WO2023009554A1 - Methods to reduce adverse effects of gene or biologics therapy - Google Patents

Methods to reduce adverse effects of gene or biologics therapy Download PDF

Info

Publication number
WO2023009554A1
WO2023009554A1 PCT/US2022/038397 US2022038397W WO2023009554A1 WO 2023009554 A1 WO2023009554 A1 WO 2023009554A1 US 2022038397 W US2022038397 W US 2022038397W WO 2023009554 A1 WO2023009554 A1 WO 2023009554A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
linker
c6alkyl
formula
immunoglobulin
Prior art date
Application number
PCT/US2022/038397
Other languages
French (fr)
Inventor
Milind Deshpande
Jesse Jingyang CHEN
Mark George Saulnier
Srinivasa Karra
Jason Allan Wiles
Kevin Tyler SPROTT
Soumya Ray
Original Assignee
Avilar Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Avilar Therapeutics, Inc. filed Critical Avilar Therapeutics, Inc.
Publication of WO2023009554A1 publication Critical patent/WO2023009554A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • 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
    • A61K47/54Medicinal 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 compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • 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
    • A61K47/54Medicinal 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 compound
    • A61K47/55Medicinal 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 compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • This invention provides methods to modulate an immune response to a therapeutic, for example gene therapy, by administering an immunoglobulin degrading molecule that has an asialoglycoprotein receptor (ASGPR) Binding Ligand bound to an Immunoglobulin Targeting Ligand or a pharmaceutically acceptable salt thereof.
  • ASGPR asialoglycoprotein receptor
  • Elevated immune responses to treatments has been most prevalent in gene therapies and recombinant therapeutic protein treatments, mitigating a promising area of medical advancement for a number of disorders.
  • gene therapies rely on viral vectors, such as adenovirus vectors and adeno-associated virus vectors (AAV), for delivering gene therapy targets to cells.
  • viral vectors such as adenovirus vectors and adeno-associated virus vectors (AAV)
  • AAV adeno-associated virus vectors
  • Many patients have pre-existing immunity to the adenovirus or AAV vector which can be further triggered by administration of the vector.
  • the increasing immune response from the patient results in the escalated removal of the vector before they can reach their target cells.
  • ongoing therapy requires either increasingly larger doses of the vector being delivered, risking damage to the patient's liver, or discontinuation of the therapy due to inefficacy.
  • many patients suffering from disorders, such as genetic disorders or cancers are unable to receive ongoing or repeat treatments using such viral vectors that might
  • Factors influencing immunogenicity include recombinant protein dependent factors such as the primary amino acid sequence, and glycosylation patterns (Hermeling et al. (2004) Structure-immunogenicity relationships of therapeutic proteins. Pharm. Res. 21, 897-903).
  • Other factors influencing immunogenicity include treatment dependent factors such as the dose of the protein, the route of administration, and the duration of treatment, patient HLA alleles, underlying genetic defects, and product dependent factors such as the mechanical processing, manufacture, and contaminants contained in the therapeutic protein formulation (see Ross et al. (2000) Immunogenicity of interferon -beta in multiple sclerosis patients: influence of preparation, dosage, dose frequency, and route of administration. Danish Multiple Sclerosis Study Group. Ann. Neurol.
  • Novel methods of mitigating a subject’s immune response to therapy comprising administering an effective amount of an heterobifunctional extracellular immunoglobulin degrader or a pharmaceutically acceptable salt thereof in combination with, in advance of, or following the therapy.
  • the immunoglobulin degraders described herein can sequester and inactivate or degrade antibodies, including anti -drug antibodies (AD As), such as immunoglobulin G (IgG), immunoglobulin A (IgA), immunoglobulin E (IgE), or immunoglobulin M (IgM) antibodies which would otherwise impede the desired therapy and cause unwanted adverse effects.
  • AD As anti -drug antibodies
  • IgG immunoglobulin G
  • IgA immunoglobulin A
  • IgE immunoglobulin E
  • IgM immunoglobulin M
  • the immunoglobulin heterobifunctional degrader comprises a noncovalent ligand for IgG optionally linked to an asialoglycoprotein receptor (ASGPR) ligand.
  • ASGPR asialoglycoprotein receptor
  • This heterobifunctional immunoglobulin degrader binds an immunoglobulin protein, such as IgG, that is in extracellular circulation and then ASGPR mediates endocytosis and degradation of the antibody, for example by cellular lysosomes in the liver.
  • a method described herein results in a mitigated effect of the immune response to therapy. In other embodiments, a method described herein completely inhibits the effects of the immune response to therapy.
  • the asialoglycoprotein receptor is a Ca 2+ - dependent lectin that is primarily expressed in parenchymal hepatocyte cells.
  • the main role of ASGPR is to help regulate serum glycoprotein levels by mediating endocytosis of desialylated glycoproteins.
  • the receptor binds ligands with a terminal galactose or N-acetylgalactosamine.
  • Asialoglycoproteins bind to ASGPRs and are then cleared by receptor-mediated endocytosis.
  • Avilar Therapeutics has described a series of heterobifunctional degraders of extracellular proteins using the ASGPR-mediated degradation mechanism as described above (WO 2021/155317).
  • Other publications describing various utilizations of the ASGPR mechanism include: U.S. Patent Nos. 9,340,553; 9,617,293; 10,039,778; 10,376,531, and 10,813,942 assigned to Pfizer Inc.; Sanhueza et al. ( JACS , 2017, 139, 3528); Petrov et al.
  • the immunoglobulin degrading compound is of Formula I, Formula II, or Formula III is provided: wherein ASPGR Binding Ligand is a compound selected from: one of R 1 or R 5 is a bond to Linker A ; the other of R 1 and R 5 is independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR 6 , C0-C6alkyl-SR 6 , C 0 -C 6 alkyl-NR 6 R 7 , C 0 -C 6 alkyl-C(O)R 3 , C 0 -C 6 alkyl-S(O)R 3 , C 0 -C 6 alkyl-C(S)R
  • a compound when “optionally substituted” it may be substituted as allowed by valence with one or more groups selected from alkyl (including C1-C4alkyl), alkenyl (including C 2 -C 4 alkenyl), alkynyl (including C 2 -C 4 alkynyl), haloalkyl (including C 1 -C 4 haloalkyl), -OR 6 , F, Cl, Br, I, -NR 6 R 7 , heteroalkyl, heterocycle, heteroaryl, aryl, cyano, nitro, hydroxyl, azide, amide, -SR 3 , -S(O)(NR 6 )R 3 , -NR 8 C(O)R 3 , -C(O)NR 6 R 7 , -C(O)OR 3 , -C(O)R 3 , -SF , and , wherein the optional substituent is selected such that a stable compound results.
  • alkyl including C1-C4
  • an immunoglobulin degrading compound of Formula I-A, Formula II-A, or Formula III-A is provided: or a pharmaceutically acceptable salt thereof.
  • ASGPR Binding Ligand is a compound selected from: or a pharmaceutically acceptable salt thereof.
  • the heterobifunctional extracellular immunoglobulin degrader is provided as an isotopically enriched immunoglobulin degrader, for example an immunoglobulin degrader with at least one desired isotopic substitution of an atom, at an amount above the natural abundance of the isotope.
  • deuterium can replace one or more hydrogens in the immunoglobulin degrader and 13 C can replace one or more carbon atoms.
  • the isotopic substitution is in one or more positions of the ASGPR Ligand.
  • the isotopic substitution is in one or more positions of the Linker portion of the molecule.
  • the isotopic substitution is in one or more positions of the Immunoglobulin Targeting Ligand portion of the molecule.
  • the immunoglobulin heterobifunctional degrader can be administered to a patient suffering from an immune response due to the administration of a therapeutic according to the severity of the response.
  • the response may be severe and life threatening and the patient can be administered the immunoglobulin heterobifunctional degrader one or more time a day, for example, once a day, twice a day, three times a day, or four time a day.
  • the immunoglobulin heterobifunctional degrader can be administered one or more times a day for multiple days, for example at least 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days or more until the immune response subsides.
  • the methods of the invention may be used with any gene or biologies therapy that generates or implicates an undesirable immune response.
  • Methods of the invention may be used, in particular, with a gene therapy, for example a gene therapy comprising a vector delivery including, for example CRISPR-based gene therapy.
  • the gene therapy can be a somatic-cell-targeting gene therapy.
  • the gene therapy can be a germline-cell-targeting cell gene therapy.
  • the therapy can be a chimeric antigen receptor T-cell (CAR T- cell) therapy.
  • the vector may be a viral vector, for example but not limited to an adenovirus- associated virus (AAV) vector, lentivirus vector, adenovirus vector, respiratory syncytial virus (RSV) vector, herpes simplex virus (HSV) vector, poxvirus, or vaccinia virus.
  • AAV adenovirus- associated virus
  • RSV respiratory syncytial virus
  • HSV herpes simplex virus
  • poxvirus poxvirus
  • vaccinia virus vaccinia virus.
  • the viral vector is AAV.
  • the vector is a lipid nanoparticle (LNP).
  • the replacement of a defective gene expressing a mutated or non-functional protein with a normal or wild-type gene expressing a functional protein may result in the induction of an immune reaction to the normal protein in the host.
  • the heterobifunctional extracellular immunoglobulin degrader is administered to a subject receiving gene therapy to replace a defective protein in order to reduce the effects of an immune response to a normal, functionalized or wild-type protein.
  • the heterobifunctional extracellular immunoglobulin degrader is administered once a week, once every two weeks, once every three weeks, or once a month.
  • the therapy can be a recombinant protein therapy.
  • the recombinant protein is a recombinant Factor VIII protein.
  • the recombinant protein is insulin.
  • the therapy is an anti-tumor necrosis factor a (TNFa) monoclonal antibody therapy.
  • TNFa anti-tumor necrosis factor a
  • the monoclonal antibody is adalimumab, infliximab, or golimumab.
  • the therapy is the anti-TNFa therapy etanercept, a soluble TNF receptor IgG Fc fusion protein.
  • the therapy is the anti-TNFa therapy certolizumab, a pegylated anti-TNFa antibody fragment.
  • the therapy is a cytokine inhibitor therapy.
  • the therapy is an anti-IL-12/IL-23 therapy, for example, ustekinumab.
  • the therapy is an anti-IL-17 therapy, for example, secukinumab.
  • the therapy is an anti-IL-6 therapy, for example tocilizumab or sarilumab.
  • the therapy is an anti-IL2 receptor therapy, for example, daclizumab or basiliximab.
  • the therapy is an anti-IIb therapy, for example cankinumab.
  • the therapy is an anti-CD20 therapy, for example, rituximab, tositumomab, ofatumumab, or ibritumomab.
  • the therapy is an anti-CD28 therapy, for example, abatacept.
  • the therapy is an anti-CD3 therapy, for example muromanab.
  • the therapy is an anti-CDl la therapy, for example, efalizumab.
  • the therapy is an anti-CD52 therapy, for example alemtuzumab.
  • the therapy is an anti-CD33 therapy, for example gemtuzumab.
  • the therapy is an anti-human C5 complement therapy, for example eculizumab or ultomiris.
  • the therapy is an anti-IgE therapy, for example omalizumab.
  • the therapy is an RSV prophylaxis therapy, for example palivizumab.
  • the therapy is a PTCA adjunct therapy, for example, abciximab.
  • the therapy is a diagnostic therapy, for example, nofetumumab. capromab, fanolesomab, arcitumomab, or imciromab.
  • the therapy is a cancer therapy, for example, trastuzumab, cetuximab, bevacizumab, or panitumumab.
  • the therapy is a macular degeneration therapy, for example ranibizumab.
  • the therapy is a rheumatoid arthritis or Crohn’s disease therapy, for example cetolizumab pegol.
  • the therapy is a bone loss therapy, for example denosumab.
  • a heterobifunctional immunoglobulin degrader may be provided simultaneous with, prior to, or following the therapy administration of an effective amount to the subject.
  • the heterobifunctional extracellular immunoglobulin degrader is provided to the subject along with the first administration of the therapy to the subject.
  • the immunoglobulin degrader is provided during subsequent administrations of the therapy to the subject.
  • the immunoglobulin degrader may be administered prior to the therapy being readministered to the subject or the immunoglobulin degrader may be conducted after the subject has been administered the therapy at least once.
  • the immunoglobulin degrader may be administered 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 days following administration of the therapy.
  • the immunoglobulin degrader may be administered once a week, once every two weeks, once every three weeks, or once a month following administration of the therapy.
  • aspects of the invention include a method for administering an effective amount of an extracellular immunoglobulin degrader (a heterobifunctional compound) comprising a first ligand configured to bind IgG and a second ligand configured to bind an asialoglycoprotein receptor (ASGPR), optimally connected with a linker, to thereby sequester and degrade immunoglobulin G (IgG), immunoglobulin A (IgA), immunoglobulin E (IgE), or immunoglobulin M (IgM) in the subject in a manner that a therapy can be re-administered to the subject and produce a therapeutic effect in the subject that would not otherwise be achieved due to IgG adversely affecting the therapy.
  • an extracellular immunoglobulin degrader a heterobifunctional compound
  • A immunoglobulin A
  • IgE immunoglobulin E
  • IgM immunoglobulin M
  • the Immunoglobulin Targeting Ligand is covalently bound to Linker in the immunoglobulin degrader compound through the Anchor Bond (which is the chemical bond between the Immunoglobulin Targeting Ligand and either Linker B, Linker C or Linker D). This bond can be placed at any location on the ligand that does not unacceptably disrupt the ability of the Immunoglobulin Targeting Ligand to bind to the Target Immunoglobulin.
  • the Anchor Bond is depicted on the nonlimiting examples of Immunoglobulin Targeting Ligands in the figures as:
  • FIG. 1A provides a non-limiting list of Immunoglobulin Targeting Ligands that target Immunoglobulin A (IgA).
  • FIG. IB provides a non-limiting list of Immunoglobulin Targeting Ligands that target Immunoglobulin G (IgG).
  • FIG. 1C-1G provides a non-limiting list of Immunoglobulin Targeting Ligands that target Immunoglobulin E (IgE).
  • FIG. 2 provides non-limiting examples of IgG protein degrading compounds for use in the present invention.
  • the present invention provides methods for administering a cell, gene, or biologies therapy in combination with a method for mitigating a subject's immune response to produce a therapeutic effect in the subject that would not otherwise be achieved due to the subject's immune response to the cell, gene, or biologies therapy.
  • the subject is generally a human, but can also be a non-human mammal. Methods of the invention can be used to modulate the immune response to any therapy that triggers an immune response.
  • the invention may use a bifunctional compound comprising a first ligand configured to bind to the antibody, for example an IgG, and a second ligand configured to bind an asialoglycoprotein receptor (ASGPR) optimally connected with a linker.
  • ASGPR asialoglycoprotein receptor
  • the bifunctional compound sequesters the antibody of interest and then ASGPR mediates endocytosis and degradation of the antibody, for example by cellular lysosomes, thereby mediating a subject's immune response.
  • one or more additional therapeutically active agents is administered in addition to the heterobifunctional extracellular immunoglobulin degrading compound and the cell, gene, or biologies therapy.
  • the therapeutics and/or immunoglobulin degrading compound or composition can be administered concurrently with, prior to, or after, administration of the cell, gene, or biologies therapy.
  • each agent will be administered at a dose and/or on a time schedule determined for that agent.
  • the additional therapeutically active agent used in this combination can be administered together in a single composition or administered separately in different compositions.
  • the particular combination used in a regimen will take into account compatibility of the inventive therapeutics and/or immunoglobulin degrading molecule with the additional therapeutically active agent and/or the desired therapeutic effect to be achieved.
  • additional therapeutically active agents used in combination be used at levels that do not exceed the levels at which they are used individually. In some embodiments, the levels used in combination will be lower than those used individually.
  • the present invention may be used in the delivery of any cell, gene, or biologies therapy, for example a recombinant protein or antibody therapy, that triggers an immune response.
  • the present invention may be used together with gene therapies.
  • Gene therapies are therapeutics which focus on the genetic modification of cells to produce a therapeutic effect.
  • the gene therapy is delivered via a vector, for example a viral vector, capable of inducing an immune response in the host, which can reduce the effectiveness or ability to readminister the viral vector in the host.
  • the viral vector is a AAV vector, lentivirus vector, adenovirus vector, respiratory syncytial virus (RSV) vector, herpes simplex virus (HSV) vector, poxvirus, or vaccinia virus.
  • the replacement of a defective gene expressing a mutated or non-functional protein with a normal or wild-type gene expressing a functional protein may result in the induction of an immune reaction to the normal protein in the host.
  • the heterobifunctional extracellular immunoglobulin degrader is administered to a subject receiving gene therapy to replace a defective protein in order to reduce the effects of an immune response to a normal, functionalized or wild-type protein.
  • the heterobifunctional extracellular immunoglobulin degrader is administered once a week, once every two weeks, once every three weeks, or once a month.
  • an effective amount of an immunoglobulin degrading compound described herein, for example and IgG degrading compound is administered before, concurrently with, or after administration of gene therapy.
  • Gene therapies may be provided according to the invention to treat a number of disorders, for example Achondroplasia, Alpha-I Antitrypsin Deficiency, Antiphospholipid Syndrome, Autosomal Dominant Polycystic Kidney Disease, Charcot-Marie-Tooth, cancer, Cri du chat, Crohn's Disease, Cystic fibrosis, Dercum Disease, Duane Syndrome, Duchenne Muscular Dystrophy, Factor V Leiden Thrombophilia, Familial Hypercholesterolemia, Familial Mediterranean Fever, Fragile X Syndrome, Gaucher Disease, Hemochromatosis, Hemophilia, Holoprosencephaly, Huntington's disease, Klinefelter syndrome, Marfan syndrome, Myotonic Dystrophy, Neurofibromatosis, Noonan Syndrome, Osteogenesis Imperfecta, Parkinson's disease, Phenylketonuria, Tru Anomaly, Porphyria, Progeria, Retinitis Pigmentosa, Se
  • the disorder to be treated is caused by a loss-of-function mutation disorder. In certain embodiments, the disorder to be treated is a monogenic disorder. In certain embodiments, the disorder to be treated is caused by a gain-of- toxicity mutation. In certain embodiments, the disorder to be treated is caused by recessive compound heterozygous mutations.
  • the disorder is a cancer.
  • cancer include squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinomas, and renal cell carcinomas, cancer of the bladder, bowel, breast, cervix, colon, esophagus, head, kidney, liver, lung, neck, ovary, pancreas, prostate, and stomach; leukemias; benign and malignant lymphomas, particularly Burkitfs lymphoma and Non-Hodgkin's lymphoma; benign and malignant melanomas; myeloproliferative diseases; sarcomas, including Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcomas, peripheral neuroepithelioma, synovial sarcoma, gliomas, astrocytomas, oligodendrogliomas, ependymomas
  • Additional cancers which may be treated using the disclosed compounds according to the present invention include, for example, acute granulocytic leukemia, acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), adenocarcinoma, adenosarcoma, adrenal cancer, adrenocortical carcinoma, anal cancer, anaplastic astrocytoma, angiosarcoma, appendix cancer, astrocytoma, Basal cell carcinoma, B-Cell lymphoma, bile duct cancer, bladder cancer, bone cancer, bone marrow cancer, bowel cancer, brain cancer, brain stem glioma, breast cancer, triple (estrogen, progesterone and HER-2) negative breast cancer, double negative breast cancer (two of estrogen, progesterone and HER-2 are negative), single negative (one of estrogen, progesterone and HER-2 is negative), estrogen-receptor positive, HER2negative breast cancer, estrogen receptor-negative breast cancer, estrogen receptor positive breast
  • the methods of the present invention may mediate an immune response to vectors used for the delivery of therapeutics, for example by sequestering IgG's reactive to a given vector.
  • Vectors are particularly useful in delivering nucleic acid molecules to cells, for example for gene therapies and vaccines.
  • an effective amount of a heterobifunctional immunoglobulin degrading compound described herein, for example an IgG degrading compound is administered before, concurrently with, or after administration of a delivery vector.
  • Vectors may be divided broadly into viral vectors and non-viral vectors.
  • viral vectors or virus-associated vectors are known in the art. Such vectors can be used as carriers of a nucleic acid construct into the cell. Constructs may be integrated and packaged into non-replicating, defective viral genomes like Adenovirus, Adeno-associated virus (AAV), or Herpes simplex virus (HSV) or others, including retroviral and lentiviral vectors, for infection or transduction into cells.
  • Adenovirus Adeno-associated virus
  • HSV Herpes simplex virus
  • retroviral and lentiviral vectors for infection or transduction into cells.
  • retroviral and lentiviral vectors for infection or transduction into cells.
  • retroviral and lentiviral vectors for infection or transduction into cells.
  • retroviral and lentiviral vectors for infection or transduction into cells.
  • retroviral and lentiviral vectors for infection or transduction into cells.
  • retroviral and lentiviral vectors for infection or transduction into cells.
  • retroviruses such as
  • Adeno-associated viral vectors are a family of medium-sized, non- enveloped viruses used as vectors. More than 30 naturally occurring serotypes of AAV are available. Many natural variants in the AAV capsid exist.
  • AAV serotypes include, but are not limited to, AAV serotypes AAVI, AAV2, AAV3, AAV3B, AAV4, AAV5, AAV6, AAV7, AAV7M8, AAV8, AAV8bp, AAV9, AAV10, AAVI 1, AAV 12, AAV13, AAVAnc80, AAVrhlO, AAVPHP.B, and variants of the known or mentioned AAVs or AAVs yet to be discovered or variants or mixtures thereof (see, e.g., WO 2005/033321, which is incorporated herein by reference).
  • LUXTURNA ® voretigene neparvovec-rzyl
  • ZOLGENSMA ® ona shogene abeparvovec-xioi
  • LUXTURNA ® is an AAV serotype 2 gene therapy for the treatment of patients having biallelic RPE65 mutation-associated retinal dystrophy
  • ZOLGENSMA ® is an AAV serotype 9 gene therapy for the treatment of pediatric patients less than 2 years of age having bi-all elic SMN1 mutation-associated spinal muscular atrophy (SMA).
  • SMA spinal muscular atrophy
  • the AAV capsid is selected from AAVI capsid, AAV8 capsid, or AAV9 capsid, or variants thereof.
  • the AAV comprises an AAV capsid having one or substitutions of the amino acid tyrosine (Y) to the amino acid phenylalanine (F) on the surface of the capsid protein.
  • the AAV capsid comprises an AAV capsid having one or more substitutions of the amino acid tyrosine (Y) to the amino acid phenylalanine (F) on the surface of the AAV capsid protein.
  • AAV capsid having tyrosine to phenylalanine mutations are known in the art, as described for example in US 8445267, US 8802440, US 9157098, US 9611302, US 9775918, US 9920097, US 10011640, US 10294281, US 10723768, US 10815279, and US 10934327, each of which is incorporated by refence.
  • the AAV capsid is an AAV capsid having substitutions selected from Y272F, Y444F, Y447F, T491 V, T494F, Y500F, Y730F, Y733F or a combination thereof.
  • present invention comprises a method that mediates an immune response to the administration of a recombinant adeno-associated virus (rAAV) containing a nucleic acid encoding a therapeutic polypeptide, thereby increasing the efficacy and/or reducing the needed dosage of therapies that use vector deliveries.
  • the rAAV comprises an AAV capsid and the nucleic acid sequence encoding the therapeutic polypeptide in the AAV capsid, wherein the nucleic acid sequence encoding the therapeutic polypeptide is operably linked to a promoter as described herein and AAV inverted terminal repeats (ITRs) required for packaging a nucleic acid into the capsid.
  • ITRs AAV inverted terminal repeats
  • the nucleic acid to be packaged comprises therapeutic polypeptide encoding nucleic acid sequences, promoter sequences, and optionally other regulatory sequences as described herein, which are flanked by packaging signals of the AAV genome to facilitate efficient packaging of the nucleic acid into the AAV capsid for delivery and expression in the target cell and/or organ.
  • the nucleic acid sequence encoding the therapeutic polypeptide is flanked at the 5’ and 3’ ends by AAV ITRs.
  • a 5’ AAV ITR, the nucleic acid sequence encoding the therapeutic polypeptide, and a 3’ AAV ITR comprise the nucleic acid that is packaged into the AAV capsid.
  • the term “ITR” is defined as a cis genetic element responsible for facilitating the replication and packaging of a nucleic acid during AAV vector production necessary for rAAV generation.
  • the ITRs are derived from a different AAV variant than the AAV capsid variant.
  • the source of the ITRs are of one AAV variant and the AAV capsid is from another AAV variant, the resulting vector is defined as pseudotyped.
  • the ITRs are derived from the AAV2 variant.
  • the ITRs are derived from deleted, or shortened, version of the AAV2 variant (AITR).
  • an AAV vector genome comprises an AAV 5’ ITR, a nucleic acid encoding a polypeptide operably linked to a promoter and optionally linked to one or more regulatory sequences, and an AAV 3’ ITR.
  • scAAV selfcomplementary AAV
  • the term “Self-complementary AAV” refers to a construct in which a coding nucleic acid sequence carried by an rAAV has been designed to form an intra-molecular double-stranded DNA template.
  • the two complementary regions of scAAV Upon infection, rather than waiting for cell-mediated second strand synthesis, the two complementary regions of scAAV will associate to form one double stranded DNA (dsDNA) unit that is amenable to immediate replication and transcription.
  • the ITRs to be used are selected from wild- type, full-length, engineered, shortened, deleted, or a combination thereof.
  • An engineered AAV may be, without limitation, a pseudotyped AAV, a chimeric AAV capsid, a recombinant AAV capsid, or a “humanized” AAV capsid.
  • a deleted, or shortened, version of the 5’ ITR termed 5’ AITR, has been described in which the D-sequence and terminal resolution sites (TRs) are deleted.
  • the ITR comprises a shortened ITR of the AAV2 variant comprises 130 base pairs (bps), wherein the external “a” element is deleted.
  • the shorted AAV2 ITR is lengthened during vector DNA amplification using the internal “a” element as a template.
  • full-length AAV 5’ and 3’ ITRs are used.
  • wild-type AAV 5’ and 3’ ITRs are used.
  • engineered ITRs are used. Suitable AAV ITR sequences are known in the art (see, e.g., Yan et al., J Virol. 79(1):364- 379(2005), incorporated herein by reference).
  • the nucleic acid to be packaged comprises one or more regulatory elements such that the total rAAV vector genome is about 2.0 to about 5.5 kilobases (kb) in size.
  • the one or more regulatory sequences are selected such that the total rAAV vector genome is about 2.1, 2.3, 2.8, 3.1, 3.2, 3.3, 4.0 kb, 4.7 kb, or 5.2 kb in size.
  • the rAAV vector genome approximates the size of a native AAV genome.
  • the one or more regulatory sequences are selected such that the total rAAV vector genome is about 4.7 kb in size.
  • the one or more regulatory sequences are selected such that the total rAAV vector genome is about 5.2 kb in size.
  • the nucleic acid to be packaged comprises one or more regulatory elements selected from a promoter, a Woodchuck Hepatitis virus (WHP) posttranscriptional regulatory element (WPRE), a polyadenylation (poly A) signal, an intron, or a combination thereof.
  • WP Woodchuck Hepatitis virus
  • WPRE Woodchuck Hepatitis virus
  • poly A polyadenylation
  • the one or more regulatory elements are operably linked to the nucleic acid encoding a therapeutic polypeptide.
  • a packaging cell line is used for production of a vector (e.g., a recombinant AAV).
  • a host cell may be a prokaryotic or eukaryotic cell (e.g., human, insect, or yeast) that contains exogenous or heterologous DNA that has been introduced into the cell by any means, e.g., electroporation, calcium phosphate precipitation, microinjection, transformation, viral infection, transfection, liposome delivery, membrane fusion techniques, high velocity DNA- coated pellets, viral infection, and protoplast fusion.
  • Examples of host cells may include, but are not limited to an isolated cell, a cell culture, an Escherichia coli cell, a yeast cell, a human cell, a non-human cell, a mammalian cell, a non-mammalian cell, an insect cell, a HEK-293 cell, a liver cell, a kidney cell, a cell of the central nervous system, a neuron, a glial cell, or a stem cell.
  • the nucleic acid encoding a therapeutic polypeptide is optimized for expression in the desirable subject species, for example, in humans. Codon-optimized coding nucleic acid sequences can be designed by various different methods known to those skilled in the art.
  • the entire length of the open reading frame (ORF) of the nucleic acid encoding a therapeutic polypeptide is modified.
  • a fragment of the open reading frame (ORF) of the nucleic acid encoding a therapeutic polypeptide is modified.
  • the nucleic acid encoding a therapeutic polypeptide is optimized for tropism towards the desirable target organ.
  • the target organ is selected from muscle, eye, liver, brain, or a combination thereof.
  • the nucleic acid encoding a therapeutic polypeptide is optimized for tropism towards the desirable target cell-type.
  • target cell refers to any cell in which the expression of the therapeutic polypeptide is desired.
  • the term “target cell” is intended to reference the cells of the subject being treated.
  • the target cell-type is selected from neurons, glia, photoreceptors, retinal pigment epithelial (RPE) cells, hepatocytes, myocytes, skeletal muscle cells, heart cells, or a combination thereof.
  • the AAV vector is formulated in a buffer/carrier suitable for infusion in a subject, for example a human subject.
  • the viral vector e.g, rAAV
  • the rAAV is delivered systemically.
  • the rAAV is delivered intravenously, intraperitoneally, intranasally, or via inhalation.
  • the buffer/carrier comprises a component that prevents the AAV from attaching to the infusion tube.
  • the dosage of the vector is about 1 x 10 9 GC to about 1 x 10 13 genome copies (GC) per dose.
  • the dose of the vector administered to a patient is at least about 1.0 x 10 9 GC/kg , about 1.5 x 10 9 GC/kg , about 2.0 x 10 9 GC/g, about 2.5 x 10 9 GC/kg , about 3.0 x 10 9 GC/kg , about 3.5 x 10 9 GC/kg , about 4.0 x 10 9 GC/kg , about 4.5 x 10 9 GC/kg , about 5.0 x 10 9 GC/kg , about 5.5 x 10 9 GC/kg , about 6.0 x 10 9 GC/kg , about 6.5 x 10 9 GC/kg , about 7.0 x 10 9 GC/kg , about 7.5 x 10 9 GC
  • the AAV capsid includes variants which may include up to about 10% variation from any described or known AAV capsid sequence.
  • the AAV capsid shares about 90% identity to about 99.9 % identity, about 95% to about 99% identity, or about 97% to about 98% identity to an AAV capsid provided herein and/or known in the art.
  • the AAV capsid shares at least 95% identity with an AAV capsid.
  • the comparison may be made over any of the variable proteins (e.g., vpl, vp2, or vp3).
  • percent (%) identity in the context of nucleic acid sequences or amino acid sequences refers to the residues in the two sequences which are the same when aligned for correspondence. Unless otherwise specified, it is to be understood that a percentage of identity is a minimum level of identity and encompasses all higher levels of identity up to 100% identity to the reference sequence. For example, “95% identity” and “at least 95% identity” may be used interchangeably and include 95, 96, 97, 98, 99 up to 100% identity to the referenced sequence, and all fractions therebetween.
  • the length of sequence identity comparison may be over the full-length of the genome, the full-length of a gene coding sequence, a fragment of a gene coding sequence, a fragment of at least about 500 to about 5000 nucleotides, the full-length of an amino acid sequence, a fragment of an amino acid sequence, a fragment of at least about 8 to about 200 amino acids, is desired.
  • identity refers to “identity”, “homology”, or “similarity” between two different sequences, “identity”, “homology” or “similarity” is determined in reference to “aligned” sequences.
  • Aligned sequences or “alignments” refer to more than one nucleic acid sequences or amino acid sequences, often containing corrections for missing or additional bases or amino acids as compared to a reference sequence.
  • Multiple sequence alignment programs are also available for nucleic acid sequences and amino acid sequences. Examples of such programs include, “Clustal Omega”, “Clustal W”, “CAP Sequence Assembly”, “BLAST”, “MAP”, and “MEME”, which are accessible through Web Servers on the internet. Other sources for such programs are known to those skilled in the art.
  • Non-viral delivery systems may also be used together with the present invention, for example a liposome, lipid nanoparticle, or other lipid formulations.
  • a nucleic acid for delivery may be associated with a lipid.
  • the nucleic acid associated with a lipid may be encapsulated in the aqueous interior of a liposome, interspersed within the lipid bilayer of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the oligonucleotide, entrapped in a liposome, complexed with a liposome, dispersed in a solution containing a lipid, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with a micelle, or otherwise associated with a lipid.
  • the nucleic acid for delivery may be encapsulated in a lipid nanoparticle.
  • Lipid nanoparticles can be comprised of a cationic lipid, an anionic lipid, a polyethylene glycol functionalized lipid, and steroids.
  • Lipid, lipid/DNA or lipid/expression vector associated compositions are not limited to any particular structure in solution. For example, they may be present in a bilayer structure, as micelles, or with collapsed structure. They may also simply be interspersed in a solution, possibly forming aggregates that are not uniform in size or shape. Lipids are fatty substances which may be naturally occurring or synthetic lipids.
  • lipids include the fatty droplets that naturally occur in the cytoplasm as well as the class of compounds which contain long-chain aliphatic hydrocarbons and their derivatives, such as fatty acids, alcohols, amines, amino alcohols, and aldehydes.
  • the methods of the present invention mediate an immune response to any vector used for the delivery of therapeutics, thereby increasing the efficacy and/or reducing the needed dosage of therapies that use vector deliveries.
  • the present invention comprises the use of an effective amount of an immunoglobulin degrading compound described herein, for example and IgG degrading compound, which is to be administered before, concurrently with, or after a vector for the delivery of a therapeutic polypeptide.
  • Immunoglobulin G is the main type of antibody found in all body fluids (for example, blood and extracellular fluid) and protects against bacterial and viral infections. It represents approximately 75% of serum antibodies in humans and is thus the most common type of antibody found in circulation. IgG antibodies are generated following class switching and maturation of the antibody response, thus they participate predominantly in the secondary immune response.
  • IgG antibodies frequently mediate a subject's response to therapies, diminishing the effectiveness of those therapies.
  • IgG can be divided into 4 distinct subclasses: IgGl, IgG2, IgG3, & IgG4.
  • the evolution of IgG subclass switches is regulated by interaction with T cells and follows a I-way direction: IgG3 to IgGl to IgG2 to IgG4.
  • the differences between subclasses are mainly in their size and the configuration of the hinge region, glycosylation sites, and structures, as well as a few key amino acids that impact the ability to interact with complement and FC receptors.
  • IgGl and IgG3 are monomeric, having 2 heavy chains & 2 light chains, and bivalent, having 2 variable regions.
  • IgG2 has a disulfide bond pattern which allows for two monomeric IgG2 antibodies to form a dimeric and tetravalent structure through unique inter-molecule disulfide bonds.
  • IgG4 has two intrachain disulfide bonds that can be reduced, which generates a monovalent structure.
  • the monovalent structures can reform the disulfide bonds, but may not be the same IgG4 monovalent chain, meaning the resulting IgG4 will be a bivalent monomer but will have two different variable regions. Binding of IgG can be accomplished through the use of an IgG-specific ligand.
  • the ligand may bind to the FC region of IgG.
  • the IgG-specific ligand may be a Fc-binding peptide, such as a Fc-BP2.
  • the IgG-specific ligand may be Fc-111. or a pharmaceutically acceptable salt thereof.
  • the Protein Data Bank website provides the crystal structure of IgG searchable by 1H3X (Krapp, S., et al., J. Mol. Biol., 2003, 325: 979); and 5V43 (Lee, C.H., et al., Nat. Immunol., 2017, 18: 889-898); as well as the crystal structure of IgG bound to various compounds searchable by 5YC5 (Kiyoshi M., et al., Sci. Rep., 2018, 8: 3955-3955); 5XJE (Sakae Y., et al., Sci. Rep., 2017, 7: 13780-13780); 5GSQ (Chen, C. L., et al., ACS Chem.
  • Kiyoshi, M., et al. provides insight into the structural basis for binding of human IgGl to its high-affinity human receptor FcyRI. (Kiyosi M., et al., Nat Commun., 2015, 6, 6866).
  • IgG Targeting Ligands are provided in Fig. IB. Additional representative IgG Targeting Ligands include: wherein X R is O, S, NH, or N-C 1 -C 3 alkyl; and X M is O, S, NH, or N-C 1 -C 3 alkyl. In other embodiments the IgG Targeting Ligand is selected from: , In some embodiments, the IgG Targeting Ligand is a group according to the chemical structure: wherein R N02 is a dinitrophenyl group optionally linked through CH2, S(O), S(O)2, - S(O)2O, - OS(O)2, or OS(O)2O.
  • the IgG Targeting Ligand is selected from: ted from O, CH2, NH, N-C1-C3 alkyl, NC(O)C1-C3 alkyl, S(O), S(O)2, -S(O) 2 O, - OS(O) 2 , or OS(O) 2 O.
  • the IgG Targeting Ligand is a 3-indoleacetic acid group according to the chemical structure: where k”” is 1-4 (preferably 2-3, most often 3) or a group.
  • the IgG Targeting Ligand is a peptide.
  • Nonlimiting examples of IgG Targeting Ligand peptides include:
  • PAM (RTY)4K2KG (Fassina, et al, J. Mol. Recognit. 1996, 9, 564-569) comprising SEQ ID NO: 1 RTYK and SEQ ID NO:2 RTYKKG
  • D-PAM wherein the amino acids of the PAM sequence are all D-amino acids (Verdoliva, et al, J. Immunol. Methods, 2002, 271, 77-88) SEQ ID NO:3 RTYK (D-amino acids) and SEQ ID NO:4 RTYKKG (D-amino acids).
  • D-RAM-F wherein the amino acids of the PAM sequence are all D-amino acids with further modifications wherein the four N-terminal arginines are acetylated with phenylacetic acid (Dinon, et al J. Mol. Recognit. 2011, 24, 1087-1094) SEQ ID NO:5 RTYK (D-amino acids, N- terminal arginine acetylated with phenylacetic acid) and SEQ ID NO: 6 RTYKKG (D-amino acids, N-terminal arginine acetylated with phenylacetic acid).
  • SEQ ID NO:30 HWRGWV (Yang, et al., J Peptide Res. 2006, 66, 1 1 0-137)
  • SEQ ID NO:31 HYFKFD (Yang, et al, J. Chromatogr. A 2009, 1216, 910-918)
  • SEQ ID NO:32 HFRRHL (Menegatti, et al, J. Chromatogr. A 2016, 1445, 93-104)
  • SEQ ID NO:33 HWCitGWV (Menegatti, et al, J. Chromatogr. A 2016, 1445, 93-
  • SEQ ID NO:36 DAAG Mall Synthetic peptide ligand, Lund, et al, J. Chromatogr. A 2012, 1225, 158- 167;
  • SEQ ID NO:37 cyclo[(Na-Ac) S(A)-RWHYFK-Lact-E] (Menegatti, et al, Anal. Chem. 2013, 85, 9229-9237);
  • SEQ ID NO:38 cyclo[(Na-Ac)-Dap(A)-RWHYFK-Lact-E] (Menegatti, et al, Anal. Chem. 2013, 85, 9229-9237);
  • SEQ ID NO:40 NKFRGKYK (Sugita, et al, Biochem. Eng. J. 2013, 79, 33-40); SEQ ID NO:41 NARKFYKG (Sugita, et al, Biochem. Eng. J. 2013, 79, 33-40); SEQ ID NO:42 FYWHCLDE (Zhao, et al, Biochem. Eng. J. 2014, 88, 1-11); SEQ ID NO:43 FYCHWALE (Zhao, et al, J Chromatogr. A 2014, 1355, 107-114); SEQ ID NO:44 FYCHTIDE (Zhao, et al., Z Chromatogr.
  • the IgG Targeting Ligand is specific for IgG4.
  • the IgG4 specific Targeting Ligand is described in Gunnarsson et al. Biomolecular Engineering 2006, 23, 111-117.
  • the IgG4 specific targeting ligand is selected from SEQ ID NO : 55 FDLLEHFY and SEQ ID NO:56 DLLHHFDYF.
  • Additional IgG Targeting Ligands include
  • the immunoglobulin degrading compound is selected from: or a pharmaceutically acceptable salt thereof; or a pharmaceutically acceptable salt thereof;
  • IgG degrading compounds include:
  • a hydroxyl, amine, amide, or carboxylic acid group in an Immunoglobulin Targeting Ligand drawn herein is capped with a protecting group.
  • a protecting group for example in this embodiment:
  • an Immunoglobulin Targeting Ligand drawn herein is used as the attachment point to Linker instead of the drawn attachment point.
  • the immunoglobulin degrading compound is selected from:
  • the immunoglobulin degrading compound is selected from:
  • the Immunoglobulin Targeting Ligand is: In certain embodiments the Immunoglobulin Targeting Ligand is:
  • Immunoglobulin E (IgE)
  • Immunoglobulin E is a strong mediator of allergic disease, including but not limited to, atopic asthma, allergic rhinitis, atopic dermatitis, cutaneous contact hypersensitivity, IgE- mediated food allergy, IgE-mediated animal allergies, allergic conjunctivitis, allergic urticaria, anaphylactic shock, nasal polyposis, keratoconjunctivitis, mastocytosis, eosinophilic gastrointestinal disease, bullous pemphigoid, chemotherapy induced hypersensitivity reaction, seasonal allergic rhinitis, interstitial cystitis, eosinophilic esophagitis, angioedema, acute interstitial nephritis, atopic eczema, eosinophilic bronchitis, chronic obstructive pulmonary disease, gastroenteritis, hyper-IgE syndrome (Job's Syndrome), IgE monoclonal gammopathy, monoclonal gammopathy, mono
  • the immunoglobulin degrading compound is: or a pharmaceutically acceptable salt thereof.
  • the immunoglobulin degrading compound is: or a pharmaceutically acceptable salt thereof. In certain embodiments the immunoglobulin degrading compound is: or a pharmaceutically acceptable salt thereof.
  • the immunoglobulin degrading compound is: or a pharmaceutically acceptable salt thereof.
  • the immunoglobulin degrading compound is: or a pharmaceutically acceptable salt thereof.
  • the Immunoglobulin Targeting Ligand is:
  • IgE degrading compounds include:
  • Immunoglobulin A (IgA)
  • IgA immunoglobulin A
  • IgA nephropathy also known as Berger’s disease
  • celiac disease also known as Berger’s disease
  • HSP Henoch-Schonlein purpura
  • IgA pemphigus IgA pemphigus
  • dermatitis herpetiformis IgA herpetiformis
  • IBD inflammatory bowel disease
  • Sjogren's syndrome ankylosing spondylitis
  • IgA multiple myeloma a-chain disease
  • IgA monoclonal gammopathy monoclonal gammopathy of undetermined significance (MGUS)
  • MGUS monoclonal gammopathy of undetermined significance
  • IgA-specific Immunoglobulin Targeting Ligand used is an Opt peptide. Variations and derivatives of the IgA-specific Opt peptide suitable for use as IgA-specific Immunoglobulin Targeting Ligands are described in Hatanaka et al. Journal of Biological Chemistry , 287(57) 43126-43136. In certain embodiments, the IgA-specific Immunoglobulin Targeting Ligand is Opt-1. In certain embodiments, the IgA-specific
  • Immunoglobulin Targeting Ligand is Opt-2.
  • Immunoglobulin Targeting Ligand is Opt-3.
  • the immunoglobulin degrading compound is: or a pharmaceutically acceptable salt thereof. In certain embodiments the immunoglobulin degrading compound is: or a pharmaceutically acceptable salt thereof.
  • the immunoglobulin degrading compound is: or a pharmaceutically acceptable salt thereof.
  • the Immunoglobulin Targeting Ligand is: .
  • Linkers In non-limiting embodiments, Linker A and Linker B are independently selected from: ; wherein: R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , and R 20 are independently at each occurrence selected from the group consisting of a bond, alkyl, -C(O)-, -C(O)O-, -OC(O)-, -SO 2 -, -S(O)-, -C(S)-, -C(O)NR 6 -, -NR 6 C(O)-, -O-, -S-, -NR 6 -, -C(R 21 R 21 )-, -P(O)(R 3 )O-, -P(O)(R 3 )-, a divalent residue of a natural or unnatural amino acid, alkenyl, alkynyl,
  • a divalent residue of an amino acid is selected from
  • amino acid can be oriented in either direction and wherein the amino acid can be in the L- or D-form.
  • a divalent residue of a dicarboxylic acid is generated from a nucleophilic addition reaction:
  • xx is independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, and 25.
  • yy is independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, and 25.
  • a divalent residue of a dicarboxylic acid is generated from a condensation reaction:
  • Non-limiting embodiments of a divalent residue of a saturated monocarboxylic acid is selected from butyric acid (-0C(0)(CH2)2CH2-), caproic acid (-0C(0)(CH2)4CH2-), caprylic acid (-0C(0)(CH2)5CH2-), capric acid (-0C(0)(CH2)8CH2-), lauric acid (-OC(0)(CH2)IOCH2-), myristic acid (-0C(0)(CH2)i2CH2-), pentadecanoic acid (-OC(O)(CH 2 ) 13 CH 2 -), palmitic acid (-OC(O)(CH 2 ) 14 CH 2 -), stearic acid (-OC(O)(CH 2 ) 16 CH 2 -), behenic acid (-OC(O)(CH2)20CH2-), and lignoceric acid (-OC(O)(CH2)22CH2-);
  • Non-limiting embodiments of a divalent residue of a fatty acid include residues selected from linoleic
  • Linker C is selected from: . wherein: R 22 is independently at each occurrence selected from the group consisting of alkyl, -C(O)N-, -NC(O)-, -N-, -C(R 21 )-, -P(O)O-, -P(O)-, -P(O)(NR 6 R 7 )N-, alkenyl, haloalkyl, aryl, heterocycle, and heteroaryl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R 21 ; and the remaining variables are as defined herein.
  • Linker D is selected from: ; wherein: R 32 is independently at each occurrence selected from the group consisting of alkyl, N + X " , -C-, alkenyl, haloalkyl, aryl, heterocycle, and heteroaryl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R 21 ;
  • X- is an anionic group, for example Br- or Cl -; and all other variables are as defined herein.
  • Linker A is selected from: each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
  • Linker A is selected from: each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein. In certain embodiments Linker A is selected from: each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
  • Linker A is selected from: each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
  • Linker A is selected from: each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
  • Linker A is selected from: each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
  • Linker A is selected from: wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
  • Linker A is selected from: wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
  • Linker A is selected from: wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
  • Linker B , Linker C , or Linker D is selected from: wherein tt is independently selected from 1, 2, or 3 and ss is 3 minus tt. In certain embodiments Linker B , Linker C , or Linker B is selected from: wherein tt and ss are as defined herein.
  • Linker B is selected from:
  • each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence; and tt and ss are as defined herein.
  • Linker B is selected from: wo 2023/009554
  • each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence; and tt and ss are as defined herein.
  • Linker B is selected from: wherein each heteroaryl and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence; and tt and ss are as defined herein.
  • Linker A is selected from: In certain embodiments Linker B is selected from:
  • the Linker A is selected from
  • Linker A is selected from wherein each is optionally substituted with 1, 2, 3, or 4 substituents substituent selected from R 21 . In certain embodiments Linker A is selected from:
  • Linker A is selected from: In certain embodiments Linker A is selected from:
  • Linker A is selected from: In certain embodiments Linker A is selected from:
  • the Linker B is selected from
  • the Linker B is selected from
  • the Linker B is selected from wherein each is optionally substituted with 1, 2, 3, or 4 substituents substituent selected from R 21 .
  • Linker B is selected from: In certain embodiments Linker B is selected from:
  • Linker B -Linker A is selected from: In certain embodiments Linker B -Linker A is selected from:
  • the Linker C is selected from
  • the Linker C is selected from In certain embodiments, the Linker C is selected from
  • Linker c -(Linker A ) 2 is selected from: In certain embodiments Linker c -(Linker A ) 2 is selected from: In certain embodiments Linker c -(Linker A ) 2 is selected from:
  • the Linker D is selected from
  • the Linker D is selected from In certain embodiments, the Linker D is selected from wherein each is optionally substituted with 1, 2, 3, or 4 substituents are selected from R 21 . In certain embodiments, Linker B -(Linker A ) is selected from In certain embodiments, Linker°-(Linker A ) is selected from
  • Linker B is selected from: wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence. In certain embodiments Linker B is selected from: wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
  • Linker B is selected from: wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
  • Linker B is selected from:
  • each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
  • Linker A is selected from: each of which is substituted with 1 or 2 optional substituents .
  • Linker A is bond
  • Linker A is attached to the ASGPR Binding Ligand and the right side is attached to Linker B , Linker C , or Linker D .
  • the right side of Linker A is attached to the ASGPR Binding Ligand and the right side is attached to Linker B , Linker C , or Linker D .
  • the left side of Linker B is attached to the Extracellular Targeting Ligand and the right side is attached to Linker A .
  • the right side of Linker B is attached to the Extracellular Targeting Ligand and the left side is attached to Linker A .
  • Linker B is bond
  • a linker is provided as described above wherein a , replaced with a for example where Linker B is drawn as it is in this embodiment.
  • a linker is provided as described above wherein a is replaced with a , for example where Linker B is drawn as this embodiment.
  • a linker is provided as described above wherein a is replaced with a
  • the asialoglycoprotein receptor is a Ca 2 +-dependent receptor.
  • the primary endogenous role of ASGPRs is to help regulate serum glycoprotein levels by mediating endocytosis of glycoproteins.
  • the receptor binds ligands with a terminal galactose or N- acetylgalactosamine.
  • the C 3 - and C 4 - hydroxyl groups bind to Ca 2 +, with the N-acetyl position also been considered important to binding activity.
  • Asialoglycoproteins bind to ASGPRs and are then cleared by receptor-mediated endocytosis.
  • the receptor and the protein are dissociated in the acidic endosomal compartment and the protein is eventually degraded by lysosomes.
  • the receptor is endocytosed and recycled constitutively from the endosome back to the plasma membrane about every 1 5 minutes regardless of whether or not a glycoprotein is bound. Internalization rate of the receptor may be altered by presence of a bound ligand.
  • the ligand of the immunoglobulin degrading compound that binds to ASGPR may be an asialoglycoprotein or a derivative thereof.
  • the ASGPR is comprised of two homologous subunits known as HI and H2. Various ratios of HI and H2 form functional homo- and hetero-oligomers with different conformations, but the most abundant conformation is a trimer composed of two HI and one H2 subunits.
  • the ASGPR is composed of a cytoplasmic domain, a transmembrane domain, a stalk region, and a carbohydrate recognition domain (CRD). Both the HI and H2 subunit form the CRD, and expression of both subunits mediates endocytosis of asialoglycoproteins.
  • ASGPRs are primarily expressed on hepatocytes, and hepatocytes exhibit a high exposition of ASGPR binding cites (approximately 100,000 — 500,000 binding sites per cell).
  • any ligand of ASGPR may be used in immunoglobulin degrading compounds of the invention.
  • ligands that may be used with the invention are described in Stokmaier, Bioorg. Med. Chem., 2009, 17, 7254, which describes the synthesis of a series of D-GalNAc derivatives where the anomeric OH group is removed and the acetamido group is replaced with a 4-substituted 1,2, 3 -triazole moiety, and Mamidyala, JACS, 2012, 134, 1978, which describes compounds derived from 2-azidogalactosyl analogs where the anomeric position is occupied by either a B-methyl or a B-4-methoxy-phenyl group and the azide group is replaced with an amide or a triazole, the entirety of the contest of each of which are incorporated by reference herein.
  • ASGPR ligands that may be used with the invention are described in PCT Application No. PCT/US21/15939, filed January 29, 2021, and U.S. Provisional Application No. 63/183,450, filed May 3, 2021, the entirety of the contents of each of which are incorporated by reference herein.
  • the ASGPR ligand may include derivatives of six-carbon pyranose moieties, specifically galactose and talose. These two sugars differ in the stereochemistry of the C 2 substituent.
  • the "down" C 2 configuration corresponds to the stereochemistry of galactose, while the C 2 substituent in the "up” configuration corresponds to the stereochemistry oftalose.
  • substituents may be at the C 2 position of the two sugars.
  • the immunoglobulin degrading compounds may utilize a 2:1 ratio of ASGPR ligands to antibody binding ligands. Multiple ASGPR ligands may bind ASGPR more tightly and have increased degradation efficacy.
  • the immunoglobulin degrading compounds may have a 1:1 ratio of ASGPR binding ligands to antibody binding ligands.
  • the compounds may utilize a heteroaryl amine substituents at the C 2 position. The substituents may increase efficacy of the ligand, preferable in compounds with a 1:1 ratio of ASGPR ligand to antibody ligand.
  • the immunoglobulin degrading molecule is:
  • the immunoglobulin degrading molecule is: or a pharmaceutically acceptable salt thereof.
  • immunoglobulin degrading compounds may comprise a ligand that binds to IgG, for example an FC binding peptide, for example Fc-111, Fc-BP2, or derivatives thereof that bind the FC portion of the antibody and thus facilitate the selective recruitment of antibody to hepatocytes for degradation
  • immunoglobulin degrading compounds may comprise a ligand that binds to IgA, for example an FC binding peptide, for example an Opt class of peptides.
  • Opt class peptides are advantageous in that they are highly selective for IgA and thus facilitate the selective recruitment of IgA to hepatocytes for degradation.
  • the immunoglobulin degrading molecule may be. or a pharmaceutically acceptable salt thereof.
  • the immunoglobulin degrading molecule of the present invention may be provided as an isotopically enriched immunoglobulin degrading molecule with at least one desired isotopic substitution of an atom, at an amount above the natural abundance of the isotope.
  • deuterium can replace one or more hydrogens in the bispecific compound and 13 c can replace one or more carbon atoms.
  • the isotopic substitution is in one or more positions of the ASGPR ligand.
  • the isotopic substitution is in one or more positions of a linker portion of the molecule.
  • the isotopic substitution is in the antibody ligand portion of the molecule.
  • the ASGPR ligand may be a molecule according to the Formula IV, Formula V, Formula
  • R 1 and R 5 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle, heterocycloalkyl, haloalkoxy, C 0 -C 6 alkyl-OR 6 , C 0 -C 6 alkyl-SR 6 , C 0 -C 6 alkyl-NR 6 R 7 , C 0 -C 6 alkyl-C(O)R 3 , C 0 -C 6 alkyl-S(O)R 3 , C 0 -C 6 alkyl-C(S)R 3 , C 0 -C 6 alkyl- S(O)2R 3 , C0-C6alkyl-N(R 8 )-C(
  • the ASGPR Binding Ligand is of Formula: or a pharmaceutically acceptable salt thereof.
  • the ASGPR ligand or ASGPR ligand-Linker- may be a compound according to the Formula:
  • ASGPR ligands and ASGPR ligand-Linkers are typically attached to the Immunoglobulin Targeting Ligand through the C6 position.
  • the ASGPR ligand or ASGPR ligand-Linker is a compound of Formula:
  • the ASGPR ligand is of structure:
  • the ASGPR ligand is of structure:
  • the ASGPR ligand is of structure:
  • the ASGPR ligand is of structure: Compositions and administration
  • heterobifunctional extracellular immunoglobulin degrading compound for use in the present invention or a pharmaceutically acceptable salt, solvate or prodrug thereof as disclosed herein can be administered as a pharmaceutical composition that includes an effective amount for a subject in need of such treatment to mediate the subject’s immune response to a therapy.
  • the present invention provides a treatment regimen for use in mediating a subject’s immune response to a cell, gene or biologies therapy that includes administration of an effective amount of a pharmaceutical composition comprising an immunoglobulin degrading bifunctional molecule of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog such as a deuterated derivative, or prodrug thereof, and a pharmaceutically acceptable excipient.
  • a pharmaceutical composition comprising an immunoglobulin degrading bifunctional molecule of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog such as a deuterated derivative, or prodrug thereof, and a pharmaceutically acceptable excipient.
  • the cell, gene, or biologies therapy and heterobifunctional immunoglobulin degrading molecule is present in an effective amount, for example a therapeutically effective amount or a prophylactically effective amount.
  • the heterobifunctional immunoglobulin degrading molecule mediates the subject's immune response, the effective amount of the therapeutic to be delivered may be reduced.
  • the treatment regimen of the present invention that includes administering an effective amount of therapeutics and/or immunoglobulin degrading molecules can be administered in any manner that allows the immunoglobulin degrading molecule to bind to the immunoglobulin, typically in the blood stream, and carry it to the ASGPR-bearing hepatocyte cells on the liver for endocytosis and degradation.
  • Methods to deliver the therapeutics and immunoglobulin degrading molecules of the present invention may include oral, intravenous, sublingual, subcutaneous, parenteral, buccal, rectal, intra-aortal, intracranial, subdermal or transnasal, or by other means, in dosage unit formulations containing one or more conventional pharmaceutically acceptable carriers, as appropriate.
  • the treatment regimen of the present invention includes administering an effective amount of therapeutics and/or immunoglobulin degrading molecules which may be administered intravenously.
  • the therapeutics and/or immunoglobulin degrading molecule may be formulated in a liquid dosage form for intravenous injection, such as a buffered solution, for example a phosphate buffered solution and saline buffered solution. The solution may be buffered with multiple salts.
  • the treatment regimen of the present invention includes administering an effective amount of therapeutics and/or immunoglobulin degrading molecule which may be administered orally.
  • the therapeutics and/or immunoglobulin degrading molecule may be formulated in capsules, tablets, and powders, for example a gel containing capsule.
  • the treatment regimen of the present invention includes administering an effective amount of therapeutics and/or heterobifunctional immunoglobulin degrading molecule which may be administered subcutaneously.
  • the therapeutics and immunoglobulin degrading molecule may be formulated in a liquid dosage form for subcutaneous injection, such as a buffered solution, for example a phosphate buffered solution and saline buffered solution.
  • the solution may be buffered with multiple salts.
  • a pharmaceutically acceptable salt means a salt of the described therapeutics and/or heterobifunctional immunoglobulin degrading molecule which is suitable for administration to a subject without undue toxicity, irritation, or allergic response, and commensurate with a reasonable benefit to risk ratio, and effective for its intended use.
  • a pharmaceutically acceptable salt means a relatively nontoxic, inorganic and organic acid addition salts of the presently disclosed therapeutics and/or immunoglobulin degrading molecules.
  • salts can be prepared during the final isolation and purification of the therapeutics and/or immunoglobulin degrading molecules or by separately reacting the purified therapeutics and/or immunoglobulin degrading molecule in its free form with a suitable organic or inorganic acid and then isolating the salt thus formed.
  • Acid addition salts of the basic therapeutics and/or immunoglobulin degrading molecules are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt in the conventional manner.
  • the free base form can be regenerated by contacting the salt form with a base and isolating the free base in the conventional manner.
  • the free base forms may differ from their respective salt forms in certain physical properties such as solubility in polar solvents.
  • Pharmaceutically acceptable base addition salts may be formed with metals or amines, such as alkali and alkaline earth metal hydroxides, or of organic amines.
  • metals used as cations include, but are not limited to, sodium, potassium, magnesium, calcium, and the like.
  • suitable amines include, but are not limited to, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine, and procaine.
  • the base addition salts of acidic therapeutics and/or immunoglobulin degrading molecules are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner.
  • the free acid form can be regenerated by contacting the salt form with an acid and isolating the free acid in a conventional manner.
  • the free acid forms may differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents.
  • Salts can be prepared from inorganic acids sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, nitric, phosphoric, sulfuric, hydrobromic, hydriodic, phosphorus, and the like.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, lactobionate, laurylsulphonate and isethionate salts, and the like.
  • Salts can also be prepared from organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl -substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. and the like.
  • organic acids such as aliphatic mono- and dicarboxylic acids, phenyl -substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. and the like.
  • Representative salts include acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, phthalate, benzenesulfonate, toluenesulfonate, phenyl acetate, citrate, lactate, maleate, tartrate, methanesulfonate, and the like.
  • Pharmaceutically acceptable salts can include cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium and the like, as well as non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to, ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. Also contemplated are the salts of amino acids such as arginate, gluconate, galacturonate, and the like. See, for example, Berge et ah, J. Pharm. Sci., 1977, 66, 1-19, which is incorporated herein by reference.
  • compositions include inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Additional acceptable excipients include cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, perfuming agents, etc., and combinations thereof.
  • Diluents may include calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and combinations thereof.
  • Granulating and/or dispersing agents may include potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation — exchange resins, calcium carbonate, silicates, sodium carbonate, cross — linked poly(vinyl — pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross — linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium compounds, etc., and combinations thereof.
  • Surface active agents and/or emulsifiers may include natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and Veegum [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g.
  • natural emulsifiers e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin
  • colloidal clays e.g. bentonite [aluminum silicate] and Veegum [magnes
  • stearyl alcohol cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g.
  • polyoxyethylene monostearate [Myij 45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and Solutol), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g., Cremophor), polyoxyethylene ethers, (e.g .
  • Binding agents may include starch (e.g. cornstarch and starch paste), gelatin, sugars (e.g.
  • natural and synthetic gums e.g. acacia, sodium alginate, extract of Irish moss,
  • Preservatives may include antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, etc., and/or combinations thereof.
  • Antioxidants may include alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabi sulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabi sulfite, and sodium sulfite.
  • the pharmaceutical composition is in a dosage form that contains from about 0.1 mg to about 1500 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of the active immunoglobulin degrading molecule and optionally from about 0.1 mg to about 1500 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of an additional active agent in a unit dosage form.
  • Examples are dosage forms with at least about 0.1, 1, 5, 10, 25, 50, 100, 200, 250, 300, 400, 500, 600, 700, 750, 800, 850, 900, 950, or 1,000 mg of active immunoglobulin degrading molecule, or its salt.
  • the dosage form has at most about 0.1, 1, 5, 10, 25, 50, 100, 200, 250, 300, 400, 500, 600, 700, 750, 800, 850, 900, 950, or 1,000 mg of active immunoglobulin degrading molecule, or its salt.
  • the dose ranges from about 0.01-100 mg/kg of patient bodyweight, for example about 0.01 mg/kg, about 0.05 mg/kg, about 0. 1 mg/kg, about 0.5 mg/kg, about 1 mg/kg, about 1.5 mg/kg, about 2 mg/kg, about 2.5 mg/kg, about 3 mg/kg, about 3.5 mg/kg, about 4 mg/kg, about 4.5 mg/kg, about 5 mg/kg, about 110 mg/kg, about 115 mg/kg, about 20 mg/kg, about 25 mg/kg, about 30 mg/kg, about 35 mg/kg, about 40 mg/kg, about 45 mg/kg, about 50 mg/kg, about 55 mg/kg, about 60 mg/kg, about 65 mg/kg, about 70 mg/kg, about 75 mg/kg, about 80 mg/kg, about 85 mg/kg, about 90 mg/kg, about 95 mg/kg, or about 100 mg/kg.
  • heterobifunctional immunoglobulin degrading molecules disclosed herein are used as described are administered once a day (QD), twice a day (BID), or three times a day (TID).
  • immunoglobulin degrading molecules disclosed herein are used as described are administered at least once a day for at least 1 day, at least 2 days, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least 14 days, at least
  • the heterobifunctional immunoglobulin degrading molecule is administered once a day, twice a day, three times a day, or four times a day.
  • the pharmaceutical composition may be formulated as any pharmaceutically useful form, for example, a pill, capsule, tablet, an injection or infusion solution, a syrup, an inhalation formulation, a suppository, a buccal or sublingual formulation, a parenteral formulation, or in a medical device.
  • Some dosage forms, such as tablets and capsules, can be subdivided into suitably sized unit doses containing appropriate quantities of the active components, e.g., an effective amount to achieve the desired purpose.
  • Carriers include excipients and diluents and must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated.
  • the carrier can be inert, or it can possess pharmaceutical benefits of its own.
  • the amount of carrier employed in conjunction with the therapeutics and/or immunoglobulin degrading molecule is sufficient to provide a practical quantity of material for administration per unit dose of the therapeutics and/or immunoglobulin degrading molecule. If provided as in a liquid, it can be a solution or a suspension.
  • Representative carriers include phosphate buffered saline, water, solvent(s), diluents, pH modifying agents, preservatives, antioxidants, suspending agents, wetting agent, viscosity agents, tonicity agents, stabilizing agents, and combinations thereof.
  • the carrier is an aqueous carrier.
  • aqueous carries include, but are not limited to, an aqueous solution or suspension, such as saline, plasma, bone marrow aspirate, buffers, such as Hank's Buffered Salt Solution (HBSS), HEPES (4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid), Ringers buffer, ProVisc@, diluted ProVisc@, Provisc@ diluted with PBS, Krebs buffer, Dulbecco's PBS, normal PBS, sodium hyaluronate solution (HA, 5 mg/mL in PBS), citrate buffer, simulated body fluids, plasma platelet concentrate and tissue culture medium or an aqueous solution or suspension comprising an organic solvent.
  • HBSS Hank's Buffered Salt Solution
  • HEPES 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid
  • Ringers buffer such as Hank's Buffered Salt Solution (HBSS), HEPES (4-(2-
  • Acceptable solutions include, for example, water, Ringer' s solution and isotonic sodium chloride solutions.
  • the formulation may also be a sterile solution, suspension, or emulsion in a non-toxic diluent or solvent such as 1,3-butanediol.
  • Viscosity agents may be added to the pharmaceutical composition to increase the viscosity of the composition as desired.
  • useful viscosity agents include, but are not limited to, hyaluronic acid, sodium hyaluronate, carbomers, polyacrylic acid, cellulosic derivatives, polycarbophil, polyvinylpyrrolidone, gelatin, dextin, polysaccharides, polyacrylamide, polyvinyl alcohol (including partially hydrolyzed polyvinyl acetate), polyvinyl acetate, derivatives thereof and mixtures thereof.
  • Solutions, suspensions, or emulsions for administration may be buffered with an effective amount necessary to maintain a pH suitable for the selected administration.
  • Suitable buffers are well known by those skilled in the art. Some examples of useful buffers are acetate, borate, carbonate, citrate, and phosphate buffers.
  • Solutions, suspensions, or emulsions for topical, for example, ocular administration may also contain one or more tonicity agents to adjust the isotonic range of the formulation. Suitable tonicity agents are well known in the art. Some examples include glycerin, mannitol, sorbitol, sodium chloride, and other electrolytes.
  • Classes of carriers include, but are not limited to binders, buffering agents, coloring agents, diluents, disintegrants, emulsifiers, flavorants, glidants, lubricants, preservatives, stabilizers, surfactants, tableting agents, and wetting agents.
  • Some carriers may be listed in more than one class, for example vegetable oil may be used as a lubricant in some formulations and a diluent in others.
  • Exemplary pharmaceutically acceptable carriers include sugars, starches, celluloses, powdered tragacanth, malt, gelatin, talc, and vegetable oils.
  • Optional active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the therapeutics and/or immunoglobulin degrading molecule of the present invention.
  • the pharmaceutical compositions/combinations can be formulated for oral administration. These compositions can contain any amount of active therapeutics and/or immunoglobulin degrading molecule that achieves the desired result, for example between 0.1 and 99 weight % (wt.%) of the therapeutics and/or immunoglobulin degrading molecule and usually at least about 5 wt.% of the therapeutics and/or immunoglobulin degrading molecule. Some embodiments contain from about 25 wt. % to about 50 wt. % or from about 5 wt. % to about 75 wt. % of the therapeutics and/or immunoglobulin degrading molecule. Enteric coated oral tablets may also be used to enhance bioavailability of the therapeutics and/or immunoglobulin degrading molecules for an oral route of administration.
  • Formulations suitable for rectal administration are typically presented as unit dose suppositories. These may be prepared by admixing the active therapeutics and/or immunoglobulin degrading molecule with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
  • conventional solid carriers for example, cocoa butter
  • Therapeutics and/or immunoglobulin degrading molecules and pharmaceutically acceptable composition, salts, isotopic analogs, or prodrugs thereof may be formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions comprising a therapeutics and/or immunoglobulin degrading molecule as described herein will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject or organism will depend upon a variety of factors including the disease, disorder, or condition being treated and the severity of the disorder; the activity of the specific therapeutics and/or immunoglobulin degrading molecule employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific therapeutics and/or immunoglobulin degrading molecule employed; the duration of the treatment; drugs used in combination or coincidental with the specific therapeutics and/or immunoglobulin degrading molecule employed; and like factors well known in the medical arts.
  • the therapeutics and/or immunoglobulin degrading molecules and compositions provided herein can be administered by any route, including enteral (e.g., oral), parenteral, intravenous, intramuscular, intra — arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, creams, and/or drops), mucosal, nasal, buccal, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; and/or as an oral spray, nasal spray, and/or aerosol.
  • enteral e.g., oral
  • parenteral intravenous, intramuscular, intra — arterial, intramedullary
  • intrathecal subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal
  • topical as by powders, ointments, creams, and/
  • Specifically contemplated routes are oral administration, intravenous administration (e.g., systemic intravenous injection), regional administration via blood and/or lymph supply, and/or direct administration to an affected site.
  • intravenous administration e.g., systemic intravenous injection
  • regional administration via blood and/or lymph supply
  • direct administration to an affected site.
  • the most appropriate route of administration will depend upon a variety of factors including the nature of the agent (e.g., its stability in the environment of the gastrointestinal tract), the condition of the subject (e.g., whether the subject is able to tolerate oral administration).
  • the exact amount of the therapeutic and/or immunoglobulin degrading molecule required to achieve an effective amount will vary from subject to subject, depending, for example, on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular therapeutics and/or immunoglobulin degrading molecule(s), mode of administration, and the like.
  • the desired dosage can be delivered using any frequency determined to be useful by the health care provider, including three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks.
  • the desired dosage can be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
  • therapeutics and/or immunoglobulin degrading molecules or compositions can be administered in combination with one or more additional therapeutically active agents.
  • the therapeutics and/or immunoglobulin degrading molecules or compositions can be administered in combination with additional therapeutically active agents that improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body.
  • the therapy employed may achieve a desired effect for the same disorder (for example, a therapeutic and/or immunoglobulin degrading molecule can be administered in combination with an anti — inflammatory agent, anti — cancer agent, immunosuppressant, etc.), and/or it may achieve different effects (e.g., control of adverse side — effects, e.g., emesis controlled by an antiemetic).
  • the therapeutics and/or immunoglobulin degrading molecule or composition can be administered concurrently with, prior to, or subsequent to, one or more additional therapeutically active agents.
  • each agent will be administered at a dose and/or on a time schedule determined for that agent.
  • the additional therapeutically active agent utilized in this combination can be administered together in a single composition or administered separately in different compositions.
  • the particular combination to employ in a regimen will take into account compatibility of the inventive therapeutics and/or immunoglobulin degrading molecule with the additional therapeutically active agent and/or the desired therapeutic effect to be achieved.
  • additional therapeutically active agents utilized in combination be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually.
  • Additional therapeutically active agents may include, but are not limited to, small organic molecules such as drug compounds (e.g., compounds approved by the Food and Drugs Administration as provided in the Code of Federal Regulations (CFR)), peptides, proteins, carbohydrates, monosaccharides, oligosaccharides, polysaccharides, nucleoproteins, mucoproteins, lipoproteins, synthetic polypeptides or proteins, small molecules linked to proteins, glycoproteins, steroids, nucleic acids, DNAs, RNAs, nucleotides, nucleosides, oligonucleotides, antisense oligonucleotides, lipids, hormones, vitamins and cells.
  • the additional therapeutically active agent is an anti -cancer agent, e.g., radiation therapy and/or one or more chemotherapeutic agents.
  • a treatment regimen comprising the administration of a therapeutic and/or immunoglobulin degrading molecule of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog (such as a deuterated derivative), or prodrug thereof in combination or in alternation with at least one additional therapeutic agent.
  • the combinations and/or alternations can be administered for beneficial, additive, or synergistic effect in the treatment of immunoglobulin-mediated disorders.
  • the methods of the present invention mediate an immune response to any additional excipient or combinations used for the delivery of the therapeutics or the immunoglobulin degrading molecule by the action of the immunoglobulin degrading molecule itself.
  • a method for administering a therapy comprising: sequestering immunoglobulin G (IgG) in a subject so that a therapy can be initiated and readministered to the subject and produce a therapeutic effect in the subject that would not otherwise be achieved due to IgG interacting with the therapy.
  • IgG immunoglobulin G
  • a method for administering a therapy comprising: providing to a subject an immunoglobulin degrading compound comprising a first ligand configured to bind IgG, and a second ligand configured to bind an asialoglycoprotein receptor (ASGPR), to thereby sequester and degrade immunoglobulin G (IgG) in the subject so that a therapy can be re-administered to the subject and produce a therapeutic effect in the subject that would not otherwise be achieved due to IgG interacting with the therapy.
  • an immunoglobulin degrading compound comprising a first ligand configured to bind IgG, and a second ligand configured to bind an asialoglycoprotein receptor (ASGPR), to thereby sequester and degrade immunoglobulin G (IgG) in the subject so that a therapy can be re-administered to the subject and produce a therapeutic effect in the subject that would not otherwise be achieved due to IgG interacting with the therapy.
  • the viral vector is an adenovirus-associated virus (AAV) vector.
  • AAV adenovirus-associated virus

Abstract

This invention provides methods to modulate an immune response to a therapeutic, for example gene therapy, by administering an immunoglobulin degrading molecule that has an asialoglycoprotein receptor (ASGPR) Binding Ligand bound to an Immunoglobulin Targeting Ligand or a pharmaceutically acceptable salt thereof.

Description

METHODS TO REDUCE THE ADVERSE EFFECTS OF GENE OR BIOLOGICS THERAPY
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application No. 63/225,746, filed July 26, 2021, the entirety of which is hereby incorporated by reference for all purposes.
FIELD OF THE INVENTION
This invention provides methods to modulate an immune response to a therapeutic, for example gene therapy, by administering an immunoglobulin degrading molecule that has an asialoglycoprotein receptor (ASGPR) Binding Ligand bound to an Immunoglobulin Targeting Ligand or a pharmaceutically acceptable salt thereof.
INCORPORATION BY REFERENCE
The contents of the text file named “19121-010W01_SequenceListing_ST26” which was created on July 25, 2022 and is 59.2 KB in size, are hereby incorporated by reference in their entirety.
BACKGROUND OF THE INVENTION
Recent medical advances have resulted in novel and improved biological agents useful to treat a host of maladies that affect humans. Biological agents are structurally immunogenic, and therefore usually elicit a minor, subclinical and transient phenomenon (see, e.g., Vande Casteele et al. Antibody response to infliximab and its impact on pharmacokinetics can be transient. Am J Gastroenterology 2013; 108: 962-971). While immune responses to drug products such as vaccines may be desirable, when a therapeutic protein or viral vector elicits an unwanted immune response, the safety and efficacy of the biologic drug can be adversely affected.
Occasionaly, complete inhibition of the biological effects of the drug may occur. Additional serious consequences include the development of serious adverse immune reactions (e.g., anaphylaxis), and autoimmune-like outcomes when endogenous human proteins are targeted by anti-drug antibodies (ADAs).
Elevated immune responses to treatments has been most prevalent in gene therapies and recombinant therapeutic protein treatments, mitigating a promising area of medical advancement for a number of disorders. For example, many gene therapies rely on viral vectors, such as adenovirus vectors and adeno-associated virus vectors (AAV), for delivering gene therapy targets to cells. Many patients have pre-existing immunity to the adenovirus or AAV vector which can be further triggered by administration of the vector. Also, with each delivery of the vector, the increasing immune response from the patient results in the escalated removal of the vector before they can reach their target cells. As a result, ongoing therapy requires either increasingly larger doses of the vector being delivered, risking damage to the patient's liver, or discontinuation of the therapy due to inefficacy. As a consequence, many patients suffering from disorders, such as genetic disorders or cancers, are unable to receive ongoing or repeat treatments using such viral vectors that might otherwise save or improve their lives.
Similarly, the development of an immune response to recombinant protein therapies has been a significant challenge. The development of ADAs can lead to cross-reactivity with the endogenous protein, leading to life-threatening conditions (see, e.g., Macdougall, (2007) Epoetin- induced pure red cell aplasia: diagnosis and treatment. Curr. Opin. Nephrol. Hypertens. 16, 585— 588). Little is known, however, about the immunological mechanisms underlying the unwanted immune response against human homolog protein therapeutics. Sauerbom et ah, Immunological mechanism underlying the immune response to recombinant human protein therapeutics, Trends in Pharmacological Sciences, Vol. 31 (2), 2010, pgs. 53-59). Factors influencing immunogenicity include recombinant protein dependent factors such as the primary amino acid sequence, and glycosylation patterns (Hermeling et al. (2004) Structure-immunogenicity relationships of therapeutic proteins. Pharm. Res. 21, 897-903). Other factors influencing immunogenicity include treatment dependent factors such as the dose of the protein, the route of administration, and the duration of treatment, patient HLA alleles, underlying genetic defects, and product dependent factors such as the mechanical processing, manufacture, and contaminants contained in the therapeutic protein formulation (see Ross et al. (2000) Immunogenicity of interferon -beta in multiple sclerosis patients: influence of preparation, dosage, dose frequency, and route of administration. Danish Multiple Sclerosis Study Group. Ann. Neurol. 48, 706-712; Rosenberg, A.S. (2006) Effects of protein aggregates: an immunologic perspective. AAPS J. 8, E501-E507). Nevertheless, the development of ADA responses and ADA-mediated adverse events can hamper the development and widespread use of promising therapeutics.
It is an object of the invention to provide a method to reduce the adverse effects of gene or biologies therapy. SUMMARY OF THE INVENTION
Novel methods of mitigating a subject’s immune response to therapy, for example cell, gene, or biologies therapy, are provided comprising administering an effective amount of an heterobifunctional extracellular immunoglobulin degrader or a pharmaceutically acceptable salt thereof in combination with, in advance of, or following the therapy. The immunoglobulin degraders described herein can sequester and inactivate or degrade antibodies, including anti -drug antibodies (AD As), such as immunoglobulin G (IgG), immunoglobulin A (IgA), immunoglobulin E (IgE), or immunoglobulin M (IgM) antibodies which would otherwise impede the desired therapy and cause unwanted adverse effects. In certain embodiments, the immunoglobulin heterobifunctional degrader comprises a noncovalent ligand for IgG optionally linked to an asialoglycoprotein receptor (ASGPR) ligand. This heterobifunctional immunoglobulin degrader binds an immunoglobulin protein, such as IgG, that is in extracellular circulation and then ASGPR mediates endocytosis and degradation of the antibody, for example by cellular lysosomes in the liver. In certain embodiments, a method described herein results in a mitigated effect of the immune response to therapy. In other embodiments, a method described herein completely inhibits the effects of the immune response to therapy.
Other heterobifunctional molecules have been described that use the asialoglycoprotein receptor to degrade extracellular proteins. The asialoglycoprotein receptor (ASGPR) is a Ca2+- dependent lectin that is primarily expressed in parenchymal hepatocyte cells. The main role of ASGPR is to help regulate serum glycoprotein levels by mediating endocytosis of desialylated glycoproteins. The receptor binds ligands with a terminal galactose or N-acetylgalactosamine. Asialoglycoproteins bind to ASGPRs and are then cleared by receptor-mediated endocytosis. The receptor and the protein are dissociated in the acidic endosomal compartment and the protein is eventually degraded by lysosomes. Avilar Therapeutics has described a series of heterobifunctional degraders of extracellular proteins using the ASGPR-mediated degradation mechanism as described above (WO 2021/155317). Other publications describing various utilizations of the ASGPR mechanism include: U.S. Patent Nos. 9,340,553; 9,617,293; 10,039,778; 10,376,531, and 10,813,942 assigned to Pfizer Inc.; Sanhueza et al. ( JACS , 2017, 139, 3528); Petrov et al. ( Bioorganic andMedicinal Chemistry Letters, 2018, 28, 382); WO 2018/223073 and WO2018/223081 assigned to Pfizer Inc. and Wave Life Sciences Ltd.; WO 2018/223056 assigned to Wave Sciences Ltd.; Schmidt et al. ( Nucleic Acids Research , 2017, 45, 2294); Huang et al. (Bioconjugate Chem. 2017, 28, 283); WO 2019/199621, WO 2019/199634, WO 2021/072246, and WO 2021/072269 assigned to Yale University; WO 2020/132100 assigned to The Board of Trustees of the Leland Stanford Junior University; WO 2021/142377, WO 2021/263060, WO 2021/263061, and WO 2022/150721 assigned to Lycia Therapeutics Inc.; Banik et al. (Nature, 2020, 584, 291); and an article from the Bertozzi group titled “LYTACs that engage the asialoglycoprotein receptor for targeted protein degradation” (Ahn, et al. Nat. Chem. Biol. (2021)), published in the journal Nature Chemical Biology.
In certain embodiments the immunoglobulin degrading compound is of Formula I, Formula II, or Formula III is provided:
Figure imgf000006_0001
wherein ASPGR Binding Ligand is a compound selected from:
Figure imgf000007_0001
one of R1 or R5 is a bond to LinkerA; the other of R1 and R5 is independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl- S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; R3 at each occurrence is independently selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, -OR8, and -NR8R9; R6 and R7 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, haloalkyl, heteroaryl, heterocycle, -alkyl-OR8, -alkyl-NR8R9, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R8 and R9 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocycle; R10 is selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R65, R66, and R67 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl-S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; LinkerA is a bond or a moiety that covalently links LinkerB, LinkerC, or LinkerD to the ASGPR Binding Ligand; LinkerB is a bond or a moiety that covalently links LinkerA to an Immunoglobulin Targeting Ligand; LinkerC is a chemical group that links each LinkerA to the Immunoglobulin Targeting Ligand; LinkerD is a chemical group that links each LinkerA to the Immunoglobulin Targeting Ligand; Immunoglobulin Targeting Ligand is a Ligand that binds to an immunoglobulin, for example IgG or IgA. and when a compound is “optionally substituted” it may be substituted as allowed by valence with one or more groups selected from alkyl (including C1-C4alkyl), alkenyl (including C2-C4alkenyl), alkynyl (including C2-C4alkynyl), haloalkyl (including C1-C4haloalkyl), -OR6, F, Cl, Br, I, -NR6R7, heteroalkyl, heterocycle, heteroaryl, aryl, cyano, nitro, hydroxyl, azide, amide, -SR3, -S(O)(NR6)R3, -NR8C(O)R3, -C(O)NR6R7, -C(O)OR3, -C(O)R3, -SF
Figure imgf000009_0001
, and , wherein the optional substituent is selected such that a stable compound results. In certain aspects an immunoglobulin degrading compound of Formula I-A, Formula II-A, or Formula III-A is provided:
Figure imgf000009_0002
or a pharmaceutically acceptable salt thereof. In certain embodiments ASGPR Binding Ligand is a compound selected from:
Figure imgf000010_0001
or a pharmaceutically acceptable salt thereof.
In certain embodiments the heterobifunctional extracellular immunoglobulin degrader is provided as an isotopically enriched immunoglobulin degrader, for example an immunoglobulin degrader with at least one desired isotopic substitution of an atom, at an amount above the natural abundance of the isotope. For example, deuterium can replace one or more hydrogens in the immunoglobulin degrader and 13C can replace one or more carbon atoms. In one embodiment, the isotopic substitution is in one or more positions of the ASGPR Ligand. In another embodiment, the isotopic substitution is in one or more positions of the Linker portion of the molecule. In another embodiment, the isotopic substitution is in one or more positions of the Immunoglobulin Targeting Ligand portion of the molecule.
In certain aspects, the immunoglobulin heterobifunctional degrader can be administered to a patient suffering from an immune response due to the administration of a therapeutic according to the severity of the response. In some embodiments, the response may be severe and life threatening and the patient can be administered the immunoglobulin heterobifunctional degrader one or more time a day, for example, once a day, twice a day, three times a day, or four time a day. In some embodiments, the immunoglobulin heterobifunctional degrader can be administered one or more times a day for multiple days, for example at least 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days or more until the immune response subsides.
Advantageously, the methods of the invention may be used with any gene or biologies therapy that generates or implicates an undesirable immune response. Methods of the invention may be used, in particular, with a gene therapy, for example a gene therapy comprising a vector delivery including, for example CRISPR-based gene therapy. In certain non-limiting embodiments, the gene therapy can be a somatic-cell-targeting gene therapy. In certain nonlimiting embodiments, the gene therapy can be a germline-cell-targeting cell gene therapy. In certain non-limiting embodiments, the therapy can be a chimeric antigen receptor T-cell (CAR T- cell) therapy. The vector may be a viral vector, for example but not limited to an adenovirus- associated virus (AAV) vector, lentivirus vector, adenovirus vector, respiratory syncytial virus (RSV) vector, herpes simplex virus (HSV) vector, poxvirus, or vaccinia virus. In some embodiments, the viral vector is AAV. In certain non-limiting embodiments, the vector is a lipid nanoparticle (LNP).
In addition, the replacement of a defective gene expressing a mutated or non-functional protein with a normal or wild-type gene expressing a functional protein may result in the induction of an immune reaction to the normal protein in the host. In certain embodiments, the heterobifunctional extracellular immunoglobulin degrader is administered to a subject receiving gene therapy to replace a defective protein in order to reduce the effects of an immune response to a normal, functionalized or wild-type protein. In some embodiments, the heterobifunctional extracellular immunoglobulin degrader is administered once a week, once every two weeks, once every three weeks, or once a month.
In some embodiments, the therapy can be a recombinant protein therapy.
In some embodiments, the recombinant protein is a recombinant Factor VIII protein.
In some embodiments, the recombinant protein is insulin.
In some embodiments, the therapy is an anti-tumor necrosis factor a (TNFa) monoclonal antibody therapy. In some embodiments, the monoclonal antibody is adalimumab, infliximab, or golimumab.
In some embodiments, the therapy is the anti-TNFa therapy etanercept, a soluble TNF receptor IgG Fc fusion protein. In some embodiments, the therapy is the anti-TNFa therapy certolizumab, a pegylated anti-TNFa antibody fragment. In some embodiments, the therapy is a cytokine inhibitor therapy. In some embodiments, the therapy is an anti-IL-12/IL-23 therapy, for example, ustekinumab. In some embodiments, the therapy is an anti-IL-17 therapy, for example, secukinumab. In some embodiments, the therapy is an anti-IL-6 therapy, for example tocilizumab or sarilumab. In some embodiments, the therapy is an anti-IL2 receptor therapy, for example, daclizumab or basiliximab. In some embodiments, the therapy is an anti-IIb therapy, for example cankinumab.
In some embodiments, the therapy is an anti-CD20 therapy, for example, rituximab, tositumomab, ofatumumab, or ibritumomab. In some embodiments, the therapy is an anti-CD28 therapy, for example, abatacept. In some embodiment, the therapy is an anti-CD3 therapy, for example muromanab. In some embodiments, the therapy is an anti-CDl la therapy, for example, efalizumab. In some embodiments, the therapy is an anti-CD52 therapy, for example alemtuzumab. In some embodiments, the therapy is an anti-CD33 therapy, for example gemtuzumab.
In some embodiments, the therapy is an anti-human C5 complement therapy, for example eculizumab or ultomiris.
In some embodiments, the therapy is an anti-IgE therapy, for example omalizumab.
In some embodiments, the therapy is an RSV prophylaxis therapy, for example palivizumab.
In some embodiments, the therapy is a PTCA adjunct therapy, for example, abciximab.
In some embodiments, the therapy is a diagnostic therapy, for example, nofetumumab. capromab, fanolesomab, arcitumomab, or imciromab. In some embodiments, the therapy is a cancer therapy, for example, trastuzumab, cetuximab, bevacizumab, or panitumumab.
In some embodiments, the therapy is a macular degeneration therapy, for example ranibizumab.
In some embodiments, the therapy is a rheumatoid arthritis or Crohn’s disease therapy, for example cetolizumab pegol.
In some embodiments, the therapy is a bone loss therapy, for example denosumab.
A heterobifunctional immunoglobulin degrader may be provided simultaneous with, prior to, or following the therapy administration of an effective amount to the subject. In certain embodiments the heterobifunctional extracellular immunoglobulin degrader is provided to the subject along with the first administration of the therapy to the subject. In certain embodiments the immunoglobulin degrader is provided during subsequent administrations of the therapy to the subject. The immunoglobulin degrader may be administered prior to the therapy being readministered to the subject or the immunoglobulin degrader may be conducted after the subject has been administered the therapy at least once. In some embodiments, the immunoglobulin degrader may be administered 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more than 10 days following administration of the therapy. In some embodiments, the immunoglobulin degrader may be administered once a week, once every two weeks, once every three weeks, or once a month following administration of the therapy.
Aspects of the invention include a method for administering an effective amount of an extracellular immunoglobulin degrader (a heterobifunctional compound) comprising a first ligand configured to bind IgG and a second ligand configured to bind an asialoglycoprotein receptor (ASGPR), optimally connected with a linker, to thereby sequester and degrade immunoglobulin G (IgG), immunoglobulin A (IgA), immunoglobulin E (IgE), or immunoglobulin M (IgM) in the subject in a manner that a therapy can be re-administered to the subject and produce a therapeutic effect in the subject that would not otherwise be achieved due to IgG adversely affecting the therapy.
BRIEF DESCRIPTION OF THE FIGURES
The Immunoglobulin Targeting Ligand is covalently bound to Linker in the immunoglobulin degrader compound through the Anchor Bond (which is the chemical bond between the Immunoglobulin Targeting Ligand and either Linker B, Linker C or Linker D). This bond can be placed at any location on the ligand that does not unacceptably disrupt the ability of the Immunoglobulin Targeting Ligand to bind to the Target Immunoglobulin. The Anchor Bond is depicted on the nonlimiting examples of Immunoglobulin Targeting Ligands in the figures as:
Figure imgf000013_0001
FIG. 1A provides a non-limiting list of Immunoglobulin Targeting Ligands that target Immunoglobulin A (IgA).
FIG. IB provides a non-limiting list of Immunoglobulin Targeting Ligands that target Immunoglobulin G (IgG).
FIG. 1C-1G provides a non-limiting list of Immunoglobulin Targeting Ligands that target Immunoglobulin E (IgE). FIG. 2 provides non-limiting examples of IgG protein degrading compounds for use in the present invention.
DETAILED DESCRIPTION
The present invention provides methods for administering a cell, gene, or biologies therapy in combination with a method for mitigating a subject's immune response to produce a therapeutic effect in the subject that would not otherwise be achieved due to the subject's immune response to the cell, gene, or biologies therapy. As provided herein, the subject is generally a human, but can also be a non-human mammal. Methods of the invention can be used to modulate the immune response to any therapy that triggers an immune response. For example, without being limited to a mechanism of action, the invention may use a bifunctional compound comprising a first ligand configured to bind to the antibody, for example an IgG, and a second ligand configured to bind an asialoglycoprotein receptor (ASGPR) optimally connected with a linker. The bifunctional compound sequesters the antibody of interest and then ASGPR mediates endocytosis and degradation of the antibody, for example by cellular lysosomes, thereby mediating a subject's immune response.
In certain embodiments one or more additional therapeutically active agents is administered in addition to the heterobifunctional extracellular immunoglobulin degrading compound and the cell, gene, or biologies therapy. The therapeutics and/or immunoglobulin degrading compound or composition can be administered concurrently with, prior to, or after, administration of the cell, gene, or biologies therapy. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent. It will further be appreciated that the additional therapeutically active agent used in this combination can be administered together in a single composition or administered separately in different compositions. The particular combination used in a regimen will take into account compatibility of the inventive therapeutics and/or immunoglobulin degrading molecule with the additional therapeutically active agent and/or the desired therapeutic effect to be achieved. In general, it is expected that additional therapeutically active agents used in combination be used at levels that do not exceed the levels at which they are used individually. In some embodiments, the levels used in combination will be lower than those used individually. Therapies
Advantageously, the present invention may be used in the delivery of any cell, gene, or biologies therapy, for example a recombinant protein or antibody therapy, that triggers an immune response.
For example, the present invention may be used together with gene therapies. Gene therapies are therapeutics which focus on the genetic modification of cells to produce a therapeutic effect. In some embodiments, the gene therapy is delivered via a vector, for example a viral vector, capable of inducing an immune response in the host, which can reduce the effectiveness or ability to readminister the viral vector in the host. In some embodiments, the viral vector is a AAV vector, lentivirus vector, adenovirus vector, respiratory syncytial virus (RSV) vector, herpes simplex virus (HSV) vector, poxvirus, or vaccinia virus. In addition, the replacement of a defective gene expressing a mutated or non-functional protein with a normal or wild-type gene expressing a functional protein may result in the induction of an immune reaction to the normal protein in the host. In certain embodiments, the heterobifunctional extracellular immunoglobulin degrader is administered to a subject receiving gene therapy to replace a defective protein in order to reduce the effects of an immune response to a normal, functionalized or wild-type protein. In some embodiments, the heterobifunctional extracellular immunoglobulin degrader is administered once a week, once every two weeks, once every three weeks, or once a month. In certain aspects an effective amount of an immunoglobulin degrading compound described herein, for example and IgG degrading compound, is administered before, concurrently with, or after administration of gene therapy.
Gene therapies may be provided according to the invention to treat a number of disorders, for example Achondroplasia, Alpha-I Antitrypsin Deficiency, Antiphospholipid Syndrome, Autosomal Dominant Polycystic Kidney Disease, Charcot-Marie-Tooth, cancer, Cri du chat, Crohn's Disease, Cystic fibrosis, Dercum Disease, Duane Syndrome, Duchenne Muscular Dystrophy, Factor V Leiden Thrombophilia, Familial Hypercholesterolemia, Familial Mediterranean Fever, Fragile X Syndrome, Gaucher Disease, Hemochromatosis, Hemophilia, Holoprosencephaly, Huntington's disease, Klinefelter syndrome, Marfan syndrome, Myotonic Dystrophy, Neurofibromatosis, Noonan Syndrome, Osteogenesis Imperfecta, Parkinson's disease, Phenylketonuria, Poland Anomaly, Porphyria, Progeria, Retinitis Pigmentosa, Severe Combined Immunodeficiency (SCID), Sickle cell disease, Spinal Muscular Atrophy, Tay-Sachs disease, Thalassemia, Trimethylaminuria, Turner Syndrome, Velocardiofacial Syndrome, WAGR Syndrome, or Wilson Disease. In certain embodiments, the disorder to be treated is caused by a loss-of-function mutation disorder. In certain embodiments, the disorder to be treated is a monogenic disorder. In certain embodiments, the disorder to be treated is caused by a gain-of- toxicity mutation. In certain embodiments, the disorder to be treated is caused by recessive compound heterozygous mutations.
In certain embodiments the disorder is a cancer. Non-limiting examples of cancer include squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinomas, and renal cell carcinomas, cancer of the bladder, bowel, breast, cervix, colon, esophagus, head, kidney, liver, lung, neck, ovary, pancreas, prostate, and stomach; leukemias; benign and malignant lymphomas, particularly Burkitfs lymphoma and Non-Hodgkin's lymphoma; benign and malignant melanomas; myeloproliferative diseases; sarcomas, including Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcomas, peripheral neuroepithelioma, synovial sarcoma, gliomas, astrocytomas, oligodendrogliomas, ependymomas, gliobastomas, neuroblastomas, ganglioneuromas, gangliogliomas, medulloblastomas, pineal cell tumors, meningiomas, meningeal sarcomas, neurofibromas, and Schwannomas; bowel cancer, breast cancer, prostate cancer, cervical cancer, uterine cancer, lung cancer, ovarian cancer, testicular cancer, thyroid cancer, astrocytoma, esophageal cancer, pancreatic cancer, stomach cancer, liver cancer, colon cancer, melanoma; carcinosarcoma, Hodgkin's disease, Wilms' tumor and teratocarcinomas. Additional cancers which may be treated using the disclosed compounds according to the present invention include, for example, acute granulocytic leukemia, acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), adenocarcinoma, adenosarcoma, adrenal cancer, adrenocortical carcinoma, anal cancer, anaplastic astrocytoma, angiosarcoma, appendix cancer, astrocytoma, Basal cell carcinoma, B-Cell lymphoma, bile duct cancer, bladder cancer, bone cancer, bone marrow cancer, bowel cancer, brain cancer, brain stem glioma, breast cancer, triple (estrogen, progesterone and HER-2) negative breast cancer, double negative breast cancer (two of estrogen, progesterone and HER-2 are negative), single negative (one of estrogen, progesterone and HER-2 is negative), estrogen-receptor positive, HER2negative breast cancer, estrogen receptor-negative breast cancer, estrogen receptor positive breast cancer, metastatic breast cancer, luminal A breast cancer, luminal B breast cancer, Her2-negative breast cancer, HER2-positive or negative breast cancer, progesterone receptor-negative breast cancer, progesterone receptor-positive breast cancer, recurrent breast cancer, carcinoid tumors, cervical cancer, cholangiocarcinoma, chondrosarcoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), colon cancer, colorectal cancer, craniopharyngioma, cutaneous lymphoma, cutaneous melanoma, diffuse astrocytoma, ductal carcinoma in situ (DCIS), endometrial cancer, ependymoma, epithelioid sarcoma, esophageal cancer, ewing sarcoma, extrahepatic bile duct cancer, eye cancer, fallopian tube cancer, fibrosarcoma, gallbladder cancer, gastric cancer, gastrointestinal cancer, gastrointestinal carcinoid cancer, gastrointestinal stromal tumors (GIST), germ cell tumor glioblastoma multiforme (GBM), glioma, hairy cell leukemia, head and neck cancer, hemangioendothelioma, Hodgkin lymphoma, hypopharyngeal cancer, infiltrating ductal carcinoma (IDC), infiltrating lobular carcinoma (ILC), inflammatory breast cancer (IBC), intestinal Cancer, intrahepatic bile duct cancer, invasive/infiltrating breast cancer, Islet cell cancer, jaw cancer, Kaposi sarcoma, kidney cancer, laryngeal cancer, leiomyosarcoma, leptomeningeal metastases, leukemia, lip cancer, liposarcoma, liver cancer, lobular carcinoma in situ, low-grade astrocytoma, lung cancer, lymph node cancer, lymphoma, male breast cancer, medullary carcinoma, medulloblastoma, melanoma, meningioma, Merkel cell carcinoma, mesenchymal chondrosarcoma, mesenchymous, mesothelioma metastatic breast cancer, metastatic melanoma metastatic squamous neck cancer, mixed gliomas, monodermal teratoma, mouth cancer mucinous carcinoma, mucosal melanoma, multiple myeloma, Mycosis Fungoides, myelodysplastic syndrome, nasal cavity cancer, nasopharyngeal cancer, neck cancer, neuroblastoma, neuroendocrine tumors (NETs), non-Hodgkin's lymphoma, non-small cell lung cancer (NSCLC), oat cell cancer, ocular cancer, ocular melanoma, oligodendroglioma, oral cancer, oral cavity cancer, oropharyngeal cancer, osteogenic sarcoma, osteosarcoma, ovarian cancer, ovarian epithelial cancer ovarian germ cell tumor, ovarian primary peritoneal carcinoma, ovarian sex cord stromal tumor, Paget's disease, pancreatic cancer, papillary carcinoma, paranasal sinus cancer, parathyroid cancer, pelvic cancer, penile cancer, peripheral nerve cancer, peritoneal cancer, pharyngeal cancer, pheochromocytoma, pilocytic astrocytoma, pineal region tumor, pineoblastoma, pituitary gland cancer, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, renal cell carcinoma, renal pelvis cancer, rhabdomyosarcoma, salivary gland cancer, soft tissue sarcoma, bone sarcoma, sarcoma, sinus cancer, skin cancer, small cell lung cancer (SCLC), small intestine cancer, spinal cancer, spinal column cancer, spinal cord cancer, squamous cell carcinoma, stomach cancer, synovial sarcoma, T-cell lymphoma, testicular cancer, throat cancer, thymoma/thymic carcinoma, thyroid cancer, tongue cancer, tonsil cancer, transitional cell cancer, tubal cancer, tubular carcinoma, undiagnosed cancer, ureteral cancer, urethral cancer, uterine adenocarcinoma, uterine cancer, uterine sarcoma, vaginal cancer, vulvar cancer, T-cell lineage acute lymphoblastic leukemia (T-ALL), T-cell lineage lymphoblastic lymphoma (T-LL), peripheral T-cell lymphoma, Adult T-cell leukemia, Pre-B ALL, Pre-B lymphomas, large B-cell lymphoma, Burkitt's lymphoma, B-cell ALL, Philadelphia chromosome positive ALL, Philadelphia chromosome positive CML, juvenile myelomonocytic leukemia (JMML), acute promyelocytic leukemia (a subtype of AML), large granular lymphocytic leukemia, Adult T-cell chronic leukemia, diffuse large B cell lymphoma, follicular lymphoma; Mucosa-Associated Lymphatic Tissue lymphoma (MALT), small cell lymphocytic lymphoma, mediastinal large B cell lymphoma, nodal marginal zone B cell lymphoma (NMZL); splenic marginal zone lymphoma (SMZL); intravascular large B-cell lymphoma; primary effusion lymphoma; or lymphomatoid granulomatosis;; B-cell prolymphocytic leukemia; splenic lymphoma/leukemia, unclassifiable, splenic diffuse red pulp small B-cell lymphoma; lymphoplasmacytic lymphoma; heavy chain diseases, for example, Alpha heavy chain disease, Gamma heavy chain disease, Mu heavy chain disease, plasma cell myeloma, solitary plasmacytoma of bone; extraosseous plasmacytoma; primary cutaneous follicle center lymphoma, T cell/hi stocyte rich large B-cell lymphoma, DLBCL associated with chronic inflammation; Epstein-Barr virus (EBV)+ DLBCL of the elderly; primary mediastinal (thymic) large B-cell lymphoma, primary cutaneous DLBCL -leg type, ALK+ large B-cell lymphoma, plasmablastic lymphoma; large B-cell lymphoma arising in HHV8 -associated multicentric, Castleman disease; B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma, or B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma.
Advantageously, the methods of the present invention may mediate an immune response to vectors used for the delivery of therapeutics, for example by sequestering IgG's reactive to a given vector. Vectors are particularly useful in delivering nucleic acid molecules to cells, for example for gene therapies and vaccines. In certain aspects an effective amount of a heterobifunctional immunoglobulin degrading compound described herein, for example an IgG degrading compound, is administered before, concurrently with, or after administration of a delivery vector. Vectors may be divided broadly into viral vectors and non-viral vectors.
Many viral vectors or virus-associated vectors are known in the art. Such vectors can be used as carriers of a nucleic acid construct into the cell. Constructs may be integrated and packaged into non-replicating, defective viral genomes like Adenovirus, Adeno-associated virus (AAV), or Herpes simplex virus (HSV) or others, including retroviral and lentiviral vectors, for infection or transduction into cells. For example, vectors derived from retroviruses, such as the lentivirus, may be used for long-term gene transfer. Lentiviral vectors are particularly advantageous because they can transduce non-proliferating cells, such as hepatocytes. They also have the added advantage of low immunogenicity. Adeno-associated viral vectors (AAVs) are a family of medium-sized, non- enveloped viruses used as vectors. More than 30 naturally occurring serotypes of AAV are available. Many natural variants in the AAV capsid exist. AAV serotypes include, but are not limited to, AAV serotypes AAVI, AAV2, AAV3, AAV3B, AAV4, AAV5, AAV6, AAV7, AAV7M8, AAV8, AAV8bp, AAV9, AAV10, AAVI 1, AAV 12, AAV13, AAVAnc80, AAVrhlO, AAVPHP.B, and variants of the known or mentioned AAVs or AAVs yet to be discovered or variants or mixtures thereof (see, e.g., WO 2005/033321, which is incorporated herein by reference). Currently, two AAV vectors, LUXTURNA® (voretigene neparvovec-rzyl) and ZOLGENSMA® (onasemnogene abeparvovec-xioi), are approved by the US Food and Drug Administration (FDA). LUXTURNA® is an AAV serotype 2 gene therapy for the treatment of patients having biallelic RPE65 mutation-associated retinal dystrophy. ZOLGENSMA® is an AAV serotype 9 gene therapy for the treatment of pediatric patients less than 2 years of age having bi-all elic SMN1 mutation-associated spinal muscular atrophy (SMA). As of July 2022, there were 292 ongoing clinical trials involving the administration of AAVs registered at ClinicalTrials.gov.
In certain embodiments, the AAV capsid is selected from AAVI capsid, AAV8 capsid, or AAV9 capsid, or variants thereof. In certain embodiments the AAV comprises an AAV capsid having one or substitutions of the amino acid tyrosine (Y) to the amino acid phenylalanine (F) on the surface of the capsid protein. In certain embodiments, the AAV capsid comprises an AAV capsid having one or more substitutions of the amino acid tyrosine (Y) to the amino acid phenylalanine (F) on the surface of the AAV capsid protein. AAV capsid having tyrosine to phenylalanine mutations are known in the art, as described for example in US 8445267, US 8802440, US 9157098, US 9611302, US 9775918, US 9920097, US 10011640, US 10294281, US 10723768, US 10815279, and US 10934327, each of which is incorporated by refence. In certain embodiments, the AAV capsid is an AAV capsid having substitutions selected from Y272F, Y444F, Y447F, T491 V, T494F, Y500F, Y730F, Y733F or a combination thereof.
In certain embodiments, present invention comprises a method that mediates an immune response to the administration of a recombinant adeno-associated virus (rAAV) containing a nucleic acid encoding a therapeutic polypeptide, thereby increasing the efficacy and/or reducing the needed dosage of therapies that use vector deliveries. The rAAV comprises an AAV capsid and the nucleic acid sequence encoding the therapeutic polypeptide in the AAV capsid, wherein the nucleic acid sequence encoding the therapeutic polypeptide is operably linked to a promoter as described herein and AAV inverted terminal repeats (ITRs) required for packaging a nucleic acid into the capsid. The nucleic acid to be packaged comprises therapeutic polypeptide encoding nucleic acid sequences, promoter sequences, and optionally other regulatory sequences as described herein, which are flanked by packaging signals of the AAV genome to facilitate efficient packaging of the nucleic acid into the AAV capsid for delivery and expression in the target cell and/or organ. In certain embodiments, the nucleic acid sequence encoding the therapeutic polypeptide is flanked at the 5’ and 3’ ends by AAV ITRs. For example, a 5’ AAV ITR, the nucleic acid sequence encoding the therapeutic polypeptide, and a 3’ AAV ITR comprise the nucleic acid that is packaged into the AAV capsid. The term “ITR” is defined as a cis genetic element responsible for facilitating the replication and packaging of a nucleic acid during AAV vector production necessary for rAAV generation. In certain embodiments, the ITRs are derived from a different AAV variant than the AAV capsid variant. In those embodiments wherein the source of the ITRs are of one AAV variant and the AAV capsid is from another AAV variant, the resulting vector is defined as pseudotyped. In certain embodiments, the ITRs are derived from the AAV2 variant. In certain embodiments, the ITRs are derived from deleted, or shortened, version of the AAV2 variant (AITR), Typically, an AAV vector genome comprises an AAV 5’ ITR, a nucleic acid encoding a polypeptide operably linked to a promoter and optionally linked to one or more regulatory sequences, and an AAV 3’ ITR. Other arrangements comprising addition of further elements or subtraction of said elements may be suitable. In certain embodiments, a selfcomplementary AAV (scAAV) is provided. As defined herein, the term “Self-complementary AAV” refers to a construct in which a coding nucleic acid sequence carried by an rAAV has been designed to form an intra-molecular double-stranded DNA template. Upon infection, rather than waiting for cell-mediated second strand synthesis, the two complementary regions of scAAV will associate to form one double stranded DNA (dsDNA) unit that is amenable to immediate replication and transcription. In certain embodiments, the ITRs to be used are selected from wild- type, full-length, engineered, shortened, deleted, or a combination thereof. An engineered AAV may be, without limitation, a pseudotyped AAV, a chimeric AAV capsid, a recombinant AAV capsid, or a “humanized” AAV capsid. A deleted, or shortened, version of the 5’ ITR, termed 5’ AITR, has been described in which the D-sequence and terminal resolution sites (TRs) are deleted. In certain embodiments, the ITR comprises a shortened ITR of the AAV2 variant comprises 130 base pairs (bps), wherein the external “a” element is deleted. In certain embodiments, the shorted AAV2 ITR is lengthened during vector DNA amplification using the internal “a” element as a template. In certain embodiments, full-length AAV 5’ and 3’ ITRs are used. In certain embodiments, wild-type AAV 5’ and 3’ ITRs are used. In certain embodiments, engineered ITRs are used. Suitable AAV ITR sequences are known in the art (see, e.g., Yan et al., J Virol. 79(1):364- 379(2005), incorporated herein by reference).
In certain embodiments, the nucleic acid to be packaged comprises one or more regulatory elements such that the total rAAV vector genome is about 2.0 to about 5.5 kilobases (kb) in size. In certain embodiments, the one or more regulatory sequences are selected such that the total rAAV vector genome is about 2.1, 2.3, 2.8, 3.1, 3.2, 3.3, 4.0 kb, 4.7 kb, or 5.2 kb in size. In certain embodiments, the rAAV vector genome approximates the size of a native AAV genome. In certain embodiments, the one or more regulatory sequences are selected such that the total rAAV vector genome is about 4.7 kb in size. In certain embodiments, the one or more regulatory sequences are selected such that the total rAAV vector genome is about 5.2 kb in size. In certain embodiments, the nucleic acid to be packaged comprises one or more regulatory elements selected from a promoter, a Woodchuck Hepatitis virus (WHP) posttranscriptional regulatory element (WPRE), a polyadenylation (poly A) signal, an intron, or a combination thereof. In certain embodiments, the one or more regulatory elements are operably linked to the nucleic acid encoding a therapeutic polypeptide.
In certain embodiments, a packaging cell line is used for production of a vector (e.g., a recombinant AAV). A host cell may be a prokaryotic or eukaryotic cell (e.g., human, insect, or yeast) that contains exogenous or heterologous DNA that has been introduced into the cell by any means, e.g., electroporation, calcium phosphate precipitation, microinjection, transformation, viral infection, transfection, liposome delivery, membrane fusion techniques, high velocity DNA- coated pellets, viral infection, and protoplast fusion. Examples of host cells may include, but are not limited to an isolated cell, a cell culture, an Escherichia coli cell, a yeast cell, a human cell, a non-human cell, a mammalian cell, a non-mammalian cell, an insect cell, a HEK-293 cell, a liver cell, a kidney cell, a cell of the central nervous system, a neuron, a glial cell, or a stem cell. In certain embodiments, the nucleic acid encoding a therapeutic polypeptide is optimized for expression in the desirable subject species, for example, in humans. Codon-optimized coding nucleic acid sequences can be designed by various different methods known to those skilled in the art. In certain embodiments, the entire length of the open reading frame (ORF) of the nucleic acid encoding a therapeutic polypeptide is modified. In certain embodiments, a fragment of the open reading frame (ORF) of the nucleic acid encoding a therapeutic polypeptide is modified. In certain embodiments, the nucleic acid encoding a therapeutic polypeptide is optimized for tropism towards the desirable target organ. In certain embodiments, the target organ is selected from muscle, eye, liver, brain, or a combination thereof. In certain embodiments, the nucleic acid encoding a therapeutic polypeptide is optimized for tropism towards the desirable target cell-type. As defined herein, the term “target cell” refers to any cell in which the expression of the therapeutic polypeptide is desired. In certain embodiments, the term “target cell” is intended to reference the cells of the subject being treated. In certain embodiments, the target cell-type is selected from neurons, glia, photoreceptors, retinal pigment epithelial (RPE) cells, hepatocytes, myocytes, skeletal muscle cells, heart cells, or a combination thereof. In certain embodiments, the AAV vector is formulated in a buffer/carrier suitable for infusion in a subject, for example a human subject. In certain embodiments, the viral vector (e.g, rAAV) is delivered systemically. In certain embodiments, the rAAV is delivered intravenously, intraperitoneally, intranasally, or via inhalation. In certain embodiments, the buffer/carrier comprises a component that prevents the AAV from attaching to the infusion tube. In certain embodiments, the dosage of the vector is about 1 x 109 GC to about 1 x 1013 genome copies (GC) per dose. In certain embodiments, the dose of the vector administered to a patient is at least about 1.0 x 109 GC/kg , about 1.5 x 109 GC/kg , about 2.0 x 109 GC/g, about 2.5 x 109 GC/kg , about 3.0 x 109 GC/kg , about 3.5 x 109 GC/kg , about 4.0 x 109 GC/kg , about 4.5 x 109 GC/kg , about 5.0 x 109 GC/kg , about 5.5 x 109 GC/kg , about 6.0 x 109 GC/kg , about 6.5 x 109 GC/kg , about 7.0 x 109 GC/kg , about 7.5 x 109 GC/kg , about 8.0 x 109 GC/kg , about 8.5 x 109 GC/kg , about 9.0 x 109 GC/kg , about 9.5 x 109 GC/kg , about 1.0 x 1010 GC/kg , about 1.5 x 1010 GC/kg , about 2.0 x 1010 GC/kg , about 2.5 x 1010 GC/kg , about 3.0 x 1010 GC/kg , about 3.5 x 1010 GC/kg , about 4.0 x 1010 GC/kg , about 4.5 x 1010 GC/kg , about 5.0 x 1010 GC/kg , about 5.5 x 1010 GC/kg , about 6.0 x 1010 GC/kg , about 6.5 x 1010 GC/kg , about 7.0 x 1010 GC/kg , about 7.5 x 1010 GC/kg , about 8.0 x 1010 GC/kg , about 8.5 x 1010 GC/kg , about 9.0 x 1010 GC/kg , about 9.5 x 1010 GC/kg , about 1.0 x 1011 GC/kg , about 1.5 x 1011 GC/kg , about 2.0 x 1011 GC/kg , about 2.5 x 1011 GC/kg , about 3.0 x 1011 GC/kg , about 3.5 x 1011 GC/kg , about 4.0 x 1011 GC/kg , about 4.5 x 1011 GC/kg , about 5.0 x 1011 GC/kg , about 5.5 x 1011 GC/kg , about 6.0 x 1011 GC/kg , about 6.5 x 1011 GC/kg , about 7.0 x 1011 GC/kg , about 7.5 x 1011 GC/kg , about 8.0 x 1011 GC/kg , about 8.5 x 1011 GC/kg , about 9.0 x 1011 GC/kg , about 9.5 x 1011 GC/kg , about 1.0 x 1012 GC/kg , about 1.5 x 1012 GC/kg , about 2.0 x 1012 GC/kg , about 2.5 x 1012 GC/kg , about 3.0 x 1012 GC/kg , about 3.5 x 1012 GC/kg , about 4.0 x 1012 GC/kg , about 4.5 x 1012 GC/kg , 48 about 5.0 x 1012 GC/kg , about 5.5 x 1012 GC/kg , about 6.0 x 1012 GC/kg , about 6.5 x 1012 GC/kg , about 7.0 x 1012 GC/kg , about 7.5 x 1012 GC/kg , about 8.0 x 1012 GC/kg , about 8.5 x 1012 GC/kg , about 9.0 x 1012 GC/kg , about 9.5 x 1012 GC/kg , about 1.0 x 1013 GC/kg , about 1.5 x 1013 GC/kg , about 2.0 x 1013 GC/kg , about 2.5 x 1013 GC/kg , about 3.0 x 1013 GC/kg , about 3.5 x 1013 GC/kg , about 4.0 x 1013 GC/kg , about 4.5 x 1013 GC/kg , about 5.0 x 1013 GC/kg , about 5.5 x 1013 GC/kg , about 6.0 x 1013 GC/kg , about 6.5 x 1013 GC/kg , about 7.0 x 1013 GC/kg , about 7.5 x 1013 GC/kg , about 8.0 x 1013 GC/kg , about 8.5 x 1013 GC/kg , about 9.0 x 1013 GC/kg , about 9.5 x 1013 GC/kg , or about 1.0 x 1014 GC/kg.As used herein, relating to AAV, the term “variant” means any AAV sequence which is derived from a known AAV sequence, including those with a conservative amino acid replacement, and those sharing at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or greater sequence identity over the amino acid or nucleic acid sequence. In certain embodiments, the AAV capsid includes variants which may include up to about 10% variation from any described or known AAV capsid sequence. In certain embodiments, the AAV capsid shares about 90% identity to about 99.9 % identity, about 95% to about 99% identity, or about 97% to about 98% identity to an AAV capsid provided herein and/or known in the art. In one embodiment, the AAV capsid shares at least 95% identity with an AAV capsid. When determining the percent identity of an AAV capsid, the comparison may be made over any of the variable proteins (e.g., vpl, vp2, or vp3). The term “percent (%) identity”, “sequence identity”, “percent sequence identity”, or “percent identical” in the context of nucleic acid sequences or amino acid sequences refers to the residues in the two sequences which are the same when aligned for correspondence. Unless otherwise specified, it is to be understood that a percentage of identity is a minimum level of identity and encompasses all higher levels of identity up to 100% identity to the reference sequence. For example, “95% identity” and “at least 95% identity” may be used interchangeably and include 95, 96, 97, 98, 99 up to 100% identity to the referenced sequence, and all fractions therebetween. The length of sequence identity comparison may be over the full-length of the genome, the full-length of a gene coding sequence, a fragment of a gene coding sequence, a fragment of at least about 500 to about 5000 nucleotides, the full-length of an amino acid sequence, a fragment of an amino acid sequence, a fragment of at least about 8 to about 200 amino acids, is desired. Generally, when referring to “identity”, “homology”, or “similarity” between two different sequences, “identity”, “homology” or “similarity” is determined in reference to “aligned” sequences. “Aligned” sequences or “alignments” refer to more than one nucleic acid sequences or amino acid sequences, often containing corrections for missing or additional bases or amino acids as compared to a reference sequence. Multiple sequence alignment programs are also available for nucleic acid sequences and amino acid sequences. Examples of such programs include, “Clustal Omega”, “Clustal W”, “CAP Sequence Assembly”, “BLAST”, “MAP”, and “MEME”, which are accessible through Web Servers on the internet. Other sources for such programs are known to those skilled in the art.
Non-viral delivery systems may also be used together with the present invention, for example a liposome, lipid nanoparticle, or other lipid formulations. For example, a nucleic acid for delivery may be associated with a lipid. The nucleic acid associated with a lipid may be encapsulated in the aqueous interior of a liposome, interspersed within the lipid bilayer of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the oligonucleotide, entrapped in a liposome, complexed with a liposome, dispersed in a solution containing a lipid, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with a micelle, or otherwise associated with a lipid. The nucleic acid for delivery may be encapsulated in a lipid nanoparticle. Lipid nanoparticles can be comprised of a cationic lipid, an anionic lipid, a polyethylene glycol functionalized lipid, and steroids. Lipid, lipid/DNA or lipid/expression vector associated compositions are not limited to any particular structure in solution. For example, they may be present in a bilayer structure, as micelles, or with collapsed structure. They may also simply be interspersed in a solution, possibly forming aggregates that are not uniform in size or shape. Lipids are fatty substances which may be naturally occurring or synthetic lipids. For example, lipids include the fatty droplets that naturally occur in the cytoplasm as well as the class of compounds which contain long-chain aliphatic hydrocarbons and their derivatives, such as fatty acids, alcohols, amines, amino alcohols, and aldehydes.
Advantageously, the methods of the present invention mediate an immune response to any vector used for the delivery of therapeutics, thereby increasing the efficacy and/or reducing the needed dosage of therapies that use vector deliveries. In certain embodiments, the present invention comprises the use of an effective amount of an immunoglobulin degrading compound described herein, for example and IgG degrading compound, which is to be administered before, concurrently with, or after a vector for the delivery of a therapeutic polypeptide.
Immunoglobulin G
Immunoglobulin G (IgG) is the main type of antibody found in all body fluids (for example, blood and extracellular fluid) and protects against bacterial and viral infections. It represents approximately 75% of serum antibodies in humans and is thus the most common type of antibody found in circulation. IgG antibodies are generated following class switching and maturation of the antibody response, thus they participate predominantly in the secondary immune response.
As a consequence, IgG antibodies frequently mediate a subject's response to therapies, diminishing the effectiveness of those therapies.
IgG can be divided into 4 distinct subclasses: IgGl, IgG2, IgG3, & IgG4. The evolution of IgG subclass switches is regulated by interaction with T cells and follows a I-way direction: IgG3 to IgGl to IgG2 to IgG4. The differences between subclasses are mainly in their size and the configuration of the hinge region, glycosylation sites, and structures, as well as a few key amino acids that impact the ability to interact with complement and FC receptors.
IgGl and IgG3 are monomeric, having 2 heavy chains & 2 light chains, and bivalent, having 2 variable regions. IgG2 has a disulfide bond pattern which allows for two monomeric IgG2 antibodies to form a dimeric and tetravalent structure through unique inter-molecule disulfide bonds. IgG4 has two intrachain disulfide bonds that can be reduced, which generates a monovalent structure. In addition, the monovalent structures can reform the disulfide bonds, but may not be the same IgG4 monovalent chain, meaning the resulting IgG4 will be a bivalent monomer but will have two different variable regions. Binding of IgG can be accomplished through the use of an IgG-specific ligand. For example, the ligand may bind to the FC region of IgG. For example, the IgG-specific ligand may be a Fc-binding peptide, such as a Fc-BP2. In certain aspects , the IgG-specific ligand may be Fc-111.
Figure imgf000026_0001
or a pharmaceutically acceptable salt thereof.
The Protein Data Bank website provides the crystal structure of IgG searchable by 1H3X (Krapp, S., et al., J. Mol. Biol., 2003, 325: 979); and 5V43 (Lee, C.H., et al., Nat. Immunol., 2017, 18: 889-898); as well as the crystal structure of IgG bound to various compounds searchable by 5YC5 (Kiyoshi M., et al., Sci. Rep., 2018, 8: 3955-3955); 5XJE (Sakae Y., et al., Sci. Rep., 2017, 7: 13780-13780); 5GSQ (Chen, C. L., et al., ACS Chem. Biol., 2017, 12: 1335-1345); and 1HZH (Saphire E. O., et al., Science, 2001, 293: 1155-1159). Additionally, Kiyoshi, M., et al., provides insight into the structural basis for binding of human IgGl to its high-affinity human receptor FcyRI. (Kiyosi M., et al., Nat Commun., 2015, 6, 6866).
Representative IgG Targeting Ligands are provided in Fig. IB. Additional representative IgG Targeting Ligands include:
Figure imgf000027_0001
Figure imgf000027_0002
wherein XR is O, S, NH, or N-C1-C3 alkyl; and XM is O, S, NH, or N-C1-C3 alkyl. In other embodiments the IgG Targeting Ligand is selected from: ,
Figure imgf000027_0003
In some embodiments, the IgG Targeting Ligand is a group according to the chemical structure:
Figure imgf000028_0003
wherein RN02 is a dinitrophenyl group optionally linked through CH2, S(O), S(O)2, - S(O)2O, - OS(O)2, or OS(O)2O. In certain embodiments the IgG Targeting Ligand is selected from:
Figure imgf000028_0004
ted from O, CH2, NH, N-C1-C3 alkyl, NC(O)C1-C3 alkyl, S(O), S(O)2, -S(O)2O, - OS(O)2, or OS(O)2O. In some embodiments, the IgG Targeting Ligand is a 3-indoleacetic acid group according to the chemical structure: where k”” is 1-4 (preferably 2-3, most often 3) or a
Figure imgf000028_0001
group.
Figure imgf000028_0002
In some embodiments, the IgG Targeting Ligand is a peptide. Nonlimiting examples of IgG Targeting Ligand peptides include:
PAM (RTY)4K2KG (Fassina, et al, J. Mol. Recognit. 1996, 9, 564-569) comprising SEQ ID NO: 1 RTYK and SEQ ID NO:2 RTYKKG
Figure imgf000029_0001
D-PAM, wherein the amino acids of the PAM sequence are all D-amino acids (Verdoliva, et al, J. Immunol. Methods, 2002, 271, 77-88) SEQ ID NO:3 RTYK (D-amino acids) and SEQ ID NO:4 RTYKKG (D-amino acids).
D-RAM-F, wherein the amino acids of the PAM sequence are all D-amino acids with further modifications wherein the four N-terminal arginines are acetylated with phenylacetic acid (Dinon, et al J. Mol. Recognit. 2011, 24, 1087-1094) SEQ ID NO:5 RTYK (D-amino acids, N- terminal arginine acetylated with phenylacetic acid) and SEQ ID NO: 6 RTYKKG (D-amino acids, N-terminal arginine acetylated with phenylacetic acid).
SEQ ID NO:7 TWKTSRISIF (Krook, et al,.J. Immunol. Methods 1998, 221, 151-157)
SEQ ID NO:8 FGRLVSSIRY (Krook, et al, J. Immunol. Methods 1998, 221, 151-157)
SEQ ID NO: 9 Fc-III (DC AWHLGEL VWCT-NH2) (DeLano et al, Science 2000, 287, 1279-1283)
Figure imgf000030_0001
SEQ ID NO: 10 FCBP-Ser DSAWHLGELWST (see W02014010813) SEQ ID NO: 11 DCHKRSFWADNCT (see W02014010813) SEQ ID NO: 12 DCRTQFRPNQTCT (see W02014010813) SEQ ID NO: 13 DCQLCDFWRTRCT (see W02014010813) SEQ ID NO: 14 DCFEDFNEQRTCT (see W02014010813) SEQ ID NO: 15 DCLAKFLKGKDCT (see W02014010813) SEQ ID NO: 16 DCWHRRTHKTFCT (see W02014010813) SEQ ID NO: 17 DCRTIQTRSCT (see W02014010813) SEQ ID NO: 18 DCIKLAQLHSVCT (see W02014010813) SEQ ID NO: 19 DCWRHRNATEWCT (see W02014010813) SEQ ID NO:20 DCQNWIKDVHKCT (see W02014010813) SEQ ID NO:21 DC AWHLGEL VWCT (see W02014010813) SEQ ID NO:22 DCAFHLGELVWCT (see W02014010813) SEQ ID NO:23 DCAYHLGELVWCT (see W02014010813) SEQ ID NO:24 FcBP-1 DPWVLEGLHWALP (Kang, et al, J. Chromatogr. A 2016, 1466, 105-1 12)
Figure imgf000031_0003
SEQ ID NO:25 FcBP-2 DPTCWVLEGLHWACDLP (Dias, et al, J. Am. Chem. Soc. 2006, 128, 2726-2732)
Figure imgf000031_0002
SEQ ID NO:26 Fc-lll-4c CTCWVLEGLHWACDC (Gong, et al, Bioconjug. Chem. 2016, 27, 1569-1573)
Figure imgf000031_0001
SEQ ID NO:27 EPIHRSTLTALL (Ehrlich, et al, J. Biochem. Biophys. Method 2001, 49, 443— 454)
SEQ ID NO:28 APAR (Camperi, et al, Biotechnol. Lett. 2003, 25, 1545-1548) FcRM (CFHH)2KG(FC Receptor Mimetic, Verdoliva, etal., ChemBioChem 2005, 6, 1242- 1253) SEQ ID NO:29 CFHH
Figure imgf000032_0001
SEQ ID NO:30 HWRGWV (Yang, et al., J Peptide Res. 2006, 66, 1 1 0-137) SEQ ID NO:31 HYFKFD (Yang, et al, J. Chromatogr. A 2009, 1216, 910-918) SEQ ID NO:32 HFRRHL (Menegatti, et al, J. Chromatogr. A 2016, 1445, 93-104) SEQ ID NO:33 HWCitGWV (Menegatti, et al, J. Chromatogr. A 2016, 1445, 93-
104)
SEQ ID NO:34 HWmetCitGWmetV (US 10,266,566) SEQ ID NO:35 D2AAG (Small Synthetic peptide ligand, Lund, et al, J. Chromatogr. A 2012, 1225, 158- 167)
SEQ ID NO:36 DAAG (Small Synthetic peptide ligand, Lund, et al, J. Chromatogr. A 2012, 1225, 158- 167);
SEQ ID NO:37 cyclo[(Na-Ac) S(A)-RWHYFK-Lact-E] (Menegatti, et al, Anal. Chem. 2013, 85, 9229-9237);
SEQ ID NO:38 cyclo[(Na-Ac)-Dap(A)-RWHYFK-Lact-E] (Menegatti, et al, Anal. Chem. 2013, 85, 9229-9237);
SEQ ID NO:39 cyclo[Link M-WFRHYK] (Menegatti, et al, Biotechnol. Bioeng. 2013, 110, 857-870);
SEQ ID NO:40 NKFRGKYK (Sugita, et al, Biochem. Eng. J. 2013, 79, 33-40); SEQ ID NO:41 NARKFYKG (Sugita, et al, Biochem. Eng. J. 2013, 79, 33-40); SEQ ID NO:42 FYWHCLDE (Zhao, et al, Biochem. Eng. J. 2014, 88, 1-11); SEQ ID NO:43 FYCHWALE (Zhao, et al, J Chromatogr. A 2014, 1355, 107-114); SEQ ID NO:44 FYCHTIDE (Zhao, et al., Z Chromatogr. A 2014, 1359, 100-111); SEQ ID NO:45 Dual 1/3 (F YWHCLDE-F Y CHTIDE) (Zhao, et al, J. Chromatogr. A 2014, 1369, 64-72); SEQ ID NO:46 RRGW (Tsai, et al, Anal. Chem. 2014, 86, 293 1-2938); SEQ ID NO:47 KHRFNKD (Yoo and Choi, BioChip J. 2015, 10, 88-94); SEQ ID NO:48 CPSTHWK (Sun et al. Polymers 2018, 10, 778); SEQ ID NO:49 NVQYFAV (Sun et al. Polymers 2018, 10, 778); SEQ ID NO:50 ASHTQKS (Sun et al. Polymers 2018, 10, 778); SEQ ID NO:51 QPQMSHM (Sun et al. Polymers 2018, 10, 778); SEQ ID NO: 52 TNIESLK (Sun et al. Polymers 2018, 10, 778); SEQ ID NO:53 NCHKCWN (Sun et al. Polymers 2018, 10, 778); SEQ ID NO: 54 SHLSKNF (Sun et al. Polymers 2018, 10, 778).
In some embodiments the IgG Targeting Ligand is specific for IgG4.
In some embodiments the IgG4 specific Targeting Ligand is described in Gunnarsson et al. Biomolecular Engineering 2006, 23, 111-117.
In some embodiments the IgG4 specific targeting ligand is selected from SEQ ID NO : 55 FDLLEHFY and SEQ ID NO:56 DLLHHFDYF.
Additional IgG Targeting Ligands include
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
In certain embodiments the immunoglobulin degrading compound is selected from:
Figure imgf000036_0001
or a pharmaceutically acceptable salt thereof;
Figure imgf000036_0002
or a pharmaceutically acceptable salt thereof;
Figure imgf000037_0001
or a pharmaceutically acceptable salt thereof;
Figure imgf000038_0002
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
or a pharmaceutically acceptable salt thereof;
Figure imgf000040_0002
or a pharmaceutically acceptable salt thereof;
Figure imgf000041_0001
or a pharmaceutically acceptable salt thereof;
Figure imgf000041_0002
or a pharmaceutically acceptable salt thereof. Non-limiting examples of IgG degrading compounds include:
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
or a pharmaceutically acceptable salt thereof. In alternative embodiments a hydroxyl, amine, amide, or carboxylic acid group in an Immunoglobulin Targeting Ligand drawn herein is capped with a protecting group. For example in this embodiment:
Figure imgf000057_0001
In alternative embodiments a hydroxyl, amine, amide, or carboxylic acid group in an Immunoglobulin Targeting Ligand drawn herein is used as the attachment point to Linker instead of the drawn attachment point. For example, in this embodiment:
Figure imgf000057_0002
In certain embodiments the immunoglobulin degrading compound is selected from:
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
or a pharmaceutically acceptable salt thereof. In certain embodiments the immunoglobulin degrading compound is selected from:
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
In certain embodiments the Immunoglobulin Targeting Ligand is:
Figure imgf000082_0001
In certain embodiments the Immunoglobulin Targeting Ligand is:
Figure imgf000082_0002
In certain embodiments the Immunoglobulin Targeting Ligand is:
Figure imgf000083_0001
Immunoglobulin E (IgE)
Immunoglobulin E (IgE) is a strong mediator of allergic disease, including but not limited to, atopic asthma, allergic rhinitis, atopic dermatitis, cutaneous contact hypersensitivity, IgE- mediated food allergy, IgE-mediated animal allergies, allergic conjunctivitis, allergic urticaria, anaphylactic shock, nasal polyposis, keratoconjunctivitis, mastocytosis, eosinophilic gastrointestinal disease, bullous pemphigoid, chemotherapy induced hypersensitivity reaction, seasonal allergic rhinitis, interstitial cystitis, eosinophilic esophagitis, angioedema, acute interstitial nephritis, atopic eczema, eosinophilic bronchitis, chronic obstructive pulmonary disease, gastroenteritis, hyper-IgE syndrome (Job's Syndrome), IgE monoclonal gammopathy, monoclonal gammopathy of undetermined significance (MGUS), pemphigus vulgaris, mucus membrane pemphigoid, chronic urticaria, autoimmune uveitis, rheumatoid arthritis, autoimmune pancreatitis, and allergic rhinoconjunctivitis among others. In certain embodiments the immunoglobulin degrading compound is:
Figure imgf000084_0001
or a pharmaceutically acceptable salt thereof.
In certain embodiments the immunoglobulin degrading compound is:
Figure imgf000084_0002
or a pharmaceutically acceptable salt thereof.
In certain embodiments the immunoglobulin degrading compound is:
Figure imgf000084_0003
or a pharmaceutically acceptable salt thereof. In certain embodiments the immunoglobulin degrading compound is:
Figure imgf000085_0001
or a pharmaceutically acceptable salt thereof.
In certain embodiments the immunoglobulin degrading compound is:
Figure imgf000085_0002
or a pharmaceutically acceptable salt thereof.
In certain embodiments the immunoglobulin degrading compound is:
Figure imgf000085_0003
or a pharmaceutically acceptable salt thereof. In certain embodiments the Immunoglobulin Targeting Ligand is:
Figure imgf000086_0001
In certain embodiments the IgE Targeting Ligand is selected from
Figure imgf000086_0002
Figure imgf000087_0001
Non-limiting examples of IgE degrading compounds include:
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Immunoglobulin A (IgA)
Aberrant expression of immunoglobulin A (IgA) mediates a range of autoimmune and immune-mediated disorders, including IgA nephropathy (also known as Berger’s disease), celiac disease, Crohn’s disease, Henoch-Schonlein purpura (HSP) (also known as IgA vasculitis), IgA pemphigus, dermatitis herpetiformis, inflammatory bowel disease (IBD), Sjogren's syndrome, ankylosing spondylitis, alcoholic liver cirrhosis, acquired immunodeficiency syndrome, IgA multiple myeloma, a-chain disease, IgA monoclonal gammopathy, monoclonal gammopathy of undetermined significance (MGUS), linear IgA bullous dermatosis, rheumatoid arthritis, ulcerative colitis, and primary glomerulonephritis, among others.
Specific degradation of IgA can be accomplished through the use of an IgA-specific Immunoglobulin Targeting Ligand. In certain embodiments, the Immunoglobulin Targeting Ligand used is an Opt peptide. Variations and derivatives of the IgA-specific Opt peptide suitable for use as IgA-specific Immunoglobulin Targeting Ligands are described in Hatanaka et al. Journal of Biological Chemistry , 287(57) 43126-43136. In certain embodiments, the IgA-specific Immunoglobulin Targeting Ligand is Opt-1. In certain embodiments, the IgA-specific
Immunoglobulin Targeting Ligand is Opt-2. In certain embodiments, the IgA-specific
Immunoglobulin Targeting Ligand is Opt-3.
In certain embodiments the immunoglobulin degrading compound is:
Figure imgf000091_0001
or a pharmaceutically acceptable salt thereof. In certain embodiments the immunoglobulin degrading compound is:
Figure imgf000092_0001
or a pharmaceutically acceptable salt thereof.
In certain embodiments the immunoglobulin degrading compound is:
Figure imgf000092_0002
or a pharmaceutically acceptable salt thereof.
In certain embodiments the Immunoglobulin Targeting Ligand is: .
Figure imgf000093_0002
Linkers In non-limiting embodiments, LinkerA and LinkerB are independently selected from: ; wherein:
Figure imgf000093_0001
R11, R12, R13, R14, R15, R16, R17, R18, R19, and R20 are independently at each occurrence selected from the group consisting of a bond, alkyl, -C(O)-, -C(O)O-, -OC(O)-, -SO2-, -S(O)-, -C(S)-, -C(O)NR6-, -NR6C(O)-, -O-, -S-, -NR6-, -C(R21R21)-, -P(O)(R3)O-, -P(O)(R3)-, a divalent residue of a natural or unnatural amino acid, alkenyl, alkynyl, haloalkyl, alkoxy, aryl, heterocycle, heteroaryl, -CH2CH2-[O-(CH2)2]n-O-, -CH2CH2-[O-(CH2)2]n-NR6-, -CH2CH2-[O- (CH2)2]n-, -[-(CH2)2-O-]n-, -[O-(CH2)2]n-, -[O-CH(CH3)C(O)]n-, -[C(O)-CH(CH3)-O]n-, -[O-CH2C(O)]n-, -[C(O)-CH2-O]n-, a divalent residue of a fatty acid, a divalent residue of an unsaturated or saturated mono- or di-carboxylic acid; each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R21; n is independently selected at each instance from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; R21 is independently at each occurrence selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, F, Cl, Br, I, hydroxyl, alkoxy, azide, amino, cyano, -NR6R7, -NR8S02R3, -NR8S(0)R3, haloalkyl, heteroalkyl, aryl, heteroaryl, and heterocycle; and the remaining variables are as defined herein.
In one embodiment LinkerA is bond and LinkerB is
Figure imgf000094_0001
In one embodiment LinkerB is bond and LinkerA is
Figure imgf000094_0002
In one embodiment, a divalent residue of an amino acid is selected from
Figure imgf000094_0003
Figure imgf000095_0001
wherein the amino acid can be oriented in either direction and wherein the amino acid can be in the L- or D-form.
In one embodiment, a divalent residue of a dicarboxylic acid is generated from a nucleophilic addition reaction:
Figure imgf000095_0002
Non-limiting embodiments of a divalent residue of a dicarboxylic acid generated from a nucleophilic addition reaction include:
Figure imgf000096_0001
As used in the embodiments herein, xx is independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, and 25.
As used in the embodiments herein, yy is independently selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, and 25.
In one embodiment, a divalent residue of a dicarboxylic acid is generated from a condensation reaction:
Figure imgf000096_0002
Non-limiting embodiments of a divalent residue of a dicarboxylic acid generated from a condensation include:
Figure imgf000097_0001
Non-limiting embodiments of a divalent residue of a saturated dicarboxylic acid include:
Figure imgf000097_0002
Non-limiting embodiments of a divalent residue of a saturated dicarboxylic acid include:
Figure imgf000097_0003
Non-limiting embodiments of a divalent residue of a saturated monocarboxylic acid is selected from butyric acid (-0C(0)(CH2)2CH2-), caproic acid (-0C(0)(CH2)4CH2-), caprylic acid (-0C(0)(CH2)5CH2-), capric acid (-0C(0)(CH2)8CH2-), lauric acid (-OC(0)(CH2)IOCH2-), myristic acid (-0C(0)(CH2)i2CH2-), pentadecanoic acid (-OC(O)(CH2)13CH2-), palmitic acid (-OC(O)(CH2)14CH2-), stearic acid (-OC(O)(CH2)16CH2-), behenic acid (-OC(O)(CH2)20CH2-), and lignoceric acid (-OC(O)(CH2)22CH2-); Non-limiting embodiments of a divalent residue of a fatty acid include residues selected from linoleic acid, palmitoleic acid, vaccenic acid, paullinic acid, oleic acid, elaidic acid, gondoic acid, gadoleic acid, nervonic acid, myristoleic acid, and erucic acid:
Figure imgf000098_0001
Non-limiting embodiments of a divalent residue of a fatty acid is selected from linoleic acid (-C(O)(CH2)7(CH)2CH2(CH)2(CH2)4CH2-), docosahexaenoic acid (–C(O)(CH2)2(CHCHCH2)6CH2-), eicosapentaenoic acid (-C(O)(CH2)3(CHCHCH2)5CH2-), alpha-linolenic acid (–C(O)(CH2)7(CHCHCH2)3CH2-) stearidonic acid (-C(O)(CH2)4(CHCHCH2)4CH2-), y-linolenic acid (-C(O)(CH2)4(CHCHCH2)3(CH2)3CH2-), arachidonic acid (-C(O)(CH2)3,(CHCHCH2)4(CH2)4CH2-), docosatetraenoic acid (-C(O)(CH2)5(CHCHCH2)4(CH2)4CH2-), palmitoleic acid (-C(O)(CH2)7CHCH(CH2)5CH2-), vaccenic acid (-C(O)(CH2)9CHCH(CH2)5CH2-), paullinic acid (-C(O)(CH2)11CHCH(CH2)5CH2-), oleic acid (-C(O)(CH2)7CHCH(CH2)7CH2-), elaidic acid (-C(O)(CH2)7CHCH(CH2)7CH2-), gondoic acid (-C(O)(CH2)9CHCH(CH2)7CH2-), gadoleic acid (- C(O)(CH2)7CHCH(CH2)9CH2-), nervonic acid (-C(O)(CH2)13CHCH(CH2)7CH2-), mead acid (- C(O)(CH2)3(CHCHCH2)3(CH2)6CH2-), myristoleic acid (-C(O)(CH2)7CHCH(CH2)3CH2-), and erucic acid (-C(O)(CH2)11CHCH(CH2)7CH2-). In certain embodiments LinkerC is selected from: . wherein:
Figure imgf000099_0001
R22 is independently at each occurrence selected from the group consisting of alkyl, -C(O)N-, -NC(O)-, -N-, -C(R21)-, -P(O)O-, -P(O)-, -P(O)(NR6R7)N-, alkenyl, haloalkyl, aryl, heterocycle, and heteroaryl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R21; and the remaining variables are as defined herein. In certain embodiments LinkerD is selected from: ; wherein:
Figure imgf000099_0002
R32 is independently at each occurrence selected from the group consisting of alkyl, N+X", -C-, alkenyl, haloalkyl, aryl, heterocycle, and heteroaryl, each of which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from R21;
X- is an anionic group, for example Br- or Cl-; and all other variables are as defined herein.
In certain embodiments LinkerA is selected from:
Figure imgf000100_0002
each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
In certain embodiments LinkerA is selected from:
Figure imgf000100_0001
each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein. In certain embodiments LinkerA is selected from:
Figure imgf000101_0001
each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
In certain embodiments LinkerA is selected from:
Figure imgf000101_0002
each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
In certain embodiments LinkerA is selected from:
Figure imgf000101_0003
each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
In certain embodiments LinkerA is selected from:
Figure imgf000101_0004
each of which is optionally substituted with 1, 2, 3, or 4 optional substituents as defined herein.
In certain embodiments LinkerA is selected from:
Figure imgf000102_0001
wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
In certain embodiments LinkerA is selected from:
Figure imgf000102_0002
wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
In certain embodiments LinkerA is selected from:
Figure imgf000102_0003
Figure imgf000103_0001
wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
In certain embodiments LinkerB is selected from:
Figure imgf000103_0002
In certain embodiments LinkerB is selected from:
Figure imgf000103_0003
Figure imgf000104_0001
In certain embodiments LinkerB, LinkerC, or LinkerD is selected from:
Figure imgf000104_0002
Figure imgf000105_0001
wherein tt is independently selected from 1, 2, or 3 and ss is 3 minus tt. In certain embodiments LinkerB, LinkerC, or LinkerB is selected from:
Figure imgf000105_0002
wherein tt and ss are as defined herein.
In certain embodiments LinkerB, LinkerC, or LinkerD is selected from:
Figure imgf000105_0003
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence; and tt and ss are as defined herein.
In certain embodiments LinkerB, LinkerC, or LinkerD is selected from:
Figure imgf000110_0002
wo 2023/009554
Figure imgf000111_0001
Figure imgf000112_0001
wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence; and tt and ss are as defined herein. In certain embodiments LinkerB, LinkerC, or LinkerD is selected from:
Figure imgf000113_0001
wherein each heteroaryl and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence; and tt and ss are as defined herein.
In certain embodiments LinkerA is selected from:
Figure imgf000113_0002
In certain embodiments LinkerA is selected from:
Figure imgf000113_0003
Figure imgf000114_0001
In certain embodiments LinkerB is selected from:
Figure imgf000115_0001
In certain embodiments LinkerB is selected from:
Figure imgf000115_0002
In certain embodiments LinkerB is selected from:
Figure imgf000115_0003
Figure imgf000116_0001
In certain embodiments LinkerB is selected from:
Figure imgf000117_0001
Figure imgf000118_0002
In certain embodiments LinkerC is selected from:
Figure imgf000118_0001
Figure imgf000119_0001
In certain embodiments LinkerC is selected from:
Figure imgf000119_0002
Figure imgf000120_0001
In certain embodiments LinkerC is selected from:
Figure imgf000120_0002
Figure imgf000121_0001
In certain embodiments LinkerC is selected from:
Figure imgf000121_0002
Figure imgf000122_0001
In certain embodiments LinkerC is selected from:
Figure imgf000122_0002
Figure imgf000123_0001
Figure imgf000124_0001
In certain embodiments, the LinkerA is selected from
Figure imgf000124_0002
Figure imgf000125_0001
In certain embodiments, the LinkerA is selected from
Figure imgf000125_0002
Figure imgf000126_0001
wherein each is optionally substituted with 1, 2, 3, or 4 substituents substituent selected from R21. In certain embodiments LinkerA is selected from:
Figure imgf000126_0002
In certain embodiments LinkerA is selected from:
Figure imgf000126_0003
In certain embodiments LinkerA is selected from:
Figure imgf000126_0004
In certain embodiments LinkerA is selected from:
Figure imgf000127_0001
In certain embodiments LinkerA is selected from:
Figure imgf000127_0002
In certain embodiments LinkerA is selected from:
Figure imgf000127_0003
Figure imgf000128_0001
In certain embodiments LinkerA is selected from:
Figure imgf000128_0002
Figure imgf000129_0001
In certain embodiments LinkerA is selected from:
Figure imgf000129_0002
In certain embodiments LinkerA is selected from:
Figure imgf000130_0001
In certain embodiments LinkerA is selected from:
Figure imgf000130_0002
In certain embodiments, the LinkerB is selected from
Figure imgf000130_0003
In certain embodiments, the LinkerB is selected from
Figure imgf000130_0004
In certain embodiments, the LinkerB is selected from
Figure imgf000130_0005
Figure imgf000131_0001
wherein each is optionally substituted with 1, 2, 3, or 4 substituents substituent selected from R21.
In certain embodiments LinkerB is selected from:
Figure imgf000131_0002
In certain embodiments LinkerB is selected from:
Figure imgf000131_0003
In certain embodiments LinkerB is selected from:
Figure imgf000131_0004
In certain embodiments LinkerB is selected from:
Figure imgf000132_0001
In certain embodiments LinkerB is selected from:
Figure imgf000132_0002
In certain embodiments LinkerB is selected from:
Figure imgf000132_0003
Figure imgf000133_0001
In certain embodiments LinkerB is selected from:
Figure imgf000133_0002
In certain embodiments LinkerB -LinkerA is selected from:
Figure imgf000133_0003
In certain embodiments LinkerB -LinkerA is selected from:
Figure imgf000134_0001
In certain embodiments, the LinkerC is selected from
Figure imgf000134_0002
In certain embodiments, the LinkerC is selected from
Figure imgf000134_0003
In certain embodiments, the LinkerC is selected from
Figure imgf000135_0001
In certain embodiments LinkerC is selected from:
Figure imgf000135_0002
Figure imgf000136_0001
In certain embodiments LinkerC is selected from:
Figure imgf000136_0002
Figure imgf000137_0002
In certain embodiments Linkerc-(LinkerA)2 is selected from:
Figure imgf000137_0001
In certain embodiments Linkerc-(LinkerA)2 is selected from:
Figure imgf000138_0001
In certain embodiments Linkerc-(LinkerA)2 is selected from:
Figure imgf000139_0001
Figure imgf000140_0003
In certain embodiments, the LinkerD is selected from
Figure imgf000140_0001
In certain embodiments, the LinkerD is selected from
Figure imgf000140_0002
In certain embodiments, the LinkerD is selected from
Figure imgf000141_0001
wherein each is optionally substituted with 1, 2, 3, or 4 substituents are selected from R21. In certain embodiments, LinkerB -(LinkerA) is selected from
Figure imgf000141_0002
In certain embodiments, Linker°-(LinkerA) is selected from
Figure imgf000142_0001
In certain embodiments LinkerB is selected from:
Figure imgf000142_0002
wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence. In certain embodiments LinkerB is selected from:
Figure imgf000143_0001
wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
In certain embodiments LinkerB is selected from:
Figure imgf000143_0002
wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
In certain embodiments LinkerB, LinkerC, or LinkerD is selected from:
Figure imgf000143_0003
Figure imgf000144_0003
wherein each heteroaryl, heterocycle, cycloalkyl, and aryl can optionally be substituted with 1, 2, 3, or 4 of any combination of halogen, alkyl, haloalkyl, aryl, heteroaryl, heterocycle, or cycloalkyl, as allowed by valence.
In certain embodiments LinkerA is selected from:
Figure imgf000144_0002
In certain embodiments LinkerA is selected from:
Figure imgf000144_0001
each of which is substituted with 1 or 2 optional substituents
Figure imgf000144_0004
.
In certain embodiments LinkerA is bond.
In certain embodiments the left side of LinkerA is attached to the ASGPR Binding Ligand and the right side is attached to LinkerB, LinkerC, or LinkerD.
In certain embodiments the right side of LinkerA is attached to the ASGPR Binding Ligand and the right side is attached to LinkerB, LinkerC, or LinkerD.
In certain embodiments LinkerB is selected from:
Figure imgf000144_0005
Figure imgf000145_0001
In certain embodiments the left side of LinkerB is attached to the Extracellular Targeting Ligand and the right side is attached to LinkerA.
In certain embodiments the right side of LinkerB is attached to the Extracellular Targeting Ligand and the left side is attached to LinkerA.
In certain embodiments LinkerB is bond.
In alternative embodiments a linker is provided as described above wherein a
Figure imgf000145_0002
, replaced with a
Figure imgf000145_0004
for example where LinkerB is drawn as it is
Figure imgf000145_0006
Figure imgf000145_0005
in this embodiment.
In alternative embodiments a linker is provided as described above wherein a is
Figure imgf000145_0003
replaced with a , for example where LinkerB is drawn as
Figure imgf000145_0007
Figure imgf000146_0001
this embodiment.
In alternative embodiments a linker is provided as described above wherein a
Figure imgf000146_0002
is replaced with a
Figure imgf000146_0003
Asialoglycoprotein Receptor
The asialoglycoprotein receptor (ASGPR) is a Ca2+-dependent receptor. The primary endogenous role of ASGPRs is to help regulate serum glycoprotein levels by mediating endocytosis of glycoproteins. The receptor binds ligands with a terminal galactose or N- acetylgalactosamine. The C3- and C4- hydroxyl groups bind to Ca2+, with the N-acetyl position also been considered important to binding activity.
Figure imgf000146_0004
N-acetyl galactosamine
Asialoglycoproteins bind to ASGPRs and are then cleared by receptor-mediated endocytosis. The receptor and the protein are dissociated in the acidic endosomal compartment and the protein is eventually degraded by lysosomes. The receptor is endocytosed and recycled constitutively from the endosome back to the plasma membrane about every 1 5 minutes regardless of whether or not a glycoprotein is bound. Internalization rate of the receptor may be altered by presence of a bound ligand.
Accordingly, in aspects of the invention, the ligand of the immunoglobulin degrading compound that binds to ASGPR may be an asialoglycoprotein or a derivative thereof.
The ASGPR is comprised of two homologous subunits known as HI and H2. Various ratios of HI and H2 form functional homo- and hetero-oligomers with different conformations, but the most abundant conformation is a trimer composed of two HI and one H2 subunits. The ASGPR is composed of a cytoplasmic domain, a transmembrane domain, a stalk region, and a carbohydrate recognition domain (CRD). Both the HI and H2 subunit form the CRD, and expression of both subunits mediates endocytosis of asialoglycoproteins. ASGPRs are primarily expressed on hepatocytes, and hepatocytes exhibit a high exposition of ASGPR binding cites (approximately 100,000 — 500,000 binding sites per cell).
It is understood that any ligand of ASGPR may be used in immunoglobulin degrading compounds of the invention. For example, ligands that may be used with the invention are described in Stokmaier, Bioorg. Med. Chem., 2009, 17, 7254, which describes the synthesis of a series of D-GalNAc derivatives where the anomeric OH group is removed and the acetamido group is replaced with a 4-substituted 1,2, 3 -triazole moiety, and Mamidyala, JACS, 2012, 134, 1978, which describes compounds derived from 2-azidogalactosyl analogs where the anomeric position is occupied by either a B-methyl or a B-4-methoxy-phenyl group and the azide group is replaced with an amide or a triazole, the entirety of the contest of each of which are incorporated by reference herein.
ASGPR ligands that may be used with the invention are described in PCT Application No. PCT/US21/15939, filed January 29, 2021, and U.S. Provisional Application No. 63/183,450, filed May 3, 2021, the entirety of the contents of each of which are incorporated by reference herein.
For example, the ASGPR ligand may include derivatives of six-carbon pyranose moieties, specifically galactose and talose. These two sugars differ in the stereochemistry of the C 2 substituent. The "down" C 2 configuration corresponds to the stereochemistry of galactose, while the C2 substituent in the "up" configuration corresponds to the stereochemistry oftalose. In aspects of the invention, substituents may be at the C 2 position of the two sugars.
In aspects of the invention the immunoglobulin degrading compounds may utilize a 2:1 ratio of ASGPR ligands to antibody binding ligands. Multiple ASGPR ligands may bind ASGPR more tightly and have increased degradation efficacy. In aspects of the present invention the immunoglobulin degrading compounds may have a 1:1 ratio of ASGPR binding ligands to antibody binding ligands. For example, the compounds may utilize a heteroaryl amine substituents at the C 2 position. The substituents may increase efficacy of the ligand, preferable in compounds with a 1:1 ratio of ASGPR ligand to antibody ligand. For example, in certain embodiments the immunoglobulin degrading molecule is:
Figure imgf000148_0001
or a pharmaceutically acceptable salt thereof.
In aspects of the present invention, the immunoglobulin degrading molecule is:
Figure imgf000148_0002
or a pharmaceutically acceptable salt thereof.
It is understood that any ligand configured to bind to an antibody may be used with the present invention. For example, immunoglobulin degrading compounds may comprise a ligand that binds to IgG, for example an FC binding peptide, for example Fc-111, Fc-BP2, or derivatives thereof that bind the FC portion of the antibody and thus facilitate the selective recruitment of antibody to hepatocytes for degradation, immunoglobulin degrading compounds may comprise a ligand that binds to IgA, for example an FC binding peptide, for example an Opt class of peptides. Opt class peptides are advantageous in that they are highly selective for IgA and thus facilitate the selective recruitment of IgA to hepatocytes for degradation.
For example, in aspects of the invention the immunoglobulin degrading molecule may be.
Figure imgf000149_0001
or a pharmaceutically acceptable salt thereof.
In aspects of the invention, the immunoglobulin degrading molecule of the present invention may be provided as an isotopically enriched immunoglobulin degrading molecule with at least one desired isotopic substitution of an atom, at an amount above the natural abundance of the isotope. For example, deuterium can replace one or more hydrogens in the bispecific compound and 13 c can replace one or more carbon atoms. In aspects of the invention, the isotopic substitution is in one or more positions of the ASGPR ligand. In another aspect, the isotopic substitution is in one or more positions of a linker portion of the molecule. In another embodiment, the isotopic substitution is in the antibody ligand portion of the molecule.
The ASGPR ligand may be a molecule according to the Formula IV, Formula V, Formula
VI, Formula VII, Formula VIII, Formula IX, Formula X, or Formula XI is provided:
Figure imgf000149_0002
or a ph
Figure imgf000150_0001
armaceutically acceptable salt thereof; wherein: R1 and R5 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl- S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; R3 at each occurrence is independently selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, -OR8, and -NR8R9; R6 and R7 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, haloalkyl, heteroaryl, heterocycle, -alkyl-OR8, -alkyl-NR8R9, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R8 and R9 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocycle; R10 is selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R25 is selected from the group consisting of heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl-S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; R65, R66, and R67 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl-S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; R68, R69, and R70 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, F, Cl, Br, I, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl-S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, heteroaryl, aryl, and C0-C6alkyl-O-S(O)2R3 each of which is optionally substituted with 1, 2, 3, or 4 substituents; and when a compound is “optionally substituted” it may be substituted as allowed by valence with one or more groups selected from alkyl (including C1-C4alkyl), alkenyl (including C2- C4alkenyl), alkynyl (including C2-C4alkynyl), haloalkyl (including C1-C4haloalkyl), -OR6, F, Cl, Br, I, -NR6R7, heteroalkyl, heterocycle, heteroaryl, aryl, cyano, nitro, hydroxyl, azide, amide, - SR3, -S(O)(NR6)R3, -NR8C(O)R3, -C(O)NR6R7, -C(O)OR3, -C(O)R3, -SF5, , , , and , wherein the optional substituent is selected such that a stable compound results. In certain embodiments the ASGPR Binding Ligand is selected from: or a pharmaceutically acceptable salt thereof.
In an alternative aspect the ASGPR Binding Ligand is of Formula:
Figure imgf000153_0001
or a pharmaceutically acceptable salt thereof.
For example, the ASGPR ligand or ASGPR ligand-Linker- may be a compound according to the Formula:
Figure imgf000153_0002
Figure imgf000154_0001
Figure imgf000155_0001
These ASGPR ligands and ASGPR ligand-Linkers are typically attached to the Immunoglobulin Targeting Ligand through the C6 position. For example, in certain embodiments the ASGPR ligand or ASGPR ligand-Linker is a compound of Formula:
Figure imgf000155_0002
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0005
In certain aspects the ASGPR ligand is of structure:
Figure imgf000158_0001
In certain aspects the ASGPR ligand is of structure:
Figure imgf000158_0002
In certain aspects the ASGPR ligand is of structure:
Figure imgf000158_0003
In certain aspects the ASGPR ligand is of structure:
Figure imgf000158_0004
Compositions and administration
The heterobifunctional extracellular immunoglobulin degrading compound for use in the present invention or a pharmaceutically acceptable salt, solvate or prodrug thereof as disclosed herein can be administered as a pharmaceutical composition that includes an effective amount for a subject in need of such treatment to mediate the subject’s immune response to a therapy.
In aspects of the invention, the present invention provides a treatment regimen for use in mediating a subject’s immune response to a cell, gene or biologies therapy that includes administration of an effective amount of a pharmaceutical composition comprising an immunoglobulin degrading bifunctional molecule of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog such as a deuterated derivative, or prodrug thereof, and a pharmaceutically acceptable excipient. In aspects of the invention, the cell, gene, or biologies therapy and heterobifunctional immunoglobulin degrading molecule is present in an effective amount, for example a therapeutically effective amount or a prophylactically effective amount. For example, because the heterobifunctional immunoglobulin degrading molecule mediates the subject's immune response, the effective amount of the therapeutic to be delivered may be reduced.
The treatment regimen of the present invention that includes administering an effective amount of therapeutics and/or immunoglobulin degrading molecules can be administered in any manner that allows the immunoglobulin degrading molecule to bind to the immunoglobulin, typically in the blood stream, and carry it to the ASGPR-bearing hepatocyte cells on the liver for endocytosis and degradation. Methods to deliver the therapeutics and immunoglobulin degrading molecules of the present invention may include oral, intravenous, sublingual, subcutaneous, parenteral, buccal, rectal, intra-aortal, intracranial, subdermal or transnasal, or by other means, in dosage unit formulations containing one or more conventional pharmaceutically acceptable carriers, as appropriate.
In aspects of the invention, the treatment regimen of the present invention includes administering an effective amount of therapeutics and/or immunoglobulin degrading molecules which may be administered intravenously. The therapeutics and/or immunoglobulin degrading molecule may be formulated in a liquid dosage form for intravenous injection, such as a buffered solution, for example a phosphate buffered solution and saline buffered solution. The solution may be buffered with multiple salts. In aspects of the invention, the treatment regimen of the present invention includes administering an effective amount of therapeutics and/or immunoglobulin degrading molecule which may be administered orally. The therapeutics and/or immunoglobulin degrading molecule may be formulated in capsules, tablets, and powders, for example a gel containing capsule.
In aspects of the invention, the treatment regimen of the present invention includes administering an effective amount of therapeutics and/or heterobifunctional immunoglobulin degrading molecule which may be administered subcutaneously. The therapeutics and immunoglobulin degrading molecule may be formulated in a liquid dosage form for subcutaneous injection, such as a buffered solution, for example a phosphate buffered solution and saline buffered solution. The solution may be buffered with multiple salts.
In aspects of the invention, a pharmaceutically acceptable salt means a salt of the described therapeutics and/or heterobifunctional immunoglobulin degrading molecule which is suitable for administration to a subject without undue toxicity, irritation, or allergic response, and commensurate with a reasonable benefit to risk ratio, and effective for its intended use. A pharmaceutically acceptable salt means a relatively nontoxic, inorganic and organic acid addition salts of the presently disclosed therapeutics and/or immunoglobulin degrading molecules. These salts can be prepared during the final isolation and purification of the therapeutics and/or immunoglobulin degrading molecules or by separately reacting the purified therapeutics and/or immunoglobulin degrading molecule in its free form with a suitable organic or inorganic acid and then isolating the salt thus formed. Acid addition salts of the basic therapeutics and/or immunoglobulin degrading molecules are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt in the conventional manner. The free base form can be regenerated by contacting the salt form with a base and isolating the free base in the conventional manner. The free base forms may differ from their respective salt forms in certain physical properties such as solubility in polar solvents. Pharmaceutically acceptable base addition salts may be formed with metals or amines, such as alkali and alkaline earth metal hydroxides, or of organic amines. Examples of metals used as cations, include, but are not limited to, sodium, potassium, magnesium, calcium, and the like. Examples of suitable amines include, but are not limited to, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N-methylglucamine, and procaine. The base addition salts of acidic therapeutics and/or immunoglobulin degrading molecules are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. The free acid form can be regenerated by contacting the salt form with an acid and isolating the free acid in a conventional manner. The free acid forms may differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents.
Salts can be prepared from inorganic acids sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, nitric, phosphoric, sulfuric, hydrobromic, hydriodic, phosphorus, and the like. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, lactobionate, laurylsulphonate and isethionate salts, and the like. Salts can also be prepared from organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl -substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. and the like. Representative salts include acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, phthalate, benzenesulfonate, toluenesulfonate, phenyl acetate, citrate, lactate, maleate, tartrate, methanesulfonate, and the like. Pharmaceutically acceptable salts can include cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium and the like, as well as non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to, ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. Also contemplated are the salts of amino acids such as arginate, gluconate, galacturonate, and the like. See, for example, Berge et ah, J. Pharm. Sci., 1977, 66, 1-19, which is incorporated herein by reference.
Pharmaceutically acceptable excipients include inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Additional acceptable excipients include cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, perfuming agents, etc., and combinations thereof.
Diluents may include calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and combinations thereof.
Granulating and/or dispersing agents may include potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation — exchange resins, calcium carbonate, silicates, sodium carbonate, cross — linked poly(vinyl — pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross — linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium compounds, etc., and combinations thereof.
Surface active agents and/or emulsifiers may include natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and Veegum [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g. stearyl alcohol, cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g. polyoxyethylene sorbitan monolaurate [Tween 20], polyoxyethylene sorbitan [Tween 60], polyoxyethylene sorbitan monooleate [Tween 80], sorbitan monopalmitate [Span 40], sorbitan monostearate [Span 60], sorbitan tristearate [Span 65], glyceryl monooleate, sorbitan monooleate [Span 80]), polyoxyethylene esters (e.g. polyoxyethylene monostearate [Myij 45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and Solutol), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g., Cremophor), polyoxyethylene ethers, (e.g . polyoxyethylene lauryl ether [Brij 30]), poly(vinyl — pyrrolidone), di ethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, Pluronic F 68, Poloxamer 188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or combinations thereof. Binding agents may include starch (e.g. cornstarch and starch paste), gelatin, sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol, etc.), natural and synthetic gums (e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl — pyrrolidone), magnesium aluminum silicate (Veegum), and larch arabogalactan), alginates, polyethylene oxide, polyethylene glycol, inorganic calcium salts, silicic acid, polymethacrylates, waxes, water, alcohol, etc., and/or combinations thereof.
Preservatives may include antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, etc., and/or combinations thereof.
Antioxidants may include alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabi sulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabi sulfite, and sodium sulfite.
Any dosage form can be used that achieves the desired results. In aspects of the invention, the pharmaceutical composition is in a dosage form that contains from about 0.1 mg to about 1500 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of the active immunoglobulin degrading molecule and optionally from about 0.1 mg to about 1500 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of an additional active agent in a unit dosage form. Examples are dosage forms with at least about 0.1, 1, 5, 10, 25, 50, 100, 200, 250, 300, 400, 500, 600, 700, 750, 800, 850, 900, 950, or 1,000 mg of active immunoglobulin degrading molecule, or its salt. In certain embodiments the dosage form has at most about 0.1, 1, 5, 10, 25, 50, 100, 200, 250, 300, 400, 500, 600, 700, 750, 800, 850, 900, 950, or 1,000 mg of active immunoglobulin degrading molecule, or its salt.
In aspects of the invention, the dose ranges from about 0.01-100 mg/kg of patient bodyweight, for example about 0.01 mg/kg, about 0.05 mg/kg, about 0. 1 mg/kg, about 0.5 mg/kg, about 1 mg/kg, about 1.5 mg/kg, about 2 mg/kg, about 2.5 mg/kg, about 3 mg/kg, about 3.5 mg/kg, about 4 mg/kg, about 4.5 mg/kg, about 5 mg/kg, about 110 mg/kg, about 115 mg/kg, about 20 mg/kg, about 25 mg/kg, about 30 mg/kg, about 35 mg/kg, about 40 mg/kg, about 45 mg/kg, about 50 mg/kg, about 55 mg/kg, about 60 mg/kg, about 65 mg/kg, about 70 mg/kg, about 75 mg/kg, about 80 mg/kg, about 85 mg/kg, about 90 mg/kg, about 95 mg/kg, or about 100 mg/kg.
In aspects of the invention, heterobifunctional immunoglobulin degrading molecules disclosed herein are used as described are administered once a day (QD), twice a day (BID), or three times a day (TID). In some embodiments, immunoglobulin degrading molecules disclosed herein are used as described are administered at least once a day for at least 1 day, at least 2 days, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least 14 days, at least
15 days, at least 16 days, at least 17 days, at least 18 days, at least 19 days, at least 20 days, at least
21 days, at least 22 days, at least 23 days, at least 24 days, at least 25 days, at least 26 days, at least
27 days, at least 28 days, at least 29 days, at least 30 days, at least 31 days, at least 35 days, at least
45 days, at least 60 days, at least 75 days, at least 90 days, at least 120 days, at least 150 days, at least 180 days, or longer.
In aspects of the invention, the heterobifunctional immunoglobulin degrading molecule is administered once a day, twice a day, three times a day, or four times a day.
The pharmaceutical composition may be formulated as any pharmaceutically useful form, for example, a pill, capsule, tablet, an injection or infusion solution, a syrup, an inhalation formulation, a suppository, a buccal or sublingual formulation, a parenteral formulation, or in a medical device. Some dosage forms, such as tablets and capsules, can be subdivided into suitably sized unit doses containing appropriate quantities of the active components, e.g., an effective amount to achieve the desired purpose.
Carriers include excipients and diluents and must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated. The carrier can be inert, or it can possess pharmaceutical benefits of its own. The amount of carrier employed in conjunction with the therapeutics and/or immunoglobulin degrading molecule is sufficient to provide a practical quantity of material for administration per unit dose of the therapeutics and/or immunoglobulin degrading molecule. If provided as in a liquid, it can be a solution or a suspension.
Representative carriers include phosphate buffered saline, water, solvent(s), diluents, pH modifying agents, preservatives, antioxidants, suspending agents, wetting agent, viscosity agents, tonicity agents, stabilizing agents, and combinations thereof. In some embodiments, the carrier is an aqueous carrier. Examples of aqueous carries include, but are not limited to, an aqueous solution or suspension, such as saline, plasma, bone marrow aspirate, buffers, such as Hank's Buffered Salt Solution (HBSS), HEPES (4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid), Ringers buffer, ProVisc@, diluted ProVisc@, Provisc@ diluted with PBS, Krebs buffer, Dulbecco's PBS, normal PBS, sodium hyaluronate solution (HA, 5 mg/mL in PBS), citrate buffer, simulated body fluids, plasma platelet concentrate and tissue culture medium or an aqueous solution or suspension comprising an organic solvent. Acceptable solutions include, for example, water, Ringer' s solution and isotonic sodium chloride solutions. The formulation may also be a sterile solution, suspension, or emulsion in a non-toxic diluent or solvent such as 1,3-butanediol.
Viscosity agents may be added to the pharmaceutical composition to increase the viscosity of the composition as desired. Examples of useful viscosity agents include, but are not limited to, hyaluronic acid, sodium hyaluronate, carbomers, polyacrylic acid, cellulosic derivatives, polycarbophil, polyvinylpyrrolidone, gelatin, dextin, polysaccharides, polyacrylamide, polyvinyl alcohol (including partially hydrolyzed polyvinyl acetate), polyvinyl acetate, derivatives thereof and mixtures thereof.
Solutions, suspensions, or emulsions for administration may be buffered with an effective amount necessary to maintain a pH suitable for the selected administration. Suitable buffers are well known by those skilled in the art. Some examples of useful buffers are acetate, borate, carbonate, citrate, and phosphate buffers. Solutions, suspensions, or emulsions for topical, for example, ocular administration may also contain one or more tonicity agents to adjust the isotonic range of the formulation. Suitable tonicity agents are well known in the art. Some examples include glycerin, mannitol, sorbitol, sodium chloride, and other electrolytes.
Classes of carriers include, but are not limited to binders, buffering agents, coloring agents, diluents, disintegrants, emulsifiers, flavorants, glidants, lubricants, preservatives, stabilizers, surfactants, tableting agents, and wetting agents. Some carriers may be listed in more than one class, for example vegetable oil may be used as a lubricant in some formulations and a diluent in others. Exemplary pharmaceutically acceptable carriers include sugars, starches, celluloses, powdered tragacanth, malt, gelatin, talc, and vegetable oils. Optional active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the therapeutics and/or immunoglobulin degrading molecule of the present invention. The pharmaceutical compositions/combinations can be formulated for oral administration. These compositions can contain any amount of active therapeutics and/or immunoglobulin degrading molecule that achieves the desired result, for example between 0.1 and 99 weight % (wt.%) of the therapeutics and/or immunoglobulin degrading molecule and usually at least about 5 wt.% of the therapeutics and/or immunoglobulin degrading molecule. Some embodiments contain from about 25 wt. % to about 50 wt. % or from about 5 wt. % to about 75 wt. % of the therapeutics and/or immunoglobulin degrading molecule. Enteric coated oral tablets may also be used to enhance bioavailability of the therapeutics and/or immunoglobulin degrading molecules for an oral route of administration.
Formulations suitable for rectal administration are typically presented as unit dose suppositories. These may be prepared by admixing the active therapeutics and/or immunoglobulin degrading molecule with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
Therapeutics and/or immunoglobulin degrading molecules and pharmaceutically acceptable composition, salts, isotopic analogs, or prodrugs thereof, may be formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions comprising a therapeutics and/or immunoglobulin degrading molecule as described herein will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject or organism will depend upon a variety of factors including the disease, disorder, or condition being treated and the severity of the disorder; the activity of the specific therapeutics and/or immunoglobulin degrading molecule employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific therapeutics and/or immunoglobulin degrading molecule employed; the duration of the treatment; drugs used in combination or coincidental with the specific therapeutics and/or immunoglobulin degrading molecule employed; and like factors well known in the medical arts.
The therapeutics and/or immunoglobulin degrading molecules and compositions provided herein can be administered by any route, including enteral (e.g., oral), parenteral, intravenous, intramuscular, intra — arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, creams, and/or drops), mucosal, nasal, buccal, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; and/or as an oral spray, nasal spray, and/or aerosol. Specifically contemplated routes are oral administration, intravenous administration (e.g., systemic intravenous injection), regional administration via blood and/or lymph supply, and/or direct administration to an affected site. In general, the most appropriate route of administration will depend upon a variety of factors including the nature of the agent (e.g., its stability in the environment of the gastrointestinal tract), the condition of the subject (e.g., whether the subject is able to tolerate oral administration).
The exact amount of the therapeutic and/or immunoglobulin degrading molecule required to achieve an effective amount will vary from subject to subject, depending, for example, on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular therapeutics and/or immunoglobulin degrading molecule(s), mode of administration, and the like. The desired dosage can be delivered using any frequency determined to be useful by the health care provider, including three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage can be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
It will be also appreciated that therapeutics and/or immunoglobulin degrading molecules or compositions, as described herein, can be administered in combination with one or more additional therapeutically active agents. The therapeutics and/or immunoglobulin degrading molecules or compositions can be administered in combination with additional therapeutically active agents that improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body. It will also be appreciated that the therapy employed may achieve a desired effect for the same disorder (for example, a therapeutic and/or immunoglobulin degrading molecule can be administered in combination with an anti — inflammatory agent, anti — cancer agent, immunosuppressant, etc.), and/or it may achieve different effects (e.g., control of adverse side — effects, e.g., emesis controlled by an antiemetic).
The therapeutics and/or immunoglobulin degrading molecule or composition can be administered concurrently with, prior to, or subsequent to, one or more additional therapeutically active agents. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent. It will further be appreciated that the additional therapeutically active agent utilized in this combination can be administered together in a single composition or administered separately in different compositions. The particular combination to employ in a regimen will take into account compatibility of the inventive therapeutics and/or immunoglobulin degrading molecule with the additional therapeutically active agent and/or the desired therapeutic effect to be achieved. In general, it is expected that additional therapeutically active agents utilized in combination be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually.
Additional therapeutically active agents may include, but are not limited to, small organic molecules such as drug compounds (e.g., compounds approved by the Food and Drugs Administration as provided in the Code of Federal Regulations (CFR)), peptides, proteins, carbohydrates, monosaccharides, oligosaccharides, polysaccharides, nucleoproteins, mucoproteins, lipoproteins, synthetic polypeptides or proteins, small molecules linked to proteins, glycoproteins, steroids, nucleic acids, DNAs, RNAs, nucleotides, nucleosides, oligonucleotides, antisense oligonucleotides, lipids, hormones, vitamins and cells. In certain embodiments, the additional therapeutically active agent is an anti -cancer agent, e.g., radiation therapy and/or one or more chemotherapeutic agents.
In certain aspects, a treatment regimen is provided comprising the administration of a therapeutic and/or immunoglobulin degrading molecule of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog (such as a deuterated derivative), or prodrug thereof in combination or in alternation with at least one additional therapeutic agent. The combinations and/or alternations can be administered for beneficial, additive, or synergistic effect in the treatment of immunoglobulin-mediated disorders.
Advantageously, the methods of the present invention mediate an immune response to any additional excipient or combinations used for the delivery of the therapeutics or the immunoglobulin degrading molecule by the action of the immunoglobulin degrading molecule itself.
Additional Embodiments
1. In certain embodiments a method for administering a therapy is provided, the method comprising: sequestering immunoglobulin G (IgG) in a subject so that a therapy can be initiated and readministered to the subject and produce a therapeutic effect in the subject that would not otherwise be achieved due to IgG interacting with the therapy.
2. The method of embodiment 1, wherein sequestering is accomplishing by providing to the subject an immunoglobulin degrading compound comprising: a first ligand configured to bind IgG; and a second ligand configured to bind an asialoglycoprotein receptor (ASGPR).
3. The method of embodiment 1, wherein the therapy is a gene therapy.
4. The method of embodiment 3, wherein the gene therapy comprises use of a vector.
5. The method of embodiment 4, wherein the vector is a viral vector.
6. The method of embodiment 5, wherein the viral vector is an adenovirus-associated virus (AAV) vector.
7. The method of embodiment 2, wherein the immunoglobulin degrading compound sequesters the IgG in the liver for degradation.
8. The method of embodiment 1, wherein the method is conducted prior to the therapy being administered to the subject.
9. A method for administering a therapy, the method comprising: providing to a subject an immunoglobulin degrading compound comprising a first ligand configured to bind IgG, and a second ligand configured to bind an asialoglycoprotein receptor (ASGPR), to thereby sequester and degrade immunoglobulin G (IgG) in the subject so that a therapy can be re-administered to the subject and produce a therapeutic effect in the subject that would not otherwise be achieved due to IgG interacting with the therapy.
10. The method of embodiment 9, wherein the first and second ligand are coupled via one or more linker molecules.
11 . The method of embodiment 9, wherein the therapy is a gene therapy.
12. The method of embodiment 11, wherein the gene therapy comprises use of a vector.
13. The method of embodiment 12, wherein the vector is a viral vector.
14. The method of embodiment 13, wherein the viral vector is an adenovirus-associated virus (AAV) vector.
15. The method of embodiment 9, wherein the immunoglobulin degrading compound sequesters the IgG in the liver for degradation. 16. The method of embodiment 9, wherein sequestering does not provoke an immune response in the subject.
17. The method of embodiment 9, wherein the method is conducted prior to the therapy being readministered to the subject.
18. The method of embodiment 17, wherein the method is conducted after the subject has been administered the therapy at least once.
References and citations to other documents, such as patents, patent applications, patent publications, journals, books, papers, web contents, have been made throughout this disclosure. All such documents are hereby incorporated herein by reference in their entirety for all purposes.
Various modifications of the invention and many further embodiments thereof, in addition to those shown and described herein, will become apparent to those skilled in the art from the full contents of this document, including references to the scientific and patent literature cited herein. The subject matter herein contains important information, exemplification and guidance that can be adapted to the practice of this invention in its various embodiments and equivalents thereof.

Claims

CLAIMS We Claim
1. A method for treating a disorder in a human comprising administering a compound of Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof before, after, or concurrently with the administration of a gene therapy or an adenovirus-associated virus (AAV) vector, wherein:
Figure imgf000171_0001
ASPGR Binding Ligand is a compound selected from:
Figure imgf000171_0002
Figure imgf000172_0001
one of R1 or R5 is a bond to LinkerA; the other of R1 and R5 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl- S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; R3 at each occurrence is independently selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, -OR8, and -NR8R9; R6 and R7 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, haloalkyl, heteroaryl, heterocycle, -alkyl-OR8, -alkyl-NR8R9, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R8 and R9 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocycle; R10 is selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R65, R66, and R67 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl-S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1,
2, 3, or 4 substituents; LinkerA is a bond or a moiety that covalently links LinkerB, LinkerC, or LinkerD to the ASGPR Binding Ligand; LinkerB is a bond or a moiety that covalently links LinkerA to the Immunoglobulin Targeting Ligand; LinkerC is a chemical group that links each LinkerA to the Immunoglobulin Targeting Ligand; LinkerD is a chemical group that links each LinkerA to the Immunoglobulin Targeting Ligand; Immunoglobulin Targeting Ligand is a Ligand that binds to an immunoglobulin; and when a compound is “optionally substituted” it may be substituted as allowed by valence with one or more groups selected from alkyl, alkenyl, alkynyl, haloalkyl, -OR6, F, Cl, Br, I, -NR6R7, heteroalkyl, heterocycle, heteroaryl, aryl, cyano, nitro, hydroxyl, azide, amide, -SR3, -S(O)(NR6)R3, -NR8C(O)R3, -C(O)NR6R7, -C(O)OR3, -C(O)R3, -SF5 , and
Figure imgf000173_0001
, wherein the optional substituent is selected such that a stable compound results.
Figure imgf000173_0002
2. The method of claim 1, wherein a gene therapy is administered.
3. The method of claim 1 or 2, wherein an AAV is administered.
4. The method of any one of claims 1-3, wherein the immunoglobulin is IgG.
5. The method of any one of claims 1-4, wherein the disorder is a loss of function mutation disorder.
6. The method of any one of claims 1-4, wherein the disorder is a monogenic disorder.
7. The method of any one of claims 1-4, wherein the disorder is a disorder caused by a gain of toxicity mutation.
8. The method of any one of claims 1-4, wherein the disorder is a disorder caused by recessive compound heterozygous mutations.
9. The method of any one of claims 1-8, wherein the disorder is selected from Achondroplasia, Alpha-I Antitrypsin Deficiency, Antiphospholipid Syndrome, Autosomal Dominant Polycystic Kidney Disease, Charcot-Marie-Tooth, cancer, Cri du chat, Crohn's Disease, Cystic fibrosis, Dercum Disease, Duane Syndrome, Duchenne Muscular Dystrophy, Factor V Leiden Thrombophilia, Familial Hypercholesterolemia, Familial Mediterranean Fever, Fragile X Syndrome, Gaucher Disease, Hemochromatosis, Hemophilia, Holoprosencephaly, Huntington's disease, Klinefelter syndrome, Marfan syndrome, Myotonic Dystrophy, Neurofibromatosis, Noonan Syndrome, Osteogenesis Imperfecta, Parkinson's disease, Phenylketonuria, Poland Anomaly, Porphyria, Progeria, Retinitis Pigmentosa, Severe Combined Immunodeficiency (SCID), Sickle cell disease, Spinal Muscular Atrophy, Tay- Sachs disease, Thalassemia, Trimethylaminuria, Turner Syndrome, Velocardiofacial Syndrome, WAGR Syndrome, and Wilson Disease.
10. The method of any one of claims 1-8, wherein the disorder is spinal muscular atrophy.
11. The method of any one of claims 1-8, wherein the disorder is mutation-associated retinal dystrophy.
12. The method of any one of claims 1-11, wherein a compound of Formula I is administered.
13. The method of any one of claims 1-11, wherein a compound of Formula II is administered.
14. The method of any one of claims 1-13, wherein LinkerA- A SGPR Binding Ligand is
Figure imgf000174_0001
15. The method of any one of claims 1-13, wherein LinkerA- A SGPR Binding Ligand is
Figure imgf000175_0001
16. The method of any one of claims 1-13, wherein LinkerA- A SGPR Binding Ligand is
Figure imgf000175_0002
17. The method of any one of claims 1-13, wherein LinkerA- A SGPR Binding Ligand is
Figure imgf000175_0003
18. The method of any one of claims 1-17, wherein the Immunoglobulin Targeting Ligand is a compound selected from Figure 1.
19. A method for reducing the effects of an immune response in a human subject, wherein the immune response is induced by the administration of a therapeutic agent, the method comprising administering a compound of Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof before, after, or concurrently with the administration of the therapeutic agent, wherein:
Figure imgf000175_0004
Figure imgf000176_0001
ASPGR Binding Ligand is a compound selected from:
Figure imgf000176_0002
Figure imgf000177_0001
one of R1 or R5 is a bond to LinkerA; the other of R1 and R5 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl- S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; R3 at each occurrence is independently selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, -OR8, and -NR8R9; R6 and R7 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, haloalkyl, heteroaryl, heterocycle, -alkyl-OR8, -alkyl-NR8R9, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R8 and R9 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocycle; R10 is selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R65, R66, and R67 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl-S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; LinkerA is a bond or a moiety that covalently links LinkerB, LinkerC, or LinkerD to the ASGPR Binding Ligand; LinkerB is a bond or a moiety that covalently links LinkerA to the Immunoglobulin Targeting Ligand; LinkerC is a chemical group that links each LinkerA to the Immunoglobulin Targeting Ligand; LinkerD is a chemical group that links each LinkerA to the Immunoglobulin Targeting Ligand; Immunoglobulin Targeting Ligand is a Ligand that binds to an immunoglobulin; and when a compound is “optionally substituted” it may be substituted as allowed by valence with one or more groups selected from alkyl, alkenyl, alkynyl, haloalkyl, -OR6, F, Cl, Br, I, -NR6R7, heteroalkyl, heterocycle, heteroaryl, aryl, cyano, nitro, hydroxyl, azide, amide, -SR3, -S(O)(NR6)R3, -NR8C(O)R3, -C(O)NR6R7, -C(O)OR3, -C(O)R3, -SF5, , , , and , wherein the optional substituent is selected such that a stable compound results.
20. The method of claim 19, wherein the therapeutic agent comprises a vector.
21. The method of claim 20, wherein the vector comprises a viral vector.
22. ‘The method of claim 21, wherein the viral vector is selected from the group consisting of an adenovirus-associated virus (AAV) vector, a lentivirus vector, an adenovirus vector, a respiratory syncytial virus (RSV) vector, a herpes simplex virus (HSV) vector, a poxvirus, or a vaccinia virus.
23. The method of claim 22, wherein the viral vector is an AAV vector.
24. The method of claim 19, wherein the therapeutic agent is a cell therapy.
25. The method of claim 24, wherein the cell therapy is a chimeric antigen receptor (CAR) – T cell therapy.
26. The method of claim 19, wherein the therapeutic agent is a gene therapy.
27. The method of claim 26, wherein the gene therapy replaces a defective endogenous protein.
28. The method of claim 19, wherein the therapeutic agent is a recombinant protein.
29. The method of claim 28, wherein the recombinant protein is selected from the group consisting of adalimumab, infliximab, golimumab. etanercept, certolizumab, ustekinumab, secukinumab, tocilizumab, sarilumab, daclizumab, basiliximab, cankinumab, rituximab, tositumomab, ofatumumab, ibritumomab, abatacept, muromanab, efalizumab, alemtuzumab, gemtuzumab, eculizumab, ultomiris, omalizumab, palivizumab, abciximab, nofetumumab. capromab, fanolesomab, arcitumomab, imciromab, trastuzumab, cetuximab, bevacizumab, panitumumab, ranibizumab, cetolizumab pegol, and denosumab.
30. The method of claim 19, wherein the recombinant protein is a recombinant Factor VIII protein.
31. The method of claim 19, wherein the recombinant protein is recombinant insulin.
32. The method of any of claims 19-31, wherein Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof is administered prior to the administration of the therapeutic agent.
33. The method of any of claims 19-31, wherein Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof is administered concurrently with the administration of the therapeutic agent.
34. The method of any of claims 19-31, wherein Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof is administered after the administration of the therapeutic agent.
35. The method of claim 34, wherein Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof is administered two or more times after the administration of the therapeutic agent.
36. The method of any one of claims 19-35, wherein a compound of Formula I is administered.
37. The method of any one of claims 19-35, wherein a compound of Formula II is administered.
38. The method of any one of claims 19-35, wherein Linkei^-ASGPR Binding Ligand is
Figure imgf000179_0001
39. The method of any one of claims 19-37, wherein Linkei^-ASGPR Binding Ligand is
Figure imgf000180_0001
40. The method of any one of claims 19-37, wherein Linkei^-ASGPR Binding Ligand is
Figure imgf000180_0002
41. The method of any one of claims 19-37, wherein Linkei^-ASGPR Binding Ligand is
Figure imgf000180_0003
42. The method of any one of claims 19-41, wherein the Immunoglobulin Targeting Ligand is a compound selected from Figure 1.
43. Use of a compound of Formula I, Formula II, or Formula III, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for reducing the effects of an immune response in a human subject, wherein the immune response is induced by the administration of a therapeutic agent, wherein the effects of the immune response are reduced by administering the compound of Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof before, after, or concurrently with the administration of the therapeutic agent, wherein:
Figure imgf000180_0004
Figure imgf000181_0001
ASPGR Binding Ligand is a compound selected from:
Figure imgf000181_0002
Figure imgf000182_0001
one of R or R5 is a bond to Linker ; the other of R1 and R5 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl- S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; R3 at each occurrence is independently selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, -OR8, and -NR8R9; R6 and R7 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, haloalkyl, heteroaryl, heterocycle, -alkyl-OR8, -alkyl-NR8R9, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R8 and R9 are independently selected at each occurrence from hydrogen, heteroalkyl, alkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, and heterocycle; R10 is selected from hydrogen, alkyl, heteroalkyl, haloalkyl, arylalkyl, heteroarylalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocycle, C(O)R3, S(O)R3, C(S)R3, and S(O)2R3; R65, R66, and R67 are independently selected from hydrogen, heteroalkyl, C0-C6alkyl-cyano, alkyl, alkenyl, alkynyl, haloalkyl, F, Cl, Br, I, heterocycle, heterocycloalkyl, haloalkoxy, C0-C6alkyl-OR6, C0-C6alkyl-SR6, C0-C6alkyl-NR6R7, C0-C6alkyl-C(O)R3, C0-C6alkyl-S(O)R3, C0-C6alkyl-C(S)R3, C0-C6alkyl-S(O)2R3, C0-C6alkyl-N(R8)-C(O)R3, C0-C6alkyl-N(R8)-S(O)R3, C0-C6alkyl-N(R8)-C(S)R3, C0-C6alkyl-N(R8)-S(O)2R3 C0-C6alkyl-O-C(O)R3, C0-C6alkyl-O-S(O)R3, C0-C6alkyl-O-C(S)R3, -N=S(O)(R3)2, C0-C6alkylN3, and C0-C6alkyl-O-S(O)2R3, each of which is optionally substituted with 1, 2, 3, or 4 substituents; LinkerA is a bond or a moiety that covalently links LinkerB, LinkerC, or LinkerD to the ASGPR Binding Ligand; LinkerB is a bond or a moiety that covalently links LinkerA to the Immunoglobulin Targeting Ligand; LinkerC is a chemical group that links each LinkerA to the Immunoglobulin Targeting Ligand; LinkerD is a chemical group that links each LinkerA to the Immunoglobulin Targeting Ligand; Immunoglobulin Targeting Ligand is a Ligand that binds to an immunoglobulin; and when a compound is “optionally substituted” it may be substituted as allowed by valence with one or more groups selected from alkyl, alkenyl, alkynyl, haloalkyl, -OR6, F, Cl, Br, I, -NR6R7, heteroalkyl, heterocycle, heteroaryl, aryl, cyano, nitro, hydroxyl, azide, amide, -SR3, -S(O)(NR6)R3, -NR8C(O)R3, -C(O)NR6R7, -C(O)OR3, -C(O)R3, -SF5, , , , and , wherein the optional substituent is selected such that a stable compound results.
44. The use of claim 43, wherein the therapeutic agent comprises a vector.
45. The use of claim 44, wherein the vector comprises a viral vector.
46. ‘The use of claim 45, wherein the viral vector is selected from the group consisting of an adenovirus-associated virus (AAV) vector, a lentivirus vector, an adenovirus vector, a respiratory syncytial virus (RSV) vector, a herpes simplex virus (HSV) vector, a poxvirus, or a vaccinia virus.
47. The use of claim 46, wherein the viral vector is an AAV vector.
48. The use of claim 43, wherein the therapeutic agent is a cell therapy.
49. The use of claim 47, wherein the cell therapy is a chimeric antigen receptor (CAR) – T cell therapy.
50. The use of claim 43, wherein the therapeutic agent is a gene therapy.
51. The use of claim 50, wherein the gene therapy replaces a defective endogenous protein.
52. The use of claim 43, wherein the therapeutic agent is a recombinant protein.
53. The use of claim 52, wherein the recombinant protein is selected from the group consisting of adalimumab, infliximab, golimumab. etanercept, certolizumab, ustekinumab, secukinumab, tocilizumab, sarilumab, daclizumab, basiliximab, cankinumab, rituximab, tositumomab, ofatumumab, ibritumomab, abatacept, muromanab, efalizumab, alemtuzumab, gemtuzumab, eculizumab, ultomiris, omalizumab, palivizumab, abciximab, nofetumumab. capromab, fanolesomab, arcitumomab, imciromab, trastuzumab, cetuximab, bevacizumab, panitumumab, ranibizumab, cetolizumab pegol, and denosumab.
54. The use of claim 43, wherein the recombinant protein is a recombinant Factor VIII protein.
55. The use of claim 43, wherein the recombinant protein is recombinant insulin.
56. The use of any of claims 43-55, wherein Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof is administered prior to the administration of the therapeutic agent.
57. The use of any of claims 43-55, wherein Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof is administered concurrently with the administration of the therapeutic agent.
58. The use of any of claims 43-55, wherein Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof is administered after the administration of the therapeutic agent.
59. The use of claim 58, wherein Formula I, Formula II, or Formula III or a pharmaceutically acceptable salt thereof is administered two or more times after the administration of the therapeutic agent.
60. The use of any one of claims 43-59, wherein a compound of Formula I is administered.
61. The use of any one of claims 43-59, wherein a compound of Formula II is administered.
62. The use of any one of claims 43-61, wherein LinkerA-ASGPR Binding Ligand is
Figure imgf000184_0001
63. The use of any one of claims 43-61, wherein LinkerA-ASGPR Binding Ligand is
Figure imgf000185_0001
64. The use of any one of claims 43-61, wherein LinkerA-ASGPR Binding Ligand is
Figure imgf000185_0002
65. The use of any one of claims 43-61, wherein LinkerA-ASGPR Binding Ligand is
Figure imgf000185_0003
66. The use of any one of claims 43-65, wherein the Immunoglobulin Targeting Ligand is a compound selected from Figure 1.
PCT/US2022/038397 2021-07-26 2022-07-26 Methods to reduce adverse effects of gene or biologics therapy WO2023009554A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163225746P 2021-07-26 2021-07-26
US63/225,746 2021-07-26

Publications (1)

Publication Number Publication Date
WO2023009554A1 true WO2023009554A1 (en) 2023-02-02

Family

ID=85087227

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/038397 WO2023009554A1 (en) 2021-07-26 2022-07-26 Methods to reduce adverse effects of gene or biologics therapy

Country Status (1)

Country Link
WO (1) WO2023009554A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160207953A1 (en) * 2014-05-19 2016-07-21 Pfizer Inc. Substituted-6,8-dioxabicyclo[3.2.1]octane-2,3-diol compounds as targeting agents of asgpr
WO2019199621A1 (en) * 2018-04-09 2019-10-17 Yale University Bi-functional molecules to degrade circulating proteins
US20200190515A1 (en) * 2017-06-02 2020-06-18 Wave Life Sciences Ltd. Oligonucleotide compositions and methods of use thereof
WO2021155317A1 (en) * 2020-01-31 2021-08-05 Avilar Therapeutics, Inc. Asgpr-binding compounds for the degradation of extracellular proteins

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160207953A1 (en) * 2014-05-19 2016-07-21 Pfizer Inc. Substituted-6,8-dioxabicyclo[3.2.1]octane-2,3-diol compounds as targeting agents of asgpr
US20200190515A1 (en) * 2017-06-02 2020-06-18 Wave Life Sciences Ltd. Oligonucleotide compositions and methods of use thereof
WO2019199621A1 (en) * 2018-04-09 2019-10-17 Yale University Bi-functional molecules to degrade circulating proteins
WO2021155317A1 (en) * 2020-01-31 2021-08-05 Avilar Therapeutics, Inc. Asgpr-binding compounds for the degradation of extracellular proteins

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SREEMAN K. MAMIDYALA: "Glycomimetic Ligands for the Human Asialoglycoprotein Receptor", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 134, no. 4, 24 January 2012 (2012-01-24), pages 1978 - 1981, XP055048572, DOI: 10.1021/ja2104679 *

Similar Documents

Publication Publication Date Title
US20220395544A1 (en) Compositions for the treatment of disease
US20240124889A1 (en) Compositions and methods for the vectored augmentation of protein destruction, expression and/or regulation
RU2423383C2 (en) Ox40l antibodies
AU2018255293B2 (en) Regulation of RAN translation by PKR and eIF2a-P pathways
CN114144197A (en) Fully human post-translationally modified antibody therapeutics
EP3963055A1 (en) Vectorized antibodies (vab) and uses thereof
Rosenberg et al. AAVrh. 10-mediated expression of an anti-cocaine antibody mediates persistent passive immunization that suppresses cocaine-induced behavior
US8597647B1 (en) Humanized anti-IL-20 antibody and uses thereof
EP3126385B1 (en) Humanized pertussis antibodies and uses thereof
US9682100B2 (en) Cationic polyamines for treatment of viruses
US20230226214A1 (en) Bifunctional bridging compositions for viral transduction
TW202016310A (en) CODON-OPTIMIZED ACID α-GLUCOSIDASE EXPRESSION CASSETTES AND METHODS OF USING SAME
US20220401464A1 (en) Treatment of cyr61- and vegf-mediated conditions
JP2022515570A (en) Treatment of influenza with substituted polycyclic pyridone derivatives and prodrugs thereof in subjects with influenza and complication risk factors
WO2023009554A1 (en) Methods to reduce adverse effects of gene or biologics therapy
JP2020537675A (en) Treatment of neuropathies with DNA constructs that reduce interference from gabapentinoids and express HGF variants
WO2021231575A1 (en) Immunosuppressive agents and viral delivery re-dosing methods for gene therapy
US20240075155A1 (en) Compositions comprising chemerin and methods of use thereof
AU2020216501A1 (en) One-step gene therapy for duchenne muscular dystrophy via gene replacement and anti-inflammation
Wu et al. Half-Life Extension Enhances Drug Efficacy in Adeno-Associated Virus Delivered Gene Therapy
WO2022092326A1 (en) Gdf15 modulator for use in inhibition of ocular tissue fibrosis
JP2024510760A (en) PEGylated T cell engager with dual specificity for CD3 and CD19
BR112021000772A2 (en) ANTIBODIES AGAINST CAMPILOBACTER SPECIES
WO2024049878A2 (en) Bioreducible polymer and use thereof
Castro American Society of Gene and Cell Therapy (ASGCT)-19th Annual Meeting. Washington, DC, USA-May 4-7, 2016

Legal Events

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

Ref document number: 22850208

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE