WO2021222175A1 - Methods for suppressing immune response in gene therapy - Google Patents

Methods for suppressing immune response in gene therapy Download PDF

Info

Publication number
WO2021222175A1
WO2021222175A1 PCT/US2021/029300 US2021029300W WO2021222175A1 WO 2021222175 A1 WO2021222175 A1 WO 2021222175A1 US 2021029300 W US2021029300 W US 2021029300W WO 2021222175 A1 WO2021222175 A1 WO 2021222175A1
Authority
WO
WIPO (PCT)
Prior art keywords
signaling pathway
disrupter
aav
gene therapy
administering
Prior art date
Application number
PCT/US2021/029300
Other languages
French (fr)
Inventor
Roland Herzog
Kumar Sandeep
Original Assignee
The Trustees Of Indiana University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Trustees Of Indiana University filed Critical The Trustees Of Indiana University
Publication of WO2021222175A1 publication Critical patent/WO2021222175A1/en

Links

Classifications

    • 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
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • C07K16/245IL-1
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/15Animals comprising multiple alterations of the genome, by transgenesis or homologous recombination, e.g. obtained by cross-breeding
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0387Animal model for diseases of the immune system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the general field of the present disclosure is gene therapy for patients with hemophilia or other inherited diseases. More particularly, the present disclosure addresses the problem that gene therapy often is complicated by cytotoxic immune responses, which prevent desired gene therapy outcomes.
  • Adeno-associated virus was first discovered from laboratory adenovirus preparations in the mid-1960s and found in human tissues soon after. Several important aspects of AAV were characterized, including its genome configuration and composition, DNA replication and transcription, infectious latency and virion assembly. Subsequently, investigators successfully cloned the wild-type AAV2 sequence into plasmids, which enabled genetic studies and sequencing of the entire AAV2 genome. These early investigations provided fundamental knowledge that led to the use of AAV as a gene delivery vehicle. Since the advent of AAV vectors, their use as a biotherapy has also advanced the understanding of virus-host interactions that govern the transduction pathway of AAV.
  • AAVs are the leading platform for in vivo delivery of gene therapies.
  • the first recombinant AAV gene therapy product alipogene tiparvovec (GLYBERA®)
  • GLYBERA® alipogene tiparvovec
  • LUXTURNA® voretigene neparvovec-rzyl
  • AAV gene therapy research has focused on the liver, striated muscles and the CNS. Almost all natural AAV capsids can transduce liver efficiently following systemic administration. Thus, recombinant AAVs provide a robust liver-targeting platform to treat a variety of diseases such as hemophilia A and hemophilia B, familial hypercholesterolemia, ornithine transcarbamylase deficiency and Crigler-Najjar syndrome. Certain capsids can target multiple muscle types throughout the body, enabling recombinant AAV gene therapies to be developed for multiple muscle diseases, especially those afflicting muscles of the entire body, such as Duchenne muscular dystrophy and the like. In addition, transduced muscle can serve as a bio factory to produce secreted therapeutic proteins for the treatment of non-muscle diseases. Several genes involved in signaling and metabolism have been tested to treat heart failure.
  • FDA US Food and Drug Administration
  • LUXTURNA® voretigene neparvovec-rzyl
  • AAV9 and AAVrh.10 have allowed the vectors to cross the blood brain barrier to transduce neurons and glia.
  • systemic recombinant AAV administration can be used to target diseases that afflict widespread regions of the CNS, including spinal muscular atrophy, amyotrophic lateral sclerosis, Canavan disease, GM1 gangliosidosis and mucopolysaccharidosis type III.
  • Hemophilia is a disease of humans and other mammals wherein a gene encoding a blood coagulation factor contains a mutation such that the encoded protein does not function normally in the cascade process.
  • WFH World Federation of Hemophilia
  • hemophilia A is about 6 times more common than hemophilia B.
  • the promising outcome of liver-directed gene transfer in both hemophilia A and B patients provides a great deal of hope in curing hemophilic patients with a single treatment.
  • adjunct immune modulation may further enforce tolerance to the factor VIII (FVIII) or FIX transgene products.
  • CD8 + T cell responses against the transgene product were observed in patients receiving AAV gene therapy for muscular dystrophy or al -antitrypsin deficiency (Calcedo, R. et al. Class I-restricted T-cell responses to a polymorphic peptide in a gene therapy clinical trial for alpha- 1 -antitrypsin deficiency. PNAS 114, 1655-1659 (2017); Mendell, J.R. et al. Dystrophin immunity in Duchenne's muscular dystrophy. NEJM 363, 1429-1437 (2010)).
  • liver has been shown to induce immunological tolerance to transgene product, concerns remain that AAV encoded therapeutic protein might be recognized as non-self protein and would be a potential target of immune cells. Moreover, an inflammatory milieu due to innate sensing of AAV capsid or other concurrent immune responses could provide relay signal to activate anti-transgene immune response. Thus far, however, there is a scarcity of data on: i) immune responses to transgene in AAV mediated liver gene transfer, ii) innate sensors involved in transgene specific immunity, and, iii) underlying mechanisms of transgene-specific immune responses. Additionally, ongoing clinical studies involving liver gene transfer in hemophilia patients rely on general immune suppression to subdue immune responses to vector and/or transgene product.
  • TLR Toll-like receptors
  • MyD88 myeloid differentiation primary response protein 88
  • FIG. la-e depicts the results of experiments showing that IL-1R1 - MyD88 signaling is essential for CD8 + T cell response to AAV encoded transgene in liver, which can be prevented by neutralization of IL-la and IL-Ib.
  • FIG. la Experimental timeline showing the AAV8-OVA vector administration via tail vein to either C57BL/6-WT mice or knockout of either TLR2 or TLR9 or MyD88 or IL-1R1. To block IL-1 signaling C57BL/6-WT mice were treated with 200ug of either anti-IL-la or anti-IL-Ib or both antibodies via intraperitoneal route.
  • FIG. la depicts the results of experiments showing that IL-1R1 - MyD88 signaling is essential for CD8 + T cell response to AAV encoded transgene in liver, which can be prevented by neutralization of IL-la and IL-Ib.
  • FIG. la Experimental timeline showing the AAV8-OVA vector administration via tail vein to either C57BL/6-WT
  • FIG. lb Similar to wild-type control mice, TLR2 and 9 deficient mice induced CD8 + T cell response whereas mice deficient for MyD88 and IL-1R1 failed to generate CD8 + T cell response against transgene.
  • FIG lc Both cytokines, IL-la and IL-Ib are known to activate IL-1R1. Blocking of either of these cytokines with anti- IL-la and anti-IL-Ib antibodies not only reduced the number of animals making these cellular responses but also reduced the frequency of these CD8 + T cells in these animals. Combination treatment with both anti-IL-la and IL-Ib antibodies further reduced the number of animals that induced transgene specific CD8 + T cell response.
  • FIG. 2a-e shows the results that redundant and non-redundant functions of TLR9 and IL1R signaling in CD8 + T cell activation are vector dose-dependent in AAV muscle gene transfer.
  • FIG. 2a Experimental timeline showing the two doses of AAV 1 -OVA vector administration via muscle to either C57BL/6-WT mice or TLR9 or MyD88 _/ or IL- 1 R 1 knock-out mice.
  • TLR9 knock-out mice were treated with 200ug of either anti -IL- la or IL-Ib or both antibodies via intraperitoneal route.
  • FIG. 2d Blocking of either of cytokine IL-la and IL-Ib with anti-IL-la and IL-Ib antibodies or combination treatment with both anti- IL-la and IL-Ib antibodies reduced the frequency of these CD8 + T cells in TLR9 knock-out mice administrated with the high-dose of AAV1.
  • FIG. 2e Summary of ova-specific CD8 + T cell frequencies in either C57BL/6-WT mice or either IL-1R1 or TLR9 knock-out mice untreated and TLR9 knock-out mice treated with either anti-IL-la or IL-Ib or both IL-la and IL-Ib antibodies or isotype control IgG 2 weeks after high dose of AAVl-OVA administration.
  • the present disclosure provides methods for inhibiting an immune response in a subj ect undergoing AAV gene therapy comprising co-administering to the subject with an AAV gene therapy vector an effective amount of an interleukin-1 (IL-1) signaling pathway disrupter, the IL- 1 signaling pathway disrupter being effective to suppress a cytotoxic CD8 + T cell response against the AAV vector or a transgene product expressed from the AAV vector.
  • the AAV vector can in some embodiments is a liver-targeting AAV or a liver gene transfer vector.
  • the IL-1 signaling pathway disrupter comprises an agonist of the IL-1 signaling pathway that can effectively deactivate IL-la or IL-Ib or both IL-la or IL-Ib.
  • the methods include an IL-1 signaling pathway disrupter that comprises an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor.
  • the IL-1 signaling pathway disrupter comprises an agent effective to deactivate MyD88.
  • co-administration of the signaling pathway disrupter to the subject will occur at a suitable time before, during or following gene therapy treatment either with a single administration or periodically as needed.
  • Yet another embodiment of the current disclosure is a method of treating a mammal comprising: (a) administering a recombinant AAV vector to a mammal; and (b) co-administering an IL-1 signaling pathway disrupter to the mammal in an amount effective to have a therapeutic effect on said mammal and wherein said therapeutic effect is a suppression of an immune response to the gene therapy.
  • the mammal is a human patient in need of gene therapy treatment.
  • the IL-1 signaling pathway disrupter is one or more of an antibody against IL-la, an antibody against IL-Ib or an antibody against an IL-1 receptor.
  • the IL-1 signaling pathway disrupter is a drug such an IL-1 receptor antagonist.
  • the methods include co-administration of the IL-1 signaling pathway disrupter into various tissues of the mammal that include but are not limited to, muscle, the brain, intraperitoneally, intramuscularly, directly into the bone marrow or the like. Administration can also be oral.
  • the IL-1 signaling pathway disrupter is injected at a single site per dose or as multiple doses at multiple sites.
  • the materials to accomplish any of the methods can be packed into a kit that includes all the necessary components to carry out said methods.
  • a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual values within that range, for example, 1.1, 2, 2.3, 4.62, 5, and 5.9. This applies regardless of the breadth of the range.
  • the upper and lower limits of these intervening ranges may independently be included in the smaller ranges, and are also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the disclosure, unless the context clearly dictates otherwise.
  • items included in a list in the form of “at least one of A, B, and C” can mean (A); (B); (C); (A and B); (B and C); (A and C); or (A, B, and C).
  • items listed in the form of “at least one of A, B, or C” can mean (A); (B); (C); (A and B); (B and C); (A and C); or (A, B, and C).
  • the term “about” in reference to a number or range of numbers is understood to mean the stated number and numbers +/- 10% thereof, or 10% below the lower listed limit and 10% above the higher listed limit for the values listed for a range.
  • the present disclosure is based on the discovery that co-administration of an interleukin- 1 (IL-1) signaling pathway disrupter to a mammal with a gene delivery vehicle results in suppression of a cytotoxic CD8 + T cell response against the AAV vector or a transgene product expressed from the AAV vector.
  • IL-1 interleukin- 1
  • suppress or “suppression” or other formatives thereof, and also the terms “inhibit” or “inhibition” or other formatives thereof, when used herein in relation to an immune response, are intended to refer to a reduction in or prevention of the immune response.
  • the disclosure is not limited solely to the treatment of hemophilia. Rather, the disclosure should be construed to include co-administration of an IL-1 signaling pathway disrupter with a variety of DNA encoding gene products that are useful for the treatment of other disease states in a mammal.
  • the gene therapy vector is a liver-targeting AAV, such as, for example, a liver gene transfer vector.
  • the AAV gene therapy comprises AAV mediated liver gene therapy.
  • Alternative DNA incorporated into the AAV gene therapy vector and associated disease states include, but are not limited to: DNA encoding glucose-6-phosphatase, associated with glycogen storage deficiency type 1A; DNA encoding phosphoenolpyruvate-carboxykinase, associated with Pepck deficiency; DNA encoding galactose- 1 phosphate uridyl transferase, associated with galactosemia; DNA encoding phenylalanine hydroxylase, associated with phenylketonuria; DNA encoding branched chain . alpha.
  • DNA encoding fumarylacetoacetate hydrolase associated with tyrosinemia type 1
  • DNA encoding methylmalonyl-CoA mutase associated with methylmalonic acidemia
  • DNA encoding medium chain acyl CoA dehydrogenase associated with medium chain acetyl CoA deficiency
  • DNA encoding ornithine transcarbamylase associated with ornithine transcarbamylase deficiency
  • DNA encoding argininosuccinic acid synthetase associated with citrullinemia
  • DNA encoding low density lipoprotein receptor protein associated with familial hypercholesterolemia
  • DNA encoding UDP-glucouronosyltransferase associated with Crigler-Najjar disease
  • DNA encoding adenosine deaminase associated with severe combined immunodeficiency disease
  • Table 1 is a representation of non-limiting examples.
  • the methods of the disclosure can include without limitation, gene replacement therapies in which the ultimate goal is to deliver a gene product to compensate for loss-of-function mutations.
  • Gene replacement is suitable for treating recessive monogenic diseases.
  • a non-limiting example of gene replacement target is the treatment of hemophilia A or B.
  • the methods of the disclosure can include without limitation, gene silencing where the therapeutic goal is to silence genes that produce toxic mutations.
  • gene silencing where the therapeutic goal is to silence genes that produce toxic mutations.
  • One nonlimiting example is Huntington disease.
  • the methods of the disclosure can include without limitation gene addition therapies.
  • Such therapies can target complex genetic diseases and acquired diseases including but not limited to heart failure and infectious diseases.
  • Gene addition can modulate these diseases in multiple ways, such as supplying neurotrophic factors for neurological diseases and tuning signaling pathways for heart failure, neurotrophic factors for neurological diseases, and tuning signaling pathways for heart failure and cancer.
  • Additional examples of gene addition strategies that the methods of the disclosure can target employ recombinant AAV delivery of genes encoding recombinant antibodies that can neutralize deadly viral infections.
  • Such therapies would utilize intramuscular delivery and transform the transduced muscle cells into a biofactory to produce therapeutic antibodies that are secreted into the bloodstream.
  • Such a strategy could target such infections and HIV, Hepatitis B or Hepatitis C.
  • This disclosure is further based on the discovery that an antibody against IL-1 alpha is particularly effective to suppress an immune response against an AAV vector or a transgene product expressed from the AAV vector. It is contemplated that the delivery of an IL-1 signaling pathway disrupter to a mammal provides a therapeutic benefit to a mammal receiving gene therapy. [0046] The disclosure also includes a method of treating a mammal, preferably, a human, undergoing gene therapy.
  • the present disclosure provides a method for inhibiting an immune response in a subject undergoing AAV gene therapy comprising co-administering to the subject with an AAV gene therapy vector an effective amount of an interleukin- 1 (IL-1) signaling pathway disrupter, the IL-1 signaling pathway disrupter being effective to suppress a cytotoxic CD8 + T cell response against the AAV vector or a transgene product expressed from the AAV vector.
  • the AAV vector can in some embodiments is a liver-targeting AAV or a liver gene transfer vector.
  • the IL-1 signaling pathway disrupter of certain embodiments of the present disclosure may be formulated with a pharmaceutical vehicle or diluent for oral, intravenous, subcutaneous, intranasal, intrabronchial or rectal administration.
  • the pharmaceutical composition can be formulated in a classical manner using solid or liquid vehicles, diluents and additives appropriate to the desired mode of administration. Orally, the composition can be administered in the form of tablets, capsules, granules, powders and the like with at least one vehicle, e.g., starch, calcium carbonate, sucrose, lactose, gelatin, etc.
  • the preparation may also be emulsified.
  • the active ingredient is often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, e.g., water, saline, dextrose, glycerol, ethanol or the like and combination thereof.
  • the composition may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, or adjuvants which enhance the effectiveness of the composition.
  • a preparation for parental administration includes sterilized water, suspension, emulsion, and suppositories.
  • emulsifying agents propylene glycol, polyethylene glycol, olive oil, ethyloleate, etc. may be used.
  • suppositories traditional binders and carriers may include polyalkene glycol, triglyceride, witepsol, macrogol, tween 61, cocoa butter, glycerogelatin, etc.
  • pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like can be used as excipients.
  • the IL-1 signaling pathway disrupter comprises an antagonist of the IL-1 signaling pathway that can effectively deactivate IL-la or IL-Ib or both IL-la or IL-Ib.
  • the methods include an IL-1 signaling pathway disrupter that comprises an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor.
  • the IL-1 signaling pathway disrupter comprises an agent effective to deactivate MyD88.
  • co-administration of the signaling pathway disrupter to the subject occurs at a suitable time before, during or following gene therapy treatment either with a single administration or periodically as needed.
  • Yet another embodiment of the current disclosure is a method of treating a mammal comprising: (a) administering a recombinant AAV vector to a mammal; and (b) co-administering an IL-1 signaling pathway disrupter to the mammal in an amount effective to have a therapeutic effect on said mammal and wherein said therapeutic effect is a suppression of an immune response to the gene therapy.
  • the mammal is a human patient in need of gene therapy treatment.
  • the IL-1 signaling pathway disrupter is one or more of an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor.
  • the IL-1 signaling pathway disrupter is a drug such an IL-1 receptor antagonist.
  • the methods include co-administration of the IL-1 signaling pathway disrupter into various tissues of the mammal that include but are not limited to, muscle, the brain, intraperitoneally, intramuscularly, directly into the bone marrow or the like. Administration can also be oral.
  • the IL-1 signaling pathway disrupter is injected at a single site per dose or as multiple doses at multiple sites.
  • the materials to accomplish any of the methods can be packed into a kit that includes all the necessary components to carry out said methods.
  • liver gene transfer is capable of inducing immunological tolerance to AAV encoded transgene products
  • levels of gene expression as determined by vector dose and design play a critical role in eliciting an adaptive CD8 + T cell response to an AAV encoded transgene (FIG. 1), resulting in loss of the model antigen ovalbumin (OVA) or of FIX expression in hemophilia B mice (Hoffman, B.E. et al. Nonredundant roles of IL-10 and TGF-beta in suppression of immune responses to hepatic AAV-factor IX gene transfer.
  • OVA model antigen ovalbumin
  • FIX FIX expression in hemophilia B mice
  • low levels of hepatic gene expression may elicit an adaptive CD8 + T cell response to an AAV encoded transgene, resulting in loss of the model antigen ovalbumin (OVA) in C57BL/6 mice or of FIX expression in hemophilia B mice.
  • OVA model antigen ovalbumin
  • PBMCs Peripheral blood mononuclear cells
  • DCs dendritic cells
  • cDCs conventional DCs
  • pDCs plasmacytoid DCs
  • TLR9 in pDCs senses AAV genome leading to secretion of type I interferon, which in turn activates cDCs in trans to cross present AAV capsid antigen during CD8 + T-cell activation.
  • This same murine model was used in further studies described below, which demonstrate that unlike AAV, innate sensing of AAV encoded transgene in liver is not regulated by TLR2 or TLR9. Furthermore, the results show a critical role of IL-l/MyD88 signaling pathway in eliciting transgene specific adaptive immune response. Surprisingly, IL-1 mediated signaling has not previously been implicated in adaptive immune response to AAV encoded transgene.
  • the murine model is also used to establish a similar model that establishes a mechanism by which innate immunity against AAV causes IL-l/MyD88 signaling that subsequently drives CD8 + T cell activation against the transgene product in the liver.
  • TLR9-MyD88 pathway is essential for capsid-specific CD8 + T-cell responses.
  • a modified AAV serotype 2 capsid containing immunodominant CD8 + T-cell epitope (SIINFEKL) of the model antigen OVA was used to study capsid specific CD8 + T cells response in C57BL/6 mice. Mice were injected with lxlO 11 vg/mouse of AAV2-SIINFEKL vector via intramuscular route. Using the H2-Kb -SIINFEKL tetramer, it was demonstrated a vector dose dependent capsid-specific CD8 + T cell.
  • CD8 + T cell response peaked between 7 to 10 days post-injection, and largely subsided by day 21. Induced cytotoxic T lymphocytes (CTLs) were functional in an in vivo killing assay. In contrast to wild type mice, TLR9 _/ and MyD88 _/ mice failed to respond to the AAV capsid. No reduction in the CD8 + T-cell response was observed in TLR2 mice, in line with the previous observations that this receptor has limited impact on the adaptive response to AAV.
  • CTLs cytotoxic T lymphocytes
  • IL-1 Receptor IL-1R
  • MyD88-dependent CD8 + T Cell Responses to Hepatic AAV Gene Transfer IL-1 Receptor
  • PBMCs were analyzed by flow cytometry for OVA-specific CD8 + T cells using a class I MHC tetramer. In 50-75% of wild type mice, OVA-specific CD8 + T cells were observed at frequencies of 2-32%.
  • innate sensor TLR9 was dispensable for transgene specific CD8 + T cell response, as 60% of TLR9 mice developed a response, i.e., were positive for OVA specific CD8 + T cells. Similarly, some of the TLR2 mice (37%) also had OVA specific CD8 + T cells, indicating that TLR2 is dispensable for these cellular immune responses. No significant differences were seen in TRIF . IFNaR V or DA5 _/ mice. Recently, double stranded RNA sensors such as RIG-I and MDA-5 have been shown to play critical role in immune responses to AAV mediated gene therapy (Shao, W. et al.
  • RNA sensing Double-stranded RNA innate immune response activation from long-term adeno-associated virus vector transduction. JCI Insight 3 (2016)); however, this study did not imply any role for RNA sensing, as DA-5 _/ mice were observed to have transgene specific CD8 + T cell response similar to WT mice. Interestingly, adaptor protein MyD88 _/ mice did not elicit CD8 + T cell response to OVA, implying an important role for MyD88 in mediating transgene specific cellular immune response. [0081] Since MyD88 is an essential adaptor protein not only for TLRs but also for interleukin- 1 (IL-1) signaling pathways, I L- 1 R 1 mice were analyzed.
  • IL-1 interleukin- 1
  • CD4-deficient mice also failed to elicit an immune response to OVA, likely reflecting a requirement for CD4 + T help. While MyD88 and CD4 were also required for antibody responses against the viral capsid and the transgene product, IL1-R was not.
  • TLR2- and ILlR-deficient mice had a modest reduction in antibody formation against ovalbumin, and these strains as well as MyD88 /_ mice showed a significant increase in circulating ovalbumin levels (reaching 100-200 ng/ml).
  • FIG. la-e depicts the results of experiments showing that IL-1R1 - MyD88 signaling is essential for CD8 + T cell response to AAV encoded transgene in liver, which can be prevented by depletion/neutralization of IL-la and IL-Ib.
  • FIG. la Experimental timeline showing the AAV8- OVA vector administration via tail vein to either C57BL/6-WT mice or knockout of either TLR2 or TLR9 or MyD88 or IL-1R1. To block IL-1 signaling C57BL/6-WT mice were treated with 200ug of either anti-IL-la or anti-IL-Ib or both antibodies via intraperitoneal route.
  • FIG. la depicts the results of experiments showing that IL-1R1 - MyD88 signaling is essential for CD8 + T cell response to AAV encoded transgene in liver, which can be prevented by depletion/neutralization of IL-la and IL-Ib.
  • FIG. la Experimental timeline showing the AAV8- OVA vector administration via tail
  • FIG. lb Similar to wild-type control mice, TLR2 and 9 deficient mice induced CD8 + T cell response whereas mice deficient for MyD88 and IL-1R1 failed to generate CD8 + T cell response against transgene.
  • FIG lc Both cytokines, IL-la and IL-Ib are known to activate IL-1R1. Blocking of either of these cytokines with anti-IL-la and anti -IL-1 b antibodies not only reduced the number of animals making these cellular responses but also reduced the frequency of these CD8 + T cells in these animals. Combination treatment with both anti-IL-la and IL-Ib antibodies further reduced the number of animals that induced transgene specific CD8 + T cell response.
  • FIG. le Ovalbumin levels in C57BL/6-WT untreated and C57BL/6-WT mice treated with either anti-IL-la or IL-Ib or both IL-la and IL-Ib antibodies 4 weeks after AAV8-OVA administration.
  • FIG. 2a-e shows the results that redundant and non-redundant functions of TLR9 and IL1R signaling in CD8 + T cell activation are vector dose-dependent in AAV muscle gene transfer.
  • both TLR9-MyD88 and IL-lRl-MyD88 signaling pathways are required for a CD8 + T cell response against the transgene product.
  • elimination of either prevents the response.
  • these pathways are redundant, so that both need to be blocked to prevent the response.
  • FIG. 2a Experimental timeline showing the two doses of AAV 1 -OVA vector administration via muscle to either C57BL/6-WT mice or TLR9 or MyD88 _/ or I L- 1 R 1 /_ knock-out mice.
  • TLR9 knockout mice were treated with 200ug of either anti-IL-la or anti-IL-Ib or both antibodies via intraperitoneal route.
  • FIG. 2b TLR9 and IL-1R1 deficient mice showed CD8 + T cell responses similar to wild-type control mice, whereas mice deficient for MyD88 failed to generate CD8 + T cell responses against transgene at the high dose (2xlO n vg).
  • FIG. 2c Mice deficient for MyD88 or TLR9 or IL-1R1 had much reduced CD8 + T cell responses against transgene at the low dose (2xl0 10 vg).
  • FIG. 2d Blocking of either of cytokine IL-la and IL-Ib with anti-IL-la and anti-IL-Ib antibodies or combination treatment with both anti-IL-la and anti-IL-Ib antibodies reduced the frequency of these CD8 + T cells in TLR9 knock out mice administrated with the high-dose of AAV1.
  • IL-1 interleukin- 1
  • IL-1 signaling plays a significant role in innate sensing and eliciting adaptive immune responses to various bacteria and viruses (Zwijnenburg, et ak, IL-1 receptor type 1 gene-deficient mice demonstrate an impaired host defense against pneumococcal meningitis. J. Immun. 170, 4724-4730 (2003); Schmitz et ak, Interleukin-1 is responsible for acute lung immunopathology but increases survival of respiratory influenza virus infection. J. Virol. 79, 6441- 6448 (2005); Pang et al., IL-1R signaling in dendritic cells replaces pattern-recognition receptors in promoting CD8 + T cell responses to influenza A virus. Nature Immun. 14, 246-253 (2013)).
  • cytokines IL-la and IL-Ib are both ligands for type I receptor IL-1R1. These cytokines share only 24% amino acid homology, and yet have largely identical biological functions. Though both IL-la and IL-Ib primarily have pro-inflammatory functions, they have been implicated in mediating adaptive immune responses (Voronov, E. et al. Unique Versus Redundant Functions of IL-1 alpha and IL-1 beta in the Tumor Microenvironment. Frontiers Immunol. 4, 177 (2013); Voronov et al., Targeting the Tumor Microenvironment by Intervention in Interleukin-1 Biology. Current Pharmac. Design 23, 4893-4905 (2017); Dinarello, C.A.
  • mice were intravenously (IV) injected with AAV8 vector encoding OVA and also intraperitoneally (IP) injected selected mice with blocking antibodies to IL-la and IL-Ib.
  • IV intravenously
  • IP intraperitoneally
  • mice were injected IP with either anti-IL-la or anti-IL-Ib.
  • mice were IV injected with AAV8 vector encoding OVA.
  • the mice in each group were further treated with either anti- IL-la or anti-IL-Ib twice per week for four weeks. Following four weeks, cellular immune responses to transgene and humoral immune responses to transgene and AAV capsid were evaluated.
  • OVA specific CD8 + T cells were quantified using a tetramer for H2-Kb restricted SIINFEKL peptide.
  • a method for inhibiting an immune response in a subj ect undergoing AAV gene therapy that includes co-administering to the subject with an AAV gene therapy vector an effective amount of an interleukin-1 (IL-1) signaling pathway disrupter, the IL-1 signaling pathway disrupter being effective to suppress a cytotoxic CD8 + T cell response against the AAV vector or a transgene product expressed from the AAV vector.
  • IL-1 interleukin-1
  • the IL- 1 signaling pathway disrupter comprises one or more of an antagonist of the IL-1 signaling pathway, an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor, or an agent effective to deactivate MyD88.
  • the IL- 1 signaling pathway disrupter comprises an agent effective to deactivate IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor.
  • a method in accordance with any other embodiment wherein administering the signaling pathway disrupter to the subject can be accomplished within an appropriate time of a gene therapy treatment.
  • a method of treating a mammal that includes: (a) administering a recombinant adeno associated virus vector to a mammal; and (b) co-administering an IL-1 signaling pathway disrupter to the mammal in an amount effective to have a therapeutic effect on said mammal and wherein said therapeutic effect is a suppression of an immune response to the gene therapy.
  • IL-1 signaling pathway disrupter is one or more of an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor, or an agent effective to deactivate MyD88 or an IL-1 receptor antagonist
  • the methods include co administration of the IL-1 signaling pathway disrupter into various tissues of the mammal that include but are not limited to, muscle, the brain, intraperitoneally, intramuscularly, directly into the bone marrow or the like. Administration can also be oral.
  • a method in accordance with any other embodiment wherein the materials to accomplish any of the methods can be packed into a kit that includes all the necessary components to carry out said methods.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • Environmental Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Plant Pathology (AREA)
  • Animal Husbandry (AREA)
  • Endocrinology (AREA)
  • Mycology (AREA)
  • Transplantation (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Virology (AREA)

Abstract

Gene therapy for patients with hemophilia or other inherited diseases is often complicated by cytotoxic CD8+ T cell responses, which prevent desired gene therapy outcomes. Using a murine model, a therapeutic protocol was discovered against such responses by targeted disruption of the interleukin- 1 signaling pathway by co-administration of an IL-1 signaling pathway disrupter with the gene therapy.

Description

METHODS FOR SUPRESSING IMMUNE RESPONSE IN GENE THERAPY
[0001] REFERENCE TO GOVERNMENT GRANTS
[0002] This invention was made with government support under AI051390 and HL131093 awarded by National Institutes of Health. The government has certain rights.
[0003] FIELD OF THE INVENTION
[0004] The general field of the present disclosure is gene therapy for patients with hemophilia or other inherited diseases. More particularly, the present disclosure addresses the problem that gene therapy often is complicated by cytotoxic immune responses, which prevent desired gene therapy outcomes.
[0005] BACKGROUND
[0006] Adeno-associated virus (AAV) was first discovered from laboratory adenovirus preparations in the mid-1960s and found in human tissues soon after. Several important aspects of AAV were characterized, including its genome configuration and composition, DNA replication and transcription, infectious latency and virion assembly. Subsequently, investigators successfully cloned the wild-type AAV2 sequence into plasmids, which enabled genetic studies and sequencing of the entire AAV2 genome. These early investigations provided fundamental knowledge that led to the use of AAV as a gene delivery vehicle. Since the advent of AAV vectors, their use as a biotherapy has also advanced the understanding of virus-host interactions that govern the transduction pathway of AAV.
[0007] Today, recombinant AAVs are the leading platform for in vivo delivery of gene therapies. The first recombinant AAV gene therapy product, alipogene tiparvovec (GLYBERA®), was approved by the European Medicines Agency to treat lipoprotein lipase deficiency in 2012, while the approval of voretigene neparvovec-rzyl (LUXTURNA®), the first recombinant AAV gene therapy product licensed in the United States, followed 5 years later. Although the clinical success of recombinant AAV gene therapy is encouraging, several limitations and challenges of this gene delivery platform persist, including, for example, issues with recombinant AAV manufacturing and more importantly, immunological barriers to delivery.
[0008] Most recombinant AAV gene therapy research has focused on the liver, striated muscles and the CNS. Almost all natural AAV capsids can transduce liver efficiently following systemic administration. Thus, recombinant AAVs provide a robust liver-targeting platform to treat a variety of diseases such as hemophilia A and hemophilia B, familial hypercholesterolemia, ornithine transcarbamylase deficiency and Crigler-Najjar syndrome. Certain capsids can target multiple muscle types throughout the body, enabling recombinant AAV gene therapies to be developed for multiple muscle diseases, especially those afflicting muscles of the entire body, such as Duchenne muscular dystrophy and the like. In addition, transduced muscle can serve as a bio factory to produce secreted therapeutic proteins for the treatment of non-muscle diseases. Several genes involved in signaling and metabolism have been tested to treat heart failure.
[0009] Recombinant AAV gene therapy focused on the CNS, including the brain and eye, is also under clinical development. As noted, the first recombinant AAV gene therapy drug approved by the US Food and Drug Administration (FDA), voretigene neparvovec-rzyl (LUXTURNA®), treats patients with an inherited form of vision loss caused by RPE65 gene mutations. Regarding the brain, some investigators have found that direct intraparenchymal recombinant AAV injections result in localized distribution of recombinant AAV and are ideal for the treatment of CNS diseases that afflict a defined region of the brain, such as the putamen in Parkinson disease. Delivery to the cerebrospinal fluid space by intrathecal injection, on the other hand, can achieve broader CNS distribution. Alternatively, intravenous delivery of certain serotype vectors, such as AAV9 and AAVrh.10, has allowed the vectors to cross the blood brain barrier to transduce neurons and glia. Thus, systemic recombinant AAV administration can be used to target diseases that afflict widespread regions of the CNS, including spinal muscular atrophy, amyotrophic lateral sclerosis, Canavan disease, GM1 gangliosidosis and mucopolysaccharidosis type III.
[0010] Hemophilia is a disease of humans and other mammals wherein a gene encoding a blood coagulation factor contains a mutation such that the encoded protein does not function normally in the cascade process. According to a recent report of the World Federation of Hemophilia (WFH), there are 28,775 patients worldwide with hemophilia B and 143,523 patients who have hemophilia A. There are around 20,000 hemophilia patients in United States with hemophilia A being about 6 times more common than hemophilia B. The promising outcome of liver-directed gene transfer in both hemophilia A and B patients provides a great deal of hope in curing hemophilic patients with a single treatment. However, although hepatic gene transfer has the potential to induce immune tolerance, adjunct immune modulation may further enforce tolerance to the factor VIII (FVIII) or FIX transgene products.
[0011] Clinical studies have shown that immune responses to AAV vectors and their transgene products represent one of the greatest hurdles to the success of gene therapy. These immune responses encompass both cellular responses against AAV transduced cells and humoral immune responses against the AAV capsid. It took investigators by surprise when two patients in AAV mediated clinical trial of hemophilia B developed cellular immunogenicity against the viral capsid that led to a sudden decline in factor IX (FIX) expression associated with asymptomatic transient transaminitis (Manno, C.S. et al. Successful transduction of liver in hemophilia by AAV -Factor IX and limitations imposed by the host immune response. Nature Med. 12, 342-347 (2006)).
[0012] Similarly, CD8+ T cell responses against the transgene product were observed in patients receiving AAV gene therapy for muscular dystrophy or al -antitrypsin deficiency (Calcedo, R. et al. Class I-restricted T-cell responses to a polymorphic peptide in a gene therapy clinical trial for alpha- 1 -antitrypsin deficiency. PNAS 114, 1655-1659 (2017); Mendell, J.R. et al. Dystrophin immunity in Duchenne's muscular dystrophy. NEJM 363, 1429-1437 (2010)).
[0013] Thus, there is a need to better understand host immune responses directed against AAV vectors.
[0014] Though liver has been shown to induce immunological tolerance to transgene product, concerns remain that AAV encoded therapeutic protein might be recognized as non-self protein and would be a potential target of immune cells. Moreover, an inflammatory milieu due to innate sensing of AAV capsid or other concurrent immune responses could provide relay signal to activate anti-transgene immune response. Thus far, however, there is a scarcity of data on: i) immune responses to transgene in AAV mediated liver gene transfer, ii) innate sensors involved in transgene specific immunity, and, iii) underlying mechanisms of transgene-specific immune responses. Additionally, ongoing clinical studies involving liver gene transfer in hemophilia patients rely on general immune suppression to subdue immune responses to vector and/or transgene product. These immunosuppressive agents increase the risk of opportunistic infections along with their various side effects. Identifying the immune players and understanding the underlying mechanisms of these immune responses (to AAV-capsid and transgene product) would provide new potential targets and pathways that could specifically be blocked (contrary to general immune suppression) to prevent immune responses in the context of liver gene therapy.
[0015] Prior studies have implicated innate immune sensors such as Toll-like receptors (TLR) 2 and 9 and their downstream adaptor molecule myeloid differentiation primary response protein 88 (MyD88) in sensing viral capsid and genome and mediating adaptive immune response against AAV (Martino, A.T. et al. The genome of self-complementary adeno-associated viral vectors increases Toll-like receptor 9-dependent innate immune responses in the liver. Blood 117, 6459-6468 (2011); Hosel, M. et al. Toll-like receptor 2-mediated innate immune response in human nonparenchymal liver cells toward adeno-associated viral vectors. Hepatology 55, 287-297 (2012); Sudres, M. et al. MyD88 signaling in B cells regulates the production of Thl -dependent antibodies to AAV. Molecular Therapy 20, 1571-1581 (2012); Rogers, G.L. et al. Unique Roles of TLR9- and MyD88-Dependent and -Independent Pathways in Adaptive Immune Responses to AAV-Mediated Gene Transfer. J. Innate Immunity 7, 302-314 (2015)).
[0016] While establishment of a link between TLR9-MyD88 signaling and CD8+ T cell responses to capsid and transgene products has prompted the development of CpG-depleted expression cassettes, little is known about the innate immune sensors and involved mechanisms that play a critical role in eliciting an adaptive immune response to AAV encoded transgene in the setting of liver gene transfer.
[0017] What is needed are methods to prevent or alleviate the cytotoxic immune responses that prevent desired AAV vector gene therapy outcomes. The present disclosure addresses this need.
[0018] BRIEF DESCRIPTION OF THE DRAWINGS
[0019] FIG. la-e depicts the results of experiments showing that IL-1R1 - MyD88 signaling is essential for CD8+ T cell response to AAV encoded transgene in liver, which can be prevented by neutralization of IL-la and IL-Ib. FIG. la: Experimental timeline showing the AAV8-OVA vector administration via tail vein to either C57BL/6-WT mice or knockout of either TLR2 or TLR9 or MyD88 or IL-1R1. To block IL-1 signaling C57BL/6-WT mice were treated with 200ug of either anti-IL-la or anti-IL-Ib or both antibodies via intraperitoneal route. FIG. lb: Similar to wild-type control mice, TLR2 and 9 deficient mice induced CD8+ T cell response whereas mice deficient for MyD88 and IL-1R1 failed to generate CD8+ T cell response against transgene. FIG lc: Both cytokines, IL-la and IL-Ib are known to activate IL-1R1. Blocking of either of these cytokines with anti- IL-la and anti-IL-Ib antibodies not only reduced the number of animals making these cellular responses but also reduced the frequency of these CD8+ T cells in these animals. Combination treatment with both anti-IL-la and IL-Ib antibodies further reduced the number of animals that induced transgene specific CD8+ T cell response. FIG. Id: Ovalbumin levels in C57BL/6-WT and different knockout mice 4 weeks after AAV8-OVA administration. Significantly higher levels of ovalbumin were detected in mice knockout for MyD88, IL-1R1 and TLR2 as compare to C57BL/6-WT mice. FIG. le: Ovalbumin levels in C57BL/6-WT untreated and C57BL/6-WT mice treated with either anti-IL-la or IL-Ib or both IL-la and IL-Ib antibodies 4 weeks after AAV8-OVA administration. [0020] FIG. 2a-e shows the results that redundant and non-redundant functions of TLR9 and IL1R signaling in CD8+ T cell activation are vector dose-dependent in AAV muscle gene transfer. At a lower vector dose, both TLR9-MyD88 and IL-lRl-MyD88 signaling pathways are required for a CD8+ T cell response against the transgene product. Thus, elimination of either prevents the response. At a higher vector dose, these pathways are redundant, so that both need to be blocked to prevent the response. FIG. 2a) Experimental timeline showing the two doses of AAV 1 -OVA vector administration via muscle to either C57BL/6-WT mice or TLR9 or MyD88_/ or IL- 1 R 1 knock-out mice. To block IL-1 signaling, TLR9 knock-out mice were treated with 200ug of either anti -IL- la or IL-Ib or both antibodies via intraperitoneal route. FIG. 2b) TLR9 and IL-1R1 deficient mice showed CD8+ T cell responses similar to wild-type control mice, whereas mice deficient for MyD88 failed to generate CD8+ T cell responses against transgene at the high dose (2xlOnvg). FIG. 2c) Mice deficient for MyD88 or TLR9 or IL-1R1 had much reduced CD8+ T cell responses against transgene at the low dose (2xl010vg). FIG. 2d) Blocking of either of cytokine IL-la and IL-Ib with anti-IL-la and IL-Ib antibodies or combination treatment with both anti- IL-la and IL-Ib antibodies reduced the frequency of these CD8+T cells in TLR9 knock-out mice administrated with the high-dose of AAV1. FIG. 2e) Summary of ova-specific CD8+ T cell frequencies in either C57BL/6-WT mice or either IL-1R1 or TLR9 knock-out mice untreated and TLR9 knock-out mice treated with either anti-IL-la or IL-Ib or both IL-la and IL-Ib antibodies or isotype control IgG 2 weeks after high dose of AAVl-OVA administration.
[0021] SUMMARY OF THE DISCLOSURE
[0022] The present disclosure provides methods for inhibiting an immune response in a subj ect undergoing AAV gene therapy comprising co-administering to the subject with an AAV gene therapy vector an effective amount of an interleukin-1 (IL-1) signaling pathway disrupter, the IL- 1 signaling pathway disrupter being effective to suppress a cytotoxic CD8+ T cell response against the AAV vector or a transgene product expressed from the AAV vector. The AAV vector can in some embodiments is a liver-targeting AAV or a liver gene transfer vector.
[0023] In other embodiments, the IL-1 signaling pathway disrupter comprises an agonist of the IL-1 signaling pathway that can effectively deactivate IL-la or IL-Ib or both IL-la or IL-Ib. In some further embodiments, the methods include an IL-1 signaling pathway disrupter that comprises an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor. [0024] Alternatively, the IL-1 signaling pathway disrupter comprises an agent effective to deactivate MyD88.
[0025] In any of the methods of the disclosed embodiments, co-administration of the signaling pathway disrupter to the subject will occur at a suitable time before, during or following gene therapy treatment either with a single administration or periodically as needed.
[0026] Yet another embodiment of the current disclosure is a method of treating a mammal comprising: (a) administering a recombinant AAV vector to a mammal; and (b) co-administering an IL-1 signaling pathway disrupter to the mammal in an amount effective to have a therapeutic effect on said mammal and wherein said therapeutic effect is a suppression of an immune response to the gene therapy. In alternate embodiments, the mammal is a human patient in need of gene therapy treatment.
[0027] In various embodiments, the IL-1 signaling pathway disrupter is one or more of an antibody against IL-la, an antibody against IL-Ib or an antibody against an IL-1 receptor.
[0028] In still other embodiments, the IL-1 signaling pathway disrupter is a drug such an IL-1 receptor antagonist.
[0029] In yet other embodiments, the methods include co-administration of the IL-1 signaling pathway disrupter into various tissues of the mammal that include but are not limited to, muscle, the brain, intraperitoneally, intramuscularly, directly into the bone marrow or the like. Administration can also be oral.
[0030] In some embodiments, the IL-1 signaling pathway disrupter is injected at a single site per dose or as multiple doses at multiple sites.
[0031] In some embodiments, the materials to accomplish any of the methods can be packed into a kit that includes all the necessary components to carry out said methods.
[0032] These and other embodiments and features of the disclosure will become more apparent through reference to the following description, the accompanying figures, and the claims. Furthermore, it is to be understood that the features of the various embodiments described herein are not mutually exclusive and can exist in various combinations and permutations. [0033] DETAILED DESCRIPTION
[0034] Throughout this disclosure, various quantities, such as amounts, sizes, dimensions, proportions and the like, are presented in a range format. It should be understood that the description of a quantity in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of any embodiment. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as all individual numerical values within that range unless the context clearly dictates otherwise. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual values within that range, for example, 1.1, 2, 2.3, 4.62, 5, and 5.9. This applies regardless of the breadth of the range. The upper and lower limits of these intervening ranges may independently be included in the smaller ranges, and are also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the disclosure, unless the context clearly dictates otherwise.
[0035] The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of any embodiment. As used herein, the singular forms “a,” “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. It will be further understood that the terms “includes”, “comprises”, “including” and/or “comprising,” when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof. As used herein, the term “and/or” includes any and all combinations of one or more of the associated listed items. Additionally, it should be appreciated that items included in a list in the form of “at least one of A, B, and C” can mean (A); (B); (C); (A and B); (B and C); (A and C); or (A, B, and C). Similarly, items listed in the form of “at least one of A, B, or C” can mean (A); (B); (C); (A and B); (B and C); (A and C); or (A, B, and C).
[0036] Unless specifically stated or obvious from context, as used herein, the term “about” in reference to a number or range of numbers is understood to mean the stated number and numbers +/- 10% thereof, or 10% below the lower listed limit and 10% above the higher listed limit for the values listed for a range. [0037] The present disclosure is based on the discovery that co-administration of an interleukin- 1 (IL-1) signaling pathway disrupter to a mammal with a gene delivery vehicle results in suppression of a cytotoxic CD8+ T cell response against the AAV vector or a transgene product expressed from the AAV vector. The terms “suppress” or “suppression” or other formatives thereof, and also the terms “inhibit” or “inhibition” or other formatives thereof, when used herein in relation to an immune response, are intended to refer to a reduction in or prevention of the immune response.
[0038] While methods for inhibiting an immune response in a subject undergoing adeno associated virus (AAV) gene therapy disclosed herein are particularly useful for gene therapy vectors designed for the treatment of hemophilia, the disclosure is not limited solely to the treatment of hemophilia. Rather, the disclosure should be construed to include co-administration of an IL-1 signaling pathway disrupter with a variety of DNA encoding gene products that are useful for the treatment of other disease states in a mammal. In some embodiments, the gene therapy vector is a liver-targeting AAV, such as, for example, a liver gene transfer vector. In some embodiments, the AAV gene therapy comprises AAV mediated liver gene therapy. Alternative DNA incorporated into the AAV gene therapy vector and associated disease states include, but are not limited to: DNA encoding glucose-6-phosphatase, associated with glycogen storage deficiency type 1A; DNA encoding phosphoenolpyruvate-carboxykinase, associated with Pepck deficiency; DNA encoding galactose- 1 phosphate uridyl transferase, associated with galactosemia; DNA encoding phenylalanine hydroxylase, associated with phenylketonuria; DNA encoding branched chain . alpha. -ketoacid dehydrogenase, associated with Maple syrup urine disease; DNA encoding fumarylacetoacetate hydrolase, associated with tyrosinemia type 1; DNA encoding methylmalonyl-CoA mutase, associated with methylmalonic acidemia; DNA encoding medium chain acyl CoA dehydrogenase, associated with medium chain acetyl CoA deficiency; DNA encoding ornithine transcarbamylase, associated with ornithine transcarbamylase deficiency; DNA encoding argininosuccinic acid synthetase, associated with citrullinemia; DNA encoding low density lipoprotein receptor protein, associated with familial hypercholesterolemia; DNA encoding UDP-glucouronosyltransferase, associated with Crigler-Najjar disease; DNA encoding adenosine deaminase, associated with severe combined immunodeficiency disease; DNA encoding hypoxanthine guanine phosphoribosyl transferase, associated with Gout and Lesch-Nyan syndrome; DNA encoding biotinidase, associated with biotinidase deficiency; DNA encoding b- glucocerebrosidase, associated with Gaucher disease; DNA encoding 3-glucuronidase, associated with Sly syndrome; DNA encoding peroxisome membrane protein 70 kDa, associated with Zellweger syndrome; DNA encoding porphobilinogen deaminase, associated with acute intermittent porphyria; DNA encoding a-1 antitrypsin for treatment of a-1 antitrypsin deficiency (emphysema); DNA encoding erythropoietin for treatment of anemia due to thalassemia or to renal failure; and, DNA encoding insulin for treatment of diabetes.
[0039] Specifically, the types of diseases, tissues and AAV targets that are contemplated by the current disclosure include those shown in Table 1. It should be noted that Table 1 is a representation of non-limiting examples.
[0040] TABLE T
Figure imgf000010_0001
Figure imgf000011_0001
A1 AT, od-antitrypsin; AADC, aromatic 1-amino acid decarboxylase; AMD, age-related macular degeneration; ARSB, arylsulfatase B; CLN2, neuronal ceroid lipofuscinosis type 2; CMT1A, Charcot-Marie-Tootli disease type 1A; CNGB3, cyclic nucleotide-gated channel-P3; DMD, Duclienne muscular dystrophy; DYSF, dysferlin; FH, familial hypercliolesterolaemia; FVIII, factor VIII; G6PC, glucose-6-pliosphatase catalytic subunit; GAA, ot- glucosidase; GAN, gigaxonin; GDNF, glial cell line-derived neurotrophic factor; GSDla, glycogen storage disease type la; LCA, Leber congenital amaurosis; LDLR, low-density lipoprotein receptor; LHON, Leber hereditary optic neuropathy; MPS, mucopolysaccharidosis; MTM, myotubular myopathy; NAGLU, V-a-acetylglucosaminidase; NTF3, neurotrophin 3; OTC, ornithine transcarbamylase; REP1, RAB escort protein 1; RLBPl, retinaldehydebinding protein 1; RP, retinitis pigmentosa; RPE65, retinal pigment epithelium-specific 65 kDa protein; RPGR, retinitis pigmentosa GTPase regulator; RSI, retinoschisin 1; SGSFI, Y-sulfoglucosaminc sulfohydrolase; SMA, spinal muscular atrophy; SMN, survival of motor neuron; UGT1A1, UDP glucuronosyltransferase family 1 member Al; VEGF, vascular endothelial growth factor; ZFN, zinc -finger-containing protein. (Adapted in part from: Wang et al., “Adeno-associated virus vector as a platform for gene therapy delivery,” Nature Reviews 18: 2019, pp. 358-378).
[0041] The methods of the disclosure can include without limitation, gene replacement therapies in which the ultimate goal is to deliver a gene product to compensate for loss-of-function mutations. Gene replacement is suitable for treating recessive monogenic diseases. A non-limiting example of gene replacement target is the treatment of hemophilia A or B.
[0042] In other embodiments, the methods of the disclosure can include without limitation, gene silencing where the therapeutic goal is to silence genes that produce toxic mutations. One nonlimiting example is Huntington disease.
[0043] In still other embodiments, the methods of the disclosure can include without limitation gene addition therapies. Such therapies can target complex genetic diseases and acquired diseases including but not limited to heart failure and infectious diseases. Gene addition can modulate these diseases in multiple ways, such as supplying neurotrophic factors for neurological diseases and tuning signaling pathways for heart failure, neurotrophic factors for neurological diseases, and tuning signaling pathways for heart failure and cancer.
[0044] Additional examples of gene addition strategies that the methods of the disclosure can target employ recombinant AAV delivery of genes encoding recombinant antibodies that can neutralize deadly viral infections. Such therapies would utilize intramuscular delivery and transform the transduced muscle cells into a biofactory to produce therapeutic antibodies that are secreted into the bloodstream. Such a strategy could target such infections and HIV, Hepatitis B or Hepatitis C.
[0045] This disclosure is further based on the discovery that an antibody against IL-1 alpha is particularly effective to suppress an immune response against an AAV vector or a transgene product expressed from the AAV vector. It is contemplated that the delivery of an IL-1 signaling pathway disrupter to a mammal provides a therapeutic benefit to a mammal receiving gene therapy. [0046] The disclosure also includes a method of treating a mammal, preferably, a human, undergoing gene therapy. The present disclosure provides a method for inhibiting an immune response in a subject undergoing AAV gene therapy comprising co-administering to the subject with an AAV gene therapy vector an effective amount of an interleukin- 1 (IL-1) signaling pathway disrupter, the IL-1 signaling pathway disrupter being effective to suppress a cytotoxic CD8+ T cell response against the AAV vector or a transgene product expressed from the AAV vector. The AAV vector can in some embodiments is a liver-targeting AAV or a liver gene transfer vector. [0047] The IL-1 signaling pathway disrupter of certain embodiments of the present disclosure may be formulated with a pharmaceutical vehicle or diluent for oral, intravenous, subcutaneous, intranasal, intrabronchial or rectal administration. The pharmaceutical composition can be formulated in a classical manner using solid or liquid vehicles, diluents and additives appropriate to the desired mode of administration. Orally, the composition can be administered in the form of tablets, capsules, granules, powders and the like with at least one vehicle, e.g., starch, calcium carbonate, sucrose, lactose, gelatin, etc. The preparation may also be emulsified. The active ingredient is often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, e.g., water, saline, dextrose, glycerol, ethanol or the like and combination thereof. In addition, if desired, the composition may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, or adjuvants which enhance the effectiveness of the composition. A preparation for parental administration includes sterilized water, suspension, emulsion, and suppositories. For the emulsifying agents, propylene glycol, polyethylene glycol, olive oil, ethyloleate, etc. may be used. For suppositories, traditional binders and carriers may include polyalkene glycol, triglyceride, witepsol, macrogol, tween 61, cocoa butter, glycerogelatin, etc. In addition, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like can be used as excipients.
[0048] In other embodiments, the IL-1 signaling pathway disrupter comprises an antagonist of the IL-1 signaling pathway that can effectively deactivate IL-la or IL-Ib or both IL-la or IL-Ib. In some further embodiments, the methods include an IL-1 signaling pathway disrupter that comprises an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor.
[0049] Alternatively, the IL-1 signaling pathway disrupter comprises an agent effective to deactivate MyD88.
[0050] In any of the methods of the disclosed embodiments, co-administration of the signaling pathway disrupter to the subject occurs at a suitable time before, during or following gene therapy treatment either with a single administration or periodically as needed.
[0051] Yet another embodiment of the current disclosure is a method of treating a mammal comprising: (a) administering a recombinant AAV vector to a mammal; and (b) co-administering an IL-1 signaling pathway disrupter to the mammal in an amount effective to have a therapeutic effect on said mammal and wherein said therapeutic effect is a suppression of an immune response to the gene therapy. In alternate embodiments, the mammal is a human patient in need of gene therapy treatment.
[0052] In various embodiments, the IL-1 signaling pathway disrupter is one or more of an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor.
[0053] In still other embodiments, the IL-1 signaling pathway disrupter is a drug such an IL-1 receptor antagonist.
[0054] In yet other embodiments, the methods include co-administration of the IL-1 signaling pathway disrupter into various tissues of the mammal that include but are not limited to, muscle, the brain, intraperitoneally, intramuscularly, directly into the bone marrow or the like. Administration can also be oral.
[0055] In some embodiments, the IL-1 signaling pathway disrupter is injected at a single site per dose or as multiple doses at multiple sites.
[0056] In some embodiments, the materials to accomplish any of the methods can be packed into a kit that includes all the necessary components to carry out said methods.
[0057] Further reference is made to the following experimental examples.
[0058] EXAMPLES
[0059] The following examples are given for the purpose of illustrating various embodiments of the invention and are not meant to limit the present disclosure in any fashion. The present examples, along with the methods described herein are presently representative of preferred embodiments, are provided only as examples, and are not intended as limitations on the scope of the invention. Changes therein and other uses which are encompassed within the spirit of the disclosure as defined by the scope of the claims will occur to those skilled in the art. [0060] EXAMPLE 1
[0061] Induction of CD8+ T cell response to AAV encoded transgene in the liver is determined by the vector dose
[0062] Initial Studies
[0063] Although liver gene transfer is capable of inducing immunological tolerance to AAV encoded transgene products, levels of gene expression as determined by vector dose and design play a critical role in eliciting an adaptive CD8+ T cell response to an AAV encoded transgene (FIG. 1), resulting in loss of the model antigen ovalbumin (OVA) or of FIX expression in hemophilia B mice (Hoffman, B.E. et al. Nonredundant roles of IL-10 and TGF-beta in suppression of immune responses to hepatic AAV-factor IX gene transfer. Molecular Therapy: 19, 1263-1272 (2011); Kumar, et al., The balance between CD8+ T Cell-Mediated Clearance of AAV -Encoded Antigen in the Liver and Tolerance Is Dependent on the Vector Dose. Molecular Therapy: 25, 880- 891 (2017); Markusic, D.M. et al. High-efficiency transduction and correction of murine hemophilia B using AAV2 vectors devoid of multiple surface-exposed tyrosines. Molecular Therapy: 18, 2048-2056 (2010).
[0064] For instance, low levels of hepatic gene expression may elicit an adaptive CD8+ T cell response to an AAV encoded transgene, resulting in loss of the model antigen ovalbumin (OVA) in C57BL/6 mice or of FIX expression in hemophilia B mice.
[0065] In a murine model of hepatic gene transfer, the inventors have shown that transgene expression and generation of CD8+ T cell response to an AAV encoded antigen are vector dose dependent. Using AAV serotype 8 (AAV8) to deliver OVA in a murine model of liver gene transfer we showed that vector dose plays a critical role in establishing an immune response / tolerance to transgene. Wild-type (WT) C57BL/6 male mice were injected with three doses (low: 1 x 108 vg, medium: 1 c 109 vg, and high: 1 c 1010 vg) of AAV8 expressing full-length OVA (AAV 8-OVA) via the tail vein. Peripheral blood mononuclear cells (PBMCs) from these animals were tested for OVA-specific CD8+T cells over a period of 16 weeks. At the low and high doses, no immune response to OVA was observed. However, at the mid dose, tetramer1 CD8+ T cells were detected. Thirty-fifty percent of the mice in this dose group had circulating OVA-specific CD8+ T cells with an average highest frequency of ~15% at 4 weeks post injection (PI). Although a slight decline in frequency was observed at 6 and 8 weeks PI, nearly constant levels of these CD8+ T cells persisted throughout the course of the study. Using additional animals injected with this vector dose, we confirmed by qPCR that liver was the only detectable source of OVA expression.
[0066] In this murine model a systemic cellular immune response is elicited against transgene at a particular vector dose. A vector dose lower or higher than this optimum dose would lead to immunological ignorance or tolerance to transgene respectively (Kumar, et al.), thereby enabling us to identify and study the immunological pathways that regulates transgene specific immune responses. Moreover, this model led the inventors to uncover a novel immune signaling pathway that leads to CD8+ T cell activation in hepatic AAV gene transfer. An important implication of this work is that some CD8+ T cell responses to AAV gene transfer will not be affected by TLR9 inhibition or elimination of CpG motifs from the vector genome (to reduce TLR9 signaling). [0067] Studies using this murine model of liver gene transfer demonstrated how different subsets of dendritic cells (DCs), in particular conventional DCs (cDCs) and plasmacytoid DCs (pDCs) work in concert to sense AAV capsid and elicit an adaptive immune response against AAV capsid. Specifically, the inventors showed that TLR9 in pDCs senses AAV genome leading to secretion of type I interferon, which in turn activates cDCs in trans to cross present AAV capsid antigen during CD8+ T-cell activation.
[0068] The data also demonstrate that that inhibiting either TLR9 sensing or type I interferon signaling can block unwanted immune responses against AAV capsid and therefore can be used as therapeutic interventions during gene therapy.
[0069] This same murine model was used in further studies described below, which demonstrate that unlike AAV, innate sensing of AAV encoded transgene in liver is not regulated by TLR2 or TLR9. Furthermore, the results show a critical role of IL-l/MyD88 signaling pathway in eliciting transgene specific adaptive immune response. Surprisingly, IL-1 mediated signaling has not previously been implicated in adaptive immune response to AAV encoded transgene. The murine model is also used to establish a similar model that establishes a mechanism by which innate immunity against AAV causes IL-l/MyD88 signaling that subsequently drives CD8+ T cell activation against the transgene product in the liver. Interestingly, the delayed CD8+ T cell- mediated loss of transgene expression that was observed upon hepatic gene transfer in mice follows a similar time course to the response against AAV capsid in humans and may therefore also have implications on T cell responses to capsid. [0070] EXAMPLE 2
[0071] The TLR9-MyD88 pathway is essential for capsid-specific CD8+ T-cell responses. [0072] Initial Studies:
[0073] A modified AAV serotype 2 capsid containing immunodominant CD8+ T-cell epitope (SIINFEKL) of the model antigen OVA was used to study capsid specific CD8+ T cells response in C57BL/6 mice. Mice were injected with lxlO11 vg/mouse of AAV2-SIINFEKL vector via intramuscular route. Using the H2-Kb -SIINFEKL tetramer, it was demonstrated a vector dose dependent capsid-specific CD8+ T cell.
[0074] The CD8+ T cell response peaked between 7 to 10 days post-injection, and largely subsided by day 21. Induced cytotoxic T lymphocytes (CTLs) were functional in an in vivo killing assay. In contrast to wild type mice, TLR9_/ and MyD88_/ mice failed to respond to the AAV capsid. No reduction in the CD8+ T-cell response was observed in TLR2 mice, in line with the previous observations that this receptor has limited impact on the adaptive response to AAV.
[0075] EXAMPLE 3
[0076] IL-1 Receptor (IL-1R) / MyD88-dependent CD8+ T Cell Responses to Hepatic AAV Gene Transfer.
[0077] To understand if AAV sensing (capsid or genome) by innate sensors could lead to an adaptive immune response to AAV encoded transgene, the role of innate sensors (such as TLRs 2, 9 and MyD88) was investigated in eliciting a CD8+ T cell response to AAV encoded transgene in liver. In order to understand the underlying mechanisms of transgene specific CD8+ T cell activation in the hepatic microenvironment, it was examined whether innate sensing of AAV has an impact on the adaptive immune response to AAV encoded transgene. Several prior studies demonstrated a dependence of CD8+ T cell responses against capsid and transgene product on TLR9-MyD88 signaling (reflecting endosomal sensing of the AAV genome) (Rogers, G.L. et al. Unique Roles of TLR9- and MyD88-Dependent and -Independent Pathways in Adaptive Immune Responses to AAV -Mediated Gene Transfer. J. Innate Immun. 7, 302-314 (2015); Zhu et al., The TLR9-MyD88 pathway is critical for adaptive immune responses to adeno-associated virus gene therapy vectors in mice. J. Clin Invest. 119, 2388-2398 (2009); Faust, S.M. et al. CpG-depleted adeno-associated virus vectors evade immune detection. J. Clin Invest 123, 2994-3001 (2013); Martino, A.T. et al. Engineered AAV vector minimizes in vivo targeting of transduced hepatocytes by capsid-specific CD8+ T cells. Blood 121, 2224-2233 (2013); Rogers, G.L. et al. Plasmacytoid and conventional dendritic cells cooperate in cross priming AAV capsid-specific CD8+ T cells. Blood 129, 3184-3195 (2017)). Surprisingly, it was discovered that in a murine model of hepatic gene transfer with hepatotropic AAV8 vector encoding OVA demonstrate that innate sensors TLR2 and TLR9 are dispensable for the CD8+ T cell response to transgene; however, adaptor protein MyD88 is required. Furthermore, the data reveals an important role for interleukin-1 (IL- 1) signaling pathway in eliciting transgene specific T cell response during AAV mediated liver gene transfer.
[0078] Materials and Methods.
[0079] Wild-type C57BL/6 mice and various knockout mice of specific innate sensing molecules (such as TLRs 2, 9 and MyD88) on C57BL/6 background (n=8 per experimental group) were intravenously injected with lxl09vg particles of hepatotropic AAV8 virus expressing OVA, which we previously found to activate CD8+ T cells (Mol Ther 25:880, 2017). Four weeks after vector administration, PBMCs were analyzed by flow cytometry for OVA-specific CD8+ T cells using a class I MHC tetramer. In 50-75% of wild type mice, OVA-specific CD8+ T cells were observed at frequencies of 2-32%.
[0080] Unlike previous studies with AAV-capsid specific CD8+ T cell responses, innate sensor TLR9 was dispensable for transgene specific CD8+ T cell response, as 60% of TLR9 mice developed a response, i.e., were positive for OVA specific CD8+ T cells. Similarly, some of the TLR2 mice (37%) also had OVA specific CD8+ T cells, indicating that TLR2 is dispensable for these cellular immune responses. No significant differences were seen in TRIF . IFNaR V or DA5_/ mice. Recently, double stranded RNA sensors such as RIG-I and MDA-5 have been shown to play critical role in immune responses to AAV mediated gene therapy (Shao, W. et al. Double-stranded RNA innate immune response activation from long-term adeno-associated virus vector transduction. JCI Insight 3 (2018)); however, this study did not imply any role for RNA sensing, as DA-5_/ mice were observed to have transgene specific CD8+ T cell response similar to WT mice. Interestingly, adaptor protein MyD88_/ mice did not elicit CD8+ T cell response to OVA, implying an important role for MyD88 in mediating transgene specific cellular immune response. [0081] Since MyD88 is an essential adaptor protein not only for TLRs but also for interleukin- 1 (IL-1) signaling pathways, I L- 1 R 1 mice were analyzed. Similar to MyD88 /_ mice, IL- 1 R 1 mice did not show OVA specific CD8+ T cells (p=0.0063, 0.0075 respectively), indicating that transgene specific adaptive responses are mediated via IL-lRl/MyD88 signaling. CD4-deficient mice also failed to elicit an immune response to OVA, likely reflecting a requirement for CD4+ T help. While MyD88 and CD4 were also required for antibody responses against the viral capsid and the transgene product, IL1-R was not. Surprisingly, in comparison to wild-type mice, TLR9 mice had a significantly (p=<0.0001) diminished antibody response to AAV capsid (but not the transgene product). TLR2- and ILlR-deficient mice had a modest reduction in antibody formation against ovalbumin, and these strains as well as MyD88 /_ mice showed a significant increase in circulating ovalbumin levels (reaching 100-200 ng/ml).
[0082] FIG. la-e depicts the results of experiments showing that IL-1R1 - MyD88 signaling is essential for CD8+ T cell response to AAV encoded transgene in liver, which can be prevented by depletion/neutralization of IL-la and IL-Ib. FIG. la: Experimental timeline showing the AAV8- OVA vector administration via tail vein to either C57BL/6-WT mice or knockout of either TLR2 or TLR9 or MyD88 or IL-1R1. To block IL-1 signaling C57BL/6-WT mice were treated with 200ug of either anti-IL-la or anti-IL-Ib or both antibodies via intraperitoneal route. FIG. lb: Similar to wild-type control mice, TLR2 and 9 deficient mice induced CD8+ T cell response whereas mice deficient for MyD88 and IL-1R1 failed to generate CD8+ T cell response against transgene. FIG lc: Both cytokines, IL-la and IL-Ib are known to activate IL-1R1. Blocking of either of these cytokines with anti-IL-la and anti -IL-1 b antibodies not only reduced the number of animals making these cellular responses but also reduced the frequency of these CD8+T cells in these animals. Combination treatment with both anti-IL-la and IL-Ib antibodies further reduced the number of animals that induced transgene specific CD8+ T cell response. FIG. Id: Ovalbumin levels in C57BL/6-WT and different knockout mice 4 weeks after AAV8-OVA administration. Significantly higher levels of ovalbumin were detected in mice knockout for MyD88, IL-1R1 and TLR2 as compare to C57BL/6-WT mice. FIG. le: Ovalbumin levels in C57BL/6-WT untreated and C57BL/6-WT mice treated with either anti-IL-la or IL-Ib or both IL-la and IL-Ib antibodies 4 weeks after AAV8-OVA administration. [0083] EXAMPLE 4
[0084] Redundant and non-redundant functions of TLR9 and IL1R signaling in CD8+ T cell activation are vector dose-dependent in AAV muscle gene transfer
[0085] FIG. 2a-e shows the results that redundant and non-redundant functions of TLR9 and IL1R signaling in CD8+ T cell activation are vector dose-dependent in AAV muscle gene transfer. At a lower vector dose, both TLR9-MyD88 and IL-lRl-MyD88 signaling pathways are required for a CD8+ T cell response against the transgene product. Thus, elimination of either prevents the response. At a higher vector dose, these pathways are redundant, so that both need to be blocked to prevent the response. FIG. 2a) Experimental timeline showing the two doses of AAV 1 -OVA vector administration via muscle to either C57BL/6-WT mice or TLR9 or MyD88_/ or I L- 1 R 1 /_ knock-out mice. To block IL-1 signaling, TLR9 knockout mice were treated with 200ug of either anti-IL-la or anti-IL-Ib or both antibodies via intraperitoneal route. FIG. 2b) TLR9 and IL-1R1 deficient mice showed CD8+ T cell responses similar to wild-type control mice, whereas mice deficient for MyD88 failed to generate CD8+ T cell responses against transgene at the high dose (2xlOnvg). FIG. 2c) Mice deficient for MyD88 or TLR9 or IL-1R1 had much reduced CD8+ T cell responses against transgene at the low dose (2xl010vg). FIG. 2d) Blocking of either of cytokine IL-la and IL-Ib with anti-IL-la and anti-IL-Ib antibodies or combination treatment with both anti-IL-la and anti-IL-Ib antibodies reduced the frequency of these CD8+T cells in TLR9 knock out mice administrated with the high-dose of AAV1. FIG. 2e) Summary of ova-specific CD8+ T cell frequencies in either C57BL/6-WT mice or either IL-1R1 or TLR9 knockout mice untreated and TLR9 knockout mice treated with either anti-IL-la or anti-IL-Ib or both antibodies or isotype control IgG 2 weeks after high dose of AAV 1 -OVA administration.
[0086] EXAMPLE 5
[0087] Identify specific cytokines in interleukin- 1 (IL-1) signaling pathway responsible for signaling to AAV liver gene transfer
[0088] Beside TLRs, IL-1 signaling plays a significant role in innate sensing and eliciting adaptive immune responses to various bacteria and viruses (Zwijnenburg, et ak, IL-1 receptor type 1 gene-deficient mice demonstrate an impaired host defense against pneumococcal meningitis. J. Immun. 170, 4724-4730 (2003); Schmitz et ak, Interleukin-1 is responsible for acute lung immunopathology but increases survival of respiratory influenza virus infection. J. Virol. 79, 6441- 6448 (2005); Pang et al., IL-1R signaling in dendritic cells replaces pattern-recognition receptors in promoting CD8+ T cell responses to influenza A virus. Nature Immun. 14, 246-253 (2013)). The founding members of IL-1 family, cytokines IL-la and IL-Ib are both ligands for type I receptor IL-1R1. These cytokines share only 24% amino acid homology, and yet have largely identical biological functions. Though both IL-la and IL-Ib primarily have pro-inflammatory functions, they have been implicated in mediating adaptive immune responses (Voronov, E. et al. Unique Versus Redundant Functions of IL-1 alpha and IL-1 beta in the Tumor Microenvironment. Frontiers Immunol. 4, 177 (2013); Voronov et al., Targeting the Tumor Microenvironment by Intervention in Interleukin-1 Biology. Current Pharmac. Design 23, 4893-4905 (2017); Dinarello, C.A. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol. Rev. 281, 8-27 (2018)). In the context of AAV gene therapy, recently published data with PBMCs (peripheral blood mononuclear cells) from healthy individuals described a critical role for IL-Ib in regulating immune response against AAV capsid (Kuranda, K. et al. Exposure to wild-type AAV drives distinct capsid immunity profiles in humans. J. Clin. Invest. 128, 5267-5279 (2018)). Having found a critical role of IL-1 signaling in adaptive immune response to AAV encoded transgene, it was investigated what specific cytokine and response mechanism is implicated in the immune response.
[0089] The murine model described above was used in which mice were intravenously (IV) injected with AAV8 vector encoding OVA and also intraperitoneally (IP) injected selected mice with blocking antibodies to IL-la and IL-Ib. For this set of experiments, injected WT C57BL/6 mice were injected IP with either anti-IL-la or anti-IL-Ib. After one day the mice were IV injected with AAV8 vector encoding OVA. The mice in each group were further treated with either anti- IL-la or anti-IL-Ib twice per week for four weeks. Following four weeks, cellular immune responses to transgene and humoral immune responses to transgene and AAV capsid were evaluated. OVA specific CD8+ T cells were quantified using a tetramer for H2-Kb restricted SIINFEKL peptide.
[0090] As will be appreciated from the descriptions herein, a wide variety of aspects and embodiments are contemplated by the present disclosure, examples of which include, without limitation, the aspects and embodiments listed below: [0091] A method for inhibiting an immune response in a subj ect undergoing AAV gene therapy that includes co-administering to the subject with an AAV gene therapy vector an effective amount of an interleukin-1 (IL-1) signaling pathway disrupter, the IL-1 signaling pathway disrupter being effective to suppress a cytotoxic CD8+ T cell response against the AAV vector or a transgene product expressed from the AAV vector.
[0092] A method in accordance with any other embodiment disclosed herein, wherein the AAV vector is a liver-targeting AAV and/or AAV mediated liver gene therapy and or a liver gene transfer vector.
[0093] A method in accordance with any other embodiment disclosed herein, wherein the IL- 1 signaling pathway disrupter comprises one or more of an antagonist of the IL-1 signaling pathway, an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor, or an agent effective to deactivate MyD88.
[0094] A method in accordance with any other embodiment disclosed herein, wherein the IL- 1 signaling pathway disrupter comprises an agent effective to deactivate IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor.
[0095] A method in accordance with any other embodiment wherein administering the signaling pathway disrupter to the subject can be accomplished within an appropriate time of a gene therapy treatment.
[0096] A method in accordance with any other embodiment wherein the signaling path disruptor is administered to the patient, before, during or following a gene therapy treatment. [0097] A method of treating a mammal that includes: (a) administering a recombinant adeno associated virus vector to a mammal; and (b) co-administering an IL-1 signaling pathway disrupter to the mammal in an amount effective to have a therapeutic effect on said mammal and wherein said therapeutic effect is a suppression of an immune response to the gene therapy.
[0098] A method in accordance with any other embodiment wherein the mammal is a patient in need of one or more gene therapy treatments.
[0099] A method in accordance with any other embodiment wherein the IL-1 signaling pathway disrupter is one or more of an antibody against IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor, or an agent effective to deactivate MyD88 or an IL-1 receptor antagonist [00100] A method in accordance with any other embodiment wherein the methods include co administration of the IL-1 signaling pathway disrupter into various tissues of the mammal that include but are not limited to, muscle, the brain, intraperitoneally, intramuscularly, directly into the bone marrow or the like. Administration can also be oral.
[00101] A method in accordance with any other embodiment wherein the IL-1 signaling pathway disrupter is injected at a single site per dose or at multiple doses at multiple sites.
[00102] A method in accordance with any other embodiment wherein the materials to accomplish any of the methods can be packed into a kit that includes all the necessary components to carry out said methods.
[00103] While embodiments of the present disclosure have been described herein, it is to be understood by those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims

1. A method for inhibiting an immune response in a subject undergoing adeno associated virus (AAV) gene therapy comprising co-administering to the subject with an AAV gene therapy vector an effective amount of an interleukin-1 (IL-1) signaling pathway disrupter, the IL-1 signaling pathway disrupter being effective to suppress a cytotoxic CD8+ T cell response against the AAV vector or a transgene product expressed from the AAV vector.
2. The method of claim 1, wherein the AAV vector is a liver-targeting AAV.
3. The method of claim 1, wherein the AAV gene therapy comprises AAV mediated liver gene therapy.
4. The method of claim 1, wherein the AAV vector is a liver gene transfer vector.
5. The method of claim 1, wherein the IL-1 signaling pathway disrupter comprises an antagonist of the IL-1 signaling pathway.
6. The method of claim 1, wherein the IL-1 signaling pathway disrupter comprises one or more of an agent effective to deactivate IL-la, IL-Ib, both IL-la or IL-Ib and/or the IL-1 receptor, or an agent effective to deactivate MyD88.
7. The method of claim 1, wherein the IL-1 signaling pathway disrupter comprises an IL-1 receptor antagonist.
8. The method of claim 6, wherein the IL-1 signaling pathway disrupter comprises one or more of an antibody against IL-la, IL-Ib, both IL-la or IL-Ib.
9. The method of claim 6, wherein the IL-1 signaling pathway disrupter comprises an agent effective to deactivate MyD88.
10. The method of claim 6, wherein the IL-1 signaling pathway disrupter comprises an antibody against the IL-1 receptor.
11. The method of any of claims 1-10, wherein the co-administering comprises administering the signaling pathway disrupter to the subject within an appropriate time before, during or following a gene therapy treatment.
12. The method of claim 1 wherein the co-administering comprises periodically administering the signaling pathway disrupter to the patient following a gene therapy treatment.
13. A method of treating a mammal comprising: (a) administering a recombinant adeno associated virus vector to a mammal; and (b) co-administering an IL-1 signaling pathway disrupter to the mammal in an amount effective to have a therapeutic effect on said mammal and wherein said therapeutic effect is a suppression of an immune response to the gene therapy.
14. The method of claim 13, wherein the mammal is a patient in need of gene therapy treatment.
15. The method of claim 13, wherein said IL-1 signaling pathway disrupter is selected from the group consisting of an antibody against IL-la and an antibody against IL-Ib.
16. The method of claim 13, wherein said IL-1 signaling pathway disrupter comprises IL-1 receptor antagonist.
17. The method of claim 13, wherein said co-administering comprises injecting the IL-1 signaling pathway disrupter into the muscle tissue of said mammal.
18. The method of claim 13, wherein said co-administering comprises injecting the IL-1 signaling pathway disrupter intraperitoneally into said mammal.
19. The method of claim 13, wherein said IL-1 signaling pathway disrupter is injected at one or more sites per dose.
20. The method of claim 13, wherein said co-administering comprises administering the IL-1 signaling pathway disrupter orally into said mammal.
PCT/US2021/029300 2020-04-27 2021-04-27 Methods for suppressing immune response in gene therapy WO2021222175A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063015712P 2020-04-27 2020-04-27
US63/015,712 2020-04-27

Publications (1)

Publication Number Publication Date
WO2021222175A1 true WO2021222175A1 (en) 2021-11-04

Family

ID=78332143

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/029300 WO2021222175A1 (en) 2020-04-27 2021-04-27 Methods for suppressing immune response in gene therapy

Country Status (1)

Country Link
WO (1) WO2021222175A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020079256A1 (en) * 2018-10-19 2020-04-23 Genethon Modulation, monitoring and prediction of the immune response directed against aav gene therapy vectors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020079256A1 (en) * 2018-10-19 2020-04-23 Genethon Modulation, monitoring and prediction of the immune response directed against aav gene therapy vectors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GOLDEN-MASON, L ET AL.: "Hepatic interleuklin 15 ( IL -15) expression: implications for local NK/NKT cell homeostasis and development", CLINICAL AND EXPERIMENTAL IMMUNOLOGY, vol. 138, no. 1, October 2004 (2004-10-01), pages 94 - 101, XP071085539, DOI: 10.1111/j.1365-2249.2004.02586.x *

Similar Documents

Publication Publication Date Title
JP6373763B2 (en) AAV vector components and methods for gene transfer into cells, organs and tissues
JP6495273B2 (en) Compositions, methods and uses for mutant AAV and gene transfer into cells, organs and tissues
JP2022065003A (en) Modified capsid proteins for enhanced delivery of parvovirus vectors
KR20160026841A (en) Vectors comprising stuffer/filler polynucleotide sequences and methods of use
US7504098B2 (en) Method for introducing a biological molecule using a viral envelope and heparin and system therefore
US20240043849A1 (en) Engineered viral vector reduces induction of inflammatory and immune responses
JP2020530834A (en) Apheresis method and use
US11981911B2 (en) Compositions and methods for inhibiting viral vector-induced inflammatory responses
JP2022530095A (en) Composition useful for the treatment of Rett syndrome
WO2023169115A1 (en) Aav vector having high affinity with nervous system, and application thereof
EP3983550A1 (en) New gene therapy constructs
WO2021222175A1 (en) Methods for suppressing immune response in gene therapy
JP2021519612A (en) Liver-tropic recombinant AAV6 vector to avoid neutralization
US20240024507A1 (en) Novel compositions with tissue-specific targeting motifs and compositions containing same
TW202305124A (en) Novel compositions with brain-specific targeting motifs and compositions containing same
Croze et al. Viral Vector Technologies and Strategies: Improving on Nature
WO2021222178A1 (en) Adjunct therapy for suppressing immune response against gene therapy
US20220133909A1 (en) Highly efficient transduction and lateral spread in the retina by a novel aav virus enhanced by rational design
WO2019155833A1 (en) Improved adeno-associated virus vector
Gabal Design and Intracellular Trafficking of Poly (L-Lysine)-Grafted-Poly (Ethylene Glycol) Fiberless Adenoviral Gene Delivery Vector
CN117801116A (en) Fusion type novel adeno-associated virus and application thereof
Meliani et al. 595. Enhanced Liver Transduction and Efficient Protection from Pre-Existing Neutralizing Antibodies with Exosome-Associated AAV8 Vectors
WO2024038002A1 (en) Prevention or mitigation of adverse effects related to recombinant viral vectors
JP2003146909A (en) Induction of immune tolerance for therapeutic polypeptide
KR20230003554A (en) Compositions and methods for reducing nuclease expression and off-target activity using promoters with low transcriptional activity

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: 21795337

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21795337

Country of ref document: EP

Kind code of ref document: A1