using amines other than those shown in FIGS. 3A-3B.
  • Procedure 4—Compounds Per FIGS. 4A-4B
  • This procedure and companion FIGS. 4A-4B illustrate another method of making compounds disclosed herein.
  • Compound 30. A suspension of compound 4 (400 mg, 1.513 mmol) in dioxane (5 mL) was treated with sodium hydroxide (10 N in water, 1.513 mL, 15.13 mmol) and stirred at 60° C. for 45 min. The reaction mixture was concentrated. The crude product was dissolved into water and purified by reverse phase chromatography on a COMBIFLASH™ unit using a 150 g C-18 column eluting with 10 mM TEAA in acetonitrile: 10 mM in water, 0-70% gradient. The desired fractions were frozen and lyophilized to yield compound 30 (150 mg, 0.727 mmol, 48.1% yield). LCMS ESI: calculated for C9H15N6=207.1 (M+H+), found 207.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 7.56 (br s, 1H), 5.53 (br s, 2H), 3.43 (br d, J=6.2 Hz, 2H), 1.57 (t, J=7.2 Hz, 2H), 1.44-1.29 (m, 2H), 0.95-0.76 (m, 3H).
  • Compound 33. A suspension of 6-fluoronicotinaldehyde 31 (1.809 g, 14.46 mmol), methyl 4-hydroxybenzoate 32 (2 g, 13.15 mmol), and K2CO3 (1.998 g, 14.46 mmol) in DMF (26.3 ml) was stirred at 110° C. for 4 h. LCMS indicated the reaction was complete. Upon cooling, the reaction was quenched with water. The resulting solid was collected by filtration and rinsed with water and dried in vacuo to yield compound 33 (3.30 g, 12.84 mmol, 95.1% yield). LCMS ESI: calculated for C14H11NO4=258.1 (M+H+), found 258.0 (M+H+). 1H NMR (400 MHz, CHLOROFORM-d) δ 10.01 (s, 1H), 8.63 (d, J=2.4 Hz, 1H), 8.23 (dd, J=8.6, 2.4 Hz, 1H), 8.17-7.97 (m, 2H), 7.27-7.22 (m, 2H), 7.10 (d, J=8.6 Hz, 1H), 3.93 (s, 3H).
  • Compound 34. A solution of compound 33 (3.76 g, 14.62 mmol) in MeOH (100 ml) was treated with NaBH4 (0.553 g, 14.62 mmol) portionwise at 0° C. and then stirred for 10 min with continued cooling. LCMS indicated the reaction was complete. Reaction was quenched by slowly adding half saturated NH4Cl. Stirring was continued for 30 min at RT. The reaction mixture was extracted with ethyl acetate. The organic extracts were dried over Na2SO4, filtered, and concentrated. The crude solid was slurried into water and collected by filtration and dried in vacuo to yield compound 34 (3.37 g, 13.00 mmol, 89% yield). LCMS ESI: calculated for C14H13NO4=260.1 (M+H+), found 260.0 (M+H+). 1H NMR (400 MHz, CHLOROFORM-d) δ 8.21 (d, J=2.2 Hz, 1H), 8.12-8.04 (m, 2H), 7.81 (dd, J=8.4, 2.4 Hz, 1H), 7.21-7.13 (m, 2H), 6.99 (d, J=8.4 Hz, 1H), 4.71 (s, 2H), 3.91 (s, 3H).
  • Compound 35. Compound 34 (7.9 g, 30.5 mmol) in DCM (75 mL) was treated with MsCl (2.61 mL, 33.5 mmol) at 0° C. After stirring at RT for 16 h, the reaction was done. The reaction was quenched with water. After extraction with DCM (3×20 mL), the combined organic extracts were dried over Na2SO4, filtered and concentrated. The crude product was purified on an ISCO silica column (80 g), eluting with ethyl acetate:hexanes, 0-70% gradient. The desired fractions were concentrated to yield compound 35 (7.47 g, 26.9 mmol, 88% yield). LCMS ESI: calculated for C14H13ClNO3=278.1 (M+H+), found 278.0 (M+H+). 1H NMR (400 MHz, CHLOROFORM-d) δ 8.19 (d, J=2.2 Hz, 1H), 8.13-8.02 (m, 2H), 7.79 (dd, J=8.5, 2.5 Hz, 1H), 7.22-7.14 (m, 2H), 6.99 (d, J=8.6 Hz, 1H), 4.57 (s, 2H), 3.92 (s, 3H).
  • Compounds 36 and 37. Compound 30 (70 mg, 0.339 mmol) in DMF (1 mL) was treated with cesium carbonate (332 mg, 1.018 mmol), followed by compound 35 (94 mg, 0.339 mmol). After stirring for 5 h at RT, the reaction was complete. After quenching with water and extraction with ethyl acetate (3×10 mL), the combined organic extracts were dried over Na2SO4, filtered and concentrated The crude product was purified on an ISCO silica column (24 g), eluted with 20% MeOH in DCM:DCM, 0-60% gradient. The desired fractions were concentrated to yield a mixture of compounds 36 and 37 (120 mg, 0.080 mmol, 79% yield), in a 1:4 ratio. LCMS ESI: calculated for C23H26N7O3=448.2 (M+H+), found 448.3 (M+H+).
  • Compounds 37a and 37b. A mixture of compounds 36 and 37 (60 mg, 0.114 mmol) in THF (2 mL) was treated with LiAlH4 (1.0 M in THF, 0.22 mL, 0.22 mmol) slowly at 0° C. under N2. The reaction mixture was stirred at 0° C. for 1 h, at which point LCMS showed completion of the reaction. Reaction was quenched by adding Na2SO4.10 H2O slowly, followed by MeOH and stirring at RT for 3 h. The solid was filtered off. The filtrate was concentrated. The residue dissolved in DMF and the products were purified via preparative LC/MS with the following conditions: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 0.1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile:water with 0.1% trifluoroacetic acid; Gradient: a 0-minute hold at 10% B, 10-45% B over 20 minutes, then a 4-minute hold at 100% B; Flow Rate: 20 mL/min. Fraction collection was triggered by MS and UV signals. Fractions containing desired product were combined and dried via centrifugal evaporation.
  • Compounds 38a and 38b. A mixture of compound 37a and compound 37b (40 mg, 0.095 mmol) in THF (1 mL) was treated with thionyl chloride (0.14 mL, 1.9 mmol). After stirring at RT for 3 h, LCMS showed completion of the reaction. The solvent was evaporated and the excess thionyl chloride was azeotropically removed with DCM. The crude chloride material was directly carried over to next step without further purification. LCMS ESI: calculated for C22H25N7O=438.2 (M+H+), found 438.1 (M+H+).
  • A mixture of the preceding chlorides (40 mg, 0.091 mmol) from preceding paragraph was dissolved in DMF (1.0 mL) and treated with 2-aminoethan-1-ol (55.0 μl, 0.91 mmol), followed by stirring at RT for 16 h. LCMS showed completion of the reaction. The mixture was purified via preparative LC/MS with the following conditions: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 10 mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate; Gradient: a 0-minute hold at 8% B, 8-48% B over 25 minutes, then a 4-min hold at 100% B; Flow Rate: 20 mL/min; Column Temperature: 25° C. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation.
  • Replacing 6-fluoronicotinaldehyde 31 with 4-fluoro-2-methoxybenzaldehyde in the scheme of FIGS. 4A-4B and generally following the procedures above, the other compounds of this disclosure can be prepared, for example: 802, 803, 808, 809, and 810.
  • Procedure 5—Compounds Per FIG. 5
  • This procedure and companion FIG. 5 relate to the synthesis of (S)—N-(3-aminohexyl)acetamide 54, which can be used as an intermediate for the synthesis of compounds disclosed herein.
  • Compound 51. A solution of (S)-3-aminohexan-1-ol 50 (1 g, 8.53 mmol) in DCM (10 mL) was treated with Hunig's base (4.47 mL, 25.6 mmol) and Boc-anhydride (2.377 mL, 10.24 mmol) and stirred for 3 h, after which LCMS showed completion of the reaction. The solvent and base were stripped off in vacuo. The crude product was purified on an ISCO machine with N2 detector using 80 g silica gel gold column with using 0-100% EtOAc/hexanes to provide fractions with the desired product, which were combined and concentrated to provide compound 51 (1.05 g, 56.6% yield). LCMS ESI: calculated for C11H23NO3=218.3 (M+H+), found 218.2 1H NMR (400 MHz, Chloroform-d) δ 3.76 (s, 1H), 3.63 (dd, J=7.6, 3.0 Hz, 2H), 1.90-1.77 (m, 1H), 1.46-1.44 (m, 10H), 1.44-1.17 (m, 4H), 1.00-0.81 (m, 5H).
  • Compound 52. To a solution of compound 51 (1050 mg, 4.83 mmol), triphenylphosphine (1648 mg, 6.28 mmol) and isoindoline-1,3-dione (924 mg, 6.28 mmol) in THF (10 mL) was slowly added DIAD (1.221 mL, 6.28 mmol). After stirring overnight at RT, LCMS showed completion of the reaction. Stripping off the solvent in a rotary evaporator and purification on an ISCO machine using 80 g silical gold column eluting with 0-50% EtOAc/hexanes provided fractions with the dsired product, which were concentrated to provide compound 52 (1.6 g, 96% yield) as a pale yellow solid. LCMS ESI: calculated for C19H26NO4=347.4 (M+H+), found 247.2 (M-Boc+H+). δ 7.83 (dd, J=5.4, 3.1 Hz, 2H), 7.70 (dd, J=5.5, 3.0 Hz, 2H), 4.40 (s, 1H), 3.75 (dd, J=8.4, 7.0 Hz, 2H), 3.64 (s, 1H), 1.69 (dq, J=15.8, 7.9 Hz, 1H), 1.52-1.44 (m, 2H), 1.42 (s, 9H), 1.39-1.28 (m, 3H), 0.91 (t, J=7.1 Hz, 3H).
  • Compound 53. Compound 52 (1.6 g, 4.62 mmol) was treated with methylamine in MeOH (15.59 g, 92 mmol). The reaction mixture was stirred for 1 h, after which LCMS showed phthalimide deprotection. The solvent and base were evaporated and the residue was dissolved in THF (5 mL) and treated with Hunig's base (1.613 mL, 9.24 mmol), followed by acetyl chloride (0.394 mL, 5.54 mmol). The reaction mixture was stirred for 1 h at RT and the solvent was evaporated. The crude product was purified on reverse phase ISCO using 80 g C-18 column eluting with 0-95% acetonitrile in water (0.05% formic acid) to provide fractions with the desired product, which were lyophilized to provide compound 53 as pale yellow paste (0.8 g). LCMS ESI: calculated for C13H26N2O3=259.3 (M+H+), found 159.1 (M-Boc+H+). δ 6.65 (s, 2H), 4.30 (d, J=9.4 Hz, 2H), 3.75-3.67 (m, 2H), 3.60 (s, 2H), 2.84 (t, J=12.7 Hz, 2H), 2.00 (s, 3H), 1.75 (dddd, J=14.1, 10.7, 5.3, 3.6 Hz, 1H), 1.44-1.24 (m, 9H), 0.91 (t, J=6.8 Hz, 3H).
  • Compound 54. Compound 53 (0.8 g, 3.10 mmol) was treated with TFA (4.7 mL, 61.9 mmol). LCMS after 30 min shows completion of reaction. TFA was evaporated. The residue was dried in a lyophilizer overnight to give compound 54 (322 mg, 65.7% yield) as pale yellow paste. LCMS ESI: calculated for C8H18N2O=159.2 (M+H+), found 159.1 (M+H+). δ 8.08 (s, 2H), 6.71 (s, 1H), 3.49 (s, 1H), 3.38-3.32 (m, 2H), 3.30 (s, 1H), 3.18 (d, J=9.7 Hz, 1H), 2.04 (s, 3H), 1.96-1.83 (m, 2H), 1.67 (ddq, J=36.7, 14.4, 7.5, 7.0 Hz, 1H), 1.40 (dq, J=15.6, 7.7 Hz, 1H), 0.94 (t, J=7.3 Hz, 3H).
  • Compound 54 can be used to make compounds such as 846 and 847, generally following the Procedure 3 and FIGS. 3A-3B, using compound 16.
  • Procedure 6—Compounds Per FIGS. 6 a -6B
  • This procedure and companion FIGS. 6A-6B illustrate another method of making compounds disclosed herein, using compound 857 as an exemplar.
  • Compound 55. To a suspension of compound 30 (1.50 g, 7.27 mmol) in DMF (25 mL) was added NBS (1.29 g, 7.27 mmol) in three portions (color changed from red to light yellow). After 10 min stirring at RT, LCMS indicated that the reaction was complete. The mixture was poured onto a small portion of ice and stirred for 16 h. The resulting solid was collected by filtration and dried in high vacuum to yield compound 55 (1.96 g, 6.9 mmol, 95% yield).
  • Compound 57 A suspension of 4-fluoro-2-methoxybenzaldehyde 56 (1.5 g, 9.73 mmol), methyl 4-hydroxybenzoate 32 (1.777 g, 11.68 mmol) and K2CO3 (2.69 g, 19.46 mmol) in DMF (19.46 ml) was stirred at 105° C. for three days, LCMS indicated that reaction was complete. It was quenched with water. The resulting precipitate of a creamy colored solid was collected by filtration and dried in vacuo to yield compound 57 (2.6 g, 9.08 mmol, 93% yield). LCMS ESI: calculated for C16H15O5=287.1 (M+H+), found 287.1 (M+H+−18).
  • Compound 58. To a stirred suspension of compound 57 (2.6 g, 9.08 mmol) in MeOH (25 mL) was added NaBH4 (0.344 g, 9.08 mmol) in portions. The reaction mixture turned clear. After 1 h, the reaction was complete. The reaction mixture was concentrated to half-volume. Water was added, followed by stirring for 30 min. The resulting solid was collected by filtration and air dried to yield compound 58 (2.50 g, 8.67 mmol, 95% yield) as a white solid, which was pure enough for use in the next step. LCMS ESI: calculated for C16H17O5=289.1 (M+H+), found 271.1 (M+H+−18).
  • Compound 59. Compound 58 (730 mg, 2.53 mmol) in THF (10 mL) was treated, with stirring, with SOCl2 (0.924 mL, 12.66 mmol). Stirring was continued at RT for 16 h. Concentration on a rotary evaporator yielded compound 59. The extra SOCl2 was removed azeotropically with DCM (3×). The resulting solid was pure enough to carry over to next step without further purification. 1H NMR (400 MHz, DMSO-d6) δ 8.07-7.83 (m, 2H), 7.46 (d, J=8.1 Hz, 1H), 7.16-7.01 (m, 2H), 6.87 (d, J=2.2 Hz, 1H), 6.66 (dd, J=8.3, 2.3 Hz, 1H), 4.73 (s, 2H), 3.84 (d, J=5.3 Hz, 6H).
  • Compound 60. To a stirred mixture of compound 55 (300 mg, 1.052 mmol) in DMF (1 mL) was added compound 59 (323 mg, 1.052 mmol). After stirring at RT for 3 h, no starting material 59 was detected. The reaction mixture was quenched with brine, extracted with DCM (3×20 mL). The combined organic extracts were dried with Na2SO4 and filtered. The filtrate was concentrated. The crude mixture was purified by ISCO silica column (40 g), eluting with EtOAc/hexanes=0-100%. The desired fractions were concentrated to yield compound 60 (166 mg, 0.299 mmol, 28.4% yield). LCMS ESI: calculated for C22H28BrN6O4=555.1, 557.1 (M+H+), found 555.2, 557.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 7.96 (d, J=8.8 Hz, 2H), 7.55-7.55 (m, 1H), 7.12-6.98 (m, 2H), 6.87 (d, J=2.2 Hz, 1H), 6.76 (t, J=5.4 Hz, 1H), 6.71 (d, J=8.1 Hz, 1H), 6.62 (dd, J=8.3, 2.3 Hz, 1H), 5.97 (s, 2H), 5.62 (s, 2H), 3.83 (s, 3H), 3.79 (s, 3H), 3.50-3.39 (m, 2H), 1.59-1.46 (m, 2H), 1.31-1.21 (m, 2H), 0.87 (t, J=7.4 Hz, 3H).
  • Compound 61. To a stirred mixture of compound 60 (160 mg, 0.288 mmol) in THF (10 mL) was added LiAlH4 (2.5M in THF) (10.93 mg, 0.288 mmol) under nitrogen at RT. After 10 mins, LCMS indicated that reaction was complete. The reaction mixture was quenched with Rochelle salt solution carefully, and stirred at RT for 18 h. The organic layer was separated and the aqueous layer was further extracted with ethyl acetate (2×50 mL). The combined organic extracts were dried over Na2SO4 and filtered. The filtrate was concentrated to yield compound 61 (140 mg, 0.265 mmol, 92.2% yield). LCMS ESI: calculated for C24H28BrN6O3=527.1, 529.1 (M+H+), found 527.2, 529.2 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 7.32 (d, J=8.6 Hz, 2H), 6.96 (d, J=8.6 Hz, 2H), 6.77-6.65 (m, 3H), 6.43 (dd, J=8.3, 2.3 Hz, 1H), 5.95 (s, 2H), 5.57 (s, 2H), 5.15 (t, J=5.6 Hz, 1H), 4.47 (d, J=5.7 Hz, 2H), 3.76 (s, 3H), 3.45 (br d, J=5.7 Hz, 2H), 1.53 (t, J=7.2 Hz, 2H), 1.31-1.20 (m, 2H), 0.88 (t, J=7.4 Hz, 3H).
  • Compound 62. A suspension of compound 61 (165 mg, 0.313 mmol) in MeOH (10 mL) and ethyl acetate (3 mL) was purged with N2. Pd-C 5% (40 mg, 0.019 mmol) was added, purged with H2, and stirred under H2 balloon for 18 h. The catalyst was then removed by filtration through a CELITE® pad. The filtrate was concentrated to yield compound 62 (157 mg, 0.350 mmol, 112% yield). LCMS ESI: calculated for C24H29N6O3=449.2 (M+H+), found 449.3 (M+H+). 1H NMR (400 MHz, DMSO-d6) δ 8.13 (br d, J=4.0 Hz, 1H), 7.75 (s, 1H), 7.61 (br s, 2H), 7.33 (d, J=8.6 Hz, 2H), 6.97 (d, J=8.6 Hz, 2H), 6.83 (d, J=8.6 Hz, 1H), 6.74 (d, J=2.2 Hz, 1H), 6.42 (dd, J=8.3, 2.3 Hz, 1H), 5.68 (s, 2H), 5.26-5.03 (m, 1H), 4.48 (d, J=5.3 Hz, 2H), 3.73 (s, 3H), 3.58 (br d, J=6.2 Hz, 2H), 1.59 (br t, J=7.3 Hz, 2H), 1.35-1.18 (m, 2H), 0.90 (t, J=7.4 Hz, 3H).
  • Compound 857 A suspension of compound 62 (100 mg, 0.223 mmol) in THF (4 mL) was treated with SOCl2 (0.081 mL, 1.115 mmol), followed by stirring at RT for 1 h. The reaction mixture was then concentrated on a rotary evaporator. The excess SOCl2 was azeotropically removed with DCM (3×), the resulting solid was pure enough for use in the next step. LCMS ESI: calculated for C24H28ClN6O2=467.2 (M+H+), found 467.2 (M+H+). The resulting solid (20 mg, 0.043 mmol) was dissolved in DMF (0.5 mL). 1-Amino-2-methylpropan-2-ol (19.09 mg, 0.214 mmol) was added to the reaction mixture and the solution was stirred at RT for 16 hrs. The reaction mixture was syringe-filtered and the crude material was purified via preparative LC/MS with the following conditions: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 0.1% trifluoroacetic acid; Mobile Phase B: 95:5 acetonitrile:water with 0.1% trifluoroacetic acid; Gradient: a 0-minute hold at 19% B, 19-59% B over 20 minutes, then a 0-minute hold at 100% B; Flow Rate: 20 mL/min; Column Temperature: 25 C. Fraction collection was triggered by MS and UV signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to yield compound 857 as a TFA salt (6.9 mg, 10.63 μmol, 24.81% yield).
  • This Procedure 6 and prior Procedure 4 can be used to make similar compounds. We have observed that alkylation of 3-bromo compound 55 yields a higher proportion of the 1H regioisomer (2H regioisomer not shown in FIG. 6A) than the alkylation of counterpart compound 36 in Procedure 4. Thus, where the 1H regioisomer is the more desired one, this Procedure 6 may be the preferable one, even though it entails the extra bromination and de-bromination steps. Table B below lists some compounds that were made by either Procedure 4 or 6. Also, 3-bromo compound 55 can be an intermediate in other synthetic pathways, as shown in Procedures hereinbelow.
  • Procedure 7—Compounds Per FIG. 7
  • This Procedure and companion FIG. 7 illustrate another method for making compounds disclosed herein, using compound 864 as an exemplar.
  • Compound 64. A solution of compound 16 (48.5 mg, 0.125 mmol) in THF (1.8 mL) was cooled to 0° C. and treated with LiAlH4 (1M in THF, 219 μL, 0.219 mmol). After 10 min, an additional portion of LiAlH4 (1M in THF, 200 μL, 0.200 mmol) was added and the reaction was stirred for another 20 min at 0° C. The reaction mixture was quenched with MeOH and Rochelle's salt (sat. aq. solution) and stirred at RT for 30 min. The mixture was extracted with EtOAc (3×). The combined organic layers were washed with H2O, dried over Na2SO4, filtered and concentrated in vacuo to give compound 64 (38 mg, 84%)). 1H NMR (400 MHz, DMSO-d6) δ 11.60-11.12 (m, 2H), 7.85-7.83 (m, 1H), 6.97 (s, 1H), 6.77 (d, J=8.0 Hz, 1H), 6.59 (d, J=7.7 Hz, 1H), 5.66 (s, 2H), 5.15 (t, J=5.7 Hz, 1H), 4.44 (d, J=5.6 Hz, 2H), 3.79 (s, 3H), 3.74 (s, 3H). LC/MS conditions B: LC RT: 0.61 min. LCMS (M+H)=360.1.
  • Compound 65. A solution of compound 64 (402 mg, 1.12 mmol) in DCM (15 mL) was treated with thionyl chloride (1.6 mL, 22 mmol). After 15 min, the reaction mixture concentrated in vacuo. The residue was redissolved in DCM and concentrated in vacuo again to yield compound 65 (422 mg, 100%). 1H NMR (400 MHz, DMSO-d6) δ 11.25-11.02 (m, 1H), 7.89-7.88 (m, 1H), 7.11 (d, J=1.5 Hz, 1H), 6.92 (dd, J=7.8, 1.5 Hz, 1H), 6.62 (d, J=7.7 Hz, 1H), 5.69 (s, 2H), 4.72 (s, 2H), 3.82 (s, 3H), 3.75 (s, 3H). One proton was not visible, likely due to overlap with water peak or proton exchange. LC/MS conditions B: LC RT: 0.80 min. LCMS (M+H)=378.0.
  • Compound 66. A solution of cyclobutanamine 5a (0.84 mL, 9.9 mmol) in THF (11 mL) was treated a solution of compound 65 (249 mg, 0.659 mmol) in THF (8 mL). The reaction mixture was stirred for 16 h at 60° C. and then concentrated in vacuo. The residue was dissolved in DCM/MeOH, absorbed onto CELITE® and purified via column chromatography (50 g C18 gold column; Mobile Phase A: 10:90 methanol:water with 0.1% trifluoroacetic acid; Mobile Phase B: 90:10 methanol:water with 0.1% trifluoroacetic acid; Flow Rate: 40 mL/min) to give compound 66 as its TFA salt (168 mg, 48% yield). 1H NMR (400 MHz, DMSO-d6) δ 11.55 (br s, 1H), 11.18-11.12 (m, 1H), 9.04-8.93 (m, 2H), 7.91-7.89 (m, 1H), 7.19 (d, J=1.4 Hz, 1H), 6.95 (dd, J=7.7, 1.3 Hz, 1H), 6.68 (d, J=7.7 Hz, 1H), 5.71 (s, 2H), 4.02-3.97 (m, 2H), 3.85 (s, 3H), 3.76 (s, 3H), 3.72-3.63 (m, 1H), 2.20-2.10 (m, 4H), 1.85-1.72 (m, 2H). LC/MS conditions B: LC RT: 0.63 min. LCMS (M+H)=413.3.
  • Compound 864. A solution of compound 66 (15 mg, 0.036 mmol) in DMF (1 mL) was treated with BOP (161 mg, 0.364 mmol), 2-ethoxyethan-1-amine 6a (32.4 mg, 0.364 mmol) and DBU (73 μL, 0.49 mmol) and stirred at RT for 18 h. The reaction mixture was concentrated under a stream of nitrogen. A solution of the crude product 67 in dioxane (0.5 mL) was treated with NaOH (10 M aq soln, 0.05 mL, 0.5 mmol) and heated to 75° C. After 1 h, the reaction mixture was neutralized with HOAc (0.03 mL, 0.5 mmol) and concentrated under a stream of nitrogen. The residue was dissolved in DMF, filtered through a PTFE frit, and the crude material was purified via preparative LC/MS with the following conditions: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 10 mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate; Gradient: a 0-minute hold at 3% B, 3-43% B over 25 minutes, then a 0-minute hold at 100% B; Flow Rate: 20 mL/min; Column Temperature: 25° C. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to give compound 864 (7.3 mg, 48% yield). LC/MS conditions C. LC RT: 0.8 min. LCMS (M+H)=426.17.
  • Additional compounds according to this disclosure can be made following this Procedure, mutatis mutandis.
  • Procedure 8—Compounds Per FIG. 8
  • This Procedure and companion FIG. 8 illustrate another method for making compounds disclosed herein, using compound 866 as an exemplar.
  • Compound 70. A solution of compound 68 (307 mg, 0.922 mmol) in acetonitrile (0.9 mL) was treated with compound 69 (321 mg, 1.01 mmol) and stirred at RT for 72 h. The reaction mixture was then heated to 60° C. for 4 h, recooled to RT and filtered. The collected solid was rinsed with acetonitrile and dried in vacuo. The filtrate was concentrated in vacuo and the residue purified by column chromatography (12 g SiO2, 0 to 30% EtOAc-hexane, gradient elution) to give a white solid, which was combined with the solid isolated by filtration to give compound 70 (392 mg, 74%). LC/MS conditions B: LC RT: 1.21 min. LCMS (M+H)=576.9.
  • Compound 71. A solution of compound 70 (144 mg, 0.251 mmol) in MeOH (1.2 mL) was treated with NaOMe (81 mg, 1.5 mmol) and stirred at RT. After 96 h, the reaction mixture was diluted with H2O and extracted with EtOAc (3×). The combined organic layers were dried over Na2SO4, filtered and concentrated in vacuo to give the compound 71 (108 mg, 100% yield). 1H NMR (400 MHz, DMSO-d6) δ 11.62-11.59 (m, 1H), 10.85-10.82 (m, 1H), 7.91 (s, 1H), 7.51 (d, J=1.4 Hz, 1H), 7.47 (dd, J=7.9, 1.4 Hz, 1H), 6.69 (d, J=7.9 Hz, 1H), 5.75 (s, 2H), 3.89 (s, 3H), 3.84 (s, 3H), 1.50 (s, 9H). LC/MS conditions B: LC RT: 0.89 min. LCMS (M+H)=430.6.
  • Compound 72. A solution of compound 71 (294 mg, 0.685 mmol) in DMSO (3.4 mL) was treated with (S)-1-((tert-butyldiphenylsilyl)oxy)hexan-3-amine (71a) hydrochloride (322 mg, 0.822 mmol), BOP (364 mg, 0.822 mmol) and DBU (516 μL, 3.43 mmol) and stirred at RT. After 3 h, the reaction mixture diluted with EtOAc and washed with H2O. The organic layer concentrated in vacuo and the residue was purified via column chromatography (24 g SiO2; 0 to 25% EtOAc-DCM, gradient elution) to give compound 72 (227.8 mg, 43%). 1H NMR (400 MHz, DMSO-d6) δ 9.00-8.93 (m, 1H), 7.92 (s, 1H), 7.53 (dd, J=8.0, 1.4 Hz, 2H), 7.46 (s, 2H), 7.44 (d, J=1.3 Hz, 1H), 7.42-7.37 (m, 1H), 7.37-7.31 (m, 3H), 7.24-7.20 (m, 2H), 7.20-7.16 (m, 1H), 6.28 (d, J=7.9 Hz, 1H), 6.20 (d, J=8.5 Hz, 1H), 5.83-5.72 (m, 2H), 4.60-4.46 (m, 1H), 3.85 (s, 3H), 3.76 (s, 3H), 3.55-3.43 (m, 2H), 1.83-1.73 (m, 1H), 1.73-1.65 (m, 1H), 1.46 (br d, J=13.6 Hz, 2H), 1.41 (s, 9H), 1.15-1.04 (m, 2H), 0.91 (s, 9H), 0.74 (t, J=7.3 Hz, 3H). LC/MS conditions B: LC RT: 1.07 min. LCMS (M+H)=767.4.
  • Compound 73. A solution of methyl ester 72 (227 mg, 0.297 mmol) in THF (4.2 mL) was cooled to 0° C. and treated with LiAlH4 (1M in THF, 327 μL, 0.327 mmol). After 10 min, additional LiAlH4 (1M in THF, 150 μL, 0.150 mmol) added. After 30 min, the reaction was quenched with MeOH and Rochelle's salt (sat aq soln) and stirred at RT for 30 minutes. The mixture was extracted with EtOAc (3×). The combined organic layers were washed with H2O, and concentrated in vacuo. The residue was purified via column chromatography (12 g SiO2; 0 to 100% EtOAc-hexane, gradient elution) to give compound 73 (91.5 mg, 42%). LC/MS conditions B: LC RT: 1.02 min. LCMS (M+H)=739.7.
  • Compound 74. A solution of compound 73 (91.5 mg, 0.124 mmol) in THF (1.2 mL) was treated with thionyl chloride (45 μL, 0.61 mmol) and stirred at RT. After 15 min, the reaction mixture concentrated in vacuo, the residue was redissolved in DCM and concentrated in vacuo to provide compound 74 (94 mg, 100%). LC/MS conditions B: LC RT: 1.09 min. LCMS (M+H)=757.3.
  • Compound 75. A solution of compound 74 (20 mg, 0.027 mmol) in DMF (0.5 mL) was treated with DIEA (70 μL, 0.40 mmol) and 3-(tert-butyl)cyclobutan-1-amine 74a (34.1 mg, 0.268 mmol) and heated to 70° C. for 1 h. The reaction mixture was cooled to RT and concentrated in vacuo. The residue was dissolved in EtOAc and washed with sat. aq NaHCO3 soln. The organic layer was concentrated in vacuo to give afford compound 75 (22 mg, 97%), which was used without further purification. LC/MS conditions B: LC RT: 0.95 min. LCMS (M+H)=848.6.
  • Compound 866. A solution of compound 75 (25 mg, 0.029 mmol) in MeOH (0.6 mL) was treated with HCl (37% aq soln, 0.3 mL, 3.7 mmol) and stirred at RT for 6 h. The reaction mixture was neutralized with NaHCO3 and extracted with DCM (3×). The combined organic layers were concentrated in vacuo. The residue was redissolved in DMF and filtered through a PTFE filter. The crude product was purified via preparative HPLC/MS conditions A-1: Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to give compound 866 (3.3 mg, 22% yield). LC/MS conditions C: LC RT=1.23 min. LCMS (M+H)=510.23.
  • Additional compounds according to this disclosure can be made following this Procedure, mutatis mutandis.
  • Procedure 9—Compounds Per FIG. 9
  • This Procedure and companion FIG. 9 illustrate another method for making compounds disclosed herein, using compounds 853 and 870 as exemplars.
  • Compound 76. A solution of compound 55 (400 mg, 1.40 mmol) and cesium carbonate (914 mg, 2.8 mmol) in DMF/acetonitrile (5 mL/5 mL) was cooled in an ice bath. A solution of methyl 4-(bromomethyl)-3-fluorobenzoate 55a (277 mg, 1.12 mmol) in acetonitrile (5 mL) was added, and the reaction stirred at 0° C. for 30 min, then at RT for a further hour. The acetonitrile was evaporated from the reaction mixture and purified using reverse-phase flash chromatography (100 g column, 10 to 60% MeCN in water containing 0.05% TFA) to give compound 76 (482 mg, 1.07 mmol, 35% purity (contaminated with the N2-regioisomer, 27% yield) as a solid. LC-MS (ES, m/z): [M+H]+=451.1, 453.2.
  • Compound 77 Compound 76 (470 mg, 1.04 mmol, 35% purity (contaminated with the N2-regioisomer) was dissolved in ethanol (50 mL). 10% Pd/C (20 mg) was added, and the reaction evacuated and purged with hydrogen six times, then stirred under a hydrogen atmosphere for 90 min. The reaction mixture was filtered through CELITE®, washed with EtOH (100 mL), and evaporated to dryness, giving compound 77 (370 mg, 0.99 mmol, 35% purity (contaminated with the N2-regioisomer), 95% yield) as a solid. LC-MS (ES, m/z): [M+H]+=373.3.
  • Compound 78. A stirred suspension of compound 77 (370 mg, 0.99 mmol, 35% purity contaminated with the N2-regioisomer) in THF (100 mL) was cooled in an ice bath. LiAlH4 (754 mg, 1.98 mmol) was added portion wise over 5 min, and the reaction was stirred at 0° C. for 20 minutes. NaOH (1N, 50 mL) was added, and the reaction stirred for 10 min at RT, transferred to a separatory funnel and extracted with EtOAc (3×40 mL). The combined organic phases were washed with brine (3×30 mL), dried (MgSO4), filtered and concentrated. Flash chromatography (40 g column, 0 to 25% MeOH in DCM) yielded compound 78 (74 mg, 0.215 mmol, 21.63% yield) as a solid. LC-MS (ES, m/z): [M+H]+=345.3.
  • Compound 853. A 20 mL scintillation vial was charged with compound 78 (25 mg, 0.073 mmol) and THF (4 mL). 3 drops of thionyl chloride were added, and the reaction mixture was stirred for 1 hour. The reaction mixture was evaporated to dryness and the residue was dissolved in acetonitrile (5 mL) and evaporated twice. The residue was dissolved in DMF (1.5 mL). Cyclobutanamine (20.7 mg, 0.29 mmol) was added, and the reaction mixture stirred at RT overnight. The reaction mixture was filtered and purified via preparative HPLC/MS conditions A-1: Fraction collection was triggered by MS and UV signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to give compound 853 (19.5 mg, 0.03 mmol, 42% yield).
  • Compound 870. A 20 mL scintillation vial was charged with compound 78 (50 mg, 0.15 mmol) and THF (4 mL). 3 Drops of thionyl chloride were added, and the reaction mixture stirred for 1 h. The reaction mixture was evaporated to dryness. The residue was dissolved in acetonitrile (5 mL) and evaporated twice. The residue was dissolved in DMF (2 mL). 2-(piperazin-1-yl)ethan-1-ol (38 mg, 0.29 mmol) was added, followed by DIPEA (0.10 mL, 0.58 mmol), and the reaction mixture stirred at 50° C. for 1 h. After cooling, the reaction mixture was filtered and purified via preparative HPLC/MS conditions A-1: Fraction collection was triggered by MS and UV signals. Fractions containing the desired product were combined and dried via centrifugal evaporation, giving compound 870 (41 mg, 0.06 mmol, 42% yield).
  • Additional compounds according to this disclosure can be made following this Procedure, mutatis mutandis.
  • Procedure 10—Compounds Per FIG. 10
  • This Procedure and companion FIG. 10 illustrate another method for making compounds disclosed herein, using compound 873 as an exemplar.
  • Compound 80. A 40 mL scintillation vial was charged with compound 79 (250 mg, 1.55 mmol), NBS (331 mg, 1.86 mmol), AIBN (50.9 mg, 0.31 mmol) and CCl4 (5 mL). The reaction mixture was stirred at 70° C. for 2 h. After cooling, the reaction mixture was evaporated to dryness. Flash chromatography (24 g column, 0 to 40% EtOAc in hexanes) gave compound 80 (189 mg, 0.787 mmol, 51% yield) as a solid. LC-MS (ES, m/z): No ion detected. 1H NMR (400 MHz, CDCl3) δ 7.33 (d, J=7.7 Hz, 1H), 6.88 (d, J=7.7 Hz, 1H), 6.84 (s, 1H), 4.53 (s, 2H), 3.92 (s, 3H), 3.74 (s, 2H).
  • Compound 81. To a stirred suspension of methyl (7-(butylamino)-1H-pyrazolo[4,3-d]pyrimidin-5-yl)carbamate (6 g, 22.7 mmol) in DMF (10 mL) and was added a solution of NBS (4.44 g, 25.0 mmol) in acetonitrile (20 mL). The reaction mixture was stirred at RT for 1 h. Water (50 mL) was added. The reaction mixture filtered. The residue was washed with water (3×30 mL) and air dried overnight to yield compound 81 (5.112 g, 14.9 mmol, 66% yield) as a solid. LC-MS (ES, m/z): [M+H]+=343.0, 345.0. 1H NMR (400 MHz, DMSO-d6) δ 12.87 (br s, 1H), 9.80 (s, 1H), 7.56 (br s, 1H), 3.62 (s, 3H), 3.54 (q, J=6.6 Hz, 2H), 1.62 (quin, J=7.2 Hz, 2H), 1.40 (dq, J=14.8, 7.4 Hz, 2H), 0.94 (t, J=7.4 Hz, 3H).
  • Compound 82. Compound 81 (1 g, 2.91 mmol) was dissolved in DMF (10 mL) and cooled in an ice bath. Cesium carbonate (1.899 g, 5.83 mmol) was added, followed by compound 80 (0.560 g, 2.33 mmol). The reaction mixture was stirred at RT for 5 h and then poured into saturated NaHCO3 solution (50 mL) and extracted with EtOAc (3×40 mL). The combined organic phases were washed with brine (4×40 mL), dried (MgSO4), filtered and concentrated. Flash chromatography (40 g column, 0 to 85% EtOAc in hexane) gave compound 82 (219 mg, 0.44 mmol, 15% yield) as a solid. LC-MS (ES, m/z): [M+H]+ 502.2, 504.2. 1H NMR (400 MHz, CDCl3) δ 7.25-7.19 (m, 1H), 6.98 (s, 1H), 6.97-6.91 (m, 1H), 5.70 (s, 2H), 5.28 (s, 1H), 3.98 (s, 3H), 3.84 (s, 3H), 3.78-3.60 (m, 4H), 1.72-1.41 (m, 2H), 1.41-1.20 (m, 2H), 0.94 (t, J=7.3 Hz, 3H).
  • Compound 83. Compound 82 (219 mg, 0.44 mmol) was dissolved in EtOH (10 mL) and 10% Pd/C (20 mg) was added. The reaction vessel was evacuated and purged with H2 six times, then stirred under an H2 atmosphere for 2 h. The reaction mixture was filtered and evaporated to dryness, giving compound 83 (188 mg, 0.44 mmol, 100% yield) as a solid. LC-MS (ES, m/z): [M+H]+ 424.4. 1H NMR (400 MHz, DMSO-d6) δ 11.71 (br s, 1H), 8.67 (br s, 1H), 8.04 (s, 1H), 7.05 (s, 1H), 6.89 (s, 2H), 5.77 (s, 2H), 4.01 (s, 2H), 3.83 (s, 3H), 3.76 (s, 3H), 3.69-3.58 (m, 2H), 1.72-1.48 (m, 2H), 1.41-1.17 (m, 2H), 0.90 (t, J=7.4 Hz, 3H).
  • Compound 873. Compound 83 (25 mg, 0.059 mmol) was dissolved in dioxane (2 mL); sodium hydroxide (0.354 mL, 1.77 mmol) was added. The reaction mixture was stirred at 80° C. for 3 h. After cooling, the reaction was neutralized with 5N HCl, then evaporated to dryness. The residue was dissolved in DMF (2 mL), filtered and purified via preparative HPLC/MS conditions A-2: Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to give compound 873 (6.2 mg, 0.013 mmol, 22%).
  • Additional compounds according to this disclosure can be made following this Procedure, mutatis mutandis.
  • Procedure 11—Compounds Per FIG. 11
  • This Procedure and companion FIG. 11 illustrate another method for making compounds disclosed herein, using compound 855 as an exemplar.
  • Compound 85. To a solution of methyl 5-methoxy-6-methylnicotinate 84 (300 mg, 1656 μmol) and NBS (413 mg, 2318 μmol) in CCl4 (3 mL), AIBN (54.4 mg, 331 μmol) was added. The reaction mixture was stirred at 80° C. for 30 min. LCMS analysis showed the reaction was complete. The reaction mixture was cooled down and diluted with DCM (2 mL). The resulting solution was purified on a 40 g RediSepRf Gold Silica column. Mobile phase A: hexanes; Mobile phase B: Ethyl acetate; Gradient: 1 min at 0% B, 15 min at 0-50% B; Flow rate: 40 mL/min; Column Temperature: 25° C. Fraction collection was triggered by UV signals at 254 nm. The fractions containing expected product were combined, concentrated and dried under high vacuum to give compound 85 (215 mg, 50%). LC/MS conditions E: LC RT: 1.57 min. [M+H]+/Z=260.0
  • Compound 86. To a solution of compound 30 (427 mg, 2070 μmol) in DMF (2 mL), Cs2CO (701 mg, 2152 μmol) was added. The reaction mixture was stirred at RT for 10 min. A solution of compound 85 (215 mg, 0.827 mmol) in acetonitrile (2 mL) was added to the reaction mixture slowly. The reaction mixture was stirred at RT for 30 min. LCMS analysis showed 3 peaks corresponding product m/z. The reaction mixture was neutralized with acetic acid (0.3 mL), diluted with water and purified by preparative HPLC/MS conditions A-3: Fraction collection was triggered by UV signals. The fractions corresponding to the three peaks were freeze-dried separately. The NMR analysis confirmed that compound corresponding to the latest peak was the desired compound 86 (76 mg, 11.9%). LC/MS conditions E: LC-RT: 1.89 min. [M+H]+/Z=386.2. 1H NMR (500 MHz, DMSO-d6) δ 8.57 (t, J=5.7 Hz, 1H), 8.50 (d, J=1.6 Hz, 1H), 7.88 (d, J=1.6 Hz, 1H), 7.83 (s, 1H), 7.76 (s, 1H), 5.95 (s, 2H), 4.00 (s, 3H), 3.89 (s, 3H), 3.55 (q, J=6.8 Hz, 2H), 1.59-1.48 (m, 2H), 1.30-1.14 (m, 2H), 0.86 (t, J=7.4 Hz, 3H).
  • Compound 87 To a solution of compound 86 (38 mg, 0.099 mmol) in THF (1 mL), LiAlH4 in THF (0.079 mL, 0.197 mmol) was added. The reaction mixture was stirred at RT for 10 min. LCMS analysis showed the reaction was complete. The reaction mixture was neutralized with acetic acid (0.2 mL), diluted with water (3 mL) and purified on HPLC/MS conditions A-3: Fraction collection was triggered by UV signals. The fractions containing expected product were combined and freeze-dried to give compound 87 (28 mg, 79%). LC/MS conditions E: LC-RT: 1.26 min. [M+H]+/Z=357.2.
  • Compound 855. To a solution of compound 87 (28 mg, 0.078 mmol) in DCM (1 mL), SOCl2 (0.144 mL, 1.972 mmol) was added. The reaction mixture was stirred at RT overnight. The reaction mixture was concentrated and co-evaporated with DCM (3×20 mL) to form crude N7-butyl-1-((5-(chloromethyl)-3-methoxypyridin-2-yl)methyl)-1H-pyrazolo[4,3-d]pyrimidine-5,7-diamine (41 mg), which was used for next step immediately without purification.
  • To a solution of the preceding compound (41 mg, 0.109 mmol, crude) in DMF (1 mL), cyclobutanamine (388 mg, 5.45 mmol) was added. The reaction mixture was stirred at RT for 3 h. LCMS analysis showed the reaction was complete. The reaction mixture was freeze-dried with acetonitrile and water. The crude material was purified via preparative HPLC with the following conditions: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 10-mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10-mM ammonium acetate; Gradient: a 0-minute hold at 8% B, 8-48% B over 20 minutes, then a 0-minute hold at 100% B; Flow rate: 20 mL/min; Column Temperature: 25 C. Fraction collection was triggered by MS and UV signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to give 6.6 mg (14.8%) of compound 855 (6.6 mg, 14.8%). LC/MS conditions: Column: Waters Acquity BEH C18, 2.1 mm×50 mm, 1.7 μm particles; Mobile Phase A: 5:95 acetonitrile:water with 10 mM ammonium acetate; Mobile Phase B: 95:5 acetonitrile:water with 10 mM ammonium acetate; Temperature: 50° C.; Gradient: 0% B to 100% B over 3 min, then a 0.50 min hold at 100% B; Flow rate: 1 mL/min; Detection: MS and UV (220 nm). LC RT=1.36 min. (M+H)=411.1
  • Additional compounds according to this disclosure can be made following this Procedure, mutatis mutandis.
  • Procedure 12—Compounds Per FIG. 12
  • This Procedure and companion FIG. 12 illustrate another method for making compounds disclosed herein, using compound 867 as an exemplar.
  • Compound 90. To a solution of compound 89 (300 mg, 0.722 mmol) dissolved in DCM (3 mL) at −78° C. was added ethyl magnesium bromide (0.633 mL, 2.165 mmol). 2-methyltetrahydrofuran was added slowly to the reaction mixture and stirred at −78° C. for 1 hr and then 0° C. for 1 h. The reaction mixture was quenched with saturated ammonium chloride solution (1 mL) and brought to RT, then diluted with DCM (10 ml), washed with water (5 mL), then followed by brine solution (5 ml). The organic layer was dried over sodium sulphate, filtered, concentrated, purified on silicagel with ethyl acetate/hexane. The product containing fractions were concentrated to afford compound 90 as thick oil (204 mg, 63.4% yield). 1H NMR (400 MHz, Chloroform-d) δ 7.58 (m, 4H), 7.42-7.26 (m, 6H), 3.75-3.57 (m, 2H), 3.31 (d, J=7.8 Hz, 1H), 2.93 (d, J=7.5 Hz, 1H), 1.75 (m, 1H), 1.68-1.47 (m, 3H), 1.06 (s, 9H), 0.98 (s, 9H), 0.87 (t, J=7.4 Hz, 3H). LCMS: M/Z=446.2.
  • Compound 91. Hydrochloric acid (194 μL, 0.774 mmol, 4 M in dioxane) was added to a solution of compound 90 (203 mg, 0.455 mmol) in diethyl ether (4554 μL) and MeOH (55.3 μL, 1.366 mmol). The mixture was stirred at RT for 1 h. The solution was concentrated to a thick oil. 20 mL of hexane was then added to the oil and the flask was kept in the freezer at −20° C. for 1 h. The white solid was collected and washed with cold hexanes, then dried under under vacuum to give compound 91 (116 mg, 0.307 mmol, 67.4% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 7.85 (s, 3H), 7.63 (ddd, J=7.7, 3.0, 1.7 Hz, 4H), 7.56-7.40 (m, 6H), 3.92-3.65 (m, 2H), 3.18 (p, J=6.3 Hz, 1H), 1.86 (dq, J=13.0, 6.5 Hz, 1H), 1.76 (dq, J=13.4, 6.4 Hz, 1H), 1.64-1.47 (m, 2H), 1.01 (s, 9H), 0.89 (t, J=7.5 Hz, 3H). LCMS: M/Z=342.2.
  • Compound 867 A solution of compound 88 (30 mg, 0.068 mmol) in DMSO (0.75 mL) was treated with compound 91 (51.3 mg, 0.136 mmol), DBU (51.1 μL, 0.339 mmol) followed by BOP (60.0 mg, 0.136 mmol) and heated at 70° C. for 2 h to give compound 92, which was not isolated. 10M aq. solution of sodium hydroxide (250 μl, 2.500 mmol) was added to the reaction mixture of compound 92 and stirred at 75° C. for overnight. The reaction mixture concentrated in vacuo and the residue was treated with MeOH (0.3 mL) and HCl (37% aq. solution, 0.3 mL, 3.7 mmol), stirred at RT for 30 min and concentrated. The crude product was dissolved in DMF, filtered through a PTFE frit, and purified via preparative HPLC conditions A-1. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to yield compound 867 as white solid (13 mg, 40% yield).
  • Additional compounds according to this disclosure can be made following this Procedure, mutatis mutandis.
  • Procedure 13—Compounds Per FIG. 13
  • This Procedure and companion FIG. 13 illustrate another method for making compounds disclosed herein, using compound 851 as an exemplar.
  • Methyl 6-(hydroxymethyl)nicotinate 94 was coupled with compound 55 and then taken to compound 851, following the reaction conditions described hereinabove for the various steps, mutatis mutandis.
  • Additional compounds according to this disclosure can be made following this Procedure, mutatis mutandis.
  • Procedure 14—Compounds Per FIG. 14
  • This Procedure and companion FIG. 14 illustrate another method for making compounds disclosed herein, using compound 877 as an exemplar.
  • Compound 102. A solution of methyl 4-nitro-1H-pyrazole-5-carboxylate 100 (1.00 g, 5.61 mmol) and K2CO3 (0.93 g, 6.73 mmol) in DMF (5.9 mL) was cooled to 0° C. and methyl 4-(bromomethyl)benzoate 101 (1.29 g, 5.61 mmol) was added in portions. The reaction mixture was allowed to warm to RT and stirred for 4 h. The reaction mixture was concentrated in vacuo. The residue was dissolved EtOAc and washed with sat. NaHCO3 and H2O (2×). The organic layer was concentrated in vacuo. The residue was dissolved in DCM and purified via column chromatography (80 g SiO2, 0 to 50% EtOAc-hexane gradient) to give compound 102 (0.37 g, 21%). 1H NMR (400 MHz, CHLOROFORM-d) δ 8.10-8.08 (m, 1H), 8.06-8.01 (m, 2H), 7.32 (d, J=8.6 Hz, 2H), 5.55 (s, 2H), 3.93 (s, 3H), 3.92-3.92 (m, 3H). LC/MS condition B. LC RT: 0.95 min. LCMS (M+H)=320.1.
  • In the preceding reaction, some of the N2 regioisomer was produced, which was removed during the SiO2 chromatography:
  • Figure US20230140047A1-20230504-C00131
  • Compound 103. Ammonium formate (292 mg, 4.64 mmol) and zinc (189 mg, 2.9 mmol) were added to a solution of compound 102 (370 mg, 1.16 mmol), in THF (1.5 ml)/MeOH (1.5 ml) at RT. The reaction was stirred at RT for 3 h and additional portions of ammonium formate (100 mg, 1.59 mmol) and zinc (100 mg, 2.04 mmol) were added. After 30 min, the reaction mixture filtered through a pad of CELITE™, and the filtrate was concentrated in vacuo to afford a white solid. The solid was suspended in EtOAc, stirred for 30 minutes and filtered. The organic filtrate was then concentrated in vacuo to give compound 103 (335 mg, 100%). 1H NMR (400 MHz, DMSO-d6) δ 7.93-7.86 (m, 2H), 7.19-7.15 (m, 3H), 5.59 (s, 2H), 5.13 (s, 2H), 3.83 (s, 3H), 3.73 (s, 3H). LC/MS condition B: LC RT: 0.78 min. LCMS (M+H)=290.1.
  • Compound 104. 1,3-bis(Methoxycarbonyl)-2-methyl-thiopseudourea 1 (271 mg, 1.27 mmol) and acetic acid (0.994 mL, 17.37 mmol) were added to a solution of compound 103 (335 mg, 1.16 mmol) in MeOH (14 mL) at RT. The reaction mixture was stirred at RT for 3 h and an additional portion of HOAc (0.5 mL, 8.7 mmol) was added. After 16 h at RT another portion of HOAc (0.3 mL, 5.2 mmol) was added. After 72 h, sodium methoxide solution (7.94 mL, 34.7 mmol, 25% in MeOH) was added to the reaction mixture. After stirring for 2 h, the reaction mixture was acidified with HOAc (3 mL) and the resulting slurry was filtered. The resulting solid was washed with H2O and MeOH, and dried overnight to afford compound 104 (383 mg, 93%). 1H NMR (400 MHz, DMSO-d6) δ 11.52-11.22 (m, 1H), 7.94-7.88 (m, 3H), 7.33 (d, J=8.3 Hz, 2H), 5.79 (s, 2H), 3.83 (s, 3H), 3.74 (s, 3H). One proton not visible, likely due to proton exchange. LC/MS condition B: LC RT: 0.80 min. LCMS (M+H)=358.2.
  • Compound 105. (S)-1-((tert-Butyldiphenylsilyl)oxy)hexan-3-amine (71a) hydrochloride (840 mg, 2.14 mmol), BOP (711 mg, 1.61 mmol) and DBU (808 μL, 5.36 mmol) were added sequentially to a RT solution of compound 104 (383 mg, 1.07 mmol) in DMF (5.4 mL). The reaction mixture was stirred for 16 h. At the end of the reaction, the mixture was diluted with EtOAc and washed with H2O (2×). The organic layer was concentrated in vacuo. The residue was dissolved in DCM and purified via column chromatography (40 g SiO2, 0 to 80% EtOAc-hexane gradient) to give provide compound 105 (424 mg, 57%). 1H NMR (400 MHz, DMSO-d6) δ 9.53-9.49 (m, 1H), 7.97 (s, 1H), 7.66 (d, J=8.4 Hz, 2H), 7.64-7.59 (m, 1H), 7.54-7.50 (m, 2H), 7.47-7.44 (m, 1H), 7.42-7.39 (m, 2H), 7.36-7.31 (m, 2H), 7.31 (br t, J=1.4 Hz, 1H), 7.19-7.12 (m, 2H), 6.94 (d, J=8.4 Hz, 2H), 6.32 (d, J=8.6 Hz, 1H), 6.03-5.84 (m, 2H), 4.56 (td, J=8.4, 4.4 Hz, 1H), 3.72 (s, 3H), 3.56 (s, 3H), 3.49-3.38 (m, 1H), 1.84-1.68 (m, 2H), 1.55-1.39 (m, 2H), 1.36-1.21 (m, 1H), 1.18-1.07 (m, 1H), 0.91-0.86 (m, 9H), 0.74 (t, J=7.3 Hz, 3H). LC/MS condition B: LC RT: 1.08 min. LCMS (M+H)=695.6.
  • Compound 106. A solution of compound 105 (424 mg, 0.610 mmol) in THF (8.7 mL) was cooled to −15° C. and LiAlH4 (1M in THF, 1.1 mL, 1.1 mmol) was added. After 15 min an additional portion of LiAlH4 (1M in THF, 0.3 mL, 0.3 mmol) was added. After 10 min the reaction was quenched with MeOH and Rochelle's salt (sat. soln) and was stirred at RT for 30 min. The mixture was extracted with EtOAc (3×). The combined organic layers were washed with H2O, and concentrated in vacuo. The residue was dissolved in DCM/MeOH, absorbed onto CELITE™ and purified via column chromatography (24 g SiO2; 0 to 100% EtOAc-hexane gradient elution) to afford compound 106 (181 mg, 44%). 1H NMR (400 MHz, DMSO-d6) δ 9.47 (s, 1H), 7.91 (s, 1H), 7.62 (ddt, J=5.7, 3.6, 1.8 Hz, 1H), 7.56 (dd, J=7.9, 1.5 Hz, 2H), 7.48-7.43 (m, 3H), 7.39-7.32 (m, 3H), 7.24-7.20 (m, 2H), 7.08 (d, J=8.1 Hz, 2H), 6.91 (d, J=8.1 Hz, 2H), 6.29 (d, J=8.6 Hz, 1H), 5.78 (d, J=1.9 Hz, 2H), 5.06 (t, J=5.6 Hz, 1H), 4.63-4.54 (m, 1H), 4.32 (d, J=5.4 Hz, 2H), 3.57 (s, 3H), 3.55-3.49 (m, 1H), 1.85-1.76 (m, 2H), 1.55-1.40 (m, 2H), 1.20-1.02 (m, 2H), 0.91 (s, 9H), 0.78 (t, J=7.3 Hz, 3H). LC/MS condition B: LC RT: 0.95 min. LCMS (M+H)=667.6.
  • Compound 107. Thionyl chloride (99 μL, 1.4 mmol) was added to a RT solution of methyl compound 106 (181 mg, 0.224 mmol) in THF (2.7 mL). After stirring for 10 min, the reaction mixture was concentrated in vacuo. The residue was redissolved in DCM and concentrated in vacuo to give compound 107 (172.2 mg, 93%). LC/MS condition B: LC RT: 1.11 min. LCMS (M+H)=685.6.
  • Compound 108. Compound 107 (23 mg, 0.034 mmol) was dissolved in DMF (0.7 mL) at RT and was treated with DIEA (88 μL, 0.50 mmol) and 3-methoxyazetidine (26a), as its hydrochloride (21 mg, 0.17 mmol). After stirring for 2 h at 70° C., the reaction mixture was concentrated in vacuo to give crude compound 108, which was used without further purification. LC/MS condition B: LC RT: 0.89 min. LCMS (M+H)=736.7.
  • Compound 877 A solution of compound 108 (25 mg, 0.034 mmol) in dioxane (0.7 mL) at RT was treated with NaOH (10M aq. soln, 0.1 mL, 1.0 mmol). The reaction was heated to 75° C. for 2 h, before the addition of another portion of NaOH (10M aq. soln, 0.15 mL, 1.5 mmol). After 4 h heating at 80° C., the reaction mixture was concentrated in vacuo. The residue was treated with MeOH (0.5 mL), HCl (37% aq. soln, 0.4 mL, 4.9 mmol) was added, stirred at RT for 60 min, and concentrated. The crude product was dissolved in DMF, filtered through a PTFE frit, and purified via preparative LC/MS Condition A-2. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to give a residue which was further purified via preparative LC/MS Conditions A-1. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to give compound 877, as its TFA salt (5.7 mg, 38%). LC/MS condition C: LC RT: 1.0 min. LCMS (M+H)=440.2.
  • Compound 874. A solution of compound 106 (20 mg, 0.030 mmol) in dioxane (0.15 mL) at RT was treated with NaOH (10M aq. soln, 0.2 mL, 2.0 mmol) and the reaction was heated to 75° C. for 1 h, before the addition of another portion of NaOH (10M aq. soln, 0.2 mL, 2.0 mmol). After 3 h heating at 75° C., the reaction mixture was concentrated in vacuo. The residue was treated with MeOH (0.5 mL) and HCl (37% aq. soln, 0.4 mL, 4.9 mmol), stirred at RT for 30 min and concentrated. The crude product was dissolved in DMF, filtered through a PTFE frit, and purified via preparative LC/MS conditions A-2. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation to give compound 874. LC/MS conditions C: LC RT: 1.07 min. LCMS (M+H)=371.2.
  • Additional compounds according to this disclosure can be made following this Procedure, mutatis mutandis.
  • Procedure 15—Compound 829
  • This procedure relates to the preparation of compounds 829 and its regiosiomer. These compounds were prepared by reacting compounds 36 and 37 with MeMgCl Grignard reagent.
  • Figure US20230140047A1-20230504-C00132
  • To a mixture of compounds 36 and 37 (60 mg, 0.134 mmol) in THF (1 mL) was added MeMgCl (0.171 mL, 0.513 mmol) at 0° C. After 1 hr, LCMS showed reaction was completed. The mixture was purified via preparative HPLC/MS with the following conditions: Column: XBridge C18, 200 mm×19 mm, 5-μm particles; Mobile Phase A: 5:95 acetonitrile:water with 0.1% TFA; Mobile Phase B: 95:5 acetonitrile:water with 0.1% TFA; Gradient: a 0-minute hold at 10% B, 10-45% B over 20 minutes, then a 4-minute hold at 100% B; Flow Rate: 20 mL/min; Column Temperature: 25° C. Fraction collection was triggered by MS signals. Fractions containing the desired product were combined and dried via centrifugal evaporation.
  • Properties
  • Analytical data for compounds of this disclosure are provided in following Table B, along with their human TLR7 (hTLR7) agonism data. In some instances, the reported activity agonism value is the average of plural assays. The association of a particular compound with a particular Procedure is illustrative, and not exhaustive. A compound could be made by one of the other Procedures and, conversely, the same Procedure could be used to make other compounds of this disclosure. The RT (retention time) column refers to the retention time per LCMS procedure B/C/D/E above, as applicable.
  • TABLE B
    hTLR7
    Cpd. EC50 LC/MS RT 1H NMR
    No. Procedure (nM) [M + H]+ (min) (500 MHz, DMSO-d6)
    800 1 58.3 409.5 1.05 δ 7.55 (s, 1H), 7.04 (s, 1H), 6.78 (d, J = 7.7 Hz,
    1H), 6.43 (dd, J = 21.5, 6.9 Hz, 2H), 5.61 (d, J =
    11.9 Hz, 4H), 3.84 (s, 3H), 3.61 (s, 1H), 3.43-
    3.37 (m, 1H), 3.18 (t, J = 7.8 Hz, 1H), 2.55 (s,
    1H), 2.09-2.02 (m, 2H), 1.91 (s, 1H), 1.77-
    1.69 (m, 2H), 1.67-1.59 (m, 1H), 1.58-1.43
    (m, 3H), 1.20 (p, J = 7.4 Hz, 2H), 0.85 (t, J =
    7.4 Hz, 3H)
    801 2 160 439.5 0.87 δ 8.22 (s, 1H), 7.73 (s, 1H), 7.18 (s, 1H), 7.01
    (d, J = 8.0 Hz, 1H), 6.91 (d, J = 7.6 Hz, 1H),
    5.71 (s, 2H), 4.13 (s, 2H), 3.92 (d, J = 9.8 Hz,
    2H), 3.77 (s, 1H), 3.64 (s, 1H), 3.57 (d, J = 6.6
    Hz, 1H), 3.30 (t, J = 11.6 Hz, 3H), 2.89 (s, 1H),
    2.73 (s, 1H), 1.99 (d, J = 12.3 Hz, 2H), 1.63-
    1.52 (m, 4H), 1.28 (h, J = 7.4 Hz, 2H), 0.88 (t,
    J = 7.3 Hz, 3H)
    802 4, 6 43.8 492.2 1.21 δ 7.55 (s, 1H), 7.36 (d, J = 8.5 Hz, 2H), 6.95 (d,
    J = 8.5 Hz, 2H), 6.76 (d, J = 2.1 Hz, 1H), 6.52 (d,
    J = 8.5 Hz, 1H), 6.49-6.43 (m, 1H), 6.40 (dd,
    J = 8.4, 2.3 Hz, 1H), 5.64 (s, 2H), 5.60 (s, 2H),
    3.80 (s, 3H), 3.78 (s, 2H), 2.65 (t, J = 5.6 Hz,
    2H), 1.56-1.42 (m, 2H), 1.29-1.16 (m, 2H),
    0.87 (t, J = 7.5 Hz, 3H).
    803 4, 6 569 449.1 1.58 δ 7.74 (s, 1H), 7.33 (br d, J = 8.2 Hz, 2H), 6.97
    (d, J = 8.2 Hz, 2H), 6.82 (d, J = 8.2 Hz, 1H), 6.74
    (d, J = 2.1 Hz, 1H), 6.47-6.41 (m, 1H), 5.68 (s,
    2H), 4.48 (br s, 2H), 3.73 (s, 3H), 3.64-3.53
    (m, 2H), 1.59 (dt, J = 14.6, 7.3 Hz, 2H), 1.27 (dt,
    J = 14.9, 7.4 Hz, 2H), 0.90 (t, J = 7.3 Hz, 3H)
    804 2 43.4 406.4 0.91 δ 8.90 (s, 1H), 8.26 (s, 1H), 7.95 (s, 1H), 7.86
    (s, 1H), 7.74 (s, 1H), 7.64 (s, 1H), 7.50 (s, 1H),
    7.15 (s, 1H), 6.88 (d, J = 7.8 Hz, 1H), 6.82 (d,
    J = 7.9 Hz, 1H), 5.71 (s, 2H), 5.33 (s, 2H), 3.77
    (s, 3H), 2.90 (s, 1H), 2.73 (d, J = 9.5 Hz, 1H),
    1.56 (p, J = 7.3 Hz, 2H), 1.23 (q, J = 7.3 Hz,
    2H), 0.85 (t, J = 7.4 Hz, 3H)
    805 2 41.4 427.5 0.94 δ 8.18 (s, 1H), 7.95 (s, 1H), 7.75 (s, 1H), 7.26
    (s, 1H), 7.02 (d, J = 7.9 Hz, 1H), 6.82 (d, J = 7.6
    Hz, 1H), 5.73 (s, 2H), 4.13 (s, 2H), 3.79 (s, 3H),
    3.56 (d, J = 7.0 Hz, 1H), 2.90 (s, 1H), 2.72 (d,
    J = 17.1 Hz, 3H), 1.58 (p, J = 7.5 Hz, 2H), 1.28
    (dd, J = 14.9, 7.5 Hz, 2H), 1.16 (s, 5H), 0.89 (t,
    J = 7.4 Hz, 3H)
    806 2 41.4 453.5 0.82 δ 8.28 (s, 1H), 7.82 (s, 1H), 7.76 (d, J = 12.9
    Hz, 1H), 7.21 (s, 1H), 7.01 (d, J = 7.8 Hz, 1H),
    6.90 (d, J = 7.7 Hz, 1H), 5.74 (s, 2H), 4.13 (s,
    1H), 3.78 (s, 3H), 3.58 (q, J = 6.7 Hz, 1H), 3.42
    (s, 0H), 2.96 (s, 1H), 2.72 (s, 1H), 2.08 (d, J =
    12.0 Hz, 2H), 1.89 (d, J = 12.8 Hz, 2H), 1.59 (h,
    J = 7.6 Hz, 2H), 1.45-1.23 (m, 4H), 1.16 (q,
    J = 11.1 Hz, 2H), 0.91 (t, J = 7.3 Hz, 3H)
    807 2 2548 439.5 0.97 δ 7.55 (s, 1H), 7.03 (s, 1H), 6.76 (d, J = 7.8 Hz,
    1H), 6.45 (d, J = 7.8 Hz, 1H), 6.39 (s, 1H), 5.61
    (d, J = 13.7 Hz, 3H), 3.89 (d, J = 19.6 Hz, 3H),
    3.84 (s, 2H), 3.40 (d, J = 6.7 Hz, 1H), 3.18 (s,
    1H), 2.44 (s, 1H), 2.23 (d, J = 8.6 Hz, 2H), 1.86
    (s, 1H), 1.75 (s, 1H), 1.48 (t, J = 7.4 Hz, 2H),
    1.41 (d, J = 9.6 Hz, 2H), 1.20 (q, J = 7.5 Hz,
    2H), 0.85 (t, J = 7.3 Hz, 3H)
    808 4, 6 157 506.2 1.22 δ 7.74 (br s, 1H), 7.51 (br d, J = 8.5 Hz, 2H), 7.05
    (br d, J = 8.5 Hz, 2H), 6.84-6.78 (m, 2H), 6.47
    (dd, J = 7.8, 2.0 Hz, 1H), 5.70 (s, 2H), 4.14 (br s,
    2H), 3.75 (s, 3H), 3.61-3.57 (m, 2H), 3.12-
    3.06 (m, 2H), 1.62-1.56 (m, 2H), 1.32-1.26
    (m, 2H), 0.90 (br t, J = 7.3 Hz, 3H).
    809 4, 6 74.5 531.2 1.43 δ 7.76 (s, 1H), 7.37-7.31 (m, 2H), 7.02-6.94
    (m, 2H), 6.85 (d, J = 7.9 Hz, 1H), 6.77 (s, 1H),
    6.46-6.41 (m, 1H), 5.72-5.69 (m, 2H), 3.73 (s,
    3H), 3.62-3.58 (m, 2H), 1.64-1.57 (m, 2H),
    1.32-1.26 (m, 2H), 0.90 (t, J = 7.3 Hz, 3H).
    810 4, 6 245 561.4 1.0 δ 7.74 (s, 1H), 7.38-7.23 (m, 2H), 6.98 (br d,
    J = 8.7 Hz, 2H), 6.82 (d, J = 8.5 Hz, 1H), 6.75 (s,
    1H), 6.42 (br d, J = 7.0 Hz, 1H), 5.68 (s, 2H),
    3.50 (s, 3H), 3.17 (s, 2H), 3.08 (br dd, J = 6.3, 2.2
    Hz, 2H), 2.97-2.88 (m, 6H), 2.65 (br s, 2H),
    1.63-1.52 (m, 2H), 1.28-1.24 (m, 2H), 0.93-
    0.81 (m, 3H).
    811 2 138 425.2 0.8 δ 7.54 (s, 1H), 7.00 (s, 1H), 6.77-6.72 (m, 1H),
    6.49-6.40 (m, 2H), 5.59 (d, J = 9.5 Hz, 3H),
    4.23 (t, J = 6.1 Hz, 1H), 3.82 (s, 3H), 3.38 (q,
    J = 6.5 Hz, 2H), 3.18 (d, J = 13.2 Hz, 2H), 1.95 (t,
    J = 5.8 Hz, 2H), 1.89 (dd, J = 7.5, 4.9 Hz, 2H),
    1.84 (s, 8H), 1.50-1.43 (m, 2H), 1.18 (q, J =
    7.4 Hz, 2H), 0.84 (t, J = 7.3 Hz, 3H)
    812 2 102 450.5 0.77 δ 8.32 (s, 1H), 7.95 (s, 1H), 7.77 (s, 1H), 7.15
    (s, 1H), 6.96 (d, J = 7.7 Hz, 1H), 6.86 (d, J = 7.6
    Hz, 1H), 5.74 (s, 2H), 4.22 (s, 1H), 3.78 (s, 2H),
    2.90 (s, 3H), 2.55 (s, 1H), 1.58 (p, J = 7.3 Hz,
    2H), 1.40 (s, 1H), 1.26 (q, J = 7.5 Hz, 2H), 0.88
    (t, J = 7.3 Hz, 3H)
    813 2 23.8 424.5 0.78 δ 8.28 (s, 1H), 7.96 (s, 1H), 7.90 (s, 0H), 7.76
    (s, 1H), 7.27 (s, 0H), 7.17 (s, 0H), 7.06 (d, J =
    13.9 Hz, 1H), 6.89 (d, J = 7.8 Hz, 1H), 6.81 (d,
    J = 7.6 Hz, 1H), 5.72 (s, 2H), 3.77 (s, 2H), 3.73
    (s, 1H), 3.58 (d, J = 6.9 Hz, 1H), 3.17 (d, J = 6.4
    Hz, 2H), 2.90 (s, 3H), 2.55 (s, 1H), 1.58 (p, J =
    7.1 Hz, 2H), 1.26 (q, J = 7.5 Hz, 2H), 0.88 (t,
    J = 7.3 Hz, 3H)
    814 2 97.6 411.5 0.83 δ 8.28 (s, 1H), 7.84 (s, 1H), 7.76 (d, J = 9.0 Hz,
    1H), 7.19 (s, 1H), 6.98 (d, J = 7.7 Hz, 1H), 6.87
    (d, J = 7.6 Hz, 1H), 5.74 (s, 2H), 4.66 (t, J = 7.4
    Hz, 2H), 4.56 (t, J = 6.6 Hz, 2H), 4.36 (q, J =
    6.5 Hz, 1H), 4.23 (d, J = 14.0 Hz, 1H), 4.07 (s,
    1H), 3.79 (s, 3H), 3.58 (d, J = 6.9 Hz, 2H), 2.51
    (s, 8H), 1.60 (q, J = 7.4 Hz, 2H), 1.28 (dt, J =
    15.3, 7.6 Hz, 2H), 0.90 (t, J = 7.3 Hz, 3H)
    815 2 33.2 439.5 1.1 δ 8.29 (d, J = 5.7 Hz, 1H), 7.96 (s, 1H), 7.89 (s,
    1H), 7.77 (d, J = 5.7 Hz, 1H), 7.15 (s, 1H), 6.97
    (d, J = 8.0 Hz, 1H), 6.85 (d, J = 7.7 Hz, 1H),
    5.75 (d, J = 10.6 Hz, 2H), 4.35 (s, 1H), 4.29 (s,
    1H), 4.25 (s, 1H), 4.07 (s, 1H), 4.00 (s, 1H),
    3.79 (d, J = 5.5 Hz, 3H), 3.57 (q, J = 6.7 Hz,
    1H), 3.43 (s, 1H), 2.90 (s, 2H), 2.73 (d, J = 9.7
    Hz, 3H), 2.55 (s, 1H), 1.73 (s, 1H), 1.58 (p, J =
    7.4 Hz, 2H), 1.27 (q, J = 7.4 Hz, 2H), 0.88 (td,
    J = 7.3, 4.3 Hz, 3H)
    816 2 92.5 399.4 1.0 δ 8.19 (s, 1H), 7.95 (s, 1H), 7.74 (s, 1H), 7.22
    (s, 1H), 7.00 (d, J = 7.8 Hz, 1H), 6.86 (d, J = 7.7
    Hz, 1H), 5.73 (s, 2H), 4.14 (s, 2H), 3.90 (s, 1H),
    3.78 (s, 3H), 3.66 (s, 1H), 3.57 (s, 1H), 3.46 (s,
    1H), 3.18 (s, 1H), 2.95 (t, J = 5.5 Hz, 2H), 2.90
    (s, 1H), 2.74 (s, 1H), 2.55 (s, 1H), 1.58 (p, J =
    7.3 Hz, 2H), 1.28 (h, J = 7.4 Hz, 2H), 0.89 (t,
    J = 7.3 Hz, 3H)
    817 3 27.8 400.4 1.17 δ 7.73 (s, 1H), 7.64 (s, 1H), 7.33 (d, J = 8.5 Hz,
    1H), 6.98 (s, 1H), 6.80 (d, J = 7.9 Hz, 1H), 6.64
    (d, J = 7.6 Hz, 1H), 5.72 (d, J = 16.4 Hz, 1H),
    5.64 (d, J = 16.3 Hz, 1H), 4.49 (d, J = 6.4 Hz,
    1H), 4.44 (s, 2H), 3.87 (s, 1H), 3.75 (s, 3H),
    3.46 (d, J = 11.3 Hz, 1H), 3.38 (d, J = 6.7 Hz,
    1H), 3.14 (s, 1H), 2.52 (s, 2H), 1.68 (dq, J =
    12.5, 6.6, 6.2 Hz, 2H), 1.47 (t, J = 5.8 Hz, 2H),
    1.09 (q, J = 9.0, 8.2 Hz, 2H), 0.78 (t, J = 7.3 Hz,
    3H)
    818 2 258 356.4 1.2 8.13 (s, 1H), 7.73 (s, 2H), 6.99 (s, 1H), 6.86-
    6.76 (m, 2H), 5.69 (s, 2H), 4.47 (s, 2H), 3.76 (s,
    3H), 3.57 (d, J = 6.7 Hz, 1H), 1.57 (p, J = 7.2
    Hz, 2H), 1.27 (dt, J = 15.0, 7.5 Hz, 2H), 1.17 (t,
    J = 7.2 Hz, 1H), 0.89 (t, J = 7.4 Hz, 3H)
    819 2 44.4 436.5 0.97 δ 8.27 (t, J = 5.7 Hz, 1H), 7.94 (s, 1H), 7.76 (d,
    J = 0.9 Hz, 1H), 7.30 (s, 1H), 7.19 (s, 0H), 7.09
    (s, 1H), 6.93 (d, J = 7.8 Hz, 1H), 6.82 (d, J = 7.7
    Hz, 1H), 5.71 (s, 2H), 4.30 (s, 1H), 4.01 (s, 1H),
    3.94 (s, 1H), 3.76 (s, 3H), 3.57 (s, 1H), 3.19 (d,
    J = 15.2 Hz, 1H), 3.02 (s, 1H), 2.89 (s, 1H),
    2.73 (s, 1H), 2.55 (s, 3H), 2.22 (s, 1H), 1.84 (d,
    J = 11.5 Hz, 1H), 1.57 (p, J = 7.2 Hz, 2H), 1.25
    (h, J = 7.5 Hz, 2H), 0.87 (t, J = 7.3 Hz, 3H)
    820 2 121 453.5 1.09 δ 7.72 (s, 1H), 7.21 (s, 1H), 7.01 (d, J = 7.8 Hz,
    1H), 6.91 (d, J = 7.7 Hz, 1H), 5.71 (s, 2H), 4.07
    (s, 2H), 3.78 (s, 3H), 3.54 (s, 1H), 2.89 (s, 1H),
    2.73 (s, 1H), 2.55 (s, 4H), 1.82-1.71 (m, 5H),
    1.59 (dd, J = 15.5, 7.8 Hz, 4H), 1.30 (q, J = 7.5
    Hz, 2H), 0.90 (t, J = 7.4 Hz, 3H)
    821 2 33.8 438.5 1.27 δ 8.30 (s, 1H), 7.94 (s, 1H), 7.77 (q, J = 1.5 Hz,
    1H), 7.16 (s, 1H), 6.97 (d, J = 7.7 Hz, 1H), 6.83
    (d, J = 7.7 Hz, 1H), 5.73 (s, 2H), 4.14 (s, 1H),
    3.78 (d, J = 1.6 Hz, 3H), 3.57 (s, 1H), 3.23 (s,
    2H), 2.89 (d, J = 1.5 Hz, 3H), 2.73 (d, J = 1.3
    Hz, 3H), 2.55 (s, 2H), 2.03 (s, 2H), 1.57 (p, J =
    7.3 Hz, 3H), 1.25 (q, J = 7.4 Hz, 3H), 0.87 (t,
    J = 7.3 Hz, 3H)
    822 2 244 475.5 1.27 δ 9.95 (s, 0H), 8.20 (d, J = 6.0 Hz, 1H), 7.73 (s,
    1H), 7.49 (s, 1H), 7.29 (s, 1H), 7.18 (s, 1H),
    7.03 (s, 1H), 6.89-6.83 (m, 2H), 6.46 (d, J =
    8.3 Hz, 1H), 5.81 (s, 1H), 5.66 (s, 1H), 4.20 (s,
    1H), 3.86 (s, 1H), 3.72 (s, 1H), 3.46 (t, J = 7.3
    Hz, 1H), 2.89 (s, 1H), 2.73 (s, 0H), 2.55 (s, 3H),
    1.59-1.52 (m, 2H), 1.24 (q, J = 7.4 Hz, 2H),
    0.86 (t, J = 7.3 Hz, 3H)
    823 2 21.5 468.6 1.01 δ 7.56 (s, 1H), 6.96 (s, 1H), 6.74 (d, J = 7.5 Hz,
    1H), 6.43 (dd, J = 12.7, 6.8 Hz, 2H), 5.70 (s,
    1H), 5.61 (s, 2H), 3.88-3.74 (m, 5H), 3.39 (d,
    J = 10.3 Hz, 2H), 2.59-2.53 (m, 9H), 2.40 (t,
    J = 6.3 Hz, 4H), 1.91 (d, J = 1.2 Hz, 3H), 1.45 (p,
    J = 7.2 Hz, 2H), 1.16 (h, J = 7.5 Hz, 2H), 0.83
    (t, J = 7.4 Hz, 3H)
    824 2 304 436.5 1.02 δ 7.55 (s, 1H), 6.90 (d, J = 1.5 Hz, 1H), 6.70 (d,
    J = 7.5 Hz, 1H), 6.42 (d, J = 7.6 Hz, 1H), 5.61
    (d, J = 17.6 Hz, 3H), 3.83 (s, 2H), 3.72 (s, 1H),
    3.39 (d, J = 6.1 Hz, 1H), 3.20 (s, 2H), 2.55 (s,
    5H), 1.87 (s, 4H), 1.52-1.42 (m, 2H), 1.19 (h,
    J = 7.3 Hz, 2H), 0.85 (t, J = 7.3 Hz, 3H)
    825 2 43.5 427.5 1.25 δ 8.24 (s, 1H), 7.94 (s, 1H), 7.81 (s, 1H), 7.76
    (s, 1H), 7.21 (s, 1H), 7.00 (d, J = 7.8 Hz, 1H),
    6.83 (d, J = 7.7 Hz, 1H), 5.74 (s, 2H), 4.25 (s,
    1H), 3.89 (s, 1H), 3.78 (s, 3H), 3.56 (d, J = 6.9
    Hz, 1H), 3.43 (s, 1H), 3.17 (s, 1H), 3.08 (s, 1H),
    2.89 (s, 1H), 2.73 (s, 1H), 2.66 (s, 3H), 2.54 (s,
    3H), 1.83 (s, 1H), 1.57 (q, J = 7.3 Hz, 2H), 1.25
    (dt, J = 14.9, 7.5 Hz, 2H), 0.87 (t, J = 7.4 Hz,
    3H)
    826 2 182 413.5 0.85 δ 8.29 (s, 1H), 7.95 (s, 1H), 7.75 (s, 1H), 7.21
    (s, 1H), 7.00 (d, J = 7.6 Hz, 1H), 6.87 (d, J = 7.7
    Hz, 1H), 5.73 (s, 2H), 4.13 (s, 2H), 3.78 (s, 2H),
    3.58 (t, J = 4.9 Hz, 2H), 3.29 (s, 2H), 3.07 (t, J =
    5.1 Hz, 2H), 2.90 (s, 2H), 2.74 (s, 2H), 1.59 (p,
    J = 7.4 Hz, 2H), 1.28 (h, J = 7.8 Hz, 2H), 0.89
    (t, J = 7.4 Hz, 3H)
    827 2 47.6 438.5 0.89 δ 7.65 (s, 1H), 6.94 (d, J = 7.9 Hz, 2H), 6.82 (d,
    J = 7.6 Hz, 1H), 5.74 (s, 1H), 5.35 (s, 2H), 3.87
    (d, J = 2.1 Hz, 2H), 3.84 (s, 9H), 3.40 (s, 2H),
    2.54 (s, 3H), 2.14 (s, 3H), 1.83 (s, 1H), 1.52 (t,
    J = 7.6 Hz, 2H), 1.28 (q, J = 7.5 Hz, 2H), 0.85 (t,
    J = 7.4 Hz, 3H)
    828 2 98.7 411.5 0.92 δ 8.17 (s, 0H), 7.72 (d, J = 2.4 Hz, 1H), 7.63 (s,
    0H), 7.24 (d, J = 4.4 Hz, 1H), 7.03 (d, J = 7.7
    Hz, 1H), 6.92 (d, J = 7.7 Hz, 1H), 5.73 (s, 2H),
    4.09 (s, 2H), 3.78 s, 3H), 3.61-3.54 (m, 1H),
    3.48 (s, 2H), 2.93 (s, 1H), 2.55 (s, 1H), 1.62 (q,
    J = 7.4 Hz, 2H), 1.38-1.27 (m, 11H), 1.21-
    1.14 (m, 1H), 0.92 (t, J = 7.4 Hz, 3H)
    829 15 1336 448.2 δ 8.15 (s, 1H), 7.82 (s, 1H), 7.74 (br d, J = 7.9
    Hz, 1H), 7.59 (br s, 1H), 7.47 (d, J = 8.5 Hz, 2H),
    7.05-6.93 (m, 3H), 5.53 (br s, 1H), 5.42 (s,
    2H), 1.55 (br t, J = 7.5 Hz, 2H), 1.43 (s, 6H),
    1.36-1.25 (m, 2H), 0.89 (br t, J = 7.5 Hz, 3H)
    830 2 30.2 423.5 0.92 δ 8.25 (s, 1H), 7.83 (s, 1H), 7.77 (s, 1H), 7.21
    (s, 1H), 6.99 (d, J = 7.7 Hz, 1H), 6.82 (d, J = 7.7
    Hz, 1H), 5.76 (s, 2H), 4.23 (s, 1H), 3.80 (s, 3H),
    3.57 (d, J = 6.6 Hz, 1H), 2.93 (s, 1H), 2.55 (s,
    4H), 1.78 (s, 1H), 1.65 (s, 1H), 1.58 (q, J = 7.3
    Hz, 3H), 1.25 (q, J = 7.4 Hz, 2H), 1.17 (t, J =
    7.2 Hz, 1H), 0.88 (t, J = 7.3 Hz, 3H)
    831 2 104 411.5 1.0 δ 7.74 (s, 1H), 7.21 (s, 1H), 6.99 (d, J = 8.0 Hz,
    1H), 6.86 (d, J = 7.7 Hz, 1H), 5.74 (s, 1H), 4.12
    (s, 1H), 3.78 (s, 3H), 3.57 (d, J = 6.6 Hz, 1H),
    3.41 (s, 1H), 2.93 (s, 3H), 2.89 (s, 1H), 2.55 (s,
    3H), 1.60 (q, J = 7.5 Hz, 3H), 1.30 (dq, J = 15.5,
    7.5 Hz, 3H), 1.17 (t, J = 7.3 Hz, 6H), 0.89 (q,
    J = 7.2 Hz, 4H)
    832 2 141 425.5 0.82 δ 8.31 (t, J = 5.6 Hz, 1H), 7.77 (d, J = 0.8 Hz,
    1H), 7.19 (s, 1H), 6.99 (d, J = 7.7 Hz, 1H), 6.85
    (d, J = 7.7 Hz, 1H), 5.75 (s, 2H), 4.24 (s, 1H),
    3.79 (s, 3H), 3.58 (t, J = 6.7 Hz, 1H), 3.09 (s,
    2H), 2.97-2.88 (m, 1H), 2.55 (d, J = 0.8 Hz,
    9H), 1.57 (p, J = 7.3 Hz, 2H), 1.25 (h, J = 7.3
    Hz, 2H), 1.17 (t, J = 7.3 Hz, 1H), 0.88 (t, J = 7.3
    Hz, 3H)
    833 2 31.1 409.5 1.11 δ 8.22 (s, 1H), 7.76 (s, 1H), 7.22 (s, 1H), 7.01
    (d, J = 7.5 Hz, 1H), 6.83 (d, J = 7.7 Hz, 1H),
    5.75 (s, 2H), 4.31 (s, 2H), 3.80 (s, 3H), 3.61-
    3.54 (m, 1H), 2.93 (s, 1H), 2.55 (s, 8H), 1.57 (p,
    J = 7.2 Hz, 2H), 1.27 (dt, J = 14.7, 7.5 Hz, 2H),
    1.17 (t, J = 7.2 Hz, 1H), 0.88 (t, J = 7.3 Hz, 3H)
    834 3 29.9 494.3 0.89 δ 7.79 (s, 1H), 7.60 (d, J = 8.5 Hz, 1H), 7.13 (s,
    1H), 6.93 (d, J = 7.8 Hz, 1H), 6.70 (d, J = 7.7
    Hz, 1H), 5.82-5.69 (m, 2H), 4.52 (d, J = 9.2
    Hz, 1H), 3.80 (s, 2H), 3.52 (s, 2H), 3.38 (t, J =
    6.5 Hz, 2H), 2.92 (d, J = 6.0 Hz, 1H), 2.55 (s,
    10H), 1.71 (q, J = 6.3 Hz, 2H), 1.53 (d, J = 8.7
    Hz, 2H), 1.16 (td, J = 8.1, 7.7, 2.7 Hz, 4H), 0.81
    (t, J = 7.3 Hz, 3H)
    835 3 89 480.9 1 δ 7.57 (s, 1H), 6.91 (s, 1H), 6.70 (d, J = 7.8 Hz,
    1H), 6.35 (d, J = 7.7 Hz, 1H), 5.71-5.61 (m,
    3H), 5.55 (d, J = 17.0 Hz, 1H), 4.31 (d, J = 9.9
    Hz, 1H), 3.84 (s, 2H), 3.79 (s, 2H), 3.44 (s, 1H),
    3.29 (d, J = 7.1 Hz, 1H), 3.26 (d, J = 48.2 Hz,
    5H), 3.17 (s, 0H), 2.55 (s, 6H), 1.84 (s, 5H),
    1.63 (dd, J = 13.7, 6.6 Hz, 1H), 1.53-1.47 (m,
    1H), 1.37 (dt, J = 34.6, 7.6 Hz, 2H), 1.08-1.01
    (m, 2H), 0.76 (t, J = 7.3 Hz, 3H)
    836 3 15.3 451.2 0.83 δ 8.63 (s, 1H), 7.76 (s, 1H), 7.52 (d, J = 7.2 Hz,
    2H), 7.35 (s, 1H), 7.14 (s, 1H), 6.85 (d, J = 7.7
    Hz, 1H), 6.69 (d, J = 7.8 Hz, 1H), 5.77 (d, J =
    16.5 Hz, 1H), 5.69 (d, J = 16.4 Hz, 1H), 5.29 (s,
    2H), 4.52 (d, J = 7.0 Hz, 1H), 3.79 (s, 3H), 3.38
    (d, J = 6.7 Hz, 1H), 3.18 (s, 0H), 2.93 (s, 2H),
    2.55 (s, 6H), 1.71 (p, J = 6.1 Hz, 2H), 1.50 (q,
    J = 7.2 Hz, 2H), 1.13 (dt, J = 35.1, 7.5 Hz, 5H),
    0.78 (t, J = 7.3 Hz, 3H)
    837 3 2.4 491.2 0.71 δ 8.67 (d, J = 5.1 Hz, 2H), 7.91 (s, 0H), 7.78 (s,
    1H), 7.71 (d, J = 8.3 Hz, 1H), 7.53 (d, J = 5.1
    Hz, 2H), 7.22 (s, 1H), 7.00 (d, J = 7.6 Hz, 1H),
    6.79 (d, J = 7.7 Hz, 1H), 5.82-5.71 (m, 2H),
    4.56 (d, J = 7.2 Hz, 1H), 4.22 (d, J = 19.9 Hz,
    4H), 3.79 (s, 3H), 3.41 (d, J = 6.3 Hz, 0H), 2.93
    (q, J = 6.7 Hz, 1H), 2.55 (s, 0H), 1.79-1.72 (m,
    2H), 1.56 (p, J = 8.0, 7.4 Hz, 2H), 1.23 (d, J =
    10.9 Hz, 1H), 1.18 (q, J = 8.3, 7.4 Hz, 4H), 0.84
    (t, J = 7.3 Hz, 3H)
    838 3 39.6 483.1 0.7 δ 7.80 (s, 1H), 7.59 (d, J = 8.5 Hz, 1H), 7.19 (s,
    1H), 6.95 (d, J = 7.9 Hz, 1H), 6.68 (d, J = 7.7
    Hz, 1H), 5.79 (d, J = 16.8 Hz, 1H), 5.74 (d, J =
    16.7 Hz, 1H), 4.57-4.51 (m, 1H), 3.81 (s, 3H),
    3.40 (d, J = 17.5 Hz, 1H), 3.22 (s, 1H), 2.97-
    2.89 (m, 2H), 2.55 (s, 1H), 2.02 (s, 1H), 1.79-
    1.68 (m, 2H), 1.56-1.50 (m, 2H), 1.16 (dt, J =
    12.8, 7.3 Hz, 5H), 0.82 (t, J = 7.3 Hz, 3H)
    839 3 242 481.1 0.74 δ 7.57 (s, 1H), 7.01 (s, 1H), 6.78 (d, J = 7.9 Hz,
    1H), 6.37 (d, J = 7.7 Hz, 1H), 5.74-5.66 (m,
    1H), 5.64 (s, 2H), 5.55 (d, J = 17.1 Hz, 1H),
    4.31 (s, 1H), 3.84 (s, 2H), 3.63 (d, J = 8.6 Hz,
    7H), 3.60 (d, J = 6.0 Hz, 6H), 3.37 (s, 1H), 3.30
    (q, J = 7.9, 7.1 Hz, 2H), 3.16 (d, J = 12.7 Hz,
    1H), 2.81 (s, 1H), 2.70 (d, J = 10.3 Hz, 1H),
    2.55 (s, 7H), 1.81 (s, 3H), 1.63 (dd, J = 13.1, 6.4
    Hz, 1H), 1.51 (dd, J = 13.3, 6.3 Hz, 2H), 1.41
    (d, J = 6.8 Hz, 1H), 1.34 (t, J = 7.4 Hz, 1H),
    1.03 (d, J = 8.0 Hz, 2H), 0.75 (t, J = 7.3 Hz, 3H)
    840 3 14.7 484.2 0.82 δ 8.97 (s, 2H), 7.85 (s, 2H), 7.77 (s, 1H), 7.70
    (d, J = 8.3 Hz, 1H), 7.24 (d, J = 1.5 Hz, 1H),
    7.02 (dd, J = 7.7, 1.5 Hz, 1H), 5.76 (d, J = 2.9
    Hz, 2H), 4.57 (dq, J = 14.2, 6.8 Hz, 2H), 4.17
    (s, 2H), 3.93 (dd, J = 11.3, 3.9 Hz, 2H), 3.80 (s,
    3H), 3.53-3.16 (m, 5H), 2.07-1.92 (m, 2H),
    1.75 (q, J = 6.6 Hz, 2H), 1.68-1.46 (m, 4H),
    1.21 (h, J = 7.3 Hz, 2H), 0.85 (t, J = 7.3 Hz, 3H)
    841 3 9.7 454.2 0.83 δ 7.92 (s, 1H), 7.56 (s, 1H), 7.05 (s, 1H), 6.78
    (d, J = 7.9 Hz, 1H), 6.40 (d, J = 7.8 Hz, 1H),
    5.73 (d, J = 8.9 Hz, 0H), 5.64 (d, J = 18.8 Hz,
    2H), 5.55 (d, J = 17.0 Hz, 1H), 4.30 (s, 1H),
    3.84 (s, 1H), 3.76-3.56 (m, 3H), 3.29 (t, J =
    6.5 Hz, 2H), 3.21-3.15 (m, 1H), 2.89 (s, 1H),
    2.73 (s, 1H), 2.55 (s, 1H), 2.03 (d, J = 9.1 Hz,
    2H), 1.74 (t, J = 9.7 Hz, 2H), 1.62 (t, J = 10.0
    Hz, 2H), 1.52 (dt, J = 16.0, 8.1 Hz, 2H), 1.37
    (dt, J = 33.8, 7.5 Hz, 2H), 1.09-1.02 (m, 2H),
    0.75 (t, J = 7.3 Hz, 3H)
    842 3 28.3 484.3 0.77 δ 7.74 (s, 1H), 7.41 (s, 1H), 7.08 (s, 1H), 6.92
    (d, J = 7.9 Hz, 1H), 6.71 (d, J = 7.9 Hz, 1H),
    5.74 (d, J = 16.7 Hz, 1H), 5.65 (d, J = 16.5 Hz,
    1H), 4.49 (s, 1H), 4.07 (s, 5H), 4.05 (s, 9H),
    3.78 (s, 3H), 3.38 (t, J = 6.1 Hz, 4H), 2.89 (s,
    1H), 2.83 (s, 1H), 2.55 (s, 13H), 1.71 (s, 2H),
    1.65 (d, J = 6.2 Hz, 1H), 1.50-1.44 (m, 2H),
    1.18-1.00 (m, 4H), 0.76 (t, J = 7.3 Hz, 3H)
    843 3 55.9 481.2 1.1 δ 7.57 (s, 1H), 7.01 (s, 1H), 6.78 (d, J = 7.9 Hz,
    1H), 6.37 (d, J = 7.7 Hz, 1H), 5.74-5.66 (m,
    1H), 5.64 (s, 2H), 5.55 (d, J = 17.1 Hz, 1H),
    4.31 (s, 1H), 3.84 (s, 2H), 3.63 (d, J = 8.6 Hz,
    7H), 3.60 (d, J = 6.0 Hz, 6H), 3.37 (s, 1H), 3.30
    (q, J = 7.9, 7.1 Hz, 2H), 3.16 (d, J = 12.7 Hz,
    1H), 2.81 (s, 1H), 2.70 (d, J = 10.3 Hz, 1H),
    2.55 (s, 7H), 1.81 (s, 3H), 1.63 (dd, J = 13.1, 6.4
    Hz, 1H), 1.51 (dd, J = 13.3, 6.3 Hz, 2H), 1.41
    (d, J = 6.8 Hz, 1H), 1.34 (t, J = 7.4 Hz, 1H),
    1.03 (d, J = 8.0 Hz, 2H), 0.75 (t, J = 7.3 Hz, 3H)
    844 3 4.6 517.4 0.73 δ 8.27 (s, 1H), 8.20 (d, J = 5.0 Hz, 1H), 7.91 (s,
    0H), 7.58 (s, 1H), 7.07-7.01 (m, 2H), 6.82 (d,
    J = 7.8 Hz, 1H), 6.41 (d, J = 7.7 Hz, 1H), 5.78
    (d, J = 8.5 Hz, 1H), 5.71-5.64 (m, 2H), 5.55
    (d, J = 17.1 Hz, 1H), 4.30 (s, 1H), 3.80 (d, J =
    30.8 Hz, 4H), 3.60 (s, 1H), 3.53 (s, 1H), 3.35-
    3.29 (m, 2H), 2.88 (s, 1H), 2.77 (d, J = 5.9 Hz,
    2H), 2.72 (s, 1H), 2.66 (d, J = 6.5 Hz, 2H), 2.55
    (s, 1H), 1.90 (s, 4H), 1.63 (d, J = 8.6 Hz, 1H),
    1.51 (s, 1H), 1.39 (s, 1H), 1.35-1.28 (m, 1H),
    1.01 (d, J = 12.4 Hz, 2H), 0.71 (t, J = 7.4 Hz,
    3H)
    845 2 21.2 439.0 0.92 δ 8.29 (s, 1H), 7.76 (s, 1H), 7.17 (s, 1H), 6.98
    (d, J = 7.8 Hz, 1H), 6.84 (d, J = 7.6 Hz, 1H),
    5.73 (s, 2H), 4.19 (s, 1H), 3.78 (s, 2H), 3.55 (s,
    1H), 3.40 (d, J = 33.6 Hz, 1H), 3.23 (s, 1H),
    3.16 (s, 1H), 2.99 (s, 1H), 2.55 (s, 9H), 2.05 (s,
    1H), 1.60-1.53 (m, 2H), 1.25 (q, J = 7.4 Hz,
    2H), 0.87 (t, J = 7.4 Hz, 3H)
    846 5 198 525.2 0.96 δ 7.82 (s, 1H), 7.76 (s, 1H), 7.58 (d, J = 8.6 Hz,
    1H), 7.20 (s, 1H), 6.99 (d, J = 8.3 Hz, 1H), 6.79
    (d, J = 7.7 Hz, 1H), 5.82 (d, J = 16.4 Hz, 1H),
    5.75 (d, J = 16.6 Hz, 1H), 4.49 (s, 1H), 4.13 (s,
    2H), 3.95-3.88 (m, 2H), 3.80-3.76 (m, 2H),
    3.56 (d, J = 29.7 Hz, 1H), 3.29 (t, J = 11.8 Hz,
    3H), 2.98 (d, J = 6.8 Hz, 2H), 2.92 (s, 1H), 2.51
    (s, 8H), 1.99 (d, J = 11.9 Hz, 2H), 1.78 (s, 3H),
    1.72 (d, J = 8.7 Hz, 1H), 1.62-1.49 (m, 4H),
    1.16 (t, J = 7.1 Hz, 3H), 0.82 (t, J = 7.2 Hz, 3H)
    847 5 187 539.1 0.81 δ 7.89 (d, J = 6.5 Hz, 1H), 7.76 (d, J = 1.2 Hz,
    1H), 7.64 (d, J = 8.6 Hz, 1H), 7.18 (d, J = 11.6
    Hz, 1H), 7.06-6.98 (m, 1H), 6.83 (d, J = 7.8
    Hz, 1H), 5.81 (d, J = 16.1 Hz, 1H), 5.74 (d, J =
    18.4 Hz, 1H), 4.50 (s, 1H), 4.09 (s, 2H), 3.83 (d,
    J = 8.8 Hz, 2H), 3.79-3.72 (m, 9H), 3.69 (s,
    1H), 3.47 (t, J = 11.9 Hz, 2H), 2.98 (d, J = 5.8
    Hz, 2H), 1.85 (s, 2H), 1.78 (s, 3H), 1.73 (d, J =
    12.3 Hz, 4H), 1.53 (dt, J = 13.9, 7.5 Hz, 2H),
    1.48 (s, 3H), 1.18 (q, J = 7.6 Hz, 2H), 0.82 (t,
    J = 7.3 Hz, 3H)
    848 3 33.0 497.9 1.02 δ 7.57 (s, 1H), 7.12 (s, 1H), 6.86 (d, J = 7.8 Hz,
    1H), 6.43 (d, J = 7.8 Hz, 1H), 5.74 (d, J = 8.2
    Hz, 1H), 5.69-5.62 (m, 3H), 5.57 (d, J = 16.9
    Hz, 1H), 4.32 (s, 1H), 3.86 (s, 3H), 3.74-3.66
    (m, 3H), 3.55 (s, 4H), 3.29 (t, J = 6.5 Hz, 2H),
    2.55 (s, 5H), 1.91 (s, 2H), 1.65 (dd, J = 13.3, 6.0
    Hz, 1H), 1.57 (s, 2H), 1.49 (s, 2H), 1.39 (dq, J =
    22.4, 7.8 Hz, 3H), 1.17 (s, 3H), 1.07 (q, J = 7.6
    Hz, 2H), 0.78 (t, J = 7.3 Hz, 3H)
    849 13 2030 440.1 0.97 δ 8.45 (d, J = 1.5 Hz, 1H), 7.73 (dd, J = 8.1, 2.0
    Hz, 1H), 7.61 (br t, J = 4.9 Hz, 1H), 7.55 (s, 1H),
    7.18 (d, J = 7.9 Hz, 1H), 5.70 (s, 2H), 5.65 (s,
    2H), 3.91 (s, 1H), 3.51-3.31 (m, 6H), 2.48-
    2.08 (m, 8H), 1.65-1.51 (m, 2H), 1.37-1.26
    (m, 2H), 0.91 (t, J = 7.3 Hz, 3H)
    850 13 1616 396.2 1.06 δ 8.45 (d, J = 1.5 Hz, 1H), 7.74 (dd, J = 7.8, 2.0
    Hz, 1H), 7.64-7.57 (m, 1H), 7.55 (s, 1H), 7.18
    (d, J = 7.9 Hz, 1H), 5.70 (s, 2H), 5.64 (s, 2H),
    2.71 (br t, J = 4.4 Hz, 4H), 2.30 (br s, 4H), 1.66-
    1.51 (m, 2H), 1.37-1.26 (m, 2H), 0.91 (t, J = 7.3
    Hz, 3H)
    851 13 1770 381.1 1.38 δ 8.52 (s, 1H), 7.96 (br d, J = 4.0 Hz, 1H), 7.83
    (dd, J = 8.0, 1.7 Hz, 1H), 7.61 (s, 1H), 7.25 (d,
    J = 8.0 Hz, 1H), 6.19 (br d, J = 2.1 Hz, 2H), 5.76
    (s, 2H), 3.86 (br s, 2H), 3.56-3.39 (m, 2H),
    2.68 (br s, 4H), 1.76 (br s, 4H), 1.59 (quin,
    J = 7.3 Hz, 2H), 1.36-1.25 (m, 2H), 0.90 (t,
    J = 7.4 Hz, 3H)
    852 13 3170 408.0 1.24 δ 8.52 (s, 1H), 7.90 (dd, J = 8.1, 1.7 Hz, 1H),
    7.78 (s, 1H), 7.48 (d, J = 7.9 Hz, 1H), 5.86 (s,
    2H), 4.09-3.88 (m, 2H), 3.65-3.55 (m, 2H),
    3.23-3.08 (m, 4H), 3.07-2.91 (m, 2H), 2.18
    (br d, J = 10.7 Hz, 1H), 1.82 (br d, J = 11.3 Hz,
    1H), 1.64 (quin, J = 7.2 Hz, 2H), 1.32 (sxt, J = 7.4
    Hz, 2H), 0.91 (t, J = 7.3 Hz, 3H).
    853 9 420 397.9 1.35 δ 8.46 (br s, 1H), 7.88 (br s, 1H), 7.81 (s, 1H),
    7.40 (d, J = 11.2 Hz, 1H), 7.27 (br d, J = 7.6 Hz,
    1H), 7.06 (t, J = 7.9 Hz, 1H), 5.90 (s, 2H), 4.04
    (s, 2H), 3.67 (br t, J = 8.1 Hz, 1H), 3.58 (br d,
    J = 6.1 Hz, 2H), 2.19-2.10 (m, 4H), 2.08 (s,
    1H), 1.85-1.69 (m, 2H), 1.60 (quin, J = 7.3 Hz,
    2H), 1.35-1.21 (m, 2H), 0.90 (t, J = 7.3 Hz, 3H)
    854 2 46.8 437.0 0.71 δ 7.55 (s, 1H), 6.99 (s, 1H), 6.80-6.74 (m, 1H),
    6.43 (d, J = 7.7 Hz, 1H), 5.65 (s, 1H), 5.59 (s,
    2H), 3.82 (s, 3H), 3.63 (s, 1H), 3.39 (d, J = 6.3
    Hz, 2H), 3.19 (d, J = 13.3 Hz, 1H), 2.91-2.83
    (m, 1H), 2.75-2.67 (m, 1H), 2.55 (s, 5H), 1.81
    (d, J = 1.7 Hz, 5H), 1.57 (d, J = 10.1 Hz, 1H),
    1.46 (p, J = 7.2 Hz, 2H), 1.17 (h, J = 7.5 Hz,
    2H), 0.83 (t, J = 7.3 Hz, 3H).
    855 11 640 411.1 1.36 δ 8.03 (t, J = 5.4 Hz, 1H), 8.02 (s, 1H), 7.51 (s,
    1H), 7.46 (s, 1H), 5.65 (s, 2H), 5.60 (s, 1H),
    3.88 (s, 3H), 3.64 (s, 1H), 3.18 (s, 2H), 3.16-
    3.10 (m, 1H), 2.10-1.99 (m, 2H), 1.92 (s, 3H),
    1.76-1.45 (m, 6H), 1.39 (h, J = 7.4 Hz, 2H),
    0.94 (t, J = 7.4 Hz, 3H)
    856 11 1160 441.3 1.28 δ 8.06 (d, J = 1.6 Hz, 1H), 7.56 (d, J = 1.5 Hz,
    1H), 7.47 (s, 1H), 5.65 (d, J = 6.9 Hz, 3H), 3.88
    (s, 2H), 3.82 (d, J = 12.6 Hz, 3H), 3.30-3.20
    (m, 2H), 3.18 (s, 1H), 3.00 (s, 1H), 2.64 (s, 1H),
    2.55 (s, 3H), 1.92 (s, 1H), 1.78 (d, J = 12.8 Hz,
    2H), 1.64 (p, J = 7.2 Hz, 2H), 1.39 (h, J = 7.4
    Hz, 2H), 1.34-1.22 (m, 2H), 0.94 (t, J = 7.4
    Hz, 3H).
    857 4, 6 11.5 519.9 1.48 δ 8.22 (br t, J = 6.0 Hz, 1H), 7.76 (s, 1H), 7.55
    (d, J = 8.5 Hz, 2H), 7.05 (d, J = 8.5 Hz, 2H), 6.86
    (d, J = 8.2 Hz, 1H), 6.78 (d, J = 2.1 Hz, 1H), 6.48
    (dd, J = 8.2, 2.1 Hz, 1H), 5.71 (s, 2H), 4.14 (s,
    2H), 3.74 (s, 3H), 3.59 (q, J = 6.5 Hz, 2H), 2.76
    (s, 2H), 1.59 (quin, J = 7.3 Hz, 2H), 1.34-1.23
    (m, 2H), 1.17 (s, 6H), 0.90 (t, J = 7.5 Hz, 3H)
    858 4, 6 10.8 532.2 1.38 δ 7.55 (s, 1H), 7.34 (d, J = 8.2 Hz, 2H), 6.93 (d,
    J = 8.5 Hz, 2H), 6.75 (d, J = 1.8 Hz, 1H), 6.52 (d,
    J = 8.2 Hz, 1H), 6.46 (br t, J = 5.2 Hz, 1H), 6.39
    (dd, J = 8.2, 2.1 Hz, 1H), 5.64 (s, 2H), 5.60 (s,
    2H), 3.71 (s, 3H), 3.42 (br d, J = 5.8 Hz, 2H),
    3.26 (td, J = 11.4, 2.0 Hz, 4H), 2.63-2.57 (m,
    1H), 1.80-1.73 (m, 2H), 1.55-1.46 (m, 2H),
    1.32-1.18 (m, 4H), 0.87 (t, J = 7.3 Hz, 3H).
    859 4, 6 13.1 531.9 1.41 δ 7.55 (s, 1H), 7.26 (d, J = 8.5 Hz, 2H), 6.92 (d,
    J = 8.5 Hz, 2H), 6.76 (d, J = 2.1 Hz, 1H), 6.53 (d,
    J = 8.2 Hz, 1H), 6.47 (br t, J = 4.0 Hz, 1H), 6.40
    (dd, J = 8.5, 2.1 Hz, 1H), 5.64 (s, 2H), 5.60 (s,
    2H), 3.80 (s, 3H), 3.46-3.35 (m, 1H), 2.69-
    2.60 (m, 2H), 2.02 (br t, J = 10.4 Hz, 2H), 1.72-
    1.64 (m, 2H), 1.50 (quin, J = 7.2 Hz, 2H), 1.42-
    1.33 (m, 2H), 1.22 (dq, J = 14.8, 7.4 Hz, 2H),
    0.86 (t, J = 7.3 Hz, 3H).
    860 4, 6 19.5 502.2 1.2 δ 7.55 (s, 1H), 7.32 (d, J = 8.4 Hz, 2H), 6.93 (d,
    J = 8.4 Hz, 2H), 6.75 (d, J = 1.9 Hz, 1H), 6.55-
    6.43 (m, 2H), 6.39 (dd, J = 8.5, 2.0 Hz, 1H), 5.65
    (s, 2H), 5.60 (s, 2H), 3.79 (s, 3H), 3.61 (s, 2H),
    3.18 (br d, J = 4.7 Hz, 1H), 2.06 (br d, J = 7.7 Hz,
    2H), 1.78-1.46 (m, 6H), 1.27-1.18 (m, 2H),
    0.87 (t, J = 7.3 Hz, 3H).
    861 3 1.2 470.3 0.82 δ 7.90-7.82 (m, 2H), 7.78 (s, 1H), 7.62-7.55
    (m, 1H), 7.17 (s, 1H), 6.95 (br d, J = 7.0 Hz, 1H),
    6.68 (d, J = 7.6 Hz, 1H), 5.84-5.70 (m, 2H),
    4.64-4.55 (m, 1H), 4.54-4.48 (m, 1H), 4.32 (s,
    2H), 4.25-4.21 (m, 2H), 4.20 (br s, 1H), 3.91
    (br d, J = 6.3 Hz, 2H), 3.80 (s, 3H), 3.23 (s, 3H),
    1.75-1.68 (m, 2H), 1.57-1.48 (m, 2H), 1.15
    (sxt, J = 7.4 Hz, 2H), 0.81 (t, J = 7.3 Hz, 3H).
    862 3 11.0 476.04 0.86 δ 7.57-7.55 (m, 1H), 6.98 (s, 1H), 6.76 (d,
    J = 7.7 Hz, 1H), 6.38 (d, J = 8.0 Hz, 1H), 5.71 (br
    d, J = 8.5 Hz, 1H), 5.68-5.62 (m, 3H), 5.59-
    5.52 (m, 1H), 4.45-4.37 (m, 1H), 4.36-4.27
    (m, 1H), 3.85 (s, 3H), 3.66 (s, 2H), 3.56 (br t,
    J = 12.5 Hz, 4H), 3.33-3.28 (m, J = 6.3 Hz, 2H),
    1.68-1.58 (m, 1H), 1.54-1.46 (m, 1H), 1.45-
    1.30 (m, 2H), 1.09-1.00 (m, 2H), 0.75 (t, J = 7.4
    Hz, 3H)
    863 3 9.5 456.2 0.77 δ 7.58-7.56 (m, 1H), 6.94 (s, 1H), 6.71 (br d,
    J = 7.6 Hz, 1H), 6.36 (d, J = 7.7 Hz, 1H), 5.70-
    5.65 (m, 2H), 5.65 (s, 2H), 5.58-5.52 (m, 1H),
    5.35-5.29 (m, 1H), 4.32 (br dd, J = 8.3, 4.3 Hz,
    1H), 4.16 (br d, J = 5.1 Hz, 1H), 3.85 (s, 3H),
    3.50 (s, 2H), 3.48-3.46 (m, 2H), 3.33-3.31 (m,
    2H), 2.72 (br t, J = 6.0 Hz, 2H), 1.68-1.59 (m,
    1H), 1.55-1.46 (m, 1H), 1.45-1.38 (m, 1H),
    1.37-1.30 (m, 1H), 1.09-0.99 (m, 2H), 0.76 (t,
    J = 7.2 Hz, 3H)
    864 7 216 426.17 0.8 δ 7.56-7.52 (m, 1H), 7.01 (s, 1H), 6.77 (br d,
    J = 7.6 Hz, 1H), 6.57 (br d, J = 7.6 Hz, 1H), 5.66
    (s, 2H), 5.55 (s, 2H), 3.83 (s, 3H), 3.63-3.54
    (m, 2H), 3.19-3.11 (m, 1H), 2.07-2.00 (m,
    2H), 1.75-1.65 (m, 2H), 1.65-1.56 (m, 1H),
    1.56-1.46 (m, 1H), 1.05 (t, J = 6.9 Hz, 3H)
    865 7 959 412.2 0.90 δ 7.56-7.52 (m, 1H), 6.99 (s, 1H), 6.76 (br d,
    J = 7.6 Hz, 1H), 6.62-6.53 (m, 1H), 5.65 (s,
    2H), 5.54 (s, 2H), 3.82 (s, 3H), 3.58 (q, J = 5.8
    Hz, 1H), 3.21 (s, 2H), 3.13-3.05 (m, 1H), 2.06-
    1.98 (m, 2H), 1.70-1.61 (m, 2H), 1.60-1.55
    (m, 1H), 1.54-1.45 (m, 1H)
    866 8 19.5 510.23 1.23 δ 9.28-9.02 (m, 1H), 7.84-7.77 (m, 1H), 7.75
    (s, 1H), 7.68-7.59 (m, 1H), 7.19 (s, 1H), 6.96
    (d, J = 7.7 Hz, 1H), 6.77 (d, J = 7.7 Hz, 1H), 5.80-
    5.66 (m, 2H), 4.60-4.49 (m, 1H), 3.99 (br s,
    2H), 3.78 (s, 3H), 3.42-3.40 (m, 2H), 2.99-
    2.97 (m, 1H), 2.22 (br d, J = 1.7 Hz, 1H), 2.07-
    2.00 (m, 1H), 1.84-1.80 (m, 1H), 1.76-1.71
    (m, 1H), 1.57-1.51 (m, 1H), 1.21-1.13 (m,
    2H), 0.83 (t, J = 7.3 Hz, 3H), 0.80-0.75 (m, 9H)
    867 12 302 470.4 0.71 δ 8.96 (s, 1H), 7.85 (s, 1H), 7.78 (s, 1H), 7.73
    (d, J = 8.1 Hz, 1H), 7.19 (d, J = 19.6 Hz, 1H),
    7.02 (d, J = 7.7 Hz, 1H), 6.84 (d, J = 7.7 Hz,
    1H), 5.82-5.70 (m, 2H), 4.46 (m, 1H), 4.16 (s,
    2H), 3.94 (d, J = 11.4 Hz, 2H), 3.80 (s, 2H),
    3.56 (s, 1H), 3.31 (t, J = 11.7 Hz, 3H), 2.00 (d,
    J = 12.5 Hz, 2H), 1.76 (p, J = 6.9 Hz, 2H), 1.62
    (tt, J = 13.2, 5.8 Hz, 4H), 0.80 (t, J = 7.4 Hz,
    3H)
    868 3 1.4 498.21 0.95 δ 7.59-7.51 (m, 1H), 6.94 (s, 1H), 6.79-6.66
    (m, 1H), 6.36 (d, J = 7.7 Hz, 1H), 5.69-5.65 (m,
    1H), 5.63 (s, 2H), 5.55-5.50 (m, 1H), 4.34-
    4.27 (m, 1H), 4.06 (quin, J = 6.1 Hz, 1H), 3.84 (s,
    3H), 3.51-3.49 (m, 4H), 3.30 (m, 2H), 2.78-
    2.73 (m, 2H), 1.90 (s, 1H), 1.66-1.59 (m, 1H),
    1.54-1.45 (m, 1H), 1.43-1.36 (m, 1H), 1.36-
    1.29 (m, 1H), 1.05-1.03 (m, 2H), 1.02 (s, 3H),
    1.01 (s, 3H), 0.74 (t, J = 7.3 Hz, 3H)
    869 12 897 484.1 1.13 δ 7.59 (s, 1H), 7.08 (s, 1H), 6.79 (d, J = 7.7 Hz,
    1H), 6.30 (d, J = 7.8 Hz, 1H), 5.67 (d, J = 17.3
    Hz, 1H), 5.62 (d, J = 10.8 Hz, 2H), 5.55 (d, J =
    17.2 Hz, 1H), 5.43 (d, J = 8.9 Hz, 1H), 4.16 (s,
    1H), 3.89 (m, 4H), 3.82 (s, 3H), 3.76 (s, 1H),
    3.70 (s, 1H), 3.25-3.15 (m, 4H), 1.88 (s, 1H),
    1.73 (m, 3H), 1.62 (m, 2H), 1.52 (m, 1H), 1.23
    (m, 2H), 0.67 (dd, J = 6.9, 2.8 Hz, 6H)
    870 9 1796 457.3 1.23 δ 8.45 (br t, J = 5.6 Hz, 1H), 7.81 (s, 1H), 7.21
    (d, J = 11.0 Hz, 1H), 7.16-7.07 (m, 1H), 6.96 (t,
    J = 7.9 Hz, 1H), 5.87 (s, 2H), 3.72 (t, J = 5.2 Hz,
    2H), 3.20-3.10 (m, 2H), 2.55 (s, 4H), 1.63-
    1.50 (m, 2H), 1.29-1.18 (m, 2H), 0.87 (t, J = 7.3
    Hz, 3H)
    871 12 61 469.9 1.04 δ 7.57 (s, 1H), 7.08 (s, 1H), 6.81 (d, J = 7.7 Hz,
    1H), 6.48 (d, J = 7.7 Hz, 1H), 5.71 (d, J = 16.9
    Hz, 1H), 5.63 (s, 2H), 5.48 (d, J = 16.9 Hz, 1H),
    4.30-4.18 (m, 1H), 3.85 (s, 3H), 3.80 (d, J =
    12.0 Hz, 2H), 3.71 (s, 2H), 3.60-3.52 (m, 1H),
    3.45-3.33 (m, 1H), 3.22 (t, J = 11.7 Hz, 1H),
    1.83-1.70 (m, 3H), 1.49 (m, 1H), 1.34-1.19
    (m, 4H), 1.09-1.03 (m, 2H), 1.03-0.94 (m,
    2H), 0.76 (t, J = 7.3 Hz, 3H)
    872 3 32.7 454.16 1.09 δ 7.57-7.54 (m, 1H), 7.05-7.01 (m, 1H), 6.76
    (d, J = 7.7 Hz, 1H), 6.40-6.37 (m, 1H), 5.66-
    5.64 (m, 1H), 5.63-5.61 (m, 2H), 5.57-5.52
    (m, 1H), 4.35-4.27 (m, 1H), 3.84 (s, 3H), 3.56
    (s, 2H), 3.29 (br t, J = 6.5 Hz, 2H), 3.18-3.06
    (m, 2H), 2.07-2.00 (m, 2H), 1.90 (s, 2H), 1.71-
    1.64 (m, 2H), 1.63-1.57 (m, 2H), 1.54-1.46
    (m, 2H), 1.44-1.37 (m, 1H), 1.36-1.30 (m,
    1H), 1.10-1.01 (m, 2H), 0.76 (t, J = 7.3 Hz, 3H)
    873 10 253 366.3 1.37 δ 7.57 (s, 1H), 7.03 (s, 1H), 6.82 (d, J = 7.9 Hz,
    1H), 6.59 (br s, 1H), 6.48 (d, J = 7.7 Hz, 1H),
    5.78 (br s, 2H), 5.63 (s, 2H), 3.98 (s, 2H), 3.84
    (s, 3H), 3.38 (br. m, 2H), 1.48 (quin, J = 7.3 Hz,
    2H), 1.25-1.12 (m, 2H), 0.85 (t, J = 7.3 Hz, 3H)
    874 14 2048 371.15 1.07 δ 7.32-7.17 (m, 2H), 6.96 (br d, J = 7.2 Hz, 2H),
    6.48-6.39 (m, 1H), 6.39-6.09 (m, 1H), 5.85-
    5.69 (m, 2H), 5.39-5.06 (m, 1H), 4.63-4.50
    (m, 1H), 4.46-4.40 (m, 2H), 4.39-4.31 (m,
    1H), 3.36-3.32 (m, 2H), 1.73-1.58 (m, 2H),
    1.41 (br d, J = 7.1 Hz, 2H), 1.08-0.89 (m, 2H),
    0.83-0.66 (m, 3H)
    875 14 58 423.91 1.03 δ 7.82-7.78 (m, 1H), 7.42 (br d, J = 8.5 Hz, 1H),
    7.37 (br d, J = 7.9 Hz, 2H), 7.01 (br d, J = 8.2 Hz,
    2H), 5.85 (s, 2H), 4.47-4.40 (m, 1H), 3.63-
    3.54 (m, 1H), 3.28 (br t, J = 6.3 Hz, 2H), 2.16-
    2.09 (m, 2H), 2.08-2.00 (m, 2H), 1.74 (br dd,
    J = 15.3, 8.9 Hz, 2H), 1.70-1.62 (m, 2H), 1.50-
    1.40 (m, 2H), 1.06-0.94 (m, 2H), 0.74 (t, J = 7.3
    Hz, 3H)
    876 14 1270 453.99 0.98 δ 7.63-7.56 (m, 1H), 7.26 (br d, J = 7.9 Hz, 2H),
    6.94 (br d, J = 7.9 Hz, 2H), 5.97 (br d, J = 8.2 Hz,
    1H), 5.69 (br d, J = 7.0 Hz, 2H), 5.59 (s, 2H),
    4.35-4.26 (m, 1H), 3.79 (br d, J = 11.6 Hz, 2H),
    3.69 (s, 1H), 3.28 (br t, J = 6.3 Hz, 2H), 3.25-
    3.16 (m, 2H), 2.67-2.58 (m, 1H), 1.79-1.70
    (m, 2H), 1.67-1.52 (m, 2H), 1.44-1.35 (m,
    2H), 1.31-1.20 (m, 2H), 1.08-0.96 (m, 2H),
    0.75 (t, J = 7.3 Hz, 3H)
    877 14 190 440.23 1.0 δ 7.83 (s, 1H), 7.49 (br d, J = 8.2 Hz, 1H), 7.39
    (br d, J = 8.2 Hz, 2H), 7.01 (br d, J = 7.9 Hz, 2H),
    5.89 (br s, 2H), 4.52-4.39 (m, 1H), 4.28 (s,
    2H), 4.22-4.18 (m, 2H), 4.18-4.14 (m, 1H),
    3.86 (br d, J = 7.6 Hz, 1H), 3.33-3.26 (m, 1H),
    3.26-3.21 (m, 1H), 3.20 (s, 3H), 1.70-1.63 (m,
    2H), 1.50-1.40 (m, 2H), 1.05-0.94 (m, 2H),
    0.74 (t, J = 7.3 Hz, 3H)
    878 14 1107 426.19 1.07 δ 7.60-7.56 (m, 1H), 7.16 (br d, J = 7.9 Hz, 2H),
    6.91 (br d, J = 8.2 Hz, 2H), 5.92 (br d, J = 8.5 Hz,
    1H), 5.75-5.63 (m, 2H), 5.59 (s, 2H), 4.35-
    4.26 (m, 1H), 4.17-4.10 (m, 1H), 3.33-3.27
    (m, 1H), 3.16 (s, 1H), 2.98 (s, 1H), 2.70 (br t,
    J = 6.1 Hz, 2H), 1.67-1.53 (m, 2H), 1.42-1.33
    (m, 2H), 1.02-0.94 (m, 2H), 0.73 (t, J = 7.3 Hz,
    3H)
    879 14 370 483.25 1.13 δ 7.92-7.87 (m, 1H), 7.86 (s, 1H), 7.55 (br d,
    J = 8.3 Hz, 1H), 7.34-7.28 (m, 3H), 7.01 (d,
    J = 8.0 Hz, 2H), 5.91 (br s, 2H), 4.54-4.44 (m,
    1H), 3.72-3.70 (m, 2H), 3.64 (br s, 2H), 3.34
    (dt, J = 10.9, 5.6 Hz, 2H), 3.30-3.24 (m, 2H),
    3.12 (br s, 2H), 1.71 (q, J = 6.1 Hz, 2H), 1.54-
    1.44 (m, 2H), 1.09-0.96 (m, 2H), 0.77 (t, J = 7.3
    Hz, 3H)
  • The foregoing detailed description of the invention includes passages that are chiefly or exclusively concerned with particular parts or aspects of the invention. It is to be understood that this is for clarity and convenience, that a particular feature may be relevant in more than just the passage in which it is disclosed, and that the disclosure herein includes all the appropriate combinations of information found in the different passages. Similarly, although the various figures and descriptions herein relate to specific embodiments of the invention, it is to be understood that where a specific feature is disclosed in the context of a particular figure or embodiment, such feature can also be used, to the extent appropriate, in the context of another figure or embodiment, in combination with another feature, or in the invention in general.
  • Further, while the present invention has been particularly described in terms of certain preferred embodiments, the invention is not limited to such preferred embodiments. Rather, the scope of the invention is defined by the appended claims.
  • ACRONYMS AND ABBREVIATIONS
  • Table C provides a list of acronyms and abbreviations used in this specification, along with their meanings.
  • TABLE C
    ACRONYM OR
    ABBREVIATION MEANING OR DEFINITION
    AIBN Azobisisobutyronitrile
    Aq. Aqueous
    Boc t-Butyloxycarbonyl
    BOP (Benzotriazol-1-yloxy)tris (dimethylamino)-
    phosphonium hexafluorophosphate (V)
    DBU 1,8-Diazabicyclo[5.4.0]undec-7-ene
    DCM Dichloromethane
    DIAD Diisopropyl azodicarboxylate
    DIPEA, DIEA N,N-diisopropylethylamine, also known as
    Hünig's base
    DMF N,N-dimethylformamide
    DMSO Dimethyl sulfoxide
    Fmoc Fluorenylmethyloxycarbonyl
    HATU Hexafluorophosphate Azabenzotriazole
    Tetramethyl Uronium; 1-[Bis (dimethylamino)-
    methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium
    3-oxide hexafluorophosphate
    Hunig's base See DIPEA, DIEA
    HPLC High pressure liquid chromatography
    LCMS, LC-MS, Liquid chromatography-mass spectrometry
    LC/MS
    MS Mass spectrometry
    MsCl Methanesylfonyl chloride, mesyl chloride
    NBS N-Bromosuccinimide
    NMR Nuclear magnetic resonance
    PEG Poly (ethylene glycol)
    PTFE Poly (tetraflurorethylene)
    RT (in the context of Room (ambient) temperature, circa 25° C.
    reaction conditions)
    RT (in context of Retention time, in min
    liquid
    chromatography)
    Sat. Saturated
    Soln Solution
    TBDPS tert-Butyldiphenylsilyl
    TEAA Triethylammonium acetate
    TFA Trifluoroacetic acid
    THF Tetrahydrofuran
  • REFERENCES
  • Full citations for the following references cited in abbreviated fashion by first author (or inventor) and date earlier in this specification are provided below. Each of these references is incorporated herein by reference for all purposes.
    • Akinbobuyi et al., Tetrahedron Lett. 2015, 56, 458, “Facile syntheses of functionalized toll-like receptor 7 agonists”.
    • Akinbobuyi et al., Bioorg. Med. Chem. Lett. 2016, 26, 4246, “Synthesis and immunostimulatory activity of substituted TLR7 agonists.”
    • Barberis et al., US 2012/0003298 A1 (2012).
    • Beesu et al., J. Med. Chem. 2017, 60, 2084, “Identification of High-Potency Human TLR8 and Dual TLR7/TLR8 Agonists in Pyrimidine-2,4-diamines.”
    • Berghofer et al., J. Immunol. 2007, 178, 4072, “Natural and Synthetic TLR7 Ligands Inhibit CpG-A- and CpG-C-Oligodeoxynucleotide-Induced IFN-α Production.”
    • Bonfanti et al., US 2014/0323441 A1 (2015) [2015a].
    • Bonfanti et al., US 2015/0299221 A1 (2015) [2015b].
    • Bonfanti et al., US 2016/0304531 A1 (2016).
    • Carson et al., US 2013/0202629 A1 (2013).
    • Carson et al., U.S. Pat. No. 8,729,088 B2 (2014).
    • Carson et al., U.S. Pat. No. 9,050,376 B2 (2015).
    • Carson et al., US 2016/0199499 A1 (2016).
    • Chan et al., Bioconjugate Chem. 2009, 20, 1194, “Synthesis and Immunological Characterization of Toll-Like Receptor 7 Agonistic Conjugates.”
    • Chan et al., Bioconjugate Chem. 2011, 22, 445, “Synthesis and Characterization of PEGylated Toll Like Receptor 7 Ligands.”
    • Chen et al., U.S. Pat. No. 7,919,498 B2 (2011).
    • Coe et al., U.S. Pat. No. 9,662,336 B2 (2017).
    • Cortez and Va, Medicinal Chem. Rev. 2018, 53, 481, “Recent Advances in Small-Molecule TLR7 Agonists for Drug Discovery”.
    • Cortez et al., US 2017/0121421 A1 (2017).
    • Cortez et al., U.S. Pat. No. 9,944,649 B2 (2018).
    • Dellaria et al., WO 2007/028129 A1 (2007).
    • Desai et al., U.S. Pat. No. 9,127,006 B2 (2015).
    • Ding et al., WO 2016/107536 A1 (2016).
    • Ding et al., US 2017/0273983 A1 (2017) [2017a].
    • Ding et al., WO 2017/076346 A1 (2017) [2017b].
    • Gadd et al., Bioconjugate Chem. 2015, 26, 1743, “Targeted Activation of Toll-Like Receptors: Conjugation of a Toll-Like Receptor 7 Agonist to a Monoclonal Antibody Maintains Antigen Binding and Specificity.”
    • Graupe et al., U.S. Pat. No. 8,993,755 B2 (2015).
    • Embrechts et al., J. Med. Chem. 2018, 61, 6236, “2,4-Diaminoquinazolines as Dual Toll Like Receptor (TLR) 7/8 Modulators for the Treatment of Hepatitis B Virus.”
    • Halcomb et al., U.S. Pat. No. 9,161,934 B2 (2015).
    • Hashimoto et al., US 2009/0118263 A1 (2009).
    • He et al., US 2019/0055246 A1 (2019) [2019a].
    • He et al., US 2019/0055247 A1 (2019) [2019b].
    • Hirota et al., U.S. Pat. No. 6,028,076 (2000).
    • Holldack et al., US 2012/0083473 A1 (2012).
    • Isobe et al., U.S. Pat. No. 6,376,501 B1 (2002).
    • Isobe et al., JP 2004137157 (2004).
    • Isobe et al., J. Med. Chem. 2006, 49 (6), 2088, “Synthesis and Biological Evaluation of Novel 9-Substituted-8-Hydroxyadenine Derivatives as Potent Interferon Inducers.”
    • Isobe et al., U.S. Pat. No. 7,521,454 B2 (2009) [2009a].
    • Isobe et al., US 2009/0105212 A1 (2009) [2009b].
    • Isobe et al., US 2011/0028715 A1 (2011).
    • Isobe et al., U.S. Pat. No. 8,148,371 B2 (2012).
    • Jensen et al., WO 2015/036044 A1 (2015).
    • Jones et al., U.S. Pat. No. 7,691,877 B2 (2010).
    • Jones et al., US 2012/0302598 A1 (2012).
    • Kasibhatla et al., U.S. Pat. No. 7,241,890 B2 (2007).
    • Koga-Yamakawa et al., Int. J. Cancer 2013, 132 (3), 580, “Intratracheal and oral administration of SM-276001: A selective TLR7 agonist, leads to antitumor efficacy in primary and metastatic models of cancer.”
    • Li et al., U.S. Pat. No. 9,902,730 B2 (2018).
    • Lioux et al., U.S. Pat. No. 9,295,732 B2 (2016).
    • Lund et al., Proc. Nat'l Acad. Sci (USA) 2004, 101 (15), 5598, “Recognition of single-stranded RNA viruses by Toll-like receptor 7.”
    • Maj et al., U.S. Pat. No. 9,173,935 B2 (2015).
    • McGowan et al., US 2016/0168150 A1 (2016) [2016a].
    • McGowan et al., U.S. Pat. No. 9,499,549 B2 (2016) [2016b].
    • McGowan et al., J. Med. Chem. 2017, 60, 6137, “Identification and Optimization of Pyrrolo[3,2-d]pyrimidine Toll-like Receptor 7 (TLR7) Selective Agonists for the Treatment of Hepatitis B.”
    • Musmuca et al., J. Chem. Information & Modeling 2009, 49 (7), 1777, “Small-Molecule Interferon Inducers. Toward the Comprehension of the Molecular Determinants through Ligand-Based Approaches.”
    • Nakamura et al., Bioorg. Med. Chem. Lett. 2013, 13, 669, “Synthesis and evaluation of 8-oxoadenine derivatives as potent Toll-like receptor agonists with high water solubility.”
    • Ogita et al., US 2007/0225303 A1 (2007).
    • Ota et al., WO 2019/124500 A1 (2019).
    • Pilatte et al., WO 2017/216293 A1 (2017).
    • Poudel et al., US 2019/0055243 A1 (2019) [2019a].
    • Poudel et al., US 2019/0055245 A1 (2019) [2019b].
    • Purandare et al., PCT Application Ser. No. PCT/US19/28697, filed Apr. 23, 2019.
    • Pryde, U.S. Pat. No. 7,642,350 B2 (2010).
    • Sato-Kaneko et al., JCI Insight 2017, 2, e93397, “Combination Immunotherapy with TLR Agonists and Checkpoint Inhibitors Suppresses Head and Neck Cancer”.
    • Smits et al., The Oncologist 2008, 13, 859, “The Use of TLR7 and TLR8 Ligands for the Enhancement of Cancer Immunotherapy”.
    • Vasilakos and Tomai, Expert Rev. Vaccines 2013, 12, 809, “The Use of Toll-like Receptor 7/8 Agonists as Vaccine Adjuvants”.
    • Vemejoul et al., US 2014/0141033 A1 (2014).
    • Young et al., US 2019/0055244 A1 (2019).
    • Yu et al., PLoS One 2013, 8 (3), e56514, “Toll-Like Receptor 7 Agonists: Chemical Feature Based Pharmacophore Identification and Molecular Docking Studies.”
    • Zhang et al., Immunity 2016, 45, 737, “Structural Analysis Reveals that Toll-like Receptor 7 Is a Dual Receptor for Guanosine and Single-Stranded RNA.”
    • Zhang et al., WO 2018/095426 A1 (2018)>
    • Zurawski et al., US 2012/0231023 A1 (2012).

Claims (16)

What is claimed is:
1. A method of treating a cancer, comprising administering to a patient suffering from such cancer a therapeutically effective combination of an anti-cancer immunotherapy agent and a compound having a structure according to formula I
Figure US20230140047A1-20230504-C00133
wherein
each X1 is independently N or CR2;
X2 is O, CH2, NH, S, or N(C1-C3 alkyl);
R1 is H, CH3(CH2)1-3, CH3(CH2)0-1O(CH2)2-3, CH3(CH2)0-3C(═O), CH3(CH2)0-1O(CH2)2-3C(═O),
Figure US20230140047A1-20230504-C00134
R2 is H, O(C1-C3 alkyl), C1-C3 alkyl, Cl, F, or CN;
R3 is H, halo, OH, CN, NH2, NH(C1-C5 alkyl), N(C1-C5 alkyl)2, NH(CH2)0-1(C3-C6 cycloalkyl), NH(C4-C8 bicycloalkyl), NH(C6-C10 spirocycloalkyl), N(C3-C6 cycloalkyl)2, NH(CH2)1-3(aryl), N((CH2)1-3(aryl))2, a cyclic amine moiety having the structure
Figure US20230140047A1-20230504-C00135
 (CH2)0-4, a 6-membered aromatic or heteroaromatic moiety or a 5-membered heteroaromatic moiety; wherein the 5-membered heteroaromatic and the 6-membered heteroaromatic moiety comprises from 1 to 4 heteroatoms independently selected from N, O, or S;
wherein
the alkyl, cycloalkyl, bicycloalkyl, spirocycloalkyl, cyclic amine, 6-membered aromatic or heteroaromatic, or 5-membered heteroaromatic moiety is optionally substituted with one or more substituents selected from OH, halo, CN, (C1-C3 alkyl), O(C1-C3 alkyl), C(═O)(Me), SO2(C1-C3 alkyl), C(═O)(Et), NH2, NH(Me), N(Me)2, NH(Et), N(Et)2, and N(C1-C3 alkyl), (CH2)1-2OH, (CH2)1-2OMe; and
wherein a CH2 group in the cycloalkyl, bicycloalkyl, spirocycloalkyl, or cyclic amine
moiety is optionally replaced by O, S, NH, N(C1-C3 alkyl), or N(Boc);
m is 0 or 1;
and
n is 1, 2, or 3;
or a pharmaceutically acceptable salt or tautomer thereof.
2. The method according to claim 1, wherein the group R1 of the compound or a pharmaceutically acceptable salt or tautomer thereof, is selected from the group consisting of
Figure US20230140047A1-20230504-C00136
3. The method according to claim 1, wherein the group R1 of the compound or a pharmaceutically acceptable salt or tautomer thereof, is
Figure US20230140047A1-20230504-C00137
4. The method according to claim 1, wherein the moiety of the compound or a pharmaceutically acceptable salt or tautomer thereof,
Figure US20230140047A1-20230504-C00138
is selected from the group consisting of
Figure US20230140047A1-20230504-C00139
5. The method according to claim 1, wherein the moiety of the compound or a pharmaceutically acceptable salt or tautomer thereof,
Figure US20230140047A1-20230504-C00140
6. The method according to claim 1, wherein the group R3 of the compound or a pharmaceutically acceptable salt or tautomer thereof, is selected from the group consisting of Cl, H,
Figure US20230140047A1-20230504-C00141
Figure US20230140047A1-20230504-C00142
Figure US20230140047A1-20230504-C00143
7. The method according to claim 1, wherein R3 of the compound or a pharmaceutically acceptable salt or tautomer thereof, is a moiety
Figure US20230140047A1-20230504-C00144
selected from the group consisting of
Figure US20230140047A1-20230504-C00145
8. The method according to claim 1, wherein the compound or a pharmaceutically acceptable salt or tautomer thereof, has a structure according to formula (I′)
Figure US20230140047A1-20230504-C00146
9. The method according to claim 1, wherein the compound or a pharmaceutically acceptable salt or tautomer thereof, has a structure according to formula Ia
Figure US20230140047A1-20230504-C00147
10. The method according to claim 9, wherein R1 of the compound or a pharmaceutically acceptable salt or tautomer thereof, is
Figure US20230140047A1-20230504-C00148
11. The method according to claim 1, wherein the compound or a pharmaceutically acceptable salt or tautomer thereof, has a structure according to formula Ib
Figure US20230140047A1-20230504-C00149
12. The method according to claim 11, wherein
Figure US20230140047A1-20230504-C00150
13. The method according to claim 1, wherein the compound is covalently bonded to a poly(ethylene glycol) moiety between 2 kDa and 40 kDa in size.
14. The method according to claim 1, wherein the anti-cancer immunotherapy agent is an antagonistic anti-CTLA-4, anti-PD-1, or anti-PD-L1 antibody.
15. The method according to claim 1, wherein the cancer is lung cancer (including non-small cell lung cancer), pancreatic cancer, kidney cancer, head and neck cancer, lymphoma (including Hodgkin's lymphoma), skin cancer (including melanoma and Merkel skin cancer), urothelial cancer (including bladder cancer), gastric cancer, hepatocellular cancer, or colorectal cancer.
16. The method according to claim 14, wherein the anti-cancer immunotherapy agent is ipilimumab, nivolumab, or pembrolizumab.
US18/081,558 2018-08-03 2022-12-14 1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR Pending US20230140047A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/081,558 US20230140047A1 (en) 2018-08-03 2022-12-14 1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862714238P 2018-08-03 2018-08-03
US16/527,802 US11554120B2 (en) 2018-08-03 2019-07-31 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor
US18/081,558 US20230140047A1 (en) 2018-08-03 2022-12-14 1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/527,802 Continuation US11554120B2 (en) 2018-08-03 2019-07-31 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor

Publications (1)

Publication Number Publication Date
US20230140047A1 true US20230140047A1 (en) 2023-05-04

Family

ID=67742964

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/527,802 Active 2040-11-18 US11554120B2 (en) 2018-08-03 2019-07-31 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor
US16/529,457 Active 2040-07-11 US11400094B2 (en) 2018-08-03 2019-08-01 2H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor
US18/081,558 Pending US20230140047A1 (en) 2018-08-03 2022-12-14 1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US16/527,802 Active 2040-11-18 US11554120B2 (en) 2018-08-03 2019-07-31 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor
US16/529,457 Active 2040-07-11 US11400094B2 (en) 2018-08-03 2019-08-01 2H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor

Country Status (19)

Country Link
US (3) US11554120B2 (en)
EP (2) EP3830089B1 (en)
JP (4) JP7337909B2 (en)
KR (2) KR20210039411A (en)
CN (2) CN112752759A (en)
AR (1) AR115885A1 (en)
AU (1) AU2019314444A1 (en)
BR (1) BR112021001503A2 (en)
CA (1) CA3108512A1 (en)
CL (1) CL2021000216A1 (en)
CO (1) CO2021000942A2 (en)
EA (1) EA202190442A1 (en)
ES (2) ES2941768T3 (en)
IL (1) IL280473A (en)
MX (1) MX2021001311A (en)
PE (1) PE20211812A1 (en)
SG (1) SG11202101031XA (en)
TW (1) TW202019933A (en)
WO (2) WO2020028608A1 (en)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2017292934B2 (en) 2016-07-07 2024-04-04 Bolt Biotherapeutics, Inc. Antibody adjuvant conjugates
CA3047403A1 (en) 2016-12-22 2018-06-28 Cadent Therapeutics, Inc. Nmda receptor modulators and uses thereof
US11554120B2 (en) * 2018-08-03 2023-01-17 Bristol-Myers Squibb Company 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor
CA3108519A1 (en) 2018-08-03 2020-04-30 Cadent Therapeutics, Inc. Heteroaromatic nmda receptor modulators and uses thereof
WO2020190725A1 (en) 2019-03-15 2020-09-24 Bolt Biotherapeutics, Inc. Immunoconjugates targeting her2
KR20220132589A (en) * 2020-01-27 2022-09-30 브리스톨-마이어스 스큅 컴퍼니 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists
CN115151546A (en) * 2020-01-27 2022-10-04 百时美施贵宝公司 C3-substituted 1H-pyrazolo [4,3-d ] pyrimidine compounds as Toll-like receptor 7 (TLR 7) agonists
KR20220132591A (en) 2020-01-27 2022-09-30 브리스톨-마이어스 스큅 컴퍼니 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists
JP2023512208A (en) 2020-01-27 2023-03-24 ブリストル-マイヤーズ スクイブ カンパニー 1H-pyrazolo[4,3-d]pyrimidine compounds as Toll-like receptor 7 (TLR7) agonists
KR20220132593A (en) 2020-01-27 2022-09-30 브리스톨-마이어스 스큅 컴퍼니 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists
US20230041738A1 (en) 2020-01-27 2023-02-09 Bristol-Myers Squibb Company 1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS
EP4097101A1 (en) * 2020-01-27 2022-12-07 Bristol-Myers Squibb Company 1h-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (tlr7) agonists
JP2023512204A (en) 2020-01-27 2023-03-24 ブリストル-マイヤーズ スクイブ カンパニー 1H-pyrazolo[4,3-d]pyrimidine compounds as Toll-like receptor 7 (TLR7) agonists
KR20220132592A (en) 2020-01-27 2022-09-30 브리스톨-마이어스 스큅 컴퍼니 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists
US20230219962A1 (en) * 2020-06-04 2023-07-13 Merck Sharp & Dohme Llc PYRAZOLO[4,3-d]PYRIMIDINE DERIVATIVES AND METHODS OF USE THEREOF FOR THE TREATMENT OF CELLULAR PROLIFERATIVE DISORDERS
WO2022156678A1 (en) * 2021-01-20 2022-07-28 上海维申医药有限公司 Macrocyclic tlr7 agonist, and preparation method therefor, pharmaceutical composition thereof and use thereof
CN115337406A (en) * 2021-05-13 2022-11-15 清华大学 Antibody drug conjugate and preparation method and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11554120B2 (en) * 2018-08-03 2023-01-17 Bristol-Myers Squibb Company 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor

Family Cites Families (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1981001145A1 (en) 1979-10-18 1981-04-30 Univ Illinois Hydrolytic enzyme-activatible pro-drugs
US4698420A (en) 1985-02-25 1987-10-06 Xoma Corporation Antibody hybrid molecules and process for their preparation
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
EP0882727B9 (en) 1996-07-03 2005-06-15 Sumitomo Pharmaceuticals Company, Limited Novel purine derivatives
TW572758B (en) 1997-12-22 2004-01-21 Sumitomo Pharma Type 2 helper T cell-selective immune response inhibitors comprising purine derivatives
US7425541B2 (en) 1998-12-11 2008-09-16 Medarex, Inc. Enzyme-cleavable prodrug compounds
CA2589418A1 (en) 1999-08-24 2001-03-01 Medarex, Inc. Human ctla-4 antibodies and their uses
AU2001286727A1 (en) 2000-08-24 2002-03-04 Coulter Pharmaceutical, Inc. Prodrugs activated by plasmin and their use in cancer chemotherapy
US7571211B1 (en) 2002-04-15 2009-08-04 Lightwaves Systems, Inc. Method for routing messages over a network based on location
ES2314042T3 (en) 2001-04-17 2009-03-16 Dainippon Sumitomo Pharma Co., Ltd. NEW DERIVATIVES OF ADENINA.
CN1463270A (en) 2001-05-31 2003-12-24 梅达莱克斯公司 Disulfide prodrugs and linkers and stablizers useful therefore
ES2364452T3 (en) 2001-06-11 2011-09-02 Medarex, Inc. METHOD FOR DESIGNING PROFESSIONAL COMPOUNDS ACTIVATED BY CD10.
US7091186B2 (en) 2001-09-24 2006-08-15 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
EP1440072A4 (en) 2001-10-30 2005-02-02 Conforma Therapeutic Corp Purine analogs having hsp90-inhibiting activity
NZ534174A (en) 2002-01-09 2007-03-30 Medarex Inc An isolated human monoclonal antibody which binds to human CD30
ES2387388T3 (en) 2002-09-27 2012-09-21 Dainippon Sumitomo Pharma Co., Ltd. Adenine compound and use thereof
JP2004137157A (en) 2002-10-16 2004-05-13 Sumitomo Pharmaceut Co Ltd Medicine comprising new adenine derivative as active ingredient
ES2340494T3 (en) 2003-04-15 2010-06-04 Glaxosmithkline Llc MUTANTS OF REPLACEMENT OF HUMAN IL-18 AND ITS CONJUGATES.
EP1648510B1 (en) 2003-07-22 2013-06-19 Bayer Pharma Aktiengesellschaft Rg1 antibodies and uses thereof
WO2005051976A2 (en) 2003-11-20 2005-06-09 Ansata Therapeutics, Inc. Protein and peptide ligation processes and one-step purification processes
CN102838675B (en) 2003-12-10 2014-07-30 梅达雷克斯有限责任公司 Ip-10 antibodies and their uses
JP5064037B2 (en) 2004-02-23 2012-10-31 ジェネンテック, インコーポレイテッド Heterocyclic self-destructive linkers and conjugates
AU2005227326B2 (en) 2004-03-24 2009-12-03 Xencor, Inc. Immunoglobulin variants outside the Fc region
US20070225303A1 (en) 2004-03-26 2007-09-27 Haruhisa Ogita 8-Oxoadenine Compound
NZ550934A (en) 2004-05-19 2010-05-28 Medarex Inc Chemical linkers and conjugates thereof
US7691962B2 (en) 2004-05-19 2010-04-06 Medarex, Inc. Chemical linkers and conjugates thereof
NZ580115A (en) 2004-09-23 2010-10-29 Genentech Inc Cysteine engineered antibody light chains and conjugates
CA2585557C (en) 2004-10-28 2009-08-18 Pharmacia & Upjohn Company Llc Pyrazolo[4,3-d] pyrimidine derivatives useful as pde-5 inhibitors
CA2598454C (en) 2005-02-18 2013-04-09 Medarex, Inc. Monoclonal antibodies against prostate specific membrane antigen (psma) lacking in fucosyl residues
DK1851250T3 (en) 2005-02-18 2012-07-09 Medarex Inc HUMANT MONOCLONAL ANTIBODY AGAINST PROSTATE-SPECIFIC MEMBRANE ANTIGEN (PSMA)
US7714016B2 (en) 2005-04-08 2010-05-11 Medarex, Inc. Cytotoxic compounds and conjugates with cleavable substrates
AU2006242920A1 (en) 2005-05-04 2006-11-09 Pfizer Limited 2-amido-6-amino-8-oxopurine derivatives as Toll-Like receptor modulators for the treatment of cancer and viral infections, such as hepatitis C
DK2161336T4 (en) 2005-05-09 2017-04-24 Ono Pharmaceutical Co Human monoclonal antibodies for programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapies
AU2006262232A1 (en) 2005-06-20 2007-01-04 Medarex, Inc. CD19 antibodies and their uses
AU2006265108C1 (en) 2005-07-01 2013-01-17 E. R. Squibb & Sons, L.L.C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
LT1912671T (en) 2005-07-18 2017-12-11 Seattle Genetics, Inc. Beta-glucuronide-linker drug conjugates
CA2620933A1 (en) 2005-09-02 2007-03-08 Pfizer Inc. Hydroxy substituted 1h-imidazopyridines and methods
US20090105212A1 (en) 2005-09-22 2009-04-23 Dainippon Sumitomo Pharma Co., Ltd. a corporation of Japan Novel adenine compound
JPWO2007034817A1 (en) 2005-09-22 2009-03-26 大日本住友製薬株式会社 New adenine compounds
EP1934261B1 (en) 2005-09-26 2014-10-29 Medarex, L.L.C. Human monoclonal antibodies to cd70
AU2006294554B2 (en) 2005-09-26 2013-03-21 E. R. Squibb & Sons, L.L.C. Antibody-drug conjugates and methods of use
KR20080068084A (en) 2005-10-26 2008-07-22 메다렉스, 인코포레이티드 Methods and compounds for preparing cc-1065 analogs
WO2007059404A2 (en) 2005-11-10 2007-05-24 Medarex, Inc. Duocarmycin derivatives as novel cytotoxic compounds and conjugates
NZ568015A (en) 2005-12-08 2012-03-30 Medarex Inc Human monoclonal antibodies to O8E
EP1957539B1 (en) 2005-12-08 2013-04-17 Medarex, Inc. Human monoclonal antibodies to protein tyrosine kinase 7 (ptk7) and their use
ES2374455T3 (en) 2006-02-17 2012-02-16 Pfizer Limited DERIVATIVES OF 3-DEAZAPURINZA AS MODULATORS OF TLR7.
CN101479379B (en) 2006-06-29 2012-01-18 利兰·斯坦福青年大学托管委员会 Cell-free synthesis of proteins containing unnatural amino acids
EP2054436A1 (en) 2006-08-18 2009-05-06 Novo Nordisk Health Care AG Transglutaminase variants with improved specificity
KR20090057072A (en) 2006-09-08 2009-06-03 암브룩스, 인코포레이티드 Site specific incorporation of non-natural amino acids by vertebrate cells
PL2486941T3 (en) 2006-10-02 2017-08-31 E. R. Squibb & Sons, L.L.C. Human antibodies that bind CXCR4 and uses thereof
TWI447124B (en) 2006-12-01 2014-08-01 Medarex Llc Human antibodies that bind cd22 and uses thereof
UY30776A1 (en) 2006-12-21 2008-07-03 Medarex Inc CD44 ANTIBODIES
TWI412367B (en) 2006-12-28 2013-10-21 Medarex Llc Chemical linkers and cleavable substrates and conjugates thereof
PL2125007T3 (en) 2007-02-07 2014-07-31 Univ California Conjugates of synthetic tlr agonists and uses therefor
CN101616911A (en) 2007-02-21 2009-12-30 梅达莱克斯公司 Chemical linkers and conjugate thereof with single amino acids
BRPI0808014A2 (en) 2007-02-22 2014-06-17 Novo Nordisk Healthcare Ag TRANSGLUTAMINASE VARIANTS WITH IMPROVED SPECIFICITY
PE20081887A1 (en) 2007-03-20 2009-01-16 Dainippon Sumitomo Pharma Co NEW ADENINE COMPOUND
CA2680783C (en) 2007-03-23 2012-04-24 Amgen Inc. Heterocyclic compounds and their uses
NZ582090A (en) 2007-06-29 2012-05-25 Gilead Sciences Inc Purine derivatives and their use as modulators of toll-like receptor 7
ES2359123T3 (en) 2007-08-03 2011-05-18 Pfizer Limited IMIDAZOPIRIDINONES.
WO2009026274A1 (en) 2007-08-22 2009-02-26 Medarex, Inc. Site-specific attachment of drugs or other agents to engineered antibodies with c-terminal extensions
CA2700860C (en) 2007-10-01 2016-07-19 Jonathan A. Terrett Human antibodies that bind mesothelin, and uses thereof
PT2364314E (en) 2008-12-09 2014-06-09 Gilead Sciences Inc Modulators of toll-like receptors
US8940501B2 (en) 2009-01-30 2015-01-27 Whitehead Institute For Biomedical Research Methods for ligation and uses thereof
US8729088B2 (en) 2009-02-11 2014-05-20 The Regents Of The University Of California Toll-like receptor modulators and treatment of diseases
US8962652B2 (en) 2009-10-22 2015-02-24 Gilead Sciences, Inc. Derivatives of purine or deazapurine useful for the treatment of (inter alia) viral infections
CA2797315C (en) 2010-04-30 2018-09-11 Telormedix Sa Phospholipid drug analogs
CN103118682A (en) 2010-04-30 2013-05-22 加利福尼亚大学校务委员会 Uses of phospholipid conjugates of synthetic TLR7 agonists
WO2011134669A1 (en) 2010-04-30 2011-11-03 Telormedix Sa Methods for inducing an immune response
WO2012038058A1 (en) 2010-09-21 2012-03-29 Telormedix Sa Treatment of conditions by toll-like receptor modulators
CA2930801C (en) 2010-11-05 2019-05-28 Rinat Neuroscience Corporation Engineered polypeptide conjugates and methods for making thereof using transglutaminase
WO2012122396A1 (en) 2011-03-08 2012-09-13 Baylor Research Institute Novel vaccine adjuvants based on targeting adjuvants to antibodies directly to antigen-presenting cells
CA2843274A1 (en) 2011-07-29 2013-02-07 Selecta Biosciences, Inc. Synthetic nanocarriers that generate humoral and cytotoxic t lymphocyte (ctl) immune responses
EA033830B1 (en) 2011-11-09 2019-11-29 Janssen Sciences Ireland Uc Adenine derivatives as activators of tlr7 toll-like receptors
SI2872515T1 (en) 2012-07-13 2016-10-28 Janssen Sciences Ireland Uc Macrocyclic purines for the treatment of viral infections
KR20150046267A (en) 2012-08-24 2015-04-29 글락소스미스클라인 엘엘씨 Pyrazolopyrimidine compounds
CN104837840B (en) 2012-10-10 2017-08-08 爱尔兰詹森科学公司 Pyrrolo- [3,2 d] pyrimidine derivatives for treating virus infection and other diseases
EP2732825B1 (en) 2012-11-19 2015-07-01 Invivogen Conjugates of a TLR7 and/or TLR8 agonist and a TLR2 agonist
DK2956173T3 (en) 2013-02-14 2017-07-17 Bristol Myers Squibb Co TUBULYSIS RELATIONSHIPS, METHODS OF PRODUCING AND USING THEREOF
US9295732B2 (en) 2013-02-22 2016-03-29 Invivogen Conjugated TLR7 and/or TLR8 and TLR2 polycationic agonists
CA2902837C (en) 2013-03-29 2021-09-07 Janssen Sciences Ireland Uc Macrocyclic deaza-purinones for the treatment of viral infections
KR102311234B1 (en) 2013-06-27 2021-10-12 얀센 사이언시즈 아일랜드 언리미티드 컴퍼니 Pyrrolo[3,2-d]pyrimidine derivatives for the treatment of viral infections and other diseases
US20160199499A1 (en) 2013-08-16 2016-07-14 The Regents Of The University Of California Uses of phospholipid conjugates of synthetic tlr7 agonists
WO2015036044A1 (en) 2013-09-13 2015-03-19 Telormedix Sa Cationic lipid vehicles for delivery of tlr7 agonists for specific targeting of human cd14+ monocytes in whole blood
US9944649B2 (en) 2014-05-01 2018-04-17 Novartis Ag Compounds and compositions as toll-like receptor 7 agonists
CN106232603B (en) 2014-05-01 2019-07-05 诺华股份有限公司 Compound and composition as 7 agonist of TOLL- sample receptor
US9695166B2 (en) * 2014-05-05 2017-07-04 Global Blood Therapeutics, Inc. Pyrazolopyridine pyrazolopyrimidine and related compounds
CN110938076B (en) 2014-08-15 2021-08-10 正大天晴药业集团股份有限公司 Pyrrolopyrimidines as TLR7 agonists
CN105732635A (en) 2014-12-29 2016-07-06 南京明德新药研发股份有限公司 Toll-like receptor 7 agonist
WO2016144608A1 (en) 2015-03-10 2016-09-15 Bristol-Myers Squibb Company Antibodies conjugatable by transglutaminase and conjugates made therefrom
US20180265851A1 (en) 2015-10-02 2018-09-20 Bristol-Myers Squibb Company Transglutaminase variants for conjugating antibodies
MA44334A (en) 2015-10-29 2018-09-05 Novartis Ag ANTIBODY CONJUGATES INCLUDING A TOLL-TYPE RECEPTOR AGONIST
JP6524355B2 (en) 2015-11-05 2019-06-05 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッドChia Tai Tianqing Pharmaceutical Group Co., Ltd. 7- (thiazol-5-yl) pyrrolopyrimidine compounds as TLR7 agonists
US20170166384A1 (en) 2015-12-11 2017-06-15 Graphic Packaging International, Inc. Container with absorption features
EP3472147B1 (en) 2016-06-16 2020-06-17 Janssen Pharmaceutica NV Azabenzimidazole derivatives as pi3k beta inhibitors
KR102497701B1 (en) 2016-09-13 2023-02-09 에프. 호프만-라 로슈 아게 Combination therapy of a TLR7 agonist and an HBV capsid assembly inhibitor
US11117898B2 (en) 2016-11-28 2021-09-14 Jiangsu Hengrui Medicine Co., Ltd. Pyrazolo-heteroaryl derivative, preparation method and medical use thereof
US10494370B2 (en) 2017-08-16 2019-12-03 Bristol-Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having a pyridine or pyrazine moiety, conjugates thereof, and methods and uses therefor
US10472361B2 (en) 2017-08-16 2019-11-12 Bristol-Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having a benzotriazole moiety, conjugates thereof, and methods and uses therefor
US10508115B2 (en) 2017-08-16 2019-12-17 Bristol-Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having heteroatom-linked aromatic moieties, conjugates thereof, and methods and uses therefor
US10487084B2 (en) 2017-08-16 2019-11-26 Bristol-Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having a heterobiaryl moiety, conjugates thereof, and methods and uses therefor
US10457681B2 (en) 2017-08-16 2019-10-29 Bristol_Myers Squibb Company Toll-like receptor 7 (TLR7) agonists having a tricyclic moiety, conjugates thereof, and methods and uses therefor
RU2020124007A (en) 2017-12-21 2022-01-21 Сумитомо Дайниппон Фарма Ко., Лтд. COMBINATION DRUG INCLUDING TLR7 AGONIST
WO2019209811A1 (en) 2018-04-24 2019-10-31 Bristol-Myers Squibb Company Macrocyclic toll-like receptor 7 (tlr7) agonists

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11554120B2 (en) * 2018-08-03 2023-01-17 Bristol-Myers Squibb Company 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses therefor

Also Published As

Publication number Publication date
ES2941520T3 (en) 2023-05-23
US20200039986A1 (en) 2020-02-06
CN112752759A (en) 2021-05-04
EP3830089B1 (en) 2023-03-08
JP2023182569A (en) 2023-12-26
US20200038403A1 (en) 2020-02-06
SG11202101031XA (en) 2021-02-25
IL280473A (en) 2021-03-01
US11554120B2 (en) 2023-01-17
AR115885A1 (en) 2021-03-10
JP7337907B2 (en) 2023-09-04
CN112513046A (en) 2021-03-16
EP3830089A1 (en) 2021-06-09
EP3830090B1 (en) 2023-03-01
KR20210039411A (en) 2021-04-09
CL2021000216A1 (en) 2021-06-04
US11400094B2 (en) 2022-08-02
WO2020028610A1 (en) 2020-02-06
EP3830090A1 (en) 2021-06-09
ES2941768T3 (en) 2023-05-25
CA3108512A1 (en) 2020-02-06
TW202019933A (en) 2020-06-01
EA202190442A1 (en) 2021-05-19
BR112021001503A2 (en) 2021-04-27
JP2023179420A (en) 2023-12-19
WO2020028608A1 (en) 2020-02-06
CO2021000942A2 (en) 2021-02-17
AU2019314444A1 (en) 2021-03-18
MX2021001311A (en) 2021-04-13
KR20210039410A (en) 2021-04-09
JP7337909B2 (en) 2023-09-04
JP2021532162A (en) 2021-11-25
JP2021533135A (en) 2021-12-02
PE20211812A1 (en) 2021-09-14

Similar Documents

Publication Publication Date Title
US20230140047A1 (en) 1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR
ES2904684T3 (en) 6-amino-7,9-dihydro-8H-purin-8-one derivatives as immunostimulant toll-like receptor 7 (TLR7) agonists
JP2020152726A (en) ANTIBODY DRUG CONJUGATES WITH CELL-PERMEABLE Bcl-xL INHIBITORS
US20200024279A1 (en) Toll-like receptor 7 (tlr7) agonists having a benzotriazole moiety, conjugates thereof, and methods and uses therefor
TW202106691A (en) Camptothecin derivatives
JP2020531469A (en) Toll-like receptor 7 (TLR7) agonists with tricyclic groups, their conjugates, and their methods and uses
JP2020143062A (en) Bcl-xL INHIBITORY COMPOUNDS HAVING LOW CELL PERMEABILITY AND ANTIBODY DRUG CONJUGATES INCLUDING THE SAME
JP2020531466A (en) 6-Amino-7,9-dihydro-8H-purine-8-one derivative as an immunostimulatory toll-like receptor 7 (TLR7) agonist
TW202016081A (en) Small molecule modulators of human sting, conjugates and therapeutic applications
EA027925B1 (en) Enediyne compounds, conjugates thereof, and methods for making and using the same
JP7023933B2 (en) Seco-cyclopropyrroloindole compound, its antibody-drug conjugate, and how to make and use it.
TW201920128A (en) Process for the preparation of TUBULYSINS and intermediates thereof

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: BRISTOL-MYERS SQUIBB COMPANY, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:POUDEL, YAM B.;GANGWAR, SANJEEV;HE, LIQI;AND OTHERS;SIGNING DATES FROM 20190916 TO 20190930;REEL/FRAME:062758/0519

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER