WO2021074695A1 - PD-L1 INHIBITOR - TGFβ INHIBITOR BISPECIFIC DRUG MOIETIES. - Google Patents

PD-L1 INHIBITOR - TGFβ INHIBITOR BISPECIFIC DRUG MOIETIES. Download PDF

Info

Publication number
WO2021074695A1
WO2021074695A1 PCT/IB2020/000899 IB2020000899W WO2021074695A1 WO 2021074695 A1 WO2021074695 A1 WO 2021074695A1 IB 2020000899 W IB2020000899 W IB 2020000899W WO 2021074695 A1 WO2021074695 A1 WO 2021074695A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
seq
polypeptide
sequence
less
Prior art date
Application number
PCT/IB2020/000899
Other languages
French (fr)
Other versions
WO2021074695A8 (en
Inventor
Amrik Basran
Emma JENKINS
Estelle ADAM
Matthew P. Vincent
Original Assignee
Avacta Life Sciences Limited
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 Avacta Life Sciences Limited filed Critical Avacta Life Sciences Limited
Publication of WO2021074695A1 publication Critical patent/WO2021074695A1/en
Publication of WO2021074695A8 publication Critical patent/WO2021074695A8/en

Links

Classifications

    • 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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/495Transforming growth factor [TGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/8139Cysteine protease (E.C. 3.4.22) inhibitors, e.g. cystatin
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2318/00Antibody mimetics or scaffolds
    • C07K2318/20Antigen-binding scaffold molecules wherein the scaffold is not an immunoglobulin variable region or antibody mimetics

Definitions

  • Immune checkpoint molecules are gaining prominence as targets for cancer immunotherapy, demonstrating durable remission of patients with metastatic lesions.
  • Antibodies targeting programmed death ligand 1 (PD-L1) such as atezolizumab, avelumab, and durvalumab have received regulatory approval. Despite showing remarkable durable remissions, these antibodies only demonstrate their efficacy in a subset of specific cancer types.
  • TGF-b Transforming growth factor-b
  • TGF-b is an immunosuppressive cytokine which is often produced in large quantities by many cell types in the tumor microenvironment, including tumor cells, regulatory T cells, and myeloid suppressor cells.
  • TGF-b is well known for its pleiotropic role from initiating to promoting tumor development and it has a negative effect on anti-tumor immunity by suppressing the effector functions of several immune effector cells such as neutrophils, macrophages, natural killer (NK) cells, CD8 cells, and CD4 T cells.
  • NK natural killer
  • TGF-b also induces the generation and recruitment of regulatory T cells to further suppress the antitumor T and NK cell responses.
  • TGF-b pathway promotes tumor immunosuppression, and its inhibition may enhance the antitumor activity of PD-(L)1 mAbs.
  • the present disclosure is directed to bifunctional proteins including at least portion of TGF Receptor II (TGF RII) polypeptide sequence that is capable of binding TGF (herein a “TGF trap” polypeptide) and at least one AFFIMER® polypeptide that binds (e.g., competitively or non-competitively) to Programmed Death Ligand 1 (PD-L1) and inhibits its interaction with PD-1 (herein a “PD-L1 binding AFFIMER® polypeptide”).
  • TGF RII TGF Receptor II
  • AFFIMER® polypeptide that binds (e.g., competitively or non-competitively) to Programmed Death Ligand 1 (PD-L1) and inhibits its interaction with PD-1
  • Such bispecific protein agents can exhibit a synergistic effect in cancer treatment, as compared to the effect of administering the two agents separately.
  • the present disclosure provides a bispecific fusion protein comprising a PD-L1 binding AFFIMER® polypeptide which binds to PD-L1 with a Kd of 1X10 -6 M or less and inhibits interaction of the PD-L1 to which it is bound with PD-1.
  • the PD-L1 binding AFFIMER® polypeptide binds human PD-L1 and blocks interactions with human PD-1. In some embodiments, the PD-L1 binding AFFIMER® polypeptide binds human PD-L1 and blocks interactions with human CD80. In some embodiments, the PD-L1 binding AFFIMER® polypeptide bind PD-L1 with a Kd of 1X10 _7 M or less, Kd of 1X10 _8 M or less, Kd of 1X10 _9 M or less, or even a Kd of lxlO _10 M or less.
  • the PD-L1 binding AFFIMER® polypeptide bind PD-L1 with a K 0ff of 10 3 s 1 or slower, 10 4 s 1 or slower, or even 10 5 s 1 or slower. In some embodiments, the PD- L1 binding AFFIMER® polypeptide bind PD-L1 with a K on of 10 3 M V 1 or faster, 10 4 M 1 s 1 or faster, 10 5 M 1 s 1 or faster, or even 10 6 M 1 s 1 or faster.
  • the PD-L1 binding AFFIMER® polypeptide bind PD-L1 with an IC50 in a competitive binding assay with human PD-1 of 1 mM or less, 100 nM or less, 40 nM or less, 20 nM or less, 10 nM or less, 1 nM or less, or even 0.1 nM or less.
  • the PD-L1 binding AFFIMER® polypeptide binds PD-L1 in a competitive binding assay with human CD80 (B7-1) with an IC50 of 1 mM or less, 100 nM or less, 40 nM or less, 20 nM or less, 10 nM or less, 1 nM or less, or 0.1 nM or less.
  • the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence represented in general formula (I)
  • FR1 is a polypeptide sequence represented by MIPGGLSEAK PATPEIQEIV DKVKPQLEEK TNETYGKLEA VQYKTQVLA (SEQ ID NO: 1) or a polypeptide sequence having at least 70% homology thereto;
  • FR2 is a polypeptide sequence represented by GTNYYIKVRA GDNKYMHLKV FKSL (SEQ ID NO: 2) or a polypeptide sequence having at least 70% homology thereto;
  • FR3 is a polypeptide sequence represented by EDLVLTGYQV DKNKDDELTG F (SEQ ID NO: 3) or a polypeptide sequence having at least 70% homology thereto; and Xaa, individually for each occurrence, is an amino acid residue; and n and m are each, independently, an integer from 3 to 20.
  • the FR1 may a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 1.
  • FR2 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 2.
  • FR3 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 2.
  • the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence represented in the general formula:
  • Xaa individually for each occurrence, is an amino acid residue; n and m are each, independently, an integer from 3 to 20;
  • Xaal is Gly, Ala, Val, Arg, Lys, Asp, or Glu;
  • Xaa2 is Gly, Ala, Val, Ser or Thr;
  • Xaa3 is Arg, Lys, Asn, Gin, Ser, Thr;
  • Xaa4 is Gly, Ala, Val, Ser or Thr;
  • Xaa5 is Ala, Val, lie, Leu, Gly or Pro;
  • Xaa6 is Gly, Ala, Val, Asp or Glu; and Xaa7 is Ala, Val, lie, Leu, Arg or Lys.
  • Xaal is Gly, Ala, Arg or Lys, more even more preferably Gly or Arg.
  • Xaa2 is Gly or Ser.
  • Xaa3 is Arg, Lys, Asn or Gin, more preferably Lys or Asn.
  • Xaa4 is Gly or Ser.
  • Xaa5 is Ala, Val, He, Leu, Gly or Pro, more preferably He, Leu or Pro, and even more preferably Leu or Pro.
  • Xaa6 is Ala, Val, Asp or Glu, even more preferably Ala or Glu.
  • Xaa7 is lie, Leu or Arg, more preferably Leu or Arg.
  • the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence represented in the general formula:
  • loop 2 is an amino acid sequence represented in the general formula (II)
  • aal represents an amino acid residue with a basic sidechain
  • aa2 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain, more preferably a small aliphatic sidechain, a neutral polar side chain or a basic or acid side chain
  • aa3 represents an amino acid residue with an aromatic or basic sidechain
  • aa4 represents an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain
  • aa5 represents an amino acid residue with a neutral polar or a charged (acidic or basic) or a small aliphatic or an aromatic sidechain
  • a neutral polar sidechain or a charged sidechain or a charged sidechain preferably a neutral polar sidechain or a charged sidechain
  • aa5 represents an amino acid residue with a neutral polar or a charged (acidic or basic) or a small aliphatic or an aromatic sidechain
  • aal represents Lys, Arg or His, more preferably Lys or Arg.
  • aa2 represents Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, more preferably Ala, Gin, Asp or Glu.
  • aa3 represents Phe, Tyr, Trp, Lys, Arg or His, preferably Phe, Tyr, Trp, more preferably His or Tyr, Trp or His.
  • aa4 represents Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, more preferably Gin, Lys, Arg, His, Asp or Glu.
  • aa5 represents Ser, Thr, Asn, Gin, Asp, Glu, Arg or His, more preferably Ser, Asn, Gin, Asp, Glu or Arg.
  • aa6 represents Phe, Tyr, Trp, Asp or Glu; preferably Trp or Asp; more preferably Trp.
  • (Xaa) n (“loop 2”) is an amino acid sequence represented in the general formula (III) -aal-aa2-aa3-Phe-Pro-aa4-aa5-Phe-Trp- (III) wherein aal represents an amino acid residue with a basic sidechain or aromatic sidechain; aa2 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain, more preferably a small aliphatic sidechain, a neutral polar side chain or a basic or acid side chain; aa3 represents an amino acid residue with an aromatic or basic sidechain, preferably Phe, Tyr, Trp, Lys, Arg or His, more preferably Phe, Tyr, Trp or His, and even more preferably Tyr, Trp or His; aa4 represents an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain
  • aal represents Lys, Arg, His, Ser, Thr, Asn or Gin, more preferably Lys, Arg, His, Asn or Gin, and even more preferably Lys or Asn.
  • aa2 represents Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, more preferably Ala, Gin, Asp or Glu.
  • aa3 represents Phe, Tyr, Trp, Lys, Arg or His, more preferably Phe, Tyr, Trp or His, and even more preferably Tyr, Trp or His.
  • aa4 represents Ala, Pro, He, Gin, Thr, Asp, Glu, Lys, Arg or His, and even more preferably Gin, Lys, Arg, His, Asp or Glu.
  • aa5 represents Ser, Thr, Asn, Gin, Asp, Glu, Arg or His, and even more preferably Ser, Asn, Gin, Asp, Glu or Arg.
  • (Xaa) n (“loop 2”) is an amino acid sequence selected from SEQ ID NOS: 6 to 41, or an amino acid sequence having at least 80% homology thereto, and more preferably an amino acid sequence having at least 85%, 90%, 95% or even 98% homology thereto.
  • (Xaa) n (“loop 2”) is an amino acid sequence selected from SEQ ID NOS: 6 to 41, or an amino acid sequence having at least 80% identity thereto, and more preferably an amino acid sequence having at least 85%, 90%, 95% or even 98% identity thereto.
  • (Xaa) m (“loop 4”) is an amino acid sequence represented in the general formula (IV) -aa7-aa8-aa9-aal0-aal l-aal2-aal3-aal4-aal5- (IV) wherein aa7 represents an amino acid residue with neutral polar or non-polar sidechain or an acidic sidechain; aa8 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain or aromatic sidechain, more preferably a charged (acidic or basic) sidechain; aa9 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain or aromatic sidechain, more preferably a neutral polar side chain or an acid side chain; aalO represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a
  • aa7 represents Gly, Ala, Val, Pro, Trp, Gin, Ser, Asp or Glu, and even more preferably Gly, Ala, Trp, Gin, Ser, Asp or Glu.
  • aa8 represents Asp, Glu, Lys, Arg, His, Gin, Ser, Thr, Asn, Ala, Val, Pro, Gly, Tyr or Phe, and even more preferably Asp, Glu, Lys, Arg, His or Gin.
  • aa9 represents Gin, Ser, Thr, Asn, Asp, Glu, Arg, Lys, Gly, Leu, Pro or Tyr, and even more preferably Gin, Thr or Asp.
  • aalO represents Asp, Glu, Arg, His, Lys, Ser, Gin, Asn, Ala, Leu, Tyr, Trp, Pro or Gly, and even more preferably Asp, Glu, His, Gin, Asn, Leu, Trp or Gly.
  • aall represents Asp, Glu, Ser, Thr, Gin, Arg, Lys, His, Val, He, Tyr or Gly and even more preferably Asp, Glu, Ser, Thr, Gin, Lys or His.
  • aal2 represents Asp, Glu, Ser, Thr, Gin, Asn, Lys, Arg, Val, Leu, lie, Trp, Tyr, Phe or Gly and even more preferably Asp, Glu, Ser, Tyr, Trp, Arg or Lys.
  • aal3 represents Ser, Thr, Gin, Asn, Val, lie, Leu, Gly, Pro, Asp, Glu, His, Arg, Trp, Tyr or Phe and even more preferably Ser, Thr, Gin, Asn, Val, He, Leu, Gly, Asp or Glu.
  • aal4 represents Ala, He, Trp, Pro, Asp, Glu, Arg, Lys, His, Ser, Thr, Gin or Asn and even more preferably Ala, Pro, Asp, Glu, Arg, Lys, Ser, Gin or Asn.
  • aal5 represents His, Arg, Lys, Asp, Ser, Thr, Gin, Asn, Ala, Val, Leu, Gly or Phe and even more preferably His, Arg, Lys, Asp, Ser, Thr, Gin or Asn.
  • (Xaa) n (“loop 4”) is an amino acid sequence selected from SEQ ID NOS: 42 to 77, or an amino acid sequence having at least 80% homology thereto, and more preferably an amino acid sequence having at least 85%, 90%, 95% or even 98% homology thereto.
  • (Xaa) n (“loop 4”) is an amino acid sequence selected from SEQ ID NOS: 42 to 77, or an amino acid sequence having at least 80% identity thereto, and more preferably an amino acid sequence having at least 85%, 90%, 95% or even 98% identity thereto.
  • the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence selected from SEQ ID NOS: 78 to 86, or an amino acid sequence having at least 70% homology thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% homology thereto.
  • the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence selected from SEQ ID NOS: 78 to 86, or an amino acid sequence having at least 70% identity thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% identity thereto.
  • the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence can be encoded by a polynucleotide having a coding sequence corresponding to nucleotides 1-336 of one of SEQ ID NOS: 87 to 95, or a coding sequence at least 70% identical thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% identity thereto.
  • the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence can be encoded by a polynucleotide having a coding sequence that hybridizes to any one of SEQ ID NOS: 87 to 95 under stringent conditions of 6X sodium chloride/sodium citrate (SSC) at 45°C followed by a wash in 0.2X SSC at 65°C.
  • the fusion proteins described herein bind PD-L1 through the PD- L1 binding AFFIMER® polypeptide in a manner competitive with PD-L1 binding by anti-PD- L1 antibodies Atezolizumab, Avelumab and/or Durvalumab.
  • the fusion proteins described herein include a PD-L1 binding AFFIMER® polypeptide that forms a crystal structure with PD-L1 comprising an interface involving at least 10 residues of PD-L1 selected from Ile-54, Tyr-56, Glu-58, Glu-60, Asp-61, Lys-62, Asn-63, Gin 66, Val-68, Val-76, Val-111, Arg-113, Met-115, He-116, Ser-117, Gly-120, Ala- 121, Asp- 122, Tyr-123, and Arg-125.
  • PD-L1 binding AFFIMER® polypeptide that forms a crystal structure with PD-L1 comprising an interface involving at least 10 residues of PD-L1 selected from Ile-54, Tyr-56, Glu-58, Glu-60, Asp-61, Lys-62, Asn-63, Gin 66, Val-68, Val-76, Val-111, Arg-113, Met-115, He
  • the fusion protein does not include an N-terminal methionine in the mature form, i.e., after cleavage of a secretion signal sequence the mature protein does not start with a methionine.
  • the mature protein may start with an lie rather than a Met.
  • the fusion proteins described herein in a manner dependent on the PD-L1 binding AFFIMER® polypeptide binding to PD-L1, (a) increases T-cell receptor signaling in subset of T cell bearing certain Vfi chains, for example, VB3, VB12, VB14, and VB17 in human PBMCs, when treated with staphylococcus enterotoxin B (SEB); (b) increases interferon-g production in an SEB assay; and/or (c) increases interleukin-2 (IL-2) production in an SEB assay in a dose dependant manner.
  • SEB staphylococcus enterotoxin B
  • IL-2 interleukin-2
  • the fusion proteins described herein in a manner dependent on the PD-L1 binding AFFIMER® polypeptide binding to PD-L1 (a) increases T-cell proliferation in a mixed lymphocyte reaction (MLR) assay; (b) increases interferon-g production in an MLR assay; and/or (c) increases interleukin-2 (IL-2) secretion in an MLR assay.
  • MLR mixed lymphocyte reaction
  • IL-2 interleukin-2
  • the TGF trap polypeptide has an amino acid sequence selected from SEQ ID NOS: 96, 97 and/or 98/99 or fragment thereof, or an amino acid sequence having at least 70% homology thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% homology thereto.
  • the TGF trap polypeptide retains at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99% of the TGF -binding activity of the wild-type sequence.
  • the present disclosure provides a bispecific fusion protein comprising a TGF trap polypeptide which binds to TGF with a Kd of 1X10 -6 M or less in a dimeric format, though more preferentially in a monomeric format, and even more preferably with a Kd of 1X10 _7 M, 1X10 _8 M or even 1X10 _9 M or less.
  • the TGF trap polypeptide is IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEV CVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDE CNDNIIF S EE YNT S NPD (SEQ ID NO: 105).
  • the fusion protein may also include (in addition to the PD-L1 binding AFFIMER® polypeptide and TGF trap polypeptides), to illustrate, at least one additional amino acid sequences selected from the group consisting of: secretion signal sequences, peptide linker sequences, affinity tags, transmembrane domains, cell surface retention sequence, substrate recognition sequences for post-translational modifications, multimerization domains to create multimeric structures of the protein aggregating through protein-protein interactions, half-life extending polypeptide moieties, polypeptides for altering tissue localization and antigen binding site of an antibody, and at least one additional AFFIMER® polypeptides binding the PD-L1 or a different target.
  • additional amino acid sequences selected from the group consisting of: secretion signal sequences, peptide linker sequences, affinity tags, transmembrane domains, cell surface retention sequence, substrate recognition sequences for post-translational modifications, multimerization domains to create multimeric structures of the protein aggregating through protein-
  • the fusion protein includes a half-life extending polypeptide moiety such as selected from the group consisting of an Fc domain or portion thereof, an albumin protein or portion thereof, an albumin-binding polypeptide moiety, transferrin or portion thereof, a transferrin-binding polypeptide moiety, fibronectin or portion thereof, or a fibronectin- binding polypeptide moiety.
  • a half-life extending polypeptide moiety such as selected from the group consisting of an Fc domain or portion thereof, an albumin protein or portion thereof, an albumin-binding polypeptide moiety, transferrin or portion thereof, a transferrin-binding polypeptide moiety, fibronectin or portion thereof, or a fibronectin- binding polypeptide moiety.
  • the fusion protein includes an Fc domain or a portion thereof, in some embodiments it is an Fc domain that retains FcRn binding.
  • the Fc domain or a portion thereof is from IgA, IgD, IgE, IgG, and IgM or a subclass (isotype) thereof such as IgGl, IgG2, IgG3, IgG4, IgAl or IgA2.
  • the fusion protein has an amino acid sequence of SEQ ID NO: 96, 97, and/or 98/99 or a sequence having at least 70% homology thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% identity thereto.
  • the Fc domain or a portion thereof retains effector function selected from Clq binding, complement dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of B cell receptor, or a combination thereof.
  • the Fc domain is selected from naturally occurring Fc domains which do not retain ADCC and/or CDC function, or Fc domains that have been engineered to have reduced or no ADCC and/or CDC function (such as the “LALA” mutation, i.e., human IgGl L234A/L235A (“hlgGl-LALA”)).
  • the fusion protein includes a half-life extending polypeptide moiety
  • that moiety increases the serum half-life of the protein by at least 5-fold relative to its absence from the protein, for example, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70- fold, 80-fold, 90-fold, 100-fold, 200-fold, 500-fold or even 1000-fold.
  • the recombinant receptor trap fusion protein includes at least one multimerization domains that induces multimerization of the recombinant receptor trap fusion protein, i.e., complexes including 2, 3, 4, 5, 6, 7, 8, 9 or even 10 recombinant receptor trap fusion proteins in a multimeric complex.
  • the disclosure features a fusion protein of the general formula A- B-C or C-B-A, wherein
  • A represents a PD-L1 binding AFFIMER® polypeptide
  • B represents an Fc polypeptide of a human IgG protein for providing serum half-life extension to the fusion protein; and C represents a human TGF RII trap polypeptide.
  • the fusion protein may further include amino acid linkers connecting each of the PD-L1 binding and/or TGF trap polypeptides to the Fc polypeptide.
  • the fusion protein includes an amino acid sequence of SEQ ID NO: 96, 97, or 98/99, which show the sequence of certain mature fusion proteins after removal of a secretion signal sequence.
  • the disclosure also features a method of promoting local depletion of TGFp.
  • the method includes administering a protein described above, where the protein binds TGF in solution, binds PD-L1 on a cell surface, and carries the bound TGF into the cell (e.g., a cancer cell).
  • the disclosure also features a method of inhibiting SMAD3 phosphorylation in a cell (e.g., a cancer cell or an immune cell), the method including exposing the cell in the tumor microenvironment to a protein described above.
  • a cell e.g., a cancer cell or an immune cell
  • the fusion protein of the disclosure is provided as a pharmaceutical preparation suitable for therapeutic use in a human patient, further comprising at least one pharmaceutically acceptable excipient, buffer, salt or the like.
  • the pharmaceutical preparation is formulated for pulmonary delivery.
  • the pharmaceutical preparation may be formulated as an intranasal formulation.
  • the pharmaceutical preparation is formulated for topical (e.g., transepithelial) delivery.
  • polynucleotides comprising a coding sequence encoding a fusion protein described above (and herein).
  • the coding sequence is operably linked to at least one transcriptional regulatory sequence, such as a promoter and/or enhancer.
  • the polynucleotide includes at least one origins of replication, minichromosome maintenance elements (MME) and/or nuclear localization elements.
  • MME minichromosome maintenance elements
  • the polynucleotide includes a polyadenylation signal sequence which is operably linked and transcribed with the coding sequence.
  • the coding sequence includes at least one intronic sequences
  • the polynucleotide includes at least one ribosome binding sites which are transcribed with the coding sequence.
  • the polynucleotide is DNA.
  • the polynucleotide is RNA, such as an mRNA.
  • viral vectors including a coding sequence encoding a bispecific fusion protein, such as proteins described above (and herein).
  • plasmid DNA in another aspect of the disclosure, there is provided plasmid DNA, plasmid Vectors or minicircles including a coding sequence encoding a bispecific fusion protein, such as proteins described above (and herein).
  • a pharmaceutical preparation suitable for therapeutic gene delivery in a human patient comprising a polynucleotide, a viral vector, a plasmid DNA, plasmid Vector or minicircle of the present disclosure, and (ii) at least one pharmaceutically acceptable excipients, buffers, salts, transfection enhancers, electroporation enhancers or the like.
  • the pharmaceutical preparation in some embodiments, is formulated for pulmonary (e.g., intranasal) delivery.
  • the pharmaceutical preparation in other embodiments, is formulated for topical (e.g., transepithelial) delivery.
  • any one of the AFFIMER® polypeptides described herein may include or exclude a signal sequence (e.g., ⁇ 15-30 amino acids present at the N-terminus of the polypeptide) or a tag sequence (e.g., C-terminal polyhistadine (e.g., HHHHHH (SEQ ID NO: 173))).
  • a signal sequence e.g., ⁇ 15-30 amino acids present at the N-terminus of the polypeptide
  • a tag sequence e.g., C-terminal polyhistadine (e.g., HHHHHH (SEQ ID NO: 173))
  • FIG. 1 Schematic representation of an example of fusion protein of the present disclosure (an Fc scaffolded embodiment), including certain of the biological/therapeutic activities conferred by each of the PD-F1 binding AFFIMER® polypeptide and the TGF trap polypeptide sequence.
  • FIG. 2. Generation of AFFIMER® libraries. Variabilized binding loops give rise to unique binding surfaces and selectable AFFIMER® binders.
  • FIG. 3 Monomeric AFFIMER® binding. AFFIMER® polypeptide binding by flow cytometry on two different cancer cell lines (AVA04-141 (SEQ ID NOs: 38 (Loop 2) and 74 (Loop 4)); AVA04-211 (SEQ ID NOs: 21 and 57); AVA04-227 (SEQ ID NOs: 10 and 46); AVA04-228 (SEQ ID NOs: 7 and 43); AVA04-231 (SEQ ID NOs: 18 and 54); AVA04-236 (SEQ ID NOs: 15 and 51); AVA04-251 (SEQ ID NOs: 39 and 40); AVA04-261 (SEQ ID NOs:
  • AVA04-269 (SEQ ID NOs: 8 and 44)).
  • FIG. 4 AFFIMER® multimers are expressed easily in Escherichia coli to high yield and purity of multiple formats (even in shaker flasks production).
  • FIGs. 5A and 5B AFFIMER® multimers bind to PD-L1 with kinetics demonstrating avidity beyond the monomeric binding domain.
  • FIGs. 6A and 6B AFFIMER® Fc fusions (e.g ., AVA04-251 hFcl) provide effector function, half-life extension and enhanced affinity (AVA04-251 (SEQ ID NO: 39 (Loop 2) and 75 (Loop 4)); AVA04-251 hFcl fusion (SEQ ID NO: 180)).
  • AVA04-251 hFcl SEQ ID NO: 39
  • AVA04-251 hFcl fusion SEQ ID NO: 180
  • FIG. 7 Single PD-L1 binding AFFIMER® polypeptide (e.g., AVA04-251 hFcl) arms match binding kinetics of PD-L1 antibody benchmarks.
  • AVA04-251 hFcl Single PD-L1 binding AFFIMER® polypeptide
  • FIG. 8 AFFIMER® polypeptide-Fc fusions (e.g., AVA04-251 hFcl) demonstrate increased serum half-life.
  • FIG. 10 Proof-of-concept demonstrating that AFFIMER® proteins (e.g., AVA04-236) can be formatted at various sites on an Fc (e.g., hlgGl Fc), and so should translate to IgG- AFFIMER® fusions.
  • Fc e.g., hlgGl Fc
  • Analytical SEC-HPLC used to assess purity (AVA04-236-6(EAAAK) hlgGl Fc (SEQ ID NO: 181).
  • FIG. 11 Illustrates the K D of several PD-L1 AFFIMER® polypeptide Fc formats determined using Biacore, showing highly flexible formatting for fine tuning of binding kinetics to suit therapeutic target. Avidity effects with the divalent Fc format clearly observed.
  • FIG. 12 Shows the calculated 3-dimensional structures of the anti-PD-Ll AFFIMER® AVA04-261 (Loop 2 SEQ ID NO: 9 and Loop 4 SEQ ID NO: 45) and human PD-L1 derived from the crystallization of the protein complex.
  • FIG. 13 From the crystal-derived structure of anti-PD-Ll AFFIMER® AVA04-261 bound to human PD-L1 derived, FIG.13 provides the hydrogen bonding interactions between amino acid residues at the interface of contact between the two proteins.
  • FIG. 14 From the crystal-derived structure of anti-PD-Ll AFFIMER® AVA04-261 bound to human PD-L1 derived, FIG. 14 provides a list of amino acid residues involved in the interface of contact between the two proteins.
  • FIGs. 15A-15C Protein production and protein characterization of the bispecific AFFIMER® polypeptide-Fc fusions.
  • FIG. 15A is a schematic showing the fusion protein - anti- PD-Ll AFFIMER-hlgG 1 LALA (L234A/L235A) Fc-TGFp Trap.
  • FIG. 15B shows the purity of the proteins and
  • FIG. 15C shows the size characterization of the reducing and non-reducing forms of the fusion proteins.
  • FIG. 16 SPR binding affinities of AFFIMER® polypeptide-Fc fusions to PD-Ll-Fc antigen (AVA27-01 (SEQ ID NO: 96); AVA27-02 (SEQ ID NO: 97); and AVA27-03 (SEQ ID NO: 98)).
  • FIG. 17 SPR binding affinities of AFFIMER® polypeptide-Fc fusions to TGF Beta antigen (AVA27-01 (SEQ ID NO: 96); AVA27-02 (SEQ ID NO: 97); and AVA27-03 (SEQ ID NO: 98)).
  • FIG. 18 Binding ELISA for human PD-L1 (hPD-Ll) (AVA04-251 hFcl and AVA27-
  • FIG. 19 Binding ELISA for murine PD-L1 (mPD-Ll) (AVA27-01; SEQ ID NO: 96).
  • FIGs. 20A and 20B Binding ELISA for TGFp.
  • FIG. 20A shows the results from detection of the AFFIMER® scaffold (AVA27-02 (SEQ ID NO: 97) and AVA27-03 (SEQ ID NO: 98)) with the anti-cystatin A antibody and
  • FIG. 20B shows the binding capacity of two constructs (AVA27-02 and AVA27-03) to TGF-b (and no biding of the parental molecules).
  • FIGs. 21A and 21B Binding ELISA for AVA27-01 (SEQ ID NO: 96).
  • FIG. 21A shows the results from detection of the AFFIMER® scaffold anti-mPDL-1 AFFIMER® polypeptide IgG with the anti-cystatin A antibody and
  • FIG. 21B shows the binding capacity of the construct to TGF-b (and no binding of the parental molecule).
  • FIGs. 22A and 22B Sandwich ELISA dual engagement results to examine binding of both targets.
  • FIG. 22A shows results from a bridging ELISA using hPD-Ll AFFIMER® polypeptide-Fc (AVA27-02 (SEQ ID NO: 97) and AVA27-03 (SEQ ID NO: 98)).
  • FIG. 22B shows results from a bridging ELISA using mPD-Ll AFFIMER® polypeptide-Fc (AVA27-01 (SEQ ID NO: 96)).
  • FIG. 23 Results from a PDLPD-Ll competitive ELISA showing that the constructs (e.g., anti-PD-Ll AFFIMER® polypeptide-Fc, AVA27-02 (SEQ ID NO: 97) and AVA27-03 (SEQ ID NO: 98)) disrupt the PD-1 :PD-L1 interaction.
  • constructs e.g., anti-PD-Ll AFFIMER® polypeptide-Fc, AVA27-02 (SEQ ID NO: 97) and AVA27-03 (SEQ ID NO: 98)
  • FIG. 24 Results from a PDLPD-Ll blockade bioluminescent reporter cell-based assay (AVA27-02 (SEQ ID NO: 97)).
  • FIG. 25 Results from a TGFP SMAD reporter assay demonstrating the ability of the fusion proteins (AVA27-01 (SEQ ID NO: 96); AVA27-02 (SEQ ID NO: 97); and AVA27-03 (SEQ ID NO: 98)) to neutralize TGFp.
  • FIGs. 26A-26B Cell Binding Assay (e.g., AVA27-02 (SEQ ID NO: 97)).
  • FIG. 27 Mouse pharmacokinetic (PK) half-life analysis (e.g., AVA27-02 (SEQ ID NO:
  • FIG. 28 Sandwich ELISA dual target engagement in serum from 120h PK timepoint (e.g., AVA27-02 (SEQ ID NO: 97)).
  • 120h PK timepoint e.g., AVA27-02 (SEQ ID NO: 97)
  • the current disclosure permits localized reduction in TGF in a tumor microenvironment by capturing the TGF using a soluble cytokine receptor (e.g., TGF RII) tethered to a PD-L1 binding AFFIMER® moiety targeting a PD-L1 found on the exterior surface of certain tumor cells and tumor stromal cells.
  • a soluble cytokine receptor e.g., TGF RII
  • This bifunctional molecule is effective precisely because the PD- L1 binding AFFIMER® polypeptide and TGF trap are physically linked.
  • the resulting advantage (over, for example, administration of the PD-L1 binding AFFIMER® polypeptide or an anti-PD-Ll antibody and the receptor trap as separate molecules) is partly because cytokines function predominantly in the local environment through autocrine and paracrine functions.
  • the PD-L1 binding AFFIMER® polypeptide directs the TGF trap to the tumor microenvironment where it can be most effective, by neutralizing
  • the PD-L1 binding AFFIMER® polypeptides of the present disclosure provide a number of advantages over antibodies, antibody fragments and other non-antibody binding proteins.
  • AFFIMER® polypeptide itself. In its monomeric form it is about 14 kDa, or 1/lOth the size of an antibody. This small size gives greater potential for increased tissue penetration, particularly in poorly vascularized and/or fibrotic target tissues (like tumors).
  • AFFIMER® polypeptides have a simple protein structures (versus multi-domain antibodies), and as the AFFIMER® polypeptides do not require disulfide bonds or other post- translational modifications for function, many of the format embodiments including these polypeptides can be manufactured in prokaryotic and eukaryotic systems.
  • libraries of AFFIMER® polypeptides such as the phage display techniques described in the appended examples
  • site directed mutagenesis the AFFIMER® polypeptides can be generated with tuneable binding kinetics with ideal ranges for therapeutic uses.
  • the AFFIMER® polypeptides can have high affinity for PD-L1, such as single digit nanomolar or lower KD for monomeric AFFIMER® polypeptides and picomolar KD and avidity in multi- valent formats.
  • the AFFIMER® polypeptides can be generated with tight binding kinetics for PD-L1, such as slow Koff rates in the 10 4 to 10 5 (s-1) range which benefits target tissue localization.
  • the PD-L1 binding AFFIMER® polypeptides of the present disclosure include AFFIMER® polypeptides with extraordinarily selectivity.
  • the PD-L1 binding AFFIMER® polypeptides can be readily formatted, allowing formats such as Fc fusions, whole antibody fusions and in-line multimers to be generated and manufactured with ease.
  • proteins including the PD-L1 binding AFFIMER® polypeptides or monomeric AFFIMER® polypeptide
  • PD-L1 binding AFFIMER® polypeptides or monomeric AFFIMER® polypeptide
  • gene delivery constructs that are introduced into the tissues of the patient, including formats where the protein is delivered systemically (such as expression from muscle tissue) or delivered locally (such as through intraturmoral gene delivery).
  • cystatin polypeptide refers to a sub-group of proteins in the cystatin superfamily, a family which encompasses proteins that contain multiple cystatin-like sequences.
  • the stefin sub-group of the cystatin family is relatively small (around 100 amino acids) single domain proteins. They receive no known post-translational modification, and lack disulphide bonds, suggesting that they will be able to fold identically in a wide range of extra- and intracellular environments.
  • Stefin A itself is a monomeric, single chain, single domain protein of 98 amino acids.
  • the structure of Stefin A has been solved, facilitating the rational mutation of Stefin A into the AFFIMER® Scaffold.
  • the only known biological activity of cystatins is the inhibition of cathepsin activity, which allowed us to exhaustively test for residual biological activity of our engineered proteins.
  • AFFIMER refers to small, highly stable proteins that are a recombinantly engineered variants of Stefin Polypeptides. AFFIMER® proteins display two peptide loops and an N-terminal sequence that can all be randomized to bind to desired target proteins with high affinity and specificity, in a similar manner to monoclonal antibodies. Stabilization of the two peptides by the Stefin protein scaffold constrains the possible conformations that the peptides can take, increasing the binding affinity and specificity compared to libraries of free peptides. These engineered non-antibody binding proteins are designed to mimic the molecular recognition characteristics of monoclonal antibodies in different applications.
  • the Stefin polypeptide includes a sequence derived from stefin A, sharing substantial identify with a stefin A wild type sequence, such as human Stefin A. It will be apparent to a person skilled in the art that modifications may be made to the scaffold sequence without departing from the disclosure.
  • an AFFIMER® Scaffold can have an amino acid sequences that is at least 25%, 35%, 45%, 55% or 60% identity to the corresponding sequences to human Stefin A, preferably at least 70%, at least 80%, at least 85%, at least 90%, at least 92%, at least 94%, at least 95% identical, e.g., where the sequence variations do not adversely affect the ability of the scaffold to bind to the desired target (such as PD-L1), and e.g., which do not restore or generate biological functions such as those which are possessed by wild type stefin A but which are abolished in mutational changes described herein.
  • the desired target such as PD-L1
  • Programmed death-ligand 1 also known as “PD-L1”, “cluster of differentiation 274”, “CD274”, “B7 homolog 1” or “B7-H1”, refers a protein that, in the case of humans, is encoded by the CD274 gene.
  • the human PD-L1 is a 40kDa type 1 transmembrane protein that plays a major role in suppressing the immune system under different circumstances.
  • a representative human PD-L1 sequence is provided by UniProtKB Primary accession number Q9NZQ7 and will include other human isoforms thereof.
  • PD-L1 binds to its receptor, PD-1, found on activated T cells, B cells, and myeloid cells, to modulate activation or inhibition.
  • PD-L1 also has an appreciable affinity for the costimulatory molecule CD80 (B7-1). Engagement of PD-L1 with its receptor PD-1 (“Programmed cell death protein 1” or “CD279”) on T cells delivers a signal that inhibits TCR-mediated activation of IL-2 production and T cell proliferation.
  • CD80 costimulatory molecule
  • Engagement of PD-L1 with its receptor PD-1 (“Programmed cell death protein 1” or “CD279”) on T cells delivers a signal that inhibits TCR-mediated activation of IL-2 production and T cell proliferation.
  • CD279 Protein 1
  • PD- L1 is considered a checkpoint, and its upregulated expression in tumors contributes to inhibition of T-cell mediated antitumor responses. While PD-L1 will be used generally with reference to PD-L1 from various mammalian species, it will be understood throughout the application that any reference to PD-L1 includes human PD-L1 and is, preferably, referring to human PD-
  • TGF RII or “TGF Receptor II” is meant a polypeptide having the wild-type human TGF Receptor Type 2 Isoform A sequence (e.g., the amino acid sequence of NCBI Reference Sequence (RefSeq) Accession No. NP_001020018 (SEQ ID NO: 100)), or a polypeptide having the wild-type human TGF Receptor Type 2 Isoform B sequence (e.g., the amino acid sequence of NCBI RefSeq Accession No. NP_003233 (SEQ ID NO: 101)) or having a sequence substantially identical the amino acid sequence of SEQ ID NO: 100 or of SEQ ID NO: 101.
  • RefSeq NCBI Reference Sequence
  • NP_001020018 SEQ ID NO: 100
  • a polypeptide having the wild-type human TGF Receptor Type 2 Isoform B sequence e.g., the amino acid sequence of NCBI RefSeq Accession No. NP_003233 (
  • the TGF RII may retain at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99% of the TGF -binding activity of the wild-type sequence.
  • the polypeptide of expressed TGF RII lacks the signal sequence.
  • a fragment of TGF RII capable of binding TGF is meant any portion of NCBI RefSeq Accession No. NP_001020018 (SEQ ID NO: 100) or of NCBI RefSeq Accession No. NP_003233 (SEQ ID NO: 101), or a sequence substantially identical to SEQ ID NO: 100 or SEQ ID NO: 101 that is at least 20 ( e.g ., at least 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 175, or 200) amino acids in length that retains at least some of the TGF -binding activity (e.g., at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99%) of the wild-type receptor or of the corresponding wild-type fragment. Typically, such fragment is a soluble fragment.
  • the TGF-b ligand binding domain of a TGF b receptor comprises SEQ ID NO: 102, SEQ ID NO: 103 or SEQ ID NO: 104, or portion thereof, or variant thereof (as described above):
  • Such fragment is a TOEbEII extra-cellular domain having the sequence of SEQ ID NO: 105.
  • the TGF-b ligand binding domain of a TGF b receptor comprises a sequence of the TGF-b type I receptor ectodomain, or portion of ectodomain, for example:
  • the TGF-b ligand binding domain of a TGF b receptor comprises a sequence of the TbK-III-E ⁇ , or portion of thereof, such as the ectodomain:
  • polypeptide and “peptide” and “protein” are used interchangeably herein and refer to polymers of amino acids of any length.
  • the polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing at least one analogs of an amino acid including, for example, unnatural amino acids
  • amino acid residue and “amino acid” are used interchangeably and means, in the context of a polypeptide, an amino acid that is participating in one more peptide bonds of the polypeptide.
  • abbreviations used herein for designating the amino acids are based on recommendations of the IUPAC-IUB Commission on Biochemical Nomenclature (see Biochemistry (1972) 11:1726-1732). For instance, Met, lie, Leu, Ala and Gly represent "residues” of methionine, isoleucine, leucine, alanine and glycine, respectively.
  • amino acid side chain is that part of an amino acid exclusive of the — CH(NH2)COOH portion, as defined by K. D. Kopple, "Peptides and Amino Acids", W. A. Benjamin Inc., New York and Amsterdam, 1966, pages 2 and 33.
  • amino acids used in the application of this disclosure are those naturally occurring amino acids found in proteins, or the naturally occurring anabolic or catabolic products of such amino acids which contain amino and carboxyl groups.
  • Particularly suitable amino acid side chains include side chains selected from those of the following amino acids: glycine, alanine, valine, cysteine, leucine, isoleucine, serine, threonine, methionine, glutamic acid, aspartic acid, glutamine, asparagine, lysine, arginine, proline, histidine, phenylalanine, tyrosine, and tryptophan, and those amino acids and amino acid analogs which have been identified as constituents of peptidylglycan bacterial cell walls.
  • Amino acid residues having “basic sidechains” include Arg, Lys and His.
  • Amino acid residues having “acidic sidechains” include Glu and Asp.
  • Amino acid residues having “neutral polar sidechains” include Ser, Thr, Asn, Gin, Cys and Tyr.
  • Amino acid residues having “neutral non-polar sidechains” include Gly, Ala, Val, lie, Leu, Met, Pro, Trp and Phe.
  • Amino acid residues having “non-polar aliphatic sidechains” include Gly, Ala, Val, He and Leu.
  • Amino acid residues having “hydrophobic sidechains” include Ala, Val, lie, Leu, Met, Phe, Tyr and Trp.
  • Amino acid residues having “small hydrophobic sidechains” include Ala and Val.
  • Amino acid residues having “aromatic sidechains” include Tyr, Trp and Phe.
  • amino acid residue further includes analogs, derivatives and congeners of any specific amino acid referred to herein, as for instance, the subject AFFIMER® polypeptides (particularly if generated by chemical synthesis) can include an amino acid analog such as, for example, cyanoalanine, canavanine, djenkolic acid, norleucine, 3-phosphoserine, homoserine, dihydroxy-phenylalanine, 5-hydroxytryptophan, 1-methylhistidine, 3-methylhistidine, diaminiopimelic acid, ornithine, or diaminobutyric acid.
  • amino acid analog such as, for example, cyanoalanine, canavanine, djenkolic acid, norleucine, 3-phosphoserine, homoserine, dihydroxy-phenylalanine, 5-hydroxytryptophan, 1-methylhistidine, 3-methylhistidine, diaminiopimelic acid, ornithine, or diaminobutyric acid.
  • (D) and (L) stereoisomers of such amino acids when the structure of the amino acid admits of stereoisomeric forms.
  • the configuration of the amino acids and amino acid residues herein are designated by the appropriate symbols (D), (L) or (DL), furthermore when the configuration is not designated the amino acid or residue can have the configuration (D), (L) or (DL).
  • the structure of some of the compounds of this disclosure includes asymmetric carbon atoms. It is to be understood accordingly that the isomers arising from such asymmetry are included within the scope of this disclosure. Such isomers can be obtained in substantially pure form by classical separation techniques and by sterically controlled synthesis.
  • a named amino acid shall be construed to include both the (D) or (L) stereoisomers.
  • nucleotide or percent “identity” in the context of two or more polynucleotides or polypeptides refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity.
  • the percent identity may be measured using sequence comparison software or algorithms or by visual inspection.
  • Various algorithms and software that may be used to obtain alignments of amino acid or nucleotide sequences are well-known in the art. These include, but are not limited to, BLAST, ALIGN, Megalign, BestFit, GCG Wisconsin Package, and variants thereof.
  • two polynucleotides or polypeptides of the disclosure are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using a sequence comparison algorithm or by visual inspection.
  • identity exists over a region of the amino acid sequences that is at least about 10 residues, at least about 20 residues, at least about 40-60 residues, at least about 60-80 residues in length or any integral value there between.
  • identity exists over a longer region than 60-80 residues, such as at least about 80- 100 residues, and in some embodiments the sequences are substantially identical over the full length of the sequences being compared, such as the coding region of a target protein or an antibody. In some embodiments, identity exists over a region of the nucleotide sequences that is at least about 10 bases, at least about 20 bases, at least about 40-60 bases, at least about 60-80 bases in length or any integral value there between.
  • identity exists over a longer region than 60-80 bases, such as at least about 80-1000 bases or more, and in some embodiments the sequences are substantially identical over the full length of the sequences being compared, such as a nucleotide sequence encoding a protein of interest.
  • a “conservative amino acid substitution” is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been generally defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • basic side chains
  • substitution of a phenylalanine for a tyrosine is a conservative substitution.
  • conservative substitutions in the sequences of the polypeptides, soluble proteins, and/or antibodies of the disclosure do not abrogate the binding of the polypeptide, soluble protein, or antibody containing the amino acid sequence, to the target binding site.
  • Methods of identifying amino acid conservative substitutions which do not eliminate binding are well-known in the art.
  • a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is "isolated” is a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is in a form not found in nature.
  • Isolated polypeptides, soluble proteins, antibodies, polynucleotides, vectors, cells, or compositions include those which have been purified to a degree that they are no longer in a form in which they are found in nature.
  • a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is isolated is substantially pure.
  • substantially pure refers to material which is at least 50% pure (i.e., free from contaminants), at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure.
  • fusion protein or "fusion polypeptide” as used herein refers to a hybrid protein expressed by a polynucleotide molecule comprising nucleotide sequences of at least two genes.
  • linker refers to a linker inserted between a first polypeptide (e.g ., copies of an AFFIMER® polypeptide) and a second polypeptide (e.g., another AFFIMER® polypeptide, an Fc domain, a ligand binding domain, etc.).
  • the linker is a peptide linker.
  • Linkers should not adversely affect the expression, secretion, or bioactivity of the polypeptides. Preferably, linkers are not antigenic and do not elicit an immune response.
  • the term “specifically binds to” or is “specific for” refers to measurable and reproducible interactions such as binding between a target and an AFFIMER® polypeptide, antibody or other binding partner, which is determinative of the presence of the target in the presence of a heterogeneous population of molecules including biological molecules.
  • an AFFIMER® polypeptide that specifically binds to a target is an AFFIMER® polypeptide that binds this target with greater affinity, avidity (if multimeric formatted), more readily, and/or with greater duration than it binds to other targets.
  • polynucleotide refers to polymers of nucleotides of any length and include DNA and RNA.
  • the nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase.
  • polynucleotide encoding refers to the order or sequence of nucleotides along a strand of deoxyribopolynucleotide deoxyribonucleotides. The order of these deoxyribonucleotides determines the order of amino acids along the polypeptide (protein) chain. Thus, a polynucleotide sequence encoding the amino acid sequence.
  • sequence when used in reference to nucleotide sequences, "sequence" as used herein, the term grammatical and other forms may comprise DNA or RNA and may be single or double stranded. Nucleic acid sequences may be mutated. Nucleic acid sequence may have any length, for example 2 to 000,000 or more nucleotides (or any integral value above or between) a polynucleotide, for example a length of from about 100 to about 10,000, or from about 200 nucleotides to about 500 nucleotides.
  • vector means a construct, which is capable of delivering, and usually expressing, at least one gene(s) or sequence(s) of interest in a host cell.
  • vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid, or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, and DNA or RNA expression vectors encapsulated in liposomes.
  • transfection refers to an exogenous polynucleotide into a eukaryotic cell. Transfection can be achieved by various means known in the art, including calcium phosphate -DNA co-precipitation, DEAE- dextran-mediated transfection, polybrene- mediated transfection, electroporation, microinjection, liposome fusion, lipofection, protoplast fusion, retroviral infection, and biolistics technology (biolistics).
  • carrier as used herein is an isolated polynucleotide comprising the isolated polynucleotide can be used to deliver a composition to the interior of the cell. It is known in the art a number of carriers including, but not limited to the linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses.
  • vector includes an autonomously replicating plasmid or vims. The term should also be construed to include facilitate transfer of polynucleotide into cells of the non-plasmid and non- viral compounds, for example, polylysine compounds, liposomes, and the like. Examples of viral vectors include, but are not limited to adenoviral vectors, adeno-associated vims vectors, retroviral vectors and the like.
  • expression vector refers to a vector comprising a recombinant polynucleotide comprising expression control sequence and a nucleotide sequence to be expressed operably linked.
  • the expression vector comprises sufficient cis-acting elements (ex acting elements) used for expression; other elements for expression can be supplied by the host cell or in vitro expression system.
  • Expression vectors include all those known in the art, such as cosmids, plasmids (e.g., naked or contained in liposomes) and viruses (e.g., lentivims, retrovimses, adenovimses and adeno-associated vimses).
  • operably linked refers to functional linkage between the regulatory sequence and a heterologous polynucleotide sequence is connected to a connection results in the expression of the latter.
  • first polynucleotide sequence and a second polynucleotide sequence is a functional relationship between the first polynucleotide sequence and the second polynucleotide sequence is operably linked.
  • the promoter affects the transcription or expression of the coding sequence
  • the promoter is operably linked to a coding sequence.
  • DNA sequencing operably linked are contiguous, and to join two protein coding regions in the same reading frame as necessary.
  • promoter is defined as a promoter DNA sequence recognized by the synthetic machinery required for the synthesis machinery of the cell specific transcription of a polynucleotide sequence or introduced.
  • inducible expression refers to expression under certain conditions, such as activation (or inactivation) of an intracellular signaling pathway or the contacting of the cells harboring the expression construct with a small molecule that regulates the expression (or degree of expression) of a gene operably linked to an inducible promoter sensitive to the concentration of the small molecule.
  • electroporation refers to the use of a transmembrane electric field pulse to induce microscopic pathways (pores) in a bio-membrane; their presence allows biomolecules such as plasmids or other oligonucleotide to pass from one side of the cellular membrane to the other.
  • checkpoint molecule refers to proteins that are expressed by tissues and/or immune cells and reduce the efficacy of an immune response in a manner dependent on the level of expression of the checkpoint molecule. When these proteins are blocked, the “brakes” on the immune system are released and, for example, T cells are able to kill cancer cells more effectively.
  • checkpoint proteins found on T cells or cancer cells include PD-l/PD- L1 and CTLA-4/B7-1/B7-2, PD-L2, NKG2A, KIR, LAG-3, TIM-3, CD96, VISTA and TIGIT.
  • checkpoint inhibitor refers to a drug entity that reverses the immunosuppressive signaling from a checkpoint molecule.
  • costimulatory molecule refers to an immune cell such as a T cell cognate binding partner which specifically binds to costimulatory ligands thereby mediating co- stimulation, such as, but not limited to proliferation.
  • Costimulatory molecules are cell surface molecules other than the antigen receptor or ligand which facilitate an effective immune response.
  • Co-stimulatory molecules include, but are not limited to MHCI molecules, BTLA receptor and Toll ligands, and 0X40, CD27, CD28, CDS, ICAM-1, LFA-1 (CDlla / CD18), ICOS (CD278) and 4-1BB (CD137).
  • costimulatory molecules include but are not limited to: CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46,
  • CD 160 CD19, CD4, CD8a, CD8p, IL2Rp , IL2Ry, IL7Ra, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CDlld, ITGAE, CD103, IT GAL, CDlla, LFA- 1, ITGAM, CD lib, ITGAX, CD 11c, ITGB1 , CD29, ITGB2, CD 18, LFA-1, ITGB7, NKG2D, NKG2C, TNFR2, TRANCE / RANKL, DNAM1 (CD226), SLAMF4 (CD244,2B4), CD84, CD96 (Tactile), CEACAM1, CRT AM, Ly9 (CD229) , CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Lyl08), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLA
  • costimulatory agonists refers to a drug entity that activates (agonizes) the costimulatory molecule, such as costimulatory ligand would do, and produces an immuno stimulatory signal or otherwise increases the potency or efficacy of an immune response.
  • chemotherapeutic agent is a chemical compound useful in the treatment of cancer.
  • examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclophosphamide (CYTOXAN); alkyl sulfonates such as busulfan, improsulfan, and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); delta-9-tetrahydrocannabinol (dronabinol, MARINOL); beta-lapachone; lapachol; colchicines; betulinic acid; a camptothecin (including the synthetic analogue topotecan (
  • calicheamicin especially calicheamicin gammall and calicheamicin omegall (see, e.g., Nicolaou et ah, Angew. Chem Inti. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; an esperamicin; as well as neocarzino statin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunombicin, detombicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including ADRIAMYCIN ® , morpholino
  • anti-hormonal agents that act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer, and are often in the form of systemic, or whole-body treatment. They may be hormones themselves.
  • SERMs selective estrogen receptor modulators
  • tamoxifen including NOLVADEX ® tamoxifen
  • raloxifene EVISTA ®
  • droloxifene 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (FARESTON ® ); anti-progesterones; estrogen receptor down-regulators (ERDs); estrogen receptor antagonists such as fulvestrant (FASLODEX ® ); agents that function to suppress or shut down the ovaries, for example, leutinizing hormone-releasing hormone (LHRH) agonists such as leuprolide acetate (LHRH) agonists such as leuprolide acetate (L
  • chemotherapeutic agents includes bisphosphonates such as clodronate (for example, BONEFOS ® or OSTAC ® ), etidronate (DIDROCAL), NE-58095, zoledronic acid/zoledronate (ZOMETA ® ), alendronate (FOSAMAX ® ), pamidronate (AREDIA ® ), tiludronate (SKELID ® ), or risedronate (ACTONEL ® ); as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); anti-sense oligonucleotides, particularly those that inhibit expression of genes in signaling pathways implicated in abherant cell proliferation, such as, for example, PKC-alpha, Raf, H-Ras, and epidermal growth factor receptor (EGF-R); vaccines such as THERATOP ® E vaccine and gene therapy vaccines, for example, ALLOVECTIN ® vaccine, LE
  • cytokine refers generically to proteins released by one cell population that act on another cell as intercellular mediators or have an autocrine effect on the cells producing the proteins.
  • cytokines include lymphokines, monokines; interleukins (“ILs”) such as IL-1, IL-la, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL10, IL- 11, IL-12, IL-13, IL-15, IL-17A-F, IL-18 to IL-29 (such as IL-23), IL-31, including PROLEUKIN rIL-2; a tumor-necrosis factor such as TNF-a or TNF-b, TGF- i-3; and other polypeptide factors including leukemia inhibitory factor ("LIF”), ciliary neurotrophic factor (“CNTF”), CNTF-like cytokine (“CLC”), cardiotrophin (“CT”), and kit
  • LIF leukemia inhibitory
  • chemokine refers to soluble factors (e.g., cytokines) that have the ability to selectively induce chemotaxis and activation of leukocytes. They also trigger processes of angiogenesis, inflammation, wound healing, and tumorigenesis.
  • cytokines include IL-8, a human homolog of murine keratinocyte chemoattractant (KC).
  • disfunctional also includes refractory or unresponsive to antigen recognition, specifically, impaired capacity to translate antigen recognition into down stream T-cell effector functions, such as proliferation, cytokine production (e.g., IL-2) and/or target cell killing.
  • T cell anergy refers to the state of unresponsiveness to antigen stimulation resulting from incomplete or insufficient signals delivered through the T-cell receptor (e.g. increase in intracellular Ca +2 in the absence of ras-activation). T cell anergy can also result upon stimulation with antigen in the absence of co-stimulation, resulting in the cell becoming refractory to subsequent activation by the antigen even in the context of costimulation.
  • the unresponsive state can often be overridden by the presence of Interleukin-2. Anergic T-cells do not undergo clonal expansion and/or acquire effector functions.
  • exhaustion refers to T cell exhaustion as a state of T cell dysfunction that arises from sustained TCR signaling that occurs during many chronic infections and cancer. It is distinguished from anergy in that it arises not through incomplete or deficient signaling, but from sustained signaling. It is defined by poor effector function, sustained expression of inhibitory receptors and a transcriptional state distinct from that of functional effector or memory T cells. Exhaustion prevents optimal control of infection and tumors.
  • Enhancing T-cell function means to induce, cause or stimulate a T-cell to have a sustained or amplified biological function, or renew or reactivate exhausted or inactive T-cells.
  • Examples of enhancing T-cell function include: increased secretion of ⁇ -interferon from CD8+ T-cells, increased proliferation, increased antigen responsiveness (e.g., viral, pathogen, or tumor clearance) relative to such levels before the intervention.
  • the level of enhancement is as least 50%, alternatively 60%, 70%, 80%, 90%, 100%, 120%, 150%, 200%. The manner of measuring this enhancement is known to one of ordinary skill in the art.
  • T cell dysfunctional disorder is a disorder or condition of T-cells characterized by decreased responsiveness to antigenic stimulation.
  • a T-cell dysfunctional disorder is a disorder that is specifically associated with inappropriate increased levels of PD-1.
  • a T-cell dysfunctional disorder can also be associated with inappropriate increased levels of PD-L1 in the tumor which gives rise to suppression of T-cell antitumor function(s).
  • a T-cell dysfunctional disorder is one in which T-cells are anergic or have decreased ability to secrete cytokines, proliferate, or execute cytolytic activity.
  • the decreased responsiveness results in ineffective control of a pathogen or tumor expressing an immunogen. Examples of T cell dysfunctional disorders characterized by T- cell dysfunction include unresolved acute infection, chronic infection and tumor immunity.
  • Tumor immunity refers to the process in which tumors evade immune recognition and clearance. Thus, as a therapeutic concept, tumor immunity is “treated” when such evasion is attenuated, and the tumors are recognized and attacked by the immune system. Examples of tumor recognition include tumor binding, tumor shrinkage and tumor clearance.
  • sustained response refers to the sustained effect on reducing tumor growth after cessation of a treatment.
  • the tumor size may remain to be the same or smaller as compared to the size at the beginning of the administration phase.
  • the sustained response has a duration at least the same as the treatment duration, at least 1.5x, 2. Ox, 2.5x, or 3. Ox length of the treatment duration.
  • cancer and “cancerous” as used herein refer to or describe the physiological condition in mammals in which a population of cells are characterized by unregulated cell growth.
  • examples of cancer include, but are not limited to, carcinoma, blastoma, sarcoma, and hematologic cancers such as lymphoma and leukemia.
  • tumor and neoplasm refer to any mass of tissue that results from excessive cell growth or proliferation, either benign (noncancerous) or malignant (cancerous) including pre-cancerous lesions. Tumor growth is generally uncontrolled and progressive, does not induce or inhibit the proliferation of normal cells.
  • Tumor can affect a variety of cells, tissues or organs, including but not limited to selected from bladder, bone, brain, breast, cartilage, glial cells, esophagus, fallopian tube, gall bladder, heart, intestine, kidney, liver, lung, lymph node, neural tissue, ovary, pancreas, prostate, skeletal muscle, skin, spinal cord, spleen, stomach, testis, thymus, thyroid, trachea, urethra, ureter, urethra, uterus, vagina organ or tissue or the corresponding cells.
  • Tumors include cancers, such as sarcoma, carcinoma, plasmacytoma or (malignant plasma cells).
  • Tumors of the present disclosure may include, but are not limited to leukemias (e.g ., acute leukemia, acute lymphoblastic leukemia, acute myeloid leukemia, acute myeloid leukemia, acute promyelocytic leukemia, acute myeloid - monocytic leukemia, acute monocytic leukemia, acute leukemia, chronic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, polycythemia vera), lymphomas (Hodgkin's disease, non-Hodgkin's disease), primary macroglobulinemia disease, heavy chain disease, and solid tumors such as sarcomas cancer (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, chordoma, endothelium sarcoma, lymphangiosarcoma, angiosarcoma, lymphangioendotheli
  • a "tumor” includes, but is not limited to: pancreatic cancer, liver cancer, lung cancer (including NSCLC), stomach cancer, esophageal cancer, head and neck squamous cell carcinoma, prostate cancer, colon cancer, breast cancer (including triple negative breast cancer), lymphoma, gallbladder cancer, renal cancer, leukemia, multiple myeloma, ovarian cancer, cervical cancer and glioma.
  • metalastasis refers to the process by which a cancer spreads or transfers from the site of origin to other regions of the body with the development of a similar cancerous lesion at the new location.
  • a “metastatic” or “metastasizing” cell is one that loses adhesive contacts with neighboring cells and migrates via the bloodstream or lymph from the primary site of disease to invade neighboring body structures.
  • cancer cell and “tumor cell” refer to the total population of cells derived from a cancer or tumor or pre-cancerous lesion, including both non-tumorigenic cells, which comprise the bulk of the cancer cell population, and tumorigenic stem cells (cancer stem cells).
  • cancer stem cells tumorigenic stem cells
  • progression free survival refers to the length of time during and after treatment during which the disease being treated (e.g ., cancer) does not get worse. Progression-free survival may include the amount of time patients have experienced a complete response or a partial response, as well as the amount of time patients have experienced stable disease.
  • ORR all response rate
  • all survival refers to the percentage of individuals in a group who are likely to be alive after a particular duration of time.
  • treatment refers to the individual trying to change the process or treatment of a clinical disease caused by intervention of a cell, may be either preventive intervention course of clinical pathology. Including but not limited to treatment to prevent the occurrence or recurrence of disease, alleviation of symptoms, reducing the direct or indirect pathological consequences of any disease, preventing metastasis, slow the rate of disease progression, amelioration or remission of disease remission or improved prognosis.
  • subject refers to any animal (e.g., a mammal), including, but not limited to, humans, non-human primates, canines, felines, rodents, and the like, which is to be the recipient of a particular treatment.
  • subject and patient are used interchangeably herein in reference to a human subject.
  • agonist and “agonistic” as used herein refer agents that are capable of, directly or indirectly, substantially inducing, activating, promoting, increasing, or enhancing the biological activity of a target or target pathway.
  • agonist is used herein to include any agent that partially or fully induces, activates, promotes, increases, or enhances the activity of a protein or other target of interest.
  • antagonist and “antagonistic” as used herein refer to or describe an agent that is capable of, directly or indirectly, partially or fully blocking, inhibiting, reducing, or neutralizing a biological activity of a target and/or pathway.
  • antagonist is used herein to include any agent that partially or fully blocks, inhibits, reduces, or neutralizes the activity of a protein or other target of interest.
  • modulation and modulate refer to a change or an alteration in a biological activity. Modulation includes, but is not limited to, stimulating an activity or inhibiting an activity. Modulation may be an increase in activity or a decrease in activity, a change in binding characteristics, or any other change in the biological, functional, or immunological properties associated with the activity of a protein, a pathway, a system, or other biological targets of interest.
  • immune response includes responses from both the innate immune system and the adaptive immune system. It includes both cell-mediated and/or humoral immune responses. It includes both T-cell and B-cell responses, as well as responses from other cells of the immune system such as natural killer (NK) cells, monocytes, macrophages, etc.
  • pharmaceutically acceptable refers to a substance approved or approvable by a regulatory agency of the Federal government or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans.
  • pharmaceutically acceptable excipient, carrier or adjuvant or “acceptable pharmaceutical carrier” refer to an excipient, carrier or adjuvant that can be administered to a subject, together with at least one agent of the present disclosure, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic effect.
  • pharmaceutically acceptable excipient, carrier, or adjuvant to be an inactive ingredient of any formulation.
  • the terms "effective amount” or “therapeutically effective amount” or “therapeutic effect” refer to an amount of a bispecific fusion protein described herein effective to "treat” a disease or disorder in a subject such as, a mammal.
  • the therapeutically effective amount of a bispecific fusion protein has a therapeutic effect and as such can boost the immune response, boost the anti-tumor response, increase cytolytic activity of immune cells, increase killing of tumor cells by immune cells, reduce the number of tumor cells; decrease tumorigenicity, tumorigenic frequency or tumorigenic capacity; reduce the number or frequency of cancer stem cells; reduce the tumor size; reduce the cancer cell population; inhibit or stop cancer cell infiltration into peripheral organs including, for example, the spread of cancer into soft tissue and bone; inhibit and stop tumor or cancer cell metastasis; inhibit and stop tumor or cancer cell growth; relieve to some extent at least one of the symptoms associated with the cancer; reduce morbidity and mortality; improve quality of life; or a combination of such effects.
  • treating or “treatment” or “to treat” or “alleviating” or “to alleviate” refer to both (1) therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder and (2) prophylactic or preventative measures that prevent or slow the development of a targeted pathologic condition or disorder.
  • prophylactic or preventative measures that prevent or slow the development of a targeted pathologic condition or disorder.
  • a subject is successfully "treated” according to the methods of the present disclosure if the patient shows at least one of the following: an increased immune response, an increased anti-tumor response, increased cytolytic activity of immune cells, increased killing of tumor cells by immune cells, a reduction in the number of or complete absence of cancer cells; a reduction in the tumor size; inhibition of or an absence of cancer cell infiltration into peripheral organs including the spread of cancer cells into soft tissue and bone; inhibition of or an absence of tumor or cancer cell metastasis; inhibition or an absence of cancer growth; relief of at least one symptoms associated with the specific cancer; reduced morbidity and mortality; improvement in quality of life; reduction in tumorigenicity; reduction in the number or frequency of cancer stem cells; or some combination of effects.
  • An AFFIMER ® polypeptide is a scaffold based on stefin A, meaning that it has a sequence which is derived from stefin A, for example, a mammalian stefin A, for example, a human stefin A.
  • Some aspects of the application provides AFFIMER® polypeptides which bind PD-L1 (also referred to as “anti-PD-Ll AFFIMER® polypeptides”) comprising an AFFIMER® in which at least one of the solvent accessible loops from the wild-type stefin A protein with amino acid sequences to provide an AFFIMER® polypeptide having the ability to bind PD-L1, preferably selectively, and preferably with Kd of 10 6 M or less.
  • the anti-PD-Ll AFFIMER® polypeptide is derived from the wild- type human stefin A protein having a backbone sequence and in which one or both of loop 2 [designated (Xaa) n ] and loop 4 [designated (Xaa) m ] are replaced with alternative loop sequences (Xaa) n and (Xaa) m , to have the general formula (i)
  • FR1 is a polypeptide sequence represented by MIPGGLSEAK PATPEIQEIV DKVKPQLEEK TNETYGKLEA VQYKTQVLA (SEQ ID NO: 1) or a polypeptide sequence having at least 70% homology thereto;
  • FR2 is a polypeptide sequence represented by GTNYYIKVRA GDNKYMHLKV FKSL (SEQ ID NO: 2) or a polypeptide sequence having at least 70% homology thereto;
  • FR3 is a polypeptide sequence represented by EDLVLTGYQV DKNKDDELTG F (SEQ ID NO: 3) or a polypeptide sequence having at least 70% homology thereto;
  • Xaa individually for each occurrence, is an amino acid residue
  • n and m are each, independently, an integer from 3 to 20.
  • FR1 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 1. In some embodiments, FR1 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% identity with SEQ ID NO: 1; In some embodiments, FR2 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 2.
  • FR2 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% identity with SEQ ID NO: 2;
  • FR3 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 3.
  • FR3 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% identity with SEQ ID NO: 3.
  • the anti-PD-Ll AFFIMER® polypeptide has an amino acid sequence represented in the general formula:
  • Xaa individually for each occurrence, is an amino acid residue; n and m are each, independently, an integer from 3 to 20; Xaal is Gly, Ala, Val, Arg, Lys, Asp, or Glu, more preferably Gly, Ala, Arg or Lys, and more even more preferably Gly or Arg; Xaa2 is Gly, Ala, Val, Ser or Thr, more preferably Gly or Ser; Xaa3 is Arg, Lys, Asn, Gin, Ser, Thr, more preferably Arg, Lys, Asn or Gin, and even more preferably Lys or Asn; Xaa4 is Gly, Ala, Val, Ser or Thr, more preferably Gly or Ser; Xaa5 is Ala, Val, lie, Leu, Gly or Pro, more preferably lie, Leu or Pro, and even more preferably Leu or Pro; Xaa6 is Gly, Ala, Val, Asp or Glu, more preferably Ala,
  • the anti-PD-Ll AFFIMER® polypeptide can have an amino acid sequence represented in the general formula:
  • MIPRGLSEAKPATPEIQEIVDKVKPQLEEKTNETYGKLEAVQYKTQVLA-(Xaa)n- STNYYIKVRAGDNKYMHLKVFNGP-(Xaa) m -ADRVLTGYQVDKNKDDELTGF (SEQ ID NO: 5), wherein Xaa, individually for each occurrence, is an amino acid residue; n and m are each, independently, an integer from 3 to 20.
  • n is 3 to 15, 3 to 12, 3 to 9, 3 to 7, 5 to 7, 5 to 9, 5 to 12, 5 to 15, 7 to 12 or 7 to 9.
  • m is 3 to 15, 3 to 12, 3 to 9, 3 to 7, 5 to 7, 5 to 9, 5 to 12, 5 to 15,
  • Xaa independently for each occurrence, is an amino acid that can be added to a polypeptide by recombinant expression in a prokaryotic or eukaryotic cell, and even more preferably one of the 20 naturally occurring amino acids.
  • (Xaa) n is an amino acid sequence represented in the general formula (II)
  • aal represents an amino acid residue with a basic sidechain, more preferably Lys, Arg or His, and even more preferably Lys or Arg
  • aa2 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain, more preferably a small aliphatic sidechain, a neutral polar side chain or a basic or acid side chain, even more preferably Ala, Pro, he, Gin, Thr, Asp, Glu, Lys, Arg or His, and even more preferably Ala, Gin, Asp or Glu
  • aa3 represents an amino acid residue with an aromatic or basic sidechain, preferably Phe, Tyr, Trp, Lys, Arg or His, more preferably Phe, Tyr, Trp, and even more preferably His or Tyr, Trp or His;
  • (Xaa) n is an amino acid sequence represented in the general formula (III)
  • aal represents an amino acid residue with a basic sidechain or aromatic sidechain, preferably Lys, Arg, His, Ser, Thr, Asn or Gin, more preferably Lys, Arg, His, Asn or Gin, and even more preferably Lys or Asn;
  • aa2 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain, more preferably a small aliphatic sidechain, a neutral polar side chain or a basic or acid side chain, even more preferably Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, and even more preferably Ala, Gin, Asp or Glu;
  • aa3 represents an amino acid residue with an aromatic or basic sidechain, preferably Phe, Tyr, Trp, Lys, Arg or His, more
  • (Xaa) n is an amino acid sequence selected from SEQ ID NOS: 6 to 40, or an amino acid sequence having at least 80%, 85%, 90%, 95% or even 98% homology with a sequence selected from SEQ ID NOS: 6 to 41. In some embodiments, (Xaa) n is an amino acid sequence having at least 80%, 85%, 90%, 95% or even 98% identity with a sequence selected from SEQ ID NO: 6 to 41.
  • (Xaa) m is an amino acid sequence represented in the general formula (IV)
  • (Xaa) m is an amino acid sequence selected from SEQ ID NOS: 42 to 77, or an amino acid sequence having at least 80%, 85%, 90%, 95% or even 98% homology with a sequence selected from SEQ ID NOS: 42 to 77. In some embodiments, (Xaa) m is an amino acid sequence having at least 80%, 85%, 90%, 95% or even 98% identity with a sequence selected from SEQ ID NO: 42 to 77.
  • the anti-PD-Ll AFFIMER® polypeptide has an amino acid sequence selected from SEQ ID NOS: 78 to 86, or an amino acid sequence having at least 70%, 75% 80%, 85%, 90%, 95% or even 98% homology with a sequence selected from SEQ ID NOS: 78 to 86. In some embodiments, the anti-PD-Ll AFFIMER® polypeptide has an amino acid sequence having at least 70%, 75% 80%, 85%, 90%, 95% or even 98% identity with a sequence selected from SEQ ID NO: 78 to 86. Table 4. Examples of anti-PD-Ll AFFIMER® Polypeptide Sequences
  • the anti-PD-Fl AFFIMER® polypeptide has an amino acid sequence that is encoded by a polynucleotide having a coding sequence corresponding to nucleotides 1-336 of one of SEQ ID NOS: 87 to 95, or an amino acid sequence that can be encoded by a polynucleotide having a coding sequence at least 70%, 75% 80%, 85%, 90%, 95% or even 98% identical with nucleotides 1-336 of one of SEQ ID NOS: 87 to 95, or an amino acid sequence that can be encoded by a polynucleotide having a coding sequence that hybridizes nucleotides 1-336 of one of SEQ ID NOS: 87 to 95 under stringent conditions (such as in the presence of 6X sodium chloride/sodium citrate (SSC) at 45°C followed by a wash in 0.2X SSC at 65°C.
  • stringent conditions such as in the presence of 6X sodium chloride/sodium citrate
  • minor modifications may also include small deletions or additions - beyond the loop 2 and loop 4 inserts described above - to the stefin A or stefin A derived sequences disclosed herein, such as addition or deletion of up to 10 amino acids relative to stefin A or the stefin A derived AFFIMER® polypeptide.
  • the PD-L1 binding AFFIMER® polypeptide portion that binds human PD-L1 as a monomer with a dissociation constant (K D ) of about 1 mM or less, about 100 nM or less, about 40 nM or less, about 20 nM or less, about 10 nM or less, about 1 nM or less, or about 0.1 nM or less.
  • K D dissociation constant
  • the PD-L1 binding AFFIMER® polypeptide portion binds human PD-L1 as a monomer with an off rate constant (K 0ff ), such as measured by Biacore, of about 10 3 s 1 (i.e., unit of 1/second) or slower; of about 10 4 s 1 or slower or even of about 10 5 s 1 or slower.
  • K 0ff off rate constant
  • the PD-L1 binding AFFIMER® polypeptide portion binds human PD-L1 as a monomer with an association constant (K on ), such as measured by Biacore, of at least about 10 3 M V 1 or faster; at least about 10 4 M 1 s 1 or faster; at least about 10 5 M V 1 or faster; or even at least about 10 6 M V 1 or faster.
  • K on association constant
  • the PD-L1 binding AFFIMER® polypeptide portion binds human PD-L1 as a monomer with an IC50 in a competitive binding assay with human PD-1 of 1 mM or less, about 100 nM or less, about 40 nM or less, about 20 nM or less, about 10 nM or less, about 1 nM or less, or about 0.1 nM or less.
  • the bispecific fusion protein has a melting temperature (Tm, i.e., temperature at which both the folded and unfolded states are equally populated) of 65 °C or higher, and preferably at least 70°C, 75°C, 80°C or even 85°C or higher.
  • Melting temperature is a particularly useful indicator of protein stability.
  • the relative proportions of folded and unfolded proteins can be determined by many techniques known to the skilled person, including differential scanning calorimetry, UV difference spectroscopy, fluorescence, circular dichroism (CD), and NMR (Pace et al. (1997) “Measuring the conformational stability of a protein” in Protein structure: A practical approach 2: 299-321).
  • the bispecific fusion protein In order for the bispecific fusion protein to be secreted, it will generally contain a signal sequence that directs the transport of the protein to the lumen of the endoplasmic reticulum and ultimately to be secreted (or retained on the cell surface if a transmembrane domain or other cell surface retention signal).
  • Signal sequences also referred to as signal peptides or leader sequences
  • signal sequences are cleaved from the protein by a signal peptidase after the proteins are transported to the endoplasmic reticulum.
  • the cleavage of the signal sequence from the polypeptide usually occurs at a specific site in the amino acid sequence and is dependent upon amino acid residues within the signal sequence.
  • the signal peptide is about 5 to about 40 amino acids in length (such as about 5 to about 7, about 7 to about 10, about 10 to about 15, about 15 to about 20, about 20 to about 25, or about 25 to about 30, about 30 to about 35, or about 35 to about 40 amino acids in length).
  • the signal peptide is a native signal peptide from a human protein.
  • the signal peptide is a non-native signal peptide.
  • the non-native signal peptide is a mutant native signal peptide from the corresponding native secreted human protein, and can include at least one (such as 2, 3, 4, 5, 6,
  • the signal peptide is a signal peptide or mutant thereof from a non- IgSF protein family, such as a signal peptide from an immunoglobulin (such as IgG heavy chain or IgG-kappa light chain), a cytokine (such as interleukin -2 (IL-2), or CD33), a serum albumin protein (e.g . HSA or albumin), a human azurocidin preprotein signal sequence, a luciferase, a trypsinogen ( e.g . chymotrypsinogen or trypsinogen) or other signal peptide able to efficiently secrete a protein from a cell.
  • a signal peptide include, but are not limited to:
  • the recombinant polypeptide comprises a signal peptide when expressed, and the signal peptide (or a portion thereof) is cleaved from the bispecific fusion protein upon secretion.
  • the subject fusion proteins may also include at least one linkers separating heterologous protein sequences or domains, i.e., separating the PD-L1 binding AFFIMER® polypeptide sequence from the TGF trap polypeptide from each other or from other polypeptides also present in the fusion protein (such as Fc domains or other domains that enhance serum half-life, provide multimerization, etc.).
  • linkers separating heterologous protein sequences or domains, i.e., separating the PD-L1 binding AFFIMER® polypeptide sequence from the TGF trap polypeptide from each other or from other polypeptides also present in the fusion protein (such as Fc domains or other domains that enhance serum half-life, provide multimerization, etc.).
  • linker refers to a linker amino acid sequence inserted between a first polypeptide (e.g ., an AFFIMER® polypeptide) and a second polypeptide (e.g., a second AFFIMER® polypeptide, an Fc region, a TGF receptor trap, albumin, etc.).
  • a first polypeptide e.g ., an AFFIMER® polypeptide
  • a second polypeptide e.g., a second AFFIMER® polypeptide, an Fc region, a TGF receptor trap, albumin, etc.
  • Empirical linkers designed by researchers are generally classified into 3 categories according to their structures: flexible linkers, rigid linkers, and in vivo cleavable linkers.
  • linkers may offer many other advantages for the production of fusion proteins, such as improving biological activity, increasing expression yield, and achieving desirable pharmacokinetic profiles. Linkers should not adversely affect the expression, secretion, or bioactivity of the fusion protein. Linkers should not be antigenic and should not elicit an immune response.
  • Suitable linkers are known to those of skill in the art and often include mixtures of glycine and serine residues and often include amino acids that are sterically unhindered. Other amino acids that can be incorporated into useful linkers include threonine and alanine residues. Linkers can range in length, for example from 1-50 amino acids in length, 1-22 amino acids in length, 1-10 amino acids in length, 1-5 amino acids in length, or 1-3 amino acids in length.
  • the linker may comprise a cleavage site.
  • the linker may comprise an enzyme cleavage site, so that the second polypeptide may be separated from the first polypeptide.
  • the linker can be characterized as flexible.
  • Flexible linkers are usually applied when the joined domains require a certain degree of movement or interaction. They are generally composed of small, non-polar (e.g. Gly) or polar (e.g. Ser or Thr) amino acids. See, for example, Argos P. (1990) “An investigation of oligopeptides linking domains in protein tertiary structures and possible candidates for general gene fusion” J Mol Biol. 211:943- 958. The small size of these amino acids provides flexibility and allows for mobility of the connecting functional domains.
  • the incorporation of Ser or Thr can maintain the stability of the linker in aqueous solutions by forming hydrogen bonds with the water molecules, and therefore reduces the unfavorable interaction between the linker and the protein moieties.
  • the most commonly used flexible linkers have sequences consisting primarily of stretches of Gly and Ser residues (“GS” linker).
  • An example of the most widely used flexible linker has the sequence of (Gly-Gly-Gly-Gly-Ser)n (SEQ ID NO: 174). By adjusting the copy number “n”, the length of this GS linker can be optimized to achieve appropriate separation of the functional domains, or to maintain necessary inter-domain interactions.
  • many other flexible linkers have been designed for recombinant fusion proteins. As These flexible linkers are also rich in small or polar amino acids such as Gly and Ser but can contain additional amino acids such as Thr and Ala to maintain flexibility, as well as polar amino acids such as Lys and Glu to improve solubility.
  • the linker can be characterized as rigid. While flexible linkers have the advantage to connect the functional domains passively and permitting certain degree of movements, the lack of rigidity of these linkers can be a limitation in certain fusion protein embodiments, such as in expression yield or biological activity. The ineffectiveness of flexible linkers in these instances was attributed to an inefficient separation of the protein domains or insufficient reduction of their interference with each other. Under these situations, rigid linkers have been successfully applied to keep a fixed distance between the domains and to maintain their independent functions.
  • rigid linkers are chosen when the spatial separation of the domains is critical to preserve the stability or bioactivity of the fusion proteins.
  • alpha helix-forming linkers with the sequence of (EAAAK)n (SEQ ID NO: 146) have been applied to the construction of many recombinant fusion proteins.
  • Another type of rigid linkers has a Pro-rich sequence, (XP)n, with X designating any amino acid, preferably Ala, Lys, or Glu.
  • linkers include:
  • linkers that may be used in the subject fusion proteins include, but are not limited to, SerGly, GGSG (SEQ ID NO: 175), GSGS (SEQ ID NO: 176), GGGS (SEQ ID NO: 177), S(GGS)n (SEQ ID NO: 178) where n is 1-7, GRA, poly(Gly), poly(Ala), GGGSGGG (SEQ ID NO: 157), ESGGGGVT (SEQ ID NO: 158), LESGGGGVT (SEQ ID NO: 159), GRAQVT (SEQ ID NO: 160), WRAQVT (SEQ ID NO: 161), and ARGRAQVT (SEQ ID NO: 162).
  • the hinge regions of the Fc fusions described below may also be considered linkers.
  • the bispecific fusion protein may not have a half-life and/or PK profile that is optimal for the route of administration, such as parenteral therapeutic dosing.
  • the term “half-life” refers to the amount of time it takes for a substance, such as a bispecific fusion protein of the present disclosure, to lose half of its pharmacologic or physiologic activity or concentration.
  • Biological half-life can be affected by elimination, excretion, degradation ( e.g ., enzymatic) of the substance, or absorption and concentration in certain organs or tissues of the body.
  • biological half-life can be assessed by determining the time it takes for the blood plasma concentration of the substance to reach half its steady state level (“plasma half-life”).
  • plasma half-life steady state level
  • half-life extending moiety refers to a pharmaceutically acceptable moiety, domain, or molecule covalently linked (“conjugated” or “fused”) to the AFFIMER® polypeptide to form the bispecific fusion proteins described herein, optionally via a non-naturally encoded amino acid, directly or via a linker, that prevents or mitigates in vivo proteolytic degradation or other activity-diminishing modification of the AFFIMER® polypeptide, increases half-life, and/or improves or alters other pharmacokinetic or biophysical properties including but not limited to increasing the rate of absorption, reducing toxicity, improving solubility, reducing protein aggregation, increasing biological activity and/or target selectivity of the modified AFFIMER® polypeptide, increasing manufacturability, and/or reducing immunogenicity of the modified AFFIMER® polypeptide, compared to a comparator such as an unconjugated form of the modified AFFIMER® polypeptide.
  • half-life extending moiety includes non- pro teinaceous, half-life extending moieties, such as a water soluble polymer such as polyethylene glycol (PEG) or discrete PEG, hydroxyethyl starch (HES), a lipid, a branched or unbranched acyl group, a branched or unbranched C8-C30 acyl group, a branched or unbranched alkyl group, and a branched or unbranched C8-C30 alkyl group; and proteinaceous half-life extending moieties, such as serum albumin, transferrin, adnectins ( e.g .
  • a water soluble polymer such as polyethylene glycol (PEG) or discrete PEG, hydroxyethyl starch (HES), a lipid, a branched or unbranched acyl group, a branched or unbranched C8-C30 acyl group, a branched or unbranche
  • albumin-binding or pharmacokinetics extending (PKE) adnectins albumin-binding or pharmacokinetics extending (PKE) adnectins), Fc domain, and unstructured polypeptide, such as XTEN and PAS polypeptide (e.g. conformationally disordered polypeptides composed of the amino acids Pro, Ala, and/or Ser), and a fragment of any of the foregoing.
  • PKE pharmacokinetics extending
  • PAS polypeptide e.g. conformationally disordered polypeptides composed of the amino acids Pro, Ala, and/or Ser
  • An examination of the crystal structure of an AFFIMER® polypeptide and its interaction with its target, such as the anti-PD-Ll AFFIMER® polypeptide complex with PD-1 shown in the FIGS. can indicate which certain amino acid residues have side chains that are fully or partially accessible to solvent.
  • the half-life extending moiety extends the half-life of the resulting bispecific fusion protein circulating in mammalian blood serum compared to the half-life of the protein that is not so conjugated to the moiety (such as relative to the AFFIMER® polypeptide alone). In some embodiments, half-life is extended by greater than or greater than about 1.2-fold, 1.5-fold, 2.0-fold, 3.0-fold, 4.0-fold., 5.0-fold, or 6.0-fold. In some embodiments, half-life is extended by more than 6 hours, more than 12 hours, more than 24 hours, more than 48 hours, more than 72 hours, more than 96 hours or more than 1 week after in vivo administration compared to the protein without the half-life extending moiety.
  • half-life extending moieties that can be used in the generation of bispecific fusion proteins of the disclosure include:
  • XTEN also known as recombinant PEG or “rPEG”
  • HAP homoamino acid polymer
  • PAS proline-alanine-serine polymer
  • ELP elastin- like peptide
  • normally long-half-life proteins such as HSA, human IgG, transferrin or fibronectin.
  • Fc fusions involve the fusion of peptides, proteins or receptor exodomains to the Fc portion of an antibody. Both Fc and albumin fusions achieve extended half-lives not only by increasing the size of the peptide drug, but both also take advantage of the body’s natural recycling mechanism: the neonatal Fc receptor, FcRn. The pH-dependent binding of these proteins to FcRn prevents degradation of the fusion protein in the endosome.
  • Fusions based on these proteins can have half-lives in the range of 3-16 days, much longer than typical PEGylated or lipidated peptides. Fusion to antibody Fc domains can improve the solubility and stability of the peptide or protein drug.
  • An example of a peptide Fc fusion is dulaglutide, a GLP-1 receptor agonist currently in late-stage clinical trials. Human serum albumin, the same protein exploited by the fatty acylated peptides is the other popular fusion partner. Albiglutide is a GLP-1 receptor agonist based on this platform.
  • Fc and albumin A major difference between Fc and albumin is the dimeric nature of Fc versus the monomeric structure of HSA leading to presentation of a fused peptide as a dimer or a monomer depending on the choice of fusion partner.
  • the dimeric nature of an AFFIMER® polypeptide-Fc fusion can produce an avidity effect if the AFFIMER® target, such as PD-L1 on tumor cells, are spaced closely enough together or are themselves dimers. This may be desirable or not depending on the target.
  • the AFFIMER® polypeptide may be part of a fusion protein with an immunoglobulin Fc domain ("Fc domain"), or a fragment or variant thereof, such as a functional Fc region.
  • Fc domain immunoglobulin Fc domain
  • an Fc fusion such as a bispecific fusion protein created as an AFFIMER® polypeptide-Fc fusion protein, is a polypeptide comprising at least one AFFIMER® sequences covalently linked through a peptide backbone (directly or indirectly) to an Fc region of an immunoglobulin.
  • An Fc-fusion may comprise, for example, the Fc region of an antibody (which facilitates effector functions and pharmacokinetics) and an AFFIMER® polypeptide sequence as part of the same polypeptide.
  • An immunoglobulin Fc region may also be linked indirectly to at least one AFFIMER® polypeptides.
  • linkers are known in the art and can optionally be used to link an Fc to a polypeptide including an AFFIMER® polypeptide sequence to generate an Fc-fusion.
  • Fc-fusions can be dimerized to form Fc-fusion homodimers, or using non-identical Fc domains, to form Fc- fusion heterodimers.
  • Fc region of human antibodies for use in generating the subject bispecific fusion proteins as AFFIMER® fusion proteins.
  • the principle rationale is to produce a stable protein, large enough to demonstrate a similar pharmacokinetic profile compared with those of antibodies, and to take advantage of the properties imparted by the Fc region; this includes the salvage neonatal FcRn receptor pathway involving FcRn- mediated recycling of the fusion protein to the cell surface post endocytosis, avoiding lysosomal degradation and resulting in release back into the bloodstream, thus contributing to an extended serum half-life.
  • Another obvious advantage is the Fc domain’s binding to Protein A, which can simplify downstream processing during production of the bispecific fusion protein and permit generation of highly pure preparation of the bispecific fusion protein.
  • an Fc domain will include the constant region of an antibody excluding the first constant region immunoglobulin domain.
  • Fc domain refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, and the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains.
  • IgA and IgM Fc may include the J chain.
  • Fc comprises immunoglobulin domains Cyl and Cy3 and the hinge between Cy 1 and Cy2.
  • the human IgG heavy chain Fc region is usually defined to comprise residues C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU index as set forth in Rabat (Rabat et ah, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, NIH, Bethesda, Md. (1991)).
  • Fc may refer to this region in isolation, or this region in the context of a whole antibody, antibody fragment, or Fc fusion protein. Polymorphisms have been observed at a number of different Fc positions and are also included as Fc domains as used herein.
  • a “functional Fc region” refers to an Fc domain or fragment thereof which retains the ability to bind FcRn.
  • a functional Fc region binds to FcRn but does not possess effector function.
  • effector functions include Clq binding; complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors ( e.g ., B cell receptor; BCR), etc.
  • effector functions can be assessed using various assays known in the art for evaluating such antibody effector functions.
  • the Fc domain is derived from an IgGl subclass, however, other subclasses (e.g., IgG2, IgG3, and IgG4) may also be used.
  • IgG2, IgG3, and IgG4 An example of a sequence of a human IgGl immunoglobulin Fc domain which can be used is:
  • the Fc region used in the fusion protein may comprise the hinge region of an Fc molecule.
  • An example hinge region comprises the core hinge residues spanning positions 1-16 (i.e., DKTHTCPPCPAPELLG (SEQ ID NO: 163) of the example human IgGl immunoglobulin Fc domain sequence provided above.
  • the AFFIMER® polypeptide-containing fusion protein may adopt a multimeric structure (e.g., dimer) owing, in part, to the cysteine residues at positions 6 and 9 within the hinge region of the example human IgGl immunoglobulin Fc domain sequence provided above.
  • the hinge region as used herein may further include residues derived from the CHI and CH2 regions that flank the core hinge sequence of the example human IgGl immunoglobulin Fc domain sequence provided above.
  • the hinge sequence may comprise or consist of GS THT CPPCP APELLG (SEQ ID NO: 164) or EPKSCDKTHTCPPCPAPELLG (SEQ ID NO: 165).
  • the hinge sequence may include at least one substitution that confer desirable pharmacokinetic, biophysical, and/or biological properties.
  • Some example hinge sequences include:
  • EPKSCDKTHTCPPCPAPELLGGPS SEQ ID NO: 179; EPKS S DKTHT CPPCP APELLGGPS (SEQ ID NO: 166); EPKS S DKTHT CPPCP APELLGGS S (SEQ ID NO: 167);
  • EPKS S GSTHTCPPCPAPELLGGS S (SEQ ID NO: 168); DKTHTCPPCPAPELLGGPS (SEQ ID NO: 169) and DKTHT CPPCP APELLGGS S (SEQ ID NO: 170).
  • the residue P at position 18 of the example human IgGl immunoglobulin Fc domain sequence provided above may be replaced with S to ablate Fc effector function; this replacement is exemplified in hinges having the sequences EPKS SDKTHTCPPCPAPELLGGS S (SEQ ID NO: 167), EPKSSGSTHTCPPCPAPELLGGSS (SEQ ID NO: 168), and DKTHTCPPCPAPELLGGSS (SEQ ID NO: 170).
  • residues DK at positions 1-2 of the example human IgGl immunoglobulin Fc domain sequence provided above may be replaced with GS to remove a potential clip site; this replacement is exemplified in the sequence EPKSSGSTHTCPPCPAPELLGGSS (SEQ ID NO: 168).
  • the C at the position 103 of the heavy chain constant region of human IgGl may be replaced with S to prevent improper cysteine bond formation in the absence of a light chain; this replacement is exemplified by EPKSSDKTHTCPPCPAPELLGGPS (SEQ ID NO: 166), EPKS SDKTHTCPPCPAPELLGGS S (SEQ ID NO: 167), and EPKSSGSTHTCPPCPAPELLGGSS (SEQ ID NO: 168).
  • the Fc is a mammalian Fc such as a human Fc, including Fc domains derived from IgGl, IgG2, IgG3 or IgG4.
  • the Fc region may possess at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with a native Fc region and/or with an Fc region of a parent polypeptide.
  • the Fc region may have at least about 90% sequence identity with a native Fc region and/or with an Fc region of a parent polypeptide.
  • the Fc domain comprises an amino acid sequence selected from SEQ ID NO: 131, or an Fc sequence from the examples provided by SEQ ID NOS: 132-144. It should be understood that the C-terminal lysine of an Fc domain is an optional component of a fusion protein comprising an Fc domain. In some embodiments, the Fc domain comprises an amino acid sequence selected from SEQ ID NOS: 131-144, except that the C-terminal lysine thereof is omitted. In some embodiments, the Fc domain comprises the amino acid sequence of SEQ ID NO: 131. In some embodiments, the Fc domain comprises the amino acid sequence of SEQ ID NO: 131 except the C-terminal lysine thereof is omitted.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcRs Fc receptors
  • cytotoxic cells e.g ., Natural Killer (NK) cells, neutrophils, and macrophages
  • NK Natural Killer
  • the fusion protein includes an Fc domain sequence for which the resulting bispecific fusion protein has no (or reduced) ADCC and/or complement activation or effector functionality.
  • the Fc domain may comprise a naturally disabled constant region of IgG2 or IgG4 isotype or a mutated IgGl constant region.
  • suitable modifications are described in EP0307434.
  • One example comprises the substitutions of alanine residues at positions 235 and 237 (EU index numbering).
  • the fusion protein includes an Fc domain sequence for which the resulting bispecific fusion protein will retain some or all Fc functionality for example will be capable of one or both of ADCC and CDC activity, as for example if the fusion protein comprises the Fc domain from human IgGl or IgG3.
  • Levels of effector function can be varied according to known techniques, for example by mutations in the CH2 domain, for example wherein the IgGl CH2 domain has at least one mutations at positions selected from 239 and 332 and 330, for example the mutations are selected from S239D and I332E and A330L such that the antibody has enhanced effector function, and/or for example altering the glycosylation profile of the antigen-binding protein of the disclosure such that there is a reduction in fucosylation of the Fc region.
  • the bispecific fusion protein is a fusion protein comprising, in addition to at least one AFFIMER® polypeptide sequence, an albumin sequence or an albumin fragment.
  • the bispecific fusion protein is conjugated to the albumin sequence or an albumin fragment through chemical linkage other than incorporation into the polypeptide sequence including the AFFIMER® polypeptide.
  • the albumin, albumin variant, or albumin fragment is human serum albumin (HSA), a human serum albumin variant, or a human serum albumin fragment.
  • HSA human serum albumin
  • Albumin serum proteins comparable to HSA are found in, for example, cynomolgus monkeys, cows, dogs, rabbits and rats.
  • bovine serum albumin is the most structurally similar to HSA. See, e.g., Kosa et ah, (2007) J Pharm Sci. 96(11):3117-24.
  • the present disclosure contemplates the use of albumin from non-human species, including, but not limited to, albumin sequence derived from cyno serum albumin or bovine serum albumin.
  • Mature HSA a 585 amino acid polypeptide (approx. 67 kDa) having a serum half-life of about 20 days, is primarily responsible for the maintenance of colloidal osmotic blood pressure, blood pH, and transport and distribution of numerous endogenous and exogenous ligands.
  • the protein has three structurally homologous domains (domains I, II and III), is almost entirely in the alpha-helical conformation, and is highly stabilized by 17 disulfide bridges.
  • the bispecific fusion protein can be an albumin fusion protein including at least one AFFIMER® polypeptides and the sequence for mature human serum albumin (SEQ ID NO: 145) or a variant or fragment thereof which maintains the PK and/or biodistribution properties of mature albumin to the extent desired in the fusion protein.
  • the albumin sequence can be set off from the AFFIMER® polypeptide sequence or other flanking sequences in the bispecific fusion protein by use of linker sequences as described above.
  • HSA human serum albumin
  • a signal peptide of 18 amino acids MKW VTFIS LLFLF S S AY S (SEQ ID NO: 109)
  • RGVFRR 6 amino acids
  • This 24 amino acid residue peptide may be referred to as the pre-pro domain.
  • the AFFIMER® polypeptide-HSA fusion proteins can be expressed and secreted using the HSA pre-pro-domain in the recombinant proteins coding sequence.
  • the AFFIMER® polypeptide-HSA fusion can be expressed and secreted through inclusion of other secretion signal sequences, such as described above.
  • the serum albumin polypeptide can be covalently coupled to the AFFIMER® polypeptide through a bond other than a backbone amide bond, such as cross-linked through chemical conjugation between amino acid sidechains on each of the albumin polypeptide and the AFFIMER® polypeptide.
  • the bispecific fusion protein can include a serum-binding moiety - either as part of a fusion protein (if also a polypeptide) with the AFFIMER® polypeptide sequence or chemically conjugated through a site other than being part of a contiguous polypeptide chain.
  • the serum-binding polypeptide is an albumin binding moiety.
  • Albumin contains multiple hydrophobic binding pockets and naturally serves as a transporter of a variety of different ligands such as fatty acids and steroids as well as different drugs. Furthermore, the surface of albumin is negatively charged making it highly water-soluble.
  • albumin binding moiety refers to any chemical group capable of binding to albumin, i.e. has albumin binding affinity.
  • Albumin binds to endogenous ligands such as fatty acids; however, it also interacts with exogenous ligands such as warfarin, penicillin and diazepam.
  • endogenous ligands such as fatty acids
  • exogenous ligands such as warfarin, penicillin and diazepam.
  • the albumin-drug complex serves as a drug reservoir that can enhance the drug biodistribution and bioavailability. Incorporation of components that mimic endogenous albumin-binding ligands, such as fatty acids, has been used to potentiate albumin association and increase drug efficacy.
  • lipidation which involves the covalent binding of fatty acids to peptide side chains.
  • PEGylation a basic mechanism of half-life extension as PEGylation, namely increasing the hydrodynamic radius to reduce renal filtration.
  • the lipid moiety is itself relatively small and the effect is mediated indirectly through the non-covalent binding of the lipid moiety to circulating albumin.
  • lipidation reduces the water-solubility of the peptide but engineering of the linker between the peptide and the fatty acid can modulate this, for example by the use of glutamate or mini PEGs within the linker.
  • Linker engineering and variation of the lipid moiety can affect self-aggregation which can contribute to increased half-life by slowing down biodistribution, independent of albumin. See, for example, Jonassen et al. (2012) Pharm Res. 29(8):2104-14.
  • a wide variety of macromolecular polymers and other molecules can be linked to the AFFIMER® polypeptides of the present disclosure to modulate biological properties of the resulting bispecific fusion protein, and/or provide new biological properties to the bispecific fusion protein.
  • These macromolecular polymers can be linked to the AFFIMER® polypeptide via a naturally encoded amino acid, via a non-naturally encoded amino acid, or any functional substituent of a natural or non-natural amino acid, or any substituent or functional group added to a natural or non-natural amino acid.
  • the molecular weight of the polymer may be of a wide range, including but not limited to, between about 100 Da and about 100,000 Da or more.
  • the molecular weight of the polymer may be between about 100 Da and about 100,000 Da, including but not limited to, 100,000 Da, 95,000 Da, 90,000 Da, 85,000 Da, 80,000 Da, 75,000 Da, 70,000 Da, 65,000 Da, 60,000 Da, 55,000 Da, 50,000 Da, 45,000 Da, 40,000 Da, 35,000 Da, 30,000 Da, 25,000 Da, 20,000 Da, 15,000 Da, 10,000 Da, 9,000 Da, 8,000 Da, 7,000 Da, 6,000 Da, 5,000 Da, 5,000 Da, 4,000 Da, 3,000 Da, 2,000 Da, 1,000 Da, 900 Da, 800 Da, 700 Da, 600 Da, 500 Da, 400 Da, 300 Da, 200 Da, and 100 Da. In some embodiments, the molecular weight of the polymer is between about 100 Da and about 50,000 Da.
  • the molecular weight of the polymer is between about 100 Da and about 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 1,000 Da and about 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 5,000 Da and about 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 10,000 Da and about 40,000 Da.
  • polymers include but are not limited to polyalkyl ethers and alkoxy-capped analogs thereof (e.g ., polyoxyethylene glycol, polyoxyethylene/propylene glycol, and methoxy or ethoxy-capped analogs thereof, especially polyoxyethylene glycol, the latter is also known as polyethylene glycol or PEG); discrete PEG (dPEG); polyvinylpyrrolidones; polyvinylalkyl ethers; polyoxazolines, polyalkyl oxazolines and polyhydroxyalkyl oxazolines; polyacrylamides, polyalkyl acrylamides, and polyhydroxyalkyl acrylamides (e.g., polyhydroxypropylmethacrylamide and derivatives thereof); polyhydroxyalkyl acrylates; polysialic acids and analogs thereof; hydrophilic peptide sequences; polysaccharides and their derivatives, including dextran and dextran derivatives, e.g., carboxymethyldextran, dextran
  • the polymer selected may be water soluble so that the bispecific fusion protein to which it is attached does not precipitate in an aqueous environment, such as a physiological environment.
  • the water soluble polymer may be any structural form including but not limited to linear, forked or branched.
  • the water soluble polymer is a poly(alkylene glycol), such as poly(ethylene glycol) (PEG), but other water soluble polymers can also be employed.
  • PEG poly(ethylene glycol)
  • the polymer may be pharmaceutically acceptable.
  • PEG polyethylene glycol molecule
  • a PEG used in the polypeptides of the disclosure terminates on one end with hydroxy or methoxy, i.e., X is H or CH 3 (“methoxy PEG”).
  • the other end of the PEG may attach to the AFFIMER® polypeptide via a naturally-occurring or non- naturally encoded amino acid.
  • the attachment may be through an amide, carbamate or urea linkage to an amine group (including but not limited to, the epsilon amine of lysine or the N-terminus) of the polypeptide.
  • the polymer is linked by a maleimide linkage to a thiol group (including but not limited to, the thiol group of cysteine) - which in the case of attachment to the AFFIMER® polypeptide sequence per se requires altering a residue in the AFFIMER® polypeptide sequence to a cysteine.
  • a maleimide linkage to a thiol group (including but not limited to, the thiol group of cysteine) - which in the case of attachment to the AFFIMER® polypeptide sequence per se requires altering a residue in the AFFIMER® polypeptide sequence to a cysteine.
  • the number of water soluble polymers linked to the AFFIMER® polypeptide can be adjusted to provide an altered (including but not limited to, increased or decreased) pharmacologic, pharmacokinetic or pharmacodynamic characteristic such as in vivo half-life in the resulting bispecific fusion protein.
  • the half-life of the resulting bispecific fusion protein is increased at least about 10, 20, 30, 40, 50, 60, 70, 80, 90 percent, 2-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 11- fold, 12-fold, 13-fold, 14-fold, 15-fold, 16-fold, 17-fold, 18-fold, 19-fold, 20-fold, 25-fold, 30- fold, 35-fold, 40-fold, 50-fold, or at least about 100-fold over an unmodified polypeptide.
  • polymer system useful to modify the PK or other biological properties of the resulting bispecific fusion protein are the use of unstructured, hydrophilic amino acid polymers that are functional analogs of PEG, particularly as part of a fusion protein with the AFFIMER® polypeptide sequence.
  • the inherent biodegradability of the polypeptide platform makes it attractive as a potentially more benign alternative to PEG.
  • Another advantage is the precise molecular structure of the recombinant molecule in contrast to the polydispersity of PEG.
  • the recombinant fusions to unstructured partners can, in many cases, be subjected to higher temperatures or harsh conditions such as HPLC purification.
  • XTEN Amunix
  • a recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner” 2009 Nat Biotechnol. 27(12): 1186-90. Enabled by the biodegradable nature of the polymer, this is much larger than the 40 KDa PEGs typically used and confers a concomitantly greater half-life extension.
  • the fusion of XTEN to the AFFIMER® containing polypeptide should result in half-life extension of the final bispecific fusion protein by 60- to 130-fold over the unmodified polypeptide.
  • PAS XL-Protein GmbH.
  • Schlapschy et al. “PASYlation: a biological alternative to PEGylation for extending the plasma half-life of pharmaceutically active proteins” 2013 Protein Eng Des Sel. 26(8):489-501.
  • a random coil polymer comprised of an even more restricted set of only three small uncharged amino acids, proline, alanine and serine.
  • the PAS modification can be genetically encoded with the AFFIMER® polypeptide sequence to produce an inline fusion protein when expressed.
  • Recombinant fusion proteins described herein can be produced by any suitable method known in the art. Such methods range from direct protein synthesis methods to constructing a DNA sequence encoding polypeptides and expressing those sequences in a suitable host. For those recombinant fusion proteins including further modifications, such as a chemical modifications or conjugation, the recombinant fusion protein can be further manipulated chemically or enzymatically after isolation form the host cell or chemical synthesis.
  • the present disclosure includes recombinant methods and polynucleotides for recombinantly expressing the recombinant fusion proteins of the present disclosure comprising (i) introducing into a host cell a polynucleotide encoding the amino acid sequence of said bispecific fusion protein, for example, wherein the polynucleotide is in a vector and/or is operably linked to a promoter; (ii) culturing the host cell (e.g ., eukaryotic or prokaryotic) under condition favorable to expression of the polynucleotide and, (iii) optionally, isolating the bispecific fusion protein from the host cell and/or medium in which the host cell is grown. See e.g., WO 04/041862, WO 2006/122786, WO 2008/020079, WO 2008/142164 or WO 2009/068627.
  • WO 04/041862 WO 2006/122786, WO 2008/020079, WO
  • a DNA sequence encoding a recombinant fusion protein of interest may be constructed by chemical synthesis using an oligonucleotide synthesizer. Oligonucleotides can be designed based on the amino acid sequence of the desired polypeptide and selecting those codons that are favored in the host cell in which the recombinant polypeptide of interest will be produced. Standard methods can be applied to synthesize a polynucleotide sequence encoding an isolated polypeptide of interest. For example, a complete amino acid sequence can be used to construct a back-translated gene. Further, a DNA oligomer containing a nucleotide sequence coding for the particular isolated polypeptide can be synthesized. For example, several small oligonucleotides coding for portions of the desired polypeptide can be synthesized and then ligated. The individual oligonucleotides typically contain 5' or 3' overhangs for complementary assembly.
  • the vector for the production of the recombinant fusion protein may be produced by recombinant DNA technology using techniques well known in the art. Methods which are well known to those skilled in the art can be used to construct expression vectors containing the recombinant bispecific fusion protein coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
  • An expression vector comprising the nucleotide sequence of a recombinant fusion protein can be transferred to a host cell by conventional techniques (e.g ., electroporation, liposomal transfection, and calcium phosphate precipitation) and the transfected cells are then cultured by conventional techniques to produce the recombinant fusion protein of the disclosure.
  • the expression of the recombinant fusion protein is regulated by a constitutive, an inducible or a tissue, specific promoter.
  • the expression vector may include an origin of replication, such as may be selected based upon the type of host cell being used for expression.
  • an origin of replication such as may be selected based upon the type of host cell being used for expression.
  • the origin of replication from the plasmid pBR322 is useful for most Gram- negative bacteria while various origins from SV40, polyoma, adenovirus, vesicular stomatitus virus (VSV) or papillomaviruses (such as HPV or BPV) are useful for cloning vectors in mammalian cells.
  • the origin of replication component is not needed for mammalian expression vectors (for example, the SV40 origin is often used because it contains the early promoter).
  • the vector may include at least one selectable marker genes, e.g., genetic elements that encode a protein necessary for the survival and growth of a host cell grown in a selective culture medium.
  • selectable marker genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, tetracycline, or kanamycin for prokaryotic host cells, (b) complement auxotrophic deficiencies of the cell; or (c) supply critical nutrients not available from complex media.
  • Preferred selectable markers are the kanamycin resistance gene, the ampicillin resistance gene, and the tetracycline resistance gene.
  • a neomycin resistance gene may also be used for selection in prokaryotic and eukaryotic host cells.
  • selection genes may be used to amplify the gene which will be expressed. Amplification is a process where genes which are in greater demand for the production of a protein critical for growth are reiterated in tandem within the chromosomes of successive generations of recombinant cells. Examples of selectable markers for mammalian cells include dihydrofolate reductase (DHFR) and thymidine kinase. The mammalian cell transformants are placed under selection pressure which only the transformants are uniquely adapted to survive by virtue of the marker present in the vector.
  • DHFR dihydrofolate reductase
  • thymidine kinase thymidine kinase
  • Selection pressure is imposed by culturing the transformed cells under conditions in which the concentration of selection agent in the medium is successively changed, thereby leading to amplification of both the selection gene and the DNA that encodes the recombinant fusion protein. As a result, increased quantities of the recombinant fusion protein are synthesized from the amplified DNA.
  • the vector may also include at least one ribosome binding site, which will be transcribed into the mRNA including the coding sequence for the recombinant fusion protein.
  • a site is characterized by a Shine-Dalgarno sequence (prokaryotes) or a Kozak sequence (eukaryotes).
  • the element is typically located 3' to the promoter and 5' to the coding sequence of the polypeptide to be expressed.
  • the Shine-Dalgarno sequence is varied but is typically a polypurine (having a high A-G content). Many Shine-Dalgarno sequences have been identified, each of which can be readily synthesized using methods set forth above and used in a prokaryotic vector.
  • the expression vectors will typically contain a promoter that is recognized by the host organism and operably linked to a polynucleotide encoding the recombinant fusion protein.
  • Either a native or heterologous promoter may be used depending the host cell used for expression and the yield desired.
  • Promoters for use with prokaryotic hosts include the beta-lactamase and lactose promoter systems; alkaline phosphatase, a tryptophan (trp) promoter system; and hybrid promoters such as the tac promoter. Other known bacterial promoters are also suitable. Their sequences have been published, and they can be ligated to a desired polynucleotide sequence(s), using linkers or adapters as desired to supply restriction sites.
  • Promoters for use with yeast hosts are also known in the art.
  • Yeast enhancers are advantageously used with yeast promoters.
  • Suitable promoters for use with mammalian host cells are well known and include those obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Virus 40 (SV40).
  • Other suitable mammalian promoters include heterologous mammalian promoters, e.g., heat-shock promoters and the actin promoter.
  • Additional promoters which may be used for expressing the selective binding agents of the disclosure include, but are not limited to: the SV40 early promoter region (Bemoist and Chambon, Nature, 290:304-310, 1981); the CMV promoter; the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto et al. (1980), Cell 22: 787-97); the herpes thymidine kinase promoter (Wagner et al. (1981), Proc. Natl. Acad. Sci. U.S.A. 78: 1444- 5); the regulatory sequences of the metallothionine gene (Brinster et al, Nature, 296; 39-42,
  • prokaryotic expression vectors such as the beta- lactamase promoter (Villa-Kamaroff, et al., Proc. Natl. Acad. Sci. U.S.A. , 75; 3727-3731, 1978); or the tac promoter (DeBoer, et al. (1983), Proc. Natl. Acad. Sci. U.S.A., 80: 21-5).
  • animal transcriptional control regions which exhibit tissue specificity and have been utilized in transgenic animals: the elastase I gene control region which is active in pancreatic acinar cells (Swift et al. (1984), Cell 38: 639-46; Omitz et al. (1986), Cold Spring Harbor Symp. Quant.
  • beta-globin gene control region which is active in myeloid cells (Mogram et al., Nature, 315 338- 340, 1985; Kollias et al. (1986), Cell 46: 89-94); the myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al. (1987), Cell, 48: 703-12); the myosin light chain-2 gene control region which is active in skeletal muscle (Sani (1985), Nature, 314: 283-6); and the gonadotropic releasing hormone gene control region which is active in the hypothalamus (Mason et al. (1986), Science 234: 1372-8).
  • An enhancer sequence may be inserted into the vector to increase transcription in eukaryotic host cells.
  • enhancer sequences available from mammalian genes are known (e.g., globin, elastase, albumin, alpha-feto-protein and insulin).
  • an enhancer from a virus will be used.
  • the SV40 enhancer, the cytomegalovirus early promoter enhancer, the polyoma enhancer, and adenovirus enhancers are examples of enhancing elements for the activation of eukaryotic promoters.
  • an enhancer may be spliced into the vector at a position 5' or 3' to the polypeptide coding region, it is typically located at a site 5' from the promoter.
  • Vectors for expressing polynucleotides include those which are compatible with bacterial, insect, and mammalian host cells. Such vectors include, inter alia, pCRIE, pCR3, and pcDNA3.1 (Invitrogen Company, San Diego, Calif.), pBSII (Stratagene Company, La Jolla, Calif.), pET15 (Novagen, Madison, Wis.), pGEX (Pharmacia Biotech, Piscataway, N.J.), pEGFP-N2 (Clontech, Palo Alto, Calif.), pETL (BlueBacII; Invitrogen), pDSR- alpha (PCT Publication No. WO90/14363) and pFastBacDual (Gibco/BRL, Grand Island, N.Y.).
  • Additional possible vectors include, but are not limited to, cosmids, plasmids or modified viruses, but the vector system must be compatible with the selected host cell.
  • Such vectors include but are not limited to plasmids such as BLUESCRIPT® plasmid derivatives (a high copy number ColEl-based phagemid, Stratagene Cloning Systems Inc., La Jolla Calif.), PCR cloning plasmids designed for cloning Taq-amplified PCR products (e.g., TOPOTM.
  • TA Cloning® Kit PCR2.1 plasmid derivatives, Invitrogen, Carlsbad, Calif.
  • mammalian, yeast or virus vectors such as a baculovirus expression system (pBacPAK plasmid derivatives, Clontech, Palo Alto, Calif.) ⁇
  • pBacPAK plasmid derivatives Clontech, Palo Alto, Calif.
  • the recombinant molecules can be introduced into host cells via transformation, transfection, infection, electroporation, or other known techniques
  • Eukaryotic and prokaryotic host cells including mammalian cells as hosts for expression of the recombinant fusion protein disclosed herein are well known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC). These include, inter alia, Chinese hamster ovary (CHO) cells, NSO, SP2 cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g ., Hep G2), A549 cells, 3T3 cells, HEK-293 cells and a number of other cell lines.
  • Mammalian host cells include human, mouse, rat, dog, monkey, pig, goat, bovine, horse and hamster cells.
  • Cell lines of particular preference are selected through determining which cell lines have high expression levels.
  • Other cell lines that may be used are insect cell lines, such as Sf9 cells, amphibian cells, bacterial cells, plant cells and fungal cells.
  • Fungal cells include yeast and filamentous fungus cells including, for example, Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia minuta ( Ogataea minuta, Pichia lindneri ), Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Kluyveromy
  • Pichia sp. any Saccharomyces sp., Hansenula polymorpha, any Kluyveromyces sp., Candida albicans, any Aspergillus sp., Trichoderma reesei, Chrysosporium lucknowense, any Fusarium sp., Yarrowia lipolytica, and Neurospora crassa.
  • host-expression vector systems may be utilized to express the recombinant fusion protein of the disclosure.
  • Such host-expression systems represent vehicles by which the coding sequences of the recombinant fusion protein may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express the recombinant fusion protein of the disclosure in situ.
  • These include, but are not limited to, microorganisms such as bacteria (e.g., E. coli and B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing fusion protein coding sequences; yeast (e.g., Saccharomyces pichia ) transformed with recombinant yeast expression vectors containing fusion protein coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the fusion protein coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus (CpMV) and tobacco mosaic virus (TMV)) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing fusion protein coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 293T, 3T3 cells, lymphotic cells (see U.S.
  • mammalian cell systems e.g., COS, CHO,
  • Per C.6 cells rat retinal cells developed by Crucell harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).
  • mammalian cells e.g., metallothionein promoter
  • mammalian viruses e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter.
  • a number of expression vectors may be advantageously selected depending upon the use intended for the recombinant fusion protein being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of the recombinant fusion protein, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al. (1983) "Easy Identification Of cDNA Clones," EMBO J.
  • fusion protein coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced;
  • pIN vectors Inouye et al. (1985) "Up- Promoter Mutations In The Lpp Gene Of Escherichia coli," Nucleic Acids Res. 13:3101-3110; Van Heeke et al. (1989) "Expression Of Human Asparagine Synthetase In Escherichia coli," J. Biol. Chem. 24:5503-5509); and the like.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to a matrix glutathione-agarose beads followed by elution in the presence of free gluta-thione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • Autographa califomica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • the fusion protein coding sequence may be cloned individually into non-essential regions (e.g., the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (e.g., the polyhedrin promoter).
  • a number of viral-based expression systems may be utilized.
  • the fusion protein coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the immunoglobulin molecule in infected hosts (see e.g., see Logan et al.
  • initiation signals may also be required for efficient translation of inserted fusion protein coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bitter et al.
  • a host cell strain may be chosen which modulates the expression of the inserted sequences or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
  • Such mammalian host cells include but are not limited to CHO, VERY, BHK, Hela, COS, MDCK, 293, 293T, 3T3, WI38, BT483, Hs578T, HTB2, BT20 and T47D, CRL7030 and Hs578Bst.
  • stable expression is contemplated.
  • cell lines which stably express an antibody of the disclosure may be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • This method may advantageously be used to engineer cell lines which express the recombinant fusion proteins of the disclosure. Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the recombinant fusion proteins.
  • a number of selection systems may be used, including but not limited to the herpes simplex vims thymidine kinase (Wigler et al. (1977) "Transfer Of Purified Herpes Virus Thymidine Kinase Gene To Cultured Mouse Cells," Cell 11:223-232), hypoxanthine-guanine phosphoribosyltransferase (Szybalska et al. (1962) "Genetics Of Human Cess Line. IV. DNA- Mediated Heritable Transformation Of A Biochemical Trait," Proc. Natl. Acad. Sci.
  • the expression levels of a recombinant fusion protein can be increased by vector amplification (for a review, see Bebbington and Hentschel, "The Use of Vectors Based On Gene Amplification For The Expression Of Cloned Genes In Mammalian Cells," in DNA CLONING, Vol. 3. (Academic Press, New York, 1987)).
  • a marker in the vector system expressing a recombinant fusion protein is amplifiable
  • increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the nucleotide sequence of the recombinant fusion protein, production of the recombinant fusion protein will also increase (Crouse et al. (1983) "Expression and Amplification Of Engineered Mouse Dihydrofolate Reductase Minigenes," Mol. Cell. Biol. 3:257-266).
  • the host cell may be co-transfected with two expression vectors, for instance the first vector encoding a heavy chain and the second vector encoding a light chain derived polypeptide, one or both of which includes an AFFIMER® polypeptide coding sequence.
  • the two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
  • a single vector may be used which encodes both heavy and light chain polypeptides.
  • the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot (1986) "Expression And Amplification Of Engineered Mouse Dihydrofolate Reductase Minigenes," Nature 322:562-565; Kohler (1980) "Immunoglobulin Chain Loss In Hybridoma Lines," Proc. Natl. Acad. Sci. (U.S.A.) 77:2197-2199).
  • the coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
  • glycoproteins produced in a particular cell line or transgenic animal will have a glycosylation pattern that is characteristic for glycoproteins produced in the cell line or transgenic animal. Therefore, the particular glycosylation pattern of the recombinant fusion protein will depend on the particular cell line or transgenic animal used to produce the protein.
  • a glycosylation pattern comprising only non-fucosylated N-glycans may be advantageous, because in the case of antibodies this has been shown to typically exhibit more potent efficacy than fucosylated counterparts both in vitro and in vivo (See for example, Shinkawa et ah, J. Biol. Chem. 278: 3466-3473 (2003); U.S. Pat. Nos. 6,946,292 and 7,214,775).
  • GS system glutamine synthetase gene expression system
  • the mammalian host cells e.g ., CHO
  • the polynucleotide encoding the immunoglobulin chain comprises a glutamine synthetase gene which complements the lack of the gene in the host cell.
  • Such host cells containing the binder or polynucleotide or vector as discussed herein as well as expression methods, as discussed herein, for making the binder using such a host cell are part of the present disclosure.
  • insect cell culture systems e.g ., baculovirus
  • Baculovirus systems for production of heterologous proteins in insect cells are well-known to those of skill in the art.
  • the recombinant fusion proteins produced by a transformed host can be purified according to any suitable method.
  • Standard methods include chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification.
  • Affinity tags such as hexa-histidine, maltose binding domain, influenza coat sequence, and glutathione-S-transferase can be attached to the protein to allow easy purification by passage over an appropriate affinity column.
  • Isolated proteins can also be physically characterized using such techniques as proteolysis, mass spectrometry (MS), nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC), and x-ray crystallography.
  • recombinant fusion proteins produced in bacterial culture can be isolated, for example, by initial extraction from cell pellets, followed by at least one concentration, salting-out, aqueous ion exchange, or size exclusion chromatography steps. HPLC can be employed for final purification steps.
  • Microbial cells employed in expression of a recombinant protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
  • Gene-based a polynucleotide encoding an AFFIMER® polypeptide can present a labor- and cost-effective alternative to the conventional production, purification and administration of the polypeptide version of the bispecific fusion protein.
  • a number of antibody expression platforms have been pursued in vivo to which delivery of a polynucleotide encoding an AFFIMER® polypeptide can be adapted: these include viral vectors, naked DNA and RNA. Gene transfer can not only enable cost-savings by reducing the cost of goods and of production but may also be able to reduce the frequency of drug administration.
  • a prolonged in vivo production of the therapeutic bispecific fusion protein by expression of the polynucleotide encoding an AFFIMER® polypeptide can contribute to (i) a broader therapeutic or prophylactic application of bispecific fusion proteins in price-sensitive conditions, (ii) an improved accessibility to therapy in both developed and developing countries, and (iii) more effective and affordable treatment modalities.
  • cells can be harvested from the host (or a donor), engineered with a polynucleotide encoding an AFFIMER® polypeptide to produce bispecific fusion proteins and re-administered to patients.
  • Intramuscular antibody gene administration has been most widely evaluated (reviewed in Deal et al. (2015) “Engineering humoral immunity as prophylaxis or therapy” Curr Opin Immunol. 35:113-22.) and carries the highest clinical translatability and application when applied to polynucleotides encoding an AFFIMER® polypeptide.
  • the inherent anatomical, cellular and physiological properties of skeletal muscle make it a stable environment for long-term polunucleotide expression and systemic circulation. Skeletal muscle is easily accessible, allowing multiple or repeated administrations.
  • the abundant blood vascular supply provides an efficient transport system for secreted therapeutic bispecific fusion proteins into the circulation.
  • the syncytial nature of muscle fibers allows dispersal of nucleotides from a limited site of penetration to a large number of neighboring nuclei within the fiber.
  • Skeletal muscle fibers are also terminally differentiated cells, and nuclei within the fibers are post-mitotic. Consequently, integration in the host genome is not a prerequisite to attain prolonged mAb expression.
  • the liver is another site often used for pre-clinical antibody gene transfer, and is typically transfected via i.v. injection, and can also be a site of gene transfer for polynucleotides either for local delivery of bispecific fusion proteins (such as in the treatment of liver cancer and/or metaplasias) or for the generation of bispecific fusion proteins that are secreted into the vascular for systemic circulation.
  • This organ has various physiological functions, including the synthesis of plasma proteins. This organ can be particularly well suited for in vivo polynucleotide expression.
  • the tumor presents another site for polynucleotide transfer, targeted either via i.v. or direct injection/electroporation.
  • intratumoral polynucleotide expression can allow for a local production of the therapeutic bispecific fusion proteins, waiving the need for high systemic bispecific fusion protein levels that might otherwise be required to penetrate and impact solid tumors.
  • a similar rationale applies for the brain, which is frequently targeted in the context of antibody gene transfer to avoid the difficulties with blood-brain barrier trafficking and would likewise be a target for delivery of polynucleotides. See, for example, Beckman et al. (2015) “Antibody constructs in cancer therapy: protein engineering strategies to improve exposure in solid tumors” Cancer 109(2): 170-9; Dronca et al.
  • cationic liposome technology can be employed, which is based on the ability of amphipathic lipids, possessing a positively charged head group and a hydrophobic lipid tail, to bind to negatively charged DNA or RNA and form particles that generally enter cells by endocytosis.
  • Some cationic liposomes also contain a neutral co-lipid, thought to enhance liposome uptake by mammalian cells. See, for example, Feigner et al. (1987) Lipofection: a highly efficient, lipid- mediated DNA-transfection procedure. MNAS 84:7413-7417; San et al. (1983) “Safety and short-term toxicity of a novel cationic lipid formulation for human gene therapy” Hum. Gene Ther. 4:781-788; Xu et al. (1996) “Mechanism of DNA release from cationic liposome/DNA complexes used in cell transfection” Biochemistry 35,:5616-5623; and Legendre et al. (1992) “Delivery of plasmid DNA into mammalian cell lines using pH-sensitive liposomes: comparison with cationic liposomes” Pharm. Res. 9, 1235-1242.
  • polycations such as poly-l-lysine and polyethylene-imine
  • polycations can be used to deliver polynucleotides.
  • These polycations complex with polynucleotides via charge interaction and aid in the condensation of DNA or RNA into nanoparticles, which are then substrates for endosome-mediated uptake.
  • cationic polynucleotide complex technologies have been developed as potential clinical products, including complexes with plasmid DNA, oligodeoxynucleotides, and various forms of synthetic RNA. Modified (and unmodified or “naked”) DNA and RNA have also been shown to mediate successful gene transfer in a number of circumstances and can also be used as systems for delivery of polynucleotides.
  • Viral vectors are currently used as a delivery vehicle in the vast majority of pre-clinical and clinical gene therapy trials and in the first to be approved directed gene therapy. See Gene Therapy Clinical Trials Worldwide 2017 (abedia.com/wiley/). The main driver thereto is their exceptional gene delivery efficiency, which reflects a natural evolutionary development; viral vector systems are attractive for gene delivery, because viruses have evolved the ability to cross through cellular membranes by infection, thereby delivering polynucleotides such as polynucleotides to target cells. Pioneered by adenoviral systems, the field of viral vector- mediated antibody gene transfer made significant strides in the past decades.
  • Muscle has emerged as the administration site of choice for prolonged mAb expression and would similarly be a suitable target tissue for prolonged bispecific fusion protein expression.
  • oncolytic viruses have a distinct advantage, as they can specifically target tumor cells, boost bispecific fusion protein expression, and amplify therapeutic responses - such as to a PD- L1 bispecific fusion protein.
  • Nonviral vectors are easily produced and do not seem to induce specific immune responses.
  • Muscle tissue is most often used as target tissue for transfection, because muscle tissue is well vascularized and easily accessible, and myocytes are long-lived cells.
  • Intramuscular injection of naked plasmid DNA results in transfection of a certain percentage of myocytes.
  • plasmid DNA encoding cytokines and cytokine/IgGl chimeric proteins has been introduced in vivo and has positively influenced (autoimmune) disease outcome.
  • intravascular delivery in which increased gene delivery and expression levels are achieved by inducing a short-lived transient high pressure in the veins.
  • Special blood-pressure cuffs that may facilitate localized uptake by temporarily increasing vascular pressure and can be adapted for use in human patients for this type of gene delivery. See, for example, Zhang et al. (2001) “Efficient expression of naked DNA delivered intraarterially to limb muscles of nonhuman primates” Hum. Gene Ther., 12:427-438
  • Increased efficiency can also be gained through other techniques, such as in which delivery of the polynucleotide is improved by use of chemical carriers — cationic polymers or lipids — or via a physical approach — gene gun delivery or electroporation.
  • chemical carriers — cationic polymers or lipids — or via a physical approach — gene gun delivery or electroporation.
  • electroporation is especially regarded as an interesting technique for nonviral gene delivery. Somiari, et al.
  • Encoded PD-L1 binding AFFIMER® polypeptides can be delivered by a wide range of gene delivery system commonly used for gene therapy including viral, non- viral, or physical.
  • Constitutive promoters such as the human elongation factor la-subunit (EFla), immediate-early cytomegalovirus (CMV), chicken b-actin (CBA) and its derivative CAG, the b glucuronidase (GUSB), or ubiquitin C (UBC) can be used to promote expression of the polynucleotide in most tissues.
  • EFla human elongation factor la-subunit
  • CMV immediate-early cytomegalovirus
  • CBA chicken b-actin
  • GUSB b glucuronidase
  • UBC ubiquitin C
  • CBA and CAG promote the larger expression among the constitutive promoters; however, their size of -1.7 kbs in comparison to CMV (-0.8 kbs) or EFla (-1.2 kbs) may limit use in vectors with packaging constraints such as AAV, particularly where bispecific fusion protein produced by expression of the polynucleotide is large.
  • the GUSB or UBC promoters can provide ubiquitous gene expression with a smaller size of 378 bps and 403 bps, respectively, but they are considerably weaker than the CMV or CBA promoter.
  • promoters can be used to mediate this specificity.
  • the nervous system promoters have been used to restrict expression to neurons, astrocytes, or oligodendrocytes.
  • the neuron- specific enolase (NSE) promoter drives stronger expression than ubiquitous promoters.
  • the platelet-derived growth factor B-chain (PDGF-b), the synapsin (Syn), and the methyl-CpG binding protein 2 (MeCP2) promoters can drive neuron- specific expression at lower levels than NSE.
  • the 680 bps-long shortened version [gfaABC(l)D] of the glial fibrillary acidic protein (GFAP, 2.2 kbs) promoter can confer higher levels of expression with the same astrocyte-specificity as the GFAP promoter.
  • Targeting oligodendrocytes can also be accomplished by the selection of the myelin basic protein (MBP) promoter, whose expression is restricted to this glial cell; however, its size of 1.9 kbs and low expression levels limit its use.
  • MBP myelin basic protein
  • a-MHC a-myosin heavy chain
  • promoters to restrict expression to only liver hepatocytes after vector-mediated gene transfer has been shown to reduce transgene- specific immune responses in systems where that is a risk, and to even induce immune tolerance to the expressed protein (Zhang et ah, 2012 Hum Gene Ther. 23(5):460-72), which for certain bispecific fusion proteins may be beneficial.
  • the a 1 -antitrypsin (hAAT; 347 bps) and the thyroxine binding globulin (TBG; -400 bps) promoters drive gene expression restricted to the liver with minimal invasion to other tissues (Yan et ah, 2012 Gene. 506(2):289-94; Cunningham et al., 2008 Mol Ther. 16(6): 1081-8).
  • a mechanism to control the duration and amount of in vivo polynucleotide expression will typically be desired.
  • inducible promoters which can be adapted for use with viral vectored- and plasmid DNA-based polynucleotide gene transfer. See Fang et al. (2007) “An antibody delivery system for regulated expression of therapeutic levels of monoclonal antibodies in vivo” Mol Ther. 5(6): 1153-9; and Perez et al. (2004) “Regulatable systemic production of monoclonal antibodies by in vivo muscle electroporation” Genet Vaccines Ther. 2(1):2.
  • viral post-transcriptional regulatory elements may be used; these cis-acting elements are required for nuclear export of intronless viral RNA (Huang and Yen, 1994 J Virol. 68(5):3193-9; and 1995 Mol Cell Biol. 15(7):3864-9).
  • HPRE Hepatitis B Vims PRE, 533 bps
  • WPRE Wideodchuck Hepatitis Vims PRE, 600 bps
  • WPRE was found to increase CMV promoter driven transgene expression, as well as increase PPE, PDGF and NSE promoter-driven transgene expression. Another effect of the WPRE can be to protect polynucleotides transgenes from silencing (Patema et al., 2000 Gene Ther. 7(15): 1304-11; Xia et al., 2007 Stem Cells Dev. 2007 Feb; 16(1): 167-76).
  • polyadenylation of a transcribed polynucleotide transcript can also be important for nuclear export, translation, and mRNA stability. Therefore, in some embodiments, the polynucleotide will include a polyadenylation signal sequence.
  • polyadenylation signal sequences include SV40 late or bovine growth hormone polyA (bGHpA) signal sequences, as well as minimal synthetic polyA (SPA) signal (Levitt et al., 1989 Genes Dev. 3(7): 1019-25; Yew et al., 1997 Hum Gene Ther. 1997 8(5):575- 84).
  • the efficiency of polyadenylation is increased by the SV40 late polyA signal upstream enhancer (USE) placed upstream of other polyA signals (Schek et al., 1992 Mol Cell Biol. 12(12):5386-93).
  • the polynucleotide will include an SV40 late + 2xUSE polyA signal.
  • the polynucleotide may be desirable for the polynucleotide to include at least one regulatory enhancers, e.g., in addition to any promoter sequences.
  • the CMV enhancer is upstream of the CMV promoter at -598 to -68 (Boshart et al., 1985 Cell. 41(2):521-30) (-600 bps) and contains transcription binding sites.
  • a CMV enhancer can be included in the construct to increase tissue- specific promoter-driven transgene expression, such as using the ANF (atrial natriuretic factor) promoter, the CC10 (club cell 10) promoter, SP-C (surfactant protein C) promoter, or the PDGF-b (platelet-derived growth factor-b) promoter (merely as examples).
  • ANF atrial natriuretic factor
  • CC10 club cell 10 promoter
  • SP-C surfactant protein C
  • PDGF-b platelet-derived growth factor-b
  • transgene expression using the CMV enhancer with a muscle-specific promoter can increase expression levels of the protein encoded by the transgene, so would be particularly useful in the current disclosure for expressing bispecific fusion proteins from polynucleotides introduced into muscle cells of a patient.
  • the subject polynucleotides may also include at least one intronic sequences.
  • the presence of an intron or intervening sequence in mRNA was first described, in vitro, to be important for mRNA processing and increased transgene expression (Huang and Gorman, 1990 Mol Cell Biol. 10(4): 1805-10; Niwa et al., 1990 Genes Dev. 4(9): 1552-9).
  • the intron(s) can be placed within the coding sequence for the bispecific fusion protein and/or can be placed between the promoter and transgene.
  • a variety of introns (Table 3) placed between the promoter and transgene were compared, in mice using AAV2, for liver transgene expression (Wu et al., 2008).
  • the MVM (minute virus of mice) intron increased transgene expression more than any other intron tested and more than 80-fold over no intron (Wu et al., 2008).
  • transgene expression was less under a CaMPKII promoter with a chimeric intron (human b-globin donor and immunoglobulin heavy chain acceptor) between the transgene and polyA signal compared to a WPRE (Choi et al., 2014).
  • a chimeric intron human b-globin donor and immunoglobulin heavy chain acceptor
  • the subject polynucleotides may also include at least one origins of replication, minichromosome maintenance elements (MME) and/or nuclear localization elements.
  • MME minichromosome maintenance elements
  • Episomal vectors of the disclosure comprise a portion of a vims genomic DNA that encodes an origin of replication (ori), which is required for such vectors to be self- replicating and, thus, to persist in a host cell over several generations.
  • an episomal vector of the disclosure also may contain at least one gene encoding viral proteins that are required for replication, i.e., replicator protein (s).
  • the replicator protein(s) which help initiate replication may be expressed in trans on another DNA molecule, such as on another vector or on the host genomic DNA, in the host cell containing a self-replicating episomal expression vector of this disclosure.
  • Preferred self-replicating episomal LCR-containing expression vectors of the disclosure do not contain viral sequences that are not required for long term stable maintenance in a eukaryotic host cell such as regions of a viral genome DNA encoding core or capsid proteins that would produce infectious viral particles or viral oncogenic sequences which may be present in the full-length viral genomic DNA molecule.
  • stable maintenance refers to the ability of a self-replicating episomal expression vector of this disclosure to persist or be maintained in non-dividing cells or in progeny cells of dividing cells in the absence of continuous selection without a significant loss (e.g ., >50%) in copy number of the vector for two, three, four, or five or more generations. In some embodiments, the vectors will be maintained over 10-15 or more cell generations.
  • transient or “short term” persistence of a plasmid in a host cell refers to the inability of a vector to replicate and segregate in a host cell in a stable manner; that is, the vector will be lost after one or two generations, or will undergo a loss of >51% of its copy number between successive generations.
  • the self-replicating function may alternatively be provided by at least one mammalian sequences such as described by Wohlge uth et al., 1996, Gene Therapy 3:503; Vos et al., 1995, Jour. Cell. Biol., Supp. 21A, 433; and Sun et al., 1994, Nature Genetics 8:33, optionally in combination with at least one sequence which may be required for nuclear retention.
  • mammalian, especially human sequences for providing the self- replicating function is that no extraneous activation factors are required which could have toxic or oncogenic properties.
  • the disclosure is not limited to any one origin of replication or any one episomal vector but encompasses the combination of the tissue-restricted control of an LCR in an episomal vector. See also WO1998007876 “Self-replicating episomal expression vectors conferring tissue- specific gene expression” and US Patent 7790446 “Vectors, cell lines and their use in obtaining extended episomal maintenance replication of hybrid plasmids and expression of gene products”
  • Epstein-Barr Virus-Based Self-Replicating Episomal Expression Vectors The latent origin oriP from Epstein-Barr Virus (EBV) is described in Yates et. al., Proc. Natl. Acad. Sci. USA 81:3806-3810 (1984); Yates et al., Nature 313:812-815 (1985); Krysan et al., Mol. Cell. Biol. 9:1026-1033 (1989); James et al. Gene 86: 233-239 (1990), Peterson and Legerski, Gene 107:279-284 (1991); and Pan et al., Som. Cell Molec. Genet. 18:163-177 (1992)).
  • EBV Epstein-Barr Virus
  • An EBV- based episomal vector useful according to the disclosure can contain the oriP region of EBV which is carried on a 2.61 kb fragment of EBV and the EBNA-1 gene which is carried on a 2.18 kb fragment of EBV.
  • the EBNA-1 protein which is the only viral gene product required to support in trans episomal replication of vectors containing oriP, may be provided on the same episomal expression vector containing oriP. It is also understood, that as with any protein such as EBNA-1 known to be required to support replication of viral plasmid in trans, the gene also may be expressed on another DNA molecule, such as a different DNA vector.
  • the episomal expression vectors of the disclosure also may be based on replication functions of the papilloma family of virus, including but not limited to Bovine Papilloma Virus (BPV) and Human Papilloma Viruses (HPVs).
  • BPV and HPVs persist as stably maintained plasmids in mammalian cells.
  • -S trans-acting factors encoded by BPV and HPVs, namely El and E2 have also been identified which are necessary and sufficient for mediate replication in many cell types via minimal origin of replication (Ustav et al., EMBO J.
  • An episomal vector useful according to the disclosure is the BPV-I vector system described in Piirsoo et al., EMBO J., 15:1 (1996) and in WO 94/12629.
  • the BPV-1 vector system described in Piirsoo et al. comprises a plasmid harboring the BPV-1 origin of replication (minimal origin plus extrachro osomal maintenance element) and optionally the El and E2 genes.
  • the BPV-1 El and E2 genes are required for stable maintenance of a BPV episomal vector. These factors ensure that the plasmid is replicated to a stable copy number of up to thirty copies per cell independent of cell cycle status.
  • the gene construct therefore persists stably in both dividing and non-dividing cells. This allows the maintenance of the gene construct in cells such as hemopoietic stem cells and more committed precursor cells.
  • the BPV origin of replication has been located at the 3’ end of the upstream regulatory region within a 60 base pair (bp) DNA fragment (nucleotides (nt) 7914 - 7927) which includes binding sites for the El and E2 replication factors.
  • the minimal origin of replication of HPV has also been characterized and located in the URR fragment (nt 7022- 7927) of HPV (see, for example, Chiang et al., Proc. Natl. Acad. Sci. USA 89:5799-5803 (1992)).
  • El refers to the protein encoded by nucleotides (nt) 849-2663 of BPV subtype 1 or by nt 832- 2779 of HPV of subtype 11, to equivalent El proteins of other papilloma viruses, or to functional fragments or mutants of a papilloma virus El protein, i.e., fragments or mutants of El which possess the replicating properties of El.
  • E2H refers to the protein encoded by nt 2594-3837 of BPV subtype 1 or by nt 2723-3823 of HPV subtype 11, to equivalent E2 proteins of other papilloma viruses, or to functional fragments or mutants of a papilloma vims E2 protein, i.e., fragments or mutants of E2 which possess the replicating properties of E2.
  • Minichromosomal maintenance element is the protein encoded by nt 2594-3837 of BPV subtype 1 or by nt 2723-3823 of HPV subtype 11, to equivalent E2 proteins of other papilloma viruses, or to functional fragments or mutants of a papilloma vims E2 protein, i.e., fragments or mutants of E2 which possess the replicating properties of E2.
  • MME refers to the extrachromosomal maintenance element of the papilloma viral genome to which viral or human proteins essential for papilloma viral replication bind, which region is essential for stable episomal maintenance of the papilloma viral MO in a host cell, as described in Piirsoo et al. (supra).
  • the MME is a sequence containing multiple binding sites for the transcriptional activator E2.
  • the MME in BPV is herein defined as the region of BPV located within the upstream regulatory region which includes a minimum of about six sequential E2 binding sites, and which gives optimum stable maintenance with about ten sequential E2 binding sites.
  • E2 binding site 9 is an example sequence for this site, as described hereinbelow, wherein the sequential sites are separated by a spacer of about 4-10 nucleotides, and optimally 6 nucleotides.
  • El and E2 can be provided to the plasmid either in cis or in trans, also as described in WO 94/12629 and in Piirsoo et al. (supra).
  • E2 binding site refers to the minimum sequence of papillomavirus double-stranded DNA to which the E2 protein binds.
  • An E2 binding site may include the sequence 5’ACCGTTGCCGGT3' (SEQ ID NO: 172), which is high affinity E2 binding site 9 of the BPV- 1 URR; alternatively, an E2 binding site may include permutations of binding site 9, which permutations are found within the URR, and fall within the generic E2 binding sequence 5' ACCN6GGT 3'.
  • One or more transcriptional activator E2 binding sites are, in most papillomaviruses, located in the upstream regulatory region, as in BPV and HPV.
  • a vector which also is useful according to the disclosure may include a region of BPV between 6959 - 7945/1 - 470 on the BPV genetic map (as described in WO 94/12629) , which region includes an origin of replication, a first promoter operatively associated with a gene of interest, the BPV El gene operatively associated with a second promoter to drive transcription of the El gene; and the BPV E2 gene operatively associated with a third promoter to drive transcription of the E2 gene.
  • El and E2 from BPV will replicate vectors containing the BPV origin or the origin of many HPV subtypes (Chiang et ah, supra). El and E2 from HPV will replicate vectors via the BPV origin and via the origin of many HPV subtypes (Chiang et al., supra). As with all vectors of the disclosure, the BPV-based episomal expression vectors of the disclosure must persist through 2-5 or more divisions of the host cell.
  • the vectors of the disclosure also may be derived from a human papovavims BK genomic DNA molecule.
  • the BK viral genome can be digested with restriction enzymes EcoRI and BamHI to produce a 5 kilobase (kb) fragment that contains the BK viral origin of replication sequences that can confer stable maintenance on vectors (see, for example, De Benedetti and Rhoads, Nucleic Acids Res . 19:1925 (1991), as can a 3.2 kb fragment of the BK vims (Cooper and Miron,
  • the polynucleotides of the present disclosure can be provided as circular or linear polynucleotides.
  • the circular and linear polynucleotides are capable of directing expression of the bispecific fusion protein coding sequence in an appropriate subject cell.
  • the at least one polynucleotide systems for expressing a bispecific fusion protein may be chimeric, meaning that at least one of its components is heterologous with respect to at least one of its other components.
  • viral gene therapy system examples include plasmid, adenovirus, adeno-associated virus (AAV), retrovirus, lentivirus, herpes simplex vims, vaccinia vims, poxvims, reovims, measles vims, Semliki Forest vims, and the like.
  • Preferred viral vectors are based on non-cytopathic eukaryotic viruses in which non- essential genes have been replaced with the polynucleotide construct carrying the polynucleotide sequences encoding the epitopes and targeting sequences of interest.
  • encoded AFFIMER® polypeptides can be delivered in vivo using adenoviruses and adeno-associated (AAV) viruses, which are double- stranded DNA viruses that have already been approved for human use in gene therapy.
  • AAV adeno-associated viruses
  • AdVs are non-enveloped, double- stranded DNA viruses that neither integrate in the host genome nor replicate during cell division.
  • AdV-mediated antibody gene transfer has shown therapeutic efficacy in a variety of different disease models advancing towards the clinic.
  • Systemic mAb expression has mostly been pursued, via s.c. and especially i.v. and intramuscular AdV injection. See Wold et al. (2013) “Adenovirus vectors for gene therapy, vaccination and cancer gene therapy” Curr Gene Ther. 13(6):421— 33; and Deal et al.
  • AdVs as oncolytic vectors
  • the use of AdVs as oncolytic vectors is a popular approach particularly for generation of encoded antibodies at the site of tumors.
  • Foreign genes delivered by current adenoviral gene delivery system are episomal, and therefore, have low genotoxicity to host cells. Therefore, gene therapy using adenoviral gene delivery systems may be considerably safe.
  • the present disclosure specifically contemplates the delivery of bispecific fusion proteins by expression of polynucleotides delivered in the form of an adenoviral vector and delivery system.
  • Adenovirus has been usually employed as a gene delivery vector because of its mid-sized genome, ease of manipulation, high titer, wide target-cell range and high infectivity. Both ends of the viral genome contains 100-200 bp ITRs (inverted terminal repeats), which are cis elements necessary for viral DNA replication and packaging.
  • the El region (E1A and E1B) of genome encodes proteins responsible for the regulation of transcription of the viral genome and a few cellular genes.
  • the E2 region (E2A and E2B) encodes proteins responsible for viral DNA replication.
  • the replication incompetent adenovirus having the deleted El region is usually used and represent one example of AdV for generating the polynucleotides of the present disclosure.
  • the deleted E3 region in adenoviral vectors may provide an insertion site for transgenes (Thimmappaya, B. et al., Cell, 31:543-551(1982); and Riordan, J. R. et al., Science, 245:1066-1073(1989)).
  • an “adenovirus expression vector” is meant to include those constructs containing adenovirus sequences sufficient to (a) support packaging of the construct and (b) to express a polynucleotide that encodes a polypeptide including a bispecific fusion protein such as a PD-L1 binding AFFIMER® (the polynucleotide sequence).
  • the sequence for polynucleotide may be inserted into the DA promoter region.
  • the recombinant adenovirus comprises deleted E1B and E3 region and the nucleotide sequence for an encoded AFFIMER® polypeptide is inserted into the deleted E1B and E3 region.
  • AAV Adeno-Associated Virus Vectors
  • AAVs are non-enveloped small, single- stranded DNA viruses capable of infecting both dividing and non-dividing cells. Similar to AdV, AAV- based vectors remain in an episomal state in the nucleus and display a limited risk of integration. In contrast to the generally limited durability of AdV-mediated gene transfer, transgene expression can persist for years following intramuscular recombinant AAV (rAAV) vector delivery.
  • Alipogene tiparvovec (GlyberaTM), an rAAV encoding the human lipoprotein lipase gene, was approved in 2012 as the first gene therapy product in Europe. Since then, various rAAV- based gene therapy products are currently under clinical evaluation. In the context of antibody gene transfer, a variety of reports have demonstrated in vivo production of an anti-human immune deficiency virus (HIV) mAb in mice following intramuscular injection of the mAb- encoding rAAV. The rAAV vector’s potential for combination therapy has also been demonstrated, i.e. by expressing two mAbs. Similar to AdV, intramuscular and i.v. rAAV administration have been most often pursued.
  • HIV anti-human immune deficiency virus
  • AAV Gene transfer virus
  • these gene transfer viruses are capable of infecting non-dividing cells and various types of cells, making them useful in constructing the polynucleotide delivery system of this disclosure.
  • the detailed descriptions for use and preparation of examples of AAV vectors are found in, for example, U.S. Pat. Nos. 5,139,941 and 4,797,368, as well as LaFace et al, Viology, 162:483486 (1988), Zhou et ah, Exp. Hematol.
  • AAV is a good choice of delivery vehicles due to its safety, i.e., genetically engineered (recombinant) does not integrate into the host genome. Likewise, AAV is not pathogenic and not associated with any disease. The removal of viral coding sequences minimizes immune reactions to viral gene expression, and therefore, recombinant AAV does not evoke an inflammatory response.
  • a recombinant AAV vims is made by co-transfecting a plasmid containing the gene of interest (i.e., the coding sequence for a bispecific fusion protein) flanked by the two AAV terminal repeats (McLaughlin et ah, J. Virol., 62:1963-1973(1988); Samulski et ah, J. Virol., 63:3822-3828(1989)) and an expression plasmid containing the wild type AAV coding sequences without the terminal repeats (McCarty et ah, J. Virol., 65:2936-2945(1991)).
  • the gene of interest i.e., the coding sequence for a bispecific fusion protein flanked by the two AAV terminal repeats
  • Samulski et ah J. Virol., 63:3822-3828(1989)
  • viral vectors containing polynucleotide are assembled from polynucleotides encoding the AFFIMER® polypeptide containing polypeptide, suitable regulatory elements and elements necessary for expression of the encoded AFFIMER® polypeptide which mediate cell transduction.
  • adeno-associated viral (AAV) vectors are employed.
  • the AAV vector is an AAV1, AAV6, or AAV8.
  • the AAV expression vector which harbors the polynucleotide sequence bounded by AAV ITRs can be constructed by directly inserting the selected sequence(s) into an AAV genome which has had the major AAV open reading frames (“ORFs”) excised therefrom.
  • ORFs major AAV open reading frames
  • expression control sequences typically include a promoter, an enhancer, such as one derived from an immunoglobulin gene, SV40, cytomegalovirus, etc. (see above), and a polyadenylation sequence which may include splice donor and acceptor sites.
  • the polyadenylation sequence generally is inserted following the transgene sequences and before the 3' ITR sequence.
  • Non-cytopathic viruses useful in the context of delivery of polynucleotides include retroviruses, the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication- deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
  • Standard protocols for producing replication-deficient retroviruses including the steps of incorporation of exogenous genetic material into a plasmid, transfection of a packaging cell lined with plasmid, production of recombinant retroviruses by the packaging cell line, collection of viral particles from tissue culture media, and infection of the target cells with viral particles) are known to those of skill in the art.
  • the bispecific fusion protein coding sequence is inserted into the viral genome in the place of certain viral sequences to produce a replication- defective virus.
  • a packaging cell line containing the gag, pol and env genes but without the LTR (long terminal repeat) and psi ( ⁇ ) components is constructed (Mann et ah, Cell, 33:153-159(1983)).
  • RNA transcript of the recombinant plasmid When a recombinant plasmid containing the cytokine gene, LTR and psi is introduced into this cell line, the psi sequence allows the RNA transcript of the recombinant plasmid to be packaged into viral particles, which are then secreted into the culture media (Nicolas and Rubinstein "Retroviral vectors," In: Vectors: A survey of molecular cloning vectors and their uses, Rodriguez and Denhardt (eds.), Stoneham: Butterworth, 494-513(1988)). The media containing the recombinant retroviruses is then collected, optionally concentrated and used for gene delivery system.
  • the retrovirus is a "gammaretroviruses", which refers to a genus of the retroviridae family.
  • gammaretroviruses include mouse stem cell virus, murine leukemia virus, feline leukemia virus, feline sarcoma virus, and avian reticuloendotheliosis viruses.
  • the retroviral vector for use in the present disclosure is a lentiviral vector, which refers to a genus of retroviruses that are capable of infecting dividing and non dividing cells and typically produce high viral titers.
  • lentiviruses include HIV (human immunodeficiency virus: including HIV type 1, and HIV type 2); equine infectious anemia virus; feline immunodeficiency vims (FIV); bovine immune deficiency vims (BIV); and simian immunodeficiency vims (SIV).
  • retroviral vectors that can be used for the delivery and expression of polynucleotide include those based upon murine leukemia vims (MuLV), gibbon ape leukemia vims (GaLV) and combinations thereof (see, e.g., Buchscher et ah, J. Virol. 66:2731-2739, 1992; Johann et ah, J. Virol. 66: 1635-1640, 1992; Sommerfelt et ah, Virol. 176:58-59, 1990; Wilson et ah, J. Virol. 63:2374-2378, 1989; Miller et ah, J. Virol. 65:2220- 2224, 1991; and PCT/US94/05700).
  • MuLV murine leukemia vims
  • GaLV gibbon ape leukemia vims
  • FVes Foamy vimses
  • This complete apathogenicity qualifies FV vectors as ideal gene transfer vehicles for genetic therapies in humans and clearly distinguishes FV vectors as gene delivery system from HIV-derived and also gammaretrovims-derived vectors.
  • retroviral vectors for use herein are described, for example, in U.S. Pat. Nos. 5,399,346 and 5,252,479; and in WIPO publications WO 92/07573, WO 90/06997, WO 89/05345, WO 92/05266 and WO 92/14829, which provide a description of methods for efficiently introducing polynucleotides into human cells using such retroviral vectors.
  • retroviral vectors include, for example, mouse mammary tumor vims vectors (e.g., Shackleford et ah, Proc. Natl. Acad. Sci. U.S. A. 85:9655-9659, 1998), lentivimses, and the like.
  • Additional retroviral viral delivery systems that can be readily adapted for delivery of a transgene encoding a PD-L1 bispecific fusion protein include, merely to illustrate Published PCT Applications WO/2010/045002, WO/2010/148203, WO/2011/126864, WO/2012/058673, WO/2014/066700, WO/2015/021077, WO/2015/148683, WO/2017/040815 - the specifications and FIGS of each of which are incorporated by reference herein.
  • a retroviral vector contains all of the cis-acting sequences necessary for the packaging and integration of the viral genome, i.e., (a) a long terminal repeat (LTR), or portions thereof, at each end of the vector; (b) primer binding sites for negative and positive strand DNA synthesis; and (c) a packaging signal, necessary for the incorporation of genomic RNA into virions. More detail regarding retroviral vectors can be found in Boesen, et ah, 1994, Biotherapy 6:291-302; Clowes, et ai, 1994, J. Clin. Invest.
  • LTR long terminal repeat
  • the retrovirus is a recombinant replication competent retrovirus comprising: a polynucleotide sequence encoding a retroviral GAG protein; a polynucleotide sequence encoding a retroviral POL protein; a polynucleotide sequence encoding a retroviral envelope; an oncoretroviral polynucleotide sequence comprising Long-Terminal Repeat (LTR) sequences at the 5' and 3' end of the oncoretroviral polynucleotide sequence; a cassette comprising an internal ribosome entry site (IRES) operably linked to a coding sequence for a bispecific fusion protein, such as for a PD-L1 bispecific fusion protein, wherein the cassette is positioned 5' to the U3 region of the 3' LTR and 3' to the sequence encoding the retroviral envelope; and cis-acting sequences for reverse transcription, packaging and integration in a target cell.
  • a ribosome entry site IVS
  • the retrovirus is a recombinant replication competent retrovirus comprising: a retroviral GAG protein; a retroviral POL protein; a retroviral envelope; a retroviral polynucleotide comprising Long-Terminal Repeat (LTR) sequences at the 3' end of the retroviral polynucleotide sequence, a promoter sequence at the 5' end of the retroviral polynucleotide, the promoter being suitable for expression in a mammalian cell, a gag polynucleotide domain, a pol polynucleotide domain and an env polynucleotide domain; a cassette comprising polynucleotide sequence, wherein the cassette is positioned 5' to the 3' LTR and is operably linked and 3' to the env polynucleotide domain encoding the retroviral envelope; and cis-acting sequences necessary for reverse transcription, packaging and integration in a target cell.
  • LTR Long-Terminal
  • the envelope is chosen from one of amphotropic, polytropic, xenotropic, 10A1, GALV, Baboon endogenous vims, RD114, rhabdovirus, alphavirus, measles or influenza vims envelopes.
  • the retroviral polynucleotide sequence is engineered from a vims selected from the group consisting of murine leukemia vims (MLV) , Moloney murine leukemia vims (MoMLV) , Feline leukemia vims (FeLV) , Baboon endogenous retrovims (BEV) , porcine endogenous vims (PERV) , the cat derived retrovims RD114, squirrel monkey retrovims, Xenotropic murine leukemia vims-related vims (XMRV) , avian reticuloendotheliosis vims (REV) , or Gibbon ape leukemia vims (GALV).
  • MMV murine leukemia vims
  • MoMLV Moloney murine leukemia vims
  • FeLV Feline leukemia vims
  • BEV Baboon endogenous retrovims
  • PERV porcine endogenous vi
  • retrovims is a gammaretro vim s .
  • a second cassette comprising a coding sequence for a second therapeutic protein, such as another checkpoint inhibitor polypeptide, a co-stimulatory polypeptide and/or a immuno stimulatory cytokine (merely as examples), e.g., downstream of the cassette.
  • the second cassette can include an internal ribosome entry site (IRES) or a minipromoter or a polIII promoter operably linked to the coding sequence for the second therapeutic protein.
  • the recombinant replication competent retrovirus is a nonlytic, amphotropic retroviral replicating vector which, preferably, selectively infects and replicates in the cells of the tumor microenvironment.
  • oncolytic viruses In the context of vectored intratumoral polynucleotide gene transfer, oncolytic viruses have a distinct advantage, as they can specifically target tumor cells, boost therapeutic bispecific fusion protein expression, and amplify antitumor therapeutic responses.
  • Oncolytic viruses which overlap with certain viral systems described above, promote anti-tumor responses through selective tumor cell killing and induction of systemic anti-tumor immunity. The mechanisms of action are not fully elucidated but are likely to depend on viral replication within transformed cells, induction of primary cell death, interaction with tumor cell anti- viral elements and initiation of innate and adaptive anti-tumor immunity. Reviewed in Kaufman et al. 2015 “Oncolytic viruses: a new class of immunotherapy drugs” Nat Rev Drug Discov. 14(9):642-62.
  • AdV poxviruses
  • coxsackieviruses poxsackieviruses
  • poliovirus measles vims
  • Newcastle disease virus reovirus
  • others have entered into early-phase clinical trials.
  • T-VEC talimogene laherparepvec
  • GM-CSF granulocyte-macrophage colony- stimulating factor
  • oncolytic vimses make them an appealing platform for polynucleotide gene transfer of the present disclosure, as transgene products can be amplified along with viral replication, thereby maximizing therapeutic effect.
  • intratumoral delivery of the polynucleotide and concomitant local expression of the bispecific fusion protein can create a better therapeutic index where dose- limiting toxicities might otherwise prevent reaching the effective intratumoral concentration for efficacy when the bispecific fusion protein is delivered (or expressed) systemically.
  • the immunomodulatory nature of these AFFIMER® polypeptides are very relevant to the use of oncolytic viruses. Indeed, for oncolytic virus therapy, it is desirable to override immune checkpoint inhibitor networks and thereby create a pro-inflammatory environment within the cancer. Numerous clinical trials are currently underway to evaluate the combination of oncolytic viruses and conventional immunomodulatory mAb administration. Kaufman et al. 2015 “Oncolytic viruses: a new class of immunotherapy drugs” Nat Rev Drug Discov. 14(9):642-62; and Lichty et al. 2014 “Going viral with cancer immunotherapy” Nat Rev Cancer. 14(8):559-67.
  • viral vectors may be employed as a gene delivery system in the present disclosure.
  • Vectors derived from viruses such as vaccinia virus (Puhlmann M. et al., Human Gene Therapy, 10:649-657(1999); Ridgeway, "Mammalian expression vectors," In: Vectors: A survey of molecular cloning vectors and their uses. Rodriguez and Denhardt, eds. Stoneham: Butterworth, 467-492(1988); Baichwal and Sugden, "Vectors for gene transfer derived from animal DNA viruses: Transient and stable expression of transferred genes," In: Kucherlapati R, ed. Gene transfer.
  • poxvirus GCE, NJL, Krupa M, Esteban M., The poxvirus vectors MVA and NYVAC as gene delivery systems for vaccination against infectious diseases and cancer Curr Gene Ther 8(2):97- 120(2008)
  • reovirus measles virus, Semliki Forest virus, and polioviruses
  • measles virus may be used in the present delivery systems for transferring the gene of interest into cells. They offer several attractive features for various mammalian cells. Also included are hepatitis B viruses.
  • pDNA After entry in the nucleus, pDNA remains in a non-replicating non integrating episomal state and is lost during the breakdown of the nuclear envelope at mitosis.
  • pDNA has no defined restrictions regarding the size of the transgene compared to viral vectors, and its modular nature allows for straightforward molecular cloning, making them easy to manipulate and design for therapeutic use. Hardee et al. 2017 “Advances in non-viral DNA vectors for gene therapy” Genes. 8(2):65. Plasmids are used in about 17% of the ongoing or completed gene therapy clinical trials and showed to be well-tolerated and safe. The method of DNA administration can greatly impact transgene expression.
  • Electroporation presents the propagation of electrical fields within tissues, which induces a transient increase in cell membrane permeability.
  • Electrotransfer of DNA is a multistep process, involving (i) electrophoretic migration of DNA towards the plasma membrane, (ii) DNA accumulation and interaction with the plasma membrane, and (iii) intracellular trafficking of the DNA to the nucleus, after which gene expression can commence.
  • Heller LC 2015 “Gene electrotransfer clinical trials” Adv Genet. 89:235-62.
  • Intramuscular, intratumoral and intradermal administration have been evaluated in clinical trials and are also suitable target tissues for electroporation of polynucleotides.
  • Hydrodynamic-based transfection utilizes the i.v. injection of high volumes of pDNA, driving DNA molecules out of the blood circulation and into tissue.
  • Other potentially less invasive physical delivery methods include sonoporation and magnetofection.
  • DNA uptake can also be improved by complexing the molecules with chemical delivery vehicles (e.g . cationic lipids or polymers and lipid nanoparticles). Such techniques can also be applied to in vivo DNA- mediated polynucleotide gene transfer.
  • polynucleotide transgene expression can be improved by modifying the make-up of pDNA constructs. See, for example, Hardee et al. 2017 “Advances in non-viral DNA vectors for gene therapy” Genes 8(2):65; and Simcikova et al. 2015 “Towards effective non-viral gene delivery vector” Biotechnol Genet Eng Rev. 31(1- 2):82-107.
  • Conventional pDNA consists of a transcription unit and bacterial backbone.
  • the transcription unit carries the polynucleotide sequence along with regulatory elements.
  • the bacterial backbone includes elements like an antibiotic resistance gene, an origin of replication, unmethylated CpG motifs, and potentially cryptic expression signals.
  • mcDNA minicircle DNA
  • Minicircles are plasmid molecules devoid of bacterial sequences, generated via a process of recombination, restriction and/or purification. Simcikova et al. 2015 supra. Elimination of the bacterial backbone has shown higher transfection efficiency and prolonged transgene expression in a variety of tissues.
  • LEC linear polynucleotide, or linear expression cassette
  • the LEC may be any linear DNA devoid of any phosphate backbone.
  • the LEC may contain a promoter, an intron, a stop codon, and/or a polyadenylation signal.
  • the expression of the polynucleotide coding sequence may be controlled by the promoter.
  • the subject polynucleotides are delivered as plasmid vectors.
  • Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et ak, 1989, cited above.
  • plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they reduced safety concerns relative to other vectors.
  • These plasmids however, having a promoter compatible with the host cell, can express a peptide epitope encoded by polynucleotide within the plasmid.
  • Other plasmids are well known to those of ordinary skill in the art.
  • Plasmids may be custom designed using restriction enzymes and ligation reactions to remove and add specific fragments of DNA. Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
  • the DNA plasmid can be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally. It may also be administered into the epidermis or a mucosal surface using a gene-gun.
  • the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
  • Kitaguchi et al. 2005 “Immune deficiency enhances expression of recombinant human antibody in mice after nonviral in vivo gene transfer” Int J Mol Med 16(4):683— 8; and Yamazaki et al. 2011 “Passive immune-prophylaxis against influenza virus infection by the expression of neutralizing anti-hemagglutinin monoclonal antibodies from plasmids” Jpn J Infect Dis.
  • pDNA electrotransfer can be enhanced by pre-treating the muscle with hyaluronidase, an enzyme that transiently breaks down hyaluronic acid, decreasing the viscosity of the extracellular matrix and facilitating DNA diffusion.
  • hyaluronidase an enzyme that transiently breaks down hyaluronic acid, decreasing the viscosity of the extracellular matrix and facilitating DNA diffusion.
  • the purpose of the plasmid is the efficient delivery of polynucleotide sequences to and expression of therapeutic bispecific fusion proteins in a cell or tissue.
  • the purpose of the plasmid may be to achieve high copy number, avoid potential causes of plasmid instability and provide a means for plasmid selection.
  • the polynucleotide cassette contains the necessary elements for expression of the polynucleotide within the cassette. Expression includes the efficient transcription of an inserted gene, polynucleotide sequence, or polynucleotide cassette with the plasmid.
  • a plasmid for expression of polynucleotide which includes an expression cassette comprising the coding sequence for the bispecific fusion protein; also referred to as a transcription unit.
  • the transcriptional unit will express the bispecific fusion protein and anything else encoded in the construct.
  • the transcription unit includes a transcriptional control sequence, which is transcriptionally linked with a cellular immune response element coding sequence.
  • Transcriptional control sequence may include promoter/enhancer sequences such as cytomegalovirus (CMV) promoter/enhancer sequences, such as described above.
  • promoter sequences suitable for expression in mammalian cells including human patient cells, are known and can similarly be used in the constructs disclosed herein.
  • the level of expression of the bispecific fusion protein will depend on the associated promoter and the presence and activation of an associated enhancer element.
  • the polynucleotide sequence (encoding the desired bispecific fusion protein) can be cloned into an expression plasmid which contains the regulatory elements for transcription, translation, RNA stability and replication (i.e., including a transcriptional control sequence).
  • expression plasmids are well known in the art and one of ordinary skill would be capable of designing an appropriate expression construct for producing a recombinant bispecific fusion protein in vivo. Minicircle
  • Minicircle (mcDNA) -based antibody gene transfer can also be adapted for delivery of polynucleotide to tissues in vivo.
  • plasmid DNA used for non-viral gene delivery can cause unacceptable inflammatory responses. Where this happens, immunotoxic responses are largely due to the presence of unmethylated CpG motifs and their associated stimulatory sequences on plasmids following bacterial propagation of plasmid DNA. Simple methylation of DNA in vitro may be enough to reduce an inflammatory response but can result in reduced gene expression. The removal of CpG islands by cloning out, or elimination of non-essential sequences has been a successful technique for reducing inflammatory responses. Yew et al. 2000 “Reduced inflammatory response to plasmid DNA vectors by elimination and inhibition of immuno stimulatory CpG motifs” Mol Ther 1(3), 255-62.
  • the "parent" plasmid is recombined into a "minicircle" which generally comprises the gene to be delivered (in this case, the polynucleotide coding sequence) and suitable control regions for its expression, and a miniplasmid which generally comprises the remainder of the parent plasmid.
  • minicircle production uses bacterial phage lambda (l) integrase mediated recombination to produce minicircle DNA. See, for example, Darquet, et al. 1997 Gene Ther 4(12): 1341-9; Darquet et al. 1999 Gene Ther 6(2): 209-18; and Kreiss, et al. 1998 Appl Micbiol Biotechnol 49(5):560-7).
  • minicircle DNA pertains to small (2-4 kb) circular plasmid derivatives that have been freed from all prokaryotic vector parts. Since minicircle DNA vectors contain no bacterial DNA sequences, they are less likely to be perceived as foreign and destroyed. As a result, these vectors can be expressed for longer periods of time compared to certain conventional plasmids. The smaller size of minicircles also extends their cloning capacity and facilitates their delivery into cells. Kits for producing minicircle DNA are known in the art and are commercially available (System Biosciences, Inc., Palo Alto, Calif.).
  • Minicircle DNA vectors achieve sustained expression reflected by active chromatin and transcriptional level.
  • a minicircle DNA vector may be produced as follows.
  • An expression cassette which comprises the polynucleotide coding sequence along with regulatory elements for its expression, is flanked by attachment sites for a recombinase.
  • a sequence encoding the recombinase is located outside of the expression cassette and includes elements for inducible expression (such as, for example, an inducible promoter).
  • the vector DNA is recombined, resulting in two distinct circular DNA molecules.
  • One of the circular DNA molecules is relatively small, forming a minicircle that comprises the expression cassette for the polynucleotide; this minicircle DNA vector is devoid of any bacterial DNA sequences.
  • the second circular DNA sequence contains the remaining vector sequence, including the bacterial sequences and the sequence encoding the recombinase.
  • the minicircle DNA containing the polynucleotide sequence can then be separately isolated and purified.
  • a minicircle DNA vector may be produced using plasmids similar to pBAD.c
  • recombinases examples include, but are not limited to, Streptomyces bacteriophage f31 integrase, Cre recombinase, and the l integrase/DNA topoisomerase IV complex. Each of these recombinases catalyzes recombination between distinct sites.
  • f31 integrase catalyzes recombination between corresponding attP and attB sites
  • Cre recombinase catalyzes recombination between loxP sites
  • the l integrase/DNA topoisomerase IV complex catalyzes recombination between bacteriophage l attP and attB sites.
  • the recombinase mediates an irreversible reaction to yield a unique population of circular products and thus high yields.
  • the recombinase mediates a reversible reaction to yield a mixture of circular products and thus lower yields.
  • the reversible reaction by Cre recombinase can be manipulated by employing mutant loxP71 and loxP66 sites, which recombine with high efficiency to yield a functionally impaired P71/66 site on the minicircle molecule and a wild-type loxP site on the minicircle molecule, thereby shifting the equilibrium towards the production of the minicircle DNA product.
  • Published US Application 20170342424 also describes a system making use of a parent plasmid which is exposed to an enzyme which causes recombination at recombination sites, thereby forming a (i) minicircle including the polynucleotide sequence and (ii) a miniplasmid comprising the remainder of the parent plasmid.
  • One recombination site is modified at the 5' end such that its reaction with the enzyme is less efficient than the wild type site, and the other recombination site is modified at the 3' end such that its reaction with the enzyme is less efficient than the wild type site, and the other recombination site is modified at the 3' end such that its reaction with the enzyme is less efficient than the wild type site, both modified sites being located in the minicircle after recombination. This favors the formation of minicircle.
  • polynucleotides for the encoded PD-L1 bispecific fusion proteins of the present disclosure include, but are not limited to, ribopolynucleotides (RNAs), deoxyribopolynucleo tides (DNAs), threose polynucleotides (TNAs), glycol polynucleotides (GNAs), peptide polynucleotides (PNAs), locked polynucleotides (LNAs, including LNA having a b- D-ribo configuration, a-LNA having an a-L-ribo configuration (a diastereomer of LNA), 2'- amino-LNA having a 2 '-amino functionalization, and 2'-amino- a-LNA having a 2'-amino functionalization), ethylene polynucleotides (ENA), cyclohexenyl polynucleotides (CeNA) or hybrids or combinations thereof.
  • mRNA presents an emerging platform for antibody gene transfer that can be adapted by those skilled in the art for delivery of polynucleotides of the present disclosure.
  • current results differ considerably, in certain instances the mRNA constructs appear to be able to rival viral vectors in terms of generated serum mAb titers.
  • Levels were in therapeutically relevant ranges within hours after mRNA administration, a marked shift in speed compared to DNA.
  • LNP lipid nanoparticles
  • mRNA-based therapeutics are expressed more transiently compared to DNA- or viral vector-mediated gene transfer, and do not pose the risk of insertional mutagenesis in the host genome.
  • mRNA production is relatively simple and inexpensive.
  • mRNA uptake can be enhanced using electroporation.
  • Broderick et al. 2017 “Enhanced delivery of DNA or RNA vaccines by electroporation” Methods Mol Biol. 2017;1499:193-200.
  • Most focus, however, has gone to non-physical transfection methods. Indeed, a variety of mRNA complexing formulations have been developed, including lipid nanoparticles (LNP), which have proven to be safe and very efficient mRNA carriers for administration in a variety of tissues and i.v. Pardi et al. 2015 “Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes” J Control Release 217:345-51. In line with this progress, IVT mRNA has reached the stage of
  • the bispecific fusion protein coding sequence can be provided as an RNA replicon comprising a 5' replication recognition sequence such as from an alphavirus 5' replication recognition sequence.
  • the RNA replicon comprises a (modified) 5' replication recognition sequence and an open reading frame encoding the bispecific fusion protein, in particular located downstream from the 5' replication recognition sequence such as that the 5' replication recognition sequence and the open reading frame do not overlap, e.g.
  • the 5' replication recognition sequence does not contain a functional initiation codon and in some embodiments does not contain any initiation codon.
  • the initiation codon of the open reading frame encoding the bispecific fusion protein is in the 5' 3' direction of the RNA replicon.
  • modified nucleosides can be incorporated into the in vitro-transcribed mRNA.
  • the IVT RNA can be 5’ capped, such an m7G5 'ppp5 'G2 '-O- ct-cappcd IVT. Efficient translation of the modified mRNA can be ensured by removing double-stranded RNA.
  • the 5' and 3' UTRs and the poly(A) tail can be optimized for improved intracellular stability and translational efficiency. See, for example, Stadler et al. (2017) Nature Medicine 23:815-817 and Kariko et al. WO/2017/036889 “Method for Reducing Immunogenicity of RNA”.
  • the mRNA that encodes the PD-L1 bispecific fusion protein may include at least one chemical modification described herein.
  • the chemical modification may be 1 -methylpseudouridine, 5-methylcytosine or 1 - methylpseudouridine and 5-methylcytosine.
  • IVT polynucleotides linear polynucleotides encoding at least one PD-L1 bispecific fusion proteins of the present disclosure which are made using only in vitro transcription (IVT) enzymatic synthesis methods are referred to as "IVT polynucleotides.” Methods of making IVT polynucleotides are known in the art and are described in PCT Application WO2013/151666, the contents of which are incorporated herein by reference in their entirety.
  • polynucleotides that encode the PD-L1 bispecific fusion protein of the present disclosure have portions or regions which differ in size and/or chemical modification pattern, chemical modification position, chemical modification percent or chemical modification population and combinations of the foregoing are known as "chimeric polynucleotides.”
  • a “chimera” according to the present disclosure is an entity having two or more incongruous or heterogeneous parts or regions.
  • a "part" or "region" of a polynucleotide is defined as any portion of the polynucleotide which is less than the entire length of the polynucleotide.
  • Such constructs are taught in for example PCT Application WO20 15/034928.
  • the polynucleotides of the present disclosure that are circular are known as “circular polynucleotides” or “circP.”
  • “circular polynucleotides” or “circP” means a single stranded circular polynucleotide which acts substantially like, and has the properties of, an RNA.
  • the term “circular” is also meant to encompass any secondary or tertiary configuration of the circP. Such constructs are taught in for example PCT Application WO2015/034925 and WO2015/034928, the contents of each of which are incorporated herein by reference in their entirety.
  • mRNA (and other polynucleotides) that can be used to encode PD-L1 bispecific fusion proteins of the present disclosure include those which can be adapted from the specifications and FIGS of, for example, PCT Publications WO2017/049275, WO2016/118724, WO2016/118725, W02016/011226, WO2015/196128, WO/2015/196130, WO/2015/196118, WO/2015/089511, with WO2015/105926 (the later titled “Polynucleotides for the In vivo Production Of Antibodies”), each of which is incorporated by reference herein.
  • Electroporation is one example of a method for introducing mRNA or other polynucleotides into a cell.
  • Lipid-containing nanoparticle compositions have proven effective as transport vehicles into cells and/or intracellular compartments for a variety of RNAs (and related polynucleotides described herein). These compositions generally include at least one "cationic" and/or ionizable lipids, phospholipids including polyunsaturated lipids, structural lipids (e.g., sterols), and lipids containing polyethylene glycol (PEG lipids). Cationic and/or ionizable lipids include, for example, amine-containing lipids that can be readily protonated. Delivery of Polynucleotide Constructs into Target Cells
  • the introduction into host cell of the gene delivery system can be performed through various methods known to those skilled in the art.
  • Physical methods to enhance delivery both viral and non-viral polynucleotides include electroporation (Neumann, E. et ah, EMBO J., 1:841(1982); and Tur-Kaspa et ah, Mol. Cell Biol., 6:716-718(1986)), gene bombardment (Yang et ah, Proc. Natl. Acad. Sci., 87:9568-9572 (1990) where DNA is loaded onto ( e.g ., gold) particles and forced to achieve penetration of the DNA into the cells, sonoporation, magnetofection, hydrodynamic delivery and the like, all of which are known to those of skill in the art.
  • the polynucleotide is introduced into target cells via electroporation.
  • Administration of the composition via electroporation may be accomplished using electroporation devices that can be configured to deliver to a desired tissue of a mammal, a pulse of energy effective to cause reversible pores to form in cell membranes, and preferable the pulse of energy is a constant current similar to a preset current input by a user.
  • the electroporation device may comprise an electroporation component and an electrode assembly or handle assembly.
  • the electroporation component may include and incorporate at least one of the various elements of the electroporation devices, including: controller, current waveform generator, impedance tester, waveform logger, input element, status reporting element, communication port, memory component, power source, and power switch.
  • the electroporation may be accomplished using an in vivo electroporation device, for example CELLECTRA EP system (VGX Pharmaceuticals, Blue Bell, Pa.) or Eigen electroporator (Genetronics, San Diego, Calif.) to facilitate transfection of cells by the plasmid.
  • CELLECTRA EP system VGX Pharmaceuticals, Blue Bell, Pa.
  • Eigen electroporator Geneetronics, San Diego, Calif.
  • the electroporation component may function as one element of the electroporation devices, and the other elements are separate elements (or components) in communication with the electroporation component.
  • the electroporation component may function as more than one element of the electroporation devices, which may be in communication with still other elements of the electroporation devices separate from the electroporation component.
  • the elements of the electroporation devices existing as parts of one electromechanical or mechanical device may not limited as the elements can function as one device or as separate elements in communication with one another.
  • the electroporation component may be capable of delivering the pulse of energy that produces the constant current in the desired tissue and includes a feedback mechanism.
  • the electrode assembly may include an electrode array having a plurality of electrodes in a spatial arrangement, wherein the electrode assembly receives the pulse of energy from the electroporation component and delivers same to the desired tissue through the electrodes. At least one of the plurality of electrodes is neutral during delivery of the pulse of energy and measures impedance in the desired tissue and communicates the impedance to the electroporation component.
  • the feedback mechanism may receive the measured impedance and can adjust the pulse of energy delivered by the electroporation component to maintain the constant current.
  • a plurality of electrodes may deliver the pulse of energy in a decentralized pattern.
  • the plurality of electrodes may deliver the pulse of energy in the decentralized pattern through the control of the electrodes under a programmed sequence, and the programmed sequence is input by a user to the electroporation component.
  • the programmed sequence may comprise a plurality of pulses delivered in sequence, wherein each pulse of the plurality of pulses is delivered by at least two active electrodes with one neutral electrode that measures impedance, and wherein a subsequent pulse of the plurality of pulses is delivered by a different one of at least two active electrodes with one neutral electrode that measures impedance.
  • the feedback mechanism may be performed by either hardware or software.
  • the feedback mechanism may be performed by an analog closed-loop circuit.
  • the feedback occurs every 50 ps, 20 ps, 10 ps or 1 ps, but in some embodiments is a real-time feedback or instantaneous (i.e., substantially instantaneous as determined by available techniques for determining response time).
  • the neutral electrode may measure the impedance in the desired tissue and communicates the impedance to the feedback mechanism, and the feedback mechanism responds to the impedance and adjusts the pulse of energy to maintain the constant current at a value similar to the preset current.
  • the feedback mechanism may maintain the constant current continuously and instantaneously during the delivery of the pulse of energy.
  • electroporation devices and electroporation methods that may facilitate delivery of the polynucleotides of the present disclosure, include those described in U.S. Pat.
  • the electroporation is carried using a minimally invasive electroporation device ("MID").
  • the device may comprise a hollow needle, DNA cassette, and fluid delivery means, wherein the device is adapted to actuate the fluid delivery means in use so as to concurrently (for example, automatically) inject the polynucleotide polynucleotide construct into body tissue during insertion of the needle into the body tissue.
  • MID minimally invasive electroporation device
  • the device may comprise a hollow needle, DNA cassette, and fluid delivery means, wherein the device is adapted to actuate the fluid delivery means in use so as to concurrently (for example, automatically) inject the polynucleotide polynucleotide construct into body tissue during insertion of the needle into the body tissue.
  • This has the advantage that the ability to inject the DNA and associated fluid gradually while the needle is being inserted leads to a more even distribution of the fluid through the body tissue. The pain experienced during injection may be reduced due to the distribution of the DNA being injected over a larger area
  • the MID may inject the polynucleotide into tissue without the use of a needle.
  • the MID may inject the polynucleotide as a small stream or jet with such force that the polynucleotide pierces the surface of the tissue and enters the underlying tissue and/or muscle.
  • the force behind the small stream or jet may be provided by expansion of a compressed gas, such as carbon dioxide through a micro-orifice within a fraction of a second. Examples of minimally invasive electroporation devices, and methods of using them, are described in published U.S. Patent Application No. 20080234655; U.S. Pat. No. 6,520,950; U.S. Pat. No. 7,171,264; U.S. Pat. No.
  • the MID may comprise an injector that creates a high-speed jet of liquid that painlessly pierces the tissue.
  • Such needle-free injectors are commercially available. Examples of needle- free injectors that can be utilized herein include those described in U.S. Pat. Nos. 3,805,783; 4,447,223; 5,505,697; and 4,342,310, the contents of each of which are herein incorporated by reference.
  • a desired polynucleotide in a form suitable for direct or indirect electro transport may be introduced (e.g ., injected) using a needle-free injector into the tissue to be treated, usually by contacting the tissue surface with the injector so as to actuate delivery of a jet of the agent, with sufficient force to cause penetration of the polynucleotide into the tissue.
  • a needle-free injector into the tissue to be treated, usually by contacting the tissue surface with the injector so as to actuate delivery of a jet of the agent, with sufficient force to cause penetration of the polynucleotide into the tissue.
  • the tissue to be treated is a mucosa, skin or muscle
  • the agent is projected towards the mucosal or skin surface with sufficient force to cause the agent to penetrate through the stratum comeum and into dermal layers, or into underlying tissue and muscle, respectively.
  • Needle-free injectors are well suited to deliver polynucleotides to all types of tissues, including into tumors (intratu
  • the MID may have needle electrodes that electroporate the tissue.
  • pulsing between multiple pairs of electrodes in a multiple electrode array for example set up in rectangular or square patterns, provides improved results over that of pulsing between a pair of electrodes.
  • Disclosed, for example, in U.S. Pat. No. 5,702,359 entitled “Needle Electrodes for Mediated Delivery of Drugs and Genes" is an array of needles wherein a plurality of pairs of needles may be pulsed during the therapeutic treatment.
  • needles were disposed in a circular array, but have connectors and switching apparatus enabling a pulsing between opposing pairs of needle electrodes.
  • a pair of needle electrodes for delivering the polynucleotide to cells may be used. Such a device and system is described in U.S. Pat. No. 6,763,264, the contents of which are herein incorporated by reference.
  • a single needle device may be used that allows injection of the DNA and electroporation with a single needle resembling a normal injection needle and applies pulses of lower voltage than those delivered by presently used devices, thus reducing the electrical sensation experienced by the patient.
  • the MID may comprise at least one electrode arrays.
  • the arrays may comprise two or more needles of the same diameter or different diameters.
  • the needles may be evenly or unevenly spaced apart.
  • the needles may be between 0.005 inches and 0.03 inches, between 0.01 inches and 0.025 inches; or between 0.015 inches and 0.020 inches.
  • the needle may be 0.0175 inches in diameter.
  • the needles may be 0.5 mm, 1.0 mm, 1.5 mm, 2.0 mm, 2.5 mm, 3.0 mm, 3.5 mm, 4.0 mm, or more spaced apart.
  • the MID may consist of a pulse generator and a two or more-needle vaccine injectors that deliver the polynucleotide and electroporation pulses in a single step.
  • the pulse generator may allow for flexible programming of pulse and injection parameters via a flash card operated personal computer, as well as comprehensive recording and storage of electroporation and patient data.
  • the pulse generator may deliver a variety of volt pulses during short periods of time. For example, the pulse generator may deliver three 15 volt pulses of 100 ms in duration.
  • the MID may be a CELLECTRA (Inovio Pharmaceuticals, Plymouth Meeting, Pa.) device and system, which is a modular electrode system, that facilitates the introduction of a macromolecule, such as polynucleotide, into cells of a selected tissue in a body.
  • the modular electrode system may comprise a plurality of needle electrodes; a hypodermic needle; an electrical connector that provides a conductive link from a programmable constant-current pulse controller to the plurality of needle electrodes; and a power source.
  • An operator can grasp the plurality of needle electrodes that are mounted on a support structure and firmly insert them into the selected tissue in a body or plant.
  • the polynucleotide is then delivered via the hypodermic needle into the selected tissue.
  • the programmable constant-current pulse controller is activated and constant-current electrical pulse is applied to the plurality of needle electrodes.
  • the applied constant-current electrical pulse facilitates the introduction of the polynucleotide into the cell between the plurality of electrodes. Cell death due to overheating of cells is minimized by limiting the power dissipation in the tissue by virtue of constant-current pulses.
  • the Cellectra device and system is described in U.S. Pat. No. 7,245,963, the contents of which are herein incorporated by reference.
  • the MID may be an Eigen 1000 system (Inovio Pharmaceuticals).
  • the Eigen 1000 system may comprise device that provides a hollow needle; and fluid delivery means, wherein the apparatus is adapted to actuate the fluid delivery means in use so as to concurrently (for example automatically) inject fluid, the described polynucleotide herein, into body tissue during insertion of the needle into the said body tissue.
  • the advantage is the ability to inject the fluid gradually while the needle is being inserted leads to a more even distribution of the fluid through the body tissue. It is also believed that the pain experienced during injection is reduced due to the distribution of the volume of fluid being injected over a larger area.
  • the automatic injection of fluid facilitates automatic monitoring and registration of an actual dose of fluid injected.
  • This data can be stored by a control unit for documentation purposes if desired.
  • the rate of injection could be either linear or non-linear and that the injection may be carried out after the needles have been inserted through the skin of the subject to be treated and while they are inserted further into the body tissue.
  • Suitable tissues into which fluid may be injected by the apparatus of the present disclosure include tumor tissue, skin and other epithelial tissues, liver tissue and muscle tissue, merely as examples.
  • the apparatus further comprises needle insertion means for guiding insertion of the needle into the body tissue.
  • the rate of fluid injection is controlled by the rate of needle insertion. This has the advantage that both the needle insertion and injection of fluid can be controlled such that the rate of insertion can be matched to the rate of injection as desired. It also makes the apparatus easier for a user to operate. If desired means for automatically inserting the needle into body tissue could be provided.
  • the depth at which muscle tissue begins could for example be taken to be a preset needle insertion depth such as a value of 4 mm which would be deemed sufficient for the needle to get through the skin layer.
  • the sensing means may comprise an ultrasound probe.
  • the sensing means may comprise a means for sensing a change in impedance or resistance.
  • the means may not as such record the depth of the needle in the body tissue but will rather be adapted to sense a change in impedance or resistance as the needle moves from a different type of body tissue into muscle. Either of these alternatives provides a relatively accurate and simple to operate means of sensing that injection may commence.
  • the depth of insertion of the needle can further be recorded if desired and could be used to control injection of fluid such that the volume of fluid to be injected is determined as the depth of needle insertion is being recorded.
  • the apparatus may further comprise: a base for supporting the needle; and a housing for receiving the base therein, wherein the base is moveable relative to the housing such that the needle is retracted within the housing when the base is in a first rearward position relative to the housing and the needle extends out of the housing when the base is in a second forward position within the housing.
  • a base for supporting the needle
  • a housing for receiving the base therein, wherein the base is moveable relative to the housing such that the needle is retracted within the housing when the base is in a first rearward position relative to the housing and the needle extends out of the housing when the base is in a second forward position within the housing.
  • the fluid delivery means may comprise piston driving means adapted to inject fluid at a controlled rate.
  • the piston driving means could for example be activated by a servo motor.
  • the piston driving means may be actuated by the base being moved in the axial direction relative to the housing.
  • alternative means for fluid delivery could be provided.
  • a closed container which can be squeezed for fluid delivery at a controlled or non- controlled rate could be provided in the place of a syringe and piston system.
  • the apparatus described above could be used for any type of injection. It is however envisaged to be particularly useful in the field of electroporation and so it may further comprise means for applying a voltage to the needle. This allows the needle to be used not only for injection but also as an electrode during, electroporation. This is particularly advantageous as it means that the electric field is applied to the same area as the injected fluid.
  • electroporation There has traditionally been a problem with electroporation in that it is very difficult to accurately align an electrode with previously injected fluid and so users have tended to inject a larger volume of fluid than is required over a larger area and to apply an electric field over a higher area to attempt to guarantee an overlap between the injected substance and the electric field.
  • both the volume of fluid injected and the size of electric field applied may be reduced while achieving a good fit between the electric field and the fluid.
  • U.S. Pat. No. 7,245,963 by Draghia-Akli, et al. describes modular electrode systems and their use for facilitating the introduction of a biomolecule into cells of a selected tissue in a body or plant.
  • the modular electrode systems may comprise a plurality of needle electrodes; a hypodermic needle; an electrical connector that provides a conductive link from a programmable constant-current pulse controller to the plurality of needle electrodes; and a power source.
  • An operator can grasp the plurality of needle electrodes that are mounted on a support structure and firmly insert them into the selected tissue in a body or plant.
  • the biomolecules are then delivered via the hypodermic needle into the selected tissue.
  • the programmable constant-current pulse controller is activated and constant-current electrical pulse is applied to the plurality of needle electrodes.
  • the applied constant-current electrical pulse facilitates the introduction of the biomolecule into the cell between the plurality of electrodes.
  • U.S. Patent Pub. 2005/0052630 submitted by Smith, et al. describes an electroporation device which may be used to effectively facilitate the introduction of a biomolecule into cells of a selected tissue in a body or plant.
  • the electroporation device comprises an electro-kinetic device (“EKD device”) whose operation is specified by software or firmware.
  • the EKD device produces a series of programmable constant-current pulse patterns between electrodes in an array based on user control and input of the pulse parameters and allows the storage and acquisition of current waveform data.
  • the electroporation device also comprises a replaceable electrode disk having an array of needle electrodes, a central injection channel for an injection needle, and a removable guide disk.
  • the entire content of U.S. Patent Pub. 2005/0052630 is hereby incorporated by reference.
  • the electrode arrays and methods described in U.S. Pat. No. 7,245,963 and U.S. Patent Pub. 2005/0052630 may be adapted for deep penetration into not only tissues such as muscle, but also other tissues or organs. Because of the configuration of the electrode array, the injection needle (to deliver the biomolecule of choice) is also inserted completely into the target organ, and the injection is administered perpendicular to the target issue, in the area that is pre delineated by the electrodes.
  • the electrodes described in U.S. Pat. No. 7,245,963 and U.S. Patent Pub. 2005/005263 are, for example, 20 mm long and 21 gauge.
  • the electric fields needed for in vivo cell electroporation are generally similar in magnitude to the fields required for cells in vitro.
  • the magnitude of the electric field range from approximately, 10 V/cm to about 1500 V/cm, 300 V/cm to 1500 V/cm, or 1000 V/cm to 1500 V/cm.
  • lower field strengths from about 10 V/cm to 100 V/cm, and more preferably from about 25 V/cm to 75 V/cm
  • the pulse length is long.
  • the nominal electric field is about 25-75 V/cm, if is preferred that the pulse length is about 10 msec.
  • the pulse length can be about 10 s to about 100 ms. There can be any desired number of pulses, typically one to 100 pulses per second. The delay between pulses sets can be any desired time, such as one second.
  • the waveform, electric field strength and pulse duration may also depend upon the type of cells and the type of molecules that are to enter the cells via electroporation.
  • electroporation devices incorporating electrochemical impedance spectroscopy (“EIS") ⁇
  • EIS electrochemical impedance spectroscopy
  • electroporation devices incorporating EIS can be found, e.g., in W02016/161201, which is hereby incorporated by reference.
  • Uptake of the polynucleotides of the present disclosure may also be enhanced by plasma electroporation also termed avalanche transfection.
  • plasma electroporation also termed avalanche transfection.
  • microsecond discharges create cavitation microbubbles at electrode surface.
  • the mechanical force created by the collapsing microbubbles combined with the magnetic field serve to increase transport efficiency across the cell membrane as compared with the diffusion mediated transport associated with conventional electroporation.
  • the technique of plasma electroporation is described in United States Patent Nos. 7,923,251 and 8,283,171. This technique may also be employed in vivo for the transformation of cells. Chaiberg, et al (2006) Investigative Ophthalmology & Visual Science 47:4083-4090; Chaiberg, et al United States Patent No 8, 101 169 Issued January 24, 2012.
  • Plasma is one of the four fundamental states of matter, the others being solid, liquid, and gas. Plasma is an electrically neutral medium of unbound positive and negative particles (i.e. the overall charge of a plasma is roughly zero).
  • a plasma can be created by heating a gas or subjecting it to a strong electromagnetic field, applied with a laser or microwave generator. This decreases or increases the number of electrons, creating positive or negative charged particles called ions (Luo, et al. (1998) Phys. Plasma 5:2868-2870) and is accompanied by the dissociation of molecular bonds, if present.
  • Cold plasmas are produced by the delivery of pulsed high voltage signals to a suitable electrode.
  • Cold plasma devices may take the form of a gas jet device or a dielectric barrier discharge (DBD) device.
  • Cold temperature plasmas have attracted a great deal of enthusiasm and interest by virtue of their provision of plasmas at relatively low gas temperatures. The provision of plasmas at such a temperature is of interest to a variety of applications, including wound healing, anti-bacterial processes, various other medical therapies and sterilization.
  • cold plasmas i.e., non-thermal plasmas
  • Cold plasma devices may take the form of a gas jet device, a dielectric barrier discharge (DBD) device or multi-frequency harmonic-rich power supply.
  • Dielectric barrier discharge device relies on a different process to generate the cold plasma.
  • a dielectric barrier discharge (DBD) device contains at least one conductive electrode covered by a dielectric layer.
  • the electrical return path is formed by the ground that can be provided by the target substrate undergoing the cold plasma treatment or by providing an in-built ground for the electrode.
  • Energy for the dielectric barrier discharge device can be provided by a high voltage power supply, such as that mentioned above. More generally, energy is input to the dielectric barrier discharge device in the form of pulsed DC electrical voltage to form the plasma discharge. By virtue of the dielectric layer, the discharge is separated from the conductive electrode and electrode etching and gas heating is reduced.
  • the pulsed DC electrical voltage can be varied in amplitude and frequency to achieve varying regimes of operation. Any device incorporating such a principle of cold plasma generation (e.g., a DBD electrode device) falls within the scope of various embodiments of the present disclosure.
  • Cold plasma has been employed to transfect cells with foreign polynucleotides.
  • transfection of tumor cells see, e.g., Connolly, et al. (2012) Human Vaccines & Immune-therapeutics 8: 1729-1733; and Connolly et al (2015) Bioelectrochemistry 103: 15-21 .
  • the transgene construct encoding the PD-L1 bispecific fusion protein of the present disclosure is delivered using an electroporation device comprising: an applicator; a plurality of electrodes extending from the applicator, the electrodes being associated with a cover area; a power supply in electrical communication with the electrodes, the power supply configured to generate at least one electroporating signals to cells within the cover area; and a guide member coupled to the electrodes, wherein the guide member is configured to adjust the cover area of the electrodes. At least a portion of the electrodes can be positioned within the applicator in a conical arrangement.
  • the at least one electroporating signals may be each associated with an electric field.
  • the device may further comprise a potentiometer coupled to the power supply and electrodes. The potentiometer may be configured to maintain the electric field substantially within a predetermined range.
  • the at least one electroporating signals may be each associated with an electric field.
  • the device may further comprise a potentiometer coupled to the power supply and the electrodes.
  • the potentiometer may be configured to maintain the electric field within a predetermined range so as to substantially prevent permanent damage in the cells within the cover area and /or substantially minimize pain. For instance, potentiometer may be configured to maintain the electric field to about 1300 V/cm.
  • the power supply may provide a first electrical signal to a first electrode and a second electrical signal to a second electrode.
  • the first and second electrical signals may combine to produce a wave having a beat frequency.
  • the first and second electrical signals may each have at least one of a unipolar waveform and a bipolar waveform.
  • the first electrical signal may have a first frequency and a first amplitude.
  • the second electrical signal may have a second frequency and a second amplitude.
  • the first frequency may be different from or the same as the second frequency.
  • the first amplitude may be different from or the same as the second amplitude.
  • the present disclosure provides a method for treating a subject having a tumor, the method comprising: injecting the tumor with an effective dose of plasmid coding for a PD-L1 bispecific fusion protein; and administering electroporation therapy to the tumor.
  • the electroporation therapy further comprises the administration of at least one voltage pulse of about 200 V/cm to about 1500 V/cm over a pulse width of about 100 microseconds to about 20 milliseconds.
  • the plasmid (or a second electroporated plasmid) further encodes at least one immuno stimulatory cytokine, such as selected from the group encoding IL-12, IL-15, and a combination of IL-12 and IL-15.
  • Polynucleotide constructs can also be encapsulated in liposomes, preferably cationic liposomes (Wong, T. K. et ah, Gene, 10:87(1980); Nicolau and Sene, Biochim. Biophys. Acta, 721:185-190 (1982); and Nicolau et ah, Methods Enzymol., 149:157-176 (1987)) or polymersomes (synthetic liposomes) which can interact with the cell membrane and fuse or undergo endocytosis to effect polynucleotide transfer into the cell.
  • the DNA also can be formed into complexes with polymers (polyplexes) or with dendrimers which can directly release their load into the cytoplasm of a cell.
  • Illustrative carriers useful in this regard include microparticles of poly(lactide-co- glycolide), polyacrylate, latex, starch, cellulose, dextran and the like.
  • Other illustrative carriers include supramolecular biovectors, which comprise a non-liquid hydrophilic core (e.g., a cross- linked polysaccharide or oligosaccharide) and, optionally, an external layer comprising an amphiphilic compound, such as a phospholipid (see e.g., U.S. Pat. No. 5,151,254 and PCT applications WO 94/20078, WO/94/23701 and WO 96/06638).
  • the amount of active agent contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented.
  • Biodegradable microspheres may be employed as carriers for compositions. Suitable biodegradable microspheres are disclosed, for example, in U.S. Pat. Nos. 4,897,268; 5,075,109; 5,928,647; 5,811,128; 5,820,883; 5,853,763; 5,814,344, 5,407,609 and 5,942,252. Modified hepatitis B core protein carrier systems such as described in WO/9940934, and references cited therein, will also be useful for many applications. Another illustrative carrier/delivery system employs a carrier comprising particulate-protein complexes, such as those described in U.S. Pat. No. 5,928,647, which can have the added benefit when used intratumorally to deliver the coding sequence for a PD-L1 AFFIMER® polypeptide.
  • Biodegradable polymeric nanoparticles facilitate nonviral polynucleotide transfer to cells.
  • positively charged (approximately 10 mV) particles are formed by the self-assembly of cationic, hydrolytically degradable poly(beta-amino esters) and plasmid DNA.
  • Polynucleotides may also be administered to cells by direct microinjection, temporary cell permeabilizations (e.g., co-administration of repressor and/or activator with a cell permeabilizing agent), fusion to membrane translocating peptides, and the like.
  • Lipid-mediated polynucleotide delivery and expression of foreign polynucleotides, including mRNA, in vitro and in vivo has been very successful.
  • Lipid based non-viral formulations provide an alternative to viral gene therapies.
  • Current in vivo lipid delivery methods use subcutaneous, intradermal, intratumoral, or intracranial injection. Advances in lipid formulations have improved the efficiency of gene transfer in vivo (see PCT Application WO 98/07408).
  • a lipid formulation composed of an equimolar ratio of 1,2- bis(oleoyloxy)-3-(trimethyl ammonio)propane (DOTAP) and cholesterol can significantly enhances systemic in vivo gene transfer.
  • DOTAP 1,2- bis(oleoyloxy)-3-(trimethyl ammonio)propane
  • the DOTAP:cholesterol lipid formulation forms unique structure termed a "sandwich liposome". This formulation is reported to "sandwich" DNA between an invaginated bi-layer or 'vase' structure.
  • Beneficial characteristics of these lipid structures include a positive p, colloidal stabilization by cholesterol, two dimensional polynucleotide packing and increased serum stability.
  • Cationic liposome technology is based on the ability of amphipathic lipids, possessing a positively charged head group and a hydrophobic lipid tail, to bind to negatively charged DNA or RNA and form particles that generally enter cells by endocytosis. Some cationic liposomes also contain a neutral co-lipid, thought to enhance liposome uptake by mammalian cells. Similarly, other polycations, such as poly-l-lysine and polyethylene-imine, complex with polynucleotides via charge interaction and aid in the condensation of DNA or RNA into nanoparticles, which are then substrates for endosome-mediated uptake.
  • pDNA plasmid DNA
  • oligodeoxynucleotides oligodeoxynucleotides
  • synthetic RNA RNA
  • the polynucleotides disclosed herein may be associated with polycationic molecules that serve to enhance uptake into cells. Complexing the polynucleotide construct with polycationic molecules also helps in packaging the construct such their size is reduced, which is believed to assist with cellular uptake. Once in the endosome, the complex dissociates due to the lower pH, and the polycationic molecules can disrupt the endosome's membrane to facilitate DNA escape into the cytoplasm before it can be degraded. Preliminary data shows that the polynucleotide construct embodiments had enhanced uptake into SCs over DCs when complexed with the polycationic molecules polylysine or polyethyleneimine.
  • polycationic molecules useful for complexing with polynucleotide constructs includes cell penetrating peptides (CPP), examples include polylysine (described above), polyarginine and Tat peptides.
  • CPP cell penetrating peptides
  • examples include polylysine (described above), polyarginine and Tat peptides.
  • Cell penetrating peptides (CPP) are small peptides which can bind to DNA and, once released, penetrate cell membranes to facilitate escape of the DNA from the endosome to the cytoplasm.
  • CPP pertains to a 27 residue chimeric peptide, termed MPG, was shown some time ago to bind ss- and ds-oligonucleotides in a stable manner, resulting in a non-covalent complex that protected the polynucleotides from degradation by DNase and effectively delivered oligonucleotides to cells in vitro (Mahapatro A, et ah, J Nanobiotechnol, 2011, 9:55).
  • the complex formed small particles of approximately 150 nm to 1 um when different peptide:DNA ratios were examined, and the 10:1 and 5:1 ratios (150 nm and 1 um respectively).
  • RNA can also be complexed by such polycationic molecules for in vivo delivery.
  • polycationic molecules that may be complexed with the polynucleotide constructs described herein include polycationic polymers commercially available as JETPRIME® and In vivo JET (Polypus-transfection, S.A., Illkirch, France).
  • the present disclosure contemplates a method of delivering an mRNA (or other polynucleotide)f encoding a PD-L1 bispecific fusion protein to a patient’s cells by administering a nanoparticle composition comprising (i) a lipid component comprising a compound of formula (I), a phospholipid, a structural lipid, and a PEG lipid; and (ii) an mRNA (or other polynucleotide)f, said administering comprising contacting said mammalian cell with said nanoparticle composition, whereby said mRNA (or other polynucleotide)f is delivered to said cell.
  • a nanoparticle composition comprising (i) a lipid component comprising a compound of formula (I), a phospholipid, a structural lipid, and a PEG lipid; and (ii) an mRNA (or other polynucleotide)f, said administering comprising contacting said mammalian cell with said nanoparticle composition
  • the PEG lipid is selected from the group consisting of a PEG- modified phosphatidylethanolamine, a PEG-modified phosphatide acid, a PEG-modified ceramide, a PEG-modified dialkylamine, a PEG-modified diacylglycerol and a PEG-modified dialkylglycerol.
  • the structural lipid is selected from the group consisting of cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, ursolic acid, and alpha- tocopherol.
  • the structural lipid is cholesterol.
  • the phospholipid includes a moiety selected from the group consisting of phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl glycerol, phosphatidyl serine, phosphatidic acid, 2-lysophosphatidyl choline, and a sphingomyelin.
  • the phospholipid includes at least one fatty acid moieties selected from the group consisting of lauric acid, myristic acid, myristoleic acid, palmitic acid, palmitoleic acid, stearic acid, oleic acid, linoleic acid, alpha-linolenic acid, erucic acid, arachidic acid, arachidonic acid, phytanoic acid, eicosapentaenoic acid, behenic acid, docosapentaenoic acid, and docosahexaenoic acid.
  • the phospholipid is selected from the group consisting of 1 ,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1 ,2-dimyristoyl-sn-glycero- phosphocholine (DMPC), 1 ,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1 ,2-dipalmitoyl- sn-glycero-3-phosphocholine (DPPC), 1 ,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1 ,2-diundecanoyl-sn-glycero-phosphocholine (DUPC), 1 -palmitoyl-2-oleoyl-sn-glycero-3- phosphocholine (POPC), 1 ,2-di-0-octadecenyl-sn-glycero-3-phosphocholine (1 8:0 Diether PC),
  • the phospholipid can be DOPE and said the component can comprise about 35 mol % to about 45 mol % said compound, about 1 0 mol % to about 20 mol % DOPE, about 38.5 mol % to about 48.5 mol % structural lipid, and about 1 .5 mol % PEG lipid.
  • the lipid component can be about 40 mol % said compound, about 15 mol % phospholipid, about 43.5 mol % structural lipid, and about 1 .5 mol % PEG lipid.
  • the wt/wt ratio of lipid component to PD-L1 bispecific fusion protein encoding mRNA is from about 5:1 to about 50:1, or about 10:1 to about 40: 1
  • the mean size of said nanoparticle composition is from about 50 nm to about 150 nm, or from about 80 nm to about 120 nm.
  • the polydispersity index of said nanoparticle composition is from about 0 to about 0.18, or from about 0.13 to about 0.17.
  • the nanoparticle composition has a zeta potential of about -10 to about +20 mV.
  • the nanoparticle composition further comprises a cationic and/or ionizable lipid selected from the group consisting of 3-(didodecylamino)-Nl ,N 1 ,4-tridodecyl-l -piperazineethanamine (KL1 0), 14,25-ditridecyl-l 5, 1 8,21 ,24-tetraaza-octatriacontane (KL25), 1 ,2-dilinoleyloxy-N,N -dimethylaminopropane (DLin-DMA), 2,2-dilinoleyl-4- dimethylaminomethyl-[l ,3]-dioxolane (DLin-K-DMA), heptatriaconta-6, 9,28,31 -tetraen-1 9-yl 4-(dimethylamino)butanoate (DLin-MC3-DMA), 2,2-dilinoleyl-4-(2-dio)butan
  • bispecific fusion proteins of the disclosure are useful in a variety of applications including, but not limited to, therapeutic treatment methods, such as immunotherapy for cancer.
  • bispecific fusion proteins described herein are useful for activating, promoting, increasing, and/or enhancing an immune response, inhibiting tumor growth, reducing tumor volume, inducing tumor regression, increasing tumor cell apoptosis, and/or reducing the tumorigenicity of a tumor.
  • the polypeptides or agents of the disclosure are also useful for immunotherapy against pathogens, such as viruses.
  • the bispecific fusion proteins described herein are useful for inhibiting viral infection, reducing viral infection, increasing virally-infected cell apoptosis, and/or increasing killing of virus-infected cells.
  • the methods of use may be in vitro, ex vivo, or in vivo methods.
  • the present disclosure provides methods for activating an immune response in a subject using a bispecific fusion protein. In some embodiments, the disclosure provides methods for promoting an immune response in a subject using a bispecific fusion protein described herein. In some embodiments, the disclosure provides methods for increasing an immune response in a subject using a bispecific fusion protein. In some embodiments, the disclosure provides methods for enhancing an immune response in a subject using a bispecific fusion protein. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing cell-mediated immunity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing Thl-type responses.
  • the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing T-cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CD4+ T-cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CD8+ T-cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CTL activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing NK cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing T-cell activity and increasing NK cell activity.
  • the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CU activity and increasing NK cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises inhibiting or decreasing the suppressive activity of Treg cells. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises inhibiting or decreasing the suppressive activity of MDSCs. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing the number of the percentage of memory T-cells. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing long-term immune memory function.
  • the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing long-term memory. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises no evidence of substantial side effects and/or immune-based toxicities. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises no evidence of cytokine release syndrome (CRS) or a cytokine storm.
  • the immune response is a result of antigenic stimulation.
  • the antigenic stimulation is a tumor cell. In some embodiments, the antigenic stimulation is cancer. In some embodiments, the antigenic stimulation is a pathogen. In some embodiments, the antigenic stimulation is a virally-infected cell.
  • a method of increasing an immune response in a subject comprises administering to the subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein a bispecific fusion protein binds human PD-L1.
  • a method of increasing an immune response in a subject comprises administering to the subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein the bispecific fusion protein is a AFFIMER® polypeptide-containing antibody or receptor trap fusion polypeptide including an AFFIMER® polypeptide that specifically binds to PD-L1.
  • a method of increasing an immune response in a subject comprises administering to the subject a therapeutically effective amount of polynucleotide, wherein the polynucleotide, when expressed in the patient, produces a recombinant bispecific fusion protein polypeptide including an anti-PD-Ll AFFIMER® polypeptide.
  • a method of activating or enhancing a persistent or long-term immune response to a tumor comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein which binds human PD- Ll.
  • a method of activating or enhancing a persistent immune response to a tumor comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein the bispecific fusion protein is an AFFIMER® polypeptide-containing antibody or receptor trap fusion polypeptide including an AFFIMER® polypeptide that specifically binds to PD-L1.
  • a method of activating or enhancing a persistent immune response to a tumor comprises administering to a subject a therapeutically effective amount of polynucleotide, wherein the polynucleotide, when expressed in the patient, produces a recombinant bispecific fusion protein polypeptide including an anti- PD-L1 AFFIMER® polypeptide.
  • a method of inducing a persistent or long-term immunity which inhibits tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein which binds human PD-L1.
  • a method of inducing a persistent immunity which inhibits tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein the bispecific fusion protein is a AFFIMER® polypeptide-containing antibody or receptor trap fusion polypeptide including an AFFIMER® polypeptide that specifically binds to PD-L1.
  • a method of inducing a persistent immunity which inhibits tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of polynucleotide, wherein the polynucleotide, when expressed in the patient, produces a recombinant bispecific fusion protein polypeptide including an anti-PD-Ll AFFIMER® polypeptide.
  • a method of inhibiting tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein which binds human PD-L1.
  • a method of inhibiting tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein the bispecific fusion protein is an AFFIMER® polypeptide-containing antibody or receptor trap fusion polypeptide including an AFFIMER® polypeptide that specifically binds to PD-L1.
  • a method of inhibiting tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of polynucleotide, wherein the polynucleotide, when expressed in the patient, produces a recombinant bispecific fusion protein polypeptide including an anti-PD-Ll AFFIMER® polypeptide.
  • the tumor expresses or overexpresses a tumor antigen that is targeted by an additional binding entity provided in the bispecific fusion protein along with the anti-PD-Ll AFFIMER® polypeptide, i.e., where the bispecific fusion protein is a bispecific or multispecific agent.
  • the method of inhibiting growth of a tumor comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein described herein.
  • the subject is a human.
  • the subject has a tumor, or the subject had a tumor which was removed.
  • the tumor is a solid tumor.
  • the tumor is a tumor selected from the group consisting of: colorectal tumor, pancreatic tumor, lung tumor, ovarian tumor, liver tumor, breast tumor, kidney tumor, prostate tumor, neuroendocrine tumor, gastrointestinal tumor, melanoma, cervical tumor, bladder tumor, glioblastoma, and head and neck tumor.
  • the tumor is a colorectal tumor.
  • the tumor is an ovarian tumor.
  • the tumor is a lung tumor.
  • the tumor is a pancreatic tumor.
  • the tumor is a melanoma tumor.
  • the tumor is a bladder tumor.
  • the subject bispecific fusion proteins can be used to treat patients suffering from cancer, such as osteosarcoma, rhabdomyosarcoma, neuroblastoma, kidney cancer, leukemia, renal transitional cell cancer, bladder cancer, Wilm's cancer, ovarian cancer, pancreatic cancer, breast cancer (including triple negative breast cancer), prostate cancer, bone cancer, lung cancer (e.g ., small cell or non-small cell lung cancer), gastric cancer, colorectal cancer, cervical cancer, synovial sarcoma, head and neck cancer, squamous cell carcinoma, multiple myeloma, renal cell cancer, retinoblastoma, hepatoblastoma, hepatocellular carcinoma, melanoma, rhabdoid tumor of the kidney, Ewing's sarcoma, chondrosarcoma, brain cancer, glioblastoma, meningioma, pituitary adenoma, vestibular schwannoma
  • cancer such
  • the cancer is a hematologic cancer.
  • the cancer is selected from the group consisting of: acute myelogenous leukemia (AML), Hodgkin lymphoma, multiple myeloma, T-cell acute lymphoblastic leukemia (T-ALL), chronic lymphocytic leukemia (CLL), hairy cell leukemia, chronic myelogenous leukemia (CML), non- Hodgkin lymphoma, diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), and cutaneous T-cell lymphoma (CTCL).
  • AML acute myelogenous leukemia
  • T-ALL T-cell acute lymphoblastic leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • non- Hodgkin lymphoma diffuse large B-cell lymphoma
  • DLBCL diffuse large B-cell lymphoma
  • MCL mantle cell lympho
  • the present disclosure also provides pharmaceutical compositions comprising a bispecific fusion protein described herein and a pharmaceutically acceptable vehicle.
  • the pharmaceutical compositions find use in immunotherapy.
  • the pharmaceutical compositions find use in immuno-oncology.
  • the compositions find use in inhibiting tumor growth.
  • the pharmaceutical compositions find use in inhibiting tumor growth in a subject ( e.g ., a human patient).
  • the compositions find use in treating cancer.
  • the pharmaceutical compositions find use in treating cancer in a subject (e.g., a human patient).
  • Formulations are prepared for storage and use by combining a purified bispecific fusion protein of the present disclosure with a pharmaceutically acceptable vehicle (e.g., a carrier or excipient).
  • a pharmaceutically acceptable vehicle e.g., a carrier or excipient.
  • pharmaceutically acceptable carriers, excipients, and/or stabilizers to be inactive ingredients of a formulation or pharmaceutical composition.
  • a bispecific fusion protein described herein is lyophilized and/or stored in a lyophilized form. In some embodiments, a formulation comprising a bispecific fusion protein described herein is lyophilized.
  • Suitable pharmaceutically acceptable vehicles include, but are not limited to, nontoxic buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives such as octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl or benzyl alcohol, alkyl parabens, such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol; low molecular weight polypeptides (e.g., less than about 10 amino acid residues); proteins such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • compositions of the present disclosure can be administered in any number of ways for either local or systemic treatment. Administration can be topical by epidermal or transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders; pulmonary by inhalation or insufflation of powders or aerosols, including by nebulizer, intratracheal, and intranasal; oral; or parenteral including intravenous, intraarterial, intratumoral, subcutaneous, intraperitoneal, intramuscular (e.g., injection or infusion), or intracranial (e.g., intrathecal or intraventricular).
  • parenteral including intravenous, intraarterial, intratumoral, subcutaneous, intraperitoneal, intramuscular (e.g., injection or infusion), or intracranial (e.g., intrathecal or intraventricular).
  • a composition is formulated for topical delivery such that the when applied to the skin, for example, the bispecific fusion protein penetrates the skin (crosses epithelial and mucosal barriers) to function systemically.
  • the therapeutic formulation can be in unit dosage form.
  • Such formulations include tablets, pills, capsules, powders, granules, solutions or suspensions in water or non-aqueous media, or suppositories.
  • solid compositions such as tablets the principal active ingredient is mixed with a pharmaceutical carrier.
  • Conventional tableting ingredients include corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and diluents (e.g., water). These can be used to form a solid preformulation composition containing a homogeneous mixture of a compound of the present disclosure, or a non-toxic pharmaceutically acceptable salt thereof.
  • the solid preformulation composition is then subdivided into unit dosage forms of a type described above.
  • the tablets, pills, etc. of the formulation or composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner composition covered by an outer component.
  • the two components can be separated by an enteric layer that serves to resist disintegration and permits the inner component to pass intact through the stomach or to be delayed in release.
  • enteric layers or coatings such materials include a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
  • microcapsules can also be entrapped in microcapsules.
  • microcapsules are prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly- (methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions as described in Remington: The Science and Practice of Pharmacy, 22.sup.nd Edition, 2012, Pharmaceutical Press, London.
  • pharmaceutical formulations include a bispecific fusion protein of the present disclosure complexed with liposomes.
  • Methods to produce liposomes are known to those of skill in the art.
  • some liposomes can be generated by reverse phase evaporation with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG- derivatized phosphatidylethanolamine (PEG-PE).
  • PEG-PE PEG- derivatized phosphatidylethanolamine
  • sustained-release preparations comprising bispecific fusion proteins described herein can be produced.
  • Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing a bispecific fusion protein, where the matrices are in the form of shaped articles ( e.g ., films or microcapsules).
  • sustained-release matrices include polyesters, hydrogels such as poly(2- hydroxyethyl-methacrylate) or poly(vinyl alcohol), polylactides, copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene- vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT.TM. (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(-)- 3-hydroxybutyric acid.
  • polyesters such as poly(2- hydroxyethyl-methacrylate) or poly(vinyl alcohol), polylactides, copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene- vinyl acetate, degradable lactic acid-glycolic acid copo
  • the method or treatment further comprises administering at least one additional immune response stimulating agent.
  • the additional immune response stimulating agent includes, but is not limited to, a colony stimulating factor (e.g., granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), granulocyte colony stimulating factor (G-CSF), stem cell factor (SCF)), an interleukin (e.g., IL- 1, IL2, IL-3, IL-7, IL-12, IL-15, IL-18), a checkpoint inhibitor, an antibody that blocks immunosuppressive functions (e.g., an anti-CTLA-4 antibody, anti-CD28 antibody, anti-CD3 antibody), a toll -like receptor (e.g., TLR4, TLR7, TLR9), or a member of the B7 family (e.g., CD80, CD86).
  • a colony stimulating factor e.g., granulocyte-macrophage colony stimulating factor (
  • An additional immune response stimulating agent can be administered prior to, concurrently with, and/or subsequently to, administration of the bispecific fusion protein.
  • Pharmaceutical compositions comprising a bispecific fusion protein and the immune response stimulating agent(s) are also provided.
  • the immune response stimulating agent comprises 1, 2, 3, or more immune response stimulating agents.
  • the method or treatment further comprises administering at least one additional therapeutic agent.
  • An additional therapeutic agent can be administered prior to, concurrently with, and/or subsequently to, administration of the bispecific fusion protein.
  • Pharmaceutical compositions comprising a bispecific fusion protein and the additional therapeutic agent(s) are also provided.
  • the at least one additional therapeutic agent comprises 1, 2, 3, or more additional therapeutic agents.
  • Combination therapy with two or more therapeutic agents often uses agents that work by different mechanisms of action, although this is not required. Combination therapy using agents with different mechanisms of action may result in additive or synergetic effects. Combination therapy may allow for a lower dose of each agent than is used in monotherapy, thereby reducing toxic side effects and/or increasing the therapeutic index of the bispecific fusion protein. Combination therapy may decrease the likelihood that resistant cancer cells will develop.
  • combination therapy comprises a therapeutic agent that affects the immune response (e.g ., enhances or activates the response) and a therapeutic agent that affects (e.g., inhibits or kills) the tumor/cancer cells.
  • the combination of a bispecific fusion protein described herein and at least one additional therapeutic agent results in additive or synergistic results.
  • the combination therapy results in an increase in the therapeutic index of the bispecific fusion protein.
  • the combination therapy results in an increase in the therapeutic index of the additional therapeutic agent(s).
  • the combination therapy results in a decrease in the toxicity and/or side effects of the bispecific fusion protein.
  • the combination therapy results in a decrease in the toxicity and/or side effects of the additional therapeutic agent(s).
  • Useful classes of therapeutic agents include, for example, anti-tubulin agents, auristatins, DNA minor groove binders, DNA replication inhibitors, alkylating agents (e.g., platinum complexes such as cisplatin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and carboplatin), anthracyclines, antibiotics, anti-folates, anti-metabolites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated pyrimidines, ionophores, lexitropsins, nitrosoureas, platinols, purine antimetabolites, puromycins, radiation sensitizers, steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like.
  • the second therapeutic agent is an alkylating agent, an antimetabolite, an antimitotic, a topoisomerase inhibitor, or an angiogenesis inhibitor.
  • Therapeutic agents that may be administered in combination with the bispecific fusion protein described herein include chemotherapeutic agents.
  • the method or treatment involves the administration of a bispecific fusion protein of the present disclosure in combination with a chemotherapeutic agent or in combination with a cocktail of chemotherapeutic agents.
  • Treatment with a bispecific fusion protein can occur prior to, concurrently with, or subsequent to administration of chemotherapies.
  • Combined administration can include co-administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive administration in either order but generally within a time period such that all active agents can exert their biological activities simultaneously.
  • Preparation and dosing schedules for such chemotherapeutic agents can be used according to manufacturers' instructions or as determined empirically by the skilled practitioner. Preparation and dosing schedules for such chemotherapy are also described in The Chemotherapy Source Book, 4.sup.th Edition, 2008, M. C. Perry, Editor, Lippincott, Williams & Wilkins, Philadelphia, Pa.
  • Chemotherapeutic agents useful in the present disclosure include, but are not limited to, alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamime; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard;
  • paclitaxel TAXOL
  • docetaxel TAXOTERE
  • chlorambucil gemcitabine
  • 6- thioguanine mercaptopurine
  • platinum analogs such as cisplatin and carboplatin
  • vinblastine platinum
  • etoposide VP-16
  • ifosfamide mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; ibandronate; CPT11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; esperamicins; capecitabine (XELODA); and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • DMFO difluoromethylornithine
  • XELODA retinoic acid
  • esperamicins capecitabine
  • Chemotherapeutic agents also include anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (FARESTON); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • the additional therapeutic agent is cisplatin.
  • the additional therapeutic agent is carboplatin.
  • the chemotherapeutic agent is a topoisomerase inhibitor.
  • Topoisomerase inhibitors are chemotherapy agents that interfere with the action of a topoisomerase enzyme (e.g ., topoisomerase I or II).
  • Topoisomerase inhibitors include, but are not limited to, doxorubicin HC1, daunombicin citrate, mitoxantrone HC1, actinomycin D, etoposide, topotecan HC1, teniposide (VM-26), and irinotecan, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these.
  • the additional therapeutic agent is irinotecan.
  • the chemotherapeutic agent is an anti-metabolite.
  • An anti metabolite is a chemical with a structure that is similar to a metabolite required for normal biochemical reactions, yet different enough to interfere with at least one normal functions of cells, such as cell division.
  • Anti-metabolites include, but are not limited to, gemcitabine, fluorouracil, capecitabine, methotrexate sodium, ralitrexed, pemetrexed, tegafur, cytosine arabinoside, thioguanine, 5-azacytidine, 6-mercaptopurine, azathioprine, 6-thioguanine, pentostatin, fludarabine phosphate, and cladribine, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these.
  • the additional therapeutic agent is gemcitabine.
  • the chemotherapeutic agent is an antimitotic agent, including, but not limited to, agents that bind tubulin.
  • the agent is a taxane.
  • the agent is paclitaxel or docetaxel, or a pharmaceutically acceptable salt, acid, or derivative of paclitaxel or docetaxel.
  • the agent is paclitaxel (TAXOL), docetaxel (TAXOTERE), albumin-bound paclitaxel (nab-paclitaxel; ABRAXANE), DHA-paclitaxel, or PG-paclitaxel.
  • the antimitotic agent comprises a vinca alkaloid, such as vincristine, vinblastine, vinorelbine, or vindesine, or pharmaceutically acceptable salts, acids, or derivatives thereof.
  • the antimitotic agent is an inhibitor of kinesin Eg5 or an inhibitor of a mitotic kinase such as Aurora A or Plkl.
  • the additional therapeutic agent is paclitaxel.
  • the additional therapeutic agent is nab-paclitaxel.
  • an additional therapeutic agent comprises an agent such as a small molecule.
  • treatment can involve the combined administration of a bispecific fusion protein of the present disclosure with a small molecule that acts as an inhibitor against tumor-associated antigens including, but not limited to, EGFR, HER2 (ErbB2), and/or VEGF.
  • tumor-associated antigens including, but not limited to, EGFR, HER2 (ErbB2), and/or VEGF.
  • a bispecific fusion protein of the present disclosure is administered in combination with a protein kinase inhibitor selected from the group consisting of: gefitinib (IRESSA), erlotinib (TARCEVA), sunitinib (SUTENT), lapatanib, vandetanib (ZACTIMA), AEE788, CI-1033, cediranib (RECENTIN), sorafenib (NEXAVAR), and pazopanib (GW786034B).
  • IRESSA gefitinib
  • TARCEVA sunitinib
  • ZACTIMA ZACTIMA
  • AEE788, CI-1033 cediranib
  • sorafenib NEXAVAR
  • GW786034B pazopanib
  • an additional therapeutic agent comprises an mTOR inhibitor.
  • the additional therapeutic agent is a small molecule that inhibits a cancer stem cell pathway.
  • the additional therapeutic agent is an inhibitor of the Notch pathway.
  • the additional therapeutic agent is an inhibitor of the Wnt pathway.
  • the additional therapeutic agent is an inhibitor of the BMP pathway.
  • the additional therapeutic agent is an inhibitor of the Hippo pathway.
  • the additional therapeutic agent is an inhibitor of the mTOR/AKR pathway.
  • the additional therapeutic agent is an inhibitor of the RSPO/LGR pathway.
  • an additional therapeutic agent comprises a biological molecule, such as an antibody.
  • treatment can involve the combined administration of a bispecific fusion protein of the present disclosure with antibodies against tumor-associated antigens including, but not limited to, antibodies that bind EGFR, HER2/ErbB2, and/or VEGF.
  • the additional therapeutic agent is an antibody specific for a cancer stem cell marker.
  • the additional therapeutic agent is an antibody that binds a component of the Notch pathway.
  • the additional therapeutic agent is an antibody that binds a component of the Wnt pathway.
  • the additional therapeutic agent is an antibody that inhibits a cancer stem cell pathway.
  • the additional therapeutic agent is an inhibitor of the Notch pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the Wnt pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the BMP pathway. In some embodiments, the additional therapeutic agent is an antibody that inhibits b- catenin signaling. In some embodiments, the additional therapeutic agent is an antibody that is an angiogenesis inhibitor (e.g ., an anti- VEGF or VEGF receptor antibody).
  • angiogenesis inhibitor e.g ., an anti- VEGF or VEGF receptor antibody.
  • the additional therapeutic agent is bevacizumab (AVASTIN), ramucirumab, trastuzumab (HERCEPTIN), pertuzumab (OMNITARG), panitumumab (VECTIBIX), nimotuzumab, zalutumumab, or cetuximab (ERBITETX).
  • the additional therapeutic agent is an antibody that modulates the immune response.
  • the additional therapeutic agent is an anti-PD- 1 antibody, an anti-LAG-3 antibody, an anti-CTLA-4 antibody, an anti-TIM-3 antibody, or an anti-TIGIT antibody.
  • treatment with a bispecific fusion protein described herein can include combination treatment with other biologic molecules, such as at least one cytokines (e.g ., lymphokines, interleukins, tumor necrosis factors, and/or growth factors) or can be accompanied by surgical removal of tumors, removal of cancer cells, or any other therapy deemed necessary by a treating physician.
  • the additional therapeutic agent is an immune response stimulating agent.
  • the bispecific fusion protein can be combined with a growth factor selected from the group consisting of: adrenomedullin (AM), angiopoietin (Ang), BMPs, BDNF, EGF, erythropoietin (EPO), FGF, GDNF, G-CSF, GM-CSF, GDF9, HGF, HDGF, IGF, migration-stimulating factor, myostatin (GDF-8), NGF, neurotrophins, PDGF, thrombopoietin, TGF-a, TGF-b, TNF-a, VEGF, P1GF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-12, IL-15, and IL-18.
  • a growth factor selected from the group consisting of: adrenomedullin (AM), angiopoietin (Ang), BMPs, BDNF, EGF, erythropoi
  • the additional therapeutic agent is an immune response stimulating agent.
  • the immune response stimulating agent is selected from the group consisting of granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), granulocyte colony stimulating factor (G-CSF), interleukin 3 (IF-3), interleukin 12 (IF-12), interleukin 1 (IF-1), interleukin 2 (IF-2), B7-1 (CD80), B7-2 (CD86), 4- IBB ligand, anti-CD3 antibody, anti-CTFA-4 antibody, anti-TIGIT antibody, anti-PD- 1 antibody, anti-FAG-3 antibody, and anti-TIM-3 antibody.
  • an immune response stimulating agent is selected from the group consisting of: a modulator of PD-1 activity, a modulator of PD- F2 activity, a modulator of CTFA-4 activity, a modulator of CD28 activity, a modulator of CD80 activity, a modulator of CD86 activity, a modulator of 4- IBB activity, an modulator of 0X40 activity, a modulator of KIR activity, a modulator of Tim-3 activity, a modulator of FAG3 activity, a modulator of CD27 activity, a modulator of CD40 activity, a modulator of GITR activity, a modulator of TIGIT activity, a modulator of CD20 activity, a modulator of CD96 activity, a modulator of IDOl activity, a cytokine, a chemokine, an interferon, an interleukin, a lymphokine, a member of the tumor necrosis factor (TNF) family, and an immuno stimulatory oligonucle
  • an immune response stimulating agent is selected from the group consisting of: a PD-1 antagonist, a PD-L2 antagonist, a CTLA-4 antagonist, a CD80 antagonist, a CD86 antagonist, a KIR antagonist, a Tim-3 antagonist, a LAG3 antagonist, a TIGIT antagonist, a CD20 antagonist, a CD96 antagonist, and/or an IDOl antagonist.
  • the PD- 1 antagonist is an antibody that specifically binds PD-1.
  • the antibody that binds PD-1 is KEYTRUDA (MK-3475), pidilizumab (CT-011), nivolumab (OPDIVO, BMS-936558, MDX- 1106), MEDI0680 (AMP-514), REGN2810, BGB-A317, PDR-001, or STI-A1110.
  • the antibody that binds PD-1 is described in PCT Publication WO 2014/179664, for example, an antibody identified as APE2058, APE1922, APE1923, APE1924, APE 1950, or APE 1963, or an antibody containing the CDR regions of any of these antibodies.
  • the PD-1 antagonist is a fusion protein that includes PD-L2, for example, AMP- 224.
  • the PD-1 antagonist is a peptide inhibitor, for example, AUNP-12.
  • the CTLA-4 antagonist is an antibody that specifically binds CTLA-4.
  • the antibody that binds CTLA-4 is ipilimumab (YERVOY) or tremelimumab (CP-675,206).
  • the CTLA-4 antagonist a CTLA-4 fusion protein, for example, KAHR-102.
  • the LAG3 antagonist is an antibody that specifically binds LAG3.
  • the antibody that binds LAG3 is IMP701, IMP731, BMS-986016, LAG525, and GSK2831781.
  • the LAG3 antagonist includes a soluble LAG3 receptor, for example, IMP321.
  • the KIR antagonist is an antibody that specifically binds KIR.
  • the antibody that binds KIR is lirilumab.
  • an immune response stimulating agent is selected from the group consisting of: a CD28 agonist, a 4- IBB agonist, an 0X40 agonist, a CD27 agonist, a CD80 agonist, a CD86 agonist, a CD40 agonist, and a GITR agonist p
  • the 0X40 agonist includes 0X40 ligand, or an OX40-binding portion thereof.
  • the 0X40 agonist may be MEDI6383.
  • the 0X40 agonist is an antibody that specifically binds 0X40.
  • the antibody that binds 0X40 is MED 16469, MEDI0562, or MOXR0916 (RG7888).
  • the 0X40 agonist is a vector ( e.g ., an expression vector or virus, such as an adenovirus) capable of expressing 0X40 ligand.
  • the OX40-expressing vector is Delta-24-RGDOX or DNX2401.
  • the 4-1BB (CD137) agonist is a binding molecule, such as an anticalin.
  • the anticalin is PRS-343.
  • the 4- IBB agonist is an antibody that specifically binds 4- IBB.
  • antibody that binds 4-1BB is PF-2566 (PF-05082566) or urelumab (B MS-663513).
  • the CD27 agonist is an antibody that specifically binds CD27.
  • the antibody that binds CD27 is varlilumab (CDX-1127).
  • the GITR agonist comprises GITR ligand or a GITR-binding portion thereof.
  • the GITR agonist is an antibody that specifically binds GITR.
  • the antibody that binds GITR is TRX518, MK-4166, or INBRX- 110.
  • immune response stimulating agents include, but are not limited to, cytokines such as chemokines, interferons, interleukins, lymphokines, and members of the tumor necrosis factor (TNF) family.
  • immune response stimulating agents include immunostimulatory oligonucleotides, such as CpG dinucleotides.
  • an immune response stimulating agent includes, but is not limited to, anti-PD-1 antibodies, anti-PD-L2 antibodies, anti-CTLA-4 antibodies, anti-CD28 antibodies, anti-CD80 antibodies, anti-CD86 antibodies, anti-4- IBB antibodies, anti-OX40 antibodies, anti- KIR antibodies, anti-Tim-3 antibodies, anti-LAG3 antibodies, anti-CD27 antibodies, anti-CD40 antibodies, anti-GITR antibodies, anti-TIGIT antibodies, anti-CD20 antibodies, anti-CD96 antibodies, or anti-IDOl antibodies.
  • the bispecific fusion proteins disclosed herein may be used alone, or in association with radiation therapy.
  • the bispecific fusion proteins disclosed herein may be used alone, or in association with targeted therapies.
  • targeted therapies include: hormone therapies, signal transduction inhibitors (e.g ., EGFR inhibitors, such as cetuximab (Erbitux) and erlotinib (Tarceva)); HER2 inhibitors (e.g., trastuzumab (Herceptin) and pertuzumab (Perjeta)); BCR-ABL inhibitors (such as imatinib (Gleevec) and dasatinib (Sprycel)); ALK inhibitors (such as crizotinib (Xalkori) and ceritinib (Zykadia)); BRAF inhibitors (such as vemurafenib (Zelboraf) and dabrafenib (Tafinlar)), gene expression modulators, apoptosis inducers (e.g., bortezomib (Velcade) and car
  • the bispecific fusion proteins of the disclosure may be used in combination with an anti-cancer therapeutic agent or immunomodulatory drug such as an immunomodulatory receptor inhibitor, e.g., an antibody or antigen-binding fragment thereof that specifically binds to the receptor.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a Tim-3 pathway antagonist, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a Vista pathway antagonist, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a BTLA pathway antagonist, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a LAG-3 pathway antagonist, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a TIGIT pathway antagonist, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-PDLl antibody
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with BMS-936559, MSB0010718C or MPDL3280A), for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CTLA4 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CSl antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti- KIR2DL 1/2/3 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CD137 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-GITR antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-PD-L2 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILTl antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT2 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT3 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT4 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT5 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT6 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT7 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT8 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CD40 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-OX40 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DLl antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DL2/3 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DL4 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DL5A antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DL5B antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR3DLl antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR3DL2 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR3DL3 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-NKG2A antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-NKG2C antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ICOS antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-SIRP. alpha antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CD47 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-4- 1 BB antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti- IL- 10 antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-TSLP antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with IL-10 or PEGylated IL-10, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti- APRIL antibody, for example, as part of a pharmaceutical composition.
  • an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CD27 antibody, for example, as part of a pharmaceutical composition.
  • a bispecific fusion protein of the disclosure is administered in association with a STING agonist, for example, as part of a pharmaceutical composition.
  • the cyclic-di-nucleotides (CDNs) cyclic-di-AMP (produced by Listeria monocytogenes and other bacteria) and its analogs cyclic-di-GMP and cyclic-GMP-AMP are recognized by the host cell as a pathogen associated molecular pattern (PAMP), which bind to the pathogen recognition receptor (PRR) known as Stimulator of INterferon Genes (STING).
  • PAMP pathogen associated molecular pattern
  • PRR pathogen recognition receptor
  • STING is an adaptor protein in the cytoplasm of host mammalian cells which activates the TANK binding kinase (TBK1)-IRL3 and the NL-.kappa.B signaling axis, resulting in the induction of IRN-b and other gene products that strongly activate innate immunity. It is now recognized that STING is a component of the host cytosolic surveillance pathway, that senses infection with intracellular pathogens and in response induces the production of ILN-a, leading to the development of an adaptive protective pathogen-specific immune response consisting of both antigen- specific CD4+ and CD8+ T cells as well as pathogen-specific antibodies.
  • a bispecific fusion protein of the disclosure is administered in association with an Akt inhibitor.
  • AKT inhibitors include GDC0068 (also known as GDC-0068, ipatasertib and RG7440), MK-2206, perifosine (also known as KRX-0401), GSK690693, AT7867, triciribine, CCT128930, A-674563, PHT-427, Akti-1/2, afuresertib (also known as GSK2110183), AT13148, GSK2141795, BAY1125976, uprosertib (aka GSK2141795), Akt Inhibitor VIII (l,3-dihydro-l-[l-[[4-(6-phenyl-lH- imidazo [4,5-g] quinoxalin-7-yl)phenyl] m- ethyl] -4-piperidinyl] -2H-benzimidazol
  • Additional Akt inhibitors include: ATP-competitive inhibitors, e.g. isoquinoline-5- sulfonamides (e.g., H-8, H- 89, NL-71-101), azepane derivatives (e.g., (-)-balanol derivatives), aminofurazans (e.g., GSK690693), heterocyclic rings (e.g., 7-azaindole, 6-phenylpurine derivatives, pyrrolo[2,3- d]pyrimidine derivatives, CCT128930, 3-aminopyrrolidine, anilinotriazole derivatives, spiroindoline derivatives, AZD5363, A-674563, A-443654), phenylpyrazole derivatives (e.g., AT7867, AT13148), thiophenecarboxamide derivatives (e.g., Afuresertib (GSK2110183), 2- pyrimidyl-5
  • alpha.- and 3 -acetoxy-timcallic acids acetoxy-tirucallic acid
  • irreversible inhibitors e.g., natural products, antibiotics, Lactoquinomycin, Frenolicin B, kalafungin, medermycin, Boc-Phe-vinyl ketone, 4- hydroxynonenal (4-HNE), 1,6-naphthyridinone derivatives, and imidazo- 1,2-pyridine derivatives.
  • a bispecific fusion protein of the disclosure is administered in association with a MEK inhibitor.
  • MEK inhibitors include AZD6244 (Selumetinib), PD0325901, GSK1120212 (Trametinib), U0126-EtOH, PD184352, RDEA119 (Rafametinib), PD98059, BIX 02189, MEK162 (Binimetinib), AS-703026 (Pimasertib), SL-327, BIX02188, AZD8330, TAK-733, cobimetinib and PD318088.
  • a bispecific fusion protein of the disclosure is administered in association with both an anthracycline such as doxorubicin and cyclophosphamide, including pegylated liposomal doxorubicin.
  • a bispecific fusion protein of the disclosure is administered in association with both an anti-CD20 antibody and an anti-CD3 antibody, or a bispecific CD20/CD3 binder (including a CD20/CD3 BiTE).
  • a bispecific fusion protein of the disclosure is administered in association with a CD73 inhibitor, a CD39 inhibitor or both.
  • These inhibitors can be CD73 binders or CD39 binders (such as antibody, antibody fragments or antibody mimetics) that inhibit the ectonucleosidase activity.
  • the inhibitor may be a small molecule inhibitor of the ectonucleosidase activity, such as 6-N,N-Diethyl-P-y-dibromomethylene-D- adenosine-5 '-triphosphate trisodium salt hydrate, PSB069, PSB 06126,
  • a bispecific fusion protein of the disclosure is administered in association with an inhibitor poly ADP ribose polymerase (PARP).
  • PARP inhibitors include Olaparib, Niraparib, Rucaparib, Talazoparib, Veliparib, CEP9722, MK4827 and BGB-290.
  • a bispecific fusion protein of the disclosure is administered in association with an oncolytic vims.
  • an oncolytic virus is Talimogene Laherparepvec.
  • a bispecific fusion protein of the disclosure is administered in association with an CSF-1 antagonist, such as an agent that binds to CSF-1 or CSF1R and inhibits the interaction of CSF-1 with CSF1R on macrophage.
  • an CSF- 1 antagonist such as an agent that binds to CSF-1 or CSF1R and inhibits the interaction of CSF-1 with CSF1R on macrophage.
  • An example of a CSF- 1 antagonist includes Emactuzumab and FPA008.
  • a bispecific fusion protein of the disclosure is administered in association with an anti-CD38 antibody.
  • An example of an anti-CD39 antibodies include Daratumumab and Isatuximab.
  • a bispecific fusion protein of the disclosure is administered in association with an anti-CD40 antibody.
  • An example of an anti-CD40 antibodies include Selicrelumab and Dacetuzumab.
  • a bispecific fusion protein of the disclosure is administered in association with an inhibitor of anaplatic lymphoma kinase (AFK).
  • AFK anaplatic lymphoma kinase
  • An example of AFK inhibitors include Alectinib, Crizotinib and Ceritinib.
  • a bispecific fusion protein of the disclosure is administered in association with multikinase inhibitor that inhibits at least one selected from the group consisting of the family members of VEGFR, PDGFR and FGFR, or an anti-angiogenesis inhibitor.
  • multikinase inhibitor that inhibits at least one selected from the group consisting of the family members of VEGFR, PDGFR and FGFR, or an anti-angiogenesis inhibitor.
  • inhibitors include Axitinib, Cediranib, Finifanib, Motesanib, Nintedanib, Pazopanib, Ponatinib, Regorafenib, Sorafenib, Sunitinib, Tivozanib, Vatalanib, LY2874455, or SU5402.
  • a bispecific fusion protein of the disclosure is administered in conjunction with at least one vaccine intended to stimulate an immune response to at least one predetermined antigens.
  • the antigen(s) may be administered directly to the individual, or may be expressed within the individual from, for example, a tumor cell vaccine (e.g ., GVAX) which may be autologous or allogenic, a dendritic cell vaccine, a DNA vaccine, an RNA vaccine, a viral-based vaccine, a bacterial or yeast vaccine (e.g., a Listeria monocytogenes or Saccharomyces cerevisiae), etc. See, e.g., Guo et al., Adv. Cancer Res.
  • the target antigen may also be a fragment or fusion polypeptide comprising an immunologically active portion of the antigens listed in the table.
  • a bispecific fusion protein of the disclosure is administered in association with at least one antiemetics including, but not limited to: casopitant (GlaxoSmithKline), Netupitant (MGI-Helsinn) and other NK-1 receptor antagonists, palonosetron (sold as Aloxi by MGI Pharma), aprepitant (sold as Emend by Merck and Co.; Rahway, N.J.), diphenhydramine (sold as Benadryl by Pfizer; New York, N.Y.), hydroxyzine (sold as Atarax by Pfizer; New York, N.Y.), metoclopramide (sold as Reglan by AH Robins Co,; Richmond, Va.), lorazepam (sold as Ativan by Wyeth; Madison, N.J.), alprazolam (sold as Xanax by Pfizer; New York, N.Y.), haloperidol (sold as Haldol by Ortho-Mc
  • casopitant Gax
  • a bispecific fusion protein is administered in association with an agent which treats or prevents such a deficiency, such as, e.g., filgrastim, PEG-filgrastim, erythropoietin, epoetin alfa or darbepoetin alfa.
  • a bispecific fusion protein of the disclosure is administered in association with anti-cancer radiation therapy.
  • the radiation therapy is external beam therapy (EBT): a method for delivering a beam of high-energy X-rays to the location of the tumor. The beam is generated outside the patient ( e.g ., by a linear accelerator) and is targeted at the tumor site. These X-rays can destroy the cancer cells and careful treatment planning allows the surrounding normal tissues to be spared. No radioactive sources are placed inside the patient's body.
  • the radiation therapy is proton beam therapy: a type of conformal therapy that bombards the diseased tissue with protons instead of X-rays.
  • the radiation therapy is conformal external beam radiation therapy: a procedure that uses advanced technology to tailor the radiation therapy to an individual's body structures.
  • the radiation therapy is brachytherapy: the temporary placement of radioactive materials within the body, usually employed to give an extra dose— or boost— of radiation to an area.
  • the treatment involves the administration of a bispecific fusion protein of the present disclosure in combination with anti viral therapy.
  • Treatment with a bispecific fusion protein can occur prior to, concurrently with, or subsequent to administration of antiviral therapy.
  • the anti- viral drug used in combination therapy will depend upon the virus the subject is infected with.
  • Combined administration can include co-administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive administration in either order but generally within a time period such that all active agents can exert their biological activities simultaneously.
  • the combination of a bispecific fusion protein described herein and at least one additional therapeutic agent may be administered in any order or concurrently.
  • the bispecific fusion protein will be administered to patients that have previously undergone treatment with a second therapeutic agent.
  • the bispecific fusion protein and a second therapeutic agent will be administered substantially simultaneously or concurrently.
  • a subject may be given a bispecific fusion protein while undergoing a course of treatment with a second therapeutic agent (e.g., chemotherapy).
  • a bispecific fusion protein will be administered within 1 year of the treatment with a second therapeutic agent.
  • a bispecific fusion protein will be administered within 10, 8, 6, 4, or 2 months of any treatment with a second therapeutic agent.
  • a bispecific fusion protein will be administered within 4, 3, 2, or 1 weeks of any treatment with a second therapeutic agent. In some embodiments, a bispecific fusion protein will be administered within 5, 4, 3, 2, or 1 days of any treatment with a second therapeutic agent. It will further be appreciated that the two (or more) agents or treatments may be administered to the subject within a matter of hours or minutes (i.e., substantially simultaneously).
  • the appropriate dosage of a bispecific fusion protein of the present disclosure depends on the type of disease to be treated, the severity and course of the disease, the responsiveness of the disease, whether the bispecific fusion protein is administered for therapeutic or preventative purposes, previous therapy, the patient's clinical history, and so on, all at the discretion of the treating physician.
  • the bispecific fusion protein can be administered one time or over a series of treatments lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved ( e.g ., reduction in tumor size).
  • Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient and will vary depending on the relative potency of an individual agent. The administering physician can determine optimum dosages, dosing methodologies, and repetition rates.
  • dosage is from 0.01 mg to 100 mg/kg of body weight, from 0.1 mg to 100 mg/kg of body weight, from 1 mg to 100 mg/kg of body weight, from 1 mg to 100 mg/kg of body weight, 1 mg to 80 mg/kg of body weight from 10 mg to 100 mg/kg of body weight, from 10 mg to 75 mg/kg of body weight, or from 10 mg to 50 mg/kg of body weight.
  • the dosage of the bispecific fusion protein is from about 0.1 mg to about 20 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 0.1 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 0.25 mg/kg of body weight.
  • the dosage of the bispecific fusion protein is about 0.5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 1 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 1.5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 2 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 2.5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 7.5 mg/kg of body weight.
  • the dosage of the bispecific fusion protein is about 10 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 12.5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 15 mg/kg of body weight. In some embodiments, the dosage can be given once or more daily, weekly, monthly, or yearly. In some embodiments, the bispecific fusion protein is given once every week, once every two weeks, once every three weeks, or once every four weeks.
  • a bispecific fusion protein may be administered at an initial higher "loading" dose, followed by at least one lower doses.
  • the frequency of administration may also change.
  • a dosing regimen may comprise administering an initial dose, followed by additional doses (or "maintenance" doses) once a week, once every two weeks, once every three weeks, or once every month.
  • a dosing regimen may comprise administering an initial loading dose, followed by a weekly maintenance dose of, for example, one-half of the initial dose.
  • a dosing regimen may comprise administering an initial loading dose, followed by maintenance doses of, for example one-half of the initial dose every other week.
  • a dosing regimen may comprise administering three initial doses for 3 weeks, followed by maintenance doses of, for example, the same amount every other week.
  • any therapeutic agent may lead to side effects and/or toxicities.
  • the side effects and/or toxicities are so severe as to preclude administration of the particular agent at a therapeutically effective dose.
  • drug therapy must be discontinued, and other agents may be tried.
  • many agents in the same therapeutic class often display similar side effects and/or toxicities, meaning that the patient either has to stop therapy, or if possible, suffer from the unpleasant side effects associated with the therapeutic agent.
  • the dosing schedule may be limited to a specific number of administrations or "cycles".
  • the bispecific fusion protein is administered for 3, 4, 5, 6, 7, 8, or more cycles.
  • the bispecific fusion protein is administered every 2 weeks for 6 cycles, the bispecific fusion protein is administered every 3 weeks for 6 cycles, the bispecific fusion protein is administered every 2 weeks for 4 cycles, the bispecific fusion protein is administered every 3 weeks for 4 cycles, etc.
  • Dosing schedules can be decided upon and subsequently modified by those skilled in the art.
  • a method for treating cancer in a human subject comprises administering to the subject a therapeutically effective dose of a bispecific fusion protein in combination with a therapeutically effective dose of a chemotherapeutic agent, wherein one or both of the agents are administered according to an intermittent dosing strategy.
  • the intermittent dosing strategy comprises administering an initial dose of a bispecific fusion protein to the subject and administering subsequent doses of the bispecific fusion protein about once every 2 weeks. In some embodiments, the intermittent dosing strategy comprises administering an initial dose of a bispecific fusion protein to the subject and administering subsequent doses of the bispecific fusion protein about once every 3 weeks. In some embodiments, the intermittent dosing strategy comprises administering an initial dose of a bispecific fusion protein to the subject and administering subsequent doses of the bispecific fusion protein about once every 4 weeks. In some embodiments, the bispecific fusion protein is administered using an intermittent dosing strategy and the chemotherapeutic agent is administered weekly.
  • the disclosure also provides methods for treating subjects using a bispecific fusion protein of the disclosure, wherein the subject suffers from a viral infection.
  • the viral infection is infection with a virus selected from the group consisting of human immunodeficiency virus (HIV), hepatitis virus (A, B, or C), herpes virus (e.g ., VZV, HSV-I, HAV-6, HSV-II, and CMV, Epstein Barr virus), adenovirus, influenza virus, flaviviruses, echovirus, rhinovirus, coxsackie virus, coronavirus, respiratory syncytial virus, mumps virus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, dengue virus, papillomavirus, molluscum virus, poliovirus, rabies virus, JC virus or arboviral encephalitis virus.
  • HCV human immunodeficiency virus
  • A, B, or C hepati
  • the disclosure provides methods for treating subjects using a bispecific fusion protein thereof of the disclosure, wherein the subject suffers from a bacterial infection.
  • the bacterial infection is infection with a bacterium selected from the group consisting of Chlamydia, rickettsial bacteria, mycobacteria, staphylococci, streptococci, pneumonococci, meningococci and gonococci, klebsiella, proteus, serratia, pseudomonas, Legionella, Corynebacterium diphtheriae, Salmonella, bacilli, Vibrio cholerae, Clostridium tetan, Clostridium botulinum, Bacillus anthricis, Yersinia pestis, Mycobacterium leprae, Mycobacterium lepromatosis, and Borriella.
  • the disclosure provides methods for treating subjects using a bispecific fusion protein of the disclosure, wherein the subject suffers from a fungal infection.
  • the fungal infection is infection with a fungus selected from the group consisting of Candida (albicans, krusei, glabrata, tropicalis, etc.), Cryptococcus neoformans, Aspergillus (fumigatus, niger, etc.), Genus Mucorales (mucor, absidia, rhizopus), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum.
  • Candida albicans, krusei, glabrata, tropicalis, etc.
  • Cryptococcus neoformans Aspergillus (fumigatus, niger, etc.)
  • Genus Mucorales micor, absidia, rhizopus
  • the disclosure provides methods for treating subjects using a bispecific fusion protein of the disclosure, wherein the subject suffers from a parasitic infection.
  • the parasitic infection is infection with a parasite selected from the group consisting of Entamoeba histolytica, Balantidium coli, Naegleria fowleri, Acanthamoeba, Giardia lambia, Cryptosporidium, Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondii and Nippostrongylus brasiliensis.
  • a number of antibody expression platforms have been pursued in vivo to which delivery of polynucleotides encoding bispecific fusion proteins can be adapted: these include viral vectors, naked DNA and RNA.
  • the use of gene transfer with polynucleotides encoding bispecific fusion proteins cannot only enable cost-savings by reducing the cost of goods and of production but may also be able to reduce the frequency of drug administration.
  • a prolonged in vivo production of the therapeutic bispecific fusion protein by expression of the polynucleotides encoding bispecific fusion proteins can contribute to (i) a broader therapeutic or prophylactic application of bispecific fusion proteins in price-sensitive conditions, (ii) an improved accessibility to therapy in both developed and developing countries, and (iii) more effective and affordable treatment modalities.
  • cells can be harvested from the host (or a donor), engineered with polynucleotides encoding bispecific fusion proteins to produce bispecific fusion proteins and re-administered to patients.
  • the tumor presents a site for the transfer of polynucleotides encoding bispecific fusion proteins, targeted either via intravenous or direct injection/electroporation.
  • intratumoral expression of polynucletodies encoding bipecific proteins can allow for a local production of the therapeutic bispecific fusion proteins, waiving the need for high systemic bispecific fusion protein levels that might otherwise be required to penetrate and impact solid tumors.
  • cationic liposome technology can be employed, which is based on the ability of amphipathic lipids, possessing a positively charged head group and a hydrophobic lipid tail, to bind to negatively charged DNA or RNA and form particles that generally enter cells by endocytosis.
  • Some cationic liposomes also contain a neutral co-lipid, thought to enhance liposome uptake by mammalian cells. See, for example, Feigner et al. (1987) Lipofection: a highly efficient, lipid-mediated DNA-transfection procedure. MNAS 84:7413- 7417; San et al. (1983) “Safety and short-term toxicity of a novel cationic lipid formulation for human gene therapy” Hum. Gene Ther. 4:781-788; Xu et al. (1996) “Mechanism of DNA release from cationic liposome/DNA complexes used in cell transfection” Biochemistry 35,:5616-5623; and Legendre et al. (1992) “Delivery of plasmid DNA into mammalian cell lines using pH-sensitive liposomes: comparison with cationic liposomes” Pharm. Res. 9, 1235-1242.
  • polycations such as poly-l-lysine and polyethylene-imine
  • polyethylene-imine can be used to deliver polynucleotides encoding bispecific fusion proteins.
  • These polycations complex with nucleic acids via charge interaction and aid in the condensation of DNA or RNA into nanoparticles, which are then substrates for endosome-mediated uptake.
  • cationic nucleic acid complex technologies have been developed as potential clinical products, including complexes with plasmid DNA, oligodeoxynucleotides, and various forms of synthetic RNA.
  • Modified (and unmodified or “naked”) DNA and RNA have also been shown to mediate successful gene transfer in a number of circumstances and can also be used as systems for delivery of polynucleotides encoding bispecific fusion proteins.
  • These include the use of plasmid DNA by direct intramuscular injection, the use of intratumoral injection of plasmid DNA. See, for example, Rodrigo et al. (2012) “De novo automated design of small RNA circuits for engineering synthetic riboregulation in living cells” PNAS 109:15271-15276; Oishi et al. (2005) “Smart polyion complex micelles for targeted intracellular delivery of PEGylated antisense oligonucleotides containing acid-labile linkages” Chembiochem.
  • Viral vectors are currently used as a delivery vehicle in the vast majority of pre-clinical and clinical gene therapy trials and in the first to be approved directed gene therapy. See Gene Therapy Clinical Trials Worldwide 2017 (abedia.com/wiley/). The main driver thereto is their exceptional gene delivery efficiency, which reflects a natural evolutionary development; viral vector systems are attractive for gene delivery, because viruses have evolved the ability to cross through cellular membranes by infection, thereby delivering nucleic acids such as polynucleotides encoding bispecific fusion proteins to target cells. Pioneered by adenoviral systems, the field of viral vector-mediated antibody gene transfer made significant strides in the past decades.
  • Nonviral vectors are easily produced and do not seem to induce specific immune responses.
  • Muscle tissue is most often used as target tissue for transfection, because muscle tissue is well vascularized and easily accessible, and myocytes are long-lived cells.
  • Intramuscular injection of naked plasmid DNA results in transfection of a certain percentage of myocytes.
  • plasmid DNA encoding cytokines and cytokine/IgGl chimeric proteins has been introduced in vivo and has positively influenced (autoimmune) disease outcome.
  • intravascular delivery in which increased gene delivery and expression levels are achieved by inducing a short-lived transient high pressure in the veins.
  • Special blood-pressure cuffs that may facilitate localized uptake by temporarily increasing vascular pressure and can be adapted for use in human patients for this type of gene delivery. See, for example, Zhang et al. (2001) “Efficient expression of naked DNA delivered intraarterially to limb muscles of nonhuman primates” Hum. Gene Ther., 12:427-438
  • Increased efficiency can also be gained through other techniques, such as in which delivery of the nucleic acid is improved by use of chemical carriers — cationic polymers or lipids — or via a physical approach — gene gun delivery or electroporation.
  • chemical carriers — cationic polymers or lipids — or via a physical approach — gene gun delivery or electroporation.
  • electroporation is especially regarded as an interesting technique for nonviral gene delivery. Somiari, et al.
  • Encoded bispecific fusion proteins can be delivered by a wide range of gene delivery system commonly used for gene therapy including viral, non-viral, or physical. See, for example, Rosenberg et al., Science, 242:1575-1578, 1988, and Wolff et al., Proc. Natl. Acad. Sci. USA 86:9011-9014 (1989). Discussion of methods and compositions for use in gene therapy include Eck et al., in Goodman & Gilman's The Pharmacological Basis of Therapeutics, Ninth Edition, Hardman et al., eds., McGraw-Hill, New York, (1996), Chapter 5, pp. 77-101; Wilson, Clin.
  • Promoters are a major cis-acting element within the vector genome design that can dictate the overall strength of expression as well as cell-specificity.
  • a viral vector is used to deliver a nucleic acid encoding a bispecific fusion protein of the present disclosure.
  • viral vectors include adenoviral vectors, adeno-associated viral (AAV) vectors, and retroviral vectors.
  • AAV adeno-associated viral
  • a non-viral vector is used to deliver a nucleic acid encoding a bispecific fusion protein of the present disclosure.
  • Non-limiting examples of non-viral vectors include plasmid vectors (e.g., plasmid DNA (pDNA) delivered via, e.g., hydrodynamic -based transfection or electroporation), minicircle DNA, and RNA-mediate gene transfer (e.g., delivery of messenger RNA (mRNA) encoding a bispecific fusion protein of the present disclosure).
  • plasmid vectors e.g., plasmid DNA (pDNA) delivered via, e.g., hydrodynamic -based transfection or electroporation
  • minicircle DNA e.g., RNA-mediate gene transfer
  • mRNA messenger RNA
  • nucleic acids or polynucleotides for the encoded bispecific fusion proteins of the present disclosure include, but are not limited to, ribonucleic acids (RNAs), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs, including LNA having a b- D-ribo configuration, a-LNA having an a-L-ribo configuration (a diastereomer of LNA), 2'-amino-LNA having a 2 '-amino functionalization, and 2'-amino- a-LNA having a 2'-amino functionalization), ethylene nucleic acids (ENA), cyclohexenyl nucleic acids (CeNA) or hybrids or combinations thereof.
  • RNAs ribonucleic acids
  • DNAs deoxyribonucleic acids
  • mRNA presents an emerging platform for antibody gene transfer that can be adapted by those skilled in the art for delivery of polynucleotide constructs encoding bispecific fusion proteins of the present disclosure. Although current results differ considerably, in certain instances the mRNA constructs appear to be able to rival viral vectors in terms of generated serum mAb titers. Levels were in therapeutically relevant ranges within hours after mRNA administration, a marked shift in speed compared to DNA. The use of lipid nanoparticles (LNP) for mRNA transfection, rather than the physical methods typically required for DNA, can provide significant advantages in some embodiments towards application range.
  • LNP lipid nanoparticles
  • Nucleic acids encoding bispecific fusion proteins may be delivered g, for example, intravenously, intramuscularly, or intratumorally (e.g., by injection, electroporation or other means).
  • Nucleic acids encoding bispecific fusion proteins may be formulated, for example, in lipid nanoparticles or liposomes (e.g., cationic lipid nanoparticles or liposomes), biodegradable microsphere, or other nano- or microparticle.
  • lipid nanoparticles or liposomes e.g., cationic lipid nanoparticles or liposomes
  • biodegradable microsphere e.g., cationic lipid nanoparticles or liposomes
  • Other lipid-based (e.g., PEG lipid) and polymeric- based formulations and delivery vehicles are contemplated herein.
  • a protein comprising a (i) PD-L1 binding ALLIMER® polypeptide sequence which binds to PD-L1 with a Kd of 1X10 -6 M or less and inhibits interaction of the PD-L1 to which it is bound with PD-1; and (ii) a T ⁇ Eb trap polypeptide sequence that can bind to T ⁇ Eb with a Kd of 1X10 -6 M or less and inhibits interaction of the T ⁇ Eb to which it is bound with T ⁇ EbEII.
  • a pharmaceutical preparation compriding a protein of any one of the preceding paragraphs and a at least one pharmaceutically acceptable exicipents.
  • a polynucleotide comprising a coding sequence encoding the protein of any one of the preceding paragraphs.
  • Anti-PD-Ll AFFIMER® proteins fused to human IgGl Fc with a C-terminal TGFB trap were designed and DNA was ordered from Atum with a human CD33 leader sequence present.
  • a schematic of the AFFIMER® bispecific format is presented in FIG. 5A.
  • AFFIMER®-Fc fusion transfections of suspension HEK cell culture (Expi293F cell line; Thermo Fisher) were performed with a mammalian CMV promoter expression vector pD609 (Atum) using ExpiFectamine reagent (Thermo Fisher) following the manufacturer’s protocol. Supernatant was harvested 7 days post-transfection by centrifuging at 15,000 xg for ljiour followed by 0.
  • a second stage polishing purification was performed using size exclusion using a Superdex 26/600 column run in PBS lx (Cytiva).
  • Analytical size exclusion chromatography was carried out using a Yarra SEC-3000 column (Phenomenex) run on an Ultimate 3000 HPLC (high performance liquid chromatography) (Thermo Fisher) at 1.0 ml/min flow rate in PBS lx (FIG. 15B).
  • SDS-PAGE analysis showed protein bands of the expected molecular weight (MW) (FIG. 15C).
  • Binding affinities of purified AVA27-01, AVA27-02 and AVA27-03 (SEQ ID NO: 96, 97, and 98/99, respectively) to PD-Fl-Fc recombinant antigen were measured using surface plasmon resonance (SPR) (FIG. 16, Table 12).
  • SPR surface plasmon resonance
  • Biacore T200 kinetic analysis was performed using running buffer HBS-EP+ (Cytiva) and series S sensor CM5 chip (Cytiva) immobilized with human PD-F1 Fc (R&D Systems) or mouse PD-F1 Fc (R&D Systems) in 10 mM Sodium acetate pH4.0 (Cytiva) using amine coupling reagents (Cytiva).
  • a concentration titration of Affimer Fc fusions was run as analyte at a flow rate of 30pl/min, with an association time of 700 seconds, followed by a dissociation time of 5,000 seconds.
  • PD-F1 Fc antigen immobilized CM5 surface was regenerated with 4.5 mM NaOH (Cytiva) for 45 seconds at 20 pl/min flow rate followed by 10 mM glycine-HCl pH 3 (Cytiva) for 30 seconds at 20 ul/min.
  • Kinetic data was blank subtracted and fit to a 1:1 Fangmuir binding model (BIAcore Evalution software; Cytiva) to calculate K D values.
  • Anti-PD-Fl AFFIMER® proteins were shown to bind with pM affinites to PD-F1 Fc antigen (Table 12).
  • Binding affinities of purified AVA27-01, AVA27-02 and AVA27-03 (SEQ ID NO: 96, 97, and 98/99, respectively) to TGFp 1 antigen were measured using surface plasmon resonance (SPR) (FIG. 17, Table 13).
  • SPR surface plasmon resonance
  • Biacore 8K kinetic analysis was performed using running buffer HBS-EP+ and series S sensor CM5 chip (Cytiva) immobilized with human TGFpi in 10 mM sodium acetate pH 5.5 using amine coupling reagents (Cytiva).
  • a concentration titration of AFFIMER® proteins was run as analyte at a flow rate of 30pl/min, with an association time of 200 seconds, followed by a dissociation time of 3,000 seconds.
  • TGFP immobilized surfaces were regenerated with 4.5 mM NaOH (Cytiva) for 45 seconds at 20 pl/min flow rate followed by 10 mM glycine-HCl pH 3 (Cytiva) for 30 seconds at 20 ul/min.
  • Kinetic data was blank subtracted and fit to a 1:1 Langmuir binding model (BIAcore Insights Evalution software; Cytiva) to calculate K D values.
  • TGFP trap fused to the C-terminus of an AFFIMER® Fc protein (SEQ ID NO: 96 and 97) were shown to bind with similar affinity to the antibody trap fusion AVA27-03 (SEQ ID NO: 98/99) with the range of 15-28pM (Table 13).
  • AVA27-02 and -03 SEQ ID NO: 97 and 98/99 was assessed by direct ELISA.
  • 96-well high binding microplates were coated with 0.5 pg/ml recombinant human PD-L1/B7-H1/CD274 Fc chimera protein (R&D Systems) in carbonate/bicarbonate buffer. After coating, and following each subsequent step, plates were washed three times with PBS containing 0.05% Tween 20. Unbound sites on the plates were blocked with 5% casein in PBS. AFFIMER® polypeptide dilutions (20 nM titrated 1:5) were prepared in duplicate and added to the plates. Binding was detected using a biotinylated human cystatin A antibody (R&D Systems BAF1407) and streptavidin poly-HRP (Thermo ScientificTM N200).
  • AFFIMER®-TGFp trap molecule maintains a similar binding capacity to PD-L1 compared to the parental molecule formatted on an Fc.
  • the calculated EC50 values for AVA27-02 and the parental anti-PD-Ll AFFIMER® polypeptide are 0.09 nM and 0.05 nM, respectively.
  • AVA27-01 (SEQ ID NO: 96) to mouse PD-L1 was also evaluated by direct ELISA (FIG. 19).
  • the method was similar to the method described above but using recombinant mouse PD-L1/B7-H1/CD274 Fc chimera protein (R&D Systems) coated at 0.5 pg/ml instead.
  • the calculated EC50 value for AVA27-01 is 0.042 nM when detected with and anti-cystatin antibody.
  • AVA27-02 and AVA27-03 SEQ ID NO: 97 and 98/99 was also evaluated by direct ELISA.
  • 96 well high binding microplates were coated with 1 pg/ml recombinant human TGFP-l (R&D Systems) in carbonate/bicarbonate buffer. After coating, and following each subsequent step, plates were washed three times with PBS containing 0.05% Tween 20.
  • AFFIMER® polypeptide dilutions (100 nM titrated 1:4) were prepared in duplicate and added to the plates. Binding was detected using a biotinylated human cystatin A antibody (R&D Systems) or anti-Fc biotinylated antibody (Rockland) with streptavidin poly-HRP (Thermo Scientific). All dilutions were prepared in PBS containing 1% casein and 0.01% Tween 20. TMB substrate (Pierce) was added to develop the signal, followed by 2M sulfuric acid to stop the reaction.
  • FIG. 20A shows the results generated by detection of the AFFIMER® scaffold with the anti-cystatin A antibody. Only AVA27-02 was detected, indicating that it is the molecule containing the AFFIMER® scaffold binds TGFp.
  • FIG. 20B shows the comparable binding capacity of AVA27-02 and AVA27-03 to TGFP and no binding of both parental molecules. The calculated EC50 value for AVA27-02 was 0.13 nM when the AFFIMER® scaffold was detected and 0.33 nM when detected with an anti-Fc antibody.
  • FIG. 21A shows that only AVA27-01 (SEQ ID NO: 96) was detected, indicating that the molecule containing the AFFIMER® scaffold binds TGFp.
  • FIG. 21B shows the similar binding capacity of AVA27-01 to T ⁇ b when detected with anti-Fc. No binding of either parental molecule was detected with either of the detection antibodies. The calculated EC 50 value for
  • AVA27-01 was 0.3 nM when detected through the Affimer scaffold and 0.45 nM when detected with an anti-Fc antibody.
  • AVA27-01, AVA27-02, and AVA27-03 were assessed for their capacity to engage both targets simultaneously (FIGs. 22A and 22B).
  • High binding microplates were coated with 1 pg/ml of recombinant human TGF-b (R&D System) in carbonate bicarbonate buffer for 16 + 2 hours. After the coating, and following each subsequent step, plates were washed three times with 150 pi PBS containing 0.05% Tween 20. Unbound sites on the plates were blocked with 100 pi 5% casein in PBS. AFFIMER® polypeptides and antibodies were diluted in dilution buffer (PBST (0.01%) + 1% Casein (Sigma)) to 30nM and titrated 1:3 in a dilution plate before 50 pi were added to the plate and incubated for 90 + 15 minutes at 21 + 1°C.
  • PBST 0.01%
  • Casein Casein
  • Recombinant human PD-L1/B7-H1 Fc chimera protein (R & D Systems) or recombinant mouse PD-L1/B7-H1 Fc chimera protein (R & D Systems) were then diluted to 10 nM before 50 pi of solution was added to the respective wells. The plate was then incubated for 90 + 15 minutes at 21 + 1°C before being washed as previously described.
  • Two detection solutions were prepared, containing 200ng/ml streptavidin poly-HRP (ThermoFisher) and either 125 ng/ml of biotinylated anti-human PD-L1/B7-H1 antibody (R & D Systems) or biotinylated anti-mouse PD-L1/B7-H1 antibody (R&D Systems), 50 pi of solution was added to the plate to detect human or mouse PD-L1. The plate was then incubated for 60 + 5 minutes at 21 + 1°C, before being washed as previously described. 50 pi of room temperature TMB subtrate was added to all wells, followed by 50pl 2M sulfuric acid solution to stop the reaction.
  • Absorbance was read at 450 and 630nm and the average of the duplicate wells was used to calculate the EC50 using four parameter non-linear regression.
  • the calculated EC50 for AVA27-01 (SEQ ID NO: 96) when engaged to both targets and detected via mPD-Ll bridging was 0.22 nM.
  • the calculated EC50 values for AVA27-02 and AVA27-03 (SEQ ID NO: 97 and 98/99, respectively) were 0.19 nM and 0.12 nM, respectively indicating similar bridging capacity.
  • Example 6 AVA27-02 Competes with PD-1 and Blocks the PD-1/PD-L1 axis
  • AVA27-02 and AVA27-03 were evaluated for their ability to disrupt the PD-1:PD-L1 interaction with a competitive ELISA (FIG. 23). Briefly, microplates were coated with 0.5pg/ml reecombinant human PD-l-Fc chimera protein (R&D Systems) in carbonate/bicarbonate buffer. After the coating, and following each subsequent step, plates were washed three times with PBS containing 0.05% Tween 20. Unbound sites on the plates were blocked with 5% casein in PBS.
  • AFFIMER® polypeptide and an antibody control were diluted in duplicate, and preincubated with 6 nM recombinant human PD-L1 Fc chimera protein (R&D Systems) for 60 minutes prior to addition to the ELISA plate. Binding of PD-L1 to PD-1 was measured using a bBiotinylated human PD-L1 antibody (R&D Systems) and streptavidin poly-HRP (Thermo Scientific). All dilutions were prepared in PBS containing 1% casein and 0.01% Tween 20. TMB (Pierce) was added, followed by 2M sulfuric acid to stop the reaction. Absorbance was read at 450 and 630nm and the data normalized to percentage of inhibition.
  • FIG. 23 shows the similar inhibition capacity of AVA27-02 to PD-L1 compared to the parental molecule.
  • the calculated IC50 values for AVA27-02 and the parental anti-PD-Ll AFFIMER® Fc are 0.49 nM and 0.48 nM, respectively.
  • the assay was performed using two engineered cell lines: Jurkat T cells expressing human PD-1 and a luciferase reporter driven by an NFAT response element (NFAT-RE), and PD-L1 aAPC/CHO-Kl cells, which are CHO-K1 cells expressing human PD-L1 and an engineered cell surface protein designed to activate cognate T cell receptors (TCRs) in an antigen-independent manner.
  • NFAT-RE NFAT response element
  • PD-L1 aAPC/CHO-Kl cells which are CHO-K1 cells expressing human PD-L1 and an engineered cell surface protein designed to activate cognate T cell receptors (TCRs) in an antigen-independent manner.
  • TCRs T cell receptors
  • AFFIMER® proteins and Avelumab were diluted to 2X final concentration (20uM and 0.04 mM, respectively) in assay buffer.
  • the AFFIMER® proteins and controls were titrated down (1:2) in assay buffer to give 10-point curves in triplicate.
  • the PD-L1 aAPC/CHO-Kl (target cells, Promega) and the PD-1 effector cells (Promega) were diluted 8 xlO 5 cells/ml and lxlO 6 cells/ml, respectively, and lOul of the cell suspension was added to each well of the assay plate (Coming 3570) and the plate was incubated for 24 hours in a 37°C, 5% CO2 incubator.
  • FIG. 24 shows that AVA27-02 to PD-L1 has a similar blockade activity compared to the parental molecule.
  • the calculated IC50 value for AVA27-02 and the parental anti-PD-Ll AFFIMER® Fc were 100.2 nM and 28.9 nM, respectively.
  • a recombinant cell line HEK293T cells containing a firefly luciferase gene under the control of SMAD responsive element stably integrated into HEK293 cells was used to demonstrate the ability of AVA27-01, AVA27-02, and AVA27-03 (SEQ ID NO: 96, 97, and 98/99, respectively) to neutralize TGFP (FIG. 25). Briefly, 20 pi of O.lxlO 6 cells/ml of SBE reporter cell line (BPS Bioscience) diluted in Thaw Medium IB (BPS Bioscience) were seeded and incubated for 24 hours at 37°C + 5% CO2.
  • Example 8 AVA27-02 Binds on Cell in a Flow Cytometry Cell Binding Assay
  • cells were washed in PBS EDTA 2mM and re-suspended in Cell Staining Buffer (PBS, 1% BSA, 2 mM EDTA and 0.05% sodium Azide)) at a density of 500000 cells/ml, then 100 ul of cells/well were distributed in a 96 well-round bottom plates.
  • AFFIMER® proteins and controls were diluted in cell staining buffer and added for 60 mins at 2-8 °C .
  • Cells were washed with 150 ul of PBS 2mM EDTA then centrifuged 3 min at 350g, the supernatant was discarded by flicking the plate, wash was repeated 2 more times.
  • the calculated EC50 value for AVA27-02 and the parental anti-PD-Ll AFFIMER® Fc bound to H441 were 5.83 nM and 1.8 nM, respectively, and for huPD-Ll CHO cells 21.73 nM and 1.6 nM.
  • each timepoint have been quantify by sandwich ELISA.
  • Microplates were coated with anti-human Fc (Rockland) overnight at 4°C. After the coating, and following each subsequent step, plates were washed three times with 150 pi PBS containing 0.05% Tween 20. The unbound sites on the plates were then blocked with lOOul of saturation buffer (lx PBS + 5% casein (Sigma) and incubated for 90 minutes at 21°C.
  • the protein standards were then prepared in dilution buffer (lx PBST (0.01%) + 1% casein) at lOnM and titrated 1 in 2 for 15 points in duplicate, serum samples were diluted and titrated 1 in 2 for 8 points in duplicate. Following plate saturation, 50 m ⁇ of standard or sample were added to each well, then plates incubated for a further 90 minutes at 21°C. 50ul of detection solution containing 200ng/ml biotinylated anti-Human Fc (Rockland) was added to each well and the plates were incubated for 90 minutes at 21°C. A secondary detection was added (200ng/ml Poly-HRP Streptavidin, 50m1) to each well and incubated for 30 minutes at 21°C.
  • dilution buffer lx PBST (0.01%) + 1% casein
  • FIG. 27 shows that AVA27- 02 (SEQ ID NO: 97) had a similar PK profile to the control molecules anti-PD-Ll AFFIMER® Fc, at 120h the quantity measured in serum was 118 nM for AVA27-02 (SEQ ID NO: 97).
  • FIG. 28 shows the parallelism between the standard and the AVA27-02 (SEQ ID NO: 97) in serum showing the stability of this molecule in the animal as it still binds to both targets simultaneously.

Abstract

The present disclosure is directed to bifunctional proteins including at least portion of ΤΟΕβ Receptor II (ΤΟΡβΚΙΙ) polypeptide sequence that is capable of binding ΤΟΡβ (a "ΤΟΡβ trap" polypeptide sequence) and at least one AFFIMER® polypeptides that bind to Programmed Death Ligand 1 (PD-L1) and inhibit its interaction with PD-1 (a "PD-L1 binding AFFIMER®" polypetide seuquence). Such bispecific protein agents can exhibit a synergistic effect in cancer treatment, as compared to the effect of administering the two agents separately.

Description

PD-U1 INHIBITOR - TGF INHIBITOR BISPECIFIC DRUG MOIETIES
REUATED APPUICATION
This application claims the benefit under 35 U.S.C. § 119(e) of U.S. provisional application number 62/915,837, filed October 16, 2019, which is incorporated by reference herein in its entirety.
BACKGROUND
Significant breakthroughs have been achieved in the fields of oncogenic signaling inhibition and particularly immune-checkpoint blockade has triggered substantial enthusiasm during the last decade. Antibody-mediated blockade of negative immune-checkpoint molecules (e.g., PD-1/PD-L1, CTLA-4) has been shown to achieve profound responses in several of solid cancers. Unfortunately, these responses only occur in a subset of patients or, after initial therapy response, these tumors eventually relapse. Thus, elucidating the determinants of intrinsic or therapy-induced resistance is the key to improve outcomes and de veloping new treatment strategies. Several cytokines and growth factors are involved in the tight regulation of either antitumor immunity or immunosuppressive tumor-promoting inflammation within the tumor microenvironment (TME), of which transforming growth factor beta (TGF-b) is of particular importance.
Immune checkpoint molecules are gaining prominence as targets for cancer immunotherapy, demonstrating durable remission of patients with metastatic lesions. Antibodies targeting programmed death ligand 1 (PD-L1) such as atezolizumab, avelumab, and durvalumab have received regulatory approval. Despite showing remarkable durable remissions, these antibodies only demonstrate their efficacy in a subset of specific cancer types.
Transforming growth factor-b (TGF-b) is an immunosuppressive cytokine which is often produced in large quantities by many cell types in the tumor microenvironment, including tumor cells, regulatory T cells, and myeloid suppressor cells. TGF-b is well known for its pleiotropic role from initiating to promoting tumor development and it has a negative effect on anti-tumor immunity by suppressing the effector functions of several immune effector cells such as neutrophils, macrophages, natural killer (NK) cells, CD8 cells, and CD4 T cells. Together with other cytokines such as interleukin (IF)-2 and IF-6, TGF-b also induces the generation and recruitment of regulatory T cells to further suppress the antitumor T and NK cell responses. Moreover, it is also known for its role in regulating and promoting the accumulation of stiff fibrillary extracellular matrix composed of collagen, resulting in hindered drug transport and infiltration of immune cells into the tumor. Most importantly, high serum levels of three TGF-b isoforms, TGF-bI, TGF-P2, and TGF-P3, correlate with poor clinical outcome. The TGF-b pathway promotes tumor immunosuppression, and its inhibition may enhance the antitumor activity of PD-(L)1 mAbs.
SUMMARY
The present disclosure is directed to bifunctional proteins including at least portion of TGF Receptor II (TGF RII) polypeptide sequence that is capable of binding TGF (herein a “TGF trap” polypeptide) and at least one AFFIMER® polypeptide that binds (e.g., competitively or non-competitively) to Programmed Death Ligand 1 (PD-L1) and inhibits its interaction with PD-1 (herein a “PD-L1 binding AFFIMER® polypeptide”). Such bispecific protein agents can exhibit a synergistic effect in cancer treatment, as compared to the effect of administering the two agents separately.
In some aspects, the present disclosure provides a bispecific fusion protein comprising a PD-L1 binding AFFIMER® polypeptide which binds to PD-L1 with a Kd of 1X10-6M or less and inhibits interaction of the PD-L1 to which it is bound with PD-1.
In some embodiments, the PD-L1 binding AFFIMER® polypeptide binds human PD-L1 and blocks interactions with human PD-1. In some embodiments, the PD-L1 binding AFFIMER® polypeptide binds human PD-L1 and blocks interactions with human CD80. In some embodiments, the PD-L1 binding AFFIMER® polypeptide bind PD-L1 with a Kd of 1X10_7M or less, Kd of 1X10_8M or less, Kd of 1X10_9M or less, or even a Kd of lxlO_10M or less. In some embodiments, the PD-L1 binding AFFIMER® polypeptide bind PD-L1 with a K0ff of 103 s 1 or slower, 104 s 1 or slower, or even 105 s 1 or slower. In some embodiments, the PD- L1 binding AFFIMER® polypeptide bind PD-L1 with a Kon of 103 M V1 or faster, 104M 1s 1 or faster, 105M 1s 1 or faster, or even 106M 1s 1 or faster. In some embodiments, the PD-L1 binding AFFIMER® polypeptide bind PD-L1 with an IC50 in a competitive binding assay with human PD-1 of 1 mM or less, 100 nM or less, 40 nM or less, 20 nM or less, 10 nM or less, 1 nM or less, or even 0.1 nM or less.
In some embodiments, the PD-L1 binding AFFIMER® polypeptide binds PD-L1 in a competitive binding assay with human CD80 (B7-1) with an IC50 of 1 mM or less, 100 nM or less, 40 nM or less, 20 nM or less, 10 nM or less, 1 nM or less, or 0.1 nM or less.
In some embodiments, the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence represented in general formula (I)
FRl-(Xaa)n-FR2-(Xaa)m-FR3 (I) wherein FR1 is a polypeptide sequence represented by MIPGGLSEAK PATPEIQEIV DKVKPQLEEK TNETYGKLEA VQYKTQVLA (SEQ ID NO: 1) or a polypeptide sequence having at least 70% homology thereto;
FR2 is a polypeptide sequence represented by GTNYYIKVRA GDNKYMHLKV FKSL (SEQ ID NO: 2) or a polypeptide sequence having at least 70% homology thereto;
FR3 is a polypeptide sequence represented by EDLVLTGYQV DKNKDDELTG F (SEQ ID NO: 3) or a polypeptide sequence having at least 70% homology thereto; and Xaa, individually for each occurrence, is an amino acid residue; and n and m are each, independently, an integer from 3 to 20.
For some embodiments, the FR1 may a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 1. For some embodiments, FR2 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 2. For some embodiments, FR3 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 2.
In some embodiments, the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence represented in the general formula:
MIP-Xaal-GLSEAKPATPEIQEIVDKVKPQLEEKTNETYGKLEAVQYKTQVLA- (Xaa)n-Xaa2-TNYYIKVRAGDNKYMHLKVF-Xaa3-Xaa4-Xaa5-(Xaa)m-Xaa6-D-Xaa7- VLTGY QVDKNKDDELTGF (SEQ ID NO: 4) wherein
Xaa, individually for each occurrence, is an amino acid residue; n and m are each, independently, an integer from 3 to 20;
Xaal is Gly, Ala, Val, Arg, Lys, Asp, or Glu;
Xaa2 is Gly, Ala, Val, Ser or Thr;
Xaa3 is Arg, Lys, Asn, Gin, Ser, Thr;
Xaa4 is Gly, Ala, Val, Ser or Thr;
Xaa5 is Ala, Val, lie, Leu, Gly or Pro;
Xaa6 is Gly, Ala, Val, Asp or Glu; and Xaa7 is Ala, Val, lie, Leu, Arg or Lys.
For some embodiments, Xaal is Gly, Ala, Arg or Lys, more even more preferably Gly or Arg. For some embodiments, Xaa2 is Gly or Ser. For some embodiments, Xaa3 is Arg, Lys, Asn or Gin, more preferably Lys or Asn. For some embodiments, Xaa4 is Gly or Ser. For some embodiments, Xaa5 is Ala, Val, He, Leu, Gly or Pro, more preferably He, Leu or Pro, and even more preferably Leu or Pro. For some embodiments, Xaa6 is Ala, Val, Asp or Glu, even more preferably Ala or Glu. For some embodiments, Xaa7 is lie, Leu or Arg, more preferably Leu or Arg.
In some embodiments, the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence represented in the general formula:
MIPRGLSEAKPATPEIQEIVDKVKPQLEEKTNETYGKLEAVQYKTQVLA- (Xaa)n-STNYYIKVRAGDNKYMHLKVFNGP-(Xaa)m- ADRVLTGY QVDKNKDDELTGF (SEQ ID NO: 5) wherein Xaa, individually for each occurrence, is an amino acid residue; and n and m are each, independently, an integer from 3 to 20.
In some embodiments of the above sequences, (Xaa)n (“loop 2”) is an amino acid sequence represented in the general formula (II)
-aal-aa2-aa3-Gly-Pro-aa4-aa5-Trp-aa6- (II) wherein aal represents an amino acid residue with a basic sidechain; aa2 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain, more preferably a small aliphatic sidechain, a neutral polar side chain or a basic or acid side chain; aa3 represents an amino acid residue with an aromatic or basic sidechain; aa4 represents an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain; preferably a neutral polar sidechain or a charged (acidic or basic) sidechain; aa5 represents an amino acid residue with a neutral polar or a charged (acidic or basic) or a small aliphatic or an aromatic sidechain; preferably a neutral polar sidechain or a charged sidechain; and aa6 represents an amino acid residue with an aromatic or acid sidechain.
For some embodiments, aal represents Lys, Arg or His, more preferably Lys or Arg. For some embodiments, aa2 represents Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, more preferably Ala, Gin, Asp or Glu. For some embodiments, aa3 represents Phe, Tyr, Trp, Lys, Arg or His, preferably Phe, Tyr, Trp, more preferably His or Tyr, Trp or His. For some embodiments, aa4 represents Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, more preferably Gin, Lys, Arg, His, Asp or Glu. For some embodiments, aa5 represents Ser, Thr, Asn, Gin, Asp, Glu, Arg or His, more preferably Ser, Asn, Gin, Asp, Glu or Arg. For some embodiments, aa6 represents Phe, Tyr, Trp, Asp or Glu; preferably Trp or Asp; more preferably Trp.
In certain other embodiments of the above sequences, (Xaa)n (“loop 2”) is an amino acid sequence represented in the general formula (III) -aal-aa2-aa3-Phe-Pro-aa4-aa5-Phe-Trp- (III) wherein aal represents an amino acid residue with a basic sidechain or aromatic sidechain; aa2 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain, more preferably a small aliphatic sidechain, a neutral polar side chain or a basic or acid side chain; aa3 represents an amino acid residue with an aromatic or basic sidechain, preferably Phe, Tyr, Trp, Lys, Arg or His, more preferably Phe, Tyr, Trp or His, and even more preferably Tyr, Trp or His; aa4 represents an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain; preferably a neutral polar sidechain or a charged (acidic or basic) sidechain; more preferably Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, and even more preferably Gin, Lys, Arg, His, Asp or Glu; and aa5 represents an amino acid residue with a neutral polar or a charged (acidic or basic) or a small aliphatic or an aromatic sidechain; preferably a neutral polar sidechain or a charged sidechain; more preferably Ser, Thr, Asn, Gin, Asp, Glu, Arg or His, and even more preferably Ser, Asn, Gin, Asp, Glu or Arg.
For some embodiments, aal represents Lys, Arg, His, Ser, Thr, Asn or Gin, more preferably Lys, Arg, His, Asn or Gin, and even more preferably Lys or Asn. For some embodiments, aa2 represents Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, more preferably Ala, Gin, Asp or Glu. For some embodiments, aa3 represents Phe, Tyr, Trp, Lys, Arg or His, more preferably Phe, Tyr, Trp or His, and even more preferably Tyr, Trp or His. For some embodiments, aa4 represents Ala, Pro, He, Gin, Thr, Asp, Glu, Lys, Arg or His, and even more preferably Gin, Lys, Arg, His, Asp or Glu. For some embodiments, aa5 represents Ser, Thr, Asn, Gin, Asp, Glu, Arg or His, and even more preferably Ser, Asn, Gin, Asp, Glu or Arg.
In some embodiments of the above sequences, (Xaa)n (“loop 2”) is an amino acid sequence selected from SEQ ID NOS: 6 to 41, or an amino acid sequence having at least 80% homology thereto, and more preferably an amino acid sequence having at least 85%, 90%, 95% or even 98% homology thereto.
In some embodiments of the above sequences, (Xaa)n (“loop 2”) is an amino acid sequence selected from SEQ ID NOS: 6 to 41, or an amino acid sequence having at least 80% identity thereto, and more preferably an amino acid sequence having at least 85%, 90%, 95% or even 98% identity thereto.
In some embodiments of the above sequences, (Xaa)m (“loop 4”) is an amino acid sequence represented in the general formula (IV) -aa7-aa8-aa9-aal0-aal l-aal2-aal3-aal4-aal5- (IV) wherein aa7 represents an amino acid residue with neutral polar or non-polar sidechain or an acidic sidechain; aa8 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain or aromatic sidechain, more preferably a charged (acidic or basic) sidechain; aa9 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain or aromatic sidechain, more preferably a neutral polar side chain or an acid side chain; aalO represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain or aromatic sidechain, more preferably a neutral polar side chain or a basic or acid side chain; aal 1 represents an amino acid residue, preferably an amino acid residue with a neutral polar sidechain or a charged (acidic or basic) sidechain or a nonpolar aliphatic sidechain or an aromatic sidechain, more preferably a neutral polar side chain or a basic or acid side chain; aal 2 represents an amino acid residue, preferably an amino acid residue with a neutral polar sidechain or a charged (acidic or basic) sidechain or a nonpolar aliphatic sidechain or an aromatic sidechain, more preferably an acid side chain; aal 3 represents an amino acid residue, preferably an amino acid residue with a neutral polar sidechain or a charged (acidic or basic) sidechain or a nonpolar aliphatic sidechain or an aromatic sidechain, more preferably an acid side chain; aal 4 represents an amino acid residue, preferably an amino acid residue with a neutral polar sidechain or a charged (acidic or basic) sidechain; and aal 5 represents an amino acid residue, preferably an amino acid residue with a neutral polar or neutral non-polar sidechain or a charged (acidic or basic) sidechain.
For some embodiments, aa7 represents Gly, Ala, Val, Pro, Trp, Gin, Ser, Asp or Glu, and even more preferably Gly, Ala, Trp, Gin, Ser, Asp or Glu. For some embodiments, aa8 represents Asp, Glu, Lys, Arg, His, Gin, Ser, Thr, Asn, Ala, Val, Pro, Gly, Tyr or Phe, and even more preferably Asp, Glu, Lys, Arg, His or Gin. For some embodiments, aa9 represents Gin, Ser, Thr, Asn, Asp, Glu, Arg, Lys, Gly, Leu, Pro or Tyr, and even more preferably Gin, Thr or Asp. For some embodiments, aalO represents Asp, Glu, Arg, His, Lys, Ser, Gin, Asn, Ala, Leu, Tyr, Trp, Pro or Gly, and even more preferably Asp, Glu, His, Gin, Asn, Leu, Trp or Gly. For some embodiments, aall represents Asp, Glu, Ser, Thr, Gin, Arg, Lys, His, Val, He, Tyr or Gly and even more preferably Asp, Glu, Ser, Thr, Gin, Lys or His. For some embodiments, aal2 represents Asp, Glu, Ser, Thr, Gin, Asn, Lys, Arg, Val, Leu, lie, Trp, Tyr, Phe or Gly and even more preferably Asp, Glu, Ser, Tyr, Trp, Arg or Lys. For some embodiments, aal3 represents Ser, Thr, Gin, Asn, Val, lie, Leu, Gly, Pro, Asp, Glu, His, Arg, Trp, Tyr or Phe and even more preferably Ser, Thr, Gin, Asn, Val, He, Leu, Gly, Asp or Glu. For some embodiments, aal4 represents Ala, He, Trp, Pro, Asp, Glu, Arg, Lys, His, Ser, Thr, Gin or Asn and even more preferably Ala, Pro, Asp, Glu, Arg, Lys, Ser, Gin or Asn. For some embodiments, aal5 represents His, Arg, Lys, Asp, Ser, Thr, Gin, Asn, Ala, Val, Leu, Gly or Phe and even more preferably His, Arg, Lys, Asp, Ser, Thr, Gin or Asn.
In some embodiments of the above sequences, (Xaa)n (“loop 4”) is an amino acid sequence selected from SEQ ID NOS: 42 to 77, or an amino acid sequence having at least 80% homology thereto, and more preferably an amino acid sequence having at least 85%, 90%, 95% or even 98% homology thereto.
In some embodiments of the above sequences, (Xaa)n (“loop 4”) is an amino acid sequence selected from SEQ ID NOS: 42 to 77, or an amino acid sequence having at least 80% identity thereto, and more preferably an amino acid sequence having at least 85%, 90%, 95% or even 98% identity thereto.
In some embodiments, the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence selected from SEQ ID NOS: 78 to 86, or an amino acid sequence having at least 70% homology thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% homology thereto.
In some embodiments, the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence selected from SEQ ID NOS: 78 to 86, or an amino acid sequence having at least 70% identity thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% identity thereto.
In some embodiments, the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence can be encoded by a polynucleotide having a coding sequence corresponding to nucleotides 1-336 of one of SEQ ID NOS: 87 to 95, or a coding sequence at least 70% identical thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% identity thereto.
In some embodiments, the PD-L1 binding AFFIMER® polypeptide has an amino acid sequence can be encoded by a polynucleotide having a coding sequence that hybridizes to any one of SEQ ID NOS: 87 to 95 under stringent conditions of 6X sodium chloride/sodium citrate (SSC) at 45°C followed by a wash in 0.2X SSC at 65°C. In some embodiments, the fusion proteins described herein bind PD-L1 through the PD- L1 binding AFFIMER® polypeptide in a manner competitive with PD-L1 binding by anti-PD- L1 antibodies Atezolizumab, Avelumab and/or Durvalumab.
In some embodiments, the fusion proteins described herein include a PD-L1 binding AFFIMER® polypeptide that forms a crystal structure with PD-L1 comprising an interface involving at least 10 residues of PD-L1 selected from Ile-54, Tyr-56, Glu-58, Glu-60, Asp-61, Lys-62, Asn-63, Gin 66, Val-68, Val-76, Val-111, Arg-113, Met-115, He-116, Ser-117, Gly-120, Ala- 121, Asp- 122, Tyr-123, and Arg-125.
In some embodiments, the fusion protein does not include an N-terminal methionine in the mature form, i.e., after cleavage of a secretion signal sequence the mature protein does not start with a methionine. For instance, if the PD-L1 binding AFFIMER® polypeptide sequence is at the N-terminal end of the fusion protein, the mature protein may start with an lie rather than a Met.
In some embodiments, the fusion proteins described herein, in a manner dependent on the PD-L1 binding AFFIMER® polypeptide binding to PD-L1, (a) increases T-cell receptor signaling in subset of T cell bearing certain Vfi chains, for example, VB3, VB12, VB14, and VB17 in human PBMCs, when treated with staphylococcus enterotoxin B (SEB); (b) increases interferon-g production in an SEB assay; and/or (c) increases interleukin-2 (IL-2) production in an SEB assay in a dose dependant manner.
In some embodiments, the fusion proteins described herein, in a manner dependent on the PD-L1 binding AFFIMER® polypeptide binding to PD-L1 (a) increases T-cell proliferation in a mixed lymphocyte reaction (MLR) assay; (b) increases interferon-g production in an MLR assay; and/or (c) increases interleukin-2 (IL-2) secretion in an MLR assay.
In some embodiments, the TGF trap polypeptide has an amino acid sequence selected from SEQ ID NOS: 96, 97 and/or 98/99 or fragment thereof, or an amino acid sequence having at least 70% homology thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% homology thereto.
In some embodiments, the TGF trap polypeptide retains at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99% of the TGF -binding activity of the wild-type sequence.
In some aspects, the present disclosure provides a bispecific fusion protein comprising a TGF trap polypeptide which binds to TGF with a Kd of 1X10-6M or less in a dimeric format, though more preferentially in a monomeric format, and even more preferably with a Kd of 1X10_7M, 1X10_8M or even 1X10_9M or less. In some embodiments, the TGF trap polypeptide is IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEV CVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDE CNDNIIF S EE YNT S NPD (SEQ ID NO: 105).
In some embodiments, the fusion protein may also include (in addition to the PD-L1 binding AFFIMER® polypeptide and TGF trap polypeptides), to illustrate, at least one additional amino acid sequences selected from the group consisting of: secretion signal sequences, peptide linker sequences, affinity tags, transmembrane domains, cell surface retention sequence, substrate recognition sequences for post-translational modifications, multimerization domains to create multimeric structures of the protein aggregating through protein-protein interactions, half-life extending polypeptide moieties, polypeptides for altering tissue localization and antigen binding site of an antibody, and at least one additional AFFIMER® polypeptides binding the PD-L1 or a different target.
In some embodiments, the fusion protein includes a half-life extending polypeptide moiety such as selected from the group consisting of an Fc domain or portion thereof, an albumin protein or portion thereof, an albumin-binding polypeptide moiety, transferrin or portion thereof, a transferrin-binding polypeptide moiety, fibronectin or portion thereof, or a fibronectin- binding polypeptide moiety.
Where the fusion protein includes an Fc domain or a portion thereof, in some embodiments it is an Fc domain that retains FcRn binding.
Where the fusion protein includes an Fc domain or a portion thereof, in some embodiments the Fc domain or a portion thereof is from IgA, IgD, IgE, IgG, and IgM or a subclass (isotype) thereof such as IgGl, IgG2, IgG3, IgG4, IgAl or IgA2.
In some embodiments, the fusion protein has an amino acid sequence of SEQ ID NO: 96, 97, and/or 98/99 or a sequence having at least 70% homology thereto, and even more preferably at least 75%, 80%, 85%, 90%, 95% or even 98% identity thereto.
Where the fusion protein includes an Fc domain or a portion thereof, in some embodiments the Fc domain or a portion thereof retains effector function selected from Clq binding, complement dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of B cell receptor, or a combination thereof. In other embodiments, the Fc domain is selected from naturally occurring Fc domains which do not retain ADCC and/or CDC function, or Fc domains that have been engineered to have reduced or no ADCC and/or CDC function (such as the “LALA” mutation, i.e., human IgGl L234A/L235A (“hlgGl-LALA”)). In some embodiments, where the fusion protein includes a half-life extending polypeptide moiety, that moiety increases the serum half-life of the protein by at least 5-fold relative to its absence from the protein, for example, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70- fold, 80-fold, 90-fold, 100-fold, 200-fold, 500-fold or even 1000-fold.
In some embodiments, the recombinant receptor trap fusion protein includes at least one multimerization domains that induces multimerization of the recombinant receptor trap fusion protein, i.e., complexes including 2, 3, 4, 5, 6, 7, 8, 9 or even 10 recombinant receptor trap fusion proteins in a multimeric complex.
In some embodiments, the disclosure features a fusion protein of the general formula A- B-C or C-B-A, wherein
A represents a PD-L1 binding AFFIMER® polypeptide;
B represents an Fc polypeptide of a human IgG protein for providing serum half-life extension to the fusion protein; and C represents a human TGF RII trap polypeptide.
The fusion protein may further include amino acid linkers connecting each of the PD-L1 binding and/or TGF trap polypeptides to the Fc polypeptide.
In some embodiments, the fusion protein includes an amino acid sequence of SEQ ID NO: 96, 97, or 98/99, which show the sequence of certain mature fusion proteins after removal of a secretion signal sequence.
Table 1.
Figure imgf000012_0001
Figure imgf000013_0001
The disclosure also features a method of promoting local depletion of TGFp. The method includes administering a protein described above, where the protein binds TGF in solution, binds PD-L1 on a cell surface, and carries the bound TGF into the cell (e.g., a cancer cell).
The disclosure also features a method of inhibiting SMAD3 phosphorylation in a cell (e.g., a cancer cell or an immune cell), the method including exposing the cell in the tumor microenvironment to a protein described above.
In some embodiments, the fusion protein of the disclosure is provided as a pharmaceutical preparation suitable for therapeutic use in a human patient, further comprising at least one pharmaceutically acceptable excipient, buffer, salt or the like. In some embodiments, the pharmaceutical preparation is formulated for pulmonary delivery. For example, the pharmaceutical preparation may be formulated as an intranasal formulation. In other embodiments, the pharmaceutical preparation is formulated for topical (e.g., transepithelial) delivery.
In another aspect of the disclosure, there is provided polynucleotides comprising a coding sequence encoding a fusion protein described above (and herein). In some embodiments, the coding sequence is operably linked to at least one transcriptional regulatory sequence, such as a promoter and/or enhancer.
In some embodiments, the polynucleotide includes at least one origins of replication, minichromosome maintenance elements (MME) and/or nuclear localization elements.
In some embodiments, the polynucleotide includes a polyadenylation signal sequence which is operably linked and transcribed with the coding sequence.
In some embodiments, the coding sequence includes at least one intronic sequences
In some embodiments, the polynucleotide includes at least one ribosome binding sites which are transcribed with the coding sequence.
In some embodiments, the polynucleotide is DNA.
In some embodiments, the polynucleotide is RNA, such as an mRNA.
In another aspect of the disclosure, there is provided viral vectors including a coding sequence encoding a bispecific fusion protein, such as proteins described above (and herein).
In another aspect of the disclosure, there is provided plasmid DNA, plasmid Vectors or minicircles including a coding sequence encoding a bispecific fusion protein, such as proteins described above (and herein).
Also provided herein is a pharmaceutical preparation suitable for therapeutic gene delivery in a human patient, comprising a polynucleotide, a viral vector, a plasmid DNA, plasmid Vector or minicircle of the present disclosure, and (ii) at least one pharmaceutically acceptable excipients, buffers, salts, transfection enhancers, electroporation enhancers or the like. The pharmaceutical preparation, in some embodiments, is formulated for pulmonary (e.g., intranasal) delivery. The pharmaceutical preparation, in other embodiments, is formulated for topical (e.g., transepithelial) delivery.
It should be understood that any one of the AFFIMER® polypeptides described herein may include or exclude a signal sequence (e.g., ~ 15-30 amino acids present at the N-terminus of the polypeptide) or a tag sequence (e.g., C-terminal polyhistadine (e.g., HHHHHH (SEQ ID NO: 173))).
Further provided herein are methods comprising administering to a subject the fusion protein or polynucleotide described herein.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1. Schematic representation of an example of fusion protein of the present disclosure (an Fc scaffolded embodiment), including certain of the biological/therapeutic activities conferred by each of the PD-F1 binding AFFIMER® polypeptide and the TGF trap polypeptide sequence. FIG. 2. Generation of AFFIMER® libraries. Variabilized binding loops give rise to unique binding surfaces and selectable AFFIMER® binders.
FIG. 3. Monomeric AFFIMER® binding. AFFIMER® polypeptide binding by flow cytometry on two different cancer cell lines (AVA04-141 (SEQ ID NOs: 38 (Loop 2) and 74 (Loop 4)); AVA04-211 (SEQ ID NOs: 21 and 57); AVA04-227 (SEQ ID NOs: 10 and 46); AVA04-228 (SEQ ID NOs: 7 and 43); AVA04-231 (SEQ ID NOs: 18 and 54); AVA04-236 (SEQ ID NOs: 15 and 51); AVA04-251 (SEQ ID NOs: 39 and 40); AVA04-261 (SEQ ID NOs:
9 and 45); AVA04-269 (SEQ ID NOs: 8 and 44)).
FIG. 4. AFFIMER® multimers are expressed easily in Escherichia coli to high yield and purity of multiple formats (even in shaker flasks production).
FIGs. 5A and 5B. AFFIMER® multimers bind to PD-L1 with kinetics demonstrating avidity beyond the monomeric binding domain.
FIGs. 6A and 6B. AFFIMER® Fc fusions ( e.g ., AVA04-251 hFcl) provide effector function, half-life extension and enhanced affinity (AVA04-251 (SEQ ID NO: 39 (Loop 2) and 75 (Loop 4)); AVA04-251 hFcl fusion (SEQ ID NO: 180)).
FIG. 7. Single PD-L1 binding AFFIMER® polypeptide (e.g., AVA04-251 hFcl) arms match binding kinetics of PD-L1 antibody benchmarks.
FIG. 8. AFFIMER® polypeptide-Fc fusions (e.g., AVA04-251 hFcl) demonstrate increased serum half-life.
FIG. 9. Immunogenicity testing by human PBMC Analysis indicates core AFFIMER® sequence is not likely immunogenic in humans.
FIG. 10. Proof-of-concept demonstrating that AFFIMER® proteins (e.g., AVA04-236) can be formatted at various sites on an Fc (e.g., hlgGl Fc), and so should translate to IgG- AFFIMER® fusions. Typical (unoptimized) expression yields in the range 400-800 mg/1. Analytical SEC-HPLC used to assess purity (AVA04-236-6(EAAAK) hlgGl Fc (SEQ ID NO: 181).
FIG. 11. Illustrates the KD of several PD-L1 AFFIMER® polypeptide Fc formats determined using Biacore, showing highly flexible formatting for fine tuning of binding kinetics to suit therapeutic target. Avidity effects with the divalent Fc format clearly observed.
FIG. 12. Shows the calculated 3-dimensional structures of the anti-PD-Ll AFFIMER® AVA04-261 (Loop 2 SEQ ID NO: 9 and Loop 4 SEQ ID NO: 45) and human PD-L1 derived from the crystallization of the protein complex.
FIG. 13. From the crystal-derived structure of anti-PD-Ll AFFIMER® AVA04-261 bound to human PD-L1 derived, FIG.13 provides the hydrogen bonding interactions between amino acid residues at the interface of contact between the two proteins. FIG. 14. From the crystal-derived structure of anti-PD-Ll AFFIMER® AVA04-261 bound to human PD-L1 derived, FIG. 14 provides a list of amino acid residues involved in the interface of contact between the two proteins.
FIGs. 15A-15C. Protein production and protein characterization of the bispecific AFFIMER® polypeptide-Fc fusions. FIG. 15A is a schematic showing the fusion protein - anti- PD-Ll AFFIMER-hlgG 1 LALA (L234A/L235A) Fc-TGFp Trap. FIG. 15B shows the purity of the proteins and FIG. 15C shows the size characterization of the reducing and non-reducing forms of the fusion proteins.
FIG. 16. SPR binding affinities of AFFIMER® polypeptide-Fc fusions to PD-Ll-Fc antigen (AVA27-01 (SEQ ID NO: 96); AVA27-02 (SEQ ID NO: 97); and AVA27-03 (SEQ ID NO: 98)).
FIG. 17. SPR binding affinities of AFFIMER® polypeptide-Fc fusions to TGF Beta antigen (AVA27-01 (SEQ ID NO: 96); AVA27-02 (SEQ ID NO: 97); and AVA27-03 (SEQ ID NO: 98)).
FIG. 18. Binding ELISA for human PD-L1 (hPD-Ll) (AVA04-251 hFcl and AVA27-
02).
FIG. 19. Binding ELISA for murine PD-L1 (mPD-Ll) (AVA27-01; SEQ ID NO: 96).
FIGs. 20A and 20B. Binding ELISA for TGFp. FIG. 20A shows the results from detection of the AFFIMER® scaffold (AVA27-02 (SEQ ID NO: 97) and AVA27-03 (SEQ ID NO: 98)) with the anti-cystatin A antibody and FIG. 20B shows the binding capacity of two constructs (AVA27-02 and AVA27-03) to TGF-b (and no biding of the parental molecules).
FIGs. 21A and 21B. Binding ELISA for AVA27-01 (SEQ ID NO: 96). FIG. 21A shows the results from detection of the AFFIMER® scaffold anti-mPDL-1 AFFIMER® polypeptide IgG with the anti-cystatin A antibody and FIG. 21B shows the binding capacity of the construct to TGF-b (and no binding of the parental molecule).
FIGs. 22A and 22B. Sandwich ELISA dual engagement results to examine binding of both targets. FIG. 22A shows results from a bridging ELISA using hPD-Ll AFFIMER® polypeptide-Fc (AVA27-02 (SEQ ID NO: 97) and AVA27-03 (SEQ ID NO: 98)). FIG. 22B shows results from a bridging ELISA using mPD-Ll AFFIMER® polypeptide-Fc (AVA27-01 (SEQ ID NO: 96)).
FIG. 23. Results from a PDLPD-Ll competitive ELISA showing that the constructs (e.g., anti-PD-Ll AFFIMER® polypeptide-Fc, AVA27-02 (SEQ ID NO: 97) and AVA27-03 (SEQ ID NO: 98)) disrupt the PD-1 :PD-L1 interaction.
FIG. 24. Results from a PDLPD-Ll blockade bioluminescent reporter cell-based assay (AVA27-02 (SEQ ID NO: 97)). FIG. 25. Results from a TGFP SMAD reporter assay demonstrating the ability of the fusion proteins (AVA27-01 (SEQ ID NO: 96); AVA27-02 (SEQ ID NO: 97); and AVA27-03 (SEQ ID NO: 98)) to neutralize TGFp.
FIGs. 26A-26B. Cell Binding Assay (e.g., AVA27-02 (SEQ ID NO: 97)).
FIG. 27. Mouse pharmacokinetic (PK) half-life analysis (e.g., AVA27-02 (SEQ ID NO:
97)).
FIG. 28. Sandwich ELISA dual target engagement in serum from 120h PK timepoint (e.g., AVA27-02 (SEQ ID NO: 97)).
DETAILED DESCRIPTION
The current disclosure permits localized reduction in TGF in a tumor microenvironment by capturing the TGF using a soluble cytokine receptor (e.g., TGF RII) tethered to a PD-L1 binding AFFIMER® moiety targeting a PD-L1 found on the exterior surface of certain tumor cells and tumor stromal cells. This bifunctional molecule is effective precisely because the PD- L1 binding AFFIMER® polypeptide and TGF trap are physically linked. The resulting advantage (over, for example, administration of the PD-L1 binding AFFIMER® polypeptide or an anti-PD-Ll antibody and the receptor trap as separate molecules) is partly because cytokines function predominantly in the local environment through autocrine and paracrine functions. The PD-L1 binding AFFIMER® polypeptide directs the TGF trap to the tumor microenvironment where it can be most effective, by neutralizing the local immunosuppressive autocrine or paracrine effects of TGF .
Based on naturally occurring proteins (cy statins) and engineered to stably display two loops which create a binding surface, the PD-L1 binding AFFIMER® polypeptides of the present disclosure provide a number of advantages over antibodies, antibody fragments and other non-antibody binding proteins.
One is the small size of the AFFIMER® polypeptide itself. In its monomeric form it is about 14 kDa, or 1/lOth the size of an antibody. This small size gives greater potential for increased tissue penetration, particularly in poorly vascularized and/or fibrotic target tissues (like tumors).
AFFIMER® polypeptides have a simple protein structures (versus multi-domain antibodies), and as the AFFIMER® polypeptides do not require disulfide bonds or other post- translational modifications for function, many of the format embodiments including these polypeptides can be manufactured in prokaryotic and eukaryotic systems. The ability to utilize libraries of AFFIMER® polypeptides (such as the phage display techniques described in the appended examples) as well as site directed mutagenesis, the AFFIMER® polypeptides can be generated with tuneable binding kinetics with ideal ranges for therapeutic uses. For instance, the AFFIMER® polypeptides can have high affinity for PD-L1, such as single digit nanomolar or lower KD for monomeric AFFIMER® polypeptides and picomolar KD and avidity in multi- valent formats. The AFFIMER® polypeptides can be generated with tight binding kinetics for PD-L1, such as slow Koff rates in the 104 to 105 (s-1) range which benefits target tissue localization.
The PD-L1 binding AFFIMER® polypeptides of the present disclosure include AFFIMER® polypeptides with exquisite selectivity.
Moreover, the PD-L1 binding AFFIMER® polypeptides can be readily formatted, allowing formats such as Fc fusions, whole antibody fusions and in-line multimers to be generated and manufactured with ease.
The lack of need for disulfide bonds and post-translational modifications also permit many embodiments of proteins including the PD-L1 binding AFFIMER® polypeptides (or monomeric AFFIMER® polypeptide) to be delivered therapeutically by expression of gene delivery constructs that are introduced into the tissues of the patient, including formats where the protein is delivered systemically (such as expression from muscle tissue) or delivered locally (such as through intraturmoral gene delivery).
AFFIMER® Polypeptides
The term “stefin polypeptide” refers to a sub-group of proteins in the cystatin superfamily, a family which encompasses proteins that contain multiple cystatin-like sequences.
The stefin sub-group of the cystatin family is relatively small (around 100 amino acids) single domain proteins. They receive no known post-translational modification, and lack disulphide bonds, suggesting that they will be able to fold identically in a wide range of extra- and intracellular environments. Stefin A itself is a monomeric, single chain, single domain protein of 98 amino acids. The structure of Stefin A has been solved, facilitating the rational mutation of Stefin A into the AFFIMER® Scaffold. The only known biological activity of cystatins is the inhibition of cathepsin activity, which allowed us to exhaustively test for residual biological activity of our engineered proteins.
The term “AFFIMER” (or “AFFIMER® Scaffold” or “AFFIMER® Polypeptide”) refers to small, highly stable proteins that are a recombinantly engineered variants of Stefin Polypeptides. AFFIMER® proteins display two peptide loops and an N-terminal sequence that can all be randomized to bind to desired target proteins with high affinity and specificity, in a similar manner to monoclonal antibodies. Stabilization of the two peptides by the Stefin protein scaffold constrains the possible conformations that the peptides can take, increasing the binding affinity and specificity compared to libraries of free peptides. These engineered non-antibody binding proteins are designed to mimic the molecular recognition characteristics of monoclonal antibodies in different applications. Variations to other parts of the Stefin polypeptide can be carried out, with such variations improving the properties of these affinity reagents, such as increase stability, make them robust across a range of temperatures and pH and the like. Preferably the AFFIMER® polypeptide includes a sequence derived from stefin A, sharing substantial identify with a stefin A wild type sequence, such as human Stefin A. It will be apparent to a person skilled in the art that modifications may be made to the scaffold sequence without departing from the disclosure. In particular, an AFFIMER® Scaffold can have an amino acid sequences that is at least 25%, 35%, 45%, 55% or 60% identity to the corresponding sequences to human Stefin A, preferably at least 70%, at least 80%, at least 85%, at least 90%, at least 92%, at least 94%, at least 95% identical, e.g., where the sequence variations do not adversely affect the ability of the scaffold to bind to the desired target (such as PD-L1), and e.g., which do not restore or generate biological functions such as those which are possessed by wild type stefin A but which are abolished in mutational changes described herein.
“Programmed death-ligand 1”, also known as “PD-L1”, “cluster of differentiation 274”, “CD274”, “B7 homolog 1” or “B7-H1”, refers a protein that, in the case of humans, is encoded by the CD274 gene. The human PD-L1 is a 40kDa type 1 transmembrane protein that plays a major role in suppressing the immune system under different circumstances. A representative human PD-L1 sequence is provided by UniProtKB Primary accession number Q9NZQ7 and will include other human isoforms thereof. PD-L1 binds to its receptor, PD-1, found on activated T cells, B cells, and myeloid cells, to modulate activation or inhibition. PD-L1 also has an appreciable affinity for the costimulatory molecule CD80 (B7-1). Engagement of PD-L1 with its receptor PD-1 (“Programmed cell death protein 1” or “CD279”) on T cells delivers a signal that inhibits TCR-mediated activation of IL-2 production and T cell proliferation. In this regard, PD- L1 is considered a checkpoint, and its upregulated expression in tumors contributes to inhibition of T-cell mediated antitumor responses. While PD-L1 will be used generally with reference to PD-L1 from various mammalian species, it will be understood throughout the application that any reference to PD-L1 includes human PD-L1 and is, preferably, referring to human PD-L1 per se. TGF Trap
By "TGF RII" or "TGF Receptor II" is meant a polypeptide having the wild-type human TGF Receptor Type 2 Isoform A sequence (e.g., the amino acid sequence of NCBI Reference Sequence (RefSeq) Accession No. NP_001020018 (SEQ ID NO: 100)), or a polypeptide having the wild-type human TGF Receptor Type 2 Isoform B sequence (e.g., the amino acid sequence of NCBI RefSeq Accession No. NP_003233 (SEQ ID NO: 101)) or having a sequence substantially identical the amino acid sequence of SEQ ID NO: 100 or of SEQ ID NO: 101. The TGF RII may retain at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99% of the TGF -binding activity of the wild-type sequence. The polypeptide of expressed TGF RII lacks the signal sequence.
Human TGF RII Isoform A Precursor Polypeptide (NCBI RefSeq Accession No: NP_001020018)
MGRGLLRGLWPLHIVLWTRIASTIPPHVQKSDVEMEAQKDEI ICPSCNRTAHPLRHINNDMIVT DNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCS ITSICEKPQEVCVAVWRKNDENITLETVCHD PKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNI IFSEEYNTSNPDLLLVIFQV TGISLLPPLGVAISVIIIFYCYRVNRQQKLSSTWETGKTRKLMEFSEHCAI ILEDDRSDISSTC ANNINHNTELLPIELDTLVGKGRFAEVYKAKLKQNTSEQFETVAVKIFPYEEYASWKTEKDIFS DINLKHENILQFLTAEERKTELGKQYWLITAFHAKGNLQEYLTRHVI SWEDLRKLGSSLARGIA HLHSDHTPCGRPKMPIVHRDLKSSNILVKNDLTCCLCDFGLSLRLDPTLSVDDLANSGQVGTAR YMAPEVLESRMNLENVESFKQTDVYSMALVLWEMTSRCNAVGEVKDYEPPFGSKVREHPCVESM KDNVLRDRGRPEIPSFWLNHQGIQMVCETLTECWDHDPEARLTAQCVAERFSELEHLDRLSGRS CSEEKIPEDGSLNTTK (SEQ ID NO: 100)
Human TGF RII Isoform B Precursor Polypeptide (NCBI RefSeq Accession No: NPJ303233
MGRGLLRGLWPLHIVLWTRIASTIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQ KSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKK PGETFFMCSCSSDECNDNIIFSEEYNTSNPDLLLVIFQVTGI SLLPPLGVAISVIIIFYCYRVN RQQKLSSTWETGKTRKLMEFSEHCAI ILEDDRSDISSTCANNINHNTELLPIELDTLVGKGRFA EVYKAKLKQNTSEQFETVAVKIFPYEEYASWKTEKDIFSDINLKHENILQFLTAEERKTELGKQ YWLITAFHAKGNLQEYLTRHVISWEDLRKLGS SLARGIAHLHSDHTPCGRPKMPIVHRDLKSSN ILVKNDLTCCLCDFGLSLRLDPTLSVDDLANSGQVGTARYMAPEVLESRMNLENVESFKQTDVY SMALVLWEMTSRCNAVGEVKDYEPPFGSKVREHPCVESMKDNVLRDRGRPEIPSFWLNHQGIQM VCETLTECWDHDPEARLTAQCVAERFSELEHLDRLSGRS CSEEKIPEDGSLNTTK (SEQ ID NO: 101)
By a "fragment of TGF RII capable of binding TGF " is meant any portion of NCBI RefSeq Accession No. NP_001020018 (SEQ ID NO: 100) or of NCBI RefSeq Accession No. NP_003233 (SEQ ID NO: 101), or a sequence substantially identical to SEQ ID NO: 100 or SEQ ID NO: 101 that is at least 20 ( e.g ., at least 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 175, or 200) amino acids in length that retains at least some of the TGF -binding activity (e.g., at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99%) of the wild-type receptor or of the corresponding wild-type fragment. Typically, such fragment is a soluble fragment.
In some embodiments, the TGF-b ligand binding domain of a TGF b receptor comprises SEQ ID NO: 102, SEQ ID NO: 103 or SEQ ID NO: 104, or portion thereof, or variant thereof (as described above):
LCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFIL EDAASPKCIMKEKKKPGETFFMCSCSSDECNDNI IF (SEQ ID NO 102)
LCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPY HDFILEDAASPTCIMKEKKKPGETFFMCSCSSDECNDNI IF (SEQ ID NO 103)
ALQCFCHLCTKDNFTCVTDGLCFVSVTETTDKVIHNSMCIAEIDLIPRDRPFVCAPSSKTGSVT TTYCCNQDHCNKIEL (SEQ ID NO 104)
An example of such fragment is a TOEbEII extra-cellular domain having the sequence of SEQ ID NO: 105.
IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCS ITSICEKPQEVCVAV WRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNI IFSE EYNTSNPD (SEQ ID NO: 105).
In some embodiments, the TGF-b ligand binding domain of a TGF b receptor comprises a sequence of the TGF-b type I receptor ectodomain, or portion of ectodomain, for example:
GVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKKFDSSRDRNKPFKFMLGKQEVIRGWEEGVA QMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLKLE (SEQ ID NO: 106); or
EDPSLDRPFISEGTTLKDLIYDMTTSGSGSGLPLLVQRTIARTIVLQESIGKGRFGEVWRGKWR GEEVAVKIFSSREERSWFREAEIYQTVMLRHENILGFIAADNKDNGTWTQLWLVSDYHEHGSLF DYLNRYTVTVEGMIKLALSTASGLAHLHMEIVGTQGKPAIAHRDLKSKNILVKKNGTCCIADLG LAVRHDSATDTIDIAPNHRVGTKRYMAPEVLDDS INMKHFESFKRADIYAMGLVFWEIARRCSI GGIHEDYQLPYYDLVPSDPSVEEMRKVVCEQKLRPNIPNRWQSCEALRVMAKIMRECWYANGAA RLTALRIKKTLSQLSQQEGIKM (SEQ ID NO: 107)
In some embodiments, the TGF-b ligand binding domain of a TGF b receptor comprises a sequence of the TbK-III-Eϋ, or portion of thereof, such as the ectodomain:
MTSHYVIAIFALMSSCLATAGPEPGALCELSPVSASHPVQALMESFTVLSGCASRGTTGLPQEV HVLNLRTAGQGPGQLQREVTLHLNPISSVHIHHKSVVFLLNSPHPLVWHLKTERLATGVSRLFL VSEGSVVQFSSANFSLTAETEERNFPHGNEHLLNWARKEYGAVTSFTELKIARNI YIKVGEDQV FPPKCNIGKNFLSLNYLAEYLQPKAAEGCVMSSQPQNEEVHI IELITPNSNPYSAFQVDITIDI RPSQEDLEVVKNLILILKCKKSVNWVIKSFDVKGSLKI IAPNSIGFGKESERSMTMTKSIRDDI PSTQGNLVKWALDNGYSPITSYTMAPVANRFHLRLENNEEMGDEEVHTIPPELRILLDPGALPA LQNPPIRGGEGQNGGLPFPFPDISRRVWNEEGEDGLPRPKDPVIPS IQLFPGLREPEEVQGSVD IALSVKCDNEKMIVAVEKDSFQASGYSGMDVTLLDPTCKAKMNGTHFVLESPLNGCGTRPRWSA LDGVVYYNSIVIQVPALGDSSGWPDGYEDLESGDNGFPGDMDEGDASLFTRPEIVVFNCSLQQV RNPSSFQEQPHGNITFNMELYNTDLFLVPSQGVFSVPENGHVYVEVSVTKAEQELGFAIQTCFI SPYSNPDRMSHYTIIENICPKDESVKFYSPKRVHFP IPQADMDKKRFSFVFKPVFNTSLLFLQC ELTLCTKMEKHPQKLPKCVPPDEACTSLDAS IIWAMMQNKKTFTKPLAVIHHEAESKEKGPSMK EPNPISPPIFHGLDTLT (SEQ ID NO: 108).
Polypeptides
The terms "polypeptide" and "peptide" and "protein" are used interchangeably herein and refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides containing at least one analogs of an amino acid (including, for example, unnatural amino acids), as well as other modifications known in the art.
The terms "amino acid residue" and "amino acid" are used interchangeably and means, in the context of a polypeptide, an amino acid that is participating in one more peptide bonds of the polypeptide. In general, the abbreviations used herein for designating the amino acids are based on recommendations of the IUPAC-IUB Commission on Biochemical Nomenclature (see Biochemistry (1972) 11:1726-1732). For instance, Met, lie, Leu, Ala and Gly represent "residues" of methionine, isoleucine, leucine, alanine and glycine, respectively. By the residue is meant a radical derived from the corresponding a-amino acid by eliminating the OH portion of the carboxyl group and the H portion of the a-amino group. The term "amino acid side chain" is that part of an amino acid exclusive of the — CH(NH2)COOH portion, as defined by K. D. Kopple, "Peptides and Amino Acids", W. A. Benjamin Inc., New York and Amsterdam, 1966, pages 2 and 33.
For the most part, the amino acids used in the application of this disclosure are those naturally occurring amino acids found in proteins, or the naturally occurring anabolic or catabolic products of such amino acids which contain amino and carboxyl groups. Particularly suitable amino acid side chains include side chains selected from those of the following amino acids: glycine, alanine, valine, cysteine, leucine, isoleucine, serine, threonine, methionine, glutamic acid, aspartic acid, glutamine, asparagine, lysine, arginine, proline, histidine, phenylalanine, tyrosine, and tryptophan, and those amino acids and amino acid analogs which have been identified as constituents of peptidylglycan bacterial cell walls. Amino acid residues having “basic sidechains” include Arg, Lys and His. Amino acid residues having “acidic sidechains” include Glu and Asp. Amino acid residues having “neutral polar sidechains” include Ser, Thr, Asn, Gin, Cys and Tyr. Amino acid residues having “neutral non-polar sidechains” include Gly, Ala, Val, lie, Leu, Met, Pro, Trp and Phe. Amino acid residues having “non-polar aliphatic sidechains” include Gly, Ala, Val, He and Leu. Amino acid residues having “hydrophobic sidechains” include Ala, Val, lie, Leu, Met, Phe, Tyr and Trp. Amino acid residues having “small hydrophobic sidechains” include Ala and Val. Amino acid residues having “aromatic sidechains” include Tyr, Trp and Phe.
The term amino acid residue further includes analogs, derivatives and congeners of any specific amino acid referred to herein, as for instance, the subject AFFIMER® polypeptides (particularly if generated by chemical synthesis) can include an amino acid analog such as, for example, cyanoalanine, canavanine, djenkolic acid, norleucine, 3-phosphoserine, homoserine, dihydroxy-phenylalanine, 5-hydroxytryptophan, 1-methylhistidine, 3-methylhistidine, diaminiopimelic acid, ornithine, or diaminobutyric acid. Other naturally occurring amino acid metabolites or precursors having side chains which are suitable herein will be recognized by those skilled in the art and are included in the scope of the present disclosure.
Also included are the (D) and (L) stereoisomers of such amino acids when the structure of the amino acid admits of stereoisomeric forms. The configuration of the amino acids and amino acid residues herein are designated by the appropriate symbols (D), (L) or (DL), furthermore when the configuration is not designated the amino acid or residue can have the configuration (D), (L) or (DL). It will be noted that the structure of some of the compounds of this disclosure includes asymmetric carbon atoms. It is to be understood accordingly that the isomers arising from such asymmetry are included within the scope of this disclosure. Such isomers can be obtained in substantially pure form by classical separation techniques and by sterically controlled synthesis. For the purposes of this application, unless expressly noted to the contrary, a named amino acid shall be construed to include both the (D) or (L) stereoisomers.
The terms "identical" or percent "identity" in the context of two or more polynucleotides or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The percent identity may be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software that may be used to obtain alignments of amino acid or nucleotide sequences are well-known in the art. These include, but are not limited to, BLAST, ALIGN, Megalign, BestFit, GCG Wisconsin Package, and variants thereof. In some embodiments, two polynucleotides or polypeptides of the disclosure are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using a sequence comparison algorithm or by visual inspection. In some embodiments, identity exists over a region of the amino acid sequences that is at least about 10 residues, at least about 20 residues, at least about 40-60 residues, at least about 60-80 residues in length or any integral value there between. In some embodiments, identity exists over a longer region than 60-80 residues, such as at least about 80- 100 residues, and in some embodiments the sequences are substantially identical over the full length of the sequences being compared, such as the coding region of a target protein or an antibody. In some embodiments, identity exists over a region of the nucleotide sequences that is at least about 10 bases, at least about 20 bases, at least about 40-60 bases, at least about 60-80 bases in length or any integral value there between. In some embodiments, identity exists over a longer region than 60-80 bases, such as at least about 80-1000 bases or more, and in some embodiments the sequences are substantially identical over the full length of the sequences being compared, such as a nucleotide sequence encoding a protein of interest.
A "conservative amino acid substitution" is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been generally defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). For example, substitution of a phenylalanine for a tyrosine is a conservative substitution. Generally, conservative substitutions in the sequences of the polypeptides, soluble proteins, and/or antibodies of the disclosure do not abrogate the binding of the polypeptide, soluble protein, or antibody containing the amino acid sequence, to the target binding site. Methods of identifying amino acid conservative substitutions which do not eliminate binding are well-known in the art.
A polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is "isolated" is a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is in a form not found in nature. Isolated polypeptides, soluble proteins, antibodies, polynucleotides, vectors, cells, or compositions include those which have been purified to a degree that they are no longer in a form in which they are found in nature. In some embodiments, a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is isolated is substantially pure.
The term "substantially pure" as used herein refers to material which is at least 50% pure (i.e., free from contaminants), at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure.
The term "fusion protein" or "fusion polypeptide" as used herein refers to a hybrid protein expressed by a polynucleotide molecule comprising nucleotide sequences of at least two genes.
The term "linker" or "linker region" as used herein refers to a linker inserted between a first polypeptide ( e.g ., copies of an AFFIMER® polypeptide) and a second polypeptide (e.g., another AFFIMER® polypeptide, an Fc domain, a ligand binding domain, etc.). In some embodiments, the linker is a peptide linker. Linkers should not adversely affect the expression, secretion, or bioactivity of the polypeptides. Preferably, linkers are not antigenic and do not elicit an immune response.
As use herein, the term "specifically binds to" or is "specific for" refers to measurable and reproducible interactions such as binding between a target and an AFFIMER® polypeptide, antibody or other binding partner, which is determinative of the presence of the target in the presence of a heterogeneous population of molecules including biological molecules. For example, an AFFIMER® polypeptide that specifically binds to a target is an AFFIMER® polypeptide that binds this target with greater affinity, avidity (if multimeric formatted), more readily, and/or with greater duration than it binds to other targets.
Polynucleotides
The terms "polynucleotide" refers to polymers of nucleotides of any length and include DNA and RNA. The nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase.
As used herein, the term "polynucleotide encoding" refers to the order or sequence of nucleotides along a strand of deoxyribopolynucleotide deoxyribonucleotides. The order of these deoxyribonucleotides determines the order of amino acids along the polypeptide (protein) chain. Thus, a polynucleotide sequence encoding the amino acid sequence.
When used in reference to nucleotide sequences, "sequence" as used herein, the term grammatical and other forms may comprise DNA or RNA and may be single or double stranded. Nucleic acid sequences may be mutated. Nucleic acid sequence may have any length, for example 2 to 000,000 or more nucleotides (or any integral value above or between) a polynucleotide, for example a length of from about 100 to about 10,000, or from about 200 nucleotides to about 500 nucleotides.
The term "vector" as used herein means a construct, which is capable of delivering, and usually expressing, at least one gene(s) or sequence(s) of interest in a host cell. Examples of vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid, or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, and DNA or RNA expression vectors encapsulated in liposomes.
As used herein, the term "transfection" refers to an exogenous polynucleotide into a eukaryotic cell. Transfection can be achieved by various means known in the art, including calcium phosphate -DNA co-precipitation, DEAE- dextran-mediated transfection, polybrene- mediated transfection, electroporation, microinjection, liposome fusion, lipofection, protoplast fusion, retroviral infection, and biolistics technology (biolistics).
The term "carrier" as used herein is an isolated polynucleotide comprising the isolated polynucleotide can be used to deliver a composition to the interior of the cell. It is known in the art a number of carriers including, but not limited to the linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses. Thus, the term "vector" includes an autonomously replicating plasmid or vims. The term should also be construed to include facilitate transfer of polynucleotide into cells of the non-plasmid and non- viral compounds, for example, polylysine compounds, liposomes, and the like. Examples of viral vectors include, but are not limited to adenoviral vectors, adeno-associated vims vectors, retroviral vectors and the like.
As used herein, the term "expression vector" refers to a vector comprising a recombinant polynucleotide comprising expression control sequence and a nucleotide sequence to be expressed operably linked. The expression vector comprises sufficient cis-acting elements (ex acting elements) used for expression; other elements for expression can be supplied by the host cell or in vitro expression system. Expression vectors include all those known in the art, such as cosmids, plasmids (e.g., naked or contained in liposomes) and viruses (e.g., lentivims, retrovimses, adenovimses and adeno-associated vimses).
As used herein, the term "operably linked" refers to functional linkage between the regulatory sequence and a heterologous polynucleotide sequence is connected to a connection results in the expression of the latter. For example, when the first polynucleotide sequence and a second polynucleotide sequence is a functional relationship between the first polynucleotide sequence and the second polynucleotide sequence is operably linked. For example, if the promoter affects the transcription or expression of the coding sequence, the promoter is operably linked to a coding sequence. Typically, DNA sequencing operably linked are contiguous, and to join two protein coding regions in the same reading frame as necessary.
As used herein, the term "promoter" is defined as a promoter DNA sequence recognized by the synthetic machinery required for the synthesis machinery of the cell specific transcription of a polynucleotide sequence or introduced.
The term "constitutive expression" as used herein refers to all expressed under physiological conditions.
The term "inducible expression" as used herein refers to expression under certain conditions, such as activation (or inactivation) of an intracellular signaling pathway or the contacting of the cells harboring the expression construct with a small molecule that regulates the expression (or degree of expression) of a gene operably linked to an inducible promoter sensitive to the concentration of the small molecule.
The term "electroporation" refers to the use of a transmembrane electric field pulse to induce microscopic pathways (pores) in a bio-membrane; their presence allows biomolecules such as plasmids or other oligonucleotide to pass from one side of the cellular membrane to the other.
Checkpoint Inhibitors, Co-stimulatory Agonists and Chemotherapeutics
A "checkpoint molecule" refers to proteins that are expressed by tissues and/or immune cells and reduce the efficacy of an immune response in a manner dependent on the level of expression of the checkpoint molecule. When these proteins are blocked, the “brakes” on the immune system are released and, for example, T cells are able to kill cancer cells more effectively. Examples of checkpoint proteins found on T cells or cancer cells include PD-l/PD- L1 and CTLA-4/B7-1/B7-2, PD-L2, NKG2A, KIR, LAG-3, TIM-3, CD96, VISTA and TIGIT.
A "checkpoint inhibitor" refers to a drug entity that reverses the immunosuppressive signaling from a checkpoint molecule.
A "costimulatory molecule" refers to an immune cell such as a T cell cognate binding partner which specifically binds to costimulatory ligands thereby mediating co- stimulation, such as, but not limited to proliferation. Costimulatory molecules are cell surface molecules other than the antigen receptor or ligand which facilitate an effective immune response. Co-stimulatory molecules include, but are not limited to MHCI molecules, BTLA receptor and Toll ligands, and 0X40, CD27, CD28, CDS, ICAM-1, LFA-1 (CDlla / CD18), ICOS (CD278) and 4-1BB (CD137). Examples of costimulatory molecules include but are not limited to: CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46,
CD 160, CD19, CD4, CD8a, CD8p, IL2Rp , IL2Ry, IL7Ra, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CDlld, ITGAE, CD103, IT GAL, CDlla, LFA- 1, ITGAM, CD lib, ITGAX, CD 11c, ITGB1 , CD29, ITGB2, CD 18, LFA-1, ITGB7, NKG2D, NKG2C, TNFR2, TRANCE / RANKL, DNAM1 (CD226), SLAMF4 (CD244,2B4), CD84, CD96 (Tactile), CEACAM1, CRT AM, Ly9 (CD229) , CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Lyl08), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS , SLP-76, PAG / Cbp, CD19a, and CD83 ligand.
A "costimulatory agonists" refers to a drug entity that activates (agonizes) the costimulatory molecule, such as costimulatory ligand would do, and produces an immuno stimulatory signal or otherwise increases the potency or efficacy of an immune response.
A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclophosphamide (CYTOXAN); alkyl sulfonates such as busulfan, improsulfan, and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); delta-9-tetrahydrocannabinol (dronabinol, MARINOL); beta-lapachone; lapachol; colchicines; betulinic acid; a camptothecin (including the synthetic analogue topotecan (HYCAMTIN), CPT-11 (irinotecan, CAMPTOSAR), acetylcamptothecin, scopolectin, and 9- aminocamptothecin); bryostatin; pemetrexed; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); podophyllo toxin; podophyllinic acid; teniposide; cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; TLK- 286; CDP323, an oral alpha-4 integrin inhibitor; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlomaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e. g., calicheamicin, especially calicheamicin gammall and calicheamicin omegall (see, e.g., Nicolaou et ah, Angew. Chem Inti. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; an esperamicin; as well as neocarzino statin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunombicin, detombicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including ADRIAMYCIN®, morpholino-doxombicin, cyanomorpholino-doxombicin, 2-pyrrolino- doxorubicin, doxorubicin HC1 liposome injection (DOXIL®) and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate, gemcitabine (GEMZAR®), tegafur (UFTORAL®), capecitabine (XELODA®), an epothilone, and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, and imatinib (a 2- phenylaminopyrimidine derivative), as well as other c-Kit inhibitors; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; 2-ethylhydrazide; procarbazine; PSK polysaccharide complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine (ELDISINE®, FILDESI®); dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); thiotepa; taxoids, e.g., paclitaxel (TAXOL®), albumin-engineered nanoparticle formulation of paclitaxel (ABRAXANE®), and doxetaxel (TAXOTERE®); chloranbucil; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine (VELBAN®); platinum; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine (ONCOVIN®); oxaliplatin; leucovovin; vinorelbine (NAVELBINE®); novantrone; edatrexate; daunomycin; aminopterin; ibandronate; topoisomerase inhibitor RFS 2000; difluorometlhylomithine (DMFO); retinoids such as retinoic acid; pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone, and FOLFOX, an abbreviation for a treatment regimen with oxaliplatin (ELOXATIN®) combined with 5-FU and leucovovin.
Also included in this definition are anti-hormonal agents that act to regulate, reduce, block, or inhibit the effects of hormones that can promote the growth of cancer, and are often in the form of systemic, or whole-body treatment. They may be hormones themselves. Examples include anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX® tamoxifen), raloxifene (EVISTA®), droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (FARESTON®); anti-progesterones; estrogen receptor down-regulators (ERDs); estrogen receptor antagonists such as fulvestrant (FASLODEX®); agents that function to suppress or shut down the ovaries, for example, leutinizing hormone-releasing hormone (LHRH) agonists such as leuprolide acetate (LUPRON® and ELIGARD®), goserelin acetate, buserelin acetate and tripterelin; anti-androgens such as flutamide, nilutamide and bicalutamide; and aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, megestrol acetate (MEGASE®), exemestane (AROMASIN®), formestanie, fadrozole, vorozole (RIVISOR®), letrozole (FEMARA®), and anastrozole (ARIMIDEX®). In addition, such definition of chemotherapeutic agents includes bisphosphonates such as clodronate (for example, BONEFOS® or OSTAC®), etidronate (DIDROCAL), NE-58095, zoledronic acid/zoledronate (ZOMETA®), alendronate (FOSAMAX®), pamidronate (AREDIA®), tiludronate (SKELID®), or risedronate (ACTONEL®); as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); anti-sense oligonucleotides, particularly those that inhibit expression of genes in signaling pathways implicated in abherant cell proliferation, such as, for example, PKC-alpha, Raf, H-Ras, and epidermal growth factor receptor (EGF-R); vaccines such as THERATOP®E vaccine and gene therapy vaccines, for example, ALLOVECTIN® vaccine, LEUVECTIN® vaccine, and VAXID® vaccine; topoisomerase 1 inhibitor ( e.g ., LURTOTECAN®); an anti-estrogen such as fulvestrant; a Kit inhibitor such as imatinib or EXEL-0862 (a tyrosine kinase inhibitor); EGFR inhibitor such as erlotinib or cetuximab; an anti-VEGF inhibitor such as bevacizumab; arinotecan; rmRH (e.g., ABARELIX®); lapatinib and lapatinib ditosylate (an ErbB-2 and EGFR dual tyrosine kinase small-molecule inhibitor also known as GW572016); 17AAG (geldanamycin derivative that is a heat shock protein (Hsp) 90 poison), and pharmaceutically acceptable salts, acids or derivatives of any of the above.
As used herein, the term "cytokine" refers generically to proteins released by one cell population that act on another cell as intercellular mediators or have an autocrine effect on the cells producing the proteins. Examples of such cytokines include lymphokines, monokines; interleukins ("ILs") such as IL-1, IL-la, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL10, IL- 11, IL-12, IL-13, IL-15, IL-17A-F, IL-18 to IL-29 (such as IL-23), IL-31, including PROLEUKIN rIL-2; a tumor-necrosis factor such as TNF-a or TNF-b, TGF- i-3; and other polypeptide factors including leukemia inhibitory factor ("LIF"), ciliary neurotrophic factor ("CNTF"), CNTF-like cytokine ("CLC"), cardiotrophin ("CT"), and kit ligand ("KL"). As used herein, the term "chemokine" refers to soluble factors (e.g., cytokines) that have the ability to selectively induce chemotaxis and activation of leukocytes. They also trigger processes of angiogenesis, inflammation, wound healing, and tumorigenesis. Example chemokines include IL-8, a human homolog of murine keratinocyte chemoattractant (KC).
Treatments
The term "dysfunctional", as used herein, also includes refractory or unresponsive to antigen recognition, specifically, impaired capacity to translate antigen recognition into down stream T-cell effector functions, such as proliferation, cytokine production (e.g., IL-2) and/or target cell killing.
The term "anergy" refers to the state of unresponsiveness to antigen stimulation resulting from incomplete or insufficient signals delivered through the T-cell receptor (e.g. increase in intracellular Ca+2 in the absence of ras-activation). T cell anergy can also result upon stimulation with antigen in the absence of co-stimulation, resulting in the cell becoming refractory to subsequent activation by the antigen even in the context of costimulation. The unresponsive state can often be overridden by the presence of Interleukin-2. Anergic T-cells do not undergo clonal expansion and/or acquire effector functions.
The term "exhaustion" refers to T cell exhaustion as a state of T cell dysfunction that arises from sustained TCR signaling that occurs during many chronic infections and cancer. It is distinguished from anergy in that it arises not through incomplete or deficient signaling, but from sustained signaling. It is defined by poor effector function, sustained expression of inhibitory receptors and a transcriptional state distinct from that of functional effector or memory T cells. Exhaustion prevents optimal control of infection and tumors.
"Enhancing T-cell function" means to induce, cause or stimulate a T-cell to have a sustained or amplified biological function, or renew or reactivate exhausted or inactive T-cells. Examples of enhancing T-cell function include: increased secretion of □ -interferon from CD8+ T-cells, increased proliferation, increased antigen responsiveness (e.g., viral, pathogen, or tumor clearance) relative to such levels before the intervention. In some embodiments, the level of enhancement is as least 50%, alternatively 60%, 70%, 80%, 90%, 100%, 120%, 150%, 200%. The manner of measuring this enhancement is known to one of ordinary skill in the art.
A "T cell dysfunctional disorder" is a disorder or condition of T-cells characterized by decreased responsiveness to antigenic stimulation. In a particular embodiment, a T-cell dysfunctional disorder is a disorder that is specifically associated with inappropriate increased levels of PD-1. A T-cell dysfunctional disorder can also be associated with inappropriate increased levels of PD-L1 in the tumor which gives rise to suppression of T-cell antitumor function(s). In another embodiment, a T-cell dysfunctional disorder is one in which T-cells are anergic or have decreased ability to secrete cytokines, proliferate, or execute cytolytic activity. In a specific aspect, the decreased responsiveness results in ineffective control of a pathogen or tumor expressing an immunogen. Examples of T cell dysfunctional disorders characterized by T- cell dysfunction include unresolved acute infection, chronic infection and tumor immunity.
"Tumor immunity" refers to the process in which tumors evade immune recognition and clearance. Thus, as a therapeutic concept, tumor immunity is "treated" when such evasion is attenuated, and the tumors are recognized and attacked by the immune system. Examples of tumor recognition include tumor binding, tumor shrinkage and tumor clearance.
"Sustained response" refers to the sustained effect on reducing tumor growth after cessation of a treatment. For example, the tumor size may remain to be the same or smaller as compared to the size at the beginning of the administration phase. In some embodiments, the sustained response has a duration at least the same as the treatment duration, at least 1.5x, 2. Ox, 2.5x, or 3. Ox length of the treatment duration.
The terms "cancer" and "cancerous" as used herein refer to or describe the physiological condition in mammals in which a population of cells are characterized by unregulated cell growth. Examples of cancer include, but are not limited to, carcinoma, blastoma, sarcoma, and hematologic cancers such as lymphoma and leukemia.
The terms "tumor" and "neoplasm" as used herein refer to any mass of tissue that results from excessive cell growth or proliferation, either benign (noncancerous) or malignant (cancerous) including pre-cancerous lesions. Tumor growth is generally uncontrolled and progressive, does not induce or inhibit the proliferation of normal cells. Tumor can affect a variety of cells, tissues or organs, including but not limited to selected from bladder, bone, brain, breast, cartilage, glial cells, esophagus, fallopian tube, gall bladder, heart, intestine, kidney, liver, lung, lymph node, neural tissue, ovary, pancreas, prostate, skeletal muscle, skin, spinal cord, spleen, stomach, testis, thymus, thyroid, trachea, urethra, ureter, urethra, uterus, vagina organ or tissue or the corresponding cells. Tumors include cancers, such as sarcoma, carcinoma, plasmacytoma or (malignant plasma cells). Tumors of the present disclosure, may include, but are not limited to leukemias ( e.g ., acute leukemia, acute lymphoblastic leukemia, acute myeloid leukemia, acute myeloid leukemia, acute promyelocytic leukemia, acute myeloid - monocytic leukemia, acute monocytic leukemia, acute leukemia, chronic leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, polycythemia vera), lymphomas (Hodgkin's disease, non-Hodgkin's disease), primary macroglobulinemia disease, heavy chain disease, and solid tumors such as sarcomas cancer (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, chordoma, endothelium sarcoma, lymphangiosarcoma, angiosarcoma, lymphangioendothelio sarcoma, synovioma vioma , mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer (including triple negative breast cancer), ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinoma, carcinoma, bronchogenic carcinoma, medullary carcinoma, renal cell carcinoma, hepatoma, Nile duct carcinoma, choriocarcinoma, spermatogonia Tumor, embryonal carcinoma, Wilms' tumor, cervical cancer, uterine cancer, testicular cancer, lung carcinoma (including small cell lung carcinoma and non- small cell lung carcinoma or NSCLC), bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, schwannoma, meningioma, melanoma, neuroblastoma, retinoblastoma), esophageal cancer, gallbladder , kidney cancer, multiple myeloma. Preferably, a "tumor" includes, but is not limited to: pancreatic cancer, liver cancer, lung cancer (including NSCLC), stomach cancer, esophageal cancer, head and neck squamous cell carcinoma, prostate cancer, colon cancer, breast cancer (including triple negative breast cancer), lymphoma, gallbladder cancer, renal cancer, leukemia, multiple myeloma, ovarian cancer, cervical cancer and glioma.
The term "metastasis" as used herein refers to the process by which a cancer spreads or transfers from the site of origin to other regions of the body with the development of a similar cancerous lesion at the new location. A "metastatic" or "metastasizing" cell is one that loses adhesive contacts with neighboring cells and migrates via the bloodstream or lymph from the primary site of disease to invade neighboring body structures.
The terms "cancer cell" and "tumor cell" refer to the total population of cells derived from a cancer or tumor or pre-cancerous lesion, including both non-tumorigenic cells, which comprise the bulk of the cancer cell population, and tumorigenic stem cells (cancer stem cells). As used herein, the terms "cancer cell" or "tumor cell" will be modified by the term "non- tumorigenic" when referring solely to those cells lacking the capacity to renew and differentiate to distinguish those tumor cells from cancer stem cells.
The term "effective amount" as used herein refers to an amount to provide therapeutic or prophylactic benefit.
As used herein, "complete response" or "CR" refers to disappearance of all target lesions; "partial response" or "PR" refers to at least a 30% decrease in the sum of the longest diameters (SLD) of target lesions, taking as reference the baseline SLD; and "stable disease" or "SD" refers to neither sufficient shrinkage of target lesions to qualify for PR, nor sufficient increase to qualify for PD, taking as reference the smallest SLD since the treatment started. As used herein, "progression free survival" (PFS) refers to the length of time during and after treatment during which the disease being treated ( e.g ., cancer) does not get worse. Progression-free survival may include the amount of time patients have experienced a complete response or a partial response, as well as the amount of time patients have experienced stable disease.
As used herein, "overall response rate" (ORR) refers to the sum of complete response (CR) rate and partial response (PR) rate.
As used herein, "overall survival" refers to the percentage of individuals in a group who are likely to be alive after a particular duration of time.
The term "treatment" as used herein refers to the individual trying to change the process or treatment of a clinical disease caused by intervention of a cell, may be either preventive intervention course of clinical pathology. Including but not limited to treatment to prevent the occurrence or recurrence of disease, alleviation of symptoms, reducing the direct or indirect pathological consequences of any disease, preventing metastasis, slow the rate of disease progression, amelioration or remission of disease remission or improved prognosis.
The term "subject" refers to any animal (e.g., a mammal), including, but not limited to, humans, non-human primates, canines, felines, rodents, and the like, which is to be the recipient of a particular treatment. Typically, the terms "subject" and "patient" are used interchangeably herein in reference to a human subject.
The terms "agonist" and "agonistic" as used herein refer agents that are capable of, directly or indirectly, substantially inducing, activating, promoting, increasing, or enhancing the biological activity of a target or target pathway. The term "agonist" is used herein to include any agent that partially or fully induces, activates, promotes, increases, or enhances the activity of a protein or other target of interest.
The terms "antagonist" and "antagonistic" as used herein refer to or describe an agent that is capable of, directly or indirectly, partially or fully blocking, inhibiting, reducing, or neutralizing a biological activity of a target and/or pathway. The term "antagonist" is used herein to include any agent that partially or fully blocks, inhibits, reduces, or neutralizes the activity of a protein or other target of interest.
The terms "modulation" and "modulate" as used herein refer to a change or an alteration in a biological activity. Modulation includes, but is not limited to, stimulating an activity or inhibiting an activity. Modulation may be an increase in activity or a decrease in activity, a change in binding characteristics, or any other change in the biological, functional, or immunological properties associated with the activity of a protein, a pathway, a system, or other biological targets of interest. The term "immune response" as used herein includes responses from both the innate immune system and the adaptive immune system. It includes both cell-mediated and/or humoral immune responses. It includes both T-cell and B-cell responses, as well as responses from other cells of the immune system such as natural killer (NK) cells, monocytes, macrophages, etc.
The term "pharmaceutically acceptable" refers to a substance approved or approvable by a regulatory agency of the Federal government or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans.
The terms "pharmaceutically acceptable excipient, carrier or adjuvant" or "acceptable pharmaceutical carrier" refer to an excipient, carrier or adjuvant that can be administered to a subject, together with at least one agent of the present disclosure, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic effect. In general, those of skill in the art and the U.S. FDA consider a pharmaceutically acceptable excipient, carrier, or adjuvant to be an inactive ingredient of any formulation.
The terms "effective amount" or "therapeutically effective amount" or "therapeutic effect" refer to an amount of a bispecific fusion protein described herein effective to "treat" a disease or disorder in a subject such as, a mammal. In the case of cancer or a tumor, the therapeutically effective amount of a bispecific fusion protein has a therapeutic effect and as such can boost the immune response, boost the anti-tumor response, increase cytolytic activity of immune cells, increase killing of tumor cells by immune cells, reduce the number of tumor cells; decrease tumorigenicity, tumorigenic frequency or tumorigenic capacity; reduce the number or frequency of cancer stem cells; reduce the tumor size; reduce the cancer cell population; inhibit or stop cancer cell infiltration into peripheral organs including, for example, the spread of cancer into soft tissue and bone; inhibit and stop tumor or cancer cell metastasis; inhibit and stop tumor or cancer cell growth; relieve to some extent at least one of the symptoms associated with the cancer; reduce morbidity and mortality; improve quality of life; or a combination of such effects.
The terms "treating" or "treatment" or "to treat" or "alleviating" or "to alleviate" refer to both (1) therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder and (2) prophylactic or preventative measures that prevent or slow the development of a targeted pathologic condition or disorder. Thus those in need of treatment include those already with the disorder; those prone to have the disorder; and those in whom the disorder is to be prevented. In the case of cancer or a tumor, a subject is successfully "treated" according to the methods of the present disclosure if the patient shows at least one of the following: an increased immune response, an increased anti-tumor response, increased cytolytic activity of immune cells, increased killing of tumor cells by immune cells, a reduction in the number of or complete absence of cancer cells; a reduction in the tumor size; inhibition of or an absence of cancer cell infiltration into peripheral organs including the spread of cancer cells into soft tissue and bone; inhibition of or an absence of tumor or cancer cell metastasis; inhibition or an absence of cancer growth; relief of at least one symptoms associated with the specific cancer; reduced morbidity and mortality; improvement in quality of life; reduction in tumorigenicity; reduction in the number or frequency of cancer stem cells; or some combination of effects.
PD-L1 Binding AFFIMER® Polypeptides
An AFFIMER® polypeptide is a scaffold based on stefin A, meaning that it has a sequence which is derived from stefin A, for example, a mammalian stefin A, for example, a human stefin A. Some aspects of the application provides AFFIMER® polypeptides which bind PD-L1 (also referred to as “anti-PD-Ll AFFIMER® polypeptides”) comprising an AFFIMER® in which at least one of the solvent accessible loops from the wild-type stefin A protein with amino acid sequences to provide an AFFIMER® polypeptide having the ability to bind PD-L1, preferably selectively, and preferably with Kd of 106M or less.
In some embodiments, the anti-PD-Ll AFFIMER® polypeptide is derived from the wild- type human stefin A protein having a backbone sequence and in which one or both of loop 2 [designated (Xaa)n] and loop 4 [designated (Xaa)m] are replaced with alternative loop sequences (Xaa)n and (Xaa)m , to have the general formula (i)
FRl-(Xaa)n-FR2-(Xaa)m-FR3 (I) wherein
FR1 is a polypeptide sequence represented by MIPGGLSEAK PATPEIQEIV DKVKPQLEEK TNETYGKLEA VQYKTQVLA (SEQ ID NO: 1) or a polypeptide sequence having at least 70% homology thereto;
FR2 is a polypeptide sequence represented by GTNYYIKVRA GDNKYMHLKV FKSL (SEQ ID NO: 2) or a polypeptide sequence having at least 70% homology thereto;
FR3 is a polypeptide sequence represented by EDLVLTGYQV DKNKDDELTG F (SEQ ID NO: 3) or a polypeptide sequence having at least 70% homology thereto; and
Xaa, individually for each occurrence, is an amino acid residue, n and m are each, independently, an integer from 3 to 20.
In some embodiments, FR1 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 1. In some embodiments, FR1 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% identity with SEQ ID NO: 1; In some embodiments, FR2 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 2. In some embodiments, FR2 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% identity with SEQ ID NO: 2; In some embodiments, FR3 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% homology with SEQ ID NO: 3. In some embodiments, FR3 is a polypeptide sequence having at least 80%, 85%, 90%, 95% or even 98% identity with SEQ ID NO: 3.
In some embodiments, the anti-PD-Ll AFFIMER® polypeptide has an amino acid sequence represented in the general formula:
MIP-Xaal-GLSEAKPATPEIQEIVDKVKPQLEEKTNETYGKLEAVQYKTQVLA- (Xaa)n-Xaa2-TNYYIKVRAGDNKYMHLKVF-Xaa3-Xaa4-Xaa5-(Xaa)m-Xaa6-D-Xaa7- VLTGY QVDKNKDDELTGF (SEQ ID NO: 4) wherein
Xaa, individually for each occurrence, is an amino acid residue; n and m are each, independently, an integer from 3 to 20; Xaal is Gly, Ala, Val, Arg, Lys, Asp, or Glu, more preferably Gly, Ala, Arg or Lys, and more even more preferably Gly or Arg; Xaa2 is Gly, Ala, Val, Ser or Thr, more preferably Gly or Ser; Xaa3 is Arg, Lys, Asn, Gin, Ser, Thr, more preferably Arg, Lys, Asn or Gin, and even more preferably Lys or Asn; Xaa4 is Gly, Ala, Val, Ser or Thr, more preferably Gly or Ser; Xaa5 is Ala, Val, lie, Leu, Gly or Pro, more preferably lie, Leu or Pro, and even more preferably Leu or Pro; Xaa6 is Gly, Ala, Val, Asp or Glu, more preferably Ala, Val, Asp or Glu, and even more preferably Ala or Glu; and Xaa7 is Ala, Val, He, Leu, Arg or Lys, more preferably He, Leu or Arg, and even more preferably Leu or Arg.
For instance, the anti-PD-Ll AFFIMER® polypeptide can have an amino acid sequence represented in the general formula:
MIPRGLSEAKPATPEIQEIVDKVKPQLEEKTNETYGKLEAVQYKTQVLA-(Xaa)n- STNYYIKVRAGDNKYMHLKVFNGP-(Xaa)m-ADRVLTGYQVDKNKDDELTGF (SEQ ID NO: 5), wherein Xaa, individually for each occurrence, is an amino acid residue; n and m are each, independently, an integer from 3 to 20.
In some embodiments, n is 3 to 15, 3 to 12, 3 to 9, 3 to 7, 5 to 7, 5 to 9, 5 to 12, 5 to 15, 7 to 12 or 7 to 9.
In some embodiments, m is 3 to 15, 3 to 12, 3 to 9, 3 to 7, 5 to 7, 5 to 9, 5 to 12, 5 to 15,
7 to 12 or 7 to 9.
In some embodiments, Xaa, independently for each occurrence, is an amino acid that can be added to a polypeptide by recombinant expression in a prokaryotic or eukaryotic cell, and even more preferably one of the 20 naturally occurring amino acids. In some embodiments of the above sequences and formulas, (Xaa)n is an amino acid sequence represented in the general formula (II)
-aal-aa2-aa3-Gly-Pro-aa4-aa5-Trp-aa6- (II) wherein aal represents an amino acid residue with a basic sidechain, more preferably Lys, Arg or His, and even more preferably Lys or Arg; aa2 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain, more preferably a small aliphatic sidechain, a neutral polar side chain or a basic or acid side chain, even more preferably Ala, Pro, he, Gin, Thr, Asp, Glu, Lys, Arg or His, and even more preferably Ala, Gin, Asp or Glu; aa3 represents an amino acid residue with an aromatic or basic sidechain, preferably Phe, Tyr, Trp, Lys, Arg or His, more preferably Phe, Tyr, Trp, and even more preferably His or Tyr, Trp or His; aa4 represents an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain; preferably a neutral polar sidechain or a charged (acidic or basic) sidechain; more preferably Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, and even more preferably Gin, Lys, Arg, His, Asp or Glu; aa5 represents an amino acid residue with a neutral polar or a charged (acidic or basic) or a small aliphatic or an aromatic sidechain; preferably a neutral polar sidechain or a charged sidechain; more preferably Ser, Thr, Asn, Gin, Asp, Glu, Arg or His, and even more preferably Ser, Asn, Gin, Asp, Glu or Arg; and aa6 represents an amino acid residue with an aromatic or acid sidechain, preferably Phe, Tyr, Trp, Asp or Glu; more preferably Trp or Asp; and even more preferably Trp.
In some embodiments of the above sequences and formulas, (Xaa)n is an amino acid sequence represented in the general formula (III)
-aal-aa2-aa3-Phe-Pro-aa4-aa5-Phe-Trp- (III) wherein aal represents an amino acid residue with a basic sidechain or aromatic sidechain, preferably Lys, Arg, His, Ser, Thr, Asn or Gin, more preferably Lys, Arg, His, Asn or Gin, and even more preferably Lys or Asn; aa2 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain, more preferably a small aliphatic sidechain, a neutral polar side chain or a basic or acid side chain, even more preferably Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, and even more preferably Ala, Gin, Asp or Glu; aa3 represents an amino acid residue with an aromatic or basic sidechain, preferably Phe, Tyr, Trp, Lys, Arg or His, more preferably Phe, Tyr, Trp or His, and even more preferably Tyr, Trp or His; aa4 represents an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain; preferably a neutral polar sidechain or a charged (acidic or basic) sidechain; more preferably Ala, Pro, lie, Gin, Thr, Asp, Glu, Lys, Arg or His, and even more preferably Gin, Lys, Arg, His, Asp or Glu; and aa5 represents an amino acid residue with a neutral polar or a charged (acidic or basic) or a small aliphatic or an aromatic sidechain; preferably a neutral polar sidechain or a charged sidechain; more preferably Ser, Thr, Asn, Gin, Asp, Glu, Arg or His, and even more preferably Ser, Asn, Gin, Asp, Glu or Arg.
In some embodiments of the above sequences and formulas, (Xaa)n is an amino acid sequence selected from SEQ ID NOS: 6 to 40, or an amino acid sequence having at least 80%, 85%, 90%, 95% or even 98% homology with a sequence selected from SEQ ID NOS: 6 to 41. In some embodiments, (Xaa)n is an amino acid sequence having at least 80%, 85%, 90%, 95% or even 98% identity with a sequence selected from SEQ ID NO: 6 to 41.
Table 2. Loop 2 Sequences
Figure imgf000039_0001
Figure imgf000040_0001
In some embodiments of the above sequences and formulas, (Xaa)m is an amino acid sequence represented in the general formula (IV)
-aa7-aa8-aa9-aal0-aal l-aal2-aal3-aal4-aal5- (IV) wherein aa7 represents an amino acid residue with neutral polar or non-polar sidechain or an acidic sidechain; preferably Gly, Ala, Val, Pro, Trp, Gin, Ser, Asp or Glu, and even more preferably Gly, Ala, Trp, Gin, Ser, Asp or Glu; aa8 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain or aromatic sidechain, more preferably a charged (acidic or basic) sidechain, more preferably Asp, Glu, Lys, Arg, His, Gin, Ser, Thr, Asn, Ala, Val, Pro, Gly, Tyr or Phe, and even more preferably Asp, Glu, Lys, Arg, His or Gin; aa9 represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain or aromatic sidechain, more preferably a neutral polar side chain or an acid side chain, more preferably Gin, Ser, Thr, Asn, Asp, Glu, Arg, Lys, Gly, Leu, Pro or Tyr, and even more preferably Gin, Thr or Asp; aalO represents an amino acid residue, preferably an amino acid residue with a neutral polar or non-polar sidechain or a charged (acidic or basic) sidechain or aromatic sidechain, more preferably a neutral polar side chain or a basic or acid side chain, more preferably Asp, Glu, Arg, His, Lys, Ser, Gin, Asn, Ala, Leu, Tyr, Trp, Pro or Gly, and even more preferably Asp, Glu, His, Gin, Asn, Leu, Trp or Gly; aal 1 represents an amino acid residue, preferably an amino acid residue with a neutral polar sidechain or a charged (acidic or basic) sidechain or a nonpolar aliphatic sidechain or an aromatic sidechain, more preferably a neutral polar side chain or a basic or acid side chain, more preferably Asp, Glu, Ser, Thr, Gin, Arg, Lys, His, Val, he, Tyr or Gly and even more preferably Asp, Glu, Ser, Thr, Gin, Lys or His; aal 2 represents an amino acid residue, preferably an amino acid residue with a neutral polar sidechain or a charged (acidic or basic) sidechain or a nonpolar aliphatic sidechain or an aromatic sidechain, more preferably a an acid side chain, more preferably Asp, Glu, Ser, Thr, Gin, Asn, Lys, Arg, Val, Leu, lie, Trp, Tyr, Phe or Gly and even more preferably Asp, Glu, Ser, Tyr, Trp, Arg or Lys; aal 3 represents an amino acid residue, preferably an amino acid residue with a neutral polar sidechain or a charged (acidic or basic) sidechain or a nonpolar aliphatic sidechain or an aromatic sidechain, more preferably a an acid side chain, more preferably Ser, Thr, Gin, Asn, Val, lie, Leu, Gly, Pro, Asp, Glu, His, Arg, Trp, Tyr or Phe and even more preferably Ser, Thr, Gin, Asn, Val, He, Leu, Gly, Asp or Glu; aal 4 represents an amino acid residue, preferably an amino acid residue with a neutral polar sidechain or a charged (acidic or basic) sidechain, more preferably Ala, He, Trp, Pro, Asp, Glu, Arg, Lys, His, Ser, Thr, Gin or Asn and even more preferably Ala, Pro, Asp, Glu, Arg, Lys, Ser, Gin or Asn; and aal 5 represents an amino acid residue, preferably an amino acid residue with a neutral polar or neutral non-polar sidechain or a charged (acidic or basic) sidechain, more preferably His, Arg, Lys, Asp, Ser, Thr, Gin, Asn, Ala, Val, Leu, Gly or Phe and even more preferably His, Arg, Lys, Asp, Ser, Thr, Gin or Asn.
In some embodiments of the above sequences and formulas, (Xaa)m is an amino acid sequence selected from SEQ ID NOS: 42 to 77, or an amino acid sequence having at least 80%, 85%, 90%, 95% or even 98% homology with a sequence selected from SEQ ID NOS: 42 to 77. In some embodiments, (Xaa)m is an amino acid sequence having at least 80%, 85%, 90%, 95% or even 98% identity with a sequence selected from SEQ ID NO: 42 to 77.
Table 3. Loop 4 Sequences
Figure imgf000041_0001
Figure imgf000042_0001
In some embodiments, the anti-PD-Ll AFFIMER® polypeptide has an amino acid sequence selected from SEQ ID NOS: 78 to 86, or an amino acid sequence having at least 70%, 75% 80%, 85%, 90%, 95% or even 98% homology with a sequence selected from SEQ ID NOS: 78 to 86. In some embodiments, the anti-PD-Ll AFFIMER® polypeptide has an amino acid sequence having at least 70%, 75% 80%, 85%, 90%, 95% or even 98% identity with a sequence selected from SEQ ID NO: 78 to 86. Table 4. Examples of anti-PD-Ll AFFIMER® Polypeptide Sequences
Figure imgf000043_0001
In some embodiments, the anti-PD-Fl AFFIMER® polypeptide has an amino acid sequence that is encoded by a polynucleotide having a coding sequence corresponding to nucleotides 1-336 of one of SEQ ID NOS: 87 to 95, or an amino acid sequence that can be encoded by a polynucleotide having a coding sequence at least 70%, 75% 80%, 85%, 90%, 95% or even 98% identical with nucleotides 1-336 of one of SEQ ID NOS: 87 to 95, or an amino acid sequence that can be encoded by a polynucleotide having a coding sequence that hybridizes nucleotides 1-336 of one of SEQ ID NOS: 87 to 95 under stringent conditions (such as in the presence of 6X sodium chloride/sodium citrate (SSC) at 45°C followed by a wash in 0.2X SSC at 65°C.
Table 5. Examples of anti-PD-Ll AFFIMER® Polypeptide Coding Sequences
Figure imgf000044_0001
Figure imgf000045_0001
Furthermore, minor modifications may also include small deletions or additions - beyond the loop 2 and loop 4 inserts described above - to the stefin A or stefin A derived sequences disclosed herein, such as addition or deletion of up to 10 amino acids relative to stefin A or the stefin A derived AFFIMER® polypeptide.
In some embodiments of the bispecific fusion protein, the PD-L1 binding AFFIMER® polypeptide portion that binds human PD-L1 as a monomer with a dissociation constant (KD) of about 1 mM or less, about 100 nM or less, about 40 nM or less, about 20 nM or less, about 10 nM or less, about 1 nM or less, or about 0.1 nM or less.
In some embodiments of the bispecific fusion protein, the PD-L1 binding AFFIMER® polypeptide portion binds human PD-L1 as a monomer with an off rate constant (K0ff), such as measured by Biacore, of about 103 s 1 (i.e., unit of 1/second) or slower; of about 104 s 1 or slower or even of about 105 s 1 or slower.
In some embodiments of the bispecific fusion protein, the PD-L1 binding AFFIMER® polypeptide portion binds human PD-L1 as a monomer with an association constant (Kon), such as measured by Biacore, of at least about 103 M V1 or faster; at least about 104M 1s 1 or faster; at least about 105 M V1 or faster; or even at least about 106 M V1 or faster.
In some embodiments of the bispecific fusion protein, the PD-L1 binding AFFIMER® polypeptide portion binds human PD-L1 as a monomer with an IC50 in a competitive binding assay with human PD-1 of 1 mM or less, about 100 nM or less, about 40 nM or less, about 20 nM or less, about 10 nM or less, about 1 nM or less, or about 0.1 nM or less. In some embodiments, the bispecific fusion protein has a melting temperature (Tm, i.e., temperature at which both the folded and unfolded states are equally populated) of 65 °C or higher, and preferably at least 70°C, 75°C, 80°C or even 85°C or higher. Melting temperature is a particularly useful indicator of protein stability. The relative proportions of folded and unfolded proteins can be determined by many techniques known to the skilled person, including differential scanning calorimetry, UV difference spectroscopy, fluorescence, circular dichroism (CD), and NMR (Pace et al. (1997) "Measuring the conformational stability of a protein" in Protein structure: A practical approach 2: 299-321).
Fusions Proteins - General
In order for the bispecific fusion protein to be secreted, it will generally contain a signal sequence that directs the transport of the protein to the lumen of the endoplasmic reticulum and ultimately to be secreted (or retained on the cell surface if a transmembrane domain or other cell surface retention signal). Signal sequences (also referred to as signal peptides or leader sequences) are located at the N-terminus of nascent polypeptides. They target the polypeptide to the endoplasmic reticulum and the proteins are sorted to their destinations, for example, to the inner space of an organelle, to an interior membrane, to the cell outer membrane, or to the cell exterior via secretion. Most signal sequences are cleaved from the protein by a signal peptidase after the proteins are transported to the endoplasmic reticulum. The cleavage of the signal sequence from the polypeptide usually occurs at a specific site in the amino acid sequence and is dependent upon amino acid residues within the signal sequence.
In some embodiments, the signal peptide is about 5 to about 40 amino acids in length (such as about 5 to about 7, about 7 to about 10, about 10 to about 15, about 15 to about 20, about 20 to about 25, or about 25 to about 30, about 30 to about 35, or about 35 to about 40 amino acids in length).
In some embodiments, the signal peptide is a native signal peptide from a human protein. In other embodiments, the signal peptide is a non-native signal peptide. For example, in some embodiments, the non-native signal peptide is a mutant native signal peptide from the corresponding native secreted human protein, and can include at least one (such as 2, 3, 4, 5, 6,
7, 8, 9, or 10 or more) substitutions insertions or deletions.
In some embodiments, the signal peptide is a signal peptide or mutant thereof from a non- IgSF protein family, such as a signal peptide from an immunoglobulin (such as IgG heavy chain or IgG-kappa light chain), a cytokine (such as interleukin -2 (IL-2), or CD33), a serum albumin protein ( e.g . HSA or albumin), a human azurocidin preprotein signal sequence, a luciferase, a trypsinogen ( e.g . chymotrypsinogen or trypsinogen) or other signal peptide able to efficiently secrete a protein from a cell. Examples of signal peptides include, but are not limited to:
Table 6. Examples of Signal Sequences
Figure imgf000047_0001
In some embodiments of a secreted bispecific fusion protein, the recombinant polypeptide comprises a signal peptide when expressed, and the signal peptide (or a portion thereof) is cleaved from the bispecific fusion protein upon secretion.
The subject fusion proteins may also include at least one linkers separating heterologous protein sequences or domains, i.e., separating the PD-L1 binding AFFIMER® polypeptide sequence from the TGF trap polypeptide from each other or from other polypeptides also present in the fusion protein (such as Fc domains or other domains that enhance serum half-life, provide multimerization, etc.).
As used herein, the term "linker" refers to a linker amino acid sequence inserted between a first polypeptide ( e.g ., an AFFIMER® polypeptide) and a second polypeptide (e.g., a second AFFIMER® polypeptide, an Fc region, a TGF receptor trap, albumin, etc.). Empirical linkers designed by researchers are generally classified into 3 categories according to their structures: flexible linkers, rigid linkers, and in vivo cleavable linkers. Besides the basic role in linking the functional domains together (as in flexible and rigid linkers) or releasing free functional domain in vivo (as in in vivo cleavable linkers), linkers may offer many other advantages for the production of fusion proteins, such as improving biological activity, increasing expression yield, and achieving desirable pharmacokinetic profiles. Linkers should not adversely affect the expression, secretion, or bioactivity of the fusion protein. Linkers should not be antigenic and should not elicit an immune response.
Suitable linkers are known to those of skill in the art and often include mixtures of glycine and serine residues and often include amino acids that are sterically unhindered. Other amino acids that can be incorporated into useful linkers include threonine and alanine residues. Linkers can range in length, for example from 1-50 amino acids in length, 1-22 amino acids in length, 1-10 amino acids in length, 1-5 amino acids in length, or 1-3 amino acids in length. In some embodiments, the linker may comprise a cleavage site. In some embodiments, the linker may comprise an enzyme cleavage site, so that the second polypeptide may be separated from the first polypeptide.
In some embodiments, the linker can be characterized as flexible. Flexible linkers are usually applied when the joined domains require a certain degree of movement or interaction. They are generally composed of small, non-polar (e.g. Gly) or polar (e.g. Ser or Thr) amino acids. See, for example, Argos P. (1990) “An investigation of oligopeptides linking domains in protein tertiary structures and possible candidates for general gene fusion” J Mol Biol. 211:943- 958. The small size of these amino acids provides flexibility and allows for mobility of the connecting functional domains. The incorporation of Ser or Thr can maintain the stability of the linker in aqueous solutions by forming hydrogen bonds with the water molecules, and therefore reduces the unfavorable interaction between the linker and the protein moieties. The most commonly used flexible linkers have sequences consisting primarily of stretches of Gly and Ser residues (“GS” linker). An example of the most widely used flexible linker has the sequence of (Gly-Gly-Gly-Gly-Ser)n (SEQ ID NO: 174). By adjusting the copy number “n”, the length of this GS linker can be optimized to achieve appropriate separation of the functional domains, or to maintain necessary inter-domain interactions. Besides the GS linkers, many other flexible linkers have been designed for recombinant fusion proteins. As These flexible linkers are also rich in small or polar amino acids such as Gly and Ser but can contain additional amino acids such as Thr and Ala to maintain flexibility, as well as polar amino acids such as Lys and Glu to improve solubility.
In some embodiments, the linker can be characterized as rigid. While flexible linkers have the advantage to connect the functional domains passively and permitting certain degree of movements, the lack of rigidity of these linkers can be a limitation in certain fusion protein embodiments, such as in expression yield or biological activity. The ineffectiveness of flexible linkers in these instances was attributed to an inefficient separation of the protein domains or insufficient reduction of their interference with each other. Under these situations, rigid linkers have been successfully applied to keep a fixed distance between the domains and to maintain their independent functions.
Many natural linkers exhibited a-helical structures. The a-helical structure was rigid and stable, with intra-segment hydrogen bonds and a closely packed backbone. Therefore, the stiff a- helical linkers can act as rigid spacers between protein domains. George et al. (2002) “An analysis of protein domain linkers: their classification and role in protein folding” Protein Eng. 15(11):871-9. In general, rigid linkers exhibit relatively stiff structures by adopting a-helical structures or by containing multiple Pro residues. Under many circumstances, they separate the functional domains more efficiently than the flexible linkers. The length of the linkers can be easily adjusted by changing the copy number to achieve an optimal distance between domains. As a result, rigid linkers are chosen when the spatial separation of the domains is critical to preserve the stability or bioactivity of the fusion proteins. In this regard, alpha helix-forming linkers with the sequence of (EAAAK)n (SEQ ID NO: 146) have been applied to the construction of many recombinant fusion proteins. Another type of rigid linkers has a Pro-rich sequence, (XP)n, with X designating any amino acid, preferably Ala, Lys, or Glu.
Merely to illustrate, examples of linkers include:
Table 7. Examples of Linkers
Figure imgf000049_0001
Figure imgf000050_0001
Other linkers that may be used in the subject fusion proteins include, but are not limited to, SerGly, GGSG (SEQ ID NO: 175), GSGS (SEQ ID NO: 176), GGGS (SEQ ID NO: 177), S(GGS)n (SEQ ID NO: 178) where n is 1-7, GRA, poly(Gly), poly(Ala), GGGSGGG (SEQ ID NO: 157), ESGGGGVT (SEQ ID NO: 158), LESGGGGVT (SEQ ID NO: 159), GRAQVT (SEQ ID NO: 160), WRAQVT (SEQ ID NO: 161), and ARGRAQVT (SEQ ID NO: 162). The hinge regions of the Fc fusions described below may also be considered linkers.
Engineering PK and ADME Properties
In some embodiment, the bispecific fusion protein may not have a half-life and/or PK profile that is optimal for the route of administration, such as parenteral therapeutic dosing. The term “half-life” refers to the amount of time it takes for a substance, such as a bispecific fusion protein of the present disclosure, to lose half of its pharmacologic or physiologic activity or concentration. Biological half-life can be affected by elimination, excretion, degradation ( e.g ., enzymatic) of the substance, or absorption and concentration in certain organs or tissues of the body. In some embodiments, biological half-life can be assessed by determining the time it takes for the blood plasma concentration of the substance to reach half its steady state level (“plasma half-life”). To address this shortcoming, there are a variety of general strategies for prolongation of half-life that have been used in the case of other protein therapeutics, including the incorporation of half-life extending moieties as part of the bispecific fusion protein.
The term “half-life extending moiety” refers to a pharmaceutically acceptable moiety, domain, or molecule covalently linked (“conjugated” or “fused”) to the AFFIMER® polypeptide to form the bispecific fusion proteins described herein, optionally via a non-naturally encoded amino acid, directly or via a linker, that prevents or mitigates in vivo proteolytic degradation or other activity-diminishing modification of the AFFIMER® polypeptide, increases half-life, and/or improves or alters other pharmacokinetic or biophysical properties including but not limited to increasing the rate of absorption, reducing toxicity, improving solubility, reducing protein aggregation, increasing biological activity and/or target selectivity of the modified AFFIMER® polypeptide, increasing manufacturability, and/or reducing immunogenicity of the modified AFFIMER® polypeptide, compared to a comparator such as an unconjugated form of the modified AFFIMER® polypeptide. The term “half-life extending moiety” includes non- pro teinaceous, half-life extending moieties, such as a water soluble polymer such as polyethylene glycol (PEG) or discrete PEG, hydroxyethyl starch (HES), a lipid, a branched or unbranched acyl group, a branched or unbranched C8-C30 acyl group, a branched or unbranched alkyl group, and a branched or unbranched C8-C30 alkyl group; and proteinaceous half-life extending moieties, such as serum albumin, transferrin, adnectins ( e.g . , albumin-binding or pharmacokinetics extending (PKE) adnectins), Fc domain, and unstructured polypeptide, such as XTEN and PAS polypeptide (e.g. conformationally disordered polypeptides composed of the amino acids Pro, Ala, and/or Ser), and a fragment of any of the foregoing. An examination of the crystal structure of an AFFIMER® polypeptide and its interaction with its target, such as the anti-PD-Ll AFFIMER® polypeptide complex with PD-1 shown in the FIGS., can indicate which certain amino acid residues have side chains that are fully or partially accessible to solvent.
In some embodiments, the half-life extending moiety extends the half-life of the resulting bispecific fusion protein circulating in mammalian blood serum compared to the half-life of the protein that is not so conjugated to the moiety (such as relative to the AFFIMER® polypeptide alone). In some embodiments, half-life is extended by greater than or greater than about 1.2-fold, 1.5-fold, 2.0-fold, 3.0-fold, 4.0-fold., 5.0-fold, or 6.0-fold. In some embodiments, half-life is extended by more than 6 hours, more than 12 hours, more than 24 hours, more than 48 hours, more than 72 hours, more than 96 hours or more than 1 week after in vivo administration compared to the protein without the half-life extending moiety.
As means for further exemplification, half-life extending moieties that can be used in the generation of bispecific fusion proteins of the disclosure include:
Genetic fusion of the pharmacologically AFFIMER® polypeptide sequence to a naturally long-half-life protein or protein domain (e.g., Fc fusion, transferrin [Tf] fusion, or albumin fusion. See, for example, Beck et al. (2011) “Therapeutic Fc-fusion proteins and peptides as successful alternatives to antibodies. MAbs. 3:1-2; Czajkowsky et al. (2012) “Fc-fusion proteins: new developments and future perspectives. EMBO Mol Med. 4:1015-28; Huang et al. (2009) “Receptor-Fc fusion therapeutics, traps, and Mimetibody technology” Curr Opin Biotechnol. 2009;20:692-9; Keefe et al. (2013) “Transferrin fusion protein therapies: acetylcholine receptor-transferrin fusion protein as a model. In: Schmidt S, editor fusion protein technologies for biopharmaceuticals: applications and challenges. Hoboken: Wiley; p. 345-56; Weimer et al. (2013) “Recombinant albumin fusion proteins. In: Schmidt S, editor fusion protein technologies for biopharmaceuticals: applications and challenges. Hoboken: Wiley;
2013. p. 297-323; Walker et al. (2013) “Albumin-binding fusion proteins in the development of novel long-acting therapeutics. In: Schmidt S, editor fusion protein technologies for biopharmaceuticals: applications and challenges. Hoboken: Wiley; 2013. p. 325-43.
Genetic fusion of the pharmacologically AFFIMER® polypeptide sequence to an inert polypeptide, e.g., XTEN (also known as recombinant PEG or “rPEG”), a homoamino acid polymer (HAP; HAPylation), a proline-alanine-serine polymer (PAS; PASylation), or an elastin- like peptide (ELP; ELPylation). See, for example, Schellenberger et al. (2009) “A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner. Nat Biotechnol. 2009;27: 1186-90; Schlapschy et al. Fusion of a recombinant antibody fragment with a homo-amino-acid polymer: effects on biophysical properties and prolonged plasma half-life. Protein Eng Des Sel. 2007;20:273-84; Schlapschy (2013) PASylation: a biological alternative to PEGylation for extending the plasma half-life of pharmaceutically active proteins. Protein Eng Des Sel. 26:489-501. Floss et al. (2012) “Elastin-like polypeptides revolutionize recombinant protein expression and their biomedical application. Trends Biotechnol. 28:37-45. Floss et al. “ELP-fusion technology for biopharmaceuticals. In: Schmidt S, editor fusion protein technologies for biopharmaceuticals: application and challenges. Hoboken: Wiley; 2013. p. 372- 98.
Increasing the hydrodynamic radius by chemical conjugation of the pharmacologically active peptide or protein to repeat chemical moieties, e.g., to PEG (PEGylation) or hyaluronic acid. See, for example, Caliceti et al. (2003) “Pharmacokinetic and biodistribution properties of poly(ethylene glycol)-protein conjugates” Adv Drug Delivery Rev. 55:1261-77; Jevsevar et al. (2010) PEGylation of therapeutic proteins. Biotechnol J 5:113-28; Kontermann (2009) “Strategies to extend plasma half-lives of recombinant antibodies” BioDrugs. 23:93-109; Kang et al. (2009) “Emerging PEGylated drugs” Expert Opin Emerg Drugs. 14:363-80; and Mero et al. (2013) “Conjugation of hyaluronan to proteins” Carb Polymers. 92:2163-70.
Significantly increasing the negative charge of fusing the pharmacologically active peptide or protein by polysialylation; or, alternatively, (b) fusing a negatively charged, highly sialylated peptide (e.g., carboxy-terminal peptide [CTP; of chorionic gonadotropin (CG) b- chain]), known to extend the half-life of natural proteins such as human CG b-subunit, to the biological drug candidate. See, for example, Gregoriadis et al. (2005) “Improving the therapeutic efficacy of peptides and proteins: a role for polysialic acids” Int J Pharm. 2005; 300:125-30; Duijkers et al. “Single dose pharmacokinetics and effects on follicular growth and serum hormones of a long-acting recombinant FSH preparation (FSHCTP) in healthy pituitary- suppressed females” (2002) Hum Reprod. 17:1987-93; and Fares et al. “Design of a longacting follitropin agonist by fusing the C-terminal sequence of the chorionic gonadotropin beta subunit to the follitropin beta subunit” (1992) Proc Natl Acad Sci USA. 89:4304-8. 35; and Fares “Half- life extension through O-glycosylation.
Binding non-covalently, via attachment of a peptide or protein-binding domain to the bioactive protein, to normally long-half-life proteins such as HSA, human IgG, transferrin or fibronectin. See, for example, Andersen et al. (2011) “Extending half-life by indirect targeting of the neonatal Fc receptor (FcRn) using a minimal albumin binding domain” J Biol Chem. 286:5234-41; O’Connor-Semmes et al. (2014) “GSK2374697, a novel albumin-binding domain antibody (albudAb), extends systemic exposure of extendin-4: first study in humans — PK/PD and safety” Clin Pharmacol Ther. 2014;96:704-12. Sockolosky et al. (2014) “Fusion of a short peptide that binds immunoglobulin G to a recombinant protein substantially increases its plasma half-life in mice” PLoS One. 2014;9:el02566.
Classical genetic fusions to long-lived serum proteins offer an alternative method of half- life extension distinct from chemical conjugation to PEG or lipids. Two major proteins have traditionally been used as fusion partners: antibody Fc domains and human serum albumin (HSA). Fc fusions involve the fusion of peptides, proteins or receptor exodomains to the Fc portion of an antibody. Both Fc and albumin fusions achieve extended half-lives not only by increasing the size of the peptide drug, but both also take advantage of the body’s natural recycling mechanism: the neonatal Fc receptor, FcRn. The pH-dependent binding of these proteins to FcRn prevents degradation of the fusion protein in the endosome. Fusions based on these proteins can have half-lives in the range of 3-16 days, much longer than typical PEGylated or lipidated peptides. Fusion to antibody Fc domains can improve the solubility and stability of the peptide or protein drug. An example of a peptide Fc fusion is dulaglutide, a GLP-1 receptor agonist currently in late-stage clinical trials. Human serum albumin, the same protein exploited by the fatty acylated peptides is the other popular fusion partner. Albiglutide is a GLP-1 receptor agonist based on this platform. A major difference between Fc and albumin is the dimeric nature of Fc versus the monomeric structure of HSA leading to presentation of a fused peptide as a dimer or a monomer depending on the choice of fusion partner. The dimeric nature of an AFFIMER® polypeptide-Fc fusion can produce an avidity effect if the AFFIMER® target, such as PD-L1 on tumor cells, are spaced closely enough together or are themselves dimers. This may be desirable or not depending on the target.
Fc Fusions
In some embodiments, the AFFIMER® polypeptide may be part of a fusion protein with an immunoglobulin Fc domain ("Fc domain"), or a fragment or variant thereof, such as a functional Fc region. In this context, an Fc fusion (“Fc-fusion”), such as a bispecific fusion protein created as an AFFIMER® polypeptide-Fc fusion protein, is a polypeptide comprising at least one AFFIMER® sequences covalently linked through a peptide backbone (directly or indirectly) to an Fc region of an immunoglobulin. An Fc-fusion may comprise, for example, the Fc region of an antibody (which facilitates effector functions and pharmacokinetics) and an AFFIMER® polypeptide sequence as part of the same polypeptide. An immunoglobulin Fc region may also be linked indirectly to at least one AFFIMER® polypeptides. Various linkers are known in the art and can optionally be used to link an Fc to a polypeptide including an AFFIMER® polypeptide sequence to generate an Fc-fusion. In some embodiments, Fc-fusions can be dimerized to form Fc-fusion homodimers, or using non-identical Fc domains, to form Fc- fusion heterodimers.
There are several reasons for choosing the Fc region of human antibodies for use in generating the subject bispecific fusion proteins as AFFIMER® fusion proteins. The principle rationale is to produce a stable protein, large enough to demonstrate a similar pharmacokinetic profile compared with those of antibodies, and to take advantage of the properties imparted by the Fc region; this includes the salvage neonatal FcRn receptor pathway involving FcRn- mediated recycling of the fusion protein to the cell surface post endocytosis, avoiding lysosomal degradation and resulting in release back into the bloodstream, thus contributing to an extended serum half-life. Another obvious advantage is the Fc domain’s binding to Protein A, which can simplify downstream processing during production of the bispecific fusion protein and permit generation of highly pure preparation of the bispecific fusion protein.
In general, an Fc domain will include the constant region of an antibody excluding the first constant region immunoglobulin domain. Thus, Fc domain refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, and the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains. For IgA and IgM Fc may include the J chain. For IgG, Fc comprises immunoglobulin domains Cyl and Cy3 and the hinge between Cy 1 and Cy2. Although the boundaries of the Fc domain may vary, the human IgG heavy chain Fc region is usually defined to comprise residues C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU index as set forth in Rabat (Rabat et ah, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, NIH, Bethesda, Md. (1991)). Fc may refer to this region in isolation, or this region in the context of a whole antibody, antibody fragment, or Fc fusion protein. Polymorphisms have been observed at a number of different Fc positions and are also included as Fc domains as used herein.
In some embodiments, the Fc As used herein, a “functional Fc region” refers to an Fc domain or fragment thereof which retains the ability to bind FcRn. A functional Fc region binds to FcRn but does not possess effector function. The ability of the Fc region or fragment thereof to bind to FcRn can be determined by standard binding assays known in the art. Examples of "effector functions" include Clq binding; complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors ( e.g ., B cell receptor; BCR), etc. Such effector functions can be assessed using various assays known in the art for evaluating such antibody effector functions.
In some embodiments, the Fc domain is derived from an IgGl subclass, however, other subclasses (e.g., IgG2, IgG3, and IgG4) may also be used. An example of a sequence of a human IgGl immunoglobulin Fc domain which can be used is:
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFN WYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKAL PAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESN GQPENN YKTTPP VLDS DGS FFLY S KLT VDKS RW QQGN VFS C S VMHE ALHNH YT QKSLSLSPGK (SEQ ID NO: 131)
In some embodiments, the Fc region used in the fusion protein may comprise the hinge region of an Fc molecule. An example hinge region comprises the core hinge residues spanning positions 1-16 (i.e., DKTHTCPPCPAPELLG (SEQ ID NO: 163) of the example human IgGl immunoglobulin Fc domain sequence provided above. In some embodiments, the AFFIMER® polypeptide-containing fusion protein may adopt a multimeric structure (e.g., dimer) owing, in part, to the cysteine residues at positions 6 and 9 within the hinge region of the example human IgGl immunoglobulin Fc domain sequence provided above. In other embodiments, the hinge region as used herein, may further include residues derived from the CHI and CH2 regions that flank the core hinge sequence of the example human IgGl immunoglobulin Fc domain sequence provided above. In yet other embodiments, the hinge sequence may comprise or consist of GS THT CPPCP APELLG (SEQ ID NO: 164) or EPKSCDKTHTCPPCPAPELLG (SEQ ID NO: 165).
In some embodiments, the hinge sequence may include at least one substitution that confer desirable pharmacokinetic, biophysical, and/or biological properties. Some example hinge sequences include:
EPKSCDKTHTCPPCPAPELLGGPS (SEQ ID NO: 179; EPKS S DKTHT CPPCP APELLGGPS (SEQ ID NO: 166); EPKS S DKTHT CPPCP APELLGGS S (SEQ ID NO: 167);
EPKS S GSTHTCPPCPAPELLGGS S (SEQ ID NO: 168); DKTHTCPPCPAPELLGGPS (SEQ ID NO: 169) and DKTHT CPPCP APELLGGS S (SEQ ID NO: 170). In some embodiments, the residue P at position 18 of the example human IgGl immunoglobulin Fc domain sequence provided above may be replaced with S to ablate Fc effector function; this replacement is exemplified in hinges having the sequences EPKS SDKTHTCPPCPAPELLGGS S (SEQ ID NO: 167), EPKSSGSTHTCPPCPAPELLGGSS (SEQ ID NO: 168), and DKTHTCPPCPAPELLGGSS (SEQ ID NO: 170). In another embodiment, the residues DK at positions 1-2 of the example human IgGl immunoglobulin Fc domain sequence provided above may be replaced with GS to remove a potential clip site; this replacement is exemplified in the sequence EPKSSGSTHTCPPCPAPELLGGSS (SEQ ID NO: 168). In another embodiment, the C at the position 103 of the heavy chain constant region of human IgGl (i.e., domains CH1-CH3), may be replaced with S to prevent improper cysteine bond formation in the absence of a light chain; this replacement is exemplified by EPKSSDKTHTCPPCPAPELLGGPS (SEQ ID NO: 166), EPKS SDKTHTCPPCPAPELLGGS S (SEQ ID NO: 167), and EPKSSGSTHTCPPCPAPELLGGSS (SEQ ID NO: 168).
In some embodiments, the Fc is a mammalian Fc such as a human Fc, including Fc domains derived from IgGl, IgG2, IgG3 or IgG4. The Fc region may possess at least about 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with a native Fc region and/or with an Fc region of a parent polypeptide. In some embodiments, the Fc region may have at least about 90% sequence identity with a native Fc region and/or with an Fc region of a parent polypeptide.
In some embodiments, the Fc domain comprises an amino acid sequence selected from SEQ ID NO: 131, or an Fc sequence from the examples provided by SEQ ID NOS: 132-144. It should be understood that the C-terminal lysine of an Fc domain is an optional component of a fusion protein comprising an Fc domain. In some embodiments, the Fc domain comprises an amino acid sequence selected from SEQ ID NOS: 131-144, except that the C-terminal lysine thereof is omitted. In some embodiments, the Fc domain comprises the amino acid sequence of SEQ ID NO: 131. In some embodiments, the Fc domain comprises the amino acid sequence of SEQ ID NO: 131 except the C-terminal lysine thereof is omitted.
Table 8. Examples of Fc Domains
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
“Antibody-dependent cell-mediated cytotoxicity” or “ADCC” refers to a form of cytotoxicity in which secreted Ig bound onto Fc receptors (FcRs) present on certain cytotoxic cells ( e.g ., Natural Killer (NK) cells, neutrophils, and macrophages) enables these cytotoxic effector cells to bind specifically to an antigen-bearing target cell and subsequently kill the target cell with cytotoxins. In some embodiments, the fusion protein includes an Fc domain sequence for which the resulting bispecific fusion protein has no (or reduced) ADCC and/or complement activation or effector functionality. For example, the Fc domain may comprise a naturally disabled constant region of IgG2 or IgG4 isotype or a mutated IgGl constant region. Examples of suitable modifications are described in EP0307434. One example comprises the substitutions of alanine residues at positions 235 and 237 (EU index numbering).
In other embodiments, the fusion protein includes an Fc domain sequence for which the resulting bispecific fusion protein will retain some or all Fc functionality for example will be capable of one or both of ADCC and CDC activity, as for example if the fusion protein comprises the Fc domain from human IgGl or IgG3. Levels of effector function can be varied according to known techniques, for example by mutations in the CH2 domain, for example wherein the IgGl CH2 domain has at least one mutations at positions selected from 239 and 332 and 330, for example the mutations are selected from S239D and I332E and A330L such that the antibody has enhanced effector function, and/or for example altering the glycosylation profile of the antigen-binding protein of the disclosure such that there is a reduction in fucosylation of the Fc region.
Albumin fusion
In other embodiments, the bispecific fusion protein is a fusion protein comprising, in addition to at least one AFFIMER® polypeptide sequence, an albumin sequence or an albumin fragment. In other embodiments, the bispecific fusion protein is conjugated to the albumin sequence or an albumin fragment through chemical linkage other than incorporation into the polypeptide sequence including the AFFIMER® polypeptide. In some embodiments, the albumin, albumin variant, or albumin fragment is human serum albumin (HSA), a human serum albumin variant, or a human serum albumin fragment. Albumin serum proteins comparable to HSA are found in, for example, cynomolgus monkeys, cows, dogs, rabbits and rats. Of the non human species, bovine serum albumin (BSA) is the most structurally similar to HSA. See, e.g., Kosa et ah, (2007) J Pharm Sci. 96(11):3117-24. The present disclosure contemplates the use of albumin from non-human species, including, but not limited to, albumin sequence derived from cyno serum albumin or bovine serum albumin.
Mature HSA, a 585 amino acid polypeptide (approx. 67 kDa) having a serum half-life of about 20 days, is primarily responsible for the maintenance of colloidal osmotic blood pressure, blood pH, and transport and distribution of numerous endogenous and exogenous ligands. The protein has three structurally homologous domains (domains I, II and III), is almost entirely in the alpha-helical conformation, and is highly stabilized by 17 disulfide bridges. In some embodiments, the bispecific fusion protein can be an albumin fusion protein including at least one AFFIMER® polypeptides and the sequence for mature human serum albumin (SEQ ID NO: 145) or a variant or fragment thereof which maintains the PK and/or biodistribution properties of mature albumin to the extent desired in the fusion protein.
DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAKTCVADESA
ENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNECFLQHKDDNPNLPRLVRP
EVDVMCTAFHDNEETFLKKYLYEIARRHPYFYAPELLFFAKRYKAAFTECCQAADKAAC
LLPKLDELRDEGKASSAKQRLKCASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVT
DLTKVHTECCHGDLLECADDRADLAKYICENQDS ISSKLKECCEKPLLEKSHCIAEVEN
DEMPADLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLAKTYE
TTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGEYKFQNALLVRYTK
KVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAEDYLSVVLNQLCVLHEKTPVSDR
VTKCCTESLVNRRPCFSALEVDETYVPKEFNAETFTFHADICTLSEKERQIKKQTALVE
LVKHKPKATKEQLKAVMDDFAAFVEKCCKADDKETCFAEEGKKLVAASQAALGL
(SEQ ID NO: 145)
The albumin sequence can be set off from the AFFIMER® polypeptide sequence or other flanking sequences in the bispecific fusion protein by use of linker sequences as described above.
While unless otherwise indicated, reference herein to “albumin” or to “mature albumin” is meant to refer to HSA. However, it is noted that full-length HSA has a signal peptide of 18 amino acids (MKW VTFIS LLFLF S S AY S (SEQ ID NO: 109)) followed by a pro-domain of 6 amino acids (RGVFRR, SEQ ID NO: 171); this 24 amino acid residue peptide may be referred to as the pre-pro domain. The AFFIMER® polypeptide-HSA fusion proteins can be expressed and secreted using the HSA pre-pro-domain in the recombinant proteins coding sequence. Alternatively, the AFFIMER® polypeptide-HSA fusion can be expressed and secreted through inclusion of other secretion signal sequences, such as described above.
In alternative embodiments, rather than provided as part of a fusion protein with the AFFIMER® polypeptide, the serum albumin polypeptide can be covalently coupled to the AFFIMER® polypeptide through a bond other than a backbone amide bond, such as cross-linked through chemical conjugation between amino acid sidechains on each of the albumin polypeptide and the AFFIMER® polypeptide.
Albumin binding domain
In some embodiments, the bispecific fusion protein can include a serum-binding moiety - either as part of a fusion protein (if also a polypeptide) with the AFFIMER® polypeptide sequence or chemically conjugated through a site other than being part of a contiguous polypeptide chain.
In some embodiments, the serum-binding polypeptide is an albumin binding moiety. Albumin contains multiple hydrophobic binding pockets and naturally serves as a transporter of a variety of different ligands such as fatty acids and steroids as well as different drugs. Furthermore, the surface of albumin is negatively charged making it highly water-soluble.
The term “albumin binding moiety” as used herein refers to any chemical group capable of binding to albumin, i.e. has albumin binding affinity. Albumin binds to endogenous ligands such as fatty acids; however, it also interacts with exogenous ligands such as warfarin, penicillin and diazepam. As the binding of these drugs to albumin is reversible the albumin-drug complex serves as a drug reservoir that can enhance the drug biodistribution and bioavailability. Incorporation of components that mimic endogenous albumin-binding ligands, such as fatty acids, has been used to potentiate albumin association and increase drug efficacy.
In some embodiments, a chemical modification method that can be applied in the generation of the subject bispecific fusion proteins to increase protein half-life is lipidation, which involves the covalent binding of fatty acids to peptide side chains. Originally conceived of and developed as a method for extending the half-life of insulin, lipidation shares the same basic mechanism of half-life extension as PEGylation, namely increasing the hydrodynamic radius to reduce renal filtration. However, the lipid moiety is itself relatively small and the effect is mediated indirectly through the non-covalent binding of the lipid moiety to circulating albumin. One consequence of lipidation is that it reduces the water-solubility of the peptide but engineering of the linker between the peptide and the fatty acid can modulate this, for example by the use of glutamate or mini PEGs within the linker. Linker engineering and variation of the lipid moiety can affect self-aggregation which can contribute to increased half-life by slowing down biodistribution, independent of albumin. See, for example, Jonassen et al. (2012) Pharm Res. 29(8):2104-14.
Other examples of albumin binding moieties for use in the generation of certain bispecific fusion proteins include albumin-binding (PKE2) adnectins (See WO2011140086 “Serum Albumin Binding Molecules”, WO2015143199 “Serum albumin-binding Fibronectin Type III Domains” and WO2017053617 “Fast-off rate serum albumin binding fibronectin type iii domains”), the albumin binding domain 3 (ABD3) of protein G of Streptococcus strain G148, and the albumin binding domain antibody GSK2374697 (“AlbudAb”) or albumin binding NANOBODY® portion of ATN-103 (Ozoralizumab). PEGylation, XTEN, PAS and Other Polymers
A wide variety of macromolecular polymers and other molecules can be linked to the AFFIMER® polypeptides of the present disclosure to modulate biological properties of the resulting bispecific fusion protein, and/or provide new biological properties to the bispecific fusion protein. These macromolecular polymers can be linked to the AFFIMER® polypeptide via a naturally encoded amino acid, via a non-naturally encoded amino acid, or any functional substituent of a natural or non-natural amino acid, or any substituent or functional group added to a natural or non-natural amino acid. The molecular weight of the polymer may be of a wide range, including but not limited to, between about 100 Da and about 100,000 Da or more. The molecular weight of the polymer may be between about 100 Da and about 100,000 Da, including but not limited to, 100,000 Da, 95,000 Da, 90,000 Da, 85,000 Da, 80,000 Da, 75,000 Da, 70,000 Da, 65,000 Da, 60,000 Da, 55,000 Da, 50,000 Da, 45,000 Da, 40,000 Da, 35,000 Da, 30,000 Da, 25,000 Da, 20,000 Da, 15,000 Da, 10,000 Da, 9,000 Da, 8,000 Da, 7,000 Da, 6,000 Da, 5,000 Da, 4,000 Da, 3,000 Da, 2,000 Da, 1,000 Da, 900 Da, 800 Da, 700 Da, 600 Da, 500 Da, 400 Da, 300 Da, 200 Da, and 100 Da. In some embodiments, the molecular weight of the polymer is between about 100 Da and about 50,000 Da. In some embodiments, the molecular weight of the polymer is between about 100 Da and about 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 1,000 Da and about 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 5,000 Da and about 40,000 Da. In some embodiments, the molecular weight of the polymer is between about 10,000 Da and about 40,000 Da.
For this purpose, various methods including pegylation, polysialylation, HESylation, glycosylation, or recombinant PEG analogue fused to flexible and hydrophilic amino acid chain (500 to 600 amino acids) have been developed (See Chapman, (2002) Adv Drug Deliv Rev. 54. 531-545; Schlapschy et ah, (2007) Prot Eng Des Sel. 20, 273-283; Contermann (2011) Curr Op Biotechnol. 22, 868-876; Jevsevar et ah, (2012) Methods Mol Biol. 901, 233-246).
Examples of polymers include but are not limited to polyalkyl ethers and alkoxy-capped analogs thereof ( e.g ., polyoxyethylene glycol, polyoxyethylene/propylene glycol, and methoxy or ethoxy-capped analogs thereof, especially polyoxyethylene glycol, the latter is also known as polyethylene glycol or PEG); discrete PEG (dPEG); polyvinylpyrrolidones; polyvinylalkyl ethers; polyoxazolines, polyalkyl oxazolines and polyhydroxyalkyl oxazolines; polyacrylamides, polyalkyl acrylamides, and polyhydroxyalkyl acrylamides (e.g., polyhydroxypropylmethacrylamide and derivatives thereof); polyhydroxyalkyl acrylates; polysialic acids and analogs thereof; hydrophilic peptide sequences; polysaccharides and their derivatives, including dextran and dextran derivatives, e.g., carboxymethyldextran, dextran sulfates, aminodextran; cellulose and its derivatives, e.g., carboxymethyl cellulose, hydroxyalkyl celluloses; chitin and its derivatives, e.g., chitosan, succinyl chitosan, carboxymethylchitin, carboxymethylchitosan; hyaluronic acid and its derivatives; starches; alginates; chondroitin sulfate; albumin; pullulan and carboxymethyl pullulan; polyaminoacids and derivatives thereof, e.g., polyglutamic acids, polylysines, polyaspartic acids, polyaspartamides; maleic anhydride copolymers such as: styrene maleic anhydride copolymer, divinylethyl ether maleic anhydride copolymer; polyvinyl alcohols; copolymers thereof; terpolymers thereof; mixtures thereof; and derivatives of the foregoing.
The polymer selected may be water soluble so that the bispecific fusion protein to which it is attached does not precipitate in an aqueous environment, such as a physiological environment. The water soluble polymer may be any structural form including but not limited to linear, forked or branched. Typically, the water soluble polymer is a poly(alkylene glycol), such as poly(ethylene glycol) (PEG), but other water soluble polymers can also be employed. By way of example, PEG is used to describe some embodiments of this disclosure. For therapeutic use of the bispecific fusion protein, the polymer may be pharmaceutically acceptable.
The term “PEG” is used broadly to encompass any polyethylene glycol molecule, without regard to size or to modification at an end of the PEG, and can be represented as linked to the AFFIMER® polypeptide by the formula:
XO — (CH2CH20)n — CH2CH2 — or
XO — (CH2CH20)n — where n is 2 to 10,000 and X is H or a terminal modification, including but not limited to, a Cl-4 alkyl, a protecting group, or a terminal functional group. In some cases, a PEG used in the polypeptides of the disclosure terminates on one end with hydroxy or methoxy, i.e., X is H or CH3 (“methoxy PEG”).
It is noted that the other end of the PEG, which is shown in the above formulas by a terminal “ — ”, may attach to the AFFIMER® polypeptide via a naturally-occurring or non- naturally encoded amino acid. For instance, the attachment may be through an amide, carbamate or urea linkage to an amine group (including but not limited to, the epsilon amine of lysine or the N-terminus) of the polypeptide. Alternatively, the polymer is linked by a maleimide linkage to a thiol group (including but not limited to, the thiol group of cysteine) - which in the case of attachment to the AFFIMER® polypeptide sequence per se requires altering a residue in the AFFIMER® polypeptide sequence to a cysteine.
The number of water soluble polymers linked to the AFFIMER® polypeptide (e.g., the extent of PEGylation or glycosylation) can be adjusted to provide an altered (including but not limited to, increased or decreased) pharmacologic, pharmacokinetic or pharmacodynamic characteristic such as in vivo half-life in the resulting bispecific fusion protein. In some embodiments, the half-life of the resulting bispecific fusion protein is increased at least about 10, 20, 30, 40, 50, 60, 70, 80, 90 percent, 2-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 11- fold, 12-fold, 13-fold, 14-fold, 15-fold, 16-fold, 17-fold, 18-fold, 19-fold, 20-fold, 25-fold, 30- fold, 35-fold, 40-fold, 50-fold, or at least about 100-fold over an unmodified polypeptide.
Another variation of polymer system useful to modify the PK or other biological properties of the resulting bispecific fusion protein are the use of unstructured, hydrophilic amino acid polymers that are functional analogs of PEG, particularly as part of a fusion protein with the AFFIMER® polypeptide sequence. The inherent biodegradability of the polypeptide platform makes it attractive as a potentially more benign alternative to PEG. Another advantage is the precise molecular structure of the recombinant molecule in contrast to the polydispersity of PEG. Unlike HSA and Fc peptide fusions, in which the three-dimensional folding of the fusion partner needs to be maintained, the recombinant fusions to unstructured partners can, in many cases, be subjected to higher temperatures or harsh conditions such as HPLC purification.
One of the more advanced of this class of polypeptides is termed XTEN (Amunix) and is 864 amino acids long and comprised of six amino acids (A, E, G, P, S and T). See Schellenberger et al. “A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner” 2009 Nat Biotechnol. 27(12): 1186-90. Enabled by the biodegradable nature of the polymer, this is much larger than the 40 KDa PEGs typically used and confers a concomitantly greater half-life extension. The fusion of XTEN to the AFFIMER® containing polypeptide should result in half-life extension of the final bispecific fusion protein by 60- to 130-fold over the unmodified polypeptide.
A second polymer based on similar conceptual considerations is PAS (XL-Protein GmbH). Schlapschy et al. “PASYlation: a biological alternative to PEGylation for extending the plasma half-life of pharmaceutically active proteins” 2013 Protein Eng Des Sel. 26(8):489-501.
A random coil polymer comprised of an even more restricted set of only three small uncharged amino acids, proline, alanine and serine. AS with Fc, HAS and XTEN, the PAS modification can be genetically encoded with the AFFIMER® polypeptide sequence to produce an inline fusion protein when expressed.
Expression Methods and Systems
Recombinant fusion proteins described herein can be produced by any suitable method known in the art. Such methods range from direct protein synthesis methods to constructing a DNA sequence encoding polypeptides and expressing those sequences in a suitable host. For those recombinant fusion proteins including further modifications, such as a chemical modifications or conjugation, the recombinant fusion protein can be further manipulated chemically or enzymatically after isolation form the host cell or chemical synthesis.
The present disclosure includes recombinant methods and polynucleotides for recombinantly expressing the recombinant fusion proteins of the present disclosure comprising (i) introducing into a host cell a polynucleotide encoding the amino acid sequence of said bispecific fusion protein, for example, wherein the polynucleotide is in a vector and/or is operably linked to a promoter; (ii) culturing the host cell ( e.g ., eukaryotic or prokaryotic) under condition favorable to expression of the polynucleotide and, (iii) optionally, isolating the bispecific fusion protein from the host cell and/or medium in which the host cell is grown. See e.g., WO 04/041862, WO 2006/122786, WO 2008/020079, WO 2008/142164 or WO 2009/068627.
In some embodiments, a DNA sequence encoding a recombinant fusion protein of interest may be constructed by chemical synthesis using an oligonucleotide synthesizer. Oligonucleotides can be designed based on the amino acid sequence of the desired polypeptide and selecting those codons that are favored in the host cell in which the recombinant polypeptide of interest will be produced. Standard methods can be applied to synthesize a polynucleotide sequence encoding an isolated polypeptide of interest. For example, a complete amino acid sequence can be used to construct a back-translated gene. Further, a DNA oligomer containing a nucleotide sequence coding for the particular isolated polypeptide can be synthesized. For example, several small oligonucleotides coding for portions of the desired polypeptide can be synthesized and then ligated. The individual oligonucleotides typically contain 5' or 3' overhangs for complementary assembly.
Once a polynucleotide sequence encoding a recombinant fusion protein of the disclosure has been obtained, the vector for the production of the recombinant fusion protein may be produced by recombinant DNA technology using techniques well known in the art. Methods which are well known to those skilled in the art can be used to construct expression vectors containing the recombinant bispecific fusion protein coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. (See, for example, the techniques described in Sambrook et al, 1990, MOLECULAR CLONING, A LABORATORY MANUAL, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. and Ausubel et al. eds., 1998, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY). An expression vector comprising the nucleotide sequence of a recombinant fusion protein can be transferred to a host cell by conventional techniques ( e.g ., electroporation, liposomal transfection, and calcium phosphate precipitation) and the transfected cells are then cultured by conventional techniques to produce the recombinant fusion protein of the disclosure. In specific embodiments, the expression of the recombinant fusion protein is regulated by a constitutive, an inducible or a tissue, specific promoter.
The expression vector may include an origin of replication, such as may be selected based upon the type of host cell being used for expression. By way of example, the origin of replication from the plasmid pBR322 (Product No. 303-3s, New England Biolabs, Beverly, Mass.) is useful for most Gram- negative bacteria while various origins from SV40, polyoma, adenovirus, vesicular stomatitus virus (VSV) or papillomaviruses (such as HPV or BPV) are useful for cloning vectors in mammalian cells. Generally, the origin of replication component is not needed for mammalian expression vectors (for example, the SV40 origin is often used because it contains the early promoter).
The vector may include at least one selectable marker genes, e.g., genetic elements that encode a protein necessary for the survival and growth of a host cell grown in a selective culture medium. Typical selection marker genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, tetracycline, or kanamycin for prokaryotic host cells, (b) complement auxotrophic deficiencies of the cell; or (c) supply critical nutrients not available from complex media. Preferred selectable markers are the kanamycin resistance gene, the ampicillin resistance gene, and the tetracycline resistance gene. A neomycin resistance gene may also be used for selection in prokaryotic and eukaryotic host cells. Other selection genes may be used to amplify the gene which will be expressed. Amplification is a process where genes which are in greater demand for the production of a protein critical for growth are reiterated in tandem within the chromosomes of successive generations of recombinant cells. Examples of selectable markers for mammalian cells include dihydrofolate reductase (DHFR) and thymidine kinase. The mammalian cell transformants are placed under selection pressure which only the transformants are uniquely adapted to survive by virtue of the marker present in the vector. Selection pressure is imposed by culturing the transformed cells under conditions in which the concentration of selection agent in the medium is successively changed, thereby leading to amplification of both the selection gene and the DNA that encodes the recombinant fusion protein. As a result, increased quantities of the recombinant fusion protein are synthesized from the amplified DNA.
The vector may also include at least one ribosome binding site, which will be transcribed into the mRNA including the coding sequence for the recombinant fusion protein. For example, such a site is characterized by a Shine-Dalgarno sequence (prokaryotes) or a Kozak sequence (eukaryotes). The element is typically located 3' to the promoter and 5' to the coding sequence of the polypeptide to be expressed. The Shine-Dalgarno sequence is varied but is typically a polypurine (having a high A-G content). Many Shine-Dalgarno sequences have been identified, each of which can be readily synthesized using methods set forth above and used in a prokaryotic vector.
The expression vectors will typically contain a promoter that is recognized by the host organism and operably linked to a polynucleotide encoding the recombinant fusion protein.
Either a native or heterologous promoter may be used depending the host cell used for expression and the yield desired.
Promoters for use with prokaryotic hosts include the beta-lactamase and lactose promoter systems; alkaline phosphatase, a tryptophan (trp) promoter system; and hybrid promoters such as the tac promoter. Other known bacterial promoters are also suitable. Their sequences have been published, and they can be ligated to a desired polynucleotide sequence(s), using linkers or adapters as desired to supply restriction sites.
Promoters for use with yeast hosts are also known in the art. Yeast enhancers are advantageously used with yeast promoters. Suitable promoters for use with mammalian host cells are well known and include those obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Virus 40 (SV40). Other suitable mammalian promoters include heterologous mammalian promoters, e.g., heat-shock promoters and the actin promoter.
Additional promoters which may be used for expressing the selective binding agents of the disclosure include, but are not limited to: the SV40 early promoter region (Bemoist and Chambon, Nature, 290:304-310, 1981); the CMV promoter; the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto et al. (1980), Cell 22: 787-97); the herpes thymidine kinase promoter (Wagner et al. (1981), Proc. Natl. Acad. Sci. U.S.A. 78: 1444- 5); the regulatory sequences of the metallothionine gene (Brinster et al, Nature, 296; 39-42,
1982); prokaryotic expression vectors such as the beta- lactamase promoter (Villa-Kamaroff, et al., Proc. Natl. Acad. Sci. U.S.A. , 75; 3727-3731, 1978); or the tac promoter (DeBoer, et al. (1983), Proc. Natl. Acad. Sci. U.S.A., 80: 21-5). Also of interest are the following animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: the elastase I gene control region which is active in pancreatic acinar cells (Swift et al. (1984), Cell 38: 639-46; Omitz et al. (1986), Cold Spring Harbor Symp. Quant.
Biol. 50: 399-409; MacDonald (1987), Hepatology 7: 425-515); the insulin gene control region which is active in pancreatic beta cells (Hanahan (1985), Nature 315: 115-22); the immunoglobulin gene control region which is active in lymphoid cells (Grosschedl et al. (1984), Cell 38; 647-58; Adames et al. (1985), Nature 318; 533-8; Alexander et al. (1987), Mol. Cell. Biol. 7: 1436-44); the mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al. (1986), Cell 45: 485-95), albumin gene control region which is active in liver (Pinkert et al. (1987), Genes and Devel. 1: 268-76); the alphafetoprotein gene control region which is active in liver (Krumlauf et al. (1985), Mol. Cell. Biol. 5: 1639-48; Hammer et al. (1987), Science, 235: 53-8); the alpha 1- antitrypsin gene control region which is active in the liver (Kelsey et al. (1987), Genes and Devel. 1: 161-71); the beta-globin gene control region which is active in myeloid cells (Mogram et al., Nature, 315 338- 340, 1985; Kollias et al. (1986), Cell 46: 89-94); the myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al. (1987), Cell, 48: 703-12); the myosin light chain-2 gene control region which is active in skeletal muscle (Sani (1985), Nature, 314: 283-6); and the gonadotropic releasing hormone gene control region which is active in the hypothalamus (Mason et al. (1986), Science 234: 1372-8).
An enhancer sequence may be inserted into the vector to increase transcription in eukaryotic host cells. Several enhancer sequences available from mammalian genes are known (e.g., globin, elastase, albumin, alpha-feto-protein and insulin). Typically, however, an enhancer from a virus will be used. The SV40 enhancer, the cytomegalovirus early promoter enhancer, the polyoma enhancer, and adenovirus enhancers are examples of enhancing elements for the activation of eukaryotic promoters.
While an enhancer may be spliced into the vector at a position 5' or 3' to the polypeptide coding region, it is typically located at a site 5' from the promoter.
Vectors for expressing polynucleotides include those which are compatible with bacterial, insect, and mammalian host cells. Such vectors include, inter alia, pCRIE, pCR3, and pcDNA3.1 (Invitrogen Company, San Diego, Calif.), pBSII (Stratagene Company, La Jolla, Calif.), pET15 (Novagen, Madison, Wis.), pGEX (Pharmacia Biotech, Piscataway, N.J.), pEGFP-N2 (Clontech, Palo Alto, Calif.), pETL (BlueBacII; Invitrogen), pDSR- alpha (PCT Publication No. WO90/14363) and pFastBacDual (Gibco/BRL, Grand Island, N.Y.).
Additional possible vectors include, but are not limited to, cosmids, plasmids or modified viruses, but the vector system must be compatible with the selected host cell. Such vectors include but are not limited to plasmids such as BLUESCRIPT® plasmid derivatives (a high copy number ColEl-based phagemid, Stratagene Cloning Systems Inc., La Jolla Calif.), PCR cloning plasmids designed for cloning Taq-amplified PCR products (e.g., TOPO™. TA Cloning® Kit, PCR2.1 plasmid derivatives, Invitrogen, Carlsbad, Calif.), and mammalian, yeast or virus vectors such as a baculovirus expression system (pBacPAK plasmid derivatives, Clontech, Palo Alto, Calif.)· The recombinant molecules can be introduced into host cells via transformation, transfection, infection, electroporation, or other known techniques
Eukaryotic and prokaryotic host cells, including mammalian cells as hosts for expression of the recombinant fusion protein disclosed herein are well known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC). These include, inter alia, Chinese hamster ovary (CHO) cells, NSO, SP2 cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells ( e.g ., Hep G2), A549 cells, 3T3 cells, HEK-293 cells and a number of other cell lines. Mammalian host cells include human, mouse, rat, dog, monkey, pig, goat, bovine, horse and hamster cells. Cell lines of particular preference are selected through determining which cell lines have high expression levels. Other cell lines that may be used are insect cell lines, such as Sf9 cells, amphibian cells, bacterial cells, plant cells and fungal cells. Fungal cells include yeast and filamentous fungus cells including, for example, Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia minuta ( Ogataea minuta, Pichia lindneri ), Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Kluyveromyces lactis, Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reesei, Chrysosporium lucknowense, Fusarium sp., Fusarium gramineum, Fusarium venenatum, Physcomitrella patens and Neurospora crassa. Pichia sp., any Saccharomyces sp., Hansenula polymorpha, any Kluyveromyces sp., Candida albicans, any Aspergillus sp., Trichoderma reesei, Chrysosporium lucknowense, any Fusarium sp., Yarrowia lipolytica, and Neurospora crassa.
A variety of host-expression vector systems may be utilized to express the recombinant fusion protein of the disclosure. Such host-expression systems represent vehicles by which the coding sequences of the recombinant fusion protein may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express the recombinant fusion protein of the disclosure in situ. These include, but are not limited to, microorganisms such as bacteria (e.g., E. coli and B. subtilis ) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing fusion protein coding sequences; yeast (e.g., Saccharomyces pichia ) transformed with recombinant yeast expression vectors containing fusion protein coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the fusion protein coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus (CpMV) and tobacco mosaic virus (TMV)) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing fusion protein coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 293T, 3T3 cells, lymphotic cells (see U.S. Pat. No. 5,807,715), Per C.6 cells (rat retinal cells developed by Crucell)) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).
In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the recombinant fusion protein being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of the recombinant fusion protein, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al. (1983) "Easy Identification Of cDNA Clones," EMBO J. 2:1791-1794), in which the fusion protein coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye et al. (1985) "Up- Promoter Mutations In The Lpp Gene Of Escherichia coli," Nucleic Acids Res. 13:3101-3110; Van Heeke et al. (1989) "Expression Of Human Asparagine Synthetase In Escherichia coli," J. Biol. Chem. 24:5503-5509); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to a matrix glutathione-agarose beads followed by elution in the presence of free gluta-thione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
In an insect system, Autographa califomica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes. The virus grows in Spodoptera frugiperda cells. The fusion protein coding sequence may be cloned individually into non-essential regions (e.g., the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (e.g., the polyhedrin promoter).
In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, the fusion protein coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the immunoglobulin molecule in infected hosts (see e.g., see Logan et al. (1984) "Adenovirus Tripartite Leader Sequence Enhances Translation Of mRNAs Late After Infection," Proc. Natl. Acad. Sci. (U.S.A.) 81:3655-3659). Specific initiation signals may also be required for efficient translation of inserted fusion protein coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bitter et al.
(1987) "Expression and Secretion Vectors For Yeast," Methods in Enzymol. 153:516-544).
In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used. Such mammalian host cells include but are not limited to CHO, VERY, BHK, Hela, COS, MDCK, 293, 293T, 3T3, WI38, BT483, Hs578T, HTB2, BT20 and T47D, CRL7030 and Hs578Bst.
For long-term, high-yield production of recombinant proteins, stable expression is contemplated. For example, cell lines which stably express an antibody of the disclosure may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the recombinant fusion proteins of the disclosure. Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the recombinant fusion proteins.
A number of selection systems may be used, including but not limited to the herpes simplex vims thymidine kinase (Wigler et al. (1977) "Transfer Of Purified Herpes Virus Thymidine Kinase Gene To Cultured Mouse Cells," Cell 11:223-232), hypoxanthine-guanine phosphoribosyltransferase (Szybalska et al. (1962) "Genetics Of Human Cess Line. IV. DNA- Mediated Heritable Transformation Of A Biochemical Trait," Proc. Natl. Acad. Sci. (U.S.A.) 48:2026-2034), and adenine phosphoribosyltransferase (Lowy et al. (1980) "Isolation Of Transforming DNA: Cloning The Hamster Aprt Gene," Cell 22:817-823) genes can be employed in tk-, hgprt- or aprt- cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al. (1980) "Transformation Of Mammalian Cells With An Amplfiable Dominant- Acting Gene," Proc. Natl. Acad. Sci. (U.S.A.) 77:3567-3570; O'Hare et al. (1981) "Transformation Of Mouse Fibroblasts To Methotrexate Resistance By A Recombinant Plasmid Expressing A Prokaryotic Dihydrofolate Reductase," Proc. Natl. Acad. Sci. (U.S.A.) 78:1527-1531); gpt, which confers resistance to mycophenolic acid (Mulligan et al. (1981) "Selection For Animal Cells That Express The Escherichia coli Gene Coding For Xanthine-Guanine Phosphoribosyltransferase," Proc. Natl. Acad. Sci. (U.S.A.) 78:2072-2076); neo, which confers resistance to the aminoglycoside G-418 (Tachibana et al. (1991) "Altered Reactivity Of Immunoglobulin Produced By Human-Human Hybridoma Cells Transfected By pSV.2-Neo Gene," Cytotechnology 6(3):219-226; Tolstoshev (1993) "Gene Therapy, Concepts, Current Trials And Future Directions," Ann. Rev. Pharmacol. Toxicol. 32:573-596; Mulligan (1993) "The Basic Science Of Gene Therapy," Science 260:926-932; and Morgan et al. (1993) "Human gene therapy," Ann. Rev. Biochem. 62:191-217). Methods commonly known in the art of recombinant DNA technology which can be used are described in Ausubel et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY; Kriegler, 1990, GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY; and in Chapters 12 and 13, Dracopoli et al. (eds), 1994, CURRENT PROTOCOLS IN HUMAN GENETICS, John Wiley & Sons, NY.; Colbere-Garapin et al. (1981) "A New Dominant Hybrid Selective Marker For Higher Eukaryotic Cells," J. Mol. Biol. 150:1-14; and hygro, which confers resistance to hygromycin (Santerre et al. (1984) "Expression Of Prokaryotic Genes For Hygromycin B And G418 Resistance As Dominant-Selection Markers In Mouse L Cells," Gene 30:147-156).
The expression levels of a recombinant fusion protein can be increased by vector amplification (for a review, see Bebbington and Hentschel, "The Use of Vectors Based On Gene Amplification For The Expression Of Cloned Genes In Mammalian Cells," in DNA CLONING, Vol. 3. (Academic Press, New York, 1987)). When a marker in the vector system expressing a recombinant fusion protein is amplifiable, increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the nucleotide sequence of the recombinant fusion protein, production of the recombinant fusion protein will also increase (Crouse et al. (1983) "Expression and Amplification Of Engineered Mouse Dihydrofolate Reductase Minigenes," Mol. Cell. Biol. 3:257-266).
Where the bispecific fusion protein is an AFFIMER® antibody fusion or other multiprotein complex, the host cell may be co-transfected with two expression vectors, for instance the first vector encoding a heavy chain and the second vector encoding a light chain derived polypeptide, one or both of which includes an AFFIMER® polypeptide coding sequence. The two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides. Alternatively, a single vector may be used which encodes both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot (1986) "Expression And Amplification Of Engineered Mouse Dihydrofolate Reductase Minigenes," Nature 322:562-565; Kohler (1980) "Immunoglobulin Chain Loss In Hybridoma Lines," Proc. Natl. Acad. Sci. (U.S.A.) 77:2197-2199). The coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
In general, glycoproteins produced in a particular cell line or transgenic animal will have a glycosylation pattern that is characteristic for glycoproteins produced in the cell line or transgenic animal. Therefore, the particular glycosylation pattern of the recombinant fusion protein will depend on the particular cell line or transgenic animal used to produce the protein. In some embodiments of AFFIMER®/antibody fusions, a glycosylation pattern comprising only non-fucosylated N-glycans may be advantageous, because in the case of antibodies this has been shown to typically exhibit more potent efficacy than fucosylated counterparts both in vitro and in vivo (See for example, Shinkawa et ah, J. Biol. Chem. 278: 3466-3473 (2003); U.S. Pat. Nos. 6,946,292 and 7,214,775).
Further, expression of a bispecific fusion protein from production cell lines can be enhanced using a number of known techniques. For example, the glutamine synthetase gene expression system (the GS system) is a common approach for enhancing expression under certain conditions. The GS system is discussed in whole or part in connection with European Patent Nos. 0216846, 0256055, and 0323997 and European Patent Application No. 89303964.4. Thus, in some embodiments of the disclosure, the mammalian host cells ( e.g ., CHO) lack a glutamine synthetase gene and are grown in the absence of glutamine in the medium wherein, however, the polynucleotide encoding the immunoglobulin chain comprises a glutamine synthetase gene which complements the lack of the gene in the host cell. Such host cells containing the binder or polynucleotide or vector as discussed herein as well as expression methods, as discussed herein, for making the binder using such a host cell are part of the present disclosure.
Expression of recombinant proteins in insect cell culture systems ( e.g ., baculovirus) also offers a robust method for producing correctly folded and biologically functional proteins. Baculovirus systems for production of heterologous proteins in insect cells are well-known to those of skill in the art.
The recombinant fusion proteins produced by a transformed host can be purified according to any suitable method. Standard methods include chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification. Affinity tags such as hexa-histidine, maltose binding domain, influenza coat sequence, and glutathione-S-transferase can be attached to the protein to allow easy purification by passage over an appropriate affinity column. Isolated proteins can also be physically characterized using such techniques as proteolysis, mass spectrometry (MS), nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC), and x-ray crystallography.
In some embodiments, recombinant fusion proteins produced in bacterial culture can be isolated, for example, by initial extraction from cell pellets, followed by at least one concentration, salting-out, aqueous ion exchange, or size exclusion chromatography steps. HPLC can be employed for final purification steps. Microbial cells employed in expression of a recombinant protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
Polynucleotides for In vivo Delivery
An alternative approach to the delivery of therapeutic bispecific fusion proteins protein, such as a PD-L1 bispecific fusion protein, would be to leave the production of the therapeutic polypeptide to the body itself. A multitude of clinical studies have illustrated the utility of in vivo gene transfer into cells using a variety of different delivery systems. In vivo gene transfer seeks to administer to patients a polynucleotide encoding an AFFIMER® polypeptide, rather than the bispecific fusion protein. This allows the patient’s body to produce the therapeutic bispecific fusion protein of interest for a prolonged period of time, and secrete it either systemically or locally, depending on the production site. Gene-based a polynucleotide encoding an AFFIMER® polypeptide can present a labor- and cost-effective alternative to the conventional production, purification and administration of the polypeptide version of the bispecific fusion protein. A number of antibody expression platforms have been pursued in vivo to which delivery of a polynucleotide encoding an AFFIMER® polypeptide can be adapted: these include viral vectors, naked DNA and RNA. Gene transfer can not only enable cost-savings by reducing the cost of goods and of production but may also be able to reduce the frequency of drug administration. Overall, a prolonged in vivo production of the therapeutic bispecific fusion protein by expression of the polynucleotide encoding an AFFIMER® polypeptide can contribute to (i) a broader therapeutic or prophylactic application of bispecific fusion proteins in price-sensitive conditions, (ii) an improved accessibility to therapy in both developed and developing countries, and (iii) more effective and affordable treatment modalities. In addition to in vivo gene transfer, cells can be harvested from the host (or a donor), engineered with a polynucleotide encoding an AFFIMER® polypeptide to produce bispecific fusion proteins and re-administered to patients.
Intramuscular antibody gene administration has been most widely evaluated (reviewed in Deal et al. (2015) “Engineering humoral immunity as prophylaxis or therapy” Curr Opin Immunol. 35:113-22.) and carries the highest clinical translatability and application when applied to polynucleotides encoding an AFFIMER® polypeptide. Indeed, the inherent anatomical, cellular and physiological properties of skeletal muscle make it a stable environment for long-term polunucleotide expression and systemic circulation. Skeletal muscle is easily accessible, allowing multiple or repeated administrations. The abundant blood vascular supply provides an efficient transport system for secreted therapeutic bispecific fusion proteins into the circulation. The syncytial nature of muscle fibers allows dispersal of nucleotides from a limited site of penetration to a large number of neighboring nuclei within the fiber. Skeletal muscle fibers are also terminally differentiated cells, and nuclei within the fibers are post-mitotic. Consequently, integration in the host genome is not a prerequisite to attain prolonged mAb expression. The liver is another site often used for pre-clinical antibody gene transfer, and is typically transfected via i.v. injection, and can also be a site of gene transfer for polynucleotides either for local delivery of bispecific fusion proteins (such as in the treatment of liver cancer and/or metaplasias) or for the generation of bispecific fusion proteins that are secreted into the vascular for systemic circulation. This organ has various physiological functions, including the synthesis of plasma proteins. This organ can be particularly well suited for in vivo polynucleotide expression.
The tumor presents another site for polynucleotide transfer, targeted either via i.v. or direct injection/electroporation. Indeed, intratumoral polynucleotide expression can allow for a local production of the therapeutic bispecific fusion proteins, waiving the need for high systemic bispecific fusion protein levels that might otherwise be required to penetrate and impact solid tumors. A similar rationale applies for the brain, which is frequently targeted in the context of antibody gene transfer to avoid the difficulties with blood-brain barrier trafficking and would likewise be a target for delivery of polynucleotides. See, for example, Beckman et al. (2015) “Antibody constructs in cancer therapy: protein engineering strategies to improve exposure in solid tumors” Cancer 109(2): 170-9; Dronca et al. (2015) “Immunomodulatory antibody therapy of cancer: the closer, the better” Clin Cancer Res. 21(5):944-6; and Neves et al. (2016) “Antibody approaches to treat brain diseases” Trends Biotechnol. 34(l):36-48.
The success of gene therapy has largely been driven by improvements in nonviral and viral gene transfer vectors. An array of physical and chemical nonviral methods have been used to transfer DNA and mRNA to mammalian cells and a substantial number of these have been developed as clinical stage technologies for gene therapy, both ex vivo and in vivo , and are readily adapted for delivery of the polynucleotides of the present disclosure. To illustrate, cationic liposome technology can be employed, which is based on the ability of amphipathic lipids, possessing a positively charged head group and a hydrophobic lipid tail, to bind to negatively charged DNA or RNA and form particles that generally enter cells by endocytosis. Some cationic liposomes also contain a neutral co-lipid, thought to enhance liposome uptake by mammalian cells. See, for example, Feigner et al. (1987) Lipofection: a highly efficient, lipid- mediated DNA-transfection procedure. MNAS 84:7413-7417; San et al. (1983) “Safety and short-term toxicity of a novel cationic lipid formulation for human gene therapy” Hum. Gene Ther. 4:781-788; Xu et al. (1996) “Mechanism of DNA release from cationic liposome/DNA complexes used in cell transfection” Biochemistry 35,:5616-5623; and Legendre et al. (1992) “Delivery of plasmid DNA into mammalian cell lines using pH-sensitive liposomes: comparison with cationic liposomes” Pharm. Res. 9, 1235-1242.
Similarly, other polycations, such as poly-l-lysine and polyethylene-imine, can be used to deliver polynucleotides. These polycations complex with polynucleotides via charge interaction and aid in the condensation of DNA or RNA into nanoparticles, which are then substrates for endosome-mediated uptake. Several of these cationic polynucleotide complex technologies have been developed as potential clinical products, including complexes with plasmid DNA, oligodeoxynucleotides, and various forms of synthetic RNA. Modified (and unmodified or “naked”) DNA and RNA have also been shown to mediate successful gene transfer in a number of circumstances and can also be used as systems for delivery of polynucleotides. These include the use of plasmid DNA by direct intramuscular injection, the use of intratumoral injection of plasmid DNA. See, for example, Rodrigo et al. (2012) “De novo automated design of small RNA circuits for engineering synthetic riboregulation in living cells” PNAS 109:15271-15276; Oishi et al. (2005) “Smart polyion complex micelles for targeted intracellular delivery of PEGylated antisense oligonucleotides containing acid-labile linkages” Chembiochem. 6:718-725; Bhatt et al. (2015) “Microbeads mediated oral plasmid DNA delivery using polymethacrylate vectors: an effectual groundwork for colorectal cancer” Drug Deliv. 22:849-861; Ulmer et al. (1994) Protective immunity by intramuscular injection of low doses of influenza virus DNA vaccines” Vaccine 12: 1541-1544; and Heinzerling et al. (2005) “Intratumoral injection of DNA encoding human interleukin 12 into patients with metastatic melanoma: clinical efficacy” Hum. Gene Ther. 16:35-48.
Viral vectors are currently used as a delivery vehicle in the vast majority of pre-clinical and clinical gene therapy trials and in the first to be approved directed gene therapy. See Gene Therapy Clinical Trials Worldwide 2017 (abedia.com/wiley/). The main driver thereto is their exceptional gene delivery efficiency, which reflects a natural evolutionary development; viral vector systems are attractive for gene delivery, because viruses have evolved the ability to cross through cellular membranes by infection, thereby delivering polynucleotides such as polynucleotides to target cells. Pioneered by adenoviral systems, the field of viral vector- mediated antibody gene transfer made significant strides in the past decades. The myriad of successfully evaluated administration routes, pre-clinical models and disease indications puts the capabilities of antibody gene transfer at full display through which the skilled artisan would readily be able to identify and adapt antibody gene transfer systems and techniques for in vivo delivery of polynucleotides. Muscle has emerged as the administration site of choice for prolonged mAb expression and would similarly be a suitable target tissue for prolonged bispecific fusion protein expression. In the context of vectored intratumoral polynucleotide gene transfer, oncolytic viruses have a distinct advantage, as they can specifically target tumor cells, boost bispecific fusion protein expression, and amplify therapeutic responses - such as to a PD- L1 bispecific fusion protein.
In vivo gene transfer of polynucleotides can also be accomplished by use of nonviral vectors, such as expression plasmids. Nonviral vectors are easily produced and do not seem to induce specific immune responses. Muscle tissue is most often used as target tissue for transfection, because muscle tissue is well vascularized and easily accessible, and myocytes are long-lived cells. Intramuscular injection of naked plasmid DNA results in transfection of a certain percentage of myocytes. Using this approach, plasmid DNA encoding cytokines and cytokine/IgGl chimeric proteins has been introduced in vivo and has positively influenced (autoimmune) disease outcome.
In some instances, in order to increase transfection efficiency via so-called intravascular delivery in which increased gene delivery and expression levels are achieved by inducing a short-lived transient high pressure in the veins. Special blood-pressure cuffs that may facilitate localized uptake by temporarily increasing vascular pressure and can be adapted for use in human patients for this type of gene delivery. See, for example, Zhang et al. (2001) “Efficient expression of naked DNA delivered intraarterially to limb muscles of nonhuman primates” Hum. Gene Ther., 12:427-438
Increased efficiency can also be gained through other techniques, such as in which delivery of the polynucleotide is improved by use of chemical carriers — cationic polymers or lipids — or via a physical approach — gene gun delivery or electroporation. See Tranchant et al. (2004) “Physicochemical optimisation of plasmid delivery by cationic lipids” J. Gene Med., 6 (Suppl. 1):S24-S35; and Niidome et al. (2002) “Gene therapy progress and prospects: nonviral vectors” Gene Ther., 9:1647-1652. Electroporation is especially regarded as an interesting technique for nonviral gene delivery. Somiari, et al. (2000) “Theory and in vivo application of electroporative gene delivery” Mol. Ther. 2:178-187; and Jaroszeski et al. (1999) “ In vivo gene delivery by electroporation” Adv. Drug Delivery Rev., 35:131-137. With electroporation, pulsed electrical currents are applied to a local tissue area to enhance cell permeability, resulting in gene transfer across the membrane. Research has shown that in vivo gene delivery can be at least 10- 100 times more efficient with electroporation than without. See, for example, Aihara et al. (1998) “Gene transfer into muscle by electroporation in vivo ” Nat. Biotechnol. 16:867-870; Mir, et al. (1999) “High-efficiency gene transfer into skeletal muscle mediated by electric pulses” PNAS 96:4262-4267; Rizzuto, et al. (1999) “Efficient and regulated erythropoietin production by naked DNA injection and muscle electroporation” PNAS 96: 6417-6422; and Mathiesen (1999) “Electropermeabilization of skeletal muscle enhances gene transfer in vivo ” Gene Ther., 6:508- 514.
Encoded PD-L1 binding AFFIMER® polypeptides can be delivered by a wide range of gene delivery system commonly used for gene therapy including viral, non- viral, or physical.
See, for example, Rosenberg et al., Science, 242:1575-1578, 1988, and Wolff et al., Proc. Natl. Acad. Sci. USA 86:9011-9014 (1989). Discussion of methods and compositions for use in gene therapy include Eck et al., in Goodman & Gilman's The Pharmacological Basis of Therapeutics, Ninth Edition, Hardman et al., eds., McGraw-Hill, New York, (1996), Chapter 5, pp. 77-101; Wilson, Clin. Exp. Immunol. 107 (Suppl. 1):31-32, 1997; Wivel et al., Hematology/Oncology Clinics of North America, Gene Therapy, S. L. Eck, ed., 12(3):483-501, 1998; Romano et al., Stem Cells, 18:19-39, 2000, and the references cited therein. U.S. Pat. No. 6,080,728 also provides a discussion of a wide variety of gene delivery methods and compositions. The routes of delivery include, for example, systemic administration and administration in situ.
An effective polynucleotide gene transfer approach must be directed to the specific tissues/cells where it is needed, and the resulting transgene expression should be at a level that is appropriate to the specific application. Promoters are a major cis-acting element within the vector genome design that can dictate the overall strength of expression as well as cell- specificity.
Table 9. Examples of Ubiquitous and Cell-specific Promoters.
Figure imgf000079_0001
Figure imgf000080_0001
In some cases, ubiquitous expression of the polynucleotide in all cell types is desired. Constitutive promoters such as the human elongation factor la-subunit (EFla), immediate-early cytomegalovirus (CMV), chicken b-actin (CBA) and its derivative CAG, the b glucuronidase (GUSB), or ubiquitin C (UBC) can be used to promote expression of the polynucleotide in most tissues. Generally, CBA and CAG promote the larger expression among the constitutive promoters; however, their size of -1.7 kbs in comparison to CMV (-0.8 kbs) or EFla (-1.2 kbs) may limit use in vectors with packaging constraints such as AAV, particularly where bispecific fusion protein produced by expression of the polynucleotide is large. The GUSB or UBC promoters can provide ubiquitous gene expression with a smaller size of 378 bps and 403 bps, respectively, but they are considerably weaker than the CMV or CBA promoter. Thus, modifications to constitutive promoters in order to reduce the size without affecting its expression have been pursued and examples such as the CBh (-800 bps) and the miniCBA (-800 bps) can promote expression comparable and even higher in selected tissues (Gray et ak, Hum Gene Ther. 2011 22: 1143-1153).
When expression of the polynucleotide should be restricted to certain cell types within an organ, promoters can be used to mediate this specificity. For example, within the nervous system promoters have been used to restrict expression to neurons, astrocytes, or oligodendrocytes. In neurons, the neuron- specific enolase (NSE) promoter drives stronger expression than ubiquitous promoters. Additionally, the platelet-derived growth factor B-chain (PDGF-b), the synapsin (Syn), and the methyl-CpG binding protein 2 (MeCP2) promoters can drive neuron- specific expression at lower levels than NSE. In astrocytes, the 680 bps-long shortened version [gfaABC(l)D] of the glial fibrillary acidic protein (GFAP, 2.2 kbs) promoter can confer higher levels of expression with the same astrocyte-specificity as the GFAP promoter. Targeting oligodendrocytes can also be accomplished by the selection of the myelin basic protein (MBP) promoter, whose expression is restricted to this glial cell; however, its size of 1.9 kbs and low expression levels limit its use.
In the case of expressing the polynucleotides in skeletal muscle cells, examples of promoters based on muscle creatine kinase (MCK) and desmin (1.7 kbs) have showed a high rate of specificity (with minimal expression in the liver if desired). The promoter of the a-myosin heavy chain (a-MHC; 1.2 kbs) has shown significant cardiac specificity in comparison with other muscle promoters (Lee et ak, 2011 J Cardiol. 57(1): 115-22). In hematopoietic stem cells the synthetic MND promoter (Li et ak, 2010 J Neurosci Methods. 189(l):56-64) and the promoter contained in the 2AUCOE (ubiquitous chromatin opening element) have shown to drive a higher transgene expression in all cell lineages when compared to the EFla and CMV promoters, respectively (Zhang et ak, 2007 Blood. 110(5): 1448-57; Koldej 2013 Hum Gene Ther Clin Dev. 24(2):77-85; Dighe et ak, 2014 PLoS One. 9(8):el04805.). Conversely, using promoters to restrict expression to only liver hepatocytes after vector-mediated gene transfer has been shown to reduce transgene- specific immune responses in systems where that is a risk, and to even induce immune tolerance to the expressed protein (Zhang et ah, 2012 Hum Gene Ther. 23(5):460-72), which for certain bispecific fusion proteins may be beneficial. The a 1 -antitrypsin (hAAT; 347 bps) and the thyroxine binding globulin (TBG; -400 bps) promoters drive gene expression restricted to the liver with minimal invasion to other tissues (Yan et ah, 2012 Gene. 506(2):289-94; Cunningham et al., 2008 Mol Ther. 16(6): 1081-8).
In some embodiments, a mechanism to control the duration and amount of in vivo polynucleotide expression will typically be desired. There are a variety of inducible promoters which can be adapted for use with viral vectored- and plasmid DNA-based polynucleotide gene transfer. See Fang et al. (2007) “An antibody delivery system for regulated expression of therapeutic levels of monoclonal antibodies in vivo” Mol Ther. 5(6): 1153-9; and Perez et al. (2004) “Regulatable systemic production of monoclonal antibodies by in vivo muscle electroporation” Genet Vaccines Ther. 2(1):2. An example of a regulatable mechanism currently under clinical evaluation is an ecdysone-based gene switch activated by a small molecule ligand. Cai et al. (2016) “Plasma pharmacokinetics of veledimex, a small-molecule activator ligand for a proprietary gene therapy promoter system, in healthy subjects” Clin Pharmacol Drug Dev. 2016.
In some embodiments of the polynucleotides, viral post-transcriptional regulatory elements (PREs) may be used; these cis-acting elements are required for nuclear export of intronless viral RNA (Huang and Yen, 1994 J Virol. 68(5):3193-9; and 1995 Mol Cell Biol. 15(7):3864-9). Examples include HPRE (Hepatitis B Vims PRE, 533 bps) and WPRE (Woodchuck Hepatitis Vims PRE, 600 bps), which can increase the level of transgene expression by almost 10-fold in certain instances (Donello et al., 1998 J Virol. 72(6):5085-92). To further illustrate, using lentiviral and AAV vectors, WPRE was found to increase CMV promoter driven transgene expression, as well as increase PPE, PDGF and NSE promoter-driven transgene expression. Another effect of the WPRE can be to protect polynucleotides transgenes from silencing (Patema et al., 2000 Gene Ther. 7(15): 1304-11; Xia et al., 2007 Stem Cells Dev. 2007 Feb; 16(1): 167-76).
The polyadenylation of a transcribed polynucleotide transcript can also be important for nuclear export, translation, and mRNA stability. Therefore, in some embodiments, the polynucleotide will include a polyadenylation signal sequence. A variety of studies are available that have determined the effects of different polyA signals on gene expression and mRNA stability. Examples of polyadenylation signal sequences include SV40 late or bovine growth hormone polyA (bGHpA) signal sequences, as well as minimal synthetic polyA (SPA) signal (Levitt et al., 1989 Genes Dev. 3(7): 1019-25; Yew et al., 1997 Hum Gene Ther. 1997 8(5):575- 84). The efficiency of polyadenylation is increased by the SV40 late polyA signal upstream enhancer (USE) placed upstream of other polyA signals (Schek et al., 1992 Mol Cell Biol. 12(12):5386-93). In some embodiments, merely to illustrate, the polynucleotide will include an SV40 late + 2xUSE polyA signal.
Table 10. Examples of Polyadenylation Signals
Figure imgf000083_0001
In some embodiments, it may be desirable for the polynucleotide to include at least one regulatory enhancers, e.g., in addition to any promoter sequences. The CMV enhancer is upstream of the CMV promoter at -598 to -68 (Boshart et al., 1985 Cell. 41(2):521-30) (-600 bps) and contains transcription binding sites. In some embodiments, a CMV enhancer can be included in the construct to increase tissue- specific promoter-driven transgene expression, such as using the ANF (atrial natriuretic factor) promoter, the CC10 (club cell 10) promoter, SP-C (surfactant protein C) promoter, or the PDGF-b (platelet-derived growth factor-b) promoter (merely as examples). Altogether, the CMV enhancer increases transgene expression under different cell- specific promoters and different cell types making it a broadly applicable tool to increase transgene expression levels. In muscle, for example, in AAV expression systems transgene expression using the CMV enhancer with a muscle-specific promoter can increase expression levels of the protein encoded by the transgene, so would be particularly useful in the current disclosure for expressing bispecific fusion proteins from polynucleotides introduced into muscle cells of a patient.
The subject polynucleotides may also include at least one intronic sequences. The presence of an intron or intervening sequence in mRNA was first described, in vitro, to be important for mRNA processing and increased transgene expression (Huang and Gorman, 1990 Mol Cell Biol. 10(4): 1805-10; Niwa et al., 1990 Genes Dev. 4(9): 1552-9). The intron(s) can be placed within the coding sequence for the bispecific fusion protein and/or can be placed between the promoter and transgene. A variety of introns (Table 3) placed between the promoter and transgene were compared, in mice using AAV2, for liver transgene expression (Wu et al., 2008). The MVM (minute virus of mice) intron increased transgene expression more than any other intron tested and more than 80-fold over no intron (Wu et al., 2008). However, in cultured neurons using AAV expression cassettes, transgene expression was less under a CaMPKII promoter with a chimeric intron (human b-globin donor and immunoglobulin heavy chain acceptor) between the transgene and polyA signal compared to a WPRE (Choi et al., 2014). Together, an intron can be a valuable element to include in an expression cassette to increase transgene expression.
Table 11. Examples of Introns
Figure imgf000084_0001
Figure imgf000085_0001
In the case of episomal vectors, the subject polynucleotides may also include at least one origins of replication, minichromosome maintenance elements (MME) and/or nuclear localization elements. Episomal vectors of the disclosure comprise a portion of a vims genomic DNA that encodes an origin of replication (ori), which is required for such vectors to be self- replicating and, thus, to persist in a host cell over several generations. In addition, an episomal vector of the disclosure also may contain at least one gene encoding viral proteins that are required for replication, i.e., replicator protein (s). Optionally, the replicator protein(s) which help initiate replication may be expressed in trans on another DNA molecule, such as on another vector or on the host genomic DNA, in the host cell containing a self-replicating episomal expression vector of this disclosure. Preferred self-replicating episomal LCR-containing expression vectors of the disclosure do not contain viral sequences that are not required for long term stable maintenance in a eukaryotic host cell such as regions of a viral genome DNA encoding core or capsid proteins that would produce infectious viral particles or viral oncogenic sequences which may be present in the full-length viral genomic DNA molecule. The term "stable maintenance" herein, refers to the ability of a self-replicating episomal expression vector of this disclosure to persist or be maintained in non-dividing cells or in progeny cells of dividing cells in the absence of continuous selection without a significant loss ( e.g ., >50%) in copy number of the vector for two, three, four, or five or more generations. In some embodiments, the vectors will be maintained over 10-15 or more cell generations. In contrast, "transient" or "short term" persistence of a plasmid in a host cell refers to the inability of a vector to replicate and segregate in a host cell in a stable manner; that is, the vector will be lost after one or two generations, or will undergo a loss of >51% of its copy number between successive generations.
Several representative self-replicating, LCR-containing, episomal vectors useful in the context of the present disclosure are described further below. The self-replicating function may alternatively be provided by at least one mammalian sequences such as described by Wohlge uth et al., 1996, Gene Therapy 3:503; Vos et al., 1995, Jour. Cell. Biol., Supp. 21A, 433; and Sun et al., 1994, Nature Genetics 8:33, optionally in combination with at least one sequence which may be required for nuclear retention. The advantage of using mammalian, especially human sequences for providing the self- replicating function is that no extraneous activation factors are required which could have toxic or oncogenic properties. It will be understood by one of skill in the art that the disclosure is not limited to any one origin of replication or any one episomal vector but encompasses the combination of the tissue-restricted control of an LCR in an episomal vector. See also WO1998007876 “Self-replicating episomal expression vectors conferring tissue- specific gene expression” and US Patent 7790446 “Vectors, cell lines and their use in obtaining extended episomal maintenance replication of hybrid plasmids and expression of gene products”
Epstein-Barr Virus-Based Self-Replicating Episomal Expression Vectors. The latent origin oriP from Epstein-Barr Virus (EBV) is described in Yates et. al., Proc. Natl. Acad. Sci. USA 81:3806-3810 (1984); Yates et al., Nature 313:812-815 (1985); Krysan et al., Mol. Cell. Biol. 9:1026-1033 (1989); James et al. Gene 86: 233-239 (1990), Peterson and Legerski, Gene 107:279-284 (1991); and Pan et al., Som. Cell Molec. Genet. 18:163-177 (1992)). An EBV- based episomal vector useful according to the disclosure can contain the oriP region of EBV which is carried on a 2.61 kb fragment of EBV and the EBNA-1 gene which is carried on a 2.18 kb fragment of EBV. The EBNA-1 protein, which is the only viral gene product required to support in trans episomal replication of vectors containing oriP, may be provided on the same episomal expression vector containing oriP. It is also understood, that as with any protein such as EBNA-1 known to be required to support replication of viral plasmid in trans, the gene also may be expressed on another DNA molecule, such as a different DNA vector.
Papilloma Virus-Based, Self-Replicating, Episomal Expression Vectors. The episomal expression vectors of the disclosure also may be based on replication functions of the papilloma family of virus, including but not limited to Bovine Papilloma Virus (BPV) and Human Papilloma Viruses (HPVs). BPV and HPVs persist as stably maintained plasmids in mammalian cells. -S trans-acting factors encoded by BPV and HPVs, namely El and E2, have also been identified which are necessary and sufficient for mediate replication in many cell types via minimal origin of replication (Ustav et al., EMBO J. 10: 449-457 (1991); Ustavet al., EMBO J. 10:4231-4329, (1991); Ustav et al., Proc. Natl. Acad. Sci. USA 90: 898-902 (1993)).
An episomal vector useful according to the disclosure is the BPV-I vector system described in Piirsoo et al., EMBO J., 15:1 (1996) and in WO 94/12629. The BPV-1 vector system described in Piirsoo et al. comprises a plasmid harboring the BPV-1 origin of replication (minimal origin plus extrachro osomal maintenance element) and optionally the El and E2 genes. The BPV-1 El and E2 genes are required for stable maintenance of a BPV episomal vector. These factors ensure that the plasmid is replicated to a stable copy number of up to thirty copies per cell independent of cell cycle status. The gene construct therefore persists stably in both dividing and non-dividing cells. This allows the maintenance of the gene construct in cells such as hemopoietic stem cells and more committed precursor cells.
The BPV origin of replication has been located at the 3’ end of the upstream regulatory region within a 60 base pair (bp) DNA fragment (nucleotides (nt) 7914 - 7927) which includes binding sites for the El and E2 replication factors. The minimal origin of replication of HPV has also been characterized and located in the URR fragment (nt 7022- 7927) of HPV (see, for example, Chiang et al., Proc. Natl. Acad. Sci. USA 89:5799-5803 (1992)). As used herein, "El" refers to the protein encoded by nucleotides (nt) 849-2663 of BPV subtype 1 or by nt 832- 2779 of HPV of subtype 11, to equivalent El proteins of other papilloma viruses, or to functional fragments or mutants of a papilloma virus El protein, i.e., fragments or mutants of El which possess the replicating properties of El.
As used herein, "E2H refers to the protein encoded by nt 2594-3837 of BPV subtype 1 or by nt 2723-3823 of HPV subtype 11, to equivalent E2 proteins of other papilloma viruses, or to functional fragments or mutants of a papilloma vims E2 protein, i.e., fragments or mutants of E2 which possess the replicating properties of E2. "Minichromosomal maintenance element"
(MME) refers to the extrachromosomal maintenance element of the papilloma viral genome to which viral or human proteins essential for papilloma viral replication bind, which region is essential for stable episomal maintenance of the papilloma viral MO in a host cell, as described in Piirsoo et al. (supra). Preferably, the MME is a sequence containing multiple binding sites for the transcriptional activator E2. The MME in BPV is herein defined as the region of BPV located within the upstream regulatory region which includes a minimum of about six sequential E2 binding sites, and which gives optimum stable maintenance with about ten sequential E2 binding sites. E2 binding site 9 is an example sequence for this site, as described hereinbelow, wherein the sequential sites are separated by a spacer of about 4-10 nucleotides, and optimally 6 nucleotides. El and E2 can be provided to the plasmid either in cis or in trans, also as described in WO 94/12629 and in Piirsoo et al. (supra).
"E2 binding site" refers to the minimum sequence of papillomavirus double-stranded DNA to which the E2 protein binds. An E2 binding site may include the sequence 5’ACCGTTGCCGGT3' (SEQ ID NO: 172), which is high affinity E2 binding site 9 of the BPV- 1 URR; alternatively, an E2 binding site may include permutations of binding site 9, which permutations are found within the URR, and fall within the generic E2 binding sequence 5' ACCN6GGT 3'. One or more transcriptional activator E2 binding sites are, in most papillomaviruses, located in the upstream regulatory region, as in BPV and HPV. A vector which also is useful according to the disclosure may include a region of BPV between 6959 - 7945/1 - 470 on the BPV genetic map (as described in WO 94/12629) , which region includes an origin of replication, a first promoter operatively associated with a gene of interest, the BPV El gene operatively associated with a second promoter to drive transcription of the El gene; and the BPV E2 gene operatively associated with a third promoter to drive transcription of the E2 gene.
El and E2 from BPV will replicate vectors containing the BPV origin or the origin of many HPV subtypes (Chiang et ah, supra). El and E2 from HPV will replicate vectors via the BPV origin and via the origin of many HPV subtypes (Chiang et al., supra). As with all vectors of the disclosure, the BPV-based episomal expression vectors of the disclosure must persist through 2-5 or more divisions of the host cell.
See also US Patent 7790446 and Abroi et al. (2004) “Analysis of chromatin attachment and partitioning functions of bovine papillomavirus type 1 E2 protein. Journal of Virology 78:2100-13 which have shown that the BPV1 E2 protein dependent MME and EBV EBNA1 dependent FR segregation/partitioning activities function independently from replication of the plasmids. The stable-maintenance function of EBNA1/FR and E2/MME can be used to ensure long-time episomal maintenance for cellular replication origins.
Papovavims-Based, Self-Replicating, Episomal Expression Vectors. The vectors of the disclosure also may be derived from a human papovavims BK genomic DNA molecule. For example, the BK viral genome can be digested with restriction enzymes EcoRI and BamHI to produce a 5 kilobase (kb) fragment that contains the BK viral origin of replication sequences that can confer stable maintenance on vectors (see, for example, De Benedetti and Rhoads, Nucleic Acids Res . 19:1925 (1991), as can a 3.2 kb fragment of the BK vims (Cooper and Miron,
Human Gene Therapy 4:557 (1993)).
The polynucleotides of the present disclosure can be provided as circular or linear polynucleotides. The circular and linear polynucleotides are capable of directing expression of the bispecific fusion protein coding sequence in an appropriate subject cell. The at least one polynucleotide systems for expressing a bispecific fusion protein may be chimeric, meaning that at least one of its components is heterologous with respect to at least one of its other components.
Viral Vectors
Examples of viral gene therapy system that are readily adapted for use in the present disclosure include plasmid, adenovirus, adeno-associated virus (AAV), retrovirus, lentivirus, herpes simplex vims, vaccinia vims, poxvims, reovims, measles vims, Semliki Forest vims, and the like. Preferred viral vectors are based on non-cytopathic eukaryotic viruses in which non- essential genes have been replaced with the polynucleotide construct carrying the polynucleotide sequences encoding the epitopes and targeting sequences of interest. To further illustrate, encoded AFFIMER® polypeptides can be delivered in vivo using adenoviruses and adeno-associated (AAV) viruses, which are double- stranded DNA viruses that have already been approved for human use in gene therapy.
Adenovirus Vectors
One illustrative method for in vivo delivery of at least one polynucleotide sequences involves the use of an adenovirus (“AdV”) expression vector. AdVs are non-enveloped, double- stranded DNA viruses that neither integrate in the host genome nor replicate during cell division. AdV-mediated antibody gene transfer has shown therapeutic efficacy in a variety of different disease models advancing towards the clinic. Systemic mAb expression has mostly been pursued, via s.c. and especially i.v. and intramuscular AdV injection. See Wold et al. (2013) “Adenovirus vectors for gene therapy, vaccination and cancer gene therapy” Curr Gene Ther. 13(6):421— 33; and Deal et al. “Engineering humoral immunity as prophylaxis or therapy” 2015 Curr Opin Immunol. 35:113-22. Other routes of delivery have focused on more local mAb production, such as via intranasal, intratracheal or intrapleural administration of the encoding AdV. The use of AdVs as oncolytic vectors is a popular approach particularly for generation of encoded antibodies at the site of tumors. Foreign genes delivered by current adenoviral gene delivery system are episomal, and therefore, have low genotoxicity to host cells. Therefore, gene therapy using adenoviral gene delivery systems may be considerably safe. The present disclosure specifically contemplates the delivery of bispecific fusion proteins by expression of polynucleotides delivered in the form of an adenoviral vector and delivery system.
Adenovirus has been usually employed as a gene delivery vector because of its mid-sized genome, ease of manipulation, high titer, wide target-cell range and high infectivity. Both ends of the viral genome contains 100-200 bp ITRs (inverted terminal repeats), which are cis elements necessary for viral DNA replication and packaging. The El region (E1A and E1B) of genome encodes proteins responsible for the regulation of transcription of the viral genome and a few cellular genes. The E2 region (E2A and E2B) encodes proteins responsible for viral DNA replication. Of adenoviral vectors developed so far, the replication incompetent adenovirus having the deleted El region is usually used and represent one example of AdV for generating the polynucleotides of the present disclosure. The deleted E3 region in adenoviral vectors may provide an insertion site for transgenes (Thimmappaya, B. et al., Cell, 31:543-551(1982); and Riordan, J. R. et al., Science, 245:1066-1073(1989)).
An “adenovirus expression vector” is meant to include those constructs containing adenovirus sequences sufficient to (a) support packaging of the construct and (b) to express a polynucleotide that encodes a polypeptide including a bispecific fusion protein such as a PD-L1 binding AFFIMER® (the polynucleotide sequence). In some embodiments, the sequence for polynucleotide may be inserted into the DA promoter region. According to some embodiments, the recombinant adenovirus comprises deleted E1B and E3 region and the nucleotide sequence for an encoded AFFIMER® polypeptide is inserted into the deleted E1B and E3 region.
Adeno-Associated Virus Vectors (AAV)
AAVs (or “rAAV” for recombinant AAV) are non-enveloped small, single- stranded DNA viruses capable of infecting both dividing and non-dividing cells. Similar to AdV, AAV- based vectors remain in an episomal state in the nucleus and display a limited risk of integration. In contrast to the generally limited durability of AdV-mediated gene transfer, transgene expression can persist for years following intramuscular recombinant AAV (rAAV) vector delivery.
Alipogene tiparvovec (Glybera™), an rAAV encoding the human lipoprotein lipase gene, was approved in 2012 as the first gene therapy product in Europe. Since then, various rAAV- based gene therapy products are currently under clinical evaluation. In the context of antibody gene transfer, a variety of reports have demonstrated in vivo production of an anti-human immune deficiency virus (HIV) mAb in mice following intramuscular injection of the mAb- encoding rAAV. The rAAV vector’s potential for combination therapy has also been demonstrated, i.e. by expressing two mAbs. Similar to AdV, intramuscular and i.v. rAAV administration have been most often pursued. Reviewed in Deal et al. “Engineering humoral immunity as prophylaxis or therapy” 2015 Curr Opin Immunol. 35:113-22. A variety of additional delivery sites have also been demonstrated to achieve more local therapeutic effects, including intracranial, intranasal, intravitreal, intrathecal, intrapleural, and intraperitoneal routes. With the utility of rAAV demonstrated for antibody gene transfer, the present disclosure also specifically contemplates the use of rAAV systems for the delivery of polynucleotide sequences in vivo and the production of bispecific fusion proteins in the body of a patient as a consequence to expression of the rAAV construct.
One important feature to AAV is that these gene transfer viruses are capable of infecting non-dividing cells and various types of cells, making them useful in constructing the polynucleotide delivery system of this disclosure. The detailed descriptions for use and preparation of examples of AAV vectors are found in, for example, U.S. Pat. Nos. 5,139,941 and 4,797,368, as well as LaFace et al, Viology, 162:483486 (1988), Zhou et ah, Exp. Hematol.
(NY), 21:928-933 (1993), Walsh et al, J. Clin. Invest., 94:1440-1448(1994) and Flotte et al., Gene Therapy, 2:29-37(1995). AAV is a good choice of delivery vehicles due to its safety, i.e., genetically engineered (recombinant) does not integrate into the host genome. Likewise, AAV is not pathogenic and not associated with any disease. The removal of viral coding sequences minimizes immune reactions to viral gene expression, and therefore, recombinant AAV does not evoke an inflammatory response.
Typically, a recombinant AAV vims is made by co-transfecting a plasmid containing the gene of interest (i.e., the coding sequence for a bispecific fusion protein) flanked by the two AAV terminal repeats (McLaughlin et ah, J. Virol., 62:1963-1973(1988); Samulski et ah, J. Virol., 63:3822-3828(1989)) and an expression plasmid containing the wild type AAV coding sequences without the terminal repeats (McCarty et ah, J. Virol., 65:2936-2945(1991)). Typically, viral vectors containing polynucleotide are assembled from polynucleotides encoding the AFFIMER® polypeptide containing polypeptide, suitable regulatory elements and elements necessary for expression of the encoded AFFIMER® polypeptide which mediate cell transduction. In some embodiments, adeno-associated viral (AAV) vectors are employed. In a more specific embodiment, the AAV vector is an AAV1, AAV6, or AAV8.
The AAV expression vector which harbors the polynucleotide sequence bounded by AAV ITRs, can be constructed by directly inserting the selected sequence(s) into an AAV genome which has had the major AAV open reading frames (“ORFs”) excised therefrom.
For eukaryotic cells, expression control sequences typically include a promoter, an enhancer, such as one derived from an immunoglobulin gene, SV40, cytomegalovirus, etc. (see above), and a polyadenylation sequence which may include splice donor and acceptor sites. The polyadenylation sequence generally is inserted following the transgene sequences and before the 3' ITR sequence.
Selection of these and other common vector and regulatory elements are conventional, and many such sequences are available. See, e.g., Sambrook et ah, and references cited therein at, for example, pages 3.18-3.26 and 16.17-16.27 and Ausubel et ah, Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1989). Of course, not all vectors and expression control sequences will function equally well to express all of the transgenes of this disclosure. However, one of skill in the art may make a selection among these expression control sequences without departing from the scope of this disclosure. Suitable promoter/enhancer sequences may be selected by one of skill in the art using the guidance provided by this application. Such selection is a routine matter and is not a limitation of the molecule or construct.
Retrovirus Vectors
Non-cytopathic viruses useful in the context of delivery of polynucleotides include retroviruses, the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication- deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo. Standard protocols for producing replication-deficient retroviruses (including the steps of incorporation of exogenous genetic material into a plasmid, transfection of a packaging cell lined with plasmid, production of recombinant retroviruses by the packaging cell line, collection of viral particles from tissue culture media, and infection of the target cells with viral particles) are known to those of skill in the art.
In order to construct a retroviral vector, the bispecific fusion protein coding sequence is inserted into the viral genome in the place of certain viral sequences to produce a replication- defective virus. To produce virions, a packaging cell line containing the gag, pol and env genes but without the LTR (long terminal repeat) and psi (□) components is constructed (Mann et ah, Cell, 33:153-159(1983)). When a recombinant plasmid containing the cytokine gene, LTR and psi is introduced into this cell line, the psi sequence allows the RNA transcript of the recombinant plasmid to be packaged into viral particles, which are then secreted into the culture media (Nicolas and Rubinstein "Retroviral vectors," In: Vectors: A survey of molecular cloning vectors and their uses, Rodriguez and Denhardt (eds.), Stoneham: Butterworth, 494-513(1988)). The media containing the recombinant retroviruses is then collected, optionally concentrated and used for gene delivery system.
Successful gene transfer using such second-generation retroviral vectors has been reported. Kasahara et al. (Science, 266:1373-1376(1994)) prepared variants of moloney murine leukemia virus in which the EPO (erythropoietin) sequence is inserted in the place of the envelope region, consequently, producing chimeric proteins having novel binding properties. Likely, the present gene delivery system can be constructed in accordance with the construction strategies for the second-generation retroviral vector.
In some embodiments, the retrovirus is a "gammaretroviruses", which refers to a genus of the retroviridae family. Examples of gammaretroviruses include mouse stem cell virus, murine leukemia virus, feline leukemia virus, feline sarcoma virus, and avian reticuloendotheliosis viruses.
In some embodiments, the retroviral vector for use in the present disclosure is a lentiviral vector, which refers to a genus of retroviruses that are capable of infecting dividing and non dividing cells and typically produce high viral titers. Several examples of lentiviruses include HIV (human immunodeficiency virus: including HIV type 1, and HIV type 2); equine infectious anemia virus; feline immunodeficiency vims (FIV); bovine immune deficiency vims (BIV); and simian immunodeficiency vims (SIV).
Another class of widely used retroviral vectors that can be used for the delivery and expression of polynucleotide include those based upon murine leukemia vims (MuLV), gibbon ape leukemia vims (GaLV) and combinations thereof (see, e.g., Buchscher et ah, J. Virol. 66:2731-2739, 1992; Johann et ah, J. Virol. 66: 1635-1640, 1992; Sommerfelt et ah, Virol. 176:58-59, 1990; Wilson et ah, J. Virol. 63:2374-2378, 1989; Miller et ah, J. Virol. 65:2220- 2224, 1991; and PCT/US94/05700).
Still other retroviral vectors that can be also be used in the present disclosure include, e.g., vectors based on human foamy vims (HFV) or other vimses in the Spumavims genera. Foamy vimses (FVes) are the largest retrovimses known today and are widespread among different mammals, including all non-human primate species however are absent in humans. This complete apathogenicity qualifies FV vectors as ideal gene transfer vehicles for genetic therapies in humans and clearly distinguishes FV vectors as gene delivery system from HIV-derived and also gammaretrovims-derived vectors.
Suitable retroviral vectors for use herein are described, for example, in U.S. Pat. Nos. 5,399,346 and 5,252,479; and in WIPO publications WO 92/07573, WO 90/06997, WO 89/05345, WO 92/05266 and WO 92/14829, which provide a description of methods for efficiently introducing polynucleotides into human cells using such retroviral vectors. Other retroviral vectors include, for example, mouse mammary tumor vims vectors (e.g., Shackleford et ah, Proc. Natl. Acad. Sci. U.S. A. 85:9655-9659, 1998), lentivimses, and the like.
Additional retroviral viral delivery systems that can be readily adapted for delivery of a transgene encoding a PD-L1 bispecific fusion protein include, merely to illustrate Published PCT Applications WO/2010/045002, WO/2010/148203, WO/2011/126864, WO/2012/058673, WO/2014/066700, WO/2015/021077, WO/2015/148683, WO/2017/040815 - the specifications and FIGS of each of which are incorporated by reference herein.
In some embodiments, a retroviral vector contains all of the cis-acting sequences necessary for the packaging and integration of the viral genome, i.e., (a) a long terminal repeat (LTR), or portions thereof, at each end of the vector; (b) primer binding sites for negative and positive strand DNA synthesis; and (c) a packaging signal, necessary for the incorporation of genomic RNA into virions. More detail regarding retroviral vectors can be found in Boesen, et ah, 1994, Biotherapy 6:291-302; Clowes, et ai, 1994, J. Clin. Invest. 93:644-651; Kiem, et ah, 1994, Blood 83: 1467-1473; Salmons and Gunzberg, 1993, Human Gene Therapy 4: 129-141 ; Miller, et ah, 1993, Meth. Enzymol. 217:581- 599; and Grossman and Wilson, 1993, Curr. Opin. in Genetics and Devel. 3: 110-1 14. In some embodiments, the retrovirus is a recombinant replication competent retrovirus comprising: a polynucleotide sequence encoding a retroviral GAG protein; a polynucleotide sequence encoding a retroviral POL protein; a polynucleotide sequence encoding a retroviral envelope; an oncoretroviral polynucleotide sequence comprising Long-Terminal Repeat (LTR) sequences at the 5' and 3' end of the oncoretroviral polynucleotide sequence; a cassette comprising an internal ribosome entry site (IRES) operably linked to a coding sequence for a bispecific fusion protein, such as for a PD-L1 bispecific fusion protein, wherein the cassette is positioned 5' to the U3 region of the 3' LTR and 3' to the sequence encoding the retroviral envelope; and cis-acting sequences for reverse transcription, packaging and integration in a target cell.
In some embodiments, the retrovirus is a recombinant replication competent retrovirus comprising: a retroviral GAG protein; a retroviral POL protein; a retroviral envelope; a retroviral polynucleotide comprising Long-Terminal Repeat (LTR) sequences at the 3' end of the retroviral polynucleotide sequence, a promoter sequence at the 5' end of the retroviral polynucleotide, the promoter being suitable for expression in a mammalian cell, a gag polynucleotide domain, a pol polynucleotide domain and an env polynucleotide domain; a cassette comprising polynucleotide sequence, wherein the cassette is positioned 5' to the 3' LTR and is operably linked and 3' to the env polynucleotide domain encoding the retroviral envelope; and cis-acting sequences necessary for reverse transcription, packaging and integration in a target cell.
In some embodiments of the recombinant replication competent retrovirus, the envelope is chosen from one of amphotropic, polytropic, xenotropic, 10A1, GALV, Baboon endogenous vims, RD114, rhabdovirus, alphavirus, measles or influenza vims envelopes.
In some embodiments of the recombinant replication competent retrovirus, the retroviral polynucleotide sequence is engineered from a vims selected from the group consisting of murine leukemia vims (MLV) , Moloney murine leukemia vims (MoMLV) , Feline leukemia vims (FeLV) , Baboon endogenous retrovims (BEV) , porcine endogenous vims (PERV) , the cat derived retrovims RD114, squirrel monkey retrovims, Xenotropic murine leukemia vims-related vims (XMRV) , avian reticuloendotheliosis vims (REV) , or Gibbon ape leukemia vims (GALV).
In some embodiments of the recombinant replication competent retrovims, retrovims is a gammaretro vim s .
In some embodiments of the recombinant replication competent retrovims, there is a second cassette comprising a coding sequence for a second therapeutic protein, such as another checkpoint inhibitor polypeptide, a co-stimulatory polypeptide and/or a immuno stimulatory cytokine (merely as examples), e.g., downstream of the cassette. In certain instances, the second cassette can include an internal ribosome entry site (IRES) or a minipromoter or a polIII promoter operably linked to the coding sequence for the second therapeutic protein.
In some embodiments of the recombinant replication competent retrovirus, it is a nonlytic, amphotropic retroviral replicating vector which, preferably, selectively infects and replicates in the cells of the tumor microenvironment.
Other Viral Vectors as Expression Constructs
In the context of vectored intratumoral polynucleotide gene transfer, oncolytic viruses have a distinct advantage, as they can specifically target tumor cells, boost therapeutic bispecific fusion protein expression, and amplify antitumor therapeutic responses. Oncolytic viruses, which overlap with certain viral systems described above, promote anti-tumor responses through selective tumor cell killing and induction of systemic anti-tumor immunity. The mechanisms of action are not fully elucidated but are likely to depend on viral replication within transformed cells, induction of primary cell death, interaction with tumor cell anti- viral elements and initiation of innate and adaptive anti-tumor immunity. Reviewed in Kaufman et al. 2015 “Oncolytic viruses: a new class of immunotherapy drugs” Nat Rev Drug Discov. 14(9):642-62. Many of the oncolytic viruses that are currently in the clinic have a natural tropism for cell surface proteins that are aberrantly expressed by cancer cells. To date, AdV, poxviruses, coxsackieviruses, poliovirus, measles vims, Newcastle disease virus, reovirus, and others have entered into early-phase clinical trials. In 2015, the FDA and EM A approved talimogene laherparepvec (T-VEC, Imlygic™), an oncolytic herpes vims armed with the gene for granulocyte-macrophage colony- stimulating factor (GM-CSF). The self-perpetuating nature of oncolytic vimses makes them an appealing platform for polynucleotide gene transfer of the present disclosure, as transgene products can be amplified along with viral replication, thereby maximizing therapeutic effect. Liu et al. 2008 “Oncolytic adenoviruses for cancer gene therapy” Methods Mol Biol. 433:243-58.
In the case of bispecific fusion proteins that are large fusion proteins, i.e., which comprise other protein domains beyond a single AFFIMER® domain, local intratumoral expression can present an appealing strategy to overcome poor penetration in solid tumors if and where that might be an issue. Beckman et al. (2007) “Antibody constmcts in cancer therapy: protein engineering strategies to improve exposure in solid tumors” Cancer 109(2): 170-9; and Dronca et al. 2015 “Immunomodulatory antibody therapy of cancer: the closer, the better” Clin Cancer Res. 21(5):944-6. Likewise, intratumoral delivery of the polynucleotide and concomitant local expression of the bispecific fusion protein can create a better therapeutic index where dose- limiting toxicities might otherwise prevent reaching the effective intratumoral concentration for efficacy when the bispecific fusion protein is delivered (or expressed) systemically.
In the case of the PD-L1 bispecific fusion proteins of the present disclosure, the immunomodulatory nature of these AFFIMER® polypeptides are very relevant to the use of oncolytic viruses. Indeed, for oncolytic virus therapy, it is desirable to override immune checkpoint inhibitor networks and thereby create a pro-inflammatory environment within the cancer. Numerous clinical trials are currently underway to evaluate the combination of oncolytic viruses and conventional immunomodulatory mAb administration. Kaufman et al. 2015 “Oncolytic viruses: a new class of immunotherapy drugs” Nat Rev Drug Discov. 14(9):642-62; and Lichty et al. 2014 “Going viral with cancer immunotherapy” Nat Rev Cancer. 14(8):559-67. However, systemic treatment with checkpoint-blocking mAbs can lead to severe immune-related adverse effects, which may also be an issue for some embodiments of the subject PD-L1 bispecific fusion proteins, highlighting the opportunity for local therapies, e.g. via polynucleotide -armed oncolytic viruses. Different studies have pursued this approach and can be readily adapted for use with the subject polynucleotides. Dias et al. armed a replication-deficient and - competent oncolytic AdV with an anti-human CTLA-4 mAb. Dias et al. 2012 “Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4” Gene Ther. 19(10):988— 98. Another system recently described (and that can be adapted for use with the polynucleotides of the present disclosure) involved armed oncolytic vaccinia viruses with anti-murine programmed cell death protein 1 (PD-1) Fab, scFv or full-length mAb. Reflecting virus replication, mAb levels in the tumor peaked 3-5 days after intratumoral injection at 9 or 30 pg/ml, depending on the tumor model. Serum mAb levels followed the same trend, albeit threefold or more lower, although mAb detection was lost after 5 days. Intratumorally expressed mAbs lasted longer compared to intratumoral injection of anti- PD-1 mAb protein, with follow-up limited to 11 days after injection. Fab and scFv expression were not reported. Anti-tumor responses of the virus armed with either the anti-PD-1 scFv or mAb were superior to the unarmed virus and as effective as the combination of the unarmed virus and systemic anti-PD-1 mAb protein injections. Kleinpeter et al. 2016 “Vectorization in an oncolytic vaccinia virus of an antibody, a Fab and a scFv against programmed cell death- 1 (PD- 1) allows their intratumoral delivery and an improved tumor- growth inhibition” Oncoimmunology. 5(10):el220467 (online). Also recently, intratumoral administration of a combination of an oncolytic AdV and a helper-dependent AdV, armed with an anti-PD-Ll mini antibody (a scFv CH2-CH3 fusion protein), improved the anti-tumor effect of chimeric antigen receptor (CAR) T cell therapy in mice. The benefits of locally produced anti-PD-Ll mini antibody could not be achieved by anti-PD-Ll IgG infusion plus CAR T-cells and co- administration of an unarmed AdV. Tanoue et al 2017 “Armed oncolytic adenovirus expressing PD-L1 mini-body enhances anti-tumor effects of chimeric antigen receptor T-cells in solid tumors” Cancer Res. 77(8):2040-51. The use of that system, particularly in combination with CAR-T cell therapy, is also contemplated for use in delivering polynucleotide to a target tumor.
Other viral vectors may be employed as a gene delivery system in the present disclosure. Vectors derived from viruses such as vaccinia virus (Puhlmann M. et al., Human Gene Therapy, 10:649-657(1999); Ridgeway, "Mammalian expression vectors," In: Vectors: A survey of molecular cloning vectors and their uses. Rodriguez and Denhardt, eds. Stoneham: Butterworth, 467-492(1988); Baichwal and Sugden, "Vectors for gene transfer derived from animal DNA viruses: Transient and stable expression of transferred genes," In: Kucherlapati R, ed. Gene transfer. New York: Plenum Press, 117-148(1986) and Coupar et al., Gene, 68:1-10(1988)), lentivirus (Wang G. et al., J. Clin. Invest., 104(11):R55-62(1999)), herpes simplex virus (Chamber R., et al., Proc. Natl. Acad. Sci USA, 92:1411-1415(1995)), poxvirus (GCE, NJL, Krupa M, Esteban M., The poxvirus vectors MVA and NYVAC as gene delivery systems for vaccination against infectious diseases and cancer Curr Gene Ther 8(2):97- 120(2008)), reovirus, measles virus, Semliki Forest virus, and polioviruses may be used in the present delivery systems for transferring the gene of interest into cells. They offer several attractive features for various mammalian cells. Also included are hepatitis B viruses.
Non- Viral Vectors
In 1990, Wolff et al. showed how injection of naked plasmid DNA (pDNA) into the skeletal muscle of mice led to the local expression of the encoded protein, kick-starting the field of DNA-based therapeutics. See Wolff et al. 1990 “Direct gene transfer into mouse muscle in vivo” Science. 247(4949 Pt 1): 1465-8. The use of “pDNA” for delivering polynucleotides of the present disclosure waives the need for a virus as biological vector and presents an appealing platform for polynucleotide gene transfer. Compared to viral vectors, pDNA is considered low- immunogenic (allowing e.g. repeated dosing), is cheaper to produce, ship, and store, and has a much longer shelf-life. After entry in the nucleus, pDNA remains in a non-replicating non integrating episomal state and is lost during the breakdown of the nuclear envelope at mitosis. pDNA has no defined restrictions regarding the size of the transgene compared to viral vectors, and its modular nature allows for straightforward molecular cloning, making them easy to manipulate and design for therapeutic use. Hardee et al. 2017 “Advances in non-viral DNA vectors for gene therapy” Genes. 8(2):65. Plasmids are used in about 17% of the ongoing or completed gene therapy clinical trials and showed to be well-tolerated and safe. The method of DNA administration can greatly impact transgene expression. In vivo DNA-mediated polynucleotide gene transfer can utilize such physical methods of transfection used for antibody gene transfer, such as electroporation or hydrodynamic injection. Electroporation presents the propagation of electrical fields within tissues, which induces a transient increase in cell membrane permeability. Electrotransfer of DNA is a multistep process, involving (i) electrophoretic migration of DNA towards the plasma membrane, (ii) DNA accumulation and interaction with the plasma membrane, and (iii) intracellular trafficking of the DNA to the nucleus, after which gene expression can commence. Heller LC. 2015 “Gene electrotransfer clinical trials” Adv Genet. 89:235-62. Intramuscular, intratumoral and intradermal administration have been evaluated in clinical trials and are also suitable target tissues for electroporation of polynucleotides.
Hydrodynamic-based transfection utilizes the i.v. injection of high volumes of pDNA, driving DNA molecules out of the blood circulation and into tissue. Other potentially less invasive physical delivery methods include sonoporation and magnetofection. DNA uptake can also be improved by complexing the molecules with chemical delivery vehicles ( e.g . cationic lipids or polymers and lipid nanoparticles). Such techniques can also be applied to in vivo DNA- mediated polynucleotide gene transfer.
In addition to the choice of delivery method, polynucleotide transgene expression can be improved by modifying the make-up of pDNA constructs. See, for example, Hardee et al. 2017 “Advances in non-viral DNA vectors for gene therapy” Genes 8(2):65; and Simcikova et al. 2015 “Towards effective non-viral gene delivery vector” Biotechnol Genet Eng Rev. 31(1- 2):82-107. Conventional pDNA consists of a transcription unit and bacterial backbone. The transcription unit carries the polynucleotide sequence along with regulatory elements. The bacterial backbone includes elements like an antibiotic resistance gene, an origin of replication, unmethylated CpG motifs, and potentially cryptic expression signals. Some of these sequences are required for the production of plasmid DNA. However, in general, for therapeutic polynucleotide gene therapy the presence of a bacterial backbone will likely be counterproductive. However, there are a variety of different types of available minimal vectors that can be selected, including minicircle DNA (mcDNA) which already been used for antibody gene transfer and can be readily adapted for polynucleotide gene transfer. Minicircles are plasmid molecules devoid of bacterial sequences, generated via a process of recombination, restriction and/or purification. Simcikova et al. 2015 supra. Elimination of the bacterial backbone has shown higher transfection efficiency and prolonged transgene expression in a variety of tissues. Also provided herein is a linear polynucleotide, or linear expression cassette ("LEC"), that is capable of being efficiently delivered to a subject via electroporation and expressing the polynucleotide sequence included therein. The LEC may be any linear DNA devoid of any phosphate backbone. The LEC may contain a promoter, an intron, a stop codon, and/or a polyadenylation signal. The expression of the polynucleotide coding sequence may be controlled by the promoter.
Plasmid Vectors
In some embodiments, the subject polynucleotides are delivered as plasmid vectors. Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et ak, 1989, cited above. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they reduced safety concerns relative to other vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide epitope encoded by polynucleotide within the plasmid. Other plasmids are well known to those of ordinary skill in the art. Additionally, plasmids may be custom designed using restriction enzymes and ligation reactions to remove and add specific fragments of DNA. Plasmids may be delivered by a variety of parenteral, mucosal and topical routes. For example, the DNA plasmid can be injected by intramuscular, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally. It may also be administered into the epidermis or a mucosal surface using a gene-gun. The plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and microencapsulation.
To expand the application and efficiency of using plasmid DNA to deliver polynucleotide to tissue in vivo, different approaches can be pursued based on principles producing higher mAb expression or overall efficacy in prior art reports. A first strategy simply relies on giving multiple or repeated pDNA doses. Kitaguchi et al. 2005 “Immune deficiency enhances expression of recombinant human antibody in mice after nonviral in vivo gene transfer” Int J Mol Med 16(4):683— 8; and Yamazaki et al. 2011 “Passive immune-prophylaxis against influenza virus infection by the expression of neutralizing anti-hemagglutinin monoclonal antibodies from plasmids” Jpn J Infect Dis. 64(l):40-9. Another approach relates to the use of a delivery adjuvant. pDNA electrotransfer can be enhanced by pre-treating the muscle with hyaluronidase, an enzyme that transiently breaks down hyaluronic acid, decreasing the viscosity of the extracellular matrix and facilitating DNA diffusion. Yamazaki et al. 2011, supra; and McMahon et al. 2001 “Optimisation of electrotransfer of plasmid into skeletal muscle by pretreatment with hyaluronidase: increased expression with reduced muscle damage” Gene Ther. 8(16): 1264-70. For antibody gene transfer, this led to an increase in mAb expression by approximately 3.5-fold, achieving plasma peak titers of 3.5 pg/ml with 30 pg pDNA, and can be adapted by one skilled in the art for polynucleotide gene transfer. Still another strategy focuses on antibody or cassette engineering. Following codon-, RNA- and leader sequence-optimization, peak serum mAb or Fab titers have been attained with intramuscular electrotransfer of ‘optimized’ pDNA. See, for example, Flingai et al. 2015 “Protection against dengue disease by synthetic polynucleotide antibody prophylaxis/immunotherapy” Sci Rep. 5:12616.
The purpose of the plasmid is the efficient delivery of polynucleotide sequences to and expression of therapeutic bispecific fusion proteins in a cell or tissue. In particular, the purpose of the plasmid may be to achieve high copy number, avoid potential causes of plasmid instability and provide a means for plasmid selection. As for expression, the polynucleotide cassette contains the necessary elements for expression of the polynucleotide within the cassette. Expression includes the efficient transcription of an inserted gene, polynucleotide sequence, or polynucleotide cassette with the plasmid. Thus, in some aspects, a plasmid is provided for expression of polynucleotide which includes an expression cassette comprising the coding sequence for the bispecific fusion protein; also referred to as a transcription unit. When a plasmid is placed in an environment suitable for epitope expression, the transcriptional unit will express the bispecific fusion protein and anything else encoded in the construct. The transcription unit includes a transcriptional control sequence, which is transcriptionally linked with a cellular immune response element coding sequence. Transcriptional control sequence may include promoter/enhancer sequences such as cytomegalovirus (CMV) promoter/enhancer sequences, such as described above. However, those skilled in the art will recognize that a variety of other promoter sequences suitable for expression in mammalian cells, including human patient cells, are known and can similarly be used in the constructs disclosed herein. The level of expression of the bispecific fusion protein will depend on the associated promoter and the presence and activation of an associated enhancer element.
In some embodiments, the polynucleotide sequence (encoding the desired bispecific fusion protein) can be cloned into an expression plasmid which contains the regulatory elements for transcription, translation, RNA stability and replication (i.e., including a transcriptional control sequence). Such expression plasmids are well known in the art and one of ordinary skill would be capable of designing an appropriate expression construct for producing a recombinant bispecific fusion protein in vivo. Minicircle
Minicircle (mcDNA) -based antibody gene transfer can also be adapted for delivery of polynucleotide to tissues in vivo. Under certain circumstances, plasmid DNA used for non-viral gene delivery can cause unacceptable inflammatory responses. Where this happens, immunotoxic responses are largely due to the presence of unmethylated CpG motifs and their associated stimulatory sequences on plasmids following bacterial propagation of plasmid DNA. Simple methylation of DNA in vitro may be enough to reduce an inflammatory response but can result in reduced gene expression. The removal of CpG islands by cloning out, or elimination of non-essential sequences has been a successful technique for reducing inflammatory responses. Yew et al. 2000 “Reduced inflammatory response to plasmid DNA vectors by elimination and inhibition of immuno stimulatory CpG motifs” Mol Ther 1(3), 255-62.
Since bacterial DNA contains on average 4 times more CpG islands than mammalian DNA, a good solution is to eliminate entirely the bacterial control regions, such as the origin of replication and antibiotic resistance genes, from gene delivery vectors during the process of plasmid production. Thus, the "parent" plasmid is recombined into a "minicircle" which generally comprises the gene to be delivered (in this case, the polynucleotide coding sequence) and suitable control regions for its expression, and a miniplasmid which generally comprises the remainder of the parent plasmid.
Removal of bacterial sequences needs to be efficient, using the smallest possible excision site, whilst creating supercoiled DNA minicircles which consist solely of gene expression elements under appropriate— preferably mammalian— control regions. Some techniques for minicircle production use bacterial phage lambda (l) integrase mediated recombination to produce minicircle DNA. See, for example, Darquet, et al. 1997 Gene Ther 4(12): 1341-9; Darquet et al. 1999 Gene Ther 6(2): 209-18; and Kreiss, et al. 1998 Appl Micbiol Biotechnol 49(5):560-7).
Therefore, embodiments of polynucleotide constructs described herein may be processed in the form of minicircle DNA. Minicircle DNA pertains to small (2-4 kb) circular plasmid derivatives that have been freed from all prokaryotic vector parts. Since minicircle DNA vectors contain no bacterial DNA sequences, they are less likely to be perceived as foreign and destroyed. As a result, these vectors can be expressed for longer periods of time compared to certain conventional plasmids. The smaller size of minicircles also extends their cloning capacity and facilitates their delivery into cells. Kits for producing minicircle DNA are known in the art and are commercially available (System Biosciences, Inc., Palo Alto, Calif.). Information on minicircle DNA is provided in Dietz et al., Vector Engineering and Delivery Molecular Therapy (2013); 21 8, 1526-1535 and Hou et al., Molecular Therapy — Methods & Clinical Development, Article number: 14062 (2015) doi:10.1038/mtm.2014.62. More information on Minicircles is provided in Chen Z Y, He C Y, Ehrhardt A, Kay M A. Mol Ther. 2003 September; 8(3):495-500 and Minicircle DNA vectors achieve sustained expression reflected by active chromatin and transcriptional level. Gracey Maniar L E, Maniar J M, Chen Z Y, Lu J, Fire A Z, Kay M A. Mol Ther. 2013 January; 21(1): 131-8
As a nonlimiting example, a minicircle DNA vector may be produced as follows. An expression cassette, which comprises the polynucleotide coding sequence along with regulatory elements for its expression, is flanked by attachment sites for a recombinase. A sequence encoding the recombinase is located outside of the expression cassette and includes elements for inducible expression (such as, for example, an inducible promoter). Upon induction of recombinase expression, the vector DNA is recombined, resulting in two distinct circular DNA molecules. One of the circular DNA molecules is relatively small, forming a minicircle that comprises the expression cassette for the polynucleotide; this minicircle DNA vector is devoid of any bacterial DNA sequences. The second circular DNA sequence contains the remaining vector sequence, including the bacterial sequences and the sequence encoding the recombinase. The minicircle DNA containing the polynucleotide sequence can then be separately isolated and purified. In some embodiments, a minicircle DNA vector may be produced using plasmids similar to pBAD.c|).C31.hFIX and pBAD.c|).C31.RHB. See, e.g., Chen et al. (2003) Mol. Ther. 8:495-500.
Examples of recombinases that may be used for creating a minicircle DNA vector include, but are not limited to, Streptomyces bacteriophage f31 integrase, Cre recombinase, and the l integrase/DNA topoisomerase IV complex. Each of these recombinases catalyzes recombination between distinct sites. For example, f31 integrase catalyzes recombination between corresponding attP and attB sites, Cre recombinase catalyzes recombination between loxP sites, and the l integrase/DNA topoisomerase IV complex catalyzes recombination between bacteriophage l attP and attB sites. In some embodiments, such as, for example, with f31 integrase or with l integrase in the absence of the l is protein, the recombinase mediates an irreversible reaction to yield a unique population of circular products and thus high yields. In other embodiments, such as, for example, with Cre recombinase or with l integrase in the presence of the l protein, the recombinase mediates a reversible reaction to yield a mixture of circular products and thus lower yields. The reversible reaction by Cre recombinase can be manipulated by employing mutant loxP71 and loxP66 sites, which recombine with high efficiency to yield a functionally impaired P71/66 site on the minicircle molecule and a wild-type loxP site on the minicircle molecule, thereby shifting the equilibrium towards the production of the minicircle DNA product. Published US Application 20170342424 also describes a system making use of a parent plasmid which is exposed to an enzyme which causes recombination at recombination sites, thereby forming a (i) minicircle including the polynucleotide sequence and (ii) a miniplasmid comprising the remainder of the parent plasmid. One recombination site is modified at the 5' end such that its reaction with the enzyme is less efficient than the wild type site, and the other recombination site is modified at the 3' end such that its reaction with the enzyme is less efficient than the wild type site, and the other recombination site is modified at the 3' end such that its reaction with the enzyme is less efficient than the wild type site, both modified sites being located in the minicircle after recombination. This favors the formation of minicircle.
RNA-mediated Polynucleotide Gene Transfer
Examples of polynucleotides for the encoded PD-L1 bispecific fusion proteins of the present disclosure include, but are not limited to, ribopolynucleotides (RNAs), deoxyribopolynucleo tides (DNAs), threose polynucleotides (TNAs), glycol polynucleotides (GNAs), peptide polynucleotides (PNAs), locked polynucleotides (LNAs, including LNA having a b- D-ribo configuration, a-LNA having an a-L-ribo configuration (a diastereomer of LNA), 2'- amino-LNA having a 2 '-amino functionalization, and 2'-amino- a-LNA having a 2'-amino functionalization), ethylene polynucleotides (ENA), cyclohexenyl polynucleotides (CeNA) or hybrids or combinations thereof. mRNA presents an emerging platform for antibody gene transfer that can be adapted by those skilled in the art for delivery of polynucleotides of the present disclosure. Although current results differ considerably, in certain instances the mRNA constructs appear to be able to rival viral vectors in terms of generated serum mAb titers. Levels were in therapeutically relevant ranges within hours after mRNA administration, a marked shift in speed compared to DNA. The use of lipid nanoparticles (LNP) for mRNA transfection, rather than the physical methods typically required for DNA, can provide significant advantages in some embodiments towards application range.
In their 1990 study, Wolff et al. (1990, supra ) found that, in addition to pDNA, intramuscular injection of in vitro transcribed (IVT) mRNA also led to local expression of the encoded protein. mRNA was not pursued as actively as DNA at that time because of its low stability. Progress over the past years allowed mRNA to catch up with DNA and viral vectors as a tool for gene transfer. Reviewed in Sahin et al. (2014) “mRNA-based therapeutics: developing a new class of drugs” Nat Rev Drug Discov. 13(10):759-80. Conceptually, there are several differences with these expression platforms. mRNA does not need to enter into the nucleus to be functional. Once it reaches the cytoplasm, mRNA is translated instantly. mRNA-based therapeutics are expressed more transiently compared to DNA- or viral vector-mediated gene transfer, and do not pose the risk of insertional mutagenesis in the host genome. mRNA production is relatively simple and inexpensive. In terms of administration, mRNA uptake can be enhanced using electroporation. Broderick et al. 2017 “Enhanced delivery of DNA or RNA vaccines by electroporation” Methods Mol Biol. 2017;1499:193-200. Most focus, however, has gone to non-physical transfection methods. Indeed, a variety of mRNA complexing formulations have been developed, including lipid nanoparticles (LNP), which have proven to be safe and very efficient mRNA carriers for administration in a variety of tissues and i.v. Pardi et al. 2015 “Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes” J Control Release 217:345-51. In line with this progress, IVT mRNA has reached the stage of clinical evaluation.
Beissert et al. WO2017162266 “RNA Replicon for Versatile and Efficient Gene Expression” describes agents and methods suitable for efficient expression of AFFIMER® polypeptides of the present disclosure, such as suitable for immunotherapeutic treatment for the prevention and therapy of tumors. For instance, the bispecific fusion protein coding sequence can be provided as an RNA replicon comprising a 5' replication recognition sequence such as from an alphavirus 5' replication recognition sequence. In some embodiments, the RNA replicon comprises a (modified) 5' replication recognition sequence and an open reading frame encoding the bispecific fusion protein, in particular located downstream from the 5' replication recognition sequence such as that the 5' replication recognition sequence and the open reading frame do not overlap, e.g. the 5' replication recognition sequence does not contain a functional initiation codon and in some embodiments does not contain any initiation codon. Most preferably, the initiation codon of the open reading frame encoding the bispecific fusion protein is in the 5' 3' direction of the RNA replicon.
In some embodiments, to prevent immune activation, modified nucleosides can be incorporated into the in vitro-transcribed mRNA. In some embodiments, the IVT RNA can be 5’ capped, such an m7G5 'ppp5 'G2 '-O- ct-cappcd IVT. Efficient translation of the modified mRNA can be ensured by removing double-stranded RNA. Moreover, the 5' and 3' UTRs and the poly(A) tail can be optimized for improved intracellular stability and translational efficiency. See, for example, Stadler et al. (2017) Nature Medicine 23:815-817 and Kariko et al. WO/2017/036889 “Method for Reducing Immunogenicity of RNA”.
In some embodiments, the mRNA that encodes the PD-L1 bispecific fusion protein may include at least one chemical modification described herein. As a non-limiting example, the chemical modification may be 1 -methylpseudouridine, 5-methylcytosine or 1 - methylpseudouridine and 5-methylcytosine. In some embodiments, linear polynucleotides encoding at least one PD-L1 bispecific fusion proteins of the present disclosure which are made using only in vitro transcription (IVT) enzymatic synthesis methods are referred to as "IVT polynucleotides." Methods of making IVT polynucleotides are known in the art and are described in PCT Application WO2013/151666, the contents of which are incorporated herein by reference in their entirety.
In another embodiment, the polynucleotides that encode the PD-L1 bispecific fusion protein of the present disclosure have portions or regions which differ in size and/or chemical modification pattern, chemical modification position, chemical modification percent or chemical modification population and combinations of the foregoing are known as "chimeric polynucleotides." A "chimera" according to the present disclosure is an entity having two or more incongruous or heterogeneous parts or regions. As used herein a "part" or "region" of a polynucleotide is defined as any portion of the polynucleotide which is less than the entire length of the polynucleotide. Such constructs are taught in for example PCT Application WO20 15/034928.
In yet another embodiment, the polynucleotides of the present disclosure that are circular are known as "circular polynucleotides" or "circP." As used herein, "circular polynucleotides" or "circP" means a single stranded circular polynucleotide which acts substantially like, and has the properties of, an RNA. The term "circular" is also meant to encompass any secondary or tertiary configuration of the circP. Such constructs are taught in for example PCT Application WO2015/034925 and WO2015/034928, the contents of each of which are incorporated herein by reference in their entirety.
Examples of mRNA (and other polynucleotides) that can be used to encode PD-L1 bispecific fusion proteins of the present disclosure include those which can be adapted from the specifications and FIGS of, for example, PCT Publications WO2017/049275, WO2016/118724, WO2016/118725, W02016/011226, WO2015/196128, WO/2015/196130, WO/2015/196118, WO/2015/089511, with WO2015/105926 (the later titled “Polynucleotides for the In vivo Production Of Antibodies”), each of which is incorporated by reference herein.
Electroporation, as described below, is one example of a method for introducing mRNA or other polynucleotides into a cell.
Lipid-containing nanoparticle compositions have proven effective as transport vehicles into cells and/or intracellular compartments for a variety of RNAs (and related polynucleotides described herein). These compositions generally include at least one "cationic" and/or ionizable lipids, phospholipids including polyunsaturated lipids, structural lipids (e.g., sterols), and lipids containing polyethylene glycol (PEG lipids). Cationic and/or ionizable lipids include, for example, amine-containing lipids that can be readily protonated. Delivery of Polynucleotide Constructs into Target Cells
The introduction into host cell of the gene delivery system can be performed through various methods known to those skilled in the art.
Where the present gene delivery system is constructed on the basis of viral vector construction, delivery can be performed as conventional infection methods known in the art.
Physical methods to enhance delivery both viral and non-viral polynucleotides include electroporation (Neumann, E. et ah, EMBO J., 1:841(1982); and Tur-Kaspa et ah, Mol. Cell Biol., 6:716-718(1986)), gene bombardment (Yang et ah, Proc. Natl. Acad. Sci., 87:9568-9572 (1990) where DNA is loaded onto ( e.g ., gold) particles and forced to achieve penetration of the DNA into the cells, sonoporation, magnetofection, hydrodynamic delivery and the like, all of which are known to those of skill in the art.
Electroporation
In the past several years, there has been a great advance in the plasmid DNA delivery technology that is utilized for in vivo production of proteins. This included codon optimization for expression in human cells, RNA optimization to improve mRNA stability as well as more efficient translation at the ribosomal level, the addition of specific leader sequences to enhance translation efficiency, the creation of synthetic inserts to further enhance production in vivo and the use of improved adaptive electroporation (EP) delivery protocols to improve in vivo delivery. EP assists in the delivery of plasmid DNA by generating an electrical field that allows the DNA to pass into the cell more efficiently. In vivo electroporation is a gene delivery technique that has been used successfully for efficient delivery of plasmid DNA to many different tissues. Kim et al. “Gene therapy using plasmid DNA-encoded anti-HER2 antibody for cancers that overexpress HER2” (2016) Cancer Gene Ther. 23(10): 341-347 teaches a vector and electroporation system for intramuscular injection and in vivo electroporation of the plasmids that results in high and sustained antibody expression in sera; the plasmid and electroporation system of Kim et al. can be readily adapted for the in vivo delivery of a plasmid for expressing an encoded PD-L1 binding AFFIMER® polypeptide of the present disclosure.
Accordingly, in certain some embodiments of the present disclosure, the polynucleotide is introduced into target cells via electroporation.
Administration of the composition via electroporation may be accomplished using electroporation devices that can be configured to deliver to a desired tissue of a mammal, a pulse of energy effective to cause reversible pores to form in cell membranes, and preferable the pulse of energy is a constant current similar to a preset current input by a user. The electroporation device may comprise an electroporation component and an electrode assembly or handle assembly. The electroporation component may include and incorporate at least one of the various elements of the electroporation devices, including: controller, current waveform generator, impedance tester, waveform logger, input element, status reporting element, communication port, memory component, power source, and power switch. The electroporation may be accomplished using an in vivo electroporation device, for example CELLECTRA EP system (VGX Pharmaceuticals, Blue Bell, Pa.) or Eigen electroporator (Genetronics, San Diego, Calif.) to facilitate transfection of cells by the plasmid.
The electroporation component may function as one element of the electroporation devices, and the other elements are separate elements (or components) in communication with the electroporation component. The electroporation component may function as more than one element of the electroporation devices, which may be in communication with still other elements of the electroporation devices separate from the electroporation component. The elements of the electroporation devices existing as parts of one electromechanical or mechanical device may not limited as the elements can function as one device or as separate elements in communication with one another. The electroporation component may be capable of delivering the pulse of energy that produces the constant current in the desired tissue and includes a feedback mechanism. The electrode assembly may include an electrode array having a plurality of electrodes in a spatial arrangement, wherein the electrode assembly receives the pulse of energy from the electroporation component and delivers same to the desired tissue through the electrodes. At least one of the plurality of electrodes is neutral during delivery of the pulse of energy and measures impedance in the desired tissue and communicates the impedance to the electroporation component. The feedback mechanism may receive the measured impedance and can adjust the pulse of energy delivered by the electroporation component to maintain the constant current.
A plurality of electrodes may deliver the pulse of energy in a decentralized pattern. The plurality of electrodes may deliver the pulse of energy in the decentralized pattern through the control of the electrodes under a programmed sequence, and the programmed sequence is input by a user to the electroporation component. The programmed sequence may comprise a plurality of pulses delivered in sequence, wherein each pulse of the plurality of pulses is delivered by at least two active electrodes with one neutral electrode that measures impedance, and wherein a subsequent pulse of the plurality of pulses is delivered by a different one of at least two active electrodes with one neutral electrode that measures impedance.
The feedback mechanism may be performed by either hardware or software. The feedback mechanism may be performed by an analog closed-loop circuit. The feedback occurs every 50 ps, 20 ps, 10 ps or 1 ps, but in some embodiments is a real-time feedback or instantaneous (i.e., substantially instantaneous as determined by available techniques for determining response time). The neutral electrode may measure the impedance in the desired tissue and communicates the impedance to the feedback mechanism, and the feedback mechanism responds to the impedance and adjusts the pulse of energy to maintain the constant current at a value similar to the preset current. The feedback mechanism may maintain the constant current continuously and instantaneously during the delivery of the pulse of energy.
Examples of electroporation devices and electroporation methods that may facilitate delivery of the polynucleotides of the present disclosure, include those described in U.S. Pat.
Nos. 7,245,963; 6,302,874; 5,676,646; 6,241,701; 6,233,482; 6,216,034; 6,208,893; 6,192,270; 6,181,964; 6,150,148; 6,120,493; 6,096,020; 6,068,650; and 5,702,359, the contents of which are incorporated herein by reference in their entirety. The electroporation may be carried out via a minimally invasive device.
In some embodiments, the electroporation is carried using a minimally invasive electroporation device ("MID"). The device may comprise a hollow needle, DNA cassette, and fluid delivery means, wherein the device is adapted to actuate the fluid delivery means in use so as to concurrently (for example, automatically) inject the polynucleotide polynucleotide construct into body tissue during insertion of the needle into the body tissue. This has the advantage that the ability to inject the DNA and associated fluid gradually while the needle is being inserted leads to a more even distribution of the fluid through the body tissue. The pain experienced during injection may be reduced due to the distribution of the DNA being injected over a larger area.
The MID may inject the polynucleotide into tissue without the use of a needle. The MID may inject the polynucleotide as a small stream or jet with such force that the polynucleotide pierces the surface of the tissue and enters the underlying tissue and/or muscle. The force behind the small stream or jet may be provided by expansion of a compressed gas, such as carbon dioxide through a micro-orifice within a fraction of a second. Examples of minimally invasive electroporation devices, and methods of using them, are described in published U.S. Patent Application No. 20080234655; U.S. Pat. No. 6,520,950; U.S. Pat. No. 7,171,264; U.S. Pat. No. 6,208,893; U.S. Pat. No. 6,009,347; U.S. Pat. No. 6,120,493; U.S. Pat. No. 7,245,963; U.S. Pat. No. 7,328,064; and U.S. Pat. No. 6,763,264, the contents of each of which are herein incorporated by reference.
The MID may comprise an injector that creates a high-speed jet of liquid that painlessly pierces the tissue. Such needle-free injectors are commercially available. Examples of needle- free injectors that can be utilized herein include those described in U.S. Pat. Nos. 3,805,783; 4,447,223; 5,505,697; and 4,342,310, the contents of each of which are herein incorporated by reference.
A desired polynucleotide in a form suitable for direct or indirect electro transport may be introduced ( e.g ., injected) using a needle-free injector into the tissue to be treated, usually by contacting the tissue surface with the injector so as to actuate delivery of a jet of the agent, with sufficient force to cause penetration of the polynucleotide into the tissue. For example, if the tissue to be treated is a mucosa, skin or muscle, the agent is projected towards the mucosal or skin surface with sufficient force to cause the agent to penetrate through the stratum comeum and into dermal layers, or into underlying tissue and muscle, respectively. Needle-free injectors are well suited to deliver polynucleotides to all types of tissues, including into tumors (intratumoral delivery).
The MID may have needle electrodes that electroporate the tissue. By pulsing between multiple pairs of electrodes in a multiple electrode array, for example set up in rectangular or square patterns, provides improved results over that of pulsing between a pair of electrodes. Disclosed, for example, in U.S. Pat. No. 5,702,359 entitled "Needle Electrodes for Mediated Delivery of Drugs and Genes" is an array of needles wherein a plurality of pairs of needles may be pulsed during the therapeutic treatment. In that application, which is incorporated herein by reference as though fully set forth, needles were disposed in a circular array, but have connectors and switching apparatus enabling a pulsing between opposing pairs of needle electrodes. A pair of needle electrodes for delivering the polynucleotide to cells may be used. Such a device and system is described in U.S. Pat. No. 6,763,264, the contents of which are herein incorporated by reference. Alternatively, a single needle device may be used that allows injection of the DNA and electroporation with a single needle resembling a normal injection needle and applies pulses of lower voltage than those delivered by presently used devices, thus reducing the electrical sensation experienced by the patient.
The MID may comprise at least one electrode arrays. The arrays may comprise two or more needles of the same diameter or different diameters. The needles may be evenly or unevenly spaced apart. The needles may be between 0.005 inches and 0.03 inches, between 0.01 inches and 0.025 inches; or between 0.015 inches and 0.020 inches. The needle may be 0.0175 inches in diameter. The needles may be 0.5 mm, 1.0 mm, 1.5 mm, 2.0 mm, 2.5 mm, 3.0 mm, 3.5 mm, 4.0 mm, or more spaced apart.
The MID may consist of a pulse generator and a two or more-needle vaccine injectors that deliver the polynucleotide and electroporation pulses in a single step. The pulse generator may allow for flexible programming of pulse and injection parameters via a flash card operated personal computer, as well as comprehensive recording and storage of electroporation and patient data. The pulse generator may deliver a variety of volt pulses during short periods of time. For example, the pulse generator may deliver three 15 volt pulses of 100 ms in duration.
An example of such a MID is the Eigen 1000 system by Inovio Biomedical Corporation, which is described in U.S. Pat. No. 7,328,064, the contents of which are herein incorporated by reference.
The MID may be a CELLECTRA (Inovio Pharmaceuticals, Plymouth Meeting, Pa.) device and system, which is a modular electrode system, that facilitates the introduction of a macromolecule, such as polynucleotide, into cells of a selected tissue in a body. The modular electrode system may comprise a plurality of needle electrodes; a hypodermic needle; an electrical connector that provides a conductive link from a programmable constant-current pulse controller to the plurality of needle electrodes; and a power source. An operator can grasp the plurality of needle electrodes that are mounted on a support structure and firmly insert them into the selected tissue in a body or plant. The polynucleotide is then delivered via the hypodermic needle into the selected tissue. The programmable constant-current pulse controller is activated and constant-current electrical pulse is applied to the plurality of needle electrodes. The applied constant-current electrical pulse facilitates the introduction of the polynucleotide into the cell between the plurality of electrodes. Cell death due to overheating of cells is minimized by limiting the power dissipation in the tissue by virtue of constant-current pulses. The Cellectra device and system is described in U.S. Pat. No. 7,245,963, the contents of which are herein incorporated by reference.
The MID may be an Eigen 1000 system (Inovio Pharmaceuticals). The Eigen 1000 system may comprise device that provides a hollow needle; and fluid delivery means, wherein the apparatus is adapted to actuate the fluid delivery means in use so as to concurrently (for example automatically) inject fluid, the described polynucleotide herein, into body tissue during insertion of the needle into the said body tissue. The advantage is the ability to inject the fluid gradually while the needle is being inserted leads to a more even distribution of the fluid through the body tissue. It is also believed that the pain experienced during injection is reduced due to the distribution of the volume of fluid being injected over a larger area.
In addition, the automatic injection of fluid facilitates automatic monitoring and registration of an actual dose of fluid injected. This data can be stored by a control unit for documentation purposes if desired.
It will be appreciated that the rate of injection could be either linear or non-linear and that the injection may be carried out after the needles have been inserted through the skin of the subject to be treated and while they are inserted further into the body tissue. Suitable tissues into which fluid may be injected by the apparatus of the present disclosure include tumor tissue, skin and other epithelial tissues, liver tissue and muscle tissue, merely as examples.
The apparatus further comprises needle insertion means for guiding insertion of the needle into the body tissue. The rate of fluid injection is controlled by the rate of needle insertion. This has the advantage that both the needle insertion and injection of fluid can be controlled such that the rate of insertion can be matched to the rate of injection as desired. It also makes the apparatus easier for a user to operate. If desired means for automatically inserting the needle into body tissue could be provided.
A user could choose when to commence injection of fluid. Ideally however, injection is commenced when the tip of the needle has reached the target tissue and the apparatus may include means for sensing when the needle has been inserted to a sufficient depth for injection of the fluid to commence. This means that injection of fluid can be prompted to commence automatically when the needle has reached a desired depth (which will normally be the depth at which muscle tissue begins). The depth at which muscle tissue begins could for example be taken to be a preset needle insertion depth such as a value of 4 mm which would be deemed sufficient for the needle to get through the skin layer.
The sensing means may comprise an ultrasound probe. The sensing means may comprise a means for sensing a change in impedance or resistance. In this case, the means may not as such record the depth of the needle in the body tissue but will rather be adapted to sense a change in impedance or resistance as the needle moves from a different type of body tissue into muscle. Either of these alternatives provides a relatively accurate and simple to operate means of sensing that injection may commence. The depth of insertion of the needle can further be recorded if desired and could be used to control injection of fluid such that the volume of fluid to be injected is determined as the depth of needle insertion is being recorded.
The apparatus may further comprise: a base for supporting the needle; and a housing for receiving the base therein, wherein the base is moveable relative to the housing such that the needle is retracted within the housing when the base is in a first rearward position relative to the housing and the needle extends out of the housing when the base is in a second forward position within the housing. This is advantageous for a user as the housing can be lined up on the skin of a patient, and the needles can then be inserted into the patient's skin by moving the housing relative to the base.
As stated above, it is desirable to achieve a controlled rate of fluid injection such that the fluid is evenly distributed over the length of the needle as it is inserted into the skin. The fluid delivery means may comprise piston driving means adapted to inject fluid at a controlled rate. The piston driving means could for example be activated by a servo motor. However, the piston driving means may be actuated by the base being moved in the axial direction relative to the housing. It will be appreciated that alternative means for fluid delivery could be provided. Thus, for example, a closed container which can be squeezed for fluid delivery at a controlled or non- controlled rate could be provided in the place of a syringe and piston system.
The apparatus described above could be used for any type of injection. It is however envisaged to be particularly useful in the field of electroporation and so it may further comprise means for applying a voltage to the needle. This allows the needle to be used not only for injection but also as an electrode during, electroporation. This is particularly advantageous as it means that the electric field is applied to the same area as the injected fluid. There has traditionally been a problem with electroporation in that it is very difficult to accurately align an electrode with previously injected fluid and so users have tended to inject a larger volume of fluid than is required over a larger area and to apply an electric field over a higher area to attempt to guarantee an overlap between the injected substance and the electric field. Using the present disclosure, both the volume of fluid injected and the size of electric field applied may be reduced while achieving a good fit between the electric field and the fluid.
U.S. Pat. No. 7,245,963 by Draghia-Akli, et al. describes modular electrode systems and their use for facilitating the introduction of a biomolecule into cells of a selected tissue in a body or plant. The modular electrode systems may comprise a plurality of needle electrodes; a hypodermic needle; an electrical connector that provides a conductive link from a programmable constant-current pulse controller to the plurality of needle electrodes; and a power source. An operator can grasp the plurality of needle electrodes that are mounted on a support structure and firmly insert them into the selected tissue in a body or plant. The biomolecules are then delivered via the hypodermic needle into the selected tissue. The programmable constant-current pulse controller is activated and constant-current electrical pulse is applied to the plurality of needle electrodes. The applied constant-current electrical pulse facilitates the introduction of the biomolecule into the cell between the plurality of electrodes. The entire content of U.S. Pat. No. 7,245,963 is hereby incorporated by reference.
U.S. Patent Pub. 2005/0052630 submitted by Smith, et al. describes an electroporation device which may be used to effectively facilitate the introduction of a biomolecule into cells of a selected tissue in a body or plant. The electroporation device comprises an electro-kinetic device (“EKD device”) whose operation is specified by software or firmware. The EKD device produces a series of programmable constant-current pulse patterns between electrodes in an array based on user control and input of the pulse parameters and allows the storage and acquisition of current waveform data. The electroporation device also comprises a replaceable electrode disk having an array of needle electrodes, a central injection channel for an injection needle, and a removable guide disk. The entire content of U.S. Patent Pub. 2005/0052630 is hereby incorporated by reference.
The electrode arrays and methods described in U.S. Pat. No. 7,245,963 and U.S. Patent Pub. 2005/0052630 may be adapted for deep penetration into not only tissues such as muscle, but also other tissues or organs. Because of the configuration of the electrode array, the injection needle (to deliver the biomolecule of choice) is also inserted completely into the target organ, and the injection is administered perpendicular to the target issue, in the area that is pre delineated by the electrodes. The electrodes described in U.S. Pat. No. 7,245,963 and U.S. Patent Pub. 2005/005263 are, for example, 20 mm long and 21 gauge.
Use of in vivo electroporation enhances plasmid DNA uptake in tumor tissue, resulting in expression within the tumor, and delivers plasmids to muscle tissue, resulting in systemic expression of secreted proteins, such as cytokines (see, e.g., US8026223). Additional examples of techniques, vectors and devices for electroporating PD-L1 bispecific fusion protein transgenes into cells in vivo include PCT Publications WO/2017/106795, WO/2016/161201, WO/2016/154473, WO/2016/112359 and WO/2014/066655.
Typically, the electric fields needed for in vivo cell electroporation are generally similar in magnitude to the fields required for cells in vitro. In some embodiments, the magnitude of the electric field range from approximately, 10 V/cm to about 1500 V/cm, 300 V/cm to 1500 V/cm, or 1000 V/cm to 1500 V/cm. Alternatively, lower field strengths (from about 10 V/cm to 100 V/cm, and more preferably from about 25 V/cm to 75 V/cm) the pulse length is long. For example, when the nominal electric field is about 25-75 V/cm, if is preferred that the pulse length is about 10 msec.
The pulse length can be about 10 s to about 100 ms. There can be any desired number of pulses, typically one to 100 pulses per second. The delay between pulses sets can be any desired time, such as one second. The waveform, electric field strength and pulse duration may also depend upon the type of cells and the type of molecules that are to enter the cells via electroporation.
Also encompassed are electroporation devices incorporating electrochemical impedance spectroscopy ("EIS")· Such devices provide real-time information on in vivo, in particular, intratumoral electroporation efficiency, allowing for the the optimization of conditions.
Examples of electroporation devices incorporating EIS can be found, e.g., in W02016/161201, which is hereby incorporated by reference.
Uptake of the polynucleotides of the present disclosure may also be enhanced by plasma electroporation also termed avalanche transfection. Briefly, microsecond discharges create cavitation microbubbles at electrode surface. The mechanical force created by the collapsing microbubbles combined with the magnetic field serve to increase transport efficiency across the cell membrane as compared with the diffusion mediated transport associated with conventional electroporation. The technique of plasma electroporation is described in United States Patent Nos. 7,923,251 and 8,283,171. This technique may also be employed in vivo for the transformation of cells. Chaiberg, et al (2006) Investigative Ophthalmology & Visual Science 47:4083-4090; Chaiberg, et al United States Patent No 8, 101 169 Issued January 24, 2012.
Other alternative electroporation technologies are also contemplated. In vivo polynucleotide delivery can also be performed using cold plasma. Plasma is one of the four fundamental states of matter, the others being solid, liquid, and gas. Plasma is an electrically neutral medium of unbound positive and negative particles (i.e. the overall charge of a plasma is roughly zero). A plasma can be created by heating a gas or subjecting it to a strong electromagnetic field, applied with a laser or microwave generator. This decreases or increases the number of electrons, creating positive or negative charged particles called ions (Luo, et al. (1998) Phys. Plasma 5:2868-2870) and is accompanied by the dissociation of molecular bonds, if present.
Cold plasmas (i.e., non-thermal plasmas) are produced by the delivery of pulsed high voltage signals to a suitable electrode. Cold plasma devices may take the form of a gas jet device or a dielectric barrier discharge (DBD) device. Cold temperature plasmas have attracted a great deal of enthusiasm and interest by virtue of their provision of plasmas at relatively low gas temperatures. The provision of plasmas at such a temperature is of interest to a variety of applications, including wound healing, anti-bacterial processes, various other medical therapies and sterilization. As noted earlier, cold plasmas (i.e., non-thermal plasmas) are produced by the delivery of pulsed high voltage signals to a suitable electrode. Cold plasma devices may take the form of a gas jet device, a dielectric barrier discharge (DBD) device or multi-frequency harmonic-rich power supply.
Dielectric barrier discharge device, relies on a different process to generate the cold plasma. A dielectric barrier discharge (DBD) device contains at least one conductive electrode covered by a dielectric layer. The electrical return path is formed by the ground that can be provided by the target substrate undergoing the cold plasma treatment or by providing an in-built ground for the electrode. Energy for the dielectric barrier discharge device can be provided by a high voltage power supply, such as that mentioned above. More generally, energy is input to the dielectric barrier discharge device in the form of pulsed DC electrical voltage to form the plasma discharge. By virtue of the dielectric layer, the discharge is separated from the conductive electrode and electrode etching and gas heating is reduced. The pulsed DC electrical voltage can be varied in amplitude and frequency to achieve varying regimes of operation. Any device incorporating such a principle of cold plasma generation (e.g., a DBD electrode device) falls within the scope of various embodiments of the present disclosure.
Cold plasma has been employed to transfect cells with foreign polynucleotides. In particular, transfection of tumor cells (see, e.g., Connolly, et al. (2012) Human Vaccines & Immune-therapeutics 8: 1729-1733; and Connolly et al (2015) Bioelectrochemistry 103: 15-21 ) .
In certain illustrative embodiments, the transgene construct encoding the PD-L1 bispecific fusion protein of the present disclosure is delivered using an electroporation device comprising: an applicator; a plurality of electrodes extending from the applicator, the electrodes being associated with a cover area; a power supply in electrical communication with the electrodes, the power supply configured to generate at least one electroporating signals to cells within the cover area; and a guide member coupled to the electrodes, wherein the guide member is configured to adjust the cover area of the electrodes. At least a portion of the electrodes can be positioned within the applicator in a conical arrangement. The at least one electroporating signals may be each associated with an electric field. The device may further comprise a potentiometer coupled to the power supply and electrodes. The potentiometer may be configured to maintain the electric field substantially within a predetermined range.
The at least one electroporating signals may be each associated with an electric field. The device may further comprise a potentiometer coupled to the power supply and the electrodes.
The potentiometer may be configured to maintain the electric field within a predetermined range so as to substantially prevent permanent damage in the cells within the cover area and /or substantially minimize pain. For instance, potentiometer may be configured to maintain the electric field to about 1300 V/cm.
The power supply may provide a first electrical signal to a first electrode and a second electrical signal to a second electrode. The first and second electrical signals may combine to produce a wave having a beat frequency. The first and second electrical signals may each have at least one of a unipolar waveform and a bipolar waveform. The first electrical signal may have a first frequency and a first amplitude. The second electrical signal may have a second frequency and a second amplitude. The first frequency may be different from or the same as the second frequency. The first amplitude may be different from or the same as the second amplitude.
In some embodiments, the present disclosure provides a method for treating a subject having a tumor, the method comprising: injecting the tumor with an effective dose of plasmid coding for a PD-L1 bispecific fusion protein; and administering electroporation therapy to the tumor. In some embodiments, the electroporation therapy further comprises the administration of at least one voltage pulse of about 200 V/cm to about 1500 V/cm over a pulse width of about 100 microseconds to about 20 milliseconds. In some embodiments, the plasmid (or a second electroporated plasmid) further encodes at least one immuno stimulatory cytokine, such as selected from the group encoding IL-12, IL-15, and a combination of IL-12 and IL-15.
Transfection Enhancing Formulations
Polynucleotide constructs can also be encapsulated in liposomes, preferably cationic liposomes (Wong, T. K. et ah, Gene, 10:87(1980); Nicolau and Sene, Biochim. Biophys. Acta, 721:185-190 (1982); and Nicolau et ah, Methods Enzymol., 149:157-176 (1987)) or polymersomes (synthetic liposomes) which can interact with the cell membrane and fuse or undergo endocytosis to effect polynucleotide transfer into the cell. The DNA also can be formed into complexes with polymers (polyplexes) or with dendrimers which can directly release their load into the cytoplasm of a cell.
Illustrative carriers useful in this regard include microparticles of poly(lactide-co- glycolide), polyacrylate, latex, starch, cellulose, dextran and the like. Other illustrative carriers include supramolecular biovectors, which comprise a non-liquid hydrophilic core (e.g., a cross- linked polysaccharide or oligosaccharide) and, optionally, an external layer comprising an amphiphilic compound, such as a phospholipid (see e.g., U.S. Pat. No. 5,151,254 and PCT applications WO 94/20078, WO/94/23701 and WO 96/06638). The amount of active agent contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented.
Biodegradable microspheres (e.g., polylactate polyglycolate) may be employed as carriers for compositions. Suitable biodegradable microspheres are disclosed, for example, in U.S. Pat. Nos. 4,897,268; 5,075,109; 5,928,647; 5,811,128; 5,820,883; 5,853,763; 5,814,344, 5,407,609 and 5,942,252. Modified hepatitis B core protein carrier systems such as described in WO/9940934, and references cited therein, will also be useful for many applications. Another illustrative carrier/delivery system employs a carrier comprising particulate-protein complexes, such as those described in U.S. Pat. No. 5,928,647, which can have the added benefit when used intratumorally to deliver the coding sequence for a PD-L1 AFFIMER® polypeptide.
Biodegradable polymeric nanoparticles facilitate nonviral polynucleotide transfer to cells. Small (approximately 200 nm), positively charged (approximately 10 mV) particles are formed by the self-assembly of cationic, hydrolytically degradable poly(beta-amino esters) and plasmid DNA.
Polynucleotides may also be administered to cells by direct microinjection, temporary cell permeabilizations (e.g., co-administration of repressor and/or activator with a cell permeabilizing agent), fusion to membrane translocating peptides, and the like. Lipid-mediated polynucleotide delivery and expression of foreign polynucleotides, including mRNA, in vitro and in vivo has been very successful. Lipid based non-viral formulations provide an alternative to viral gene therapies. Current in vivo lipid delivery methods use subcutaneous, intradermal, intratumoral, or intracranial injection. Advances in lipid formulations have improved the efficiency of gene transfer in vivo (see PCT Application WO 98/07408). For instance, a lipid formulation composed of an equimolar ratio of 1,2- bis(oleoyloxy)-3-(trimethyl ammonio)propane (DOTAP) and cholesterol can significantly enhances systemic in vivo gene transfer. The DOTAP:cholesterol lipid formulation forms unique structure termed a "sandwich liposome". This formulation is reported to "sandwich" DNA between an invaginated bi-layer or 'vase' structure. Beneficial characteristics of these lipid structures include a positive p, colloidal stabilization by cholesterol, two dimensional polynucleotide packing and increased serum stability.
Cationic liposome technology is based on the ability of amphipathic lipids, possessing a positively charged head group and a hydrophobic lipid tail, to bind to negatively charged DNA or RNA and form particles that generally enter cells by endocytosis. Some cationic liposomes also contain a neutral co-lipid, thought to enhance liposome uptake by mammalian cells. Similarly, other polycations, such as poly-l-lysine and polyethylene-imine, complex with polynucleotides via charge interaction and aid in the condensation of DNA or RNA into nanoparticles, which are then substrates for endosome-mediated uptake. Several of these cationic -polynucleotide complex technologies have been developed as potential clinical products, including complexes with plasmid DNA (pDNA), oligodeoxynucleotides, and various forms of synthetic RNA, and be used as part of the delivery system for the polynucleotides of the present disclosure.
The polynucleotides disclosed herein may be associated with polycationic molecules that serve to enhance uptake into cells. Complexing the polynucleotide construct with polycationic molecules also helps in packaging the construct such their size is reduced, which is believed to assist with cellular uptake. Once in the endosome, the complex dissociates due to the lower pH, and the polycationic molecules can disrupt the endosome's membrane to facilitate DNA escape into the cytoplasm before it can be degraded. Preliminary data shows that the polynucleotide construct embodiments had enhanced uptake into SCs over DCs when complexed with the polycationic molecules polylysine or polyethyleneimine.
One example of polycationic molecules useful for complexing with polynucleotide constructs includes cell penetrating peptides (CPP), examples include polylysine (described above), polyarginine and Tat peptides. Cell penetrating peptides (CPP) are small peptides which can bind to DNA and, once released, penetrate cell membranes to facilitate escape of the DNA from the endosome to the cytoplasm. Another example of a CPP pertains to a 27 residue chimeric peptide, termed MPG, was shown some time ago to bind ss- and ds-oligonucleotides in a stable manner, resulting in a non-covalent complex that protected the polynucleotides from degradation by DNase and effectively delivered oligonucleotides to cells in vitro (Mahapatro A, et ah, J Nanobiotechnol, 2011, 9:55). The complex formed small particles of approximately 150 nm to 1 um when different peptide:DNA ratios were examined, and the 10:1 and 5:1 ratios (150 nm and 1 um respectively). Another CPP pertains to a modified tetrapeptide [tetralysine containing guanidinocarbonylpyrrole (GCP) groups (TL-GCP)], which was reported to bind with high affinity to a 6.2 kb plasmid DNA resulting in a positive charged aggregate of 700-900 nm Li et al., Agnew Chem Int Ed Enl 2015; 54(10):2941-4). RNA can also be complexed by such polycationic molecules for in vivo delivery.
Other examples of polycationic molecules that may be complexed with the polynucleotide constructs described herein include polycationic polymers commercially available as JETPRIME® and In vivo JET (Polypus-transfection, S.A., Illkirch, France).
In some embodiments, the present disclosure contemplates a method of delivering an mRNA (or other polynucleotide)f encoding a PD-L1 bispecific fusion protein to a patient’s cells by administering a nanoparticle composition comprising (i) a lipid component comprising a compound of formula (I), a phospholipid, a structural lipid, and a PEG lipid; and (ii) an mRNA (or other polynucleotide)f, said administering comprising contacting said mammalian cell with said nanoparticle composition, whereby said mRNA (or other polynucleotide)f is delivered to said cell.
In some embodiments, the PEG lipid is selected from the group consisting of a PEG- modified phosphatidylethanolamine, a PEG-modified phosphatide acid, a PEG-modified ceramide, a PEG-modified dialkylamine, a PEG-modified diacylglycerol and a PEG-modified dialkylglycerol. In some embodiments, the structural lipid is selected from the group consisting of cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, ursolic acid, and alpha- tocopherol. In some embodiments, the structural lipid is cholesterol.
In some embodiments, the phospholipid includes a moiety selected from the group consisting of phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl glycerol, phosphatidyl serine, phosphatidic acid, 2-lysophosphatidyl choline, and a sphingomyelin. In some embodiments, the phospholipid includes at least one fatty acid moieties selected from the group consisting of lauric acid, myristic acid, myristoleic acid, palmitic acid, palmitoleic acid, stearic acid, oleic acid, linoleic acid, alpha-linolenic acid, erucic acid, arachidic acid, arachidonic acid, phytanoic acid, eicosapentaenoic acid, behenic acid, docosapentaenoic acid, and docosahexaenoic acid. In some embodiments, the phospholipid is selected from the group consisting of 1 ,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1 ,2-dimyristoyl-sn-glycero- phosphocholine (DMPC), 1 ,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1 ,2-dipalmitoyl- sn-glycero-3-phosphocholine (DPPC), 1 ,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1 ,2-diundecanoyl-sn-glycero-phosphocholine (DUPC), 1 -palmitoyl-2-oleoyl-sn-glycero-3- phosphocholine (POPC), 1 ,2-di-0-octadecenyl-sn-glycero-3-phosphocholine (1 8:0 Diether PC), 1 -oleoyl-2-cholesterylhemisuccinoyl-sn-glycero-3-phosphocholine (OChemsPC), 1 -hexadecyl- sn-glycero-3-phosphocholine (C16 Lyso PC), 1 ,2-dilinolenoyl-sn-glycero-3-phosphocholine, 1 ,2-diarachidonoyl-sn-glycero-3-phosphocholine, 1 ,2-didocosahexaenoyl-sn-glycero-3- phosphocholine,l ,2-dioleoyl-sn-glycero-3-phosphoethanola mine (DOPE), 1 ,2-diphytanoyl-sn- glycero-3-phosphoethanolamine (ME 1 6.0 PE), 1 ,2-distearoyl-sn-glycero-3- phosphoethanolamine, 1 ,2-dilinoleoyl-sn-glycero-3-phosphoethanolamine, 1 ,2-dilinolenoyl-sn- glycero-3-phosphoethanolamine, 1 ,2-diarachidonoyl-sn-glycero-3-phosphoethanolamine, 1 ,2- didocosahexaenoyl-sn-glycero-3-phosphoethanolamine, 1 ,2-dioleoyl-sn-glycero-3-phospho-rac- (1 -glycerol) sodium salt (DOPG), and sphingomyelin In some embodiments, the phospholipid is DOPE or DSPC.
To further illustrate, the phospholipid can be DOPE and said the component can comprise about 35 mol % to about 45 mol % said compound, about 1 0 mol % to about 20 mol % DOPE, about 38.5 mol % to about 48.5 mol % structural lipid, and about 1 .5 mol % PEG lipid. The lipid component can be about 40 mol % said compound, about 15 mol % phospholipid, about 43.5 mol % structural lipid, and about 1 .5 mol % PEG lipid.
In some embodiments, the wt/wt ratio of lipid component to PD-L1 bispecific fusion protein encoding mRNA (or other polynucleotide) is from about 5:1 to about 50:1, or about 10:1 to about 40: 1
In some embodiments, the mean size of said nanoparticle composition is from about 50 nm to about 150 nm, or from about 80 nm to about 120 nm.
In some embodiments, the polydispersity index of said nanoparticle composition is from about 0 to about 0.18, or from about 0.13 to about 0.17.
In some embodiments, the nanoparticle composition has a zeta potential of about -10 to about +20 mV.
In some embodiments, the nanoparticle composition further comprises a cationic and/or ionizable lipid selected from the group consisting of 3-(didodecylamino)-Nl ,N 1 ,4-tridodecyl-l -piperazineethanamine (KL1 0), 14,25-ditridecyl-l 5, 1 8,21 ,24-tetraaza-octatriacontane (KL25), 1 ,2-dilinoleyloxy-N,N -dimethylaminopropane (DLin-DMA), 2,2-dilinoleyl-4- dimethylaminomethyl-[l ,3]-dioxolane (DLin-K-DMA), heptatriaconta-6, 9,28,31 -tetraen-1 9-yl 4-(dimethylamino)butanoate (DLin-MC3-DMA), 2,2-dilinoleyl-4-(2-dimethylaminoethyl)-[l ,3]- dioxolane (DLin-KC2-DMA), 1 ,2-dioleyloxy-N,N-dimethylaminopropane (DODMA), and (2R)-2-({8-[(3P)-cholest-5-en-3-yloxy]octyl}oxy)-N,N-dimethyl-3-[(9Z,l 2Z)-octadeca-9, 12- dien-1 -yl oxy]propan-l -amine (Octyl-CLinDMA (2R)).
Methods of Use
The bispecific fusion proteins of the disclosure are useful in a variety of applications including, but not limited to, therapeutic treatment methods, such as immunotherapy for cancer. In some embodiments, bispecific fusion proteins described herein are useful for activating, promoting, increasing, and/or enhancing an immune response, inhibiting tumor growth, reducing tumor volume, inducing tumor regression, increasing tumor cell apoptosis, and/or reducing the tumorigenicity of a tumor. In some embodiments, the polypeptides or agents of the disclosure are also useful for immunotherapy against pathogens, such as viruses. In some embodiments, the bispecific fusion proteins described herein are useful for inhibiting viral infection, reducing viral infection, increasing virally-infected cell apoptosis, and/or increasing killing of virus-infected cells. The methods of use may be in vitro, ex vivo, or in vivo methods.
The present disclosure provides methods for activating an immune response in a subject using a bispecific fusion protein. In some embodiments, the disclosure provides methods for promoting an immune response in a subject using a bispecific fusion protein described herein. In some embodiments, the disclosure provides methods for increasing an immune response in a subject using a bispecific fusion protein. In some embodiments, the disclosure provides methods for enhancing an immune response in a subject using a bispecific fusion protein. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing cell-mediated immunity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing Thl-type responses.
In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing T-cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CD4+ T-cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CD8+ T-cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CTL activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing NK cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing T-cell activity and increasing NK cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing CU activity and increasing NK cell activity. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises inhibiting or decreasing the suppressive activity of Treg cells. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises inhibiting or decreasing the suppressive activity of MDSCs. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing the number of the percentage of memory T-cells. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing long-term immune memory function. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises increasing long-term memory. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises no evidence of substantial side effects and/or immune-based toxicities. In some embodiments, the activating, promoting, increasing, and/or enhancing of an immune response comprises no evidence of cytokine release syndrome (CRS) or a cytokine storm. In some embodiments, the immune response is a result of antigenic stimulation. In some embodiments, the antigenic stimulation is a tumor cell. In some embodiments, the antigenic stimulation is cancer. In some embodiments, the antigenic stimulation is a pathogen. In some embodiments, the antigenic stimulation is a virally-infected cell.
In vivo and in vitro assays for determining whether a bispecific fusion protein activates, or inhibits an immune response are known in the art.
In some embodiments, a method of increasing an immune response in a subject comprises administering to the subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein a bispecific fusion protein binds human PD-L1. In some embodiments, a method of increasing an immune response in a subject comprises administering to the subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein the bispecific fusion protein is a AFFIMER® polypeptide-containing antibody or receptor trap fusion polypeptide including an AFFIMER® polypeptide that specifically binds to PD-L1. In some embodiments, a method of increasing an immune response in a subject comprises administering to the subject a therapeutically effective amount of polynucleotide, wherein the polynucleotide, when expressed in the patient, produces a recombinant bispecific fusion protein polypeptide including an anti-PD-Ll AFFIMER® polypeptide.
In some embodiments of the methods described herein, a method of activating or enhancing a persistent or long-term immune response to a tumor comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein which binds human PD- Ll. In some embodiments, a method of activating or enhancing a persistent immune response to a tumor comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein the bispecific fusion protein is an AFFIMER® polypeptide-containing antibody or receptor trap fusion polypeptide including an AFFIMER® polypeptide that specifically binds to PD-L1. In some embodiments, a method of activating or enhancing a persistent immune response to a tumor comprises administering to a subject a therapeutically effective amount of polynucleotide, wherein the polynucleotide, when expressed in the patient, produces a recombinant bispecific fusion protein polypeptide including an anti- PD-L1 AFFIMER® polypeptide.
In some embodiments of the methods described herein, a method of inducing a persistent or long-term immunity which inhibits tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein which binds human PD-L1. In some embodiments, a method of inducing a persistent immunity which inhibits tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein the bispecific fusion protein is a AFFIMER® polypeptide-containing antibody or receptor trap fusion polypeptide including an AFFIMER® polypeptide that specifically binds to PD-L1. In some embodiments, a method of inducing a persistent immunity which inhibits tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of polynucleotide, wherein the polynucleotide, when expressed in the patient, produces a recombinant bispecific fusion protein polypeptide including an anti-PD-Ll AFFIMER® polypeptide.
In some embodiments of the methods described herein, a method of inhibiting tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein which binds human PD-L1. In some embodiments, a method of inhibiting tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein described herein, wherein the bispecific fusion protein is an AFFIMER® polypeptide-containing antibody or receptor trap fusion polypeptide including an AFFIMER® polypeptide that specifically binds to PD-L1. In some embodiments, a method of inhibiting tumor relapse or tumor regrowth comprises administering to a subject a therapeutically effective amount of polynucleotide, wherein the polynucleotide, when expressed in the patient, produces a recombinant bispecific fusion protein polypeptide including an anti-PD-Ll AFFIMER® polypeptide.
In some embodiments, the tumor expresses or overexpresses a tumor antigen that is targeted by an additional binding entity provided in the bispecific fusion protein along with the anti-PD-Ll AFFIMER® polypeptide, i.e., where the bispecific fusion protein is a bispecific or multispecific agent. In some embodiments, the method of inhibiting growth of a tumor comprises administering to a subject a therapeutically effective amount of a bispecific fusion protein described herein. In some embodiments, the subject is a human. In some embodiments, the subject has a tumor, or the subject had a tumor which was removed.
In some embodiments, the tumor is a solid tumor. In some embodiments, the tumor is a tumor selected from the group consisting of: colorectal tumor, pancreatic tumor, lung tumor, ovarian tumor, liver tumor, breast tumor, kidney tumor, prostate tumor, neuroendocrine tumor, gastrointestinal tumor, melanoma, cervical tumor, bladder tumor, glioblastoma, and head and neck tumor. In some embodiments, the tumor is a colorectal tumor. In some embodiments, the tumor is an ovarian tumor. In some embodiments, the tumor is a lung tumor. In some embodiments, the tumor is a pancreatic tumor. In some embodiments, the tumor is a melanoma tumor. In some embodiments, the tumor is a bladder tumor.
To further illustrate, the subject bispecific fusion proteins can be used to treat patients suffering from cancer, such as osteosarcoma, rhabdomyosarcoma, neuroblastoma, kidney cancer, leukemia, renal transitional cell cancer, bladder cancer, Wilm's cancer, ovarian cancer, pancreatic cancer, breast cancer (including triple negative breast cancer), prostate cancer, bone cancer, lung cancer ( e.g ., small cell or non-small cell lung cancer), gastric cancer, colorectal cancer, cervical cancer, synovial sarcoma, head and neck cancer, squamous cell carcinoma, multiple myeloma, renal cell cancer, retinoblastoma, hepatoblastoma, hepatocellular carcinoma, melanoma, rhabdoid tumor of the kidney, Ewing's sarcoma, chondrosarcoma, brain cancer, glioblastoma, meningioma, pituitary adenoma, vestibular schwannoma, a primitive neuroectodermal tumor, medulloblastoma, astrocytoma, anaplastic astrocytoma, oligodendroglioma, ependymoma, choroid plexus papilloma, polycythemia vera, thrombocythemia, idiopathic myelfibrosis, soft tissue sarcoma, thyroid cancer, endometrial cancer, carcinoid cancer or liver cancer, breast cancer or gastric cancer. In some embodiments of the disclosure, the cancer is metastatic cancer, e.g., of the varieties described above.
In some embodiments, the cancer is a hematologic cancer. In some embodiment, the cancer is selected from the group consisting of: acute myelogenous leukemia (AML), Hodgkin lymphoma, multiple myeloma, T-cell acute lymphoblastic leukemia (T-ALL), chronic lymphocytic leukemia (CLL), hairy cell leukemia, chronic myelogenous leukemia (CML), non- Hodgkin lymphoma, diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), and cutaneous T-cell lymphoma (CTCL). Pharmaceutical Compositions/F ormulations
The present disclosure also provides pharmaceutical compositions comprising a bispecific fusion protein described herein and a pharmaceutically acceptable vehicle. In some embodiments, the pharmaceutical compositions find use in immunotherapy. In some embodiments, the pharmaceutical compositions find use in immuno-oncology. In some embodiments, the compositions find use in inhibiting tumor growth. In some embodiments, the pharmaceutical compositions find use in inhibiting tumor growth in a subject ( e.g ., a human patient). In some embodiments, the compositions find use in treating cancer. In some embodiments, the pharmaceutical compositions find use in treating cancer in a subject (e.g., a human patient).
Formulations are prepared for storage and use by combining a purified bispecific fusion protein of the present disclosure with a pharmaceutically acceptable vehicle (e.g., a carrier or excipient). Those of skill in the art generally consider pharmaceutically acceptable carriers, excipients, and/or stabilizers to be inactive ingredients of a formulation or pharmaceutical composition.
In some embodiments, a bispecific fusion protein described herein is lyophilized and/or stored in a lyophilized form. In some embodiments, a formulation comprising a bispecific fusion protein described herein is lyophilized.
Suitable pharmaceutically acceptable vehicles include, but are not limited to, nontoxic buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives such as octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl or benzyl alcohol, alkyl parabens, such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol; low molecular weight polypeptides (e.g., less than about 10 amino acid residues); proteins such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; carbohydrates such as monosaccharides, disaccharides, glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes such as Zn-protein complexes; and non-ionic surfactants such as TWEEN or polyethylene glycol (PEG). (Remington: The Science and Practice of Pharmacy, 22.sup.nd Edition, 2012, Pharmaceutical Press, London.).
The pharmaceutical compositions of the present disclosure can be administered in any number of ways for either local or systemic treatment. Administration can be topical by epidermal or transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders; pulmonary by inhalation or insufflation of powders or aerosols, including by nebulizer, intratracheal, and intranasal; oral; or parenteral including intravenous, intraarterial, intratumoral, subcutaneous, intraperitoneal, intramuscular (e.g., injection or infusion), or intracranial (e.g., intrathecal or intraventricular).
In some embodiments, a composition is formulated for topical delivery such that the when applied to the skin, for example, the bispecific fusion protein penetrates the skin (crosses epithelial and mucosal barriers) to function systemically.
The therapeutic formulation can be in unit dosage form. Such formulations include tablets, pills, capsules, powders, granules, solutions or suspensions in water or non-aqueous media, or suppositories. In solid compositions such as tablets the principal active ingredient is mixed with a pharmaceutical carrier. Conventional tableting ingredients include corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and diluents (e.g., water). These can be used to form a solid preformulation composition containing a homogeneous mixture of a compound of the present disclosure, or a non-toxic pharmaceutically acceptable salt thereof. The solid preformulation composition is then subdivided into unit dosage forms of a type described above. The tablets, pills, etc. of the formulation or composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner composition covered by an outer component. Furthermore, the two components can be separated by an enteric layer that serves to resist disintegration and permits the inner component to pass intact through the stomach or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials include a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.
The bispecific fusion proteins described herein can also be entrapped in microcapsules. Such microcapsules are prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly- (methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nanoparticles and nanocapsules) or in macroemulsions as described in Remington: The Science and Practice of Pharmacy, 22.sup.nd Edition, 2012, Pharmaceutical Press, London.
In some embodiments, pharmaceutical formulations include a bispecific fusion protein of the present disclosure complexed with liposomes. Methods to produce liposomes are known to those of skill in the art. For example, some liposomes can be generated by reverse phase evaporation with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG- derivatized phosphatidylethanolamine (PEG-PE). Liposomes can be extruded through filters of defined pore size to yield liposomes with the desired diameter.
In some embodiments, sustained-release preparations comprising bispecific fusion proteins described herein can be produced. Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing a bispecific fusion protein, where the matrices are in the form of shaped articles ( e.g ., films or microcapsules). Examples of sustained-release matrices include polyesters, hydrogels such as poly(2- hydroxyethyl-methacrylate) or poly(vinyl alcohol), polylactides, copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene- vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT.TM. (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(-)- 3-hydroxybutyric acid.
In some embodiments, in addition to administering a bispecific fusion protein described herein, the method or treatment further comprises administering at least one additional immune response stimulating agent. In some embodiments, the additional immune response stimulating agent includes, but is not limited to, a colony stimulating factor (e.g., granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), granulocyte colony stimulating factor (G-CSF), stem cell factor (SCF)), an interleukin (e.g., IL- 1, IL2, IL-3, IL-7, IL-12, IL-15, IL-18), a checkpoint inhibitor, an antibody that blocks immunosuppressive functions (e.g., an anti-CTLA-4 antibody, anti-CD28 antibody, anti-CD3 antibody), a toll -like receptor (e.g., TLR4, TLR7, TLR9), or a member of the B7 family (e.g., CD80, CD86). An additional immune response stimulating agent can be administered prior to, concurrently with, and/or subsequently to, administration of the bispecific fusion protein. Pharmaceutical compositions comprising a bispecific fusion protein and the immune response stimulating agent(s) are also provided. In some embodiments, the immune response stimulating agent comprises 1, 2, 3, or more immune response stimulating agents.
In some embodiments, in addition to administering a bispecific fusion protein described herein, the method or treatment further comprises administering at least one additional therapeutic agent. An additional therapeutic agent can be administered prior to, concurrently with, and/or subsequently to, administration of the bispecific fusion protein. Pharmaceutical compositions comprising a bispecific fusion protein and the additional therapeutic agent(s) are also provided. In some embodiments, the at least one additional therapeutic agent comprises 1, 2, 3, or more additional therapeutic agents.
Combination therapy with two or more therapeutic agents often uses agents that work by different mechanisms of action, although this is not required. Combination therapy using agents with different mechanisms of action may result in additive or synergetic effects. Combination therapy may allow for a lower dose of each agent than is used in monotherapy, thereby reducing toxic side effects and/or increasing the therapeutic index of the bispecific fusion protein. Combination therapy may decrease the likelihood that resistant cancer cells will develop. In some embodiments, combination therapy comprises a therapeutic agent that affects the immune response ( e.g ., enhances or activates the response) and a therapeutic agent that affects (e.g., inhibits or kills) the tumor/cancer cells.
In some embodiments of the methods described herein, the combination of a bispecific fusion protein described herein and at least one additional therapeutic agent results in additive or synergistic results. In some embodiments, the combination therapy results in an increase in the therapeutic index of the bispecific fusion protein. In some embodiments, the combination therapy results in an increase in the therapeutic index of the additional therapeutic agent(s). In some embodiments, the combination therapy results in a decrease in the toxicity and/or side effects of the bispecific fusion protein. In some embodiments, the combination therapy results in a decrease in the toxicity and/or side effects of the additional therapeutic agent(s).
Useful classes of therapeutic agents include, for example, anti-tubulin agents, auristatins, DNA minor groove binders, DNA replication inhibitors, alkylating agents (e.g., platinum complexes such as cisplatin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and carboplatin), anthracyclines, antibiotics, anti-folates, anti-metabolites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated pyrimidines, ionophores, lexitropsins, nitrosoureas, platinols, purine antimetabolites, puromycins, radiation sensitizers, steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like. In some embodiments, the second therapeutic agent is an alkylating agent, an antimetabolite, an antimitotic, a topoisomerase inhibitor, or an angiogenesis inhibitor.
Therapeutic agents that may be administered in combination with the bispecific fusion protein described herein include chemotherapeutic agents. Thus, in some embodiments, the method or treatment involves the administration of a bispecific fusion protein of the present disclosure in combination with a chemotherapeutic agent or in combination with a cocktail of chemotherapeutic agents. Treatment with a bispecific fusion protein can occur prior to, concurrently with, or subsequent to administration of chemotherapies. Combined administration can include co-administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive administration in either order but generally within a time period such that all active agents can exert their biological activities simultaneously. Preparation and dosing schedules for such chemotherapeutic agents can be used according to manufacturers' instructions or as determined empirically by the skilled practitioner. Preparation and dosing schedules for such chemotherapy are also described in The Chemotherapy Source Book, 4.sup.th Edition, 2008, M. C. Perry, Editor, Lippincott, Williams & Wilkins, Philadelphia, Pa.
Chemotherapeutic agents useful in the present disclosure include, but are not limited to, alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamime; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, caminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L- norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6- mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6- azauridine, carmofur, cytosine arabinoside, dideoxyuridine, doxifluridine, enocitabine, floxuridine, 5-FU; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenishers such as folinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK; razoxane; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (Ara-C); taxoids, e.g. paclitaxel (TAXOL) and docetaxel (TAXOTERE); chlorambucil; gemcitabine; 6- thioguanine; mercaptopurine; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; ibandronate; CPT11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; esperamicins; capecitabine (XELODA); and pharmaceutically acceptable salts, acids or derivatives of any of the above. Chemotherapeutic agents also include anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (FARESTON); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above. In some embodiments, the additional therapeutic agent is cisplatin. In some embodiments, the additional therapeutic agent is carboplatin.
In some embodiments of the methods described herein, the chemotherapeutic agent is a topoisomerase inhibitor. Topoisomerase inhibitors are chemotherapy agents that interfere with the action of a topoisomerase enzyme ( e.g ., topoisomerase I or II). Topoisomerase inhibitors include, but are not limited to, doxorubicin HC1, daunombicin citrate, mitoxantrone HC1, actinomycin D, etoposide, topotecan HC1, teniposide (VM-26), and irinotecan, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these. In some embodiments, the additional therapeutic agent is irinotecan.
In some embodiments, the chemotherapeutic agent is an anti-metabolite. An anti metabolite is a chemical with a structure that is similar to a metabolite required for normal biochemical reactions, yet different enough to interfere with at least one normal functions of cells, such as cell division. Anti-metabolites include, but are not limited to, gemcitabine, fluorouracil, capecitabine, methotrexate sodium, ralitrexed, pemetrexed, tegafur, cytosine arabinoside, thioguanine, 5-azacytidine, 6-mercaptopurine, azathioprine, 6-thioguanine, pentostatin, fludarabine phosphate, and cladribine, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these. In some embodiments, the additional therapeutic agent is gemcitabine.
In some embodiments of the methods described herein, the chemotherapeutic agent is an antimitotic agent, including, but not limited to, agents that bind tubulin. In some embodiments, the agent is a taxane. In some embodiments, the agent is paclitaxel or docetaxel, or a pharmaceutically acceptable salt, acid, or derivative of paclitaxel or docetaxel. In some embodiments, the agent is paclitaxel (TAXOL), docetaxel (TAXOTERE), albumin-bound paclitaxel (nab-paclitaxel; ABRAXANE), DHA-paclitaxel, or PG-paclitaxel. In certain alternative embodiments, the antimitotic agent comprises a vinca alkaloid, such as vincristine, vinblastine, vinorelbine, or vindesine, or pharmaceutically acceptable salts, acids, or derivatives thereof. In some embodiments, the antimitotic agent is an inhibitor of kinesin Eg5 or an inhibitor of a mitotic kinase such as Aurora A or Plkl. In some embodiments, the additional therapeutic agent is paclitaxel. In some embodiments, the additional therapeutic agent is nab-paclitaxel. In some embodiments of the methods described herein, an additional therapeutic agent comprises an agent such as a small molecule. For example, treatment can involve the combined administration of a bispecific fusion protein of the present disclosure with a small molecule that acts as an inhibitor against tumor-associated antigens including, but not limited to, EGFR, HER2 (ErbB2), and/or VEGF. In some embodiments, a bispecific fusion protein of the present disclosure is administered in combination with a protein kinase inhibitor selected from the group consisting of: gefitinib (IRESSA), erlotinib (TARCEVA), sunitinib (SUTENT), lapatanib, vandetanib (ZACTIMA), AEE788, CI-1033, cediranib (RECENTIN), sorafenib (NEXAVAR), and pazopanib (GW786034B). In some embodiments, an additional therapeutic agent comprises an mTOR inhibitor.
In some embodiments of the methods described herein, the additional therapeutic agent is a small molecule that inhibits a cancer stem cell pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the Notch pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the Wnt pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the BMP pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the Hippo pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the mTOR/AKR pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the RSPO/LGR pathway.
In some embodiments of the methods described herein, an additional therapeutic agent comprises a biological molecule, such as an antibody. For example, treatment can involve the combined administration of a bispecific fusion protein of the present disclosure with antibodies against tumor-associated antigens including, but not limited to, antibodies that bind EGFR, HER2/ErbB2, and/or VEGF. In some embodiments, the additional therapeutic agent is an antibody specific for a cancer stem cell marker. In some embodiments, the additional therapeutic agent is an antibody that binds a component of the Notch pathway. In some embodiments, the additional therapeutic agent is an antibody that binds a component of the Wnt pathway. In some embodiments, the additional therapeutic agent is an antibody that inhibits a cancer stem cell pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the Notch pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the Wnt pathway. In some embodiments, the additional therapeutic agent is an inhibitor of the BMP pathway. In some embodiments, the additional therapeutic agent is an antibody that inhibits b- catenin signaling. In some embodiments, the additional therapeutic agent is an antibody that is an angiogenesis inhibitor ( e.g ., an anti- VEGF or VEGF receptor antibody). In some embodiments, the additional therapeutic agent is bevacizumab (AVASTIN), ramucirumab, trastuzumab (HERCEPTIN), pertuzumab (OMNITARG), panitumumab (VECTIBIX), nimotuzumab, zalutumumab, or cetuximab (ERBITETX).
In some embodiments of the methods described herein, the additional therapeutic agent is an antibody that modulates the immune response. In some embodiments, the additional therapeutic agent is an anti-PD- 1 antibody, an anti-LAG-3 antibody, an anti-CTLA-4 antibody, an anti-TIM-3 antibody, or an anti-TIGIT antibody.
Furthermore, treatment with a bispecific fusion protein described herein can include combination treatment with other biologic molecules, such as at least one cytokines ( e.g ., lymphokines, interleukins, tumor necrosis factors, and/or growth factors) or can be accompanied by surgical removal of tumors, removal of cancer cells, or any other therapy deemed necessary by a treating physician. In some embodiments, the additional therapeutic agent is an immune response stimulating agent.
In some embodiments of the methods described herein, the bispecific fusion protein can be combined with a growth factor selected from the group consisting of: adrenomedullin (AM), angiopoietin (Ang), BMPs, BDNF, EGF, erythropoietin (EPO), FGF, GDNF, G-CSF, GM-CSF, GDF9, HGF, HDGF, IGF, migration-stimulating factor, myostatin (GDF-8), NGF, neurotrophins, PDGF, thrombopoietin, TGF-a, TGF-b, TNF-a, VEGF, P1GF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-12, IL-15, and IL-18.
In some embodiments of the methods described herein, the additional therapeutic agent is an immune response stimulating agent. In some embodiments, the immune response stimulating agent is selected from the group consisting of granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), granulocyte colony stimulating factor (G-CSF), interleukin 3 (IF-3), interleukin 12 (IF-12), interleukin 1 (IF-1), interleukin 2 (IF-2), B7-1 (CD80), B7-2 (CD86), 4- IBB ligand, anti-CD3 antibody, anti-CTFA-4 antibody, anti-TIGIT antibody, anti-PD- 1 antibody, anti-FAG-3 antibody, and anti-TIM-3 antibody.
In some embodiments of the methods described herein, an immune response stimulating agent is selected from the group consisting of: a modulator of PD-1 activity, a modulator of PD- F2 activity, a modulator of CTFA-4 activity, a modulator of CD28 activity, a modulator of CD80 activity, a modulator of CD86 activity, a modulator of 4- IBB activity, an modulator of 0X40 activity, a modulator of KIR activity, a modulator of Tim-3 activity, a modulator of FAG3 activity, a modulator of CD27 activity, a modulator of CD40 activity, a modulator of GITR activity, a modulator of TIGIT activity, a modulator of CD20 activity, a modulator of CD96 activity, a modulator of IDOl activity, a cytokine, a chemokine, an interferon, an interleukin, a lymphokine, a member of the tumor necrosis factor (TNF) family, and an immuno stimulatory oligonucleotide. In some embodiments of the methods described herein, an immune response stimulating agent is selected from the group consisting of: a PD-1 antagonist, a PD-L2 antagonist, a CTLA-4 antagonist, a CD80 antagonist, a CD86 antagonist, a KIR antagonist, a Tim-3 antagonist, a LAG3 antagonist, a TIGIT antagonist, a CD20 antagonist, a CD96 antagonist, and/or an IDOl antagonist.
In some embodiments of the methods described herein, the PD- 1 antagonist is an antibody that specifically binds PD-1. In some embodiments, the antibody that binds PD-1 is KEYTRUDA (MK-3475), pidilizumab (CT-011), nivolumab (OPDIVO, BMS-936558, MDX- 1106), MEDI0680 (AMP-514), REGN2810, BGB-A317, PDR-001, or STI-A1110. In some embodiments, the antibody that binds PD-1 is described in PCT Publication WO 2014/179664, for example, an antibody identified as APE2058, APE1922, APE1923, APE1924, APE 1950, or APE 1963, or an antibody containing the CDR regions of any of these antibodies. In other embodiments, the PD-1 antagonist is a fusion protein that includes PD-L2, for example, AMP- 224. In other embodiments, the PD-1 antagonist is a peptide inhibitor, for example, AUNP-12.
In some embodiments, the CTLA-4 antagonist is an antibody that specifically binds CTLA-4. In some embodiments, the antibody that binds CTLA-4 is ipilimumab (YERVOY) or tremelimumab (CP-675,206). In some embodiments, the CTLA-4 antagonist a CTLA-4 fusion protein, for example, KAHR-102.
In some embodiments, the LAG3 antagonist is an antibody that specifically binds LAG3. In some embodiments, the antibody that binds LAG3 is IMP701, IMP731, BMS-986016, LAG525, and GSK2831781. In some embodiments, the LAG3 antagonist includes a soluble LAG3 receptor, for example, IMP321.
In some embodiments, the KIR antagonist is an antibody that specifically binds KIR. In some embodiments, the antibody that binds KIR is lirilumab.
In some embodiments, an immune response stimulating agent is selected from the group consisting of: a CD28 agonist, a 4- IBB agonist, an 0X40 agonist, a CD27 agonist, a CD80 agonist, a CD86 agonist, a CD40 agonist, and a GITR agonist p In some embodiments, the 0X40 agonist includes 0X40 ligand, or an OX40-binding portion thereof. For example, the 0X40 agonist may be MEDI6383. In some embodiments, the 0X40 agonist is an antibody that specifically binds 0X40. In some embodiments, the antibody that binds 0X40 is MED 16469, MEDI0562, or MOXR0916 (RG7888). In some embodiments, the 0X40 agonist is a vector ( e.g ., an expression vector or virus, such as an adenovirus) capable of expressing 0X40 ligand. In some embodiments the OX40-expressing vector is Delta-24-RGDOX or DNX2401.
In some embodiments, the 4-1BB (CD137) agonist is a binding molecule, such as an anticalin. In some embodiments, the anticalin is PRS-343. In some embodiments, the 4- IBB agonist is an antibody that specifically binds 4- IBB. In some embodiments, antibody that binds 4-1BB is PF-2566 (PF-05082566) or urelumab (B MS-663513).
In some embodiments, the CD27 agonist is an antibody that specifically binds CD27. In some embodiments, the antibody that binds CD27 is varlilumab (CDX-1127).
In some embodiments, the GITR agonist comprises GITR ligand or a GITR-binding portion thereof. In some embodiments, the GITR agonist is an antibody that specifically binds GITR. In some embodiments, the antibody that binds GITR is TRX518, MK-4166, or INBRX- 110.
In some embodiments, immune response stimulating agents include, but are not limited to, cytokines such as chemokines, interferons, interleukins, lymphokines, and members of the tumor necrosis factor (TNF) family. In some embodiments, immune response stimulating agents include immunostimulatory oligonucleotides, such as CpG dinucleotides.
In some embodiments, an immune response stimulating agent includes, but is not limited to, anti-PD-1 antibodies, anti-PD-L2 antibodies, anti-CTLA-4 antibodies, anti-CD28 antibodies, anti-CD80 antibodies, anti-CD86 antibodies, anti-4- IBB antibodies, anti-OX40 antibodies, anti- KIR antibodies, anti-Tim-3 antibodies, anti-LAG3 antibodies, anti-CD27 antibodies, anti-CD40 antibodies, anti-GITR antibodies, anti-TIGIT antibodies, anti-CD20 antibodies, anti-CD96 antibodies, or anti-IDOl antibodies.
In some embodiments, the bispecific fusion proteins disclosed herein may be used alone, or in association with radiation therapy.
In some embodiments, the bispecific fusion proteins disclosed herein may be used alone, or in association with targeted therapies. Examples of targeted therapies include: hormone therapies, signal transduction inhibitors ( e.g ., EGFR inhibitors, such as cetuximab (Erbitux) and erlotinib (Tarceva)); HER2 inhibitors (e.g., trastuzumab (Herceptin) and pertuzumab (Perjeta)); BCR-ABL inhibitors (such as imatinib (Gleevec) and dasatinib (Sprycel)); ALK inhibitors (such as crizotinib (Xalkori) and ceritinib (Zykadia)); BRAF inhibitors (such as vemurafenib (Zelboraf) and dabrafenib (Tafinlar)), gene expression modulators, apoptosis inducers (e.g., bortezomib (Velcade) and carfilzomib (Kyprolis)), angiogenesis inhibitors (e.g., bevacizumab (Avastin) and ramucirumab (Cyramza), monoclonal antibodies attached to toxins (e.g., brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla)).
In some embodiments, the bispecific fusion proteins of the disclosure may be used in combination with an anti-cancer therapeutic agent or immunomodulatory drug such as an immunomodulatory receptor inhibitor, e.g., an antibody or antigen-binding fragment thereof that specifically binds to the receptor. In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a Tim-3 pathway antagonist, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a Vista pathway antagonist, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a BTLA pathway antagonist, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a LAG-3 pathway antagonist, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with a TIGIT pathway antagonist, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-PDLl antibody
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with BMS-936559, MSB0010718C or MPDL3280A), for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CTLA4 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CSl antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti- KIR2DL 1/2/3 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CD137 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-GITR antibody, for example, as part of a pharmaceutical composition. In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-PD-L2 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILTl antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT2 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT3 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT4 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT5 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT6 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT7 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ILT8 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CD40 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-OX40 antibody, for example, as part of a pharmaceutical composition. In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DLl antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DL2/3 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DL4 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DL5A antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR2DL5B antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR3DLl antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR3DL2 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-KIR3DL3 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-NKG2A antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-NKG2C antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-ICOS antibody, for example, as part of a pharmaceutical composition. In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-SIRP. alpha antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CD47 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-4- 1 BB antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti- IL- 10 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-TSLP antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with IL-10 or PEGylated IL-10, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti- APRIL antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, an anti-PD-1 antibody or antigen-binding fragment thereof of the disclosure is administered in association with an anti-CD27 antibody, for example, as part of a pharmaceutical composition.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with a STING agonist, for example, as part of a pharmaceutical composition. The cyclic-di-nucleotides (CDNs) cyclic-di-AMP (produced by Listeria monocytogenes and other bacteria) and its analogs cyclic-di-GMP and cyclic-GMP-AMP are recognized by the host cell as a pathogen associated molecular pattern (PAMP), which bind to the pathogen recognition receptor (PRR) known as Stimulator of INterferon Genes (STING). STING is an adaptor protein in the cytoplasm of host mammalian cells which activates the TANK binding kinase (TBK1)-IRL3 and the NL-.kappa.B signaling axis, resulting in the induction of IRN-b and other gene products that strongly activate innate immunity. It is now recognized that STING is a component of the host cytosolic surveillance pathway, that senses infection with intracellular pathogens and in response induces the production of ILN-a, leading to the development of an adaptive protective pathogen-specific immune response consisting of both antigen- specific CD4+ and CD8+ T cells as well as pathogen-specific antibodies. U.S. Pat. Nos. 7,709,458 and 7,592,326; PCT Publication Nos. W02007/054279, WO2014/093936, WO2014/179335, WO2014/189805, WO2015/185565, WO2016/096174, W02016/145102,
W 02017/027645, WO2017/027646, and WO2017/075477; and Yan et al., Bioorg. Med. Chem Lett. 18:5631-4, 2008.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with an Akt inhibitor. Examples of AKT inhibitors include GDC0068 (also known as GDC-0068, ipatasertib and RG7440), MK-2206, perifosine (also known as KRX-0401), GSK690693, AT7867, triciribine, CCT128930, A-674563, PHT-427, Akti-1/2, afuresertib (also known as GSK2110183), AT13148, GSK2141795, BAY1125976, uprosertib (aka GSK2141795), Akt Inhibitor VIII (l,3-dihydro-l-[l-[[4-(6-phenyl-lH- imidazo [4,5-g] quinoxalin-7-yl)phenyl] m- ethyl] -4-piperidinyl] -2H-benzimidazol-2-one) , Akt Inhibitor X (2-chloro-N,N-diethyl-10H-phenoxazine-10-butanamine, monohydrochloride), MK- 2206 (8-(4-(l-aminocyclobutyl)phenyl)-9-phenyl-[l,2,4]triazolo[3,4-f][- l,6]naphthyridin- 3(2H)-one), uprosertib (N-((S)-l-amino-3-(3,4-difluorophenyl)propan-2-yl)-5-chloro-4-(4- chloro-1- -methyl-lH-pyrazol-5-yl)furan-2-carboxamide), ipatasertib ((S)-2-(4-chlorophenyl)-l- (4-((5R,7R)-7-hydroxy-5-methyl-6,7-dihydro-5H-c- yclopenta[d]pyrimidin-4-yl)piperazin-l-yl)- 3-(isopropylamino)propan-l-one)- , AZD 5363 (4-Piperidinecarboxamide, 4-amino-N-[(lS)-l- (4-chlorophenyl)-3-hydroxypropyl]-l-(7H-pyrrolo[2,3-d]p- yrimidin-4-yl)), perifosine, GSK690693, GDC-0068, tricirbine, CCT128930, A-674563, PF-04691502, AT7867, miltefosine, PHT-427, honokiol, triciribine phosphate, and KP372-1A (10H-indeno[2,l- e]tetrazolo[l,5-b][l,2,4]triazin-10-one), Akt Inhibitor IX (CAS 98510-80-6). Additional Akt inhibitors include: ATP-competitive inhibitors, e.g. isoquinoline-5- sulfonamides (e.g., H-8, H- 89, NL-71-101), azepane derivatives (e.g., (-)-balanol derivatives), aminofurazans (e.g., GSK690693), heterocyclic rings (e.g., 7-azaindole, 6-phenylpurine derivatives, pyrrolo[2,3- d]pyrimidine derivatives, CCT128930, 3-aminopyrrolidine, anilinotriazole derivatives, spiroindoline derivatives, AZD5363, A-674563, A-443654), phenylpyrazole derivatives (e.g., AT7867, AT13148), thiophenecarboxamide derivatives (e.g., Afuresertib (GSK2110183), 2- pyrimidyl-5-amidothiophene derivative (DC120), uprosertib (GSK2141795); Allosteric inhibitors, e.g., 2,3-diphenylquinoxaline analogues (e.g., 2,3-diphenylquinoxaline derivatives, triazolo[3,4-f][l,6]naphthyridin-3(2H)-one derivative (MK-2206)), alkylphospholipids (e.g., Edelfosine (l-0-octadecyl-2-0-methyl-rac-glycero-3-phosphocholine, ET-18-OCH3) ilmofosine (BM 41.440), miltefosine (hexadecylphosphocholine, HePC), perifosine (D-21266), erucylphosphocholine (ErPC), erufosine (ErPC3, erucylphosphohomocholine), indole-3-carbinol analogues ( e.g ., indole-3 -carbinol, 3-chloroacetylindole, diindolylmethane, diethyl 6-methoxy- 5,7-dihydroindolo [2,3-b]carbazole-2,10-dicarboxylate (SR13668), OSU-A9), Sulfonamide derivatives (e.g., PH-316, PHT-427), thiourea derivatives (e.g., PIT-1, PIT-2, DM-PIT-1, N-[(l- methyl-lH-pyrazol-4-yl)carbonyl]-N'-(3-bromophenyl)-thiourea), purine derivatives (e.g., Triciribine (TCN, NSC 154020), triciribine mono-phosphate active analogue (TCN-P),4-amino- pyrido[2,3-d]pyrimidine derivative API-1, 3-phenyl-3H-imidazo[4,5-b]pyridine derivatives, ARQ 092), BAY 1125976, 3 -methyl-xanthine, quinoline-4-carboxamide, 2-[4-(cyclohexa-l,3- dien-l-yl)-lH-pyrazol-3-yl]phenol, 3-oxo-tirucallic acid, 3. alpha.- and 3 -acetoxy-timcallic acids, acetoxy-tirucallic acid; and irreversible inhibitors, e.g., natural products, antibiotics, Lactoquinomycin, Frenolicin B, kalafungin, medermycin, Boc-Phe-vinyl ketone, 4- hydroxynonenal (4-HNE), 1,6-naphthyridinone derivatives, and imidazo- 1,2-pyridine derivatives.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with a MEK inhibitor. Examples of MEK inhibitors include AZD6244 (Selumetinib), PD0325901, GSK1120212 (Trametinib), U0126-EtOH, PD184352, RDEA119 (Rafametinib), PD98059, BIX 02189, MEK162 (Binimetinib), AS-703026 (Pimasertib), SL-327, BIX02188, AZD8330, TAK-733, cobimetinib and PD318088.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with both an anthracycline such as doxorubicin and cyclophosphamide, including pegylated liposomal doxorubicin.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with both an anti-CD20 antibody and an anti-CD3 antibody, or a bispecific CD20/CD3 binder (including a CD20/CD3 BiTE).
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with a CD73 inhibitor, a CD39 inhibitor or both. These inhibitors can be CD73 binders or CD39 binders (such as antibody, antibody fragments or antibody mimetics) that inhibit the ectonucleosidase activity. The inhibitor may be a small molecule inhibitor of the ectonucleosidase activity, such as 6-N,N-Diethyl-P-y-dibromomethylene-D- adenosine-5 '-triphosphate trisodium salt hydrate, PSB069, PSB 06126,
Figure imgf000139_0001
Figure imgf000140_0001
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with an inhibitor poly ADP ribose polymerase (PARP). Examples of PARP inhibitors include Olaparib, Niraparib, Rucaparib, Talazoparib, Veliparib, CEP9722, MK4827 and BGB-290.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with an oncolytic vims. An example of an oncolytic virus is Talimogene Laherparepvec.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with an CSF-1 antagonist, such as an agent that binds to CSF-1 or CSF1R and inhibits the interaction of CSF-1 with CSF1R on macrophage. An example of a CSF- 1 antagonist includes Emactuzumab and FPA008.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with an anti-CD38 antibody. An example of an anti-CD39 antibodies include Daratumumab and Isatuximab.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with an anti-CD40 antibody. An example of an anti-CD40 antibodies include Selicrelumab and Dacetuzumab.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with an inhibitor of anaplatic lymphoma kinase (AFK). An example of AFK inhibitors include Alectinib, Crizotinib and Ceritinib.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with multikinase inhibitor that inhibits at least one selected from the group consisting of the family members of VEGFR, PDGFR and FGFR, or an anti-angiogenesis inhibitor. Examples of inhibitors include Axitinib, Cediranib, Finifanib, Motesanib, Nintedanib, Pazopanib, Ponatinib, Regorafenib, Sorafenib, Sunitinib, Tivozanib, Vatalanib, LY2874455, or SU5402.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in conjunction with at least one vaccine intended to stimulate an immune response to at least one predetermined antigens. The antigen(s) may be administered directly to the individual, or may be expressed within the individual from, for example, a tumor cell vaccine ( e.g ., GVAX) which may be autologous or allogenic, a dendritic cell vaccine, a DNA vaccine, an RNA vaccine, a viral-based vaccine, a bacterial or yeast vaccine (e.g., a Listeria monocytogenes or Saccharomyces cerevisiae), etc. See, e.g., Guo et al., Adv. Cancer Res. 2013; 119: 421-475; Obeid et al., Semin Oncol. 2015 August; 42(4): 549-561. The target antigen may also be a fragment or fusion polypeptide comprising an immunologically active portion of the antigens listed in the table.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with at least one antiemetics including, but not limited to: casopitant (GlaxoSmithKline), Netupitant (MGI-Helsinn) and other NK-1 receptor antagonists, palonosetron (sold as Aloxi by MGI Pharma), aprepitant (sold as Emend by Merck and Co.; Rahway, N.J.), diphenhydramine (sold as Benadryl by Pfizer; New York, N.Y.), hydroxyzine (sold as Atarax by Pfizer; New York, N.Y.), metoclopramide (sold as Reglan by AH Robins Co,; Richmond, Va.), lorazepam (sold as Ativan by Wyeth; Madison, N.J.), alprazolam (sold as Xanax by Pfizer; New York, N.Y.), haloperidol (sold as Haldol by Ortho-McNeil; Raritan, N.J.), droperidol (Inapsine), dronabinol (sold as Marinol by Solvay Pharmaceuticals, Inc.; Marietta, Ga.), dexamethasone (sold as Decadron by Merck and Co.; Rahway, N.J.), methylprednisolone (sold as Medrol by Pfizer; New York, N.Y.), prochlorperazine (sold as Compazine by Glaxosmithkline; Research Triangle Park, N.C.), granisetron (sold as Kytril by Hoffmann-La Roche Inc.; Nutley, N.J.), ondansetron (sold as Zofran by Glaxosmithkline; Research Triangle Park, N.C.), dolasetron (sold as Anzemet by Sanofi-Aventis; New York, N.Y.), tropisetron (sold as Navoban by Novartis; East Hanover, N.J.).
Other side effects of cancer treatment include red and white blood cell deficiency. Accordingly, in some embodiments of the disclosure, a bispecific fusion protein is administered in association with an agent which treats or prevents such a deficiency, such as, e.g., filgrastim, PEG-filgrastim, erythropoietin, epoetin alfa or darbepoetin alfa.
In some embodiments of the disclosure, a bispecific fusion protein of the disclosure is administered in association with anti-cancer radiation therapy. For example, in some embodiments of the disclosure, the radiation therapy is external beam therapy (EBT): a method for delivering a beam of high-energy X-rays to the location of the tumor. The beam is generated outside the patient ( e.g ., by a linear accelerator) and is targeted at the tumor site. These X-rays can destroy the cancer cells and careful treatment planning allows the surrounding normal tissues to be spared. No radioactive sources are placed inside the patient's body. In some embodiments of the disclosure, the radiation therapy is proton beam therapy: a type of conformal therapy that bombards the diseased tissue with protons instead of X-rays. In some embodiments of the disclosure, the radiation therapy is conformal external beam radiation therapy: a procedure that uses advanced technology to tailor the radiation therapy to an individual's body structures. In some embodiments of the disclosure, the radiation therapy is brachytherapy: the temporary placement of radioactive materials within the body, usually employed to give an extra dose— or boost— of radiation to an area.
In some embodiments of the methods described herein, the treatment involves the administration of a bispecific fusion protein of the present disclosure in combination with anti viral therapy. Treatment with a bispecific fusion protein can occur prior to, concurrently with, or subsequent to administration of antiviral therapy. The anti- viral drug used in combination therapy will depend upon the virus the subject is infected with.
Combined administration can include co-administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive administration in either order but generally within a time period such that all active agents can exert their biological activities simultaneously.
It will be appreciated that the combination of a bispecific fusion protein described herein and at least one additional therapeutic agent may be administered in any order or concurrently. In some embodiments, the bispecific fusion protein will be administered to patients that have previously undergone treatment with a second therapeutic agent. In certain other embodiments, the bispecific fusion protein and a second therapeutic agent will be administered substantially simultaneously or concurrently. For example, a subject may be given a bispecific fusion protein while undergoing a course of treatment with a second therapeutic agent (e.g., chemotherapy). In some embodiments, a bispecific fusion protein will be administered within 1 year of the treatment with a second therapeutic agent. In certain alternative embodiments, a bispecific fusion protein will be administered within 10, 8, 6, 4, or 2 months of any treatment with a second therapeutic agent. In certain other embodiments, a bispecific fusion protein will be administered within 4, 3, 2, or 1 weeks of any treatment with a second therapeutic agent. In some embodiments, a bispecific fusion protein will be administered within 5, 4, 3, 2, or 1 days of any treatment with a second therapeutic agent. It will further be appreciated that the two (or more) agents or treatments may be administered to the subject within a matter of hours or minutes (i.e., substantially simultaneously). For the treatment of a disease, the appropriate dosage of a bispecific fusion protein of the present disclosure depends on the type of disease to be treated, the severity and course of the disease, the responsiveness of the disease, whether the bispecific fusion protein is administered for therapeutic or preventative purposes, previous therapy, the patient's clinical history, and so on, all at the discretion of the treating physician. The bispecific fusion protein can be administered one time or over a series of treatments lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved ( e.g ., reduction in tumor size). Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient and will vary depending on the relative potency of an individual agent. The administering physician can determine optimum dosages, dosing methodologies, and repetition rates. In some embodiments, dosage is from 0.01 mg to 100 mg/kg of body weight, from 0.1 mg to 100 mg/kg of body weight, from 1 mg to 100 mg/kg of body weight, from 1 mg to 100 mg/kg of body weight, 1 mg to 80 mg/kg of body weight from 10 mg to 100 mg/kg of body weight, from 10 mg to 75 mg/kg of body weight, or from 10 mg to 50 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is from about 0.1 mg to about 20 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 0.1 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 0.25 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 0.5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 1 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 1.5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 2 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 2.5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 7.5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 10 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 12.5 mg/kg of body weight. In some embodiments, the dosage of the bispecific fusion protein is about 15 mg/kg of body weight. In some embodiments, the dosage can be given once or more daily, weekly, monthly, or yearly. In some embodiments, the bispecific fusion protein is given once every week, once every two weeks, once every three weeks, or once every four weeks.
In some embodiments, a bispecific fusion protein may be administered at an initial higher "loading" dose, followed by at least one lower doses. In some embodiments, the frequency of administration may also change. In some embodiments, a dosing regimen may comprise administering an initial dose, followed by additional doses (or "maintenance" doses) once a week, once every two weeks, once every three weeks, or once every month. For example, a dosing regimen may comprise administering an initial loading dose, followed by a weekly maintenance dose of, for example, one-half of the initial dose. Or a dosing regimen may comprise administering an initial loading dose, followed by maintenance doses of, for example one-half of the initial dose every other week. Or a dosing regimen may comprise administering three initial doses for 3 weeks, followed by maintenance doses of, for example, the same amount every other week.
As is known to those of skill in the art, administration of any therapeutic agent may lead to side effects and/or toxicities. In some cases, the side effects and/or toxicities are so severe as to preclude administration of the particular agent at a therapeutically effective dose. In some cases, drug therapy must be discontinued, and other agents may be tried. However, many agents in the same therapeutic class often display similar side effects and/or toxicities, meaning that the patient either has to stop therapy, or if possible, suffer from the unpleasant side effects associated with the therapeutic agent.
In some embodiments, the dosing schedule may be limited to a specific number of administrations or "cycles". In some embodiments, the bispecific fusion protein is administered for 3, 4, 5, 6, 7, 8, or more cycles. For example, the bispecific fusion protein is administered every 2 weeks for 6 cycles, the bispecific fusion protein is administered every 3 weeks for 6 cycles, the bispecific fusion protein is administered every 2 weeks for 4 cycles, the bispecific fusion protein is administered every 3 weeks for 4 cycles, etc. Dosing schedules can be decided upon and subsequently modified by those skilled in the art.
Thus, the present disclosure provides methods of administering to a subject the polypeptides or agents described herein comprising using an intermittent dosing strategy for administering at least one agents, which may reduce side effects and/or toxicities associated with administration of a bispecific fusion protein, chemotherapeutic agent, etc. In some embodiments, a method for treating cancer in a human subject comprises administering to the subject a therapeutically effective dose of a bispecific fusion protein in combination with a therapeutically effective dose of a chemotherapeutic agent, wherein one or both of the agents are administered according to an intermittent dosing strategy. In some embodiments, the intermittent dosing strategy comprises administering an initial dose of a bispecific fusion protein to the subject and administering subsequent doses of the bispecific fusion protein about once every 2 weeks. In some embodiments, the intermittent dosing strategy comprises administering an initial dose of a bispecific fusion protein to the subject and administering subsequent doses of the bispecific fusion protein about once every 3 weeks. In some embodiments, the intermittent dosing strategy comprises administering an initial dose of a bispecific fusion protein to the subject and administering subsequent doses of the bispecific fusion protein about once every 4 weeks. In some embodiments, the bispecific fusion protein is administered using an intermittent dosing strategy and the chemotherapeutic agent is administered weekly.
In some embodiments, the disclosure also provides methods for treating subjects using a bispecific fusion protein of the disclosure, wherein the subject suffers from a viral infection. In some embodiments, the viral infection is infection with a virus selected from the group consisting of human immunodeficiency virus (HIV), hepatitis virus (A, B, or C), herpes virus ( e.g ., VZV, HSV-I, HAV-6, HSV-II, and CMV, Epstein Barr virus), adenovirus, influenza virus, flaviviruses, echovirus, rhinovirus, coxsackie virus, coronavirus, respiratory syncytial virus, mumps virus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, dengue virus, papillomavirus, molluscum virus, poliovirus, rabies virus, JC virus or arboviral encephalitis virus.
In some embodiments, the disclosure provides methods for treating subjects using a bispecific fusion protein thereof of the disclosure, wherein the subject suffers from a bacterial infection. In some embodiments, the bacterial infection is infection with a bacterium selected from the group consisting of Chlamydia, rickettsial bacteria, mycobacteria, staphylococci, streptococci, pneumonococci, meningococci and gonococci, klebsiella, proteus, serratia, pseudomonas, Legionella, Corynebacterium diphtheriae, Salmonella, bacilli, Vibrio cholerae, Clostridium tetan, Clostridium botulinum, Bacillus anthricis, Yersinia pestis, Mycobacterium leprae, Mycobacterium lepromatosis, and Borriella.
In some embodiments, the disclosure provides methods for treating subjects using a bispecific fusion protein of the disclosure, wherein the subject suffers from a fungal infection. In some embodiments, the fungal infection is infection with a fungus selected from the group consisting of Candida (albicans, krusei, glabrata, tropicalis, etc.), Cryptococcus neoformans, Aspergillus (fumigatus, niger, etc.), Genus Mucorales (mucor, absidia, rhizopus), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum.
In some embodiments, the disclosure provides methods for treating subjects using a bispecific fusion protein of the disclosure, wherein the subject suffers from a parasitic infection. In some embodiments, the parasitic infection is infection with a parasite selected from the group consisting of Entamoeba histolytica, Balantidium coli, Naegleria fowleri, Acanthamoeba, Giardia lambia, Cryptosporidium, Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondii and Nippostrongylus brasiliensis. Gene Delivery
An alternative approach to the delivery of therapeutic bispecific fusion protein would be to leave the production of the therapeutic polypeptide to the body itself. A multitude of clinical studies have illustrated the utility of in vivo gene transfer into cells using a variety of different delivery systems. In vivo gene transfer seeks to administer to patients the bispecific fusion protein nucleotide sequence, rather than the bispecific fusion protein itself. This allows the patient’s body to produce the bispecific fusion protein of interest for a prolonged period of time, and secrete it either systemically or locally, depending on the production site. Gene-based nucleotides encoding bispecific fusion proteins can present a labor- and cost-effective alternative to the conventional production, purification and administration of the polypeptide version of the bispecific fusion protein. A number of antibody expression platforms have been pursued in vivo to which delivery of polynucleotides encoding bispecific fusion proteins can be adapted: these include viral vectors, naked DNA and RNA. The use of gene transfer with polynucleotides encoding bispecific fusion proteins cannot only enable cost-savings by reducing the cost of goods and of production but may also be able to reduce the frequency of drug administration. Overall, a prolonged in vivo production of the therapeutic bispecific fusion protein by expression of the polynucleotides encoding bispecific fusion proteins can contribute to (i) a broader therapeutic or prophylactic application of bispecific fusion proteins in price-sensitive conditions, (ii) an improved accessibility to therapy in both developed and developing countries, and (iii) more effective and affordable treatment modalities. In addition to in vivo gene transfer, cells can be harvested from the host (or a donor), engineered with polynucleotides encoding bispecific fusion proteins to produce bispecific fusion proteins and re-administered to patients.
The tumor presents a site for the transfer of polynucleotides encoding bispecific fusion proteins, targeted either via intravenous or direct injection/electroporation. Indeed, intratumoral expression of polynucletodies encoding bipecific proteins can allow for a local production of the therapeutic bispecific fusion proteins, waiving the need for high systemic bispecific fusion protein levels that might otherwise be required to penetrate and impact solid tumors. See, for example, Beckman et al. (2015) “Antibody constructs in cancer therapy: protein engineering strategies to improve exposure in solid tumors” Cancer 109(2): 170-9 and Dronca et al. (2015) “Immunomodulatory antibody therapy of cancer: the closer, the better” Clin Cancer Res. 21(5):944-6.
The success of gene therapy has largely been driven by improvements in nonviral and viral gene transfer vectors. An array of physical and chemical nonviral methods have been used to transfer DNA and mRNA to mammalian cells and a substantial number of these have been developed as clinical stage technologies for gene therapy, both ex vivo and in vivo , and are readily adapted for delivery of the polynucleotides encoding bispecific fusion proteins of the present disclosure. To illustrate, cationic liposome technology can be employed, which is based on the ability of amphipathic lipids, possessing a positively charged head group and a hydrophobic lipid tail, to bind to negatively charged DNA or RNA and form particles that generally enter cells by endocytosis. Some cationic liposomes also contain a neutral co-lipid, thought to enhance liposome uptake by mammalian cells. See, for example, Feigner et al. (1987) Lipofection: a highly efficient, lipid-mediated DNA-transfection procedure. MNAS 84:7413- 7417; San et al. (1983) “Safety and short-term toxicity of a novel cationic lipid formulation for human gene therapy” Hum. Gene Ther. 4:781-788; Xu et al. (1996) “Mechanism of DNA release from cationic liposome/DNA complexes used in cell transfection” Biochemistry 35,:5616-5623; and Legendre et al. (1992) “Delivery of plasmid DNA into mammalian cell lines using pH-sensitive liposomes: comparison with cationic liposomes” Pharm. Res. 9, 1235-1242.
Similarly, other polycations, such as poly-l-lysine and polyethylene-imine, can be used to deliver polynucleotides encoding bispecific fusion proteins. These polycations complex with nucleic acids via charge interaction and aid in the condensation of DNA or RNA into nanoparticles, which are then substrates for endosome-mediated uptake. Several of these cationic nucleic acid complex technologies have been developed as potential clinical products, including complexes with plasmid DNA, oligodeoxynucleotides, and various forms of synthetic RNA. Modified (and unmodified or “naked”) DNA and RNA have also been shown to mediate successful gene transfer in a number of circumstances and can also be used as systems for delivery of polynucleotides encoding bispecific fusion proteins. These include the use of plasmid DNA by direct intramuscular injection, the use of intratumoral injection of plasmid DNA. See, for example, Rodrigo et al. (2012) “De novo automated design of small RNA circuits for engineering synthetic riboregulation in living cells” PNAS 109:15271-15276; Oishi et al. (2005) “Smart polyion complex micelles for targeted intracellular delivery of PEGylated antisense oligonucleotides containing acid-labile linkages” Chembiochem. 6:718-725; Bhatt et al. (2015) “Microbeads mediated oral plasmid DNA delivery using polymethacrylate vectors: an effectual groundwork for colorectal cancer” Drug Deliv. 22:849-861; Ulmer et al. (1994) Protective immunity by intramuscular injection of low doses of influenza vims DNA vaccines” Vaccine 12: 1541-1544; and Heinzerling et al. (2005) “Intratumoral injection of DNA encoding human interleukin 12 into patients with metastatic melanoma: clinical efficacy” Hum. Gene Ther. 16:35-48.
Viral vectors are currently used as a delivery vehicle in the vast majority of pre-clinical and clinical gene therapy trials and in the first to be approved directed gene therapy. See Gene Therapy Clinical Trials Worldwide 2017 (abedia.com/wiley/). The main driver thereto is their exceptional gene delivery efficiency, which reflects a natural evolutionary development; viral vector systems are attractive for gene delivery, because viruses have evolved the ability to cross through cellular membranes by infection, thereby delivering nucleic acids such as polynucleotides encoding bispecific fusion proteins to target cells. Pioneered by adenoviral systems, the field of viral vector-mediated antibody gene transfer made significant strides in the past decades. The myriad of successfully evaluated administration routes, pre-clinical models and disease indications puts the capabilities of antibody gene transfer at full display through which the skilled artisan would readily be able to identify and adapt antibody gene transfer systems and techniques for in vivo delivery of polynucleotides constructs encoding bispecific fusion proteinsln the context of vectored intratumoral polynucleotides encoding bispecific fusion proteins gene transfer, oncolytic viruses have a distinct advantage, as they can specifically target tumor cells, boost bispecific fusion protein expression, and amplify therapeutic responses - such as to a PD-L1 pAFFIMER® bispecific fusion proteins.
In vivo gene transfer of polynucleotides encoding bispecific fusion proteins can also be accomplished by use of nonviral vectors, such as expression plasmids. Nonviral vectors are easily produced and do not seem to induce specific immune responses. Muscle tissue is most often used as target tissue for transfection, because muscle tissue is well vascularized and easily accessible, and myocytes are long-lived cells. Intramuscular injection of naked plasmid DNA results in transfection of a certain percentage of myocytes. Using this approach, plasmid DNA encoding cytokines and cytokine/IgGl chimeric proteins has been introduced in vivo and has positively influenced (autoimmune) disease outcome.
In some instances, in order to increase transfection efficiency via so-called intravascular delivery in which increased gene delivery and expression levels are achieved by inducing a short-lived transient high pressure in the veins. Special blood-pressure cuffs that may facilitate localized uptake by temporarily increasing vascular pressure and can be adapted for use in human patients for this type of gene delivery. See, for example, Zhang et al. (2001) “Efficient expression of naked DNA delivered intraarterially to limb muscles of nonhuman primates” Hum. Gene Ther., 12:427-438
Increased efficiency can also be gained through other techniques, such as in which delivery of the nucleic acid is improved by use of chemical carriers — cationic polymers or lipids — or via a physical approach — gene gun delivery or electroporation. See Tranchant et al. (2004) “Physicochemical optimisation of plasmid delivery by cationic lipids” J. Gene Med., 6 (Suppl. 1): S24-S35; and Niidome et al. (2002) “Gene therapy progress and prospects: nonviral vectors” Gene Ther., 9:1647-1652. Electroporation is especially regarded as an interesting technique for nonviral gene delivery. Somiari, et al. (2000) “Theory and in vivo application of electroporative gene delivery” Mol. Ther. 2:178-187; and Jaroszeski et al. (1999) “In vivo gene delivery by electroporation” Adv. Drug Delivery Rev., 35:131-137. With electroporation, pulsed electrical currents are applied to a local tissue area to enhance cell permeability, resulting in gene transfer across the membrane. Research has shown that in vivo gene delivery can be at least 10- 100 times more efficient with electroporation than without. See, for example, Aihara et al. (1998) “Gene transfer into muscle by electroporation in vivo” Nat. Biotechnol. 16:867-870; Mir, et al. (1999) “High-efficiency gene transfer into skeletal muscle mediated by electric pulses” PNAS 96:4262-4267; Rizzuto, et al. (1999) “Efficient and regulated erythropoietin production by naked DNA injection and muscle electroporation” PNAS 96: 6417-6422; and Mathiesen (1999) “Electropermeabilization of skeletal muscle enhances gene transfer in vivo” Gene Ther., 6:508- 514.
Encoded bispecific fusion proteins can be delivered by a wide range of gene delivery system commonly used for gene therapy including viral, non-viral, or physical. See, for example, Rosenberg et al., Science, 242:1575-1578, 1988, and Wolff et al., Proc. Natl. Acad. Sci. USA 86:9011-9014 (1989). Discussion of methods and compositions for use in gene therapy include Eck et al., in Goodman & Gilman's The Pharmacological Basis of Therapeutics, Ninth Edition, Hardman et al., eds., McGraw-Hill, New York, (1996), Chapter 5, pp. 77-101; Wilson, Clin.
Exp. Immunol. 107 (Suppl. 1):31-32, 1997; Wivel et al., Hematology/Oncology Clinics of North America, Gene Therapy, S. L. Eck, ed., 12(3):483-501, 1998; Romano et al., Stem Cells, 18:19- 39, 2000, and the references cited therein. U.S. Pat. No. 6,080,728 also provides a discussion of a wide variety of gene delivery methods and compositions. The routes of delivery include, for example, systemic administration and administration in situ.
An effective gene transfer approach should be directed to the specific tissues/cells where it is needed, and the resulting transgene expression should be at a level that is appropriate to the specific application. Promoters are a major cis-acting element within the vector genome design that can dictate the overall strength of expression as well as cell- specificity.
In some embodiments, a viral vector is used to deliver a nucleic acid encoding a bispecific fusion protein of the present disclosure. Non-limiting examples of viral vectors include adenoviral vectors, adeno-associated viral (AAV) vectors, and retroviral vectors. In other embodiments, a non-viral vector is used to deliver a nucleic acid encoding a bispecific fusion protein of the present disclosure. Non-limiting examples of non-viral vectors include plasmid vectors (e.g., plasmid DNA (pDNA) delivered via, e.g., hydrodynamic -based transfection or electroporation), minicircle DNA, and RNA-mediate gene transfer (e.g., delivery of messenger RNA (mRNA) encoding a bispecific fusion protein of the present disclosure). Exemplary nucleic acids or polynucleotides for the encoded bispecific fusion proteins of the present disclosure include, but are not limited to, ribonucleic acids (RNAs), deoxyribonucleic acids (DNAs), threose nucleic acids (TNAs), glycol nucleic acids (GNAs), peptide nucleic acids (PNAs), locked nucleic acids (LNAs, including LNA having a b- D-ribo configuration, a-LNA having an a-L-ribo configuration (a diastereomer of LNA), 2'-amino-LNA having a 2 '-amino functionalization, and 2'-amino- a-LNA having a 2'-amino functionalization), ethylene nucleic acids (ENA), cyclohexenyl nucleic acids (CeNA) or hybrids or combinations thereof. mRNA presents an emerging platform for antibody gene transfer that can be adapted by those skilled in the art for delivery of polynucleotide constructs encoding bispecific fusion proteins of the present disclosure. Although current results differ considerably, in certain instances the mRNA constructs appear to be able to rival viral vectors in terms of generated serum mAb titers. Levels were in therapeutically relevant ranges within hours after mRNA administration, a marked shift in speed compared to DNA. The use of lipid nanoparticles (LNP) for mRNA transfection, rather than the physical methods typically required for DNA, can provide significant advantages in some embodiments towards application range.
Nucleic acids encoding bispecific fusion proteins may be delivered g, for example, intravenously, intramuscularly, or intratumorally (e.g., by injection, electroporation or other means).
Nucleic acids encoding bispecific fusion proteins may be formulated, for example, in lipid nanoparticles or liposomes (e.g., cationic lipid nanoparticles or liposomes), biodegradable microsphere, or other nano- or microparticle. Other lipid-based (e.g., PEG lipid) and polymeric- based formulations and delivery vehicles are contemplated herein.
Additional Embodiments
The present disclosure also encompasses the embodiments described in the numbered paragraphs below.
1. A protein comprising a (i) PD-L1 binding ALLIMER® polypeptide sequence which binds to PD-L1 with a Kd of 1X10-6M or less and inhibits interaction of the PD-L1 to which it is bound with PD-1; and (ii) a TΰEb trap polypeptide sequence that can bind to TΰEb with a Kd of 1X10-6M or less and inhibits interaction of the TΰEb to which it is bound with TΰEbEII.
2. The protein of paragraph 1, including an amino acid sequence
IPRGLSEAKPATPEIQEIVDKVKPQLEEKTGETYGKLEAVQYKTQVLAFALPEFEYMSTNYYIK VRAGDNKYMHLKVFNGPPMIRRKNEVADRVLTGYQVDKNKDDELTGFAAAGGGGSGGGGSGGGG SGGGGSEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMI SRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAP IEKTIS KAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSD GSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGaGGGGSGGGGSGGGGSGGG GSGIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCS ITSICEKPQEVC VAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNII FSEEYNTSNPD (SEQ ID NO: 96) or and amino acid sequence
IPRGLSEAKPATPEIQEIVDKVKPQLEEKTGETYGKLEAVQYKTQVLAREGRQDWVLSTNYYIK VRAGDNKYMHLKVFNGPWVPFPHQQLADRVLTGYQVDKNKDDELTGFAAAGGGGSGGGGSGGGG SGGGGSEPKSCDKTHTCPPCPAPEAAGGPSVFLFPPKPKDTLMI SRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAP IEKTIS KAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSD GSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGaGGGGSGGGGSGGGGSGGG GSGIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCS ITSICEKPQEVC VAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNII FSEEYNTSNPD (SEQ ID NO: 97)
3. A pharmaceutical preparation compriding a protein of any one of the preceding paragraphs and a at least one pharmaceutically acceptable exicipents.
4. A polynucleotide comprising a coding sequence encoding the protein of any one of the preceding paragraphs.
EXAMPLES
Example 1. AFFIMER® Fc fusion Production and Protein Characterization
Anti-PD-Ll AFFIMER® proteins fused to human IgGl Fc with a C-terminal TGFB trap were designed and DNA was ordered from Atum with a human CD33 leader sequence present. A schematic of the AFFIMER® bispecific format is presented in FIG. 5A. AFFIMER®-Fc fusion transfections of suspension HEK cell culture (Expi293F cell line; Thermo Fisher) were performed with a mammalian CMV promoter expression vector pD609 (Atum) using ExpiFectamine reagent (Thermo Fisher) following the manufacturer’s protocol. Supernatant was harvested 7 days post-transfection by centrifuging at 15,000 xg for ljiour followed by 0. 22pm filtration. A first stage purification was performed on the protein batches using MabSelect PrismA protein A chromatography resin using an AKTA system (Cytiva). Resin was prepared by washing with five column volumes (CV) water, followed by equilibration with 5 CV PBS lx running buffer. Mammalian supernatant was run through the resin at a flow rate of 13.3 ml/min followed by a wash with 5 CV PBS lx. Bound AFFIMER®-Fc fusion protein was eluted in 5 CV 0.1 M acetic acid pH 3.5 and buffer exchanged into PBS lx using Centripure desalting columns (empBiotech GmbH). A second stage polishing purification was performed using size exclusion using a Superdex 26/600 column run in PBS lx (Cytiva). Analytical size exclusion chromatography (SEC) was carried out using a Yarra SEC-3000 column (Phenomenex) run on an Ultimate 3000 HPLC (high performance liquid chromatography) (Thermo Fisher) at 1.0 ml/min flow rate in PBS lx (FIG. 15B). SDS-PAGE analysis showed protein bands of the expected molecular weight (MW) (FIG. 15C).
Example 2. PD-L1 Binding Affimer Characterization
Binding affinities of purified AVA27-01, AVA27-02 and AVA27-03 (SEQ ID NO: 96, 97, and 98/99, respectively) to PD-Fl-Fc recombinant antigen were measured using surface plasmon resonance (SPR) (FIG. 16, Table 12). Biacore T200 kinetic analysis was performed using running buffer HBS-EP+ (Cytiva) and series S sensor CM5 chip (Cytiva) immobilized with human PD-F1 Fc (R&D Systems) or mouse PD-F1 Fc (R&D Systems) in 10 mM Sodium acetate pH4.0 (Cytiva) using amine coupling reagents (Cytiva). A concentration titration of Affimer Fc fusions was run as analyte at a flow rate of 30pl/min, with an association time of 700 seconds, followed by a dissociation time of 5,000 seconds. PD-F1 Fc antigen immobilized CM5 surface was regenerated with 4.5 mM NaOH (Cytiva) for 45 seconds at 20 pl/min flow rate followed by 10 mM glycine-HCl pH 3 (Cytiva) for 30 seconds at 20 ul/min. Kinetic data was blank subtracted and fit to a 1:1 Fangmuir binding model (BIAcore Evalution software; Cytiva) to calculate KD values. Anti-PD-Fl AFFIMER® proteins were shown to bind with pM affinites to PD-F1 Fc antigen (Table 12).
Table 12. Binding kinetics to human or mouse PD-F1 Fc of AVA27-01, AVA27-02 and AVA27-03 bispecific AFFIMER® Fc fusions measured using Biacore SPR
Figure imgf000152_0001
Example 3. SPR Binding Affinities of AFFIMER® Fc Fusions to TGFp Antigen
Binding affinities of purified AVA27-01, AVA27-02 and AVA27-03 (SEQ ID NO: 96, 97, and 98/99, respectively) to TGFp 1 antigen were measured using surface plasmon resonance (SPR) (FIG. 17, Table 13). Biacore 8K kinetic analysis was performed using running buffer HBS-EP+ and series S sensor CM5 chip (Cytiva) immobilized with human TGFpi in 10 mM sodium acetate pH 5.5 using amine coupling reagents (Cytiva). A concentration titration of AFFIMER® proteins was run as analyte at a flow rate of 30pl/min, with an association time of 200 seconds, followed by a dissociation time of 3,000 seconds. TGFP immobilized surfaces were regenerated with 4.5 mM NaOH (Cytiva) for 45 seconds at 20 pl/min flow rate followed by 10 mM glycine-HCl pH 3 (Cytiva) for 30 seconds at 20 ul/min. Kinetic data was blank subtracted and fit to a 1:1 Langmuir binding model (BIAcore Insights Evalution software; Cytiva) to calculate KD values. TGFP trap fused to the C-terminus of an AFFIMER® Fc protein (SEQ ID NO: 96 and 97) were shown to bind with similar affinity to the antibody trap fusion AVA27-03 (SEQ ID NO: 98/99) with the range of 15-28pM (Table 13).
Table 13. Binding kinetics to human TGFpl of AVA27-01, AVA27-02, and AVA27-03 bispecific AFFIMER® Fc fusions measured using Biacore SPR
Figure imgf000153_0001
Example 4. AVA27-02 Binding to Both PD-L1 and TGFp in Direct ELISA
The binding of AVA27-02 and -03 (SEQ ID NO: 97 and 98/99) to PD-L1 was assessed by direct ELISA.
Briefly, 96-well high binding microplates were coated with 0.5 pg/ml recombinant human PD-L1/B7-H1/CD274 Fc chimera protein (R&D Systems) in carbonate/bicarbonate buffer. After coating, and following each subsequent step, plates were washed three times with PBS containing 0.05% Tween 20. Unbound sites on the plates were blocked with 5% casein in PBS. AFFIMER® polypeptide dilutions (20 nM titrated 1:5) were prepared in duplicate and added to the plates. Binding was detected using a biotinylated human cystatin A antibody (R&D Systems BAF1407) and streptavidin poly-HRP (Thermo Scientific™ N200). All dilutions were prepared in PBS containing 1% casein and 0.01% Tween 20. TMB (Pierce) was added followed by 2M sulfuric acid to stop the reaction. Absorbance was read at 450 and 630nm and the average of the duplicate wells was used to calculate EC50 using four parameter non-linear regression. As shown in FIG. 18 shows the AVA27-02 (SEQ ID NO: 97), AFFIMER®-TGFp trap molecule maintains a similar binding capacity to PD-L1 compared to the parental molecule formatted on an Fc. The calculated EC50 values for AVA27-02 and the parental anti-PD-Ll AFFIMER® polypeptide are 0.09 nM and 0.05 nM, respectively.
The binding of AVA27-01 (SEQ ID NO: 96) to mouse PD-L1 was also evaluated by direct ELISA (FIG. 19). The method was similar to the method described above but using recombinant mouse PD-L1/B7-H1/CD274 Fc chimera protein (R&D Systems) coated at 0.5 pg/ml instead. The calculated EC50 value for AVA27-01 is 0.042 nM when detected with and anti-cystatin antibody.
The binding of AVA27-02 and AVA27-03 (SEQ ID NO: 97 and 98/99) to TGBp was also evaluated by direct ELISA.
Briefly, 96 well high binding microplates were coated with 1 pg/ml recombinant human TGFP-l (R&D Systems) in carbonate/bicarbonate buffer. After coating, and following each subsequent step, plates were washed three times with PBS containing 0.05% Tween 20.
Unbound sites on the plates were blocked with 5% casein in PBS. AFFIMER® polypeptide dilutions (100 nM titrated 1:4) were prepared in duplicate and added to the plates. Binding was detected using a biotinylated human cystatin A antibody (R&D Systems) or anti-Fc biotinylated antibody (Rockland) with streptavidin poly-HRP (Thermo Scientific). All dilutions were prepared in PBS containing 1% casein and 0.01% Tween 20. TMB substrate (Pierce) was added to develop the signal, followed by 2M sulfuric acid to stop the reaction. Absorbance was read at 450 and 630nm and the average of the duplicate wells used to calculate EC50 using four parameter non-linear regression. FIG. 20A shows the results generated by detection of the AFFIMER® scaffold with the anti-cystatin A antibody. Only AVA27-02 was detected, indicating that it is the molecule containing the AFFIMER® scaffold binds TGFp. FIG. 20B shows the comparable binding capacity of AVA27-02 and AVA27-03 to TGFP and no binding of both parental molecules. The calculated EC50 value for AVA27-02 was 0.13 nM when the AFFIMER® scaffold was detected and 0.33 nM when detected with an anti-Fc antibody. AVA27-03 (SEQ ID NO: 98/99) showed a comparable binding to TGFP with an EC50 value of 0.42 nM. FIG. 21A shows that only AVA27-01 (SEQ ID NO: 96) was detected, indicating that the molecule containing the AFFIMER® scaffold binds TGFp. FIG. 21B shows the similar binding capacity of AVA27-01 to Tΰb when detected with anti-Fc. No binding of either parental molecule was detected with either of the detection antibodies. The calculated EC 50 value for
AVA27-01 was 0.3 nM when detected through the Affimer scaffold and 0.45 nM when detected with an anti-Fc antibody.
Example 5. AVA27-01 and AVA27-02 Binding to Both Targets Simultaneously
Purified AVA27-01, AVA27-02, and AVA27-03 (SEQ ID NO: 96, 97, and 98/99, respectively) were assessed for their capacity to engage both targets simultaneously (FIGs. 22A and 22B).
High binding microplates were coated with 1 pg/ml of recombinant human TGF-b (R&D System) in carbonate bicarbonate buffer for 16 + 2 hours. After the coating, and following each subsequent step, plates were washed three times with 150 pi PBS containing 0.05% Tween 20. Unbound sites on the plates were blocked with 100 pi 5% casein in PBS. AFFIMER® polypeptides and antibodies were diluted in dilution buffer (PBST (0.01%) + 1% Casein (Sigma)) to 30nM and titrated 1:3 in a dilution plate before 50 pi were added to the plate and incubated for 90 + 15 minutes at 21 + 1°C. Recombinant human PD-L1/B7-H1 Fc chimera protein (R & D Systems) or recombinant mouse PD-L1/B7-H1 Fc chimera protein (R & D Systems) were then diluted to 10 nM before 50 pi of solution was added to the respective wells. The plate was then incubated for 90 + 15 minutes at 21 + 1°C before being washed as previously described. Two detection solutions were prepared, containing 200ng/ml streptavidin poly-HRP (ThermoFisher) and either 125 ng/ml of biotinylated anti-human PD-L1/B7-H1 antibody (R & D Systems) or biotinylated anti-mouse PD-L1/B7-H1 antibody (R&D Systems), 50 pi of solution was added to the plate to detect human or mouse PD-L1. The plate was then incubated for 60 + 5 minutes at 21 + 1°C, before being washed as previously described. 50 pi of room temperature TMB subtrate was added to all wells, followed by 50pl 2M sulfuric acid solution to stop the reaction. Absorbance was read at 450 and 630nm and the average of the duplicate wells was used to calculate the EC50 using four parameter non-linear regression. The calculated EC50 for AVA27-01 (SEQ ID NO: 96) when engaged to both targets and detected via mPD-Ll bridging was 0.22 nM. The calculated EC50 values for AVA27-02 and AVA27-03 (SEQ ID NO: 97 and 98/99, respectively) were 0.19 nM and 0.12 nM, respectively indicating similar bridging capacity.
Example 6: AVA27-02 Competes with PD-1 and Blocks the PD-1/PD-L1 axis
AVA27-02 and AVA27-03 (SEQ ID NO: 97 and 98/99, respectively) were evaluated for their ability to disrupt the PD-1:PD-L1 interaction with a competitive ELISA (FIG. 23). Briefly, microplates were coated with 0.5pg/ml reecombinant human PD-l-Fc chimera protein (R&D Systems) in carbonate/bicarbonate buffer. After the coating, and following each subsequent step, plates were washed three times with PBS containing 0.05% Tween 20. Unbound sites on the plates were blocked with 5% casein in PBS. AFFIMER® polypeptide and an antibody control were diluted in duplicate, and preincubated with 6 nM recombinant human PD-L1 Fc chimera protein (R&D Systems) for 60 minutes prior to addition to the ELISA plate. Binding of PD-L1 to PD-1 was measured using a bBiotinylated human PD-L1 antibody (R&D Systems) and streptavidin poly-HRP (Thermo Scientific). All dilutions were prepared in PBS containing 1% casein and 0.01% Tween 20. TMB (Pierce) was added, followed by 2M sulfuric acid to stop the reaction. Absorbance was read at 450 and 630nm and the data normalized to percentage of inhibition. The average of the duplicate wells was used to fit a nonlinear regression curve using a sigmoidal dose-response (variable slope) in Graphpad Prism 8. IC50 values were calculated. FIG. 23 shows the similar inhibition capacity of AVA27-02 to PD-L1 compared to the parental molecule. The calculated IC50 values for AVA27-02 and the parental anti-PD-Ll AFFIMER® Fc are 0.49 nM and 0.48 nM, respectively.
To evaluate the blockade of PD-1/PD-L1 pathway, a luciferase blockade bioluminescent reporter assay was performed (FIG. 24).
The assay was performed using two engineered cell lines: Jurkat T cells expressing human PD-1 and a luciferase reporter driven by an NFAT response element (NFAT-RE), and PD-L1 aAPC/CHO-Kl cells, which are CHO-K1 cells expressing human PD-L1 and an engineered cell surface protein designed to activate cognate T cell receptors (TCRs) in an antigen-independent manner. When the two cell types are co-cultured, the PD-1/PD-L1 interaction inhibits TCR signaling and NFAT-RE-mediated luminescence. AFFIMER® proteins and Avelumab were diluted to 2X final concentration (20uM and 0.04 mM, respectively) in assay buffer. The AFFIMER® proteins and controls were titrated down (1:2) in assay buffer to give 10-point curves in triplicate. The PD-L1 aAPC/CHO-Kl (target cells, Promega) and the PD-1 effector cells (Promega) were diluted 8 xlO5 cells/ml and lxlO6 cells/ml, respectively, and lOul of the cell suspension was added to each well of the assay plate (Coming 3570) and the plate was incubated for 24 hours in a 37°C, 5% CO2 incubator.
Following the 24 hour incubation, the plates were equilibrated to room temperature, and 20pl of Bio-Glo (Promega) was added to each well. The luminescence was then measured using a luminescence plate reader (BMG Labtech). The data was then analysed using Prism Graph Pad where the fold induction was plotted vs. log(concentration). A four-parameter fit was used to calculate IC50 values. FIG. 24 shows that AVA27-02 to PD-L1 has a similar blockade activity compared to the parental molecule. The calculated IC50 value for AVA27-02 and the parental anti-PD-Ll AFFIMER® Fc were 100.2 nM and 28.9 nM, respectively. Example 7. AVA27-02 Binds TGFp and Blocks TGFp i -SMAD Signalling
A recombinant cell line HEK293T cells containing a firefly luciferase gene under the control of SMAD responsive element stably integrated into HEK293 cells was used to demonstrate the ability of AVA27-01, AVA27-02, and AVA27-03 (SEQ ID NO: 96, 97, and 98/99, respectively) to neutralize TGFP (FIG. 25). Briefly, 20 pi of O.lxlO6 cells/ml of SBE reporter cell line (BPS Bioscience) diluted in Thaw Medium IB (BPS Bioscience) were seeded and incubated for 24 hours at 37°C + 5% CO2. Following incubation, the thaw medium IB was replaced by assay medium IB (BPS Bioscience, 79617) and the plate was incubated for a further 4 hours at 37°C + 5% CO2. Molecules of interest were diluted to 4 nM in assay medium IB and titrated 1:3 for 12 points before adding equal volumes of 300pM recombinant human TGF-b (R&D Systems). The final assay concentrations were InM of molecule of interest and 75pM recombinant human TGF-b. Following the cell incubation, the AFFIMER® proteins and controls were added to their corresponding wells in triplicate and the cells were incubated for a further 16 hours at 37°C + 5% CO2. The plates were then equilibrated to room temperature before 25ul of room temperature Bio-Glo (Promega) was added to each well. The plate was then incubated for 5 minutes in the dark before the luminescence was read using a luminometer (BMG FabTech) before luminescence was plotted using Prism 8 (GraphPad). The data was normalized against the average of 0% and 100% inhibition controls to calculate % of inhibition, and an IC50 was then determined for log(concentration) and % of inhibition using four-parameter non-linear regression. FIG. 25 shows that similar neutralization capacity to TΰRb was found for AVA27- 01, AVA27-02, and AVA27-03. The calculated IC50 values were 14.6 nM, 8.5 nM and, 7 nM respectively.
Example 8. AVA27-02 Binds on Cell in a Flow Cytometry Cell Binding Assay
To demonstrate the AVA27-02 molecule is able to engage target on cell a Flow cytometry assay was performed; various cell lines:
• a naturally PD-F1 expressing cells line H441 cells (Human Fung Tumour Cells) (FIG.
26 A); and
• a recombinant over-expressing cell line hPD-Fl CHO cells (Promega) and CHO cell line
(FIG. 26B).
Briefly, cells were washed in PBS EDTA 2mM and re-suspended in Cell Staining Buffer (PBS, 1% BSA, 2 mM EDTA and 0.05% sodium Azide)) at a density of 500000 cells/ml, then 100 ul of cells/well were distributed in a 96 well-round bottom plates. AFFIMER® proteins and controls were diluted in cell staining buffer and added for 60 mins at 2-8 °C . Cells were washed with 150 ul of PBS 2mM EDTA then centrifuged 3 min at 350g, the supernatant was discarded by flicking the plate, wash was repeated 2 more times. After washes, anti-Fc detection Ab A488 and Zombie Aqua Viability reagent (Biolegend) were added to the cells for 30 mins at 2-8 °C. After incubation, cells were wash as previously and fixed with 50 ul in the fixation buffer (Biolegend) for 15 mins. The cells were washed once as previously described and resuspended in 100 ul of cell staining buffer before acquiring the Fluorescence signal using the Guava easyCyte 12HT. The Mean Fluorescence Intensity (MFI) was plotted vs. log(concentration) using Prism Graph Pad, and a four-parameter fit was used to calculate EC50 values. The calculated EC50 value for AVA27-02 and the parental anti-PD-Ll AFFIMER® Fc bound to H441 were 5.83 nM and 1.8 nM, respectively, and for huPD-Ll CHO cells 21.73 nM and 1.6 nM.
Example 9. Pharmacokinetic (PK) study analysis in mice
C57BL6 mice >8 weeks old were randomly assigned to 2 groups (n=6/ group) and each group received one of the 2 proteins (AVA27-02 and control protein), the body weight was recorded before dosing and each animal received 5 mg/kg of tested proteins intravenously (IV) via the tail vein, each groups were subdivided in 2 groups of 3 animals be able to collect each timepoint. Blood samples were taken prior the study, at 15 min 6h, 24h,72h, 120h, 168h, 336h and 480h (full dataset for AVA27-02 (SEQ ID 118) from T168-T336h is not available at the time of filing)
At each timepoint 20-30 ul of blood were collected via the tail vein. Blood was clotted for 30 min at room temperature and spun 10 min at 14000 rpm. The supernatant was collected and spun for a second time 10 mins at 14000rpm. The supernatant was then transferred in pre labelled tubes, snap frozen and stored at -80°C.
To calculate PK parameters for AVA27-02 (SEQ ID NO: 97) each timepoint have been quantify by sandwich ELISA. Microplates were coated with anti-human Fc (Rockland) overnight at 4°C. After the coating, and following each subsequent step, plates were washed three times with 150 pi PBS containing 0.05% Tween 20. The unbound sites on the plates were then blocked with lOOul of saturation buffer (lx PBS + 5% casein (Sigma) and incubated for 90 minutes at 21°C. The protein standards were then prepared in dilution buffer (lx PBST (0.01%) + 1% casein) at lOnM and titrated 1 in 2 for 15 points in duplicate, serum samples were diluted and titrated 1 in 2 for 8 points in duplicate. Following plate saturation, 50 mΐ of standard or sample were added to each well, then plates incubated for a further 90 minutes at 21°C. 50ul of detection solution containing 200ng/ml biotinylated anti-Human Fc (Rockland) was added to each well and the plates were incubated for 90 minutes at 21°C. A secondary detection was added (200ng/ml Poly-HRP Streptavidin, 50m1) to each well and incubated for 30 minutes at 21°C.
After a final wash, 50 mΐ of TMB substrate was added and the reaction was stopped with 2M sulfuric acid. The plates were then read using a plate reader (BMG Labtech, Pherastar FSX) to measure absorbance at 450nm and 630nm. The absorbance at 630nm was then subtracted from the 450nm readout and the resulting OD was interpolated into the standard curve using Mars (BMG Labtech) to calculate concentrations for each timepoint. The FIG. 27 shows that AVA27- 02 (SEQ ID NO: 97) had a similar PK profile to the control molecules anti-PD-Ll AFFIMER® Fc, at 120h the quantity measured in serum was 118 nM for AVA27-02 (SEQ ID NO: 97). The estimated half-life for AVA27-02 and control was 4.75 and 4.4 days respectively. The ability of AVA27-02 (SEQ ID NO: 97) to still bind both target at 120h was measured using the bridging ELISA described in Example 5 using the concentration in nM calculated in serum. FIG. 28 shows the parallelism between the standard and the AVA27-02 (SEQ ID NO: 97) in serum showing the stability of this molecule in the animal as it still binds to both targets simultaneously.
All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document.
The indefinite articles “a” and “an,” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean “at least one.”
It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.
In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases “consisting of’ and “consisting essentially of’ shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.
The terms “about” and “substantially” preceding a numerical value mean ±10% of the recited numerical value.
Where a range of values is provided, each value between and including the upper and lower ends of the range are specifically contemplated and described herein.

Claims

What is claimed is: CLAIMS
1. A bispecific protein comprising: (a) a recombinantly engineered PD-Ll-binding variant of a stefin polypeptide that binds to PD-L1 with a Kd of 1X10-6M or less and inhibits interaction of the PD-L1 and PD-1; and (b) a TOEb trap polypeptide that binds to TOEb with a Kd of 1X10_6M or less and inhibits interaction of the TORb and TORb receptor, optionally TORbKII.
2. The bispecific protein of claim 1, wherein the recombinantly engineered PD-Ll-binding variant of a stefin polypeptide binds to PD-L1 with a Kd of 1X10_7M or less, a Kd of 1X10_8M or less, a Kd of 1X10_9M or less, a Kd of lxlO_10M or less, a Kd of lxlO_11M or less, or a Kd of 1X10_12M or less.
3. The bispecific protein of claim 2, wherein the TORb trap polypeptide binds to TORb with a Kd of 1X10_7M or less, a Kd of 1X10_8M or less, a Kd of 1X10_9M or less, a Kd of lxlO_10M or less, a Kd of lxlO_11M or less, or a Kd of 1X10_12M or less.
4. The bispecific protein of any one of claims 1-3, wherein the TORb trap polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98% identity, or 100% identity to the amino acid sequence of SEQ ID NO: 117.
5. The bispecific protein of any one of claims 1-3, wherein the TϋEb trap polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98% identity, or 100% identity to the amino acid sequence of SEQ ID NO: 118.
6. The bispecific protein of any one of claims 1-3, wherein the TORb trap polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98% identity, or 100% identity to the amino acid sequence of SEQ ID NO: 119/120.
7. The bispecific protein of any one of the preceding claims, wherein the recombinantly engineered PD-Ll-binding variant of a stefin polypeptide comprises a loop 2 sequence of any one of SEQ ID NOs: 6-41.
8. The bispecific protein of any one of the preceding claims, wherein the recombinantly engineered PD-Ll-binding variant of a stefin polypeptide comprises a loop 4 sequence of any one of SEQ ID NOs: 42-77.
9. The bispecific protein of any one of the preceding claims, wherein the recombinantly engineered PD-Ll-binding variant of a stefin polypeptide comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98% identity, or 100% identity to the amino acid sequence of SEQ ID NOs: 78-86.
10. The bispecific protein of any one of the preceding claims encoding by a nucleic acid sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or 100% identity to the amino acid sequence of any one of SEQ ID NOs: 87-95.
11. The bispecific protein of any one of the preceding claims, wherein the PD-Ll-binding recombinantly engineered variant of stefin polypeptide comprise an amino acid sequence represented in general formula (I)
FRl-(Xaa)n-FR2-(Xaa)m-FR3 (I), wherein
FR1 is an amino acid sequence having at least 70% identity to MIPGGLSEAK PATPEIQEIV DKVKPQLEEK TNETYGKLEA VQYKTQVLA (SEQ ID NO: 1);
FR2 is an amino acid sequence having at least 70% identity to GTNYYIKVRA GDNKYMHLKV FKSL (SEQ ID NO: 2);
FR3 is an amino acid sequence having at least 70% identity to EDLVLTGYQV DKNKDDELTG F (SEQ ID NO: 3);
Xaa, individually for each occurrence, is an amino acid; and n is an integer from 3 to 20, and m is an integer from 3 to 20.
12. The bispecific protein of claim 14, wherein:
FR1 has at least 80%, at least 82%, at least 84%, at least 86%, at least 88%, at least 90%, at least 92%, at least 94%, at least 96%, or at least 98% identity to SEQ ID NO: 1;
FR2 has at least 80%, at least 84%, at least 88%, at least 92%, or at least 96% identity to SEQ ID NO: 2; and/or.
FR3 has at least 80%, at least 85%, at least 90%, or at least 95% identity to SEQ ID NO: 3.
13. The bispecific protein of claim 14, wherein:
FR1 comprises the amino acid sequence of SEQ ID NO: 1;
FR2 comprises the amino acid sequence of SEQ ID NO: 2; and/or FR3 comprises the amino acid sequence of SEQ ID NO: 3.
14. The bispecific protein of any of claims 11-13, wherein (Xaa)n is at least 75% identical to a Loop 2 sequence selected from SEQ ID NOS: 6-41.
15. The bispecific protein of any of claims 11-14, wherein (Xaa)m is at least 75% identical to a Loop 4 sequence selected from SEQ ID NOS: 42-77.
16. The bispecific protein of any one of the preceding claims, wherein the bispecific protein includes at least one cysteine, which is optionally available for chemical conjugation, and which (optionally) is located at the C-terminal end or the N-terminal end of the bispecific protein.
17. The bispecific protein of any one of the preceding claims, wherein the PD-Ll-binding variant of a stefin polypeptide binds human PD-L1 as a monomer with an IC50 in a competitive binding assay with human PD-1 of 500 nM or less, 400 nM or less, 300 nM or less, 200 nM or less, or 100 nM or less.
18. The bispecific protein of any one of the preceding claims, wherein the TΰRb trap polypeptide binds human TGFP with an IC50 in a competitive binding assay with human TGFP of 50 pM or less, 40 pM or less, 30 pM or less, 20 pM or less, 10 pM or less, or 5 pM or less.
19. A pharmaceutical composition suitable for therapeutic use in a human subject, comprising a bispecific protein of any of any one of the preceding claims, and a pharmaceutically acceptable excipient.
20. The pharmaceutical composition of claim 19, wherein the pharmaceutical composition is formulated for pulmonary delivery or topical delivery.
21. The pharmaceutical composition of claim 20, wherein the pulmonary delivery is intranasal delivery.
22. A polynucleotide comprising a sequence encoding the bispecific protein of any of any one of the preceding claims.
23. The polynucleotide of claim 22, wherein the sequence encoding the bispecific protein is operably linked to a transcriptional regulatory sequence.
24. The polynucleotide of claim 23, wherein the transcriptional regulatory sequence is selected from the group consisting of promoters and enhancers.
25. The polynucleotide of any of claims 22-24, wherein the polynucleotide comprises a deoxyribonucleic acid (DNA) or a ribonucleic acid (RNA).
26. A viral vector comprising the polynucleotide of any of claims 22-25.
27. A plasmid or minicircle comprising the polynucleotide any of claims 22-25.
28. A cell comprising the polypeptide of any one of the preceding claims, the polynucleotide of any one of the preceding claims, the viral vector of claim 26, or the plasmid or minicircle of claim 27.
29. The bispecific protein of any one of the preceding claims for use in a method for treating cancer.
30. A method of producing the bispecific protein of any one of the preceding claims, the method comprising expressing in a host cell a nucleic acid encoding the polypeptide, and optionally isolating the polypeptide from the host cell.
PCT/IB2020/000899 2019-10-16 2020-10-15 PD-L1 INHIBITOR - TGFβ INHIBITOR BISPECIFIC DRUG MOIETIES. WO2021074695A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962915837P 2019-10-16 2019-10-16
US62/915,837 2019-10-16

Publications (2)

Publication Number Publication Date
WO2021074695A1 true WO2021074695A1 (en) 2021-04-22
WO2021074695A8 WO2021074695A8 (en) 2021-07-22

Family

ID=73646360

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2020/000899 WO2021074695A1 (en) 2019-10-16 2020-10-15 PD-L1 INHIBITOR - TGFβ INHIBITOR BISPECIFIC DRUG MOIETIES.

Country Status (2)

Country Link
TW (1) TW202128775A (en)
WO (1) WO2021074695A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022090529A1 (en) * 2020-11-02 2022-05-05 Ares Trading S.A. Combination treatment of cancer
WO2022234003A1 (en) * 2021-05-07 2022-11-10 Avacta Life Sciences Limited Cd33 binding polypeptides with stefin a protein
WO2023057567A1 (en) * 2021-10-07 2023-04-13 Avacta Life Sciences Limited Pd-l1 binding affimers

Citations (107)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3805783A (en) 1971-02-12 1974-04-23 A Ismach Hand powered hypodermic jet injector gun
US4342310A (en) 1980-07-08 1982-08-03 Istvan Lindmayer Hydro-pneumatic jet injector
US4447223A (en) 1982-04-16 1984-05-08 Cct Associates Medicament implant applicator
EP0216846A1 (en) 1985-04-01 1987-04-08 Celltech Ltd Transformed myeloma cell-line and a process for the expression of a gene coding for a eukaryotic polypeptide employing same.
EP0256055A1 (en) 1986-01-23 1988-02-24 Celltech Ltd Recombinant dna sequences, vectors containing them and method for the use thereof.
US4797368A (en) 1985-03-15 1989-01-10 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus as eukaryotic expression vector
EP0307434A1 (en) 1987-03-18 1989-03-22 Medical Res Council Altered antibodies.
WO1989005345A1 (en) 1987-12-11 1989-06-15 Whitehead Institute For Biomedical Research Genetic modification of endothelial cells
EP0323997A1 (en) 1987-07-23 1989-07-19 Celltech Limited Recombinant dna expression vectors
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
WO1990006997A1 (en) 1988-12-13 1990-06-28 United States Government As Represented By The Secretary Of The Department Of Health And Human Services Genetically engineered endothelial cells and use thereof
WO1990014363A1 (en) 1989-05-19 1990-11-29 Amgen Inc. Metalloproteinase inhibitor
US5075109A (en) 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
WO1992005266A2 (en) 1990-09-21 1992-04-02 Viagene, Inc. Packaging cells
WO1992007573A1 (en) 1990-10-31 1992-05-14 Somatix Therapy Corporation Genetic modification of endothelial cells
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
WO1992014829A1 (en) 1991-02-19 1992-09-03 The Regents Of The University Of California Viral particles having altered host range
US5151254A (en) 1989-08-29 1992-09-29 Suzuki Jidosha Kogyo Kabushiki Kaisha Exhaust gas cleaning device for internal combustion engine
US5252479A (en) 1991-11-08 1993-10-12 Research Corporation Technologies, Inc. Safe vector for gene therapy
WO1994012629A1 (en) 1992-12-02 1994-06-09 Baylor College Of Medicine Episomal vectors for gene therapy
WO1994020078A1 (en) 1993-03-02 1994-09-15 Biovector Therapeutics Sa Synthetic particulate vectors and method for preparing same
WO1994023701A1 (en) 1993-04-21 1994-10-27 Institut Pasteur Particulate vector and pharmaceutical composition containing such vector
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5407609A (en) 1989-05-04 1995-04-18 Southern Research Institute Microencapsulation process and products therefrom
WO1996006638A1 (en) 1994-08-31 1996-03-07 Biovector Therapeutics S.A. Method for enhancing immunogenicity, product obtained and pharmaceutical composition
US5505697A (en) 1994-01-14 1996-04-09 Mckinnon, Jr.; Charles N. Electrically powered jet injector
US5676646A (en) 1992-04-08 1997-10-14 Genetronics, Inc. Flow through electroporation apparatus
US5702359A (en) 1995-06-06 1997-12-30 Genetronics, Inc. Needle electrodes for mediated delivery of drugs and genes
WO1998007408A1 (en) 1996-08-19 1998-02-26 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Novel liposome complexes for increased systemic delivery
WO1998007876A2 (en) 1996-08-16 1998-02-26 Medical Research Council Self-replicating episomal expression vectors conferring tissue-specific gene expression
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US5811128A (en) 1986-10-24 1998-09-22 Southern Research Institute Method for oral or rectal delivery of microencapsulated vaccines and compositions therefor
US5928647A (en) 1993-01-11 1999-07-27 Dana-Farber Cancer Institute Inducing cytotoxic T lymphocyte responses
WO1999040934A1 (en) 1998-02-12 1999-08-19 Immune Complex, Corporation Strategically modified hepatitis b core proteins and their derivatives
US6009347A (en) 1998-01-27 1999-12-28 Genetronics, Inc. Electroporation apparatus with connective electrode template
US6068650A (en) 1997-08-01 2000-05-30 Gentronics Inc. Method of Selectively applying needle array configurations
US6080728A (en) 1996-07-16 2000-06-27 Mixson; A. James Carrier: DNA complexes containing DNA encoding anti-angiogenic peptides and their use in gene therapy
US6096020A (en) 1996-09-09 2000-08-01 Genetronics, Inc. Electroporation employing user-configured pulsing scheme
US6120493A (en) 1998-01-27 2000-09-19 Genetronics, Inc. Method for the introduction of therapeutic agents utilizing an electroporation apparatus
US6150148A (en) 1998-10-21 2000-11-21 Genetronics, Inc. Electroporation apparatus for control of temperature during the process
US6192270B1 (en) 1998-08-14 2001-02-20 Genetronics, Inc. Apparatus and method for the delivery of drugs and genes into tissue
US6208893B1 (en) 1998-01-27 2001-03-27 Genetronics, Inc. Electroporation apparatus with connective electrode template
US6216034B1 (en) 1997-08-01 2001-04-10 Genetronics, Inc. Method of programming an array of needle electrodes for electroporation therapy of tissue
US6241701B1 (en) 1997-08-01 2001-06-05 Genetronics, Inc. Apparatus for electroporation mediated delivery of drugs and genes
US6302874B1 (en) 1998-07-13 2001-10-16 Genetronics, Inc. Method and apparatus for electrically assisted topical delivery of agents for cosmetic applications
US6520950B1 (en) 1999-05-10 2003-02-18 Genetronics, Inc. Method of electroporation-enhanced delivery of active agents
WO2004041862A2 (en) 2002-11-08 2004-05-21 Ablynx N.V. Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US6763264B2 (en) 1993-04-01 2004-07-13 Genetronics, Inc. Method of treatment using electroporation mediated delivery of drugs and genes
US20050005263A1 (en) 2001-08-30 2005-01-06 Mitsuhiro Miyazaki Information processing apparatus and method
US20050052630A1 (en) 2002-03-07 2005-03-10 Advisys, Inc. Constant current electroporation device and methods of use
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
WO2006122786A2 (en) 2005-05-18 2006-11-23 Ablynx Nv Improved nanobodies™ against tumor necrosis factor-alpha
US7171264B1 (en) 1999-05-10 2007-01-30 Genetronics, Inc. Intradermal delivery of active agents by needle-free injection and electroporation
US7214775B2 (en) 1999-04-09 2007-05-08 Kyowa Hakko Kogyo Co., Ltd. Method of modulating the activity of functional immune molecules
WO2007054279A2 (en) 2005-11-08 2007-05-18 Helmholtz-Zentrum für Infektionsforschung GmbH Cyclic-dinucleotides and its conjugates as adjuvants and their uses in pharmaceutical compositions
US7245963B2 (en) 2002-03-07 2007-07-17 Advisys, Inc. Electrode assembly for constant-current electroporation and use
US7328064B2 (en) 2002-07-04 2008-02-05 Inovio As Electroporation device and injection apparatus
WO2008020079A1 (en) 2006-08-18 2008-02-21 Ablynx N.V. Amino acid sequences directed against il-6r and polypeptides comprising the same for the treatment of deseases and disorders associated with il-6-mediated signalling
WO2008142164A2 (en) 2007-05-24 2008-11-27 Ablynx N.V. Amino acid sequences directed against rank-l and polypeptides comprising the same for the treatment of bone diseases and disorders
WO2009068627A2 (en) 2007-11-27 2009-06-04 Ablynx N.V. Amino acid sequences directed against heterodimeric cytokines and/or their receptors and polypeptides comprising the same
US7592326B2 (en) 2004-03-15 2009-09-22 Karaolis David K R Method for stimulating the immune, inflammatory or neuroprotective response
WO2010045002A2 (en) 2008-09-26 2010-04-22 Tocagen Inc. Gene therapy vectors and cytosine deaminases
US7709458B2 (en) 2004-03-15 2010-05-04 David K. R. Karaolis Method for inhibiting cancer cell proliferation or increasing cancer cell apoptosis
US7790446B2 (en) 2005-02-11 2010-09-07 Kosagen Cell Factory Oü Vectors, cell lines and their use in obtaining extended episomal maintenance replication of hybrid plasmids and expression of gene products
WO2010148203A2 (en) 2009-06-17 2010-12-23 Tocagen Inc. Producer cells for replication competent retroviral vectors
US7923251B2 (en) 2005-02-23 2011-04-12 The Board Of Trustees Of The Leland Stanford Junior University Method and apparatus for avalanche-mediated transfer of agents into cells
US8026223B1 (en) 2003-05-30 2011-09-27 University Of South Florida Treating malignant tumors with high field strength electroporation of plasmids encoding IL-12
WO2011126864A2 (en) 2010-03-29 2011-10-13 Tocagen Inc. Cancer treatment with recombinant vector
WO2011140086A2 (en) 2010-05-03 2011-11-10 Bristol-Myers Squibb Company Serum albumin binding molecules
US8101169B2 (en) 2005-02-23 2012-01-24 The Board Of Trustees Of The Leland Stanford Junior University Ocular gene therapy using avalanche-mediated transfection
WO2012058673A2 (en) 2010-10-31 2012-05-03 Tocagen Inc. Enhanced cancer treatment and monitoring using recombinant vectors
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2014066655A2 (en) 2012-10-25 2014-05-01 Oncosec Medical Incorporation Electroporation device
WO2014066700A1 (en) 2012-10-25 2014-05-01 Tocagen Inc. Retroviral vector with mini-promoter cassette
WO2014093936A1 (en) 2012-12-13 2014-06-19 Aduro Biotech, Inc. Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use
WO2014179664A2 (en) 2013-05-02 2014-11-06 Anaptysbio, Inc. Antibodies directed against programmed death-1 (pd-1)
WO2014179335A1 (en) 2013-04-29 2014-11-06 Memorial Sloan Kettering Cancer Center Compositions and methods for altering second messenger signaling
WO2014189805A1 (en) 2013-05-18 2014-11-27 Auro Biotech, Inc. Compositions and methods for activating "stimulator of interferon gene"-dependent signalling
WO2015021077A1 (en) 2013-08-05 2015-02-12 Tocagen Inc. Recombinant vector with optimized a-bulge
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015089511A2 (en) 2013-12-13 2015-06-18 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
WO2015105926A1 (en) 2014-01-08 2015-07-16 Moderna Therapeutics, Inc. Polynucleotides for the in vivo production of antibodies
WO2015143199A1 (en) 2014-03-20 2015-09-24 Bristol-Myers Squibb Company Serum albumin-binding fibronectin type iii domains
WO2015148683A1 (en) 2014-03-26 2015-10-01 Tocagen Inc. A retroviral vector having immune-stimulating activity
WO2015185565A1 (en) 2014-06-04 2015-12-10 Glaxosmithkline Intellectual Property Development Limited Cyclic di-nucleotides as modulators of sting
WO2015196118A1 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
WO2015196130A2 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
WO2015196128A2 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
WO2016011226A1 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2016096174A1 (en) 2014-12-16 2016-06-23 Invivogen Fluorinated cyclic dinucleotides for cytokine induction
WO2016112359A1 (en) 2015-01-09 2016-07-14 Oncosec Medical Incorporated Method for the treatment of malignancies
WO2016118725A1 (en) 2015-01-23 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
WO2016118724A1 (en) 2015-01-21 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
US9405700B2 (en) 2010-11-04 2016-08-02 Sonics, Inc. Methods and apparatus for virtualization in an integrated circuit
WO2016145102A1 (en) 2015-03-10 2016-09-15 Aduro Biotech, Inc. Compositions and methods for activating "stimulator of interferon gene" -dependent signalling
WO2016154473A1 (en) 2015-03-26 2016-09-29 Oncosec Medical Incorporated Method for the treatment of malignancies
WO2016161201A2 (en) 2015-03-31 2016-10-06 Oncosec Medical Incorporated Systems and methods for improved tissue-sensing based electroporation
WO2017027645A1 (en) 2015-08-13 2017-02-16 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
WO2017040815A1 (en) 2015-09-04 2017-03-09 Tocagen Inc. Recombinant vectors comprising 2a peptide
WO2017036889A1 (en) 2015-08-28 2017-03-09 Biontech Rna Pharmaceuticals Gmbh Method for reducing immunogenicity of rna
WO2017049275A2 (en) 2015-09-17 2017-03-23 Moderna Therapeutics, Inc. Polynucleotides containing a stabilizing tail region
WO2017053617A1 (en) 2015-09-23 2017-03-30 Bristol-Myers Squibb Company Fast-off rate serum albumin binding fibronectin type iii domains
WO2017075477A1 (en) 2015-10-28 2017-05-04 Aduro Biotech, Inc. Compositions and methods for activating "stimulator of interferon gene"-dependent signalling
WO2017106795A1 (en) 2015-12-18 2017-06-22 Oncosec Medical Incorporated Plasmid constructs for heterologous protein expression and methods of use
WO2017162266A1 (en) 2016-03-21 2017-09-28 Biontech Rna Pharmaceuticals Gmbh Rna replicon for versatile and efficient gene expression
US20170342424A1 (en) 2001-04-10 2017-11-30 Plasmidfactory Gmbh Co & Kg Methods of producing recombinant minicircle constructs

Patent Citations (116)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3805783A (en) 1971-02-12 1974-04-23 A Ismach Hand powered hypodermic jet injector gun
US4342310A (en) 1980-07-08 1982-08-03 Istvan Lindmayer Hydro-pneumatic jet injector
US4447223A (en) 1982-04-16 1984-05-08 Cct Associates Medicament implant applicator
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US4797368A (en) 1985-03-15 1989-01-10 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus as eukaryotic expression vector
EP0216846A1 (en) 1985-04-01 1987-04-08 Celltech Ltd Transformed myeloma cell-line and a process for the expression of a gene coding for a eukaryotic polypeptide employing same.
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
EP0256055A1 (en) 1986-01-23 1988-02-24 Celltech Ltd Recombinant dna sequences, vectors containing them and method for the use thereof.
US5811128A (en) 1986-10-24 1998-09-22 Southern Research Institute Method for oral or rectal delivery of microencapsulated vaccines and compositions therefor
US5820883A (en) 1986-10-24 1998-10-13 Southern Research Institute Method for delivering bioactive agents into and through the mucosally-associated lymphoid tissues and controlling their release
US5814344A (en) 1986-10-24 1998-09-29 Southern Research Institute Method for delivering bioactive agents into and through the mucosally associated lymphoid tissues and controlling their release
US5853763A (en) 1986-10-24 1998-12-29 Southern Research Institute Method for delivering bioactive agents into and through the mucosally-associated lymphoid tissue and controlling their release
US5075109A (en) 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
US5942252A (en) 1986-10-24 1999-08-24 Southern Research Institute Method for delivering bioactive agents into and through the mucosally-associated lymphoid tissues and controlling their release
EP0307434A1 (en) 1987-03-18 1989-03-22 Medical Res Council Altered antibodies.
EP0323997A1 (en) 1987-07-23 1989-07-19 Celltech Limited Recombinant dna expression vectors
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
WO1989005345A1 (en) 1987-12-11 1989-06-15 Whitehead Institute For Biomedical Research Genetic modification of endothelial cells
WO1990006997A1 (en) 1988-12-13 1990-06-28 United States Government As Represented By The Secretary Of The Department Of Health And Human Services Genetically engineered endothelial cells and use thereof
US5407609A (en) 1989-05-04 1995-04-18 Southern Research Institute Microencapsulation process and products therefrom
WO1990014363A1 (en) 1989-05-19 1990-11-29 Amgen Inc. Metalloproteinase inhibitor
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5151254A (en) 1989-08-29 1992-09-29 Suzuki Jidosha Kogyo Kabushiki Kaisha Exhaust gas cleaning device for internal combustion engine
WO1992005266A2 (en) 1990-09-21 1992-04-02 Viagene, Inc. Packaging cells
WO1992007573A1 (en) 1990-10-31 1992-05-14 Somatix Therapy Corporation Genetic modification of endothelial cells
WO1992014829A1 (en) 1991-02-19 1992-09-03 The Regents Of The University Of California Viral particles having altered host range
US5252479A (en) 1991-11-08 1993-10-12 Research Corporation Technologies, Inc. Safe vector for gene therapy
US5676646A (en) 1992-04-08 1997-10-14 Genetronics, Inc. Flow through electroporation apparatus
WO1994012629A1 (en) 1992-12-02 1994-06-09 Baylor College Of Medicine Episomal vectors for gene therapy
US5928647A (en) 1993-01-11 1999-07-27 Dana-Farber Cancer Institute Inducing cytotoxic T lymphocyte responses
WO1994020078A1 (en) 1993-03-02 1994-09-15 Biovector Therapeutics Sa Synthetic particulate vectors and method for preparing same
US6763264B2 (en) 1993-04-01 2004-07-13 Genetronics, Inc. Method of treatment using electroporation mediated delivery of drugs and genes
WO1994023701A1 (en) 1993-04-21 1994-10-27 Institut Pasteur Particulate vector and pharmaceutical composition containing such vector
US5505697A (en) 1994-01-14 1996-04-09 Mckinnon, Jr.; Charles N. Electrically powered jet injector
WO1996006638A1 (en) 1994-08-31 1996-03-07 Biovector Therapeutics S.A. Method for enhancing immunogenicity, product obtained and pharmaceutical composition
US5702359A (en) 1995-06-06 1997-12-30 Genetronics, Inc. Needle electrodes for mediated delivery of drugs and genes
US6080728A (en) 1996-07-16 2000-06-27 Mixson; A. James Carrier: DNA complexes containing DNA encoding anti-angiogenic peptides and their use in gene therapy
WO1998007876A2 (en) 1996-08-16 1998-02-26 Medical Research Council Self-replicating episomal expression vectors conferring tissue-specific gene expression
WO1998007408A1 (en) 1996-08-19 1998-02-26 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Novel liposome complexes for increased systemic delivery
US6096020A (en) 1996-09-09 2000-08-01 Genetronics, Inc. Electroporation employing user-configured pulsing scheme
US6216034B1 (en) 1997-08-01 2001-04-10 Genetronics, Inc. Method of programming an array of needle electrodes for electroporation therapy of tissue
US6241701B1 (en) 1997-08-01 2001-06-05 Genetronics, Inc. Apparatus for electroporation mediated delivery of drugs and genes
US6068650A (en) 1997-08-01 2000-05-30 Gentronics Inc. Method of Selectively applying needle array configurations
US6233482B1 (en) 1997-08-01 2001-05-15 Genetronics, Inc. Method of electroporation mediated delivery of drugs and genes
US6181964B1 (en) 1997-08-01 2001-01-30 Genetronics, Inc. Minimally invasive apparatus and method to electroporate drugs and genes into tissue
US6120493A (en) 1998-01-27 2000-09-19 Genetronics, Inc. Method for the introduction of therapeutic agents utilizing an electroporation apparatus
US6208893B1 (en) 1998-01-27 2001-03-27 Genetronics, Inc. Electroporation apparatus with connective electrode template
US6009347A (en) 1998-01-27 1999-12-28 Genetronics, Inc. Electroporation apparatus with connective electrode template
WO1999040934A1 (en) 1998-02-12 1999-08-19 Immune Complex, Corporation Strategically modified hepatitis b core proteins and their derivatives
US6302874B1 (en) 1998-07-13 2001-10-16 Genetronics, Inc. Method and apparatus for electrically assisted topical delivery of agents for cosmetic applications
US6192270B1 (en) 1998-08-14 2001-02-20 Genetronics, Inc. Apparatus and method for the delivery of drugs and genes into tissue
US6150148A (en) 1998-10-21 2000-11-21 Genetronics, Inc. Electroporation apparatus for control of temperature during the process
US7214775B2 (en) 1999-04-09 2007-05-08 Kyowa Hakko Kogyo Co., Ltd. Method of modulating the activity of functional immune molecules
US7171264B1 (en) 1999-05-10 2007-01-30 Genetronics, Inc. Intradermal delivery of active agents by needle-free injection and electroporation
US6520950B1 (en) 1999-05-10 2003-02-18 Genetronics, Inc. Method of electroporation-enhanced delivery of active agents
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
US20170342424A1 (en) 2001-04-10 2017-11-30 Plasmidfactory Gmbh Co & Kg Methods of producing recombinant minicircle constructs
US20050005263A1 (en) 2001-08-30 2005-01-06 Mitsuhiro Miyazaki Information processing apparatus and method
US20050052630A1 (en) 2002-03-07 2005-03-10 Advisys, Inc. Constant current electroporation device and methods of use
US7245963B2 (en) 2002-03-07 2007-07-17 Advisys, Inc. Electrode assembly for constant-current electroporation and use
US7328064B2 (en) 2002-07-04 2008-02-05 Inovio As Electroporation device and injection apparatus
US20080234655A1 (en) 2002-07-04 2008-09-25 Inovio As Electroporation device and injection apparatus
WO2004041862A2 (en) 2002-11-08 2004-05-21 Ablynx N.V. Single domain antibodies directed against tumour necrosis factor-alpha and uses therefor
US8026223B1 (en) 2003-05-30 2011-09-27 University Of South Florida Treating malignant tumors with high field strength electroporation of plasmids encoding IL-12
US7592326B2 (en) 2004-03-15 2009-09-22 Karaolis David K R Method for stimulating the immune, inflammatory or neuroprotective response
US7709458B2 (en) 2004-03-15 2010-05-04 David K. R. Karaolis Method for inhibiting cancer cell proliferation or increasing cancer cell apoptosis
US7790446B2 (en) 2005-02-11 2010-09-07 Kosagen Cell Factory Oü Vectors, cell lines and their use in obtaining extended episomal maintenance replication of hybrid plasmids and expression of gene products
US8283171B2 (en) 2005-02-23 2012-10-09 The Board Of Trustees Of The Leland Stanford Junior University Method and apparatus for avalanche-mediated transfer of agents into cells
US7923251B2 (en) 2005-02-23 2011-04-12 The Board Of Trustees Of The Leland Stanford Junior University Method and apparatus for avalanche-mediated transfer of agents into cells
US8101169B2 (en) 2005-02-23 2012-01-24 The Board Of Trustees Of The Leland Stanford Junior University Ocular gene therapy using avalanche-mediated transfection
WO2006122786A2 (en) 2005-05-18 2006-11-23 Ablynx Nv Improved nanobodies™ against tumor necrosis factor-alpha
WO2007054279A2 (en) 2005-11-08 2007-05-18 Helmholtz-Zentrum für Infektionsforschung GmbH Cyclic-dinucleotides and its conjugates as adjuvants and their uses in pharmaceutical compositions
WO2008020079A1 (en) 2006-08-18 2008-02-21 Ablynx N.V. Amino acid sequences directed against il-6r and polypeptides comprising the same for the treatment of deseases and disorders associated with il-6-mediated signalling
WO2008142164A2 (en) 2007-05-24 2008-11-27 Ablynx N.V. Amino acid sequences directed against rank-l and polypeptides comprising the same for the treatment of bone diseases and disorders
WO2009068627A2 (en) 2007-11-27 2009-06-04 Ablynx N.V. Amino acid sequences directed against heterodimeric cytokines and/or their receptors and polypeptides comprising the same
WO2010045002A2 (en) 2008-09-26 2010-04-22 Tocagen Inc. Gene therapy vectors and cytosine deaminases
WO2010148203A2 (en) 2009-06-17 2010-12-23 Tocagen Inc. Producer cells for replication competent retroviral vectors
WO2011126864A2 (en) 2010-03-29 2011-10-13 Tocagen Inc. Cancer treatment with recombinant vector
WO2011140086A2 (en) 2010-05-03 2011-11-10 Bristol-Myers Squibb Company Serum albumin binding molecules
WO2012058673A2 (en) 2010-10-31 2012-05-03 Tocagen Inc. Enhanced cancer treatment and monitoring using recombinant vectors
US9405700B2 (en) 2010-11-04 2016-08-02 Sonics, Inc. Methods and apparatus for virtualization in an integrated circuit
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2014066655A2 (en) 2012-10-25 2014-05-01 Oncosec Medical Incorporation Electroporation device
WO2014066700A1 (en) 2012-10-25 2014-05-01 Tocagen Inc. Retroviral vector with mini-promoter cassette
WO2014093936A1 (en) 2012-12-13 2014-06-19 Aduro Biotech, Inc. Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use
WO2014179335A1 (en) 2013-04-29 2014-11-06 Memorial Sloan Kettering Cancer Center Compositions and methods for altering second messenger signaling
WO2014179664A2 (en) 2013-05-02 2014-11-06 Anaptysbio, Inc. Antibodies directed against programmed death-1 (pd-1)
WO2014189805A1 (en) 2013-05-18 2014-11-27 Auro Biotech, Inc. Compositions and methods for activating "stimulator of interferon gene"-dependent signalling
WO2015021077A1 (en) 2013-08-05 2015-02-12 Tocagen Inc. Recombinant vector with optimized a-bulge
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015089511A2 (en) 2013-12-13 2015-06-18 Moderna Therapeutics, Inc. Modified nucleic acid molecules and uses thereof
WO2015105926A1 (en) 2014-01-08 2015-07-16 Moderna Therapeutics, Inc. Polynucleotides for the in vivo production of antibodies
WO2015143199A1 (en) 2014-03-20 2015-09-24 Bristol-Myers Squibb Company Serum albumin-binding fibronectin type iii domains
WO2015148683A1 (en) 2014-03-26 2015-10-01 Tocagen Inc. A retroviral vector having immune-stimulating activity
WO2015185565A1 (en) 2014-06-04 2015-12-10 Glaxosmithkline Intellectual Property Development Limited Cyclic di-nucleotides as modulators of sting
WO2015196128A2 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
WO2015196130A2 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
WO2015196118A1 (en) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Alternative nucleic acid molecules and uses thereof
WO2016011226A1 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2016096174A1 (en) 2014-12-16 2016-06-23 Invivogen Fluorinated cyclic dinucleotides for cytokine induction
WO2016112359A1 (en) 2015-01-09 2016-07-14 Oncosec Medical Incorporated Method for the treatment of malignancies
WO2016118724A1 (en) 2015-01-21 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
WO2016118725A1 (en) 2015-01-23 2016-07-28 Moderna Therapeutics, Inc. Lipid nanoparticle compositions
WO2016145102A1 (en) 2015-03-10 2016-09-15 Aduro Biotech, Inc. Compositions and methods for activating "stimulator of interferon gene" -dependent signalling
WO2016154473A1 (en) 2015-03-26 2016-09-29 Oncosec Medical Incorporated Method for the treatment of malignancies
WO2016161201A2 (en) 2015-03-31 2016-10-06 Oncosec Medical Incorporated Systems and methods for improved tissue-sensing based electroporation
WO2017027645A1 (en) 2015-08-13 2017-02-16 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
WO2017027646A1 (en) 2015-08-13 2017-02-16 Merck Sharp & Dohme Corp. Cyclic di-nucleotide compounds as sting agonists
WO2017036889A1 (en) 2015-08-28 2017-03-09 Biontech Rna Pharmaceuticals Gmbh Method for reducing immunogenicity of rna
WO2017040815A1 (en) 2015-09-04 2017-03-09 Tocagen Inc. Recombinant vectors comprising 2a peptide
WO2017049275A2 (en) 2015-09-17 2017-03-23 Moderna Therapeutics, Inc. Polynucleotides containing a stabilizing tail region
WO2017053617A1 (en) 2015-09-23 2017-03-30 Bristol-Myers Squibb Company Fast-off rate serum albumin binding fibronectin type iii domains
WO2017075477A1 (en) 2015-10-28 2017-05-04 Aduro Biotech, Inc. Compositions and methods for activating "stimulator of interferon gene"-dependent signalling
WO2017106795A1 (en) 2015-12-18 2017-06-22 Oncosec Medical Incorporated Plasmid constructs for heterologous protein expression and methods of use
WO2017162266A1 (en) 2016-03-21 2017-09-28 Biontech Rna Pharmaceuticals Gmbh Rna replicon for versatile and efficient gene expression

Non-Patent Citations (217)

* Cited by examiner, † Cited by third party
Title
"NCBI", Database accession no. NP_001020018
"Remington: The Science and Practice of Pharmacy", 2012, PHARMACEUTICAL PRESS
"UniProtKB", Database accession no. Q9NZQ7
"United States Patent Office Manual of Patent Examining Procedures"
ABROI ET AL.: "Analysis of chromatin attachment and partitioning functions of bovine papillomavirus type 1 E2 protein", JOURNAL OF VIROLOGY, vol. 78, 2004, pages 2100 - 13, XP002380473
AIHARA ET AL.: "Gene transfer into muscle by electroporation in vivo", NAT. BIOTECHNOL., vol. 16, 1998, pages 867 - 870, XP001061636, DOI: 10.1038/nbt0998-867
ALEXANDER ET AL., MOL. CELL. BIOL., vol. 7, 1987, pages 1436 - 44
AMRIK BASRAN ET AL: "2248: Generation and formatting of a bispecific Affimer biotherapeutic for the inhibition of the LAG-3 and PD-L1 pathway", 1 January 2019 (2019-01-01), XP055770183, Retrieved from the Internet <URL:https://cancerres.aacrjournals.org/content/79/13_Supplement/2248> [retrieved on 20210129], DOI: 10.1158/1538-7445.AM2019-2248Published *
ANDERSEN ET AL.: "Extending half-life by indirect targeting of the neonatal Fc receptor (FcRn) using a minimal albumin binding domain", J BIOL CHEM, vol. 286, 2011, pages 5234 - 41, XP055019819, DOI: 10.1074/jbc.M110.164848
ARGOS P.: "An investigation of oligopeptides linking domains in protein tertiary structures and possible candidates for general gene fusion", J MOL BIOL., vol. 211, 1990, pages 943 - 958, XP024010410, DOI: 10.1016/0022-2836(90)90085-Z
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1989, JOHN WILEY & SONS
B GOMES ET AL: "Isolation of Affimer therapeutics with nanomolar affinity to human PD-L1 from a synthetic, phage display library", 3 October 2017 (2017-10-03), XP055609131, Retrieved from the Internet <URL:https://www.myeventflo.com/event-lecture.asp?lectID=13653> [retrieved on 20190726] *
BAICHWALSUGDEN: "Gene transfer", 1986, PLENUM PRESS, article "Vectors for gene transfer derived from animal DNA viruses: Transient and stable expression of transferred genes", pages: 117 - 148
BEBBINGTONHENTSCHEL: "DNA CLONING", vol. 3, 1987, ACADEMIC PRESS, article "The Use of Vectors Based On Gene Amplification For The Expression Of Cloned Genes In Mammalian Cells"
BECK ET AL.: "Therapeutic Fc-fusion proteins and peptides as successful alternatives to antibodies", MABS, vol. 3, 2011, pages 1 - 2
BECKMAN ET AL.: "Antibody constructs in cancer therapy: protein engineering strategies to improve exposure in solid tumors", CANCER, vol. 109, no. 2, 2007, pages 170 - 9
BECKMAN ET AL.: "Antibody constructs in cancer therapy: protein engineering strategies to improve exposure in solid tumors", CANCER, vol. 109, no. 2, 2015, pages 170 - 9
BERNOISTCHAMBON, NATURE, vol. 290, 1981, pages 304 - 310
BHATT ET AL.: "Microbeads mediated oral plasmid DNA delivery using polymethacrylate vectors: an effectual groundwork for colorectal cancer", DRUG DELIV, vol. 22, 2015, pages 849 - 861
BIOCHEMISTRY, vol. 11, 1972, pages 1726 - 1732
BITTER ET AL.: "Expression and Secretion Vectors For Yeast", METHODS IN ENZYMOL, vol. 153, 1987, pages 516 - 544
BOESEN ET AL., BIOTHERAPY, vol. 6, 1994, pages 291 - 302
BOSHART ET AL., CELL, vol. 41, no. 2, 1985, pages 521 - 30
BRINSTER ET AL., NATURE, vol. 296, 1982, pages 39 - 42
BRODERICK ET AL.: "Enhanced delivery of DNA or RNA vaccines by electroporation", METHODS MOL BIOL, vol. 1499, 2017, pages 193 - 200
BUCHSCHER ET AL., J. VIROL., vol. 66, 1992, pages 1635 - 1640
CAI ET AL.: "Plasma pharmacokinetics of veledimex, a small-molecule activator ligand for a proprietary gene therapy promoter system, in healthy subjects", CLIN PHARMACOL DRUG DEV, 2016
CALICETI ET AL.: "Pharmacokinetic and biodistribution properties of poly(ethylene glycol)-protein conjugates", ADV DRUG DELIVERY REV, vol. 55, 2003, pages 1261 - 77, XP003001867, DOI: 10.1016/S0169-409X(03)00108-X
CHAIBERG ET AL., INVESTIGATIVE OPHTHALMOLOGY & VISUAL SCIENCE, vol. 47, 2006, pages 4083 - 4090
CHAMBER R. ET AL., PROC. NATL. ACAD. SCI USA, vol. 92, 1995, pages 1411 - 1415
CHAPMAN, ADV DRUG DELIV REV, vol. 54, 2002, pages 531 - 545
CHEN ET AL., MOL. THER., vol. 8, 2003, pages 495 - 500
CHEN Z YHE C YEHRHARDT AKAY M A, MOL THER, vol. 8, no. 3, 2003, pages 495 - 500
CHIANG ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 5799 - 5803
COLBERE-GARAPIN ET AL.: "A New Dominant Hybrid Selective Marker For Higher Eukaryotic Cells", J. MOL. BIOL., vol. 150, 1981, pages 1 - 14, XP024018978, DOI: 10.1016/0022-2836(81)90321-1
CONNOLLY ET AL., BIOELECTROCHEMISTRY, vol. 103, 2015, pages 15 - 21
CONNOLLY ET AL., HUMAN VACCINES & IMMUNE-THERAPEUTICS, vol. 8, 2012, pages 1729 - 1733
CONTERMANN, CURR OP BIOTECHNOL, vol. 22, 2011, pages 868 - 876
COUPAR ET AL., GENE, vol. 68, 1988, pages 1 - 10
CROUSE ET AL.: "Expression and Amplification Of Engineered Mouse Dihydrofolate Reductase Minigenes", CELL. BIOL., vol. 3, 1983, pages 257 - 266
CUNNINGHAM ET AL., MOL THER, vol. 16, no. 6, 2008, pages 1081 - 8
CZAJKOWSKY ET AL.: "Fc-fusion proteins: new developments and future perspectives", EMBO MOL MED, vol. 4, 2012, pages 1015 - 28, XP055542855, DOI: 10.1002/emmm.201201379
DARQUET ET AL., GENE THER, vol. 4, no. 12, 1997, pages 1341 - 9
DARQUET ET AL., GENE THER, vol. 6, no. 2, 1999, pages 209 - 18
DE BENEDETTIRHOADS, NUCLEIC ACIDS RES, vol. 19, 1991, pages 1925
DEAL ET AL.: "Engineering humoral immunity as prophylaxis or therapy", CURR OPIN IMMUNOL, vol. 35, 2015, pages 113 - 22
DEBOER, PROC. NATL. ACAD. SCI. U.S.A., vol. 80, 1983, pages 21 - 5
DIAS ET AL.: "Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4", GENE THER, vol. 19, no. 10, 2012, pages 988 - 98
DIETZ ET AL., VECTOR ENGINEERING AND DELIVERY MOLECULAR THERAPY, vol. 21, no. 8, 2013, pages 1526 - 1535
DIGHE ET AL., PLOS ONE, vol. 9, no. 8, 2014, pages e104805
DONELLO ET AL., J VIROL, vol. 72, no. 6, 1998, pages 5085 - 501
DR ALASTAIR SMITH ET AL: "Non-confidential Technical Introduction to the Affimer Technology for Therapeutics and Reagents", 1 November 2017 (2017-11-01), XP055770316, Retrieved from the Internet <URL:https://www.cosmobio.co.jp/product/uploads/document/ALS_Affimer_document2.pdf> [retrieved on 20210129] *
DRONCA ET AL.: "Immunomodulatory antibody therapy of cancer: the closer, the better", CLIN CANCER RES, vol. 21, no. 5, 2015, pages 944 - 6, XP055397745, DOI: 10.1158/1078-0432.CCR-14-2111
DUIJKERS ET AL.: "Single dose pharmacokinetics and effects on follicular growth and serum hormones of a long-acting recombinant FSH preparation (FSHCTP) in healthy pituitary-suppressed females", HUM REPROD, vol. 17, 2002, pages 1987 - 93, XP055409330, DOI: 10.1093/humrep/17.8.1987
ECK ET AL.: "Goodman & Gilman's The Pharmacological Basis of Therapeutics", 1996, MCGRAW-HILL, pages: 77 - 101
FANG ET AL.: "An antibody delivery system for regulated expression of therapeutic levels of monoclonal antibodies in vivo", MOL THER, vol. 5, no. 6, 2007, pages 1153 - 9, XP002560535, DOI: 10.1038/SJ.MT.6300142
FARES ET AL.: "Design of a longacting follitropin agonist by fusing the C-terminal sequence of the chorionic gonadotropin beta subunit to the follitropin beta subunit", PROC NATL ACAD SCI USA., vol. 89, 1992, pages 4304 - 8
FEIGNER ET AL.: "Lipofection: a highly efficient, lipid-mediated DNA-transfection procedure", MNAS, vol. 84, 1987, pages 7413 - 7417, XP002015347, DOI: 10.1073/pnas.84.21.7413
FLINGAI ET AL.: "Protection against dengue disease by synthetic polynucleotide antibody prophylaxis/immunotherapy", SCI REP, vol. 5, 2015, pages 12616
FLOSS ET AL.: "Elastin-like polypeptides revolutionize recombinant protein expression and their biomedical application", TRENDS BIOTECHNOL, vol. 28, 2012, pages 37 - 45
FLOSS ET AL.: "fusion protein technologies for biopharmaceuticals: application and challenges. Hoboken", 2013, WILEY, article "ELP-fusion technology for biopharmaceuticals", pages: 372 - 98
FLOTTE ET AL., GENE THERAPY, vol. 2, 1995, pages 29 - 37
GENE THERAPY CLINICAL TRIALS WORLDWIDE, 2017, Retrieved from the Internet <URL:abedia.com/wiley>
GEORGE ET AL.: "An analysis of protein domain linkers: their classification and role in protein folding", PROTEIN ENG., vol. 15, no. 11, 2002, pages 871 - 9, XP002374925, DOI: 10.1093/protein/15.11.871
GRACEY MANIAR L EMANIAR J MCHEN Z YLU JFIRE A ZKAY M A, MOL THER, vol. 21, no. 1, January 2013 (2013-01-01), pages 131 - 8
GRAY ET AL., HUM GENE THER, vol. 22, 2011, pages 1143 - 1153
GREGORIADIS ET AL.: "Improving the therapeutic efficacy of peptides and proteins: a role for polysialic acids", INT J PHARM, vol. 300, 2005, pages 125 - 30, XP005013268, DOI: 10.1016/j.ijpharm.2005.06.007
GROSSCHEDL ET AL., CELL, vol. 38, 1984, pages 647 - 58
GROSSMANWILSON, CURR. OPIN. IN GENETICS AND DEVEL., vol. 3, no. 14, 1993, pages 110 - 1
GUO ET AL., ADV. CANCER RES., vol. 119, 2013, pages 421 - 475
HAMMER ET AL., SCIENCE, vol. 235, 1987, pages 53 - 8
HARDEE ET AL.: "Advances in non-viral DNA vectors for gene therapy", GENES, vol. 8, no. 2, 2017, pages 65, XP055472444, DOI: 10.3390/genes8020065
HEINZERLING ET AL.: "Intratumoral injection of DNA encoding human interleukin 12 into patients with metastatic melanoma: clinical efficacy", HUM. GENE THER., vol. 16, 2005, pages 35 - 48
HELLER LC.: "Gene electrotransfer clinical trials", ADV GENET, vol. 89, 2015, pages 235 - 62
HOU ET AL., MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT, 2015
HUANG ET AL.: "Receptor-Fc fusion therapeutics, traps, and Mimetibody technology", CURR OPIN BIOTECHNOL, vol. 20, 2009, pages 692 - 9, XP026778880, DOI: 10.1016/j.copbio.2009.10.010
HUANGGORMAN, MOL CELL BIOL, vol. 10, no. 4, 1990, pages 1805 - 10
HUANGYEN, J VIROL, vol. 68, no. 5, 1994, pages 3193 - 9
INOUYE ET AL.: "Up-Promoter Mutations In The Lpp Gene Of Escherichia coli", NUCLEIC ACIDS RES, vol. 13, 1985, pages 3101 - 3110, XP001316824
JAMES ET AL., GENE, vol. 86, 1990, pages 233 - 239
JAROSZESKI ET AL.: "In vivo gene delivery by electroporation", ADV. DRUG DELIVERY REV., vol. 35, 1999, pages 131 - 137
JEVSEVAR ET AL., METHODS MOL BIOL, vol. 901, 2012, pages 233 - 246
JEVSEVAR ET AL.: "PEGylation of therapeutic proteins", BIOTECHNOL J, vol. 5, 2010, pages 113 - 28
JONASSEN ET AL., PHARM RES, vol. 29, no. 8, 2012, pages 2104 - 14
KANG ET AL.: "Emerging PEGylated drugs", EXPERT OPIN EMERG DRUGS, vol. 14, 2009, pages 363 - 80, XP008164958, DOI: 10.1517/14728210902907847
KARIN M. KNUDSON ET AL: "M7824, a novel bifunctional anti-PD-L1/TGF[beta] Trap fusion protein, promotes anti-tumor efficacy as monotherapy and in combination with vaccine", ONCOIMMUNOLOGY, vol. 7, no. 5, 14 February 2018 (2018-02-14), pages e1426519, XP055665440, DOI: 10.1080/2162402X.2018.1426519 *
KASAHARA ET AL., SCIENCE, vol. 266, 1994, pages 1373 - 1376
KAUFMAN ET AL.: "Oncolytic viruses: a new class of immunotherapy drugs", NAT REV DRUG DISCOV, vol. 14, no. 9, 2015, pages 642 - 62, XP037065528, DOI: 10.1038/nrd4663
KIEM ET AL., BLOOD, vol. 83, 1994, pages 1467 - 1473
KIM ET AL.: "Gene therapy using plasmid DNA-encoded anti-HER2 antibody for cancers that overexpress HER2", CANCER GENE THER, vol. 23, no. 10, 2016, pages 341 - 347
KITAGUCHI ET AL.: "Immune deficiency enhances expression of recombinant human antibody in mice after nonviral in vivo gene transfer", INT J MOL MED, vol. 16, no. 4, 2005, pages 683 - 8, XP008118466
KLEINPETER ET AL.: "Vectorization in an oncolytic vaccinia virus of an antibody, a Fab and a scFv against programmed cell death-1 (PD-1) allows their intratumoral delivery and an improved tumor-growth inhibition", ONCOIMMUNOLOGY, vol. 5, no. 10, 2016, pages e1220467, XP055552525, DOI: 10.1080/2162402X.2016.1220467
KOHLER: "Immunoglobulin Chain Loss In Hybridoma Lines", PROC. NATL. ACAD. SCI. (U.S.A., vol. 77, 1980, pages 2197 - 2199
KOLDEJ, HUM GENE THER CLIN DEV, vol. 24, no. 2, 2013, pages 77 - 85
KOLLIAS ET AL., CELL, vol. 46, 1986, pages 485 - 94
KONTERMANN: "Strategies to extend plasma half-lives of recombinant antibodies", BIODRUGS, vol. 23, 2009, pages 93 - 109, XP008124089, DOI: 10.2165/00063030-200923020-00003
KOSA ET AL., J PHARM SCI, vol. 96, no. 11, 2007, pages 3117 - 24
KREISS ET AL., APPL MICBIOL BIOTECHNOL, vol. 49, no. 5, 1998, pages 560 - 7
KRUMLAUF ET AL., MOL. CELL. BIOL., vol. 5, 1985, pages 1639 - 48
KRUPA MESTEBAN M.: "The poxvirus vectors MVA and NYVAC as gene delivery systems for vaccination against infectious diseases and cancer", CURR GENE THER, vol. 8, no. 2, 2008, pages 97 - 120, XP009149369
KRYSAN ET AL., MOL. CELL. BIOL., vol. 9, 1989, pages 1026 - 1033
LAFACE ET AL., VIOLOGY, vol. 162, 1988, pages 483486
LEE ET AL., J CARDIOL, vol. 57, no. 1, 2011, pages 115 - 22
LEGENDRE ET AL.: "Delivery of plasmid DNA into mammalian cell lines using pH-sensitive liposomes: comparison with cationic liposomes", PHARM. RES., vol. 9, 1992, pages 1235 - 1242, XP000600707, DOI: 10.1023/A:1015836829670
LEVITT ET AL., GENES DEV, vol. 3, no. 7, 1989, pages 1019 - 25
LI ET AL., AGNEW CHEM INT ED ENL, vol. 54, no. 10, 2015, pages 2941 - 4
LI ET AL., J NEUROSCI METHODS, vol. 189, no. 1, 2010, pages 56 - 64
LICHTY ET AL.: "Going viral with cancer immunotherapy", NAT REV CANCER, vol. 14, no. 8, 2014, pages 559 - 67, XP055463372
LIU ET AL.: "Oncolytic adenoviruses for cancer gene therapy", METHODS MOL BIOL, vol. 433, 2008, pages 243 - 58
LOGAN ET AL.: "Adenovirus Tripartite Leader Sequence Enhances Translation Of mRNAs Late After Infection", PROC. NATL. ACAD. SCI. (U.S.A., vol. 81, 1984, pages 3655 - 3659
LUO ET AL., PHYS. PLASMA, vol. 5, 1998, pages 2868 - 2870
MACDONALD, HEPATOLOGY, vol. 7, 1987, pages 425 - 515
MAHAPATRO A ET AL., J NANOBIOTECHNOL, vol. 9, 2011, pages 55
MANN ET AL., CELL, vol. 33, 1983, pages 153 - 159
MASON ET AL., SCIENCE, vol. 234, 1986, pages 1372 - 8
MATHIESEN: "Electropermeabilization of skeletal muscle enhances gene transfer in vivo", GENE THER., vol. 6, 1999, pages 508 - 514, XP009003349, DOI: 10.1038/sj.gt.3300847
MCCARTY ET AL., J. VIROL., vol. 65, 1991, pages 2220 - 2224
MCLAUGHLIN ET AL., J. VIROL., vol. 62, 1988, pages 1963 - 1973
MCMAHON ET AL.: "Optimisation of electrotransfer of plasmid into skeletal muscle by pretreatment with hyaluronidase: increased expression with reduced muscle damage", GENE THER, vol. 8, no. 16, 2001, pages 1264 - 70, XP009005905, DOI: 10.1038/sj.gt.3301522
MERO ET AL.: "Conjugation of hyaluronan to proteins", CARB POLYMERS, vol. 92, 2013, pages 2163 - 70, XP028972870, DOI: 10.1016/j.carbpol.2012.11.090
MILLER ET AL., METH. ENZYMOL., vol. 217, 1993, pages 581 - 599
MIR ET AL.: "High-efficiency gene transfer into skeletal muscle mediated by electric pulses", PNAS, vol. 96, 1999, pages 4262 - 4267, XP002982026, DOI: 10.1073/pnas.96.8.4262
MOGRAM ET AL., NATURE, vol. 313, 1985, pages 812 - 815
MOL CELL BIOL, vol. 15, no. 7, 1995, pages 3864 - 9
MORGAN ET AL.: "Human gene therapy", ANN. REV. BIOCHEM., vol. 62, 1993, pages 191 - 217, XP002031972, DOI: 10.1146/annurev.bi.62.070193.001203
MULLIGAN ET AL.: "Selection For Animal Cells That Express The Escherichia coli Gene Coding For Xanthine-Guanine Phosphoribosyltransferase", PROC. NATL. ACAD. SCI. (U.S.A., vol. 78, 1981, pages 2072 - 2076
MULLIGAN: "The Basic Science Of Gene Therapy", SCIENCE, vol. 260, 1993, pages 926 - 932, XP002925334, DOI: 10.1126/science.8493530
NEUMANN, E. ET AL., EMBO J., vol. 1, 1982, pages 841
NEVES ET AL.: "Antibody approaches to treat brain diseases", TRENDS BIOTECHNOL, vol. 34, no. 1, 2016, pages 36 - 48, XP055512549, DOI: 10.1016/j.tibtech.2015.10.005
NICOLAOU ET AL., ANGEW. CHEM INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186
NICOLASRUBINSTEIN: "Vectors: A survey of molecular cloning vectors and their uses", 1988, BUTTERWORTH, article "Mammalian expression vectors", pages: 467 - 492
NICOLAU ET AL., METHODS ENZYMOL., vol. 149, 1987, pages 157 - 176
NICOLAUSENE, BIOCHIM. BIOPHYS. ACTA, vol. 721, 1982, pages 185 - 190
NIIDOME ET AL.: "Gene therapy progress and prospects: nonviral vectors", GENE THER., vol. 9, 2002, pages 1647 - 1652, XP008101843, DOI: 10.1038/SJ.GT.3301923
NIWA ET AL., GENES DEV, vol. 4, no. 9, 1990, pages 1552 - 9
OBEID ET AL., SEMIN ONCOL., vol. 42, no. 4, August 2015 (2015-08-01), pages 549 - 561
O'CONNOR-SEMMES ET AL.: "GSK2374697, a novel albumin-binding domain antibody (albudAb), extends systemic exposure of extendin-4: first study in humans-PK/PD and safety", CLIN PHARMACOL THER, vol. 96, 2014, pages 704 - 12
O'HARE ET AL.: "Transformation Of Mouse Fibroblasts To Methotrexate Resistance By A Recombinant Plasmid Expressing A Prokaryotic Dihydrofolate Reductase", PROC. NATL. ACAD. SCI. (U.S.A., vol. 78, 1981, pages 1527 - 1531, XP055600445
OISHI ET AL.: "Smart polyion complex micelles for targeted intracellular delivery of PEGylated antisense oligonucleotides containing acid-labile linkages", CHEMBIOCHEM, vol. 6, 2005, pages 718 - 725, XP003010787, DOI: 10.1002/cbic.200400334
ORNITZ ET AL., COLD SPRING HARBOR SYMP. QUANT. BIOL., vol. 50, 1986, pages 399 - 409
PACE ET AL.: "Measuring the conformational stability of a protein", PROTEIN STRUCTURE: A PRACTICAL APPROACH, vol. 2, 1997, pages 299 - 321
PAN ET AL., SOM. CELL MOLEC. GENET., vol. 18, 1992, pages 163 - 177
PARDI ET AL.: "Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes", J CONTROL RELEASE, vol. 217, 2015, pages 345 - 51, XP055722698, DOI: 10.1016/j.jconrel.2015.08.007
PATERNA ET AL., GENE THER, vol. 7, no. 15, 2000, pages 1304 - 11
PEREZ ET AL.: "Regulatable systemic production of monoclonal antibodies by in vivo muscle electroporation", GENET VACCINES THER, vol. 2, no. 1, 2004, pages 2, XP021009747, DOI: 10.1186/1479-0556-2-2
PETERSONLEGERSKI, GENE, vol. 107, 1991, pages 279 - 284
PIIRSOO ET AL., EMBO J., vol. 15, 1996, pages 1
PINKERT ET AL., GENES AND DEVEL, vol. 1, 1987, pages 161 - 71
PROUDFOOT: "Expression And Amplification Of Engineered Mouse Dihydrofolate Reductase Minigenes", NATURE, vol. 322, 1986, pages 562 - 565
PUHLMANN M. ET AL., HUMAN GENE THERAPY, vol. 10, 1999, pages 649 - 657
READHEAD ET AL., CELL, vol. 48, 1987, pages 703 - 12
RIORDAN, J. R. ET AL., SCIENCE, vol. 245, 1989, pages 1066 - 1073
RIZZUTO ET AL.: "Efficient and regulated erythropoietin production by naked DNA injection and muscle electroporation", PNAS, vol. 96, 1999, pages 6417 - 6422, XP002168199, DOI: 10.1073/pnas.96.11.6417
RODRIGO ET AL.: "De novo automated design of small RNA circuits for engineering synthetic riboregulation in living cells", PNAS, vol. 109, 2012, pages 15271 - 15276, XP055114063, DOI: 10.1073/pnas.1203831109
ROMANO ET AL., STEM CELLS, vol. 18, 2000, pages 19 - 39
ROSENBERG ET AL., SCIENCE, vol. 242, 1988, pages 1575 - 1578
RUTHER ET AL.: "Easy Identification Of cDNA Clones", EMBO J, vol. 2, 1983, pages 1791 - 1794, XP055413083, DOI: 10.1002/j.1460-2075.1983.tb01659.x
SAHIN ET AL.: "mRNA-based therapeutics: developing a new class of drugs", NAT REV DRUG DISCOV, vol. 13, no. 10, 2014, pages 759 - 80, XP055544673, DOI: 10.1038/nrd4278
SALMONSGUNZBERG, HUMAN GENE THERAPY, vol. 4, 1993, pages 129 - 141
SAMULSKI ET AL., J. VIROL., vol. 63, 1989, pages 2374 - 2378
SAN ET AL.: "Safety and short-term toxicity of a novel cationic lipid formulation for human gene therapy", HUM. GENE THER., vol. 4, 1983, pages 781 - 788, XP000565712
SANTERRE ET AL.: "Expression Of Prokaryotic Genes For Hygromycin B And G418 Resistance As Dominant-Selection Markers In Mouse L Cells", GENE, vol. 30, 1984, pages 147 - 156, XP025696524, DOI: 10.1016/0378-1119(84)90115-X
SCHEK ET AL., MOL CELL BIOL, vol. 12, no. 12, 1992, pages 5386 - 93
SCHELLENBERGER ET AL.: "A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner", NAT BIOTECHNOL, vol. 27, 2009, pages 1186 - 90
SCHELLENBERGER ET AL.: "A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner", NAT BIOTECHNOL., vol. 27, no. 12, 2009, pages 1186 - 90
SCHLAPSCHY ET AL., PROT ENG DES SEL, vol. 20, 2007, pages 273 - 283
SCHLAPSCHY ET AL.: "Fusion of a recombinant antibody fragment with a homo-amino-acid polymer: effects on biophysical properties and prolonged plasma half-life", PROTEIN ENG DES SEL, vol. 20, 2007, pages 273 - 84, XP002498446, DOI: 10.1093/protein/gzm020
SCHLAPSCHY ET AL.: "PASYlation: a biological alternative to PEGylation for extending the plasma half-life of pharmaceutically active proteins", PROTEIN ENG DES SEL, vol. 26, no. 8, 2013, pages 489 - 501, XP055195431, DOI: 10.1093/protein/gzt023
SCHLAPSCHY: "PASylation: a biological alternative to PEGylation for extending the plasma half-life of pharmaceutically active proteins", PROTEIN ENG DES SEL, vol. 26, 2013, pages 489 - 501, XP055195431, DOI: 10.1093/protein/gzt023
SHACKLEFORD ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 85, 1998, pages 9655 - 9659
SHINKAWA ET AL., J. BIOL. CHEM., vol. 278, 2003, pages 3466 - 3473
SIMCIKOVA ET AL.: "Towards effective non-viral gene delivery vector", BIOTECHNOL GENET ENG REV, vol. 31, no. 1-2, 2015, pages 82 - 107
SOCKOLOSKY ET AL.: "Fusion of a short peptide that binds immunoglobulin G to a recombinant protein substantially increases its plasma half-life in mice", PLOS ONE, vol. 9, 2014, pages e102566, XP055490311, DOI: 10.1371/journal.pone.0102566
SOMIARI ET AL.: "Theory and in vivo application of electroporative gene delivery", MOL. THER., vol. 2, 2000, pages 178 - 187, XP002965078, DOI: 10.1006/mthe.2000.0124
SOMMERFELT ET AL., VIROL, vol. 176, 1990, pages 58 - 59
STADLER ET AL., NATURE MEDICINE, vol. 23, 2017, pages 815 - 817
SUN ET AL., NATURE GENETICS, vol. 8, 1994, pages 33
SZYBALSKA ET AL.: "Genetics Of Human Cess Line. IV. DNA-Mediated Heritable Transformation Of A Biochemical Trait", PROC. NATL. ACAD. SCI. (U.S.A., vol. 48, 1962, pages 2026 - 2034
TACHIBANA ET AL.: "Altered Reactivity Of Immunoglobulin Produced By Human-Human Hybridoma Cells Transfected By pSV.2-Neo Gene", CYTOTECHNOLOGY, vol. 6, no. 3, 1991, pages 219 - 226
TANOUE ET AL.: "Armed oncolytic adenovirus expressing PD-L1 mini-body enhances anti-tumor effects of chimeric antigen receptor T-cells in solid tumors", CANCER RES, vol. 77, no. 8, 2017, pages 2040 - 51
THERAPY CLINICAL TRIALS WORLDWIDE, 2017, Retrieved from the Internet <URL:abedia.com/wiley>
THIMMAPPAYA, B. ET AL., CELL, vol. 31, 1982, pages 543 - 551
TOLSTOSHEV: "Gene Therapy, Concepts, Current Trials And Future Directions", ANN. REV. PHARMACOL. TOXICOL., vol. 32, 1993, pages 573 - 596
TRANCHANT ET AL.: "Physicochemical optimisation of plasmid delivery by cationic lipids", J. GENE MED., vol. 6, 2004, pages S24 - S35, XP002521501, DOI: 10.1002/jgm.509
TUR-KASPA ET AL., MOL. CELL BIOL., vol. 6, 1986, pages 716 - 718
ULMER ET AL.: "Protective immunity by intramuscular injection of low doses of influenza virus DNA vaccines", VACCINE, vol. 12, 1994, pages 1541 - 1544, XP023937584, DOI: 10.1016/0264-410X(94)90081-7
USTAV ET AL., EMBO J, vol. 10, 1991, pages 4231 - 4329
USTAV ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 898 - 902
VAN HEEKE ET AL.: "Expression Of Human Asparagine Synthetase In Escherichia coli", J. BIOL. CHEM., vol. 24, 1989, pages 5503 - 5509, XP002438811
VILLA-KAMAROFF ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 75, 1978, pages 3727 - 3731
VOS ET AL., JOUR. CELL. BIOL., vol. 21A, 1995, pages 433
WAGNER ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 78, 1981, pages 1444 - 5
WALKER ET AL.: "fusion protein technologies for biopharmaceuticals: applications and challenges", 2013, WILEY, article "Transferrin fusion protein therapies: acetylcholine receptor-transferrin fusion protein as a model", pages: 325 - 323
WALSH ET AL., J. CLIN. INVEST., vol. 93, 1994, pages 1440 - 1448
WANG G. ET AL., J. CLIN. INVEST., vol. 104, no. 11, 1999, pages R55 - 62
WIGLER ET AL.: "Transfer Of Purified Herpes Virus Thymidine Kinase Gene To Cultured Mouse Cells", CELL, vol. 11, 1977, pages 223 - 232, XP023910928, DOI: 10.1016/0092-8674(77)90333-6
WIGLER ET AL.: "Transformation Of Mammalian Cells With An Amplfiable Dominant-Acting Gene", PROC. NATL. ACAD. SCI. (U.S.A., vol. 77, 1980, pages 3567 - 3570
WILSON, CLIN. EXP. IMMUNOL., vol. 107, 1997, pages 31 - 32
WOHLGE ET AL., GENE THERAPY, vol. 3, 1996, pages 503 - 101
WOLD ET AL.: "Adenovirus vectors for gene therapy, vaccination and cancer gene therapy", CURR GENE THER, vol. 13, no. 6, 2013, pages 421 - 33
WOLFF ET AL., PROC. NATL. ACAD. SCI. USA, vol. 86, 1989, pages 9011 - 9014
WOLFF ET AL.: "Direct gene transfer into mouse muscle in vivo", SCIENCE, vol. 247, 1990, pages 1465 - 8, XP000293057, DOI: 10.1126/science.1690918
WONG, T. K. ET AL., GENE, vol. 10, 1980, pages 87
XIA ET AL., STEM CELLS DEV, vol. 16, no. 1, February 2007 (2007-02-01), pages 167 - 76
XU ET AL.: "Mechanism of DNA release from cationic liposome/DNA complexes used in cell transfection", BIOCHEMISTRY, vol. 35, 1996, pages 5616 - 5623, XP008122590
YAMAMOTO ET AL.: "Isolation Of Transforming DNA: Cloning The Hamster Aprt Gene", CELL, vol. 22, 1980, pages 817 - 823
YAMAZAKI ET AL.: "Passive immune-prophylaxis against influenza virus infection by the expression of neutralizing anti-hemagglutinin monoclonal antibodies from plasmids", JPN J INFECT DIS, vol. 64, no. 1, 2011, pages 40 - 9
YAN ET AL., BIOORG. MED. CHEM LETT., vol. 18, 2008, pages 5631 - 4
YAN ET AL., GENE, vol. 506, no. 2, 2012, pages 289 - 94
YANG ET AL., PROC. NATL. ACAD. SCI., vol. 87, 1990, pages 9568 - 9572
YATES, PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 3806 - 3810
YEW ET AL., HUM GENE THER, vol. 8, no. 5, 1997, pages 575 - 84
YEW ET AL.: "Reduced inflammatory response to plasmid DNA vectors by elimination and inhibition of immunostimulatory CpG motifs", MOL THER, vol. 1, no. 3, 2000, pages 255 - 62, XP001078874, DOI: 10.1006/mthe.2000.0036
ZHANG ET AL., BLOOD, vol. 110, no. 5, 2007, pages 1448 - 57
ZHANG ET AL., HUM GENE THER, vol. 23, no. 5, 2012, pages 460 - 72
ZHANG ET AL.: "Efficient expression of naked DNA delivered intraarterially to limb muscles of nonhuman primates", HUM. GENE THER., vol. 12, 2001, pages 427 - 438, XP002969821, DOI: 10.1089/10430340150504046
ZHOU ET AL., EXP. HEMATOL. (NY, vol. 21, 1993, pages 928 - 933

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022090529A1 (en) * 2020-11-02 2022-05-05 Ares Trading S.A. Combination treatment of cancer
WO2022234003A1 (en) * 2021-05-07 2022-11-10 Avacta Life Sciences Limited Cd33 binding polypeptides with stefin a protein
WO2023057567A1 (en) * 2021-10-07 2023-04-13 Avacta Life Sciences Limited Pd-l1 binding affimers

Also Published As

Publication number Publication date
TW202128775A (en) 2021-08-01
WO2021074695A8 (en) 2021-07-22

Similar Documents

Publication Publication Date Title
AU2019253221B2 (en) PD-L1 binding affimers, and uses related thereto
KR20220032568A (en) FLT3L-FC fusion protein and methods of use
WO2021074695A1 (en) PD-L1 INHIBITOR - TGFβ INHIBITOR BISPECIFIC DRUG MOIETIES.
US20210047425A1 (en) Methods and compositions for promoting and potentiating t-cell mediated immune responses through adcc targeting of cd39 expressing cells
CA2892831A1 (en) Immunotherapy with binding agents
CN112839675A (en) Compositions and methods for preventing or reversing T cell failure through exonuclease inhibition and antibody-mediated target endocytosis
US20220265847A1 (en) Methods and compositions for treating non-small cell lung cancer
JP2023539871A (en) Methods and compositions for treating glioblastoma
JP2023542080A (en) Multifunctional molecules that bind to calreticulin and their uses
JP2023522972A (en) Formulations, Dosing Regimens, and Manufacturing Processes for Heterodimeric Fc Fusion Proteins
WO2023057946A1 (en) Serum half-life extended pd-l1 binding polypeptides
US20240052061A1 (en) Methods and compositions for potentiating antitumoral immune responses through targeting of ntpdase3
KR20230028242A (en) Multifunctional molecules that bind to T cell-associated cancer cells and their uses
WO2023218243A1 (en) Lag-3/pd-l1 binding fusion proteins
WO2022234003A1 (en) Cd33 binding polypeptides with stefin a protein
WO2023057567A1 (en) Pd-l1 binding affimers
KR20230107617A (en) Combination therapy using chimeric antigen receptor (CAR)-expressing cells

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: 20816580

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: 20816580

Country of ref document: EP

Kind code of ref document: A1