WO2023211873A2 - Pharmaceutical formulations of an anti-ilt4 antibody or antigen-binding fragment thererof and methods of use - Google Patents

Pharmaceutical formulations of an anti-ilt4 antibody or antigen-binding fragment thererof and methods of use Download PDF

Info

Publication number
WO2023211873A2
WO2023211873A2 PCT/US2023/019713 US2023019713W WO2023211873A2 WO 2023211873 A2 WO2023211873 A2 WO 2023211873A2 US 2023019713 W US2023019713 W US 2023019713W WO 2023211873 A2 WO2023211873 A2 WO 2023211873A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
formulation
antibody
cdr
antigen
Prior art date
Application number
PCT/US2023/019713
Other languages
French (fr)
Inventor
Valentyn Antochshuk
Preeti G. Desai
Original Assignee
Merck Sharp & Dohme Llc
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 Merck Sharp & Dohme Llc filed Critical Merck Sharp & Dohme Llc
Publication of WO2023211873A2 publication Critical patent/WO2023211873A2/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the invention relates to pharmaceutical formulations of an antibody or antigen-binding fragment thereof that binds to human interleukin-like transcript 4 (ILT4) and their use in treating various disorders (e.g., cancer).
  • ILT4 human interleukin-like transcript 4
  • HLA human leukocyte antigen
  • HLA-G can directly inhibit immune cell function through receptor binding and/or through trogocytosis and impairment of chemotaxis (Morandi et al. Cytokine Growth Factor Review. 2014, 25: 327-335 and Lin et al. Mol Med. 2015, 21 : 782-791).
  • High expression of HLA-G in multiple tumor types including for example, colorectal, pancreatic, endometrial, lung, breast, ovarian, and gastric cancer, is associated with advanced disease stage, tumor invasiveness, metastatic potential and an unfavorable prognosis (Lin et al. Mol Med. 2015, 21 : 782-791 and Loumange et al. Int J Cancer.
  • HLA-G binding to ILT4 can directly inhibit the function of monocytes, dendritic cells, and neutrophils, thus impairing the innate immune antitumor response.
  • HLA-G human monocyte-derived dendritic cells (DCs) and ILT4-transgenic mice, HLA-G was shown to induce the development of tolerogenic APCs with arrest maturation/activation of myeloid DCs, and the induction of tolerogenic DCs by HLA-G was through disrupting the MHC class II presentation pathway (Ristich et al. Eur J Immunol. 2005, 35: 1133-1142).
  • ILT4 blockade would fill this need and would differentiate from current T-cell-targeted antibodies (e.g., anti-PDl, anti-TIGIT) by relieving suppression of tolerogenic myeloid cells in the tumor microenvironment.
  • an anti-human ILT4 (anti-ILT4) antibody or antigen-binding fragment thereof comprising: (i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM of a buffer; (iii) about 6% to about 8% weight /volume (w/v) of a non-reducing sugar;
  • the anti-lLT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21), wherein the buffer is an
  • the buffer is a selected from the group consisting of L-histidine buffer, acetate buffer, and citrate buffer.
  • the buffer is L-histidine buffer.
  • the buffer is acetate buffer.
  • the buffer is citrate buffer.
  • the non-reducing sugar is sucrose.
  • the non-ionic surfactant is polysorbate 80 (PS-80) or polysorbate 20 (PS-20). In one embodiment, the non-ionic surfactant is PS-80. In another embodiment, the non-ionic surfactant is PS-20.
  • the anti-oxidant is L-methionine.
  • an anti-ILT4 antibody or antigen-binding fragment thereof comprising: (i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM L- histidine buffer; (iii) about 6% to about 8% (w/v) sucrose; (iv) about 0.01 % to about 0.10% (w/v) PS-80; and (v) about 1 mM to about 20 mM L-methionine, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR- Hl : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO:
  • GX1X2NRPS wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the formulation comprises about 8 mM to about 12 mM L- histidine buffer.
  • the formulation comprises about 5 mM to about 10 mM L- methionine.
  • the formulation comprises about 0.01 % to about 0.05% (w/v) PS-80.
  • the formulation comprises about 10 mg/mL to about 150 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof.
  • the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL, about 12.5 mg/mL, about 15 mg/mL, about 25 mg/mL, about 50 mg/mL, about 75 mg/mL, about 100 mg/mL, about 125 mg/mL, or about 150 mg/mL. In one embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL.
  • the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 12.5 mg/mL. In yet another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 15 mg/mL. In still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 25 mg/mL. In one embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 50 mg/mL. In another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 75 mg/mL. In yet another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 100 mg/mL.
  • the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 125 mg/mL. In yet still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 150 mg/mL. In specific embodiments, the anti-ILT4 antibody or antigen-binding fragment thereof is a monoclonal antibody.
  • the formulation comprises about 25 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation comprises about 50 mg/mL of the anti- ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation comprises about 75 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation comprises about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation comprises about 125 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation has a pH range of from about 5.0 to about 6.8. In some embodiments, the formulation has a pH range of from about 5.5 to about 6.0. In other embodiments, the formulation has a pH of about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, or about 6.0. In one embodiment, the formulation has a pH of about 5.5. In another embodiment, the formulation has a pH of about 5.6. In yet another embodiment, the formulation has a pH of about 5.7. In still another embodiment, the formulation has a pH of about 5.8. In another embodiment, the formulation has a pH of about 5.9. In yet still another embodiment, the formulation has a pH of about 6.0.
  • the pharmaceutical formulation for an anti-ILT4 antibody or antigen-binding fragment thereof comprises: (i) about 50 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer, pH about 5.5; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) polysorbate 80; and (v) about 10 mM L-methionine, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSN
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein Xi is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GDSNRPS(SEQ ID NO: 52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:2 and a light chain comprising the amino acid sequence set forth in SEQ ID NOY.
  • the anti-lLT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 80 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 7.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:58.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO:2 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
  • the anti-ILT4 antibody or antigen binding fragment thereof is a monoclonal antibody comprising two heavy chains comprising the amino acid sequence set forth in SEQ ID NO:2 and two light chains comprising the amino acid sequence set forth in SEQ ID NO:7.
  • the anti-ILT4 antibody or antigen binding fragment thereof is a monoclonal antibody comprising two heavy chains consisting of the amino acid sequence set forth in SEQ ID NO:2 and two light chains consisting of the amino acid sequence set forth in SEQ ID NO:7.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO: 80 and a light chain consisting of the ammo acid sequence set forth in SEQ ID NO:7.
  • the anti-ILT4 antibody or antigen binding fragment thereof is a monoclonal antibody comprising two heavy chains comprising the amino acid sequence set forth in SEQ ID NO: 80 and two light chains comprising the amino acid sequence set forth in SEQ ID NO:7.
  • the anti-ILT4 antibody or antigen binding fragment thereof is a monoclonal antibody comprising two heavy chains consisting of the amino acid sequence set forth in SEQ ID NO: 80 and two light chains consisting of the amino acid sequence set forth in SEQ ID NO:7.
  • the formulation is a liquid formulation. In one embodiment, the formulation is a frozen formulation. In another embodiment, the liquid formulation is stored at about 5°C. In yet another embodiment, the formulation is stored frozen at about -20°C or below. In yet another embodiment, the formulation is stored frozen at about -70°C or below. In still another embodiment, the liquid formulation is a reconstituted solution from a lyophilized formulation.
  • the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultra-performance size exclusion chromatography; (ii) the turbidity of the formulation is at most about 0.135 as measured by OD350-500; (iii) the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, and/or the basic variant of the anti-ILT4 antibody or antigenbinding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography; (iv) the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging; and
  • the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultra-performance size exclusion chromatography.
  • the turbidity of the formulation is at most about 0.135 as measured by OD350-500.
  • the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, as determined by high performance ion-exchange chromatography. In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, as determined by high performance ion-exchange chromatography.
  • the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography.
  • the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%
  • the % acidic variant of the anti-lLT4 antibody or antigen-binding fragment thereof is at most about 23%
  • the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography.
  • the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging.
  • the % oxidation of one or more amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the amino acid residue is W7 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80.
  • the amino acid residue is W102 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80.
  • the amino acid residue is M253 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80. In still another embodiment, the amino acid residue is M359 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80. In yet still another embodiment, the amino acid residue is M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80.
  • the % oxidation of one amino acid residue selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the % oxidation of two amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the % oxidation of three amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the % oxidation of four ammo acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the % oxidation of all five amino acid residues from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • provided herein is a method of treating a cancer in a human patient in need thereof, comprising administering a therapeutically effective amount of the pharmaceutical formulation described herein.
  • FIGS. 1A-1I show early stability studies of various anti-ILT4 formulations, including turbidity (OD350-500) (FIGS. 1A-1C), % HMW (high molecular weight) (FIGS. 1D-1F), or % monomer (FIGS. 1G-1I) at 5°C (FIGS. 1A, ID, 1G), 25°C (FIGS. IB, IE, 1H), or 40°C (FIGS 1C, IF, II) storage conditions, as described in Example 4.
  • FIGS. 2A-2I show early stability studies of various anti-ILT4 formulations, including % acidic variants (FIGS. 2A-2C), % main peak (FIGS. 2D-2F), or % basic variants (FIGS. 2G-2I) at 5°C (FIGS. 2A, 2D, 2G), 25°C (FIGS. 2B, 2E, 2H), or 40°C (FIGS. 2C, 2F, 21) storage conditions, as described in Example 4.
  • FIGS. 3A-3I show pH ranging stability studies of various anti-ILT4 formulations, including turbidity (OD350-500) (FIGS. 3A-3C), % HMW (FIGS. 3D-3F), or % monomer (FIGS. 3G-31) at 5°C (FIGS. 3A, 3D, 3G), 25°C (FIGS. 3B, 3E, 3H), or 40°C (FIGS. 3C, 3F, 31) storage conditions, as described in Example 5.
  • turbidity OD350-500
  • % HMW FIGS. 3D-3F
  • % monomer % monomer
  • FIGS. 4A-4I show pH ranging stability studies of various anti-ILT4 formulations, including % acidic variants (FIGS. 4A-4C), % main peak (FIGS. 4D-4F), or % basic variants (FIGS. 4G-4I) at 5°C (FIGS. 4A, 4D, 4G), 25°C (FIGS. 4B, 4E, 4H), or 40°C (FIGS. 4C, 4F, 41) storage conditions, as described in Example 5.
  • % acidic variants % main peak
  • FIGS. 4G-4I % basic variants at 5°C (FIGS. 4A, 4D, 4G), 25°C (FIGS. 4B, 4E, 4H), or 40°C (FIGS. 4C, 4F, 41) storage conditions, as described in Example 5.
  • FIGS. 5A-5G show surfactant ranging stability studies of various anti-ILT4 formulations, including turbidity (OD350-500) (FIG. 5A), % HMW (FIG. 5B), % monomer (FIG. 5C), subvisible particle counts before initiation of agitation (FIG. 5D), after 3 days agitation (FIG. 5E), after 7 days agitation (FIG. 5F), or after 7 days ambient control (FIG. 5G), as described in Example 6.
  • FIGS. 6A-6K show light stress stability studies of various anti-lLT4 formulations, including turbidity (OD350-500) (FIG. 6A), % HMW (FIG.
  • FIG 7 shows the initial and 3 days agitation results from a surfactant ranging study, as described in Example 6.
  • FIG 8 shows the 7 days agitation and 7 day ambient control results from a surfactant ranging study, as described in Example 6.
  • FIG 9 shows results from a light stress study with antioxidant L-methionine, as described in Example 7.
  • PS20 (or PS-20) polysorbate 20
  • PS80 (or PS-80) polysorbate 80
  • VL immunoglobulin light chain variable region v/v volume per volume
  • “acidic variant” refers to the anti-ILT4 antibody that is more acidic (e.g., as determined by cation exchange chromatography) than the anti-ILT4 antibody main species. Such acidic variations are detected by various chromatography purification methods for separating molecule variants by charge, such as ion exchange, for example, cation exchange chromatography or WCX-10 HPLC (a weak cation exchange chromatography), optionally followed by mass spectroscopy.
  • the acidic variant has a lower isoelectric point (pl) than the main species, and can have a more acidic character due to for example, methionine oxidation, sialylation of asparagine residues or deamidated variants of the antibody, or a combination thereof.
  • the anti-ILT4 antibody acidic variants are the anti-ILT4 antibody species as identified by the acidic variants peak(s) in Figure 6D, and eluted according to the cation ion exchange method described in Example 10.
  • the “% acidic variants species” refers to the total area of acidic variants peak(s) divided by the total area of all peaks in the elution chromatogram.
  • the anti-ILT4 antibody acidic variant is as identified by peak(s) eluted prior to the main peak according to a cation ion exchange method. In another embodiment, the anti-ILT4 antibody acidic variant is as identified by peak(s) eluted prior to the main peak according to a weak cation ion exchange method. In an ion exchange chromatography method, the “% acidic variant” refers to the total area of acidic species peaks divided by the total area of all peaks in the elution chromatogram.
  • Binding affinity refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen).
  • binding affinity refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including KinExA and Biacore.
  • antibody includes, but is not limited to, monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), fully human antibodies, and chimeric antibodies.
  • antigen-binding fragment refers to antigen-binding fragments of antibodies, i. e. , antibody fragments that retain the ability to bind to the antigen bound by the full-length antibody, e.g., fragments that retain one or more CDR regions.
  • antibody binding fragments include, but are not limited to. Fab, Fab’, F(ab’)2, Fv fragments and individual antibody heavy chains or light chains, and individual heavy chain or light chain variable regions.
  • basic variant refers to the anti-ILT4 antibody that is more basic (e.g., as determined by cation exchange chromatography) than the anti-ILT4 antibody main species.
  • Such basic variants are detected by various chromatography purification methods for separating molecule variants by charge, such as ion exchange, for example, cation exchange chromatography (e.g., the method described in Example 10 or WCX-10 HPLC (a weak cation exchange chromatography), optionally followed by mass spectroscopy.
  • cation exchange chromatography e.g., the method described in Example 10 or WCX-10 HPLC (a weak cation exchange chromatography
  • mass spectroscopy e.g., the basic species has a higher pH than the main species, and can have a more basic character due to modifications or differences from the main species.
  • the anti-ILT4 antibody basic species is as identified by peak(s) eluted after the mam peak according to a cation ion exchange method. In another embodiment, the anti-ILT4 antibody basic species is as identified by peak(s) eluted after the main peak according to a weak cation ion exchange method. In an ion exchange chromatography method, the “% basic species” refers to the total area of basic species peak(s) divided by the total area of all peaks in the elution chromatogram.
  • a “Fab fragment” is comprised of one light chain and the CHI and variable regions of one heavy chain.
  • the heavy chain of a Fab molecule cannot form a disulfide bond with another heavy chain molecule.
  • An “Fab fragment” can be the product of papain cleavage of an antibody.
  • An “Fc” region contains two heavy chain fragments comprising the CHI and CH2 domains of an antibody.
  • the two heavy chain fragments are held together by two or more disulfide bonds and by hydrophobic interactions of the CH3 domains.
  • a “Fab’ fragment” contains one light chain and a portion or fragment of one heavy chain that contains the VH domain and the CHI domain and also the region between the CHI and CH2 domains, such that an interchain disulfide bond can be formed between the two heavy chains of two Fab’ fragments to form a F(ab’)2molecule.
  • a “F(ab’)2 fragment” contains two light chains and two heavy chains containing a portion of the constant region between the CHI and CH2 domains, such that an interchain disulfide bond is formed between the two heavy chains.
  • a F(ab’)2 fragment thus is composed of two Fab’ fragments that are held together by a disulfide bond between the two heavy chains.
  • An “F(ab’)2 fragment” can be the product of pepsin cleavage of an antibody.
  • the “Fv region” comprises the variable regions from both the heavy and light chains, but lacks the constant regions.
  • isolated antibody refers to the purification status and in such context means the molecule is substantially free of other biological molecules such as nucleic acids, proteins, lipids, carbohydrates, or other material such as cellular debris and growth media. Generally, the term “isolated” is not intended to refer to a complete absence of such material or to an absence of water, buffers, or salts, unless they are present in amounts that substantially interfere with experimental or therapeutic use of the binding compound as described herein.
  • main species refers to the anti-ILT4 antibody species identified as the majority of the antibody species in a mixture with one or more acidic or basic species thereof. Such main species are detected by various chromatography purification methods for separating molecule variants by charge, such as ion exchange, for example, cation exchange chromatography (e.g., the method described in Example 10) or WCX-10 HPLC (a weak cation exchange chromatography), optionally followed by mass spectroscopy.
  • the mixture can be a result of for example, antibody preparations from mammalian cells and post-translational modifications thereof, upstream and downstream processing, or storage.
  • the main species is identified as the main peak according to a cation ion exchange method.
  • the “% main” refers to the total area of main peak divided by the total area of all peaks in the elution chromatogram.
  • a Thermo Scientific ProPac WCX-10 column is used for the cation ion exchange method.
  • a Thermo Scientific ProPac WCX-10 column is used, with a Mobile Phase (A) 24 mM MES pH 6.
  • a non-linear gradient is used with: 22%-22%B for 0-0.6 min; 22%- 29%B for 0.6-15.0 min; 29%-70%B for 15.0-30.0 min; 70%-100%B for 30.0-30.5 min; and 100%-100%B from 30.5-33.0 min.
  • the cation ion exchange method is described in Example 10.
  • the term “monoclonal antibody”, as used herein, refers to a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts.
  • conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains that are often specific for different epitopes.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the invention may be made by the hybridoma method first described by Kohler et al. (1975) Nature 256: 495, or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol. Biol. 222: 581-597, for example. See also Presta (2005) J. Allergy Clin. Immunol. 116:731.
  • Fully human antibody refers to an antibody that comprises human immunoglobulin protein sequences only.
  • a fully human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell.
  • mouse antibody refers to an antibody that comprises mouse immunoglobulin sequences only.
  • a fully human antibody may contain rat carbohydrate chains if produced in a rat, in a rat cell, or in a hybndoma derived from a rat cell.
  • rat antibody refers to an antibody that comprises rat immunoglobulin sequences only.
  • the basic “antibody” structural unit comprises a tetramer.
  • each tetramer includes two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a “variable region” or “variable domain” of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy -terminal portion of the heavy chain may define a constant region primarily responsible for effector function.
  • human constant light chains are classified as kappa and lambda light chains.
  • human constant heavy chains are typically classified as mu, delta, gamma, alpha, or epsilon, and define the antibody’s isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • Subtypes of these IgG include, for example, IgGl and IgG4.
  • the invention includes anti-ILT4 antibodies and antigen-binding fragments comprising any of these light and/or heavy constant chains.
  • variable region means the segment of IgG chains which is variable in sequence between different antibodies.
  • a “variable region” of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination.
  • the variable region of the heavy chain may be referred to as “VH.”
  • the variable region of the light chain may be referred to as “VL.”
  • the variable regions of both the heavy and light chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), which are located within relatively conserved framework regions (FR). The CDRs are usually aligned by the framework regions, enabling binding to a specific epitope.
  • CDRs complementarity determining regions
  • both light and heavy chains variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • the assignment of amino acids to each domain is, generally, in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al:, National Institutes of Health, Bethesda, Md.; 5th ed.; NIH Publ. No. 91-3242 (1991); Kabat (1978) Adv. Prot. Chem. 32: 1-75; Kabat, etal., (1977) J. Biol. Chem. 252:6609-6616; Chothia, etal., (1987) J Mol. Biol. 196:901-917 or Chothia, et al., (1989) Nature 342:878-883.
  • a “CDR” refers to one of three hypervariable regions (Hl, H2, or H3) within the nonframework region of the antibody VH P-sheet framework, or one of three hypervariable regions (LI, L2, or L3) within the non-framework region of the antibody VL P-sheet framework. Accordingly, CDRs are variable region sequences interspersed within the framework region sequences. CDR regions are well known to those skilled in the art and have been defined by, for example, Kabat as the regions of most hypervariability within the antibody variable domains. CDR region sequences also have been defined structurally by Chothia as those residues that are not part of the conserved -sheet framework, and thus are able to adapt to different conformation. Both terminologies are well recognized in the art.
  • CDR region sequences have also been defined by AbM, Contact, and IMGT.
  • the positions of CDRs within a canonical antibody variable region have been determined by comparison of numerous structures (Al-Lazikani et al., 1997, J. Mol. Biol. 273:927-48; Morea et al., 2000, Methods 20:267-79). Because the number of residues within a hypervariable region varies in different antibodies, additional residues relative to the canonical positions are conventionally numbered with a, b, c and so forth next to the residue number in the canonical variable region numbering scheme (Al-Lazikani et al., supra). Such nomenclature is similarly well known to those skilled in the art.
  • the CDRs are as defined by the Kabat numbering system. In other embodiments, the CDRs are as defined by the IMGT numbering system. In yet other embodiments, the CDRs are as defined by the AbM numbering system. In still other embodiments, the CDRs are as defined by the Chothia numbering system. In yet other embodiments, the CDRs are as defined by the Contact numbering system.
  • Sequence identity refers to the degree to which the amino acids of two polypeptides are the same at equivalent positions when the two sequences are optimally aligned
  • Sequence similarity includes identical residues and non-identical, biochemically related amino acids. Biochemically related amino acids that share similar properties and may be interchangeable are discussed above.
  • Constantly modified variants or “conservative substitution” refers to substitutions of amino acids in a protein with other amino acids having similar characteristics (e.g., charge, side-chain size, hydrophobicity /hydrophilicity, backbone conformation and rigidity, etc.), such that the changes can frequently be made without altering the biological activity of the protein.
  • Those of skill in this art recognize that, in general, single amino acid substitutions in non- essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)).
  • substitutions of structurally or functionally similar amino acids are less likely to disrupt biological activity. Exemplary conservative substitutions are set forth in Table 2.
  • epitope refers to an area or region on an antigen to which an antibody or antigen-binding fragment binds. Binding of an antibody or antigen-binding fragment thereof disclosed herein to an epitope means that the antibody or antigen-binding fragment thereof binds to one or more amino acid residues within the epitope.
  • Treat” or “treating” a cancer or an infectious condition as used herein means to administer a formulation of the invention to a subject having an immune condition or cancerous condition, or diagnosed with a cancer or pathogenic infection (e.g., viral, bacterial, fungal), to achieve at least one positive therapeutic effect, such as for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral organs, or reduced rate of tumor metastasis or tumor growth.
  • a cancer or pathogenic infection e.g., viral, bacterial, fungal
  • Treatment may include one or more of the following: inducing/increasing an antitumor immune response, stimulating an immune response to a pathogen, toxin, and/or self-antigen, stimulating an immune response to a viral infection, decreasing the number of one or more tumor markers, inhibiting the growth or survival of tumor cells, eliminating or reducing the size of one or more cancerous lesions or tumors, decreasing the level of one or more tumor markers, ameliorating, reducing the severity or duration of cancer, prolonging the survival of a patient relative to the expected survival in a similar untreated patient.
  • cancer cancer
  • cancer cancerous
  • malignant refers to or describes the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • cancer examples include, but are not limited to, carcinoma, lymphoma, leukemia, blastoma, and sarcoma. More particular examples of such cancers include squamous cell carcinoma, myeloma, small-cell lung cancer, non-small cell lung cancer, glioma, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, gastrointestinal (tract) cancer, renal cancer, ovarian cancer, liver cancer, lymphoblastic leukemia, lymphocytic leukemia, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, melanoma, chondrosarcoma, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, brain cancer, stomach cancer, bladder cancer, hepatoma, breast cancer, colon carcinoma, and head and neck cancer.
  • patient refers to a mammal (e.g., rat, mouse, dog, cat, rabbit) capable of being treated with the formulations of the invention, most preferably a human.
  • the patient is an adult patient.
  • the patient is a pediatnc patient.
  • a patient “in need of treatment” is an individual diagnosed with, suspected of having, or predisposed to a disease or disorder in which a formulation of the invention is intended to treat or ameliorate (e.g., an ILT4-related disease such as cancer), or a patient for whom prevention of such a disorder is desired.
  • terapéuticaally effective amount means an amount whereby sufficient therapeutic composition or formulation is introduced to a patient to treat a diseased or condition.
  • this level may vary according to the patient’s characteristics such as age, weight, etc.
  • the term “about”, when modifying the quantity (e.g., mM, or M) of a substance or composition, the percentage (v/v or w/v) of a formulation component, the pH of a solution/formulation, or the value of a parameter characterizing a step in a method, or the like refers to variation in the numerical quantity that can occur, for example, through typical measuring, handling, and sampling procedures involved in the preparation, characterization and/or use of the substance or composition; through instrumental error in these procedures; through differences in the manufacture, source, or purity of the ingredients employed to make or use the compositions or carry out the procedures; and the like.
  • “about” can mean a variation of ⁇ 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, or 10%.
  • x% (w/v) is equivalent to x g/100 ml (for example 5% w/v equals 50 mg/ml).
  • a binding compound that consists essentially of a recited amino acid sequence may also include one or more amino acids, including substitutions of one or more amino acid residues, that do not materially affect the properties of the binding compound.
  • “Comprising” or variations such as “comprise” or “comprises” are used throughout the specification and claims in an inclusive sense, i.e., to specify the presence of the stated features but not to preclude the presence or addition of further features that may materially enhance the operation or utility of any of the embodiments of the invention, unless the context requires otherwise due to express language or necessary implication.
  • the term “buffer” encompasses those agents which maintain the solution pH of the formulations in an acceptable range, or, for lyophilized formulations of the invention, provide an acceptable solution pH prior to lyophilization.
  • lyophilization refers to a process by which the material to be dried is first frozen and then the ice or frozen solvent is removed by sublimation in a vacuum environment. Excipients may be included in pre-lyophilized formulations to enhance stability of the lyophilized product upon storage.
  • composition refers to preparations which are in such a form as to permit the active ingredients to be effective, and which contains no additional components which are toxic to the subjects to which the formulation would be administered.
  • formulation and “pharmaceutical formulation” are used interchangeably throughout.
  • “Pharmaceutically acceptable” refers to excipients (vehicles, additives) and compositions that can reasonably be administered to a subject to provide an effective dose of the active ingredient employed and that are “generally regarded as safe,” e.g., that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction, such as gastric upset and the like, when administered to a human.
  • this tenn refers to molecular entities and compositions approved by a regulatory agency of the federal or a state government or listed in the U.S. Pharmacopeia or another generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • a “reconstituted” formulation is one that has been prepared by dissolving a lyophilized protein formulation in a diluent such that the protein is dispersed in the reconstituted formulation.
  • the reconstituted formulation is suitable for administration, e.g., parenteral administration), and may optionally be suitable for subcutaneous administration.
  • a “stable” formulation is one in which the protein therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage or under stress conditions.
  • Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs. (1991) and Jones, A. Adv. Drug Delivery Rev. 10:29-90 (1993).
  • Stability can be measured at a selected temperature for a selected time period.
  • a stable formulation is a formulation with no significant changes observed at a refrigerated temperature (2-8° C) for at least 6 months.
  • a stable formulation is a formulation with no significant changes observed at a refrigerated temperature (2-8° C) for at least 12 months. In another embodiment, a stable formulation is a formulation with no significant changes observed at a refrigerated temperature (2-8° C) for at least 24 months. In another embodiment, stable formulation is a formulation with no significant changes observed at room temperature (23-27°C) for at least 3 months. In another embodiment, stable formulation is a formulation with no significant changes observed at room temperature (23-27°C) for at least 6 months. In another embodiment, stable formulation is a formulation with no significant changes observed at room temperature (23-27°C) for at least 12 months.
  • stable formulation is a formulation with no significant changes observed at room temperature (23-27°C) for at least 18 months.
  • the criteria for stability for an antibody formulation are as follows. Typically, no more than 10%, preferably 5%, of antibody monomer is degraded as measured by SEC-HPLC. Typically, the formulation is colorless, or clear to slightly opalescent by visual analysis. Typically, the concentration, pH, and osmolality of the formulation have no more than +/-10% change. Potency is typically within 60-140%, preferably 80-120% of the control or reference. Typically, no more than 10%, preferably 5% of clipping of the antibody is observed, i.e., % low molecular weight species as determined, for example, by HP-SEC. Typically, no more than 10%, preferably no more than 5% of aggregation of the antibody is observed, i.e., % high molecular weight species as determined, for example, by HP-SEC.
  • An antibody “retains its physical stability” in a pharmaceutical fomrulation, if it shows no significant increase of aggregation, precipitation, and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering, size exclusion chromatography (SEC) and dynamic light scattering.
  • SEC size exclusion chromatography
  • the changes of protein conformation can be evaluated by fluorescence spectroscopy, which determines the protein tertiary structure, and by FTIR spectroscopy, which determines the protein secondary structure.
  • An antibody “retains its chemical stability” in a pharmaceutical formulation, if it shows no significant chemical alteration. Chemical stability can be assessed by detecting and quantifying chemically altered forms of the protein. Degradation processes that often alter the protein chemical structure include hydrolysis or clipping (evaluated by methods such as size exclusion chromatography and SDS-PAGE), oxidation (evaluated by methods such as by peptide mapping in conjunction with mass spectroscopy or MALDI/TOF/MS), deamidation (evaluated by methods such as ion-exchange chromatography, capillary isoelectric focusing, peptide mapping, isoaspartic acid measurement), and isomerization (evaluated by measuring the isoaspartic acid content, peptide mapping, etc.).
  • An antibody “retains its biological activity” in a pharmaceutical formulation, if the biological activity of the antibody at a given time is within a predetermined range of the biological activity exhibited at the time the pharmaceutical formulation was prepared.
  • the biological activity of an antibody can be determined, for example, by an antigen binding assay.
  • the term “isotonic” means that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 270-328 mOsm. Slightly hypotonic pressure is 250-269 and slightly hypertonic pressure is 328-350 mOsm. Osmotic pressure can be measured, for example, using a vapor pressure or ice-freezing type osmometer.
  • the formulations disclosed herein can be used with any antibodies and antigen-binding fragments thereof (e.g., fully human antibodies) that bind to ILT4.
  • the anti- ILT4 antibody or antigen-binding fragment thereof is an antagonist.
  • the anti-ILT4 antibody or antigen-binding fragment thereof is an anti-ILT4 antibody or antigenbinding fragment thereof described herein (e.g., 1E1, 2A6, 3G7, or 2C1).
  • formulations can be used with anti-ILT4 antibodies and antigenbinding fragments thereof as set forth herein having one or more of the properties set forth below:
  • ILT4 extracellular domain or TM form of ILT4 expressed on a cell surface e.g., a pre-B cell, Chinese hamster ovary cell, U937 cell, or Jurkat JE6 cell.
  • thermogram • is characterized by a thermogram having Tm onset > 60°C, Tml- 65.2°C and Tm2 ⁇ 78.8°C;
  • HLA-G e.g., Fc fused HLA-G
  • human ILT4 e.g., ILT4 on mouse 3A9 T cells transfected with and expressing ILT4
  • IC50 0.25 micrograms/ml (+0.06 micrograms/ml), e.g., as determined by surface plasmon resonance;
  • HLA-B e.g., fluorochrome labeled dexamers of HL A- A, such as HLA*A2:01 or HLA-B such as HLA*B7:02
  • HLA-F e.g, fluorochrome labeled tetramers of HLA-F
  • ILT4 e.g, ILT4 on mouse 3A9 T cells transfected with and expressing ILT4
  • surface plasmon resonance e.g., as determined by surface plasmon resonance
  • ILT4 e.g., ILT4 on mouse 3A9 T cells transfected with and expressing ILT4
  • ANGPTL1, ANGPTL4, and/or ANGPTL7 e.g, biotinylated ANGPTL proteins, e.g, as determined by surface plasmon resonance
  • LPS lipopolysaccharide
  • PBMC peripheral blood mononuclear cell
  • mice e.g., Immuno-deficient NSG mice
  • human hematopoietic stem cells for example, which harbor peripheral human CD45+ immune cells, for example, wherein the tumor is a human skin melanoma tumor such as from the cell line SKMEL5;
  • Antibody 1E1 Q1E, S54A heavy chain (IgG4)
  • Antibody 1E1 heavy chain (IgGl)
  • CDR-H2 EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17) (e.g.,
  • CDR-H3 LPTRWVTTRYFDL (SEQ ID NO: 18)
  • Antibody 1E1 (Q1E, S54A) light chain (lambda)
  • Antibody 1E1 (Q1E, N53Q) light chain (lambda)
  • Antibody 1E1 (Q1E, N53E) light chain (lambda)
  • Antibody 1E1 light chain (lambda)
  • GX1X2NRPS wherein Xi is N,Q,E or D and X2 is S or A (SEQ ID NO: 20) (e.g., GNSNRPS (SEQ ID NO:49), GQSNRPS (SEQ ID NO:50), GESNRPS (SEQ ID NO:51), GDSNRPS (SEQ ID NO:52), GNANRPS (SEQ ID NO: 53), GQANRPS (SEQ ID NO:54), GEANRPS (SEQ ID NO:55), or GDANRPS (SEQ ID NO:56))
  • GNSNRPS SEQ ID NO:49
  • GQSNRPS SEQ ID NO:50
  • GESNRPS SEQ ID NO:51
  • GDSNRPS SEQ ID NO:52
  • GNANRPS SEQ ID NO: 53
  • GQANRPS SEQ ID NO:54
  • GEANRPS SEQ ID NO:55
  • GDANRPS SEQ ID NO:56
  • Antibodies and antigen-binding fragments thereof including the 1E1 heavy and light chain CDRs or the 1E1 VH and VL or the 1E1 heavy chain and light chain may be referred to as “1E1.”
  • Antibody 2A6 Q1E heavy' chain (IgG4) Heavy chain
  • Antibody 2A6 Q1E, S102A, M119L heavy chain (IgG4)
  • Antibody 2A6 (Q1E, D101S, M119L) heavy chain (IgG4) Heavy chain EVQLVOSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMGGIIPIFGTAN YAOKFOGRVTITADESTSTAYMELSSLRSEDTAVYYCARYFSSSGWYKGGAFDIWGQG TLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF PAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPE FLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPR EEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTL PPSQEEMTKNQVSLTCLVKGFYPS
  • compositions disclosed herein can be used with antibodies comprising SEQ ID NO: 8, 9, 10, 64, 65, or 66, and antigen-binding fragments thereof, wherein residue 1 of SEQ ID NO: 8, 9, 10, 64, 65, or 66 is Q instead of E.
  • CDR-H1 SYAIS (SEQ ID NO:22)
  • CDR-H2 GIIPIFGTANYAQKFQG (SEQ ID NO:23)
  • CDR-H3 YFX1X2SGWYKGGAFDI; wherein Xi is D or S and X2 is S or A (SEQ ID NO:24) (e.g., YFDSSGWYKGGAFD1 (SEQ ID NO:91), YFSSSGWYKGGAFD1 (SEQ ID NO:92), YFDASGWYKGGAFDI (SEQ ID NO:93), or YFSASGWYKGGAFDI (SEQ ID NO:94)) [0119] Antibody 2A6 light chain (lambda)
  • compositions disclosed herein can be used with antibodies comprising SEQ ID NO:
  • CDR-L1 TLRSGINVDTYRIH (SEQ ID NO:25)
  • CDR-L2 YKSDSDKHQGS (SEQ ID NO:26)
  • CDR-L3 AIWYSSTWV (SEQ ID NO: 27)
  • Antibodies and antigen-binding fragments thereof including the 2A6 heavy and light chain CDRs or the 2A6 VH and VL or the 2A6 heavy chain and light chain may be referred to as “2A6.”
  • Antibody 3G7 (Q1E) heavy' chain (IgG4) [0123] Antibody 3G7 (Q1E) heavy' chain (IgG4)
  • compositions disclosed herein can be used with antibodies comprising SEQ ID NO:
  • CDR-H1 SYAMH (SEQ ID NO:28)
  • CDR-H2 V1SYDGSNKYYADSVKG (SEQ ID NO:29)
  • DIOMTQSP S S VS AS VGDRVTITCRAS OGIS S WL AWYQOKPGKAPKFLIYAAS SLOS GVP S KFSGSGSGTDFTLTISSLQPEDFATYYCOOYNSYPPTFGGGTKVEIKRtVAAPSVFTFPPSD EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLTL SKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
  • CDR-L2 AASSLQS (SEQ ID NO:32)
  • Antibodies and antigen-binding fragments thereof including the 3G7 heavy and light chain CDRs or the 3G7 VH and VL or the 3G7 heavy chain and light chain may be referred to as “3G7.”
  • compositions disclosed herein can be used with antibodies comprising SEQ ID NO:
  • CDR-H2 GFDPEDGETIYAQKFQG (SEQ ID NO 35)
  • CDR-H3 AGPLYTIFGVVIIPDNWFDP (SEQ ID NO:36)
  • Antibody 2C1 light chain (Q1E) (lambda)
  • compositions disclosed herein can be used with antibodies comprising SEQ ID NO:
  • CDR-L1 TGSSSNIGAGYDVH (SEQ ID NO:37)
  • CDR-L2 GNSNRPS (SEQ ID NO:38)
  • CDR-L3 QSYDSSLSGSGVV (SEQ ID NO:39)
  • Antibodies and antigen-binding fragments thereof including the 2C1 heavy and light chain CDRs or the 2C1 VH and VL or the 2C1 heavy chain and light chain may be referred to as “2C1.”
  • a C- terminal lysine of a heavy chain immunoglobulin is absent.
  • the antibody or antigen-binding fragment thereof comprises a light chain immunoglobulin, a heavy chain immunoglobulin, or both a light and heavy chain immunoglobulin, wherein the light chain immunoglobulin comprises the amino acid sequence set forth in SEQ ID NO:3, 4, 5, 6,7, 11, 13, 15, or 45; and/or the heavy chain immunoglobulin comprises the amino acid sequence set forth in SEQ ID NO: 1, 2, 8, 9, 10, 12, 14, 44, 79, 80, 81, 82, 83, 84, 85, or 86.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 1 or 79; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 3.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 4.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 5.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 6.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 7.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 3.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 8 or 82; and a light chain immunoglobulin comprising the ammo acid sequence set forth in SEQ ID NO: 11.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 9 or 83; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 11.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 10 or 84; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 11.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 12 or 85; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 13.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 14 or 86; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 15.
  • the antibody or antigen-binding fragment thereof comprises a light chain immunoglobulin, a heavy chain immunoglobulin, or both a light and heavy chain immunoglobulin, wherein the light chain variable domain comprises the amino acid sequence set forth in SEQ ID NO:70, 71, 72, 73, 58, 74, 75, 76, or 77, and/or the heavy chain variable domain compnses the ammo acid sequence set forth in SEQ ID NO:63, 57, 64, 65, 66, 67, 68, or 69.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:63; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:70.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:71.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain vanable domain comprising the ammo acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:72.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:73.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:70.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:64; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:74.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:65; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:74.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:66; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO: 74.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:67; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:75.
  • the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:68; and a light chain variable domain comprising the ammo acid sequence set forth in SEQ ID NO:76.
  • the antibody or antigen-binding fragment thereof that binds ILT4 comprises an immunoglobulin light chain variable (VL) domain comprising a CDR-L1, CDR-L2 and CDR-L3 of 1E1 (e.g, SEQ ID NOs: 19-21); and an immunoglobulin heavy chain variable (VH) domain comprising a CDR-H1, CDR-H2 and CDR-H3 of 1E1 (e.g, SEQ ID NOs: 16-18).
  • VL immunoglobulin light chain variable
  • VH immunoglobulin heavy chain variable
  • the antibody or antigen-binding fragment thereof that binds ILT4 comprises an immunoglobulin light chain variable (VL) domain comprising a CDR-L1, CDR-L2 and CDR-L3 of 2A6 (e.g, SEQ ID NOs: 25-27); and an immunoglobulin heavy chain variable (VH) domain comprising a CDR-H1, CDR-H2 and CDR-H3 of 2A6 (e.g., SEQ ID NOs: 22-24).
  • VL immunoglobulin light chain variable
  • VH immunoglobulin heavy chain variable
  • the antibody or antigen-binding fragment thereof that binds ILT4 comprises an immunoglobulin light chain variable (VL) domain comprising a CDR-L1, CDR-L2 and CDR-L3 of 3G7 (e.g, SEQ ID NOs: 31-33); and an immunoglobulin heavy chain variable (VH) domain comprising a CDR-H1, CDR-H2 and CDR-H3 of 3G7 (e.g, SEQ ID NOs: 28-30).
  • VL immunoglobulin light chain variable
  • VH immunoglobulin heavy chain variable domain comprising a CDR-H1, CDR-H2 and CDR-H3 of 3G7
  • the antibody or antigen-binding fragment thereof that binds ILT4 comprises an immunoglobulin light chain variable (VL) domain comprising a CDR-L1, CDR-L2 and CDR-L3 of 2C1 (e.g, SEQ ID NOs: 37-39); and an immunoglobulin heavy' chain variable (VH) domain comprising a CDR-H1, CDR-H2 and CDR-H3 of 2C1 (e.g., SEQ ID NOs: 34-36).
  • VL immunoglobulin light chain variable
  • VH immunoglobulin heavy' chain variable
  • the antibody or antigen-binding fragment comprises a VH domain comprising CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GNSNRPS(SEQ ID NO: 49), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment comprises a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GQSNRPS(SEQ ID NO: 50), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GESNRPS(SEQ ID NO: 51), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21) [0167] In still another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYF
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GNANRPS(SEQ ID NO: 53), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2:
  • EINHSGSTNYNPSLKS SEQ ID NO: 47
  • a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GQANRPS(SEQ ID NO: 54), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2:
  • EINHSGSTNYNPSLKS SEQ ID NO: 47
  • a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GEANRPS(SEQ ID NO: 55), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GDANRPS(SEQ ID NO: 56), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GNSNRPS(SEQ ID NO: 49), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2:
  • EINHAGSTNYNPSLKS SEQ ID NO: 48
  • a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GQSNRPS(SEQ ID NO: 50), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising:CDR-Hl: GYYWS (SEQ ID NO: 16), CDR-H2:
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GDSNR
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GNANRPS(SEQ ID NO: 53), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2:
  • EINHAGSTNYNPSLKS SEQ ID NO: 48
  • a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GQANRPS(SEQ ID NO: 54), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2:
  • EINHAGSTNYNPSLKS SEQ ID NO: 48
  • a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GEANRPS(SEQ ID NO: 55), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: E1NHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: EPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GDANRPS(SEQ ID NO: 56), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the VL domain of antibody 1E1 (e.g, SEQ ID NO:70, 71, 72, 73, 58 or 77) and/or the VH domain of antibody 1E1 (e.g., SEQ ID NO:63, 57 or 69).
  • VL domain of antibody 1E1 e.g, SEQ ID NO:70, 71, 72, 73, 58 or 77
  • VH domain of antibody 1E1 e.g., SEQ ID NO:63, 57 or 69.
  • formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the VL domain of antibody 2A6 (e.g, SEQ ID NO:74) and/or the VH domain of antibody 2A6 (e.g., SEQ ID NO:64, 65 or 66).
  • VL domain of antibody 2A6 e.g, SEQ ID NO:74
  • VH domain of antibody 2A6 e.g., SEQ ID NO:64, 65 or 66.
  • formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the VL domain of antibody 3G7 (e.g., SEQ ID NO:75) and/or the VH domain of antibody 3G7 (e.g., SEQ ID NO:67).
  • VL domain of antibody 3G7 e.g., SEQ ID NO:75
  • VH domain of antibody 3G7 e.g., SEQ ID NO:67
  • formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the VL domain of antibody 2C1 (e.g., SEQ ID NO:76) and/or the VH domain of antibody 2C1 (e.g., SEQ ID NO:68).
  • VL domain of antibody 2C1 e.g., SEQ ID NO:76
  • VH domain of antibody 2C1 e.g., SEQ ID NO:68
  • the formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 1E1 (e.g., SEQ ID NO:3, 4, 5, 6, 7 or 45) and/or the heavy chain immunoglobulin of antibody 1E1 (e.g., SEQ ID NO: I, 2, 44, 79, 80, or 81).
  • the light chain immunoglobulin of antibody 1E1 e.g., SEQ ID NO:3, 4, 5, 6, 7 or 45
  • the heavy chain immunoglobulin of antibody 1E1 e.g., SEQ ID NO: I, 2, 44, 79, 80, or 81.
  • the formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 2A6 (e.g. , SEQ ID NO: 11) and/or the heavy chain immunoglobulin of antibody 2A6 (e.g. , SEQ ID NO:8, 9, 10, 82, 83, or 84).
  • an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 2A6 (e.g. , SEQ ID NO: 11) and/or the heavy chain immunoglobulin of antibody 2A6 (e.g. , SEQ ID NO:8, 9, 10, 82, 83, or 84).
  • the formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 3G7 (e.g, SEQ ID NO: 13) and/or the heavy chain immunoglobulin of antibody 3G7 (e.g., SEQ ID NO: 12 or 85).
  • an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 3G7 (e.g, SEQ ID NO: 13) and/or the heavy chain immunoglobulin of antibody 3G7 (e.g., SEQ ID NO: 12 or 85).
  • the formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 2C1 (e.g, SEQ ID NO: 15) and/or the heavy chain immunoglobulin of antibody 2C1 (e.g., SEQ ID NO: 14 or 86).
  • an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 2C1 (e.g, SEQ ID NO: 15) and/or the heavy chain immunoglobulin of antibody 2C1 (e.g., SEQ ID NO: 14 or 86).
  • each light chain comprises the VL or light chain immunoglobulin of antibody 1E1, 2A6, 3G7, or 2C1
  • each heavy chain comprises the VH or heavy chain immunoglobulin of antibody 1E1, 2A6, 3G7, or 2C1.
  • the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO:58 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO:57.
  • the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO:58 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO:57, wherein the light chain further comprises the amino acid sequence set forth in SEQ ID NO: 90.
  • the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO:58 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO:57, wherein the heavy chain further comprises the amino acid sequence set forth in SEQ ID NO: 89.
  • the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO:58 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO: 57, wherein the light chain further comprises the amino acid sequence set forth in SEQ ID NO:90 and the heavy chain further comprises the amino acid sequence set forth in SEQ ID NO: 89.
  • the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO: 7 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO:2.
  • the antibody consists of two heavy chains and two light chains, wherein each light chain consists of the amino acid sequence set forth in SEQ ID NO:7 and each heavy chain consists of the amino acid sequence set forth in SEQ ID NO:2.
  • the antibody or antigen-binding fragment thereof comprises a VL (with or without signal sequence), e.g., the VL in any of SEQ ID NO:58 or 70-77, having up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more conservative or non-conservative amino acid substitutions; and/or a VH (with or without signal sequence), e.g., the VH in any of SEQ ID NO:57 or 63-69, having up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more conservative or non-conservative amino acid substitutions, while still binding to ILT4.
  • VL with or without signal sequence
  • VH with or without signal sequence
  • formulations disclosed herein can be used with polypeptides comprising the amino acid sequences disclosed herein, e.g., SEQ ID NOs: 1-39, 44, 45, 47-58, 63-77, or 79-86, as well as polypeptides comprising such amino acid sequences with up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 20 or more conservative or non-conservative amino acid substitutions therein.
  • the antibody or antigen-binding fragment thereof comprises a light chain immunoglobulin, a heavy chain immunoglobulin, or both a light and heavy chain immunoglobulin, wherein the light chain immunoglobulin has at least 90% amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO:3, 4, 5, 6,7, 11, 13, 15, or 45, and/or the heavy chain immunoglobulin has at least 90% amino acid sequence identity' to the amino acid sequence set forth in SEQ ID NO: 1, 2, 8, 9, 10, 12, 14, 44, 79, 80, 81, 82, 83, 84, 85, or 86.
  • the antibody or antigen-binding fragment thereof comprises a light chain immunoglobulin, a heavy' chain immunoglobulin, or both a light and heavy chain immunoglobulin, wherein the light chain immunoglobulin comprises a light chain variable domain having at least 90% amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO:70, 71, 72, 73, 58, 74, 75, 76, or 77, and/or the heavy chain immunoglobulin comprises a heavy chain variable domain having at least 90% amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO:63, 57, 64, 65, 66, 67, 68, or 69.
  • an immunoglobulin heavy chain of an anti-ILT4 antibody or antigenbinding fragment of the invention is operably linked to a signal sequence, e.g., comprising the amino acid sequence MEWSWVFLFFLSVTTGVHS (SEQ ID NO:41) and/or an immunoglobulin light chain of an anti-ILT4 antibody or antigen-binding fragment of the invention is operably linked to a signal sequence, e.g., comprising the amino acid sequence MSVPTQVLGLLLLWLTDARC (SEQ ID NO:42).
  • an N-terminal glutamine (Q) of an immunoglobulin chain set forth herein is replaced with a pyroglutamic acid.
  • an N-terminal Q of a heavy chain immunoglobulin is replaced with a pyroglutamic acid.
  • an N-terminal Q of a light chain immunoglobulin is replaced with a pyroglutamic acid.
  • an N-terminal Q of a heavy chain immunoglobulin and an N- terminal Q of a heavy chain immunoglobulin are replaced with a pyroglutamic acid.
  • the formulations disclosed herein can also be used with antibodies or antigen-binding fragments that bind to the same epitope of ILT4 (e.g., human ILT4) as any anti-ILT4 antibody or antigen-binding fragment thereof disclosed herein (e.g., 1E1, 2A6, 3G7 or 2C1).
  • the epitope is LYREKKSASW (SEQ ID NO:59).
  • the epitope is TRIRPEL (SEQ ID NO:60).
  • the epitope is NGQF (SEQ ID NO:61).
  • the epitope is HTGRYGCQ (SEQ ID NO:62).
  • the antibody or antigen-binding fragment thereof binds to the same epitope of human ILT4 as an antibody or antigen-binding fragment thereof comprising the heavy chain and light chain amino acid sequences set forth in SEQ ID NOs: 1 and 3; 2 and 4; 2 and 5; 2 and 6; 2 and 7; 2 and 3; 8 and 11; 9 and 11; 10 and 11; 12 and 13; 14 and 15; 79 and 3; 80 and 4; 80 and 5; 80 and 6; 80 and 7; 80 and 3; 82 and 11; 83 and 11; 84 and 11; 85 and 13; and 86 and 15; respectively.
  • the antibody or antigen-binding fragment thereof binds to the same epitope of human ILT4 as an antibody or antigen-binding fragment thereof comprising the heavy chain variable domain and light chain variable domain amino acid sequences set forth in SEQ ID NOs:63 and 70; 57 and 71; 57 and 72; 57 and 73; 57 and 58; 57 and 70; 64 and 74; 65 and 74; 66 and 74; 67 and 75; 68 and 76; respectively.
  • compositions disclosed herein can be used with antibodies and antigen-binding fragments that cross-block the binding of any anti-ILT4 antibody or antigen-binding fragment thereof disclosed herein (e.g., 1E1, 2A6, 3G7 or 2C1) to ILT4 (e.g., human ILT4) or compete with any anti-ILT4 antibody or antigen-binding fragment thereof disclosed herein (e.g, 1E1, 2A6, 3G7 or 2C1) to ILT4 (e.g., human ILT4).
  • any anti-ILT4 antibody or antigen-binding fragment thereof disclosed herein e.g., 1E1, 2A6, 3G7 or 2C1
  • ILT4 e.g., human ILT4
  • cross-blocking antibodies and antigenbinding fragments thereof discussed herein can be identified based on their ability to block any of the antibodies or fragments specifically set forth herein from binding to ILT4, in binding assays (e.g., bio-layer interferometry (BLI; for example FORTEBIO OCTET binding assay; Pall ForteBio Corp; Menlo Park, CA), surface plasmon resonance (SPR), BIACore, ELISA, flow cytometry).
  • binding assays e.g., bio-layer interferometry (BLI; for example FORTEBIO OCTET binding assay; Pall ForteBio Corp; Menlo Park, CA), surface plasmon resonance (SPR), BIACore, ELISA, flow cytometry.
  • the tip of a fiberoptic probe when using BLI, is coated wdth ligand (e.g., ILT4) and acts as the biosensor wherein binding of anti- ILT4 antibody or antigen-binding fragment to the ILT4 alters the interference pattern of white light reflected from the probe layer bound to 1LT4 and an internal reference layer. The shift is indicative of ILT4/anti-ILT4 binding.
  • the ILT4 coated tip is immersed in a solution of analyte containing antibody or antigen-binding fragment, e.g., in the well of either a 96- or 384-well plate.
  • the plate is shaken during reading to create orbital flow.
  • assays can be used in which an ILT4 (e.g., human ILT4) protein is immobilized on a BLI probe or plate, a reference anti-ILT4 antibody or fragment binds to ILT4 (e.g., at saturating concentration) and a test anti-ILT4 antibody or fragment is added. The ability of the test antibody to compete with the reference antibody for 1LT4 binding is then determined.
  • ILT4 e.g., human ILT4
  • a reference anti-ILT4 antibody or fragment binds to ILT4 (e.g., at saturating concentration) and a test anti-ILT4 antibody or fragment is added. The ability of the test antibody to compete with the reference antibody for 1LT4 binding is then determined.
  • the test antibody effectively competes with the reference antibody, e.g., nanometers of light wavelength shift over time is monitored wherein a shift indicates additional binding of the test antibody and a lack of cross-blocking.
  • cross-blocking is qualitatively deemed to have occurred between the antibodies if no additional binding of test antibody is observed.
  • as a control crossblocking of the reference antibody with itself is confirmed; wherein the assay is determined to be operating correctly if the reference antibody can cross-block itself from ILT4 binding.
  • test antibody to inhibit the binding of the anti-ILT4 antibody or fragment 1E1, 2A6, 3G7 or 2C1, to ILT4 (e.g., human ILT4) demonstrates that the test antibody can cross-block the antibody or fragment for binding to ILT4 (e.g., human ILT4) and thus, may, in some cases, bind to the same epitope on ILT4 (e.g, human ILT4) as 1E1, 2A6, 3G7 and/or 2C1.
  • antibodies and fragments that bind to the same epitope as any of the anti-ILT4 antibodies or fragments of the invention also form part of the invention.
  • BLI is conducted in a sandwich format wherein a reference anti- ILT4 antibody or antigenbinding fragment is immobilized to the probe and then bound with ILT4. Test anti- ILT4 antibody or antigen-binding fragment is then tested for the ability to block binding of the references antibody or fragment.
  • the antibody or antigen-binding fragment thereof competes for binding to human ILT4 with an antibody or fragment comprising the heavy chain and light chain amino acid sequences set forth in SEQ ID NOs: 1 and 3; 2 and 4; 2 and 5; 2 and 6; 2 and 7; 2 and 3; 8 and 11; 9 and 11; 10 and 11; 12 and 13; 14 and 15; 79 and 3; 80 and 4; 80 and 5; 80 and 6; 80 and 7; 80 and 3; 82 and 11; 83 and 11; 84 and 11; 85 and 13; and 86 and 15; respectively.
  • the antibody or antigen-binding fragment thereof competes for binding to human ILT4 with an antibody or fragment comprising the heavy chain variable domain and light chain variable domain amino acid sequences set forth in SEQ ID NOs:63 and 70; 57 and 71; 57 and 72; 57 and 73; 57 and 58; 57 and 70; 64 and 74; 65 and 74; 66 and 74; 67 and 75; 68 and 76; respectively.
  • formulations disclosed herein can be used with anti-ILT4 antibodies and antigenbinding fragments thereof comprising N-linked glycans that are typically added to immunoglobulins produced in Chinese hamster ovary cells (CHO N-linked glycans) or in engineered yeast cells (engineered yeast N-linked glycans), such as, for example, Pichict pastoris.
  • the anti-lLT4 antibodies and antigen-binding fragments thereof comprise one or more of the “engineered yeast N-linked glycans” or “CHO N-linked glycans” (e.g, GO and/or GO-F and/or G1 and/or Gl-F and/or G2-F and/or Man5).
  • the anti-ILT4 antibodies and antigen-binding fragments thereof comprise the engineered yeast N- linked glycans, i.e., GO and/or G1 and/or G2, optionally, further including Man5.
  • the anti-ILT4 antibodies and antigen-binding fragments thereof comprise the CHO N-linked glycans, i.e., GO-F, Gl-F and G2-F, optionally, further including GO and/or G1 and/or G2 and/or Man5.
  • about 80% to about 95% (e.g., about 80-90%, about 85%, about 90% or about 95%) of all N-linked glycans on the anti-ILT4 antibodies and antigenbinding fragments thereof are engineered yeast N-linked glycans or CHO N-linked glycans. See Nett et al. Yeast. 28(3): 237-252 (2011); Hamilton et al. Science.
  • an engineered yeast cell is GFI5.0 or YGLY8316 or strains set forth in U.S. Patent No. 7,795,002 or Zha et al. Methods Mol Biol. 988:31-43 (2013). See also international patent application publication no. WO2013/066765.
  • anti-ILT4 antibodies and antigen-binding fragments thereof that can be used with various formulations disclosed herein (e.g, 1E1, 2A6, 3G7 and/or 2C1) can also be engineered to include modifications within the Fc region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or effector function (e.g., antigen-dependent cellular cytotoxicity).
  • modifications within the Fc region typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or effector function (e.g., antigen-dependent cellular cytotoxicity).
  • antibodies and antigenbinding fragments thereof that can be used with various formulations disclosed herein can be chemically modified (e.g., one or more chemical moieties can be attached to the antibody, such as a labeled antibody or an antibody-drug conjugate) or be modified to alter its glycosylation, again to alter one or more functional properties of the antibody.
  • the antibody or antigen-binding fragment is a monoclonal antibody.
  • the pharmaceutical formulations described herein can retain physical, chemical, and/or biological stability of an anti-ILT4 antibody (e.g., 1E1, 2A6, 3G7, or 2C1) or antigen-binding fragment thereof during storage (e g, at about 5°C for at least 6 months, 12 months, 24 months, or 36 months, at -20°C for at least 6 months, 12 months, 24 months, or 36 months, or at -70°C for at least 6 months, 12 months, 24 months, or 36 months) and/or under various stress conditions (e.g, agitation, freeze and thaw cycles, or light exposure).
  • an anti-ILT4 antibody e.g., 1E1, 2A6, 3G7, or 2C1
  • antigen-binding fragment thereof e.g., at about 5°C for at least 6 months, 12 months, 24 months, or 36 months, at -20°C for at least 6 months, 12 months, 24 months, or 36 months, or at -70°C for at least 6 months, 12
  • an anti-ILT4 monoclonal antibody e.g., 1E1, 2A6, 3G7, or 2C1 or antigen-binding fragment thereof, comprising (i) an anti-ILT4 antibody or antigen-binding fragment thereof (e.g., 1E1, 2A6, 3G7, or 2C1); (ii) a buffer (e.g., L-histidine buffer or acetate buffer); (iii) a non-reducing sugar (e.g., sucrose); (iv) a non-ionic surfactant (e.g., PS-80); and (v) an antioxidant (e.g., L-methionine).
  • a buffer e.g., L-histidine buffer or acetate buffer
  • a non-reducing sugar e.g., sucrose
  • a non-ionic surfactant e.g., PS-80
  • an antioxidant e.g., L-methionine
  • Buffers that can be used in the pharmaceutical formulations disclosed herein include, but are not limited to, succinate (sodium or potassium), L-histidine, phosphate (sodium or potassium), Tris (tris (hydroxymethyl) aminomethane), diethanolamine, citrate (sodium), acetate (sodium) and the like.
  • the buffer is present in the formulation at a concentration of about 1-20 mM (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 mM).
  • the buffer of various formulations described herein has a pH in the range of from about 4.5 to about 7.0, from about 4.5 to about 6.8, from about 5.0 to about 6.8, from about 5.0 to about 6.5, from 5.0 to about 6.0, from about 5.5 to about 6.0.
  • L-histidine and acetate buffers in the pH range of 5.0-6.8 were explored for suitability.
  • a range of pH values such as “a pH from about pH 5.5 to about 6.0,” the range is intended to be inclusive of the recited values.
  • a range from about 5.0 to about 6.0 includes 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, and 6.0.
  • the pH refers to the pH after reconstitution of the lyophilized formulation. pH is typically measured at 25 °C using a standard glass bulb pH meter.
  • a solution comprising “histidine buffer at pH X” refers to a solution at pH X and comprising the histidine buffer, i.e., the pH is intended to refer to the pH of the solution.
  • the anti-ILT4 formulation comprises a non-reducing sugar.
  • non-reducing sugar is a sugar not capable of acting as a reducing agent because it does not contain or cannot be converted to contain a free aldehyde group or a free ketone group.
  • non-reducing sugars include, but are not limited to, disaccharides such as sucrose and trehalose.
  • the non-reducing sugar is present in an amount of from about 1% (w/v) to about 10% (w/v) (about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10% (w/v)).
  • the non-reducing sugar is present in an amount from about 6% (w/v) to about 8% (w/v) (about 6, 7, or 8% (w/v)). In a further embodiment, the non-reducing sugar is present in an amount of about 6% (w/v). In a further embodiment, the non-reducing sugar is present in an amount of about 7% (w/v). In a further embodiment, the non-reducing sugar is present in an amount of about 8% (w/v). In one embodiment, the non-reducing sugar is sucrose, trehalose, or raffinose. In a further embodiment, the sucrose is present at about 6% (w/v) to about 8% (w/v). In one embodiment, the sucrose is present at about 6% (w/v). In one embodiment, the sucrose is present at about 7% (w/v). In one embodiment, the sucrose is present at about 8% (w/v).
  • the anti-ILT4 formulations also comprise a surfactant.
  • a surfactant is a surface active agent that is amphipathic in nature.
  • Surfactants can be added to the formulations herein to provide stability, reduce and/or prevent aggregation, or to prevent and/or inhibit protein damage during processing conditions such as purification, filtration, freeze-drying, transportation, storage, and delivery.
  • a surfactant can be useful for providing additional stability to the active ingredient(s).
  • Non-ionic surfactants that can be used in the formulations of the invention include, but are not limited to, polyoxyethylene sorbitan fatty acid esters (Polysorbates, sold under the trade name Tween® (Uniquema Americas LLC, Wilmington, DE)) including Polysorbate-20 (PS-20, polyoxyethylene sorbitan monolaurate), Polysorbate-40 (PS-40, polyoxyethylene sorbitan monopalmitate), Polysorbate-60 (PS-60, polyoxyethylene sorbitan monostearate), and Polysorbate-80 (PS-80, polyoxyethylene sorbitan monooleate); polyoxyethylene alkyl ethers such as Brij® 58 (Uniquema Americas LLC, Wilmington, DE) and Brij® 35; pol oxamers (e.g., poloxamer 188); Triton® X-100 (Union Carbide Corp., Houston, TX) and Triton® X-114; NP40; Span 20, Span 40, Span 60
  • the non-ionic surfactant is PS-80 or PS-20. In one embodiment, the non-ionic surfactant is PS-20. In another embodiment, the non-ionic surfactant is PS-80.
  • the amount of non-ionic surfactant to be included in the formulations is an amount sufficient to perform the desired function, i.e., a minimal amount necessary to stabilize the active pharmaceutical ingredient (i.e., the anti-ILT4 antibody or antigen-binding fragment thereof e.g., 1E1, 2A6, 3G7, or 2C1)) in the formulation. All percentages for the non-ionic surfactant are listed as % (w/v). Typically, the surfactant is present in a concentration of from about 0.008% to about 0.1% (w/v).
  • the surfactant is present in the formulation in an amount from about 0.01% to about 0.1%, from about 0.01% to about 0.09%, from about 0.01% to about 0.08%, from about 0.01% to about 0.07%, from about 0.01% to about 0.06%, from about 0.01% to about 0.05%, from about 0.01% to about 0.04%, from about 0.01% to about 0.03%, from about 0.01% to about 0.02%, from about 0.015% to about 0.04%, from about 0.015% to about 0.03%, from about 0.015% to about 0.02%, from about 0.02% to about 0.04%, from about 0.02% to about 0.035%, or from about 0.02% to about 0.03%.
  • the surfactant is present in an amount of about 0.01%, about 0.015%, about 0.02%, about 0.025%, about 0.03%, about 0.035%, or about 0.04%.
  • the formulations described herein comprise about 0.01% to about 0.04% (w/v) PS-80.
  • the formulations described herein comprise PS-80 in an amount of about 0.01%.
  • the amount of PS-80 is about 0.015%.
  • the amount of PS-80 is about 0.02%.
  • the amount of PS-80 is about 0.025%.
  • the amount of PS-80 is about 0.03%.
  • the amount of PS-80 is about 0.035%.
  • the amount of PS-80 is about 0.04%.
  • the amount of PS-80 is about 0.045%.
  • the formulations described herein also comprise methionine, or pharmaceutically acceptable salt thereof as an antioxidant.
  • the methionine is L-methionine.
  • the methionine is a pharmaceutically acceptable salt of L-methionine, such as, for example, methionine HC1.
  • methionine is present in the formulation at a concentration of about 1-20 mM (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, and 20 mM).
  • the methionine is present from about 5 mM to about 10 mM (5, 6, 7, 8, 9, and 10 mM).
  • the methionine is present at about 10 mM.
  • a pharmaceutical formulation for an anti-lLT4 antibody or antigen-binding fragment thereof comprising: (i) about 10 mg/mL to about 200 mg/rnL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM of a buffer; (iii) about 6% to about 8% weight /volume (w/v) of a non-reducing sugar; (iv) about 0.01 % to about 0.10% (w/v) of anon-ionic surfactant; and (v) about 1 mM to about 20 mM of an anti-oxidant.
  • the buffer is a selected from the group consisting of L-histidine buffer, acetate buffer, and citrate buffer.
  • the buffer is L-histidine buffer.
  • the buffer is acetate buffer.
  • the buffer is citrate buffer.
  • the non-reducing sugar is sucrose.
  • the non-ionic surfactant is PS-80 or PS-20. In one embodiment, the non-ionic surfactant is PS-80. In another embodiment, the non-ionic surfactant is PS-20.
  • the anti-oxidant is L-methionine.
  • a pharmaceutical formulation for an anti- ILT4 antibody or antigen-binding fragment thereof comprising: (i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM L-histidine buffer; (iii) about 6% to about 8% (w/v) sucrose; (iv) about 0.01 % to about 0.10% (w/v) PS-80; and (v) about 1 mM to about 20 mM L-methionine.
  • the formulation comprises about 8 mM to about 12 mM L- histidine buffer.
  • the formulation comprises about 5 mM to about 10 mM L- methionine.
  • the formulation comprises about 0.01 % to about 0.05% (w/v) PS-80.
  • the formulation comprises about 10 mg/mL to about 150 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof.
  • the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL, about 12.5 mg/mL, about 15 mg/mL, about 25 mg/mL, about 50 mg/mL, about 75 mg/mL, about 100 mg/mL, about 125 mg/mL, or about 150 mg/mL. In one embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL.
  • the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 12.5 mg/mL. In yet another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 15 mg/mL. In still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 25 mg/mL. In one embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 50 mg/mL. In another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 75 mg/mL. In yet another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 100 mg/mL.
  • the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 125 mg/mL. In yet still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 150 mg/mL.
  • the formulation comprises about 25 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation comprises about 50 mg/mL of the anti- ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation comprises about 75 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation comprises about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation comprises about 125 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
  • the formulation has a pH of about 5.0 - about 6.8. In some embodiments, the formulation has a pH of about 5.5 - about 6.0. In other embodiments, the formulation has a pH of about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, or about 6.0. In one embodiment, the formulation has a pH of about 5.5. In another embodiment, the formulation has a pH of about 5.6. In yet another embodiment, the formulation has a pH of about 5.7. In still another embodiment, the formulation has a pH of about 5.8. In another embodiment, the formulation has a pH of about 5.9. In yet still another embodiment, the formulation has a pH of about 6.0.
  • the pharmaceutical formulation for an anti-ILT4 antibody or antigen-binding fragment thereof compnses: (i) about 50 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer, pH about 5.5; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) polysorbate 80; and (v) about 10 mM L-methionine.
  • the anti-ILT4 antibody or antigen-binding fragment thereof is an anti-ILT4 antibody or antigen-binding fragment thereof described herein (e.g. , 1E1, 2A6, 3G7, or 2C1). In some embodiments of the formulations, the anti-ILT4 antibody or antigen-binding fragment thereof is 1E1 or a variant thereof. In some embodiments of the formulations, the anti-ILT4 antibody or antigen-binding fragment thereof is 2A6 or a variant thereof. In some embodiments of the formulations, the anti- ILT4 antibody or antigen-binding fragment thereof is 3G7 or a variant thereof. In some embodiments of the formulations, the anti-lLT4 antibody or antigen-binding fragment thereof is 2C1 or a variant thereof.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein Xi is N, Q, E or D and X 2 is S or A (SEQ ID NO:20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO:21).
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO:48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO:18); and a light chain variable domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GDSNRPS(SEQ ID NO:52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO:21).
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:2 and a light chain comprising the amino acid sequence set forth in SEQ ID NOY.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 80 and a light chain comprising the amino acid sequence set forth in SEQ ID NOY.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:58.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO:2 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO: 80 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
  • an anti-ILT4 antibody or antigen-binding fragment thereof comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L-histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSS
  • an anti- ILT4 antibody or antigen-binding fragment thereof comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: E1NHAGSTNYNPSLKS (SEQ ID NO:48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO:18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO:
  • an anti-ILT4 antibody or antigen-binding fragment thereof comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58.
  • an anti- ILT4 antibody or antigen-binding fragment thereof comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:2 and a light chain comprising the amino acid sequence set forth in SEQ ID NO:7.
  • an anti- ILT4 antibody or antigen-binding fragment thereof comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 80 and a light chain comprising the amino acid sequence set forth in SEQ ID NO:7.
  • an anti-ILT4 antibody or antigen-binding fragment thereof comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:58.
  • an anti-lLT4 antibody or antigen-binding fragment thereof comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO:2 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
  • an anti-ILT4 antibody or antigen-binding fragment thereof comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-lLT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO: 80 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
  • the formulation is a liquid formulation.
  • the liquid formulation is stored at about 3°C - 5°C.
  • the liquid formulation is frozen to at least -70°C or below.
  • the liquid formulation is a reconstituted solution from a lyophilized formulation.
  • the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultra-performance size exclusion chromatography; (ii) the turbidity of the formulation is at most about 0.135 as measured by OD350-500; (iii) the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, and/or the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography; (iv) the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging; and/or (v) the % oxidation of one or more amino acid
  • the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultra-performance size exclusion chromatography.
  • the turbidity of the formulation is at most about 0.135 as measured by OD350-500.
  • the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, as determined by high performance ion-exchange chromatography. In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, as determined by high performance ion-exchange chromatography.
  • the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography.
  • the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%
  • the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%
  • the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography.
  • the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging.
  • the % oxidation of one or more amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the % oxidation of one amino acid residue selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the % oxidation of two amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the % oxidation of three amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-lLT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the % oxidation of four amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • the % oxidation of all five amino acid residues from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
  • a liquid antibody formulation can be made by taking the drug substance (e.g., anti-lLT4 monoclonal antibody) which is in liquid form (e.g., anti-ILT4 monoclonal antibody in an aqueous pharmaceutical formulation) and buffer exchanging it into the desired buffer as the last step of the purification process. There is no lyophilization step in this embodiment.
  • the drug substance in the final buffer is concentrated to a desired concentration. Excipients such as sucrose and PS 80 are added to the drug substance and it is diluted using the appropriate buffer to final protein concentration.
  • the final formulated drug substance is filtered using 0.22pm filters and filled into a final container (e.g., glass vials).
  • the formulations described herein can also be reconstituted from lyophilized formulations.
  • Lyophilized formulations of therapeutic proteins provide several advantages. Lyophilized formulations in general offer better chemical stability than solution formulations, and thus increased half-life.
  • a lyophilized formulation may also be reconstituted at different concentrations depending on clinical factors, such as route of administration or dosing. For example, a lyophilized formulation may be reconstituted at a high concentration (i.e., in a small volume) if necessary for subcutaneous administration, or at a lower concentration if administered intravenously. High concentrations may also be necessary if high dosing is required for a particular subject, particularly if administered subcutaneously where injection volume must be minimized.
  • lyophilized antibody formulation is disclosed at U.S. Pat. No. 6,267,958, which is hereby incorporated by reference in its entirety.
  • Lyophilized formulations of another therapeutic protein are disclosed at U.S. Pat. No. 7,247,707, which is hereby incorporated by reference in its entirety.
  • the lyophilized formulation is prepared in anticipation of reconstitution at high concentration of drug product (DP, in an exemplary embodiment anti-lLT4 antibody or antigen-binding fragment thereof), i.e., in anticipation of reconstitution in a low volume of water. Subsequent dilution with water or isotonic buffer can then readily be used to dilute the DP to a lower concentration.
  • excipients are included in a lyophilized formulation of the invention at levels that will result in a roughly isotonic formulation when reconstituted at high DP concentration, e.g., for subcutaneous administration.
  • the lyophilized formulations of the invention are formed by lyophilization (freeze- drying) of a pre-lyophilization solution. Freeze-drying is accomplished by freezing the formulation and subsequently subliming water at a temperature suitable for primary drying.
  • the product temperature is below the eutectic point or the collapse temperature of the formulation.
  • the shelf temperature for the primary drying will range from about -30 to 25°C (provided the product remains frozen during primary drying) at a suitable pressure, ranging typically from about 50 to 250 mTorr.
  • the formulation, size and type of the container holding the sample (e.g., glass vial) and the volume of liquid will dictate the time required for drying, which can range from a few hours to several days (e.g., 40-60 hrs).
  • a secondary drying stage may be carried out at about 0-40°C, depending primarily on the type and size of container and the ty pe of protein employed. The secondary drying time is dictated by the desired residual moisture level in the product and typically takes at least about 5 hours.
  • the moisture content of a lyophilized formulation is less than about 5%, and preferably less than about 3%.
  • the pressure may be the same as that employed during the primary drying step. Freeze-drying conditions can be varied depending on the formulation and vial size.
  • the container in which reconstitution of the protein is to be carried out may, for example, be a 3, 5, 10, 20, 50 or 100 cc vial.
  • the lyophilized formulations are reconstituted prior to administration.
  • the protein may be reconstituted at a concentration of about 10, 15, 20, 25, 30, 40, 50, 60, 75, 80, 90 or 100 mg/mL or higher concentrations such as 150 mg/mL, 200 mg/mL, 250 mg/mL, or 300 mg/mL up to about 500 mg/mL.
  • the protein concentration after reconstitution is about 10-300 mg/mL.
  • the protein concentration after reconstitution is about 20-250 mg/mL.
  • the protein concentration after reconstitution is about 150-250 mg/mL.
  • the protein concentration after reconstitution is about 180-220 mg/mL.
  • the protein concentration after reconstitution is about 50-150 mg/mL.
  • the protein concentration after reconstitution is about 150 mg/mL. In one embodiment, the protein concentration after reconstitution is about 125 mg/mL. In one embodiment, the protein concentration after reconstitution is about 100 mg/mL. In one embodiment, the protein concentration after reconstitution is about 75 mg/mL. In one embodiment, the protein concentration after reconstitution is about 50 mg/mL. In one embodiment, the protein concentration after reconstitution is about 25 mg/mL. High protein concentrations are particularly useful where subcutaneous delivery of the reconstituted formulation is intended. However, for other routes of administration, such as intravenous administration, lower concentrations of the protein may be desired (e.g., from about 5-50 mg/rnL).
  • Reconstitution generally takes place at a temperature of about 25 °C to ensure complete hydration, although other temperatures may be employed as desired.
  • the time required for reconstitution will depend, e.g., on the type of diluent, amount of excipient(s) and protein.
  • exemplary diluents include sterile water, bacteriostatic water for injection (BWFI), a pH buffered solution (e.g., phosphate-buffered saline), sterile saline solution, Ringer’s solution or dextrose solution.
  • a method of treating cancer in a subject comprising administering a therapeutically effective amount of any formulation described herein to the subject.
  • the formulation is administered to the subject via intravenous administration.
  • the formulation is administered to the subject by subcutaneous administration.
  • the invention comprises a method of treating cancer in a human patient comprising administering any formulation described herein to the patient.
  • the cancer can be selected from the group consisting of: melanoma, lung cancer, head and neck cancer, bladder cancer, breast cancer, gastrointestinal cancer, multiple myeloma, hepatocellular cancer, lymphoma, renal cancer, mesothelioma, ovarian cancer, esophageal cancer, anal cancer, bi 1 i ary tract cancer, colorectal cancer, cervical cancer, thyroid cancer, salivary cancer, prostate cancer (e.g., hormone refractory prostate adenocarcinoma), pancreatic cancer, colon cancer, esophageal cancer, liver cancer, thyroid cancer, glioblastoma, glioma, and other neoplastic malignancies.
  • melanoma lung cancer, head and neck cancer, bladder cancer, breast cancer, gastrointestinal cancer, multiple myeloma, hepatocellular cancer, lymphoma, renal cancer, mesothelioma, ovarian cancer, esophageal cancer, anal cancer,
  • the lung cancer in non-small cell lung cancer.
  • the lung cancer is small-cell lung cancer.
  • the lymphoma is Hodgkin lymphoma.
  • the lymphoma is non-Hodgkin lymphoma.
  • the lymphoma is mediastinal large B-cell lymphoma.
  • the breast cancer is triple negative breast cancer.
  • the breast cancer is ER+/HER2- breast cancer.
  • the bladder cancer is urothelial cancer.
  • the head and neck cancer is nasopharyngeal cancer.
  • the cancer is thyroid cancer.
  • the cancer is salivary cancer.
  • the cancer is squamous cell carcinoma of the head and neck.
  • the cancer is a solid tumor with a high level of microsatellite instability (MSI-H).
  • the cancer is a solid tumor with a high mutational burden.
  • the cancer is metastatic colorectal cancer with high levels of microsatellite instability (MSI-H).
  • the cancer is selected from the group consisting of colorectal cancer, esophageal cancer, melanoma, non-small cell lung cancer, ovarian cancer, renal cell cancer and small cell lung cancer.
  • the cancer is a hematological malignancy.
  • the Heme malignancy is acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), diffuse large B-cell lymphoma (DLBCL), EBV-positive DLBCL, primary mediastinal large B-cell lymphoma, T-cell/histiocyte-rich large B-cell lymphoma, follicular lymphoma, Hodgkin’s lymphoma (HL), mantle cell lymphoma (MCL), multiple myeloma (MM), myeloid cell leukemia-1 protein (Mcl-1), myelodysplastic syndrome (MDS), non-Hodgkin lymphoma (NHL), or small lymphocytic lymphoma (SLL).
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • CLL chronic lymphocytic
  • Malignancies that demonstrate improved disease-free and overall survival in relation to the presence of tumor-infdtrating lymphocytes in biopsy or surgical material e.g., melanoma, colorectal, liver, kidney, stomach/esophageal, breast, pancreas, and ovarian cancer are encompassed in the methods and treatments described herein.
  • Such cancer subtypes are known to be susceptible to immune control by T lymphocytes.
  • refractory or recurrent malignancies whose growth may be inhibited using the antibodies described herein.
  • Additional cancers that can benefit from treatment with the formulations described herein include those associated with persistent infection with viruses such as human immunodeficiency viruses, hepatitis viruses class A, B and C, Epstein Barr virus, human papilloma viruses that are known to be causally related to for instance Kaposi’s sarcoma, liver cancer, nasopharyngeal cancer, lymphoma, cervical, vulval, anal, penile and oral cancers.
  • viruses such as human immunodeficiency viruses, hepatitis viruses class A, B and C, Epstein Barr virus, human papilloma viruses that are known to be causally related to for instance Kaposi’s sarcoma, liver cancer, nasopharyngeal cancer, lymphoma, cervical, vulval, anal, penile and oral cancers.
  • the formulations can also be used to prevent or treat infections and infectious disease.
  • the invention provides a method for treating chronic infection in a mammalian subject comprising administering an effective amount of a formulation of the invention to the subject.
  • the formulation is administered to the subject via intravenous administration.
  • the formulation is administered to the subject by subcutaneous administration.
  • These agents can be used alone, or in combination with vaccines, to stimulate the immune response to pathogens, toxins, and self-antigens.
  • the antibodies or antigen-binding fragment thereof can be used to stimulate immune response to viruses infectious to humans, including but not limited to: human immunodeficiency viruses, hepatitis viruses class A, B and C, Epstein Barr virus, human cytomegalovirus, human papilloma viruses, and herpes viruses.
  • Antagonist anti-PD-1 antibodies or antibody fragments can be used to stimulate immune response to infection with bacterial or fungal parasites, and other pathogens. Viral infections with hepatitis B and C and HIV are among those considered to be chronic viral infections.
  • the formulations of the invention may be administered to a patient in combination with one or more “additional therapeutic agents”.
  • the additional therapeutic agent may be a biotherapeutic agent (including but not limited to antibodies to VEGF, EGFR, Her2/neu, VEGF receptors, other grow th factor receptors, CD20, CD40, CD-40L, OX-40, 4-1BB, and ICOS), an immunogenic agent (for example, attenuated cancerous cells, tumor antigens, antigen presenting cells such as dendritic cells pulsed with tumor derived antigen or nucleic acids, immune stimulating cytokines (for example, IL-2, IFNa2, GM-CSF), and cells transfected with genes encoding immune stimulating cy tokines such as but not limited to GM-CSF).
  • a biotherapeutic agent including but not limited to antibodies to VEGF, EGFR, Her2/neu, VEGF receptors, other grow th factor receptors, CD20, CD40, CD-40L, OX-40,
  • the method further comprises administering an additional therapeutic agent.
  • the additional therapeutic agent is an anti-PD-1 antibody or antigen-binding fragment thereof, an anti-PD-Ll antibody or antigen binding fragment thereof, an anti-LAG3 antibody or antigen binding fragment thereof, an anti-TIGIT antibody or antigen binding fragment thereof, an anti- GITR antibody or antigen-binding fragment thereof, an anti-CTL4 antibody or antigen-binding fragment thereof, an anti-CD27 antibody or antigen-binding fragment thereof.
  • the additional therapeutic agent is a Newcastle disease viral vector expressing IL- 12.
  • the additional therapeutic agent is dinaciclib.
  • the additional therapeutic agent is a STING agonist.
  • Suitable routes of administration may, for example, include parenteral delivery, including intramuscular, subcutaneous, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal.
  • Drugs can be administered in a variety of conventional ways, such as intraperitoneal, parenteral, intraarterial or intravenous injection.
  • Modes of administration in which the volume of solution must be limited e.g., subcutaneous administration
  • a dosage of the additional therapeutic agent depends on several factors, including the serum or tissue turnover rate of the entity, the level of symptoms, the immunogenicity of the entity, and the accessibility of the target cells, tissue or organ in the individual being treated.
  • the dosage of the additional therapeutic agent should be an amount that provides an acceptable level of side effects. Accordingly, the dose amount and dosing frequency of each additional therapeutic agent (e.g., biotherapeutic or chemotherapeutic agent) will depend in part on the particular therapeutic agent, the severity of the cancer being treated, and patient characteristics. Guidance in selecting appropriate doses of antibodies, cytokines, and small molecules are available. See, e.g., Wawrzynczak (1996) Antibody Therapy, Bios Scientific Pub.
  • Determination of the appropriate dosage regimen may be made by the clinician, e.g., using parameters or factors known or suspected in the art to affect treatment or predicted to affect treatment, and will depend, for example, the patient’s clinical history (e.g., previous therapy), the ty pe and stage of the cancer to be treated and biomarkers of response to one or more of the therapeutic agents in the combination therapy.
  • a pharmaceutical antibody formulation can be administered by continuous infusion, or by doses at intervals of, e.g., one day, 1-7 times per week, one week, two weeks, three weeks, monthly, bimonthly, etc.
  • a preferred dose protocol is one involving the maximal dose or dose frequency that avoids significant undesirable side effects.
  • a total weekly dose is generally at least 0.05 pg/kg, 0.2 pg/kg, 0.5 pg/kg, 1 pg/kg, 10 pg/kg, 100 pg/kg, 0.2 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 10 mg/kg, 25 mg/kg, 50 mg/kg body weight or more. See, e.g., Yang et al. (2003) New Engl. J. Med.
  • a small molecule therapeutic e.g., a peptide mimetic, natural product, or organic chemical, is about the same as for an antibody or polypeptide, on a moles/kg basis.
  • Embodiments of the invention also include one or more of the biological formulations described herein (i) for use in, (ii) for use as a medicament or composition for, or (iii) for use in the preparation of a medicament for: (a) therapy (e.g., of the human body); (b) medicine; (c) induction of or increasing of an antitumor immune response; (d) decreasing the number of one or more tumor markers in a patient; (e) halting or delaying the growth of a tumor or a blood cancer; (1) halting or delaying the progression of a PD-1 -related disease or an ILT4-related disease; (g) stabilization of a PD-1 -related disease or an ILT4-related disease; (h) inhibiting the grow th or survival of tumor cells; (i) eliminating or reducing the size of one or more cancerous lesions or tumors; (j) reduction of the progression, onset or severity of a PD-l-related disease or an ILT4- related disease
  • Monoclonal, polyclonal, and humanized antibodies can be prepared (see, e.g., Sheperd and Dean (eds.) (2000) Monoclonal Antibodies, Oxford Univ. Press, New York, NY;
  • An alternative to humanization is to use human antibody libraries displayed on phage or human antibody libraries in transgenic mice (Vaughan et al. (1996) Nature Biotechnol. 14:309- 314; Barbas (1995) Nature Medicine 1:837-839; Mendez et al. (1997) Nature Genetics 15:146- 156; Hoogenboom and Chames (2000) Immunol. Today 21.Ml-MT, Barbas et al. (2001) Phage Display: A Laboratory Manual, Cold Spring Harbor Laboratory' Press, Cold Spring Harbor, New York; Kay et al. (1996) Phage Display of Peptides and Proteins: A Laboratory Manual, Academic Press, San Diego, CA; de Bruin et al. (1999) Nature Biotechnol. 17:397-399).
  • Antibodies can be conjugated, e.g., to small drug molecules, enzymes, liposomes, polyethylene glycol (PEG). Antibodies are useful for therapeutic, diagnostic, kit or other purposes, and include antibodies coupled, e.g., to dyes, radioisotopes, enzymes, or metals, e.g., colloidal gold (see, e.g., Le Doussal et al. (1991) J. Immunol. 146: 169-175; Gibellini et al.
  • Fluorescent reagents suitable for modifying nucleic acids including nucleic acid primers and probes, polypeptides, and antibodies, for use, e.g, as diagnostic reagents, are available (Molecular Probesy (2003) Catalogue, Molecular Probes, Inc., Eugene, OR; Sigma- Aldrich (2003) Catalogue, St. Louis, MO).
  • Analytical methods suitable for evaluating the product stability include size exclusion chromatography (SEC), dynamic light scattering test (DLS), differential scanning calorimetry (DSC), iso-asp quantification, potency, UV at 340 nm, UV spectroscopy, and FTIR.
  • SEC size exclusion chromatography
  • DSC differential scanning calorimetry
  • iso-asp quantification potency, UV at 340 nm, UV spectroscopy, and FTIR.
  • SEC J. Pharm. Scien., 83: 1645-1650, (1994); Pharm. Res., 11 :485 (1994); J. Pharm. Bio. Anal., 15: 1928 (1997); J. Pharm. Bio. Anal., 14: 1133-1140 (1986)
  • DSC Pharm.
  • the iso-asp content in the samples is measured using the Isoquant Isoaspartate Detection System (Promega).
  • the kit uses the enzyme Protein Isoaspartyl Methyltransferase (PIMT) to specifically detect the presence of isoaspartic acid residues in a target protein.
  • PIMT catalyzes the transfer of a methyl group from S-adenosyl-L-methionine to isoaspartic acid at the .alpha. -carboxyl position, generating S-adenosyl-L-homocysteine (SAH) in the process.
  • SAH S-adenosyl-L-homocysteine
  • the potency or bioidentity of an antibody can be measured by its ability to bind to its antigen.
  • the specific binding of an antibody to its antigen can be quantitated by any method known to those skilled in the art, for example, an immunoassay, such as ELISA (enzyme-linked immunosorbent assay).
  • Embodiment 1 provides a formulation of an anti-human immunoglobulin-like transcnpt 4 (anti-ILT4) antibody or antigen-binding fragment thereof, comprising: (i) about 10 mg/rnL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM of a buffer; (iii) about 6% to about 8% weight /volume (w/v) of a nonreducing sugar; (iv) about 0.01 % to about 0.
  • anti-ILT4 anti-human immunoglobulin-like transcnpt 4
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21), wherein the buffer is an L-histidine buffer, an acetate buffer
  • Embodiment 3 provides the formulation of embodiment 1 or 2, wherein the nonreducing sugar is sucrose.
  • Embodiment 4 provides the formulation of any one of embodiments 1-3, wherein the non-ionic surfactant is polysorbate 80.
  • Embodiment 5 provides the formulation of any one of embodiments 1-4, wherein the anti-oxidant is L-methionine.
  • Embodiment 6 provides formulation of an anti-human immunoglobulin-like transcript 4 (“anti-ILT4”) antibody or antigen-binding fragment thereof, comprising: (i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM L-histidine buffer; (iii) about 6% to about 8% weight/volume (w/v) sucrose; (iv) about 0.01 % to about 0.
  • anti-ILT4 anti-human immunoglobulin-like transcript 4
  • Embodiment 7 provides the formulation of embodiment 6, comprising about 8 mM to about
  • Embodiment 8 provides the formulation of embodiment 6 or 7, comprising about 5 mM to about 10 mM L-methionine.
  • Embodiment 9 provides the formulation of any of embodiments 6-8, comprising about 0.01 % to about 0.05% (w/v) polysorbate 80.
  • Embodiment 10 provides the formulation of any of embodiments 1-9, comprising about 10 mg/mL to about 150 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof.
  • Embodiment 11 provides the formulation of embodiment 10, wherein the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL, about 12.5 mg/mL, about 25 mg/mL, about 50 mg/mL, about 75 mg/mL, about 100 mg/mL, about 125 mg/mL, or about 150 mg/mL.
  • Embodiment 12 provides the formulation of any of embodiments 1-11, comprising about 25 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L- histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
  • Embodiment 13 provides the formulation of any of embodiments 1-11, comprising about 50 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L- histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
  • Embodiment 14 provides the formulation of any of embodiments 1-11, comprising about 75 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L- histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
  • Embodiment 15 provides the formulation of any of embodiments 1-11, comprising about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
  • Embodiment 16 provides the formulation of any of embodiments 1-11, comprising about 125 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
  • Embodiment 17 provides the formulation of any one of embodiments 1-16, wherein the formulation has a pH of about 5.0 to about 6.8.
  • Embodiment 18 provides the formulation of any one of embodiments 1-16, wherein the formulation has a pH of about 5.5 to about 6.0.
  • Embodiment 19 provides the formulation of any one of embodiments 1-16, wherein the formulation has a pH of about 5.5.
  • Embodiment 20 provides the formulation of an anti-human immunoglobulin-like transcript 4 (“anti-ILT4”) antibody or antigen-binding fragment thereof, comprising: (i) about 50 mg/rnL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L-histidine buffer, pH about 5.5; (iii) about 7% weight/volume (w/v) sucrose; (iv) about 0.025% (w/v) polysorbate 80; and (v) about 10 mM L-methionme, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and
  • Embodiment 21 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO:47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-
  • Embodiment 22 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GDSNRPS(SEQ ID NO: 52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
  • the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H
  • Embodiment 23 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising the ammo acid sequence set forth in SEQ ID NO:57 and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58.
  • Embodiment 24 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:2 and a light chain comprising the amino acid sequence set forth in SEQ ID NO:7.
  • Embodiment 25 provides the formulation of any embodiments of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 80 and a light chain comprising the amino acid sequence set forth in SEQ ID NO:7.
  • Embodiment 26 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO: 57 and a light chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:58.
  • Embodiment 27 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO:2 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
  • Embodiment 28 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO: 80 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
  • Embodiment 29 provides the formulation of any one of embodiments 1-28, wherein the formulation is a liquid formulation stored at about 3°C to about 5°C, is frozen to -20°C or below, is frozen to -70°C or below, or is a reconstituted solution from a lyophilized formulation.
  • Embodiment 30 provides the formulation of any one of embodiments 1-27, wherein after storage at about 3°C to about 5°C for 6 months: (i) the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultraperformance size exclusion chromatography; (ii) the turbidity of the formulation is at most about 0.
  • the % main peak of the anti-ILT4 antibody or antigenbinding fragment thereof is at least about 63%, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, and/or the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography;
  • Embodiment 31 provides the formulation of any of embodiments 1-30, wherein the anti- ILT4 antibody or antigen-binding fragment thereof is a monoclonal antibody.
  • Embodiment 32 provides a method of treating a cancer in a human patient in need thereof, the method comprising administering an effective amount of the formulation of any one of embodiment 1-31.
  • Embodiment 33 provides the method of embodiment 32, wherein the cancer is selected from the group consisting of colorectal cancer, esophageal cancer, melanoma, non-small cell lung cancer, ovarian cancer, renal cell cancer and small cell lung cancer.
  • Embodiment 34 provides the use of the formulation of any one of embodiments 1-31 for preparing a medicament for treating a cancer.
  • Embodiment 35 provides the use of the formulation of any one of embodiments 1-31 for the treatment of cancer in a human patient.
  • Embodiment 36 provides the use of embodiment 34, wherein the cancer is selected from the group consisting of colorectal cancer, esophageal cancer, melanoma, non-small cell lung cancer, ovarian cancer, renal cell cancer and small cell lung cancer.
  • Table 3 summarizes the pre-formulation developability plan.
  • a fully human anti-ILT4 monoclonal antibody, 1E1 with a human lambda light chain constant domain and a human IgG4- S228P heavy chain constant domain (“Antibody 1”) was tested. All formulations were prepared in 96-well Axygen Polypropylene deep well plates (Product # P-DW-l l-C) using the JANUS automated liquid handling system equipped with the Parisian pipetting arm. There was 500 pL aliquot in each well with a final concentration of 1 mg/mL Antibody 1.
  • a 200 pL aliquot was removed from the plate for initial sample analysis (20 pL for Differential Scanning Fluorimetry (DSF) thermal transition analysis and 180 pL for sub-visible particle analysis using Guava® easyCyte flow cytometer).
  • the remaining 300 pL sample in the 96-well Axygen plate was covered and tightly sealed with a NuncTM microplate sealing cap mat (Product # 276000).
  • the plate was double-packed in SCS Dri-Shield® 2000 Series Moisture Barrier Bags (Product # 700810) to prevent evaporation.
  • the sealed plate was subjected to stress for 10 days at 50°C in a thermal chamber. Protein concentration was monitored before and after thermal stress to account for any increase in protein concentration due to evaporation. The increase in protein concentration across the plate was less than 5%. All the standard precautions were observed during handling of protein solution (e g., equilibration of the solution to room temperature, minimal shaking and exposure to room light).
  • the plate was equilibrated at room temperature for 30 minutes. The plate was then carefully opened in a laminar hood. Using 200 pL multi-channel pipette, the stressed sample was mixed up to 5 times. Then 200 pL of sample was removed and dispensed to a 96-well costar clear plate (Product # 3635) and the absorbance was measured at wavelengths 280, 320, 350 and 500 nm. Following UV analysis, 180 pL of the 200 pL aliquot was carefully transferred to a Coming® 96-well clear V-bottom microplate (Product #3894) for sub-visible particle analysis.
  • the remaining 20 pL was utilized for quantitative protein purity analysis with reduced high-throughput CE-SDS using Caliper’s LabChip GXII system.
  • the remaining 100 pL stressed sample in the Axygen deep well plate was transferred to ABgene 96- well PCR plate (Product # AB-2800) along with initial (unstressed) sample for UP-SEC and cIEF analysis.
  • the chemical stability of the Antibody 1 formulation was evaluated by cIEF under different buffer, pH, and ionic strength conditions. cIEF profile showed a decrease in relative main peak area and significant increase in the relative peak area of the acidic variants after 10 days at 50°C. Addition of salt did not result in any significant improvement in the stability of the protein across studied compositions.
  • the formulation that imparted the maximum stability to Antibody 1 was identified as 10 mM Histidine in the pH range of 5.5 -6.0 or 10 mM Acetate at pH 5.6.
  • a forced degradation study was conducted on formulations including Antibody 1.
  • the tested Antibody 1 formulation contained 50 mg/mL antibody, 10 mM L-histidine buffer at pH 6.0, 7%(w/v) sucrose, and 0.02%(w/v) PS-80.
  • the Antibody 1 formulation was exposed to various stress conditions, including heat (50°C, up to 7 days), high pH (pH 10, 25°C, up to 7 days), low pH (pH 3.5, 25°C, up to 7 days), light (25°C, up to 2X ICH scale), or peroxide (0.1% tBHP, 25°C, up to 24 hours).
  • the stressed samples were visually inspected for color and particulate, and tested by UV, UP-SEC, NR-CE-SDS, R-CE-SDS, HP-IEX, DLS and LC-MS (intact and reduced mass, and reduced peptide mapping).
  • the major degradations include increase of acidic variants upon heat (50°C), high pH (pH 10.0) and light exposure stresses; increases of deamidation and isomerization upon high pH stress; increases of HMW species, basic variants, Trp oxidation upon light exposure (2X) stress; and increase of Met oxidation upon light exposure and peroxide (0.1% tBHP) stresses. There was no major degradation pathway upon low pH (pH 3.5) stress.
  • the minor degradations include increase of HMW species upon heat, high pH, and low pH stresses; increase of LMW species upon heat, high pH, low pH, and light exposure stresses; increase of turbidity upon light exposure stress; and increase of basic variants upon peroxide stress.
  • Two formulation matrixes consisted of 10 mM histidine (with or without 5 rnM L-methionine as an antioxidant) and the other two consisted of 10 mM acetate (with or without 5 mM L-methionine as an antioxidant); each of the four formulations were prepared with 7% (w/v) sucrose as a stabilizer and 0.025% w/v polysorbate 80 (PS-80) as a surfactant at pH 5.5.
  • the 10 mM L-histidine buffer consists of 0.29 mg/mL L-histidine and 1.71 mg/mL L-histidine monohydrochloride monohydrate to obtain target formulation pH of 5.5.
  • the 10 mM L-methionine in the formulation is equivalent to 1.49 mg/mL L-methionine whereas the 7% (w/v) sucrose and 0.025% (w/v) polysorbate 80 are equivalent to 70 mg/mL sucrose and 0.25 mg/mL PS-80 in the formulation, respectively.
  • the formulations were staged up to 6 months on stability under 1CH conditions: 5°C (5°C ⁇ 3°C), 25°C (25°C, 60% RH), and 40°C (40°C, 75% RH).
  • the stability plan is provided in Table 6 below.
  • the four formulations were tested for visual appearance, pH, protein concentration (UVzso), and colloidal stability by turbidity (OD350-500) measurements, and purity by ultra-performance size exclusion chromatography (UP-SEC), charge variants by high performance ion-exchange chromatography (HP-IEX), subvisible particle count by microflow imaging (MFI) and % change in oxidation.
  • UP-SEC ultra-performance size exclusion chromatography
  • HP-IEX high performance ion-exchange chromatography
  • MFI microflow imaging
  • the Antibody 1 formulations were stable up to 6 months at 5°C storage as per the stability plan.
  • the presence of 5 mM L-methionine as an antioxidant in the formulation was found to improve the colloidal stability (OD350-500) as well as reduce soluble aggregates (%HMW) to some extent under 25°C accelerated as well as 40°C stressed conditions for the anti-ILT4 monoclonal antibody formulations in the two buffer matrixes (FIGS. 1 A-1I).
  • FIGS. 1 A-1I There was no impact of 5 mM L-methionine on the charge profile by HP-IEX (FIGS. 2A-2I).
  • L-histidine was chosen as the lead buffer and acetate was chosen as back-up.
  • the pH ranging studies were conducted to determine the Antibody 1 drug product stability ⁇ 0.5 pH units from the target formulation pH of 5.5, i.e., at pH 5.0 or at pH 6.0, and to help define pH specifications for formal stability.
  • Four Antibody 1 pH ranging formulations were staged up to 6 months on stability under ICH conditions: 5°C (5°C 3°C), 25°C (25°C, 60% RH), and 40°C (40°C, 75% RH).
  • Two formulations were prepared in 10 mM histidine buffer, with 7% (w/v) sucrose as a stabilizer and 0.025% (w/v) polysorbate 80 as a surfactant; one formulation at pH 5.0 and the other at pH 6.0.
  • Two formulations were prepared in 10 mM acetate buffer, with 7% (w/v) sucrose as a stabilizer and 0.025% (w/v) polysorbate 80 as a surfactant; one formulation at pH 5.0 and the other at pH 6.0.
  • the purified Antibody I was dialyzed against each of the buffers, the pH of the final formulation was measured following the dialysis, and no pH adjustments were made after this step.
  • the final formulated drug substance was filtered to obtain drug product bulk which were subsequently filled into 2 mL glass vials, rubber stoppered, and sealed.
  • the stability plan was the same as shown in Table 6.
  • the four formulations were tested for visual appearance, pH, protein concentration (UV280), colloidal stability by turbidity (OD350- 500) measurements, purity by UP-SEC, charge variants by HP-IEX, and subvisible particle count by MFI.
  • the anti-ILT4 monoclonal antibody showed better colloidal (OD350-500), physical (%HMW and %monomer) and chemical stability at the lower pH (5.0), followed by the target pH (5.5) and the higher pH (6.0). Based on these data set, pH 5.5 was selected as the anti-ILT4 monoclonal antibody formulation pH. Table 11. pH Ranging Studies (L-Histidine Formulation pH 5.0)
  • anti-ILT4 monoclonal antibody formulations including Antibody 1 were carried out using polysorbate 80 (PS-80) to analyze the sensitivity of the antibody to agitation stress in the presence or absence of the surfactant.
  • PS-80 polysorbate 80
  • the anti-ILT4 monoclonal antibody formulations consisted of 50 mg/mL Antibody 1 in 10 rnM histidine buffer at pH 5.5 and 7% (w/v) sucrose.
  • the anti-ILT4 drug product vials were agitated at 300 rpm on Thermo Scientific Shaker up to 3 days or up to 7 days placed horizontally (at ambient conditions).
  • the same five anti-ILT4 formulations (PS-80 ranging) were placed up to 7 days at ambient room temperature without shaking and were used as respective study controls to eliminate temperature effect. All the vials (stressed and ambient control) were covered with aluminum foil to eliminate light effect.
  • the samples were analyzed for visual appearance, protein concentration (UV280), turbidity (OD350-500), subvisible particulates by MFI, purity by UP-SEC, and charge variants by HP-IEX.
  • FIGS. 5A-5G The stability trends for the five anti-ILT4 monoclonal antibody surfactant ranging formulations are provided in FIGS. 5A-5G. As seen in these figures, the absence of surfactant resulted in generation of visible particles from 3 days of agitation up to 7 days of agitation with increasing amounts. Few visible particles were also seen in the 7 days ambient control sample. There was an increase in turbidity (OD350-500), an increase in subvisible particle counts (> 2 pm, > 5 pm, > 10 pm and > 25 pm), as well as an increase in soluble aggregate levels (%HMW).
  • a third formulation consisted of 50 mg/mL of the anti-ILT4 monoclonal antibody, 10 mM L-histidine at pH 5.5, 7% (w/v) sucrose, and 0.025% (w/v) polysorbate 80 sucrose to serve as dark control (nonlight exposed) under light stress (combination of ultraviolet light-UV and cool white light-CWL or visible light) at 0.2 ICH, 0.5 ICH or 1 ICH in a Caron Photostability Chamber at ambient room temperature condition.
  • the dark control samples were covered with aluminum foil and placed in the same chamber as the light exposed samples to eliminate temperature effect. The samples were exposed to a cumulative amount of light exposure and were removed as exposure limits were completed.
  • the samples were analyzed for visual appearance, protein concentration (UVzso), turbidity (OD350-500), purity by UP-SEC, charge variants by HP-IEX, oxidation levels by reduced peptide mapping (the anti-ILT4 monoclonal antibody oxidation hotspots M359, W7, M253, W102, M429), as well as potency by Binding ELISA.
  • 10 mM L-methionine was found to improve the colloidal stability (OD350-500), reduce soluble aggregate levels, maintain the monomer content (% monomer), minimize change in charged species, and minimize oxidation of L-methionine as well as L-tryptophan residues (to some extent) in the anti-ILT4 monoclonal antibody. Moreover, the potency was found to improve in the presence of 10 mM L-methionine upon exposure to extreme light conditions (1 1CH) as shown in Figure 9.
  • the lead anti-lLT4 monoclonal antibody formulation was selected as follows: 50 mg/mL anti-ILT4 antibody, 0.29 mg/mL L-histidine, 1.71 mg/mL L-histidine monohydrochloride monohydrate, 1.49 mg/mL L-methionine, 70 mg/mL sucrose, and 0.25 mg/mL polysorbate 80.
  • the stability of the lead anti-ILT4 monoclonal antibody formulation (50 mg/mL anti- ILT4, 0.29 mg/mL L-histidine, 1.71 mg/mL L-histidine monohydrochloride monohydrate, 1.49 mg/mL L-methionine, 70 mg/mL sucrose, 0.25 mg/mL polysorbate 80, pH 5.5) was assessed under freeze-thaw stress. A 2.2 mL volume was used to fill the anti-ILT4 drug product in 2 mL Type 1 glass vial, rubber stoppered, and sealed.
  • the drug product vials were exposed up to five freeze-thaw cycles (i.e., IX F/T, 3X F/T, and 5X F/T); each cycle consisting of freezing at -80°C following by thawing at 25°C (25°C, 60% RH chamber) with 5°C control.
  • the samples were analyzed for visual appearance, protein concentration (UV280), turbidity (OD350-500), purity by UP-SEC, and charge variants by HP-IEX.
  • the lead anti-ILT4 L-histidine formulation did not show any change up to 5X freezethaw as shown in Table 15.
  • the visual appearance, turbidity (OD350-500), subvisible particulate counts, and biochemical profiles (UP-SEC and HP-IEX) were comparable to up to 5X freezethaw stress.
  • turbidity OD350-500
  • subvisible particulate counts UP-SEC and HP-IEX
  • biochemical profiles UP-SEC and HP-IEX
  • the anti-lLT4 monoclonal antibody formulation properties such as density, viscosity, osmolality, and apparent glass transition temperature (T g ’), were determined.
  • Modulated differential scanning microcalorimetry was used for determination of T g .
  • the following parameters were used during the run: temperature equilibrated at -90.0°C; modulation of ⁇ 0.5°C every 60 seconds, isothermal for 5.0 mins with ramp up of 2.0°C/min to 25.0°C.
  • the Advanced Instruments, Inc. Osmometer was calibrated using two calibration standards (100 mOsm/kg and 500 mOsm/kg) provided by Advanced Instruments, Inc., which were measured to be 100 mOsm/kg and 500 mOsm/kg, respectively (within the acceptable limits). 250 pL of sample was taken in the sample dispenser and the osmolality was measured in triplicate.
  • Anton Paar Densitometer was used for density measurement.
  • the oscillating U-tube of the densitometer was rinsed with water followed by ethanol and air-dried using in-built pump.
  • the air density in the oscillating U-tube was measured to be 0.00126 g/cm 3 at 20°C prior to sample density measurement.
  • 1 mL of the sample was filled into a sterile 1 mL syringe.
  • the sample was checked for air bubbles and gently inserted via sample port into the oscillating U-tube.
  • the densities were measured at 5°C, 20°C, and 30°C.
  • Rheosense mVROC viscometer was used for viscosity measurement. The water bath was stabilized at 20°C and a system check was performed using aquet solution. About 400 pL of sample was taken into a clean, 0.5 mL syringe and locked in the system for measurement. The viscosity was measured at 20°C using flow-rate of 50 pL/min with measurement time of 60 seconds. Viscosity values with slope fit Rsqrd value of at least 0.98 were used for recording.
  • EXAMPLE 10 Ion Exchange (1EX) method to measure acidic species of anti-lLT4 antibodies
  • IEX Ion Exchange
  • LC system Waters Alliance LC system (Milford, MA, U.S.A.)
  • Thermo Scientific’s ProPac WCX-10 p/n: 054993, particle size 10 um, diameter 4 mm, length 250 mm
  • Mobile Phase (A) 24 mM MES pH 6.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention relates to stable formulations of antibodies or antigen-binding fragments thereof that bind to human immunoglobulin-like transcript 4 (ILT4). Also provided are methods of treating various cancers using the formulations disclosed herein.

Description

PHARMACEUTICAL FORMULATIONS OF AN ANT1-1LT4 ANTIBODY OR ANTIGEN¬
BINDING FRAGMENT THEREROF AND METHODS OF USE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of priority to U.S. Provisional Application No. 63/336,670, filed April 29, 2022, the disclosure of which is incorporated herein by its entirety.
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY [0002] The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. The XML file, created on March 21, 2023, is named 25401_WO_PCT_SL.XML and is 131,718 bytes in size.
FIELD OF THE INVENTION
[0003] The invention relates to pharmaceutical formulations of an antibody or antigen-binding fragment thereof that binds to human interleukin-like transcript 4 (ILT4) and their use in treating various disorders (e.g., cancer).
BACKGROUND OF THE INVENTION
[0004] A common strategy used by tumor cells to escape innate and adaptive immune response is associated with aberrant expression of human leukocyte antigen (HLA)-G (Curigliano et al. Clin Cancer Res. 2013; 19(20): 5564-5571 and Gonzalez et al. Crit Rev Clin Lab Sci. 2012;
49(3): 63-84). HLA-G can directly inhibit immune cell function through receptor binding and/or through trogocytosis and impairment of chemotaxis (Morandi et al. Cytokine Growth Factor Review. 2014, 25: 327-335 and Lin et al. Mol Med. 2015, 21 : 782-791). High expression of HLA-G in multiple tumor types, including for example, colorectal, pancreatic, endometrial, lung, breast, ovarian, and gastric cancer, is associated with advanced disease stage, tumor invasiveness, metastatic potential and an unfavorable prognosis (Lin et al. Mol Med. 2015, 21 : 782-791 and Loumange et al. Int J Cancer. 2014, 135, 222: 581-597). Antibody-mediated blockade of HLA-G function in transgenic mouse models has been shown to inhibit tumor development and block expansion of myeloid-derived suppressor cells (MDSC) (Loumange et al. Int J Cancer. 2014, 135, 222: 581-597, Lin et al. Hum Immunol. 2013, 74: 439-446, and Agaugue et al. Blood. 2011, 117: 7021-7031). HLA-G binding to ILT4 can directly inhibit the function of monocytes, dendritic cells, and neutrophils, thus impairing the innate immune antitumor response. The interaction between HLA-G and monocytes due to ILT4 inhibits maturation of human monocyte-derived antigen-presenting cells (APCs) resulting in a reduced expression of MHC class II antigens and co-stimulatory molecules through Stat3 activation (Colonna et al. J Immunol. 1998, 160: 3096-3100; Allan et al. J Exp Med. 1999, 189(7): 1149-1155, and Liang et al. Proc Natl Sci USA. 2008, 105(24): 8357). Using human monocyte-derived dendritic cells (DCs) and ILT4-transgenic mice, HLA-G was shown to induce the development of tolerogenic APCs with arrest maturation/activation of myeloid DCs, and the induction of tolerogenic DCs by HLA-G was through disrupting the MHC class II presentation pathway (Ristich et al. Eur J Immunol. 2005, 35: 1133-1142).
[0005] An unmet medical need exists for patients having cancer that do not respond to T-cell therapy but may benefit from relief of tissue associated-macrophage/ MDSC-mediated tumor tolerance (e.g., myeloid “rich” tumors). ILT4 blockade would fill this need and would differentiate from current T-cell-targeted antibodies (e.g., anti-PDl, anti-TIGIT) by relieving suppression of tolerogenic myeloid cells in the tumor microenvironment.
SUMMARY OF THE INVENTION
[0006] In one aspect, provided herein is a formulation for an anti-human ILT4 (anti-ILT4) antibody or antigen-binding fragment thereof, comprising: (i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM of a buffer; (iii) about 6% to about 8% weight /volume (w/v) of a non-reducing sugar;
(iv) about 0.01 % to about 0.10% (w/v) of a non-ionic surfactant; and (v) about 1 mM to about 20 mM of an anti-oxidant, wherein the anti-lLT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21), wherein the buffer is an L-histidine buffer, an acetate buffer, or a citrate buffer, wherein the non-reducing sugar is a disaccharide, wherein the non-ionic surfactant is polysorbate 20 or polysorbate 80, and wherein the antioxidant is methionine.
[0007] In certain embodiments, the buffer is a selected from the group consisting of L-histidine buffer, acetate buffer, and citrate buffer. In one embodiment, the buffer is L-histidine buffer. In another embodiment, the buffer is acetate buffer. In yet another embodiment, the buffer is citrate buffer.
[0008] In some embodiments, the non-reducing sugar is sucrose.
[0009] In certain embodiments, the non-ionic surfactant is polysorbate 80 (PS-80) or polysorbate 20 (PS-20). In one embodiment, the non-ionic surfactant is PS-80. In another embodiment, the non-ionic surfactant is PS-20.
[0010] In some embodiments, the anti-oxidant is L-methionine.
[0011] Thus, in another aspect, provided herein is a formulation for an anti-ILT4 antibody or antigen-binding fragment thereof, comprising: (i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM L- histidine buffer; (iii) about 6% to about 8% (w/v) sucrose; (iv) about 0.01 % to about 0.10% (w/v) PS-80; and (v) about 1 mM to about 20 mM L-methionine, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR- Hl : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2:
GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0012] In some embodiments, the formulation comprises about 8 mM to about 12 mM L- histidine buffer.
[0013] In certain embodiments, the formulation comprises about 5 mM to about 10 mM L- methionine.
[0014] In other embodiments, the formulation comprises about 0.01 % to about 0.05% (w/v) PS-80.
[0015] In yet other embodiments, the formulation comprises about 10 mg/mL to about 150 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof. In still other embodiments, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL, about 12.5 mg/mL, about 15 mg/mL, about 25 mg/mL, about 50 mg/mL, about 75 mg/mL, about 100 mg/mL, about 125 mg/mL, or about 150 mg/mL. In one embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL. In another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 12.5 mg/mL. In yet another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 15 mg/mL. In still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 25 mg/mL. In one embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 50 mg/mL. In another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 75 mg/mL. In yet another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 100 mg/mL. In still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 125 mg/mL. In yet still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 150 mg/mL. In specific embodiments, the anti-ILT4 antibody or antigen-binding fragment thereof is a monoclonal antibody.
[0016] Thus, in one specific embodiment, the formulation comprises about 25 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0017] In another specific embodiment, the formulation comprises about 50 mg/mL of the anti- ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0018] In yet another specific embodiment, the formulation comprises about 75 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0019] In still another specific embodiment, the formulation comprises about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0020] In yet still another specific embodiment, the formulation comprises about 125 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0021] In certain embodiments of various formulations provided herein, the formulation has a pH range of from about 5.0 to about 6.8. In some embodiments, the formulation has a pH range of from about 5.5 to about 6.0. In other embodiments, the formulation has a pH of about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, or about 6.0. In one embodiment, the formulation has a pH of about 5.5. In another embodiment, the formulation has a pH of about 5.6. In yet another embodiment, the formulation has a pH of about 5.7. In still another embodiment, the formulation has a pH of about 5.8. In another embodiment, the formulation has a pH of about 5.9. In yet still another embodiment, the formulation has a pH of about 6.0.
[0022] Thus, in one specific embodiment, the pharmaceutical formulation for an anti-ILT4 antibody or antigen-binding fragment thereof comprises: (i) about 50 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer, pH about 5.5; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) polysorbate 80; and (v) about 10 mM L-methionine, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0023] In certain embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein Xi is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0024] In some embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GDSNRPS(SEQ ID NO: 52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0025] In other embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58.
[0026] In yet other embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:2 and a light chain comprising the amino acid sequence set forth in SEQ ID NOY. [0027] In still other embodiments of the various formulations provided herein, the anti-lLT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 80 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 7.
[0028] In certain embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:58.
[0029] In some embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO:2 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
[0030] In certain embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof is a monoclonal antibody comprising two heavy chains comprising the amino acid sequence set forth in SEQ ID NO:2 and two light chains comprising the amino acid sequence set forth in SEQ ID NO:7.
[0031] In certain embodiments of the various fonnulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof is a monoclonal antibody comprising two heavy chains consisting of the amino acid sequence set forth in SEQ ID NO:2 and two light chains consisting of the amino acid sequence set forth in SEQ ID NO:7.
[0032] In other embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO: 80 and a light chain consisting of the ammo acid sequence set forth in SEQ ID NO:7.
[0033] In certain embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof is a monoclonal antibody comprising two heavy chains comprising the amino acid sequence set forth in SEQ ID NO: 80 and two light chains comprising the amino acid sequence set forth in SEQ ID NO:7.
[0034] In certain embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof is a monoclonal antibody comprising two heavy chains consisting of the amino acid sequence set forth in SEQ ID NO: 80 and two light chains consisting of the amino acid sequence set forth in SEQ ID NO:7.
[0035] In one embodiment, the formulation is a liquid formulation. In one embodiment, the formulation is a frozen formulation. In another embodiment, the liquid formulation is stored at about 5°C. In yet another embodiment, the formulation is stored frozen at about -20°C or below. In yet another embodiment, the formulation is stored frozen at about -70°C or below. In still another embodiment, the liquid formulation is a reconstituted solution from a lyophilized formulation.
[0036] In certain embodiments, after the formulation is stored at about 3°C to about 5°C (e.g., 3°C, 4°C, 5°C) for 6 months, (i) the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultra-performance size exclusion chromatography; (ii) the turbidity of the formulation is at most about 0.135 as measured by OD350-500; (iii) the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, and/or the basic variant of the anti-ILT4 antibody or antigenbinding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography; (iv) the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging; and/or (v) the % oxidation of one or more amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
[0037] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultra-performance size exclusion chromatography.
[0038] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the turbidity of the formulation is at most about 0.135 as measured by OD350-500.
[0039] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, as determined by high performance ion-exchange chromatography. In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, as determined by high performance ion-exchange chromatography. In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography. In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, the % acidic variant of the anti-lLT4 antibody or antigen-binding fragment thereof is at most about 23%, and the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography.
[0040] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging.
[0041] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of one or more amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, the amino acid residue is W7 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80. In another embodiment, the amino acid residue is W102 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80. In yet another embodiment, the amino acid residue is M253 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80. In still another embodiment, the amino acid residue is M359 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80. In yet still another embodiment, the amino acid residue is M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80.
[0042] In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of one amino acid residue selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of two amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of three amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of four ammo acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of all five amino acid residues from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
[0043] In yet another aspect, provided herein is a method of treating a cancer in a human patient in need thereof, comprising administering a therapeutically effective amount of the pharmaceutical formulation described herein.
[0044] In still another aspect, provided herein is use of the pharmaceutical formulation described herein for preparing a medicament for treating a cancer.
BRIEF DESCRIPTION OF THE DRAWINGS
[0045] FIGS. 1A-1I show early stability studies of various anti-ILT4 formulations, including turbidity (OD350-500) (FIGS. 1A-1C), % HMW (high molecular weight) (FIGS. 1D-1F), or % monomer (FIGS. 1G-1I) at 5°C (FIGS. 1A, ID, 1G), 25°C (FIGS. IB, IE, 1H), or 40°C (FIGS 1C, IF, II) storage conditions, as described in Example 4.
[0046] FIGS. 2A-2I show early stability studies of various anti-ILT4 formulations, including % acidic variants (FIGS. 2A-2C), % main peak (FIGS. 2D-2F), or % basic variants (FIGS. 2G-2I) at 5°C (FIGS. 2A, 2D, 2G), 25°C (FIGS. 2B, 2E, 2H), or 40°C (FIGS. 2C, 2F, 21) storage conditions, as described in Example 4.
[0047] FIGS. 3A-3I show pH ranging stability studies of various anti-ILT4 formulations, including turbidity (OD350-500) (FIGS. 3A-3C), % HMW (FIGS. 3D-3F), or % monomer (FIGS. 3G-31) at 5°C (FIGS. 3A, 3D, 3G), 25°C (FIGS. 3B, 3E, 3H), or 40°C (FIGS. 3C, 3F, 31) storage conditions, as described in Example 5.
[0048] FIGS. 4A-4I show pH ranging stability studies of various anti-ILT4 formulations, including % acidic variants (FIGS. 4A-4C), % main peak (FIGS. 4D-4F), or % basic variants (FIGS. 4G-4I) at 5°C (FIGS. 4A, 4D, 4G), 25°C (FIGS. 4B, 4E, 4H), or 40°C (FIGS. 4C, 4F, 41) storage conditions, as described in Example 5.
[0049] FIGS. 5A-5G show surfactant ranging stability studies of various anti-ILT4 formulations, including turbidity (OD350-500) (FIG. 5A), % HMW (FIG. 5B), % monomer (FIG. 5C), subvisible particle counts before initiation of agitation (FIG. 5D), after 3 days agitation (FIG. 5E), after 7 days agitation (FIG. 5F), or after 7 days ambient control (FIG. 5G), as described in Example 6. [0050] FIGS. 6A-6K show light stress stability studies of various anti-lLT4 formulations, including turbidity (OD350-500) (FIG. 6A), % HMW (FIG. 6B), % monomer (FIG. 6C), % acidic variants (FIG. 6D), % main peak (FIG. 6E), % basic variants (FIG. 6F), % oxidation of M359 (FIG. 6G), M253 (FIG. 6H), M429 (FIG. 61), W102 (FIG. 6J), or W7 (FIG. 6K), as described in Example 7.
[0051] FIG 7 shows the initial and 3 days agitation results from a surfactant ranging study, as described in Example 6.
[0052] FIG 8 shows the 7 days agitation and 7 day ambient control results from a surfactant ranging study, as described in Example 6.
[0053] FIG 9 shows results from a light stress study with antioxidant L-methionine, as described in Example 7.
DETAILED DESCRIPTION OF THE INVENTION
Definitions and Abbreviations
[0054] As used throughout the specification and appended claims, the following abbreviations apply:
API active pharmaceutical ingredient
CDR complementarity determining region in the immunoglobulin variable regions, defined using the Kabat numbering system, unless otherwise indicated
CE-SDS capillary electrophoresis sodium dodecyl sulfate
CHO Chinese hamster ovary clEF capillary' isoelectric focusing
DLS dynamic light scattering test
DSC differential scanning calorimetry
DSF differential scanning fluorimetry
ELISA enzyme-linked immunosorbent assay
FR framework region
HC heavy chain
HMW high molecular weight
HP-IEX high performance ion-exchange chromatography
ICH International Council on Harmonization
IEX Ion exchange IgG immunoglobulin G
LC liquid chromatography
LC-MS liquid chromatography -mass spectrometry
LMW low molecular weight mAb monoclonal antibody
NR-CE-SDS non-reduced capillary electrophoresis sodium dodecyl sulfate
PS20 (or PS-20) polysorbate 20
PS80 (or PS-80) polysorbate 80
R-CE-SDS reducing capillary electrophoresis sodium dodecyl sulfate
RH relative humidity
UP-SEC ultra-performance size exclusion chromatography
VH immunoglobulin heavy chain variable region
VL immunoglobulin light chain variable region v/v volume per volume
WFI water for injection w/v weight per volume
[0055] Unless specifically defined elsewhere in this document, all technical and scientific temrs used herein have the meaning commonly understood by one of ordinary skill in the art to which this invention belongs.
[0056] As used throughout the specification and in the appended claims, the singular forms “a,”
“an,” and “the” include the plural reference unless the context clearly dictates otherwise.
[0057] Reference to “or” indicates either or both possibilities unless the context clearly dictates one of the indicated possibilities. In some cases, “and/or” was employed to highlight either or both possibilities.
[0058] As used herein, “acidic variant” refers to the anti-ILT4 antibody that is more acidic (e.g., as determined by cation exchange chromatography) than the anti-ILT4 antibody main species. Such acidic variations are detected by various chromatography purification methods for separating molecule variants by charge, such as ion exchange, for example, cation exchange chromatography or WCX-10 HPLC (a weak cation exchange chromatography), optionally followed by mass spectroscopy. Generally, the acidic variant has a lower isoelectric point (pl) than the main species, and can have a more acidic character due to for example, methionine oxidation, sialylation of asparagine residues or deamidated variants of the antibody, or a combination thereof. In one embodiment, the anti-ILT4 antibody acidic variants are the anti-ILT4 antibody species as identified by the acidic variants peak(s) in Figure 6D, and eluted according to the cation ion exchange method described in Example 10. In an ion exchange chromatography method, the “% acidic variants species” refers to the total area of acidic variants peak(s) divided by the total area of all peaks in the elution chromatogram. In one embodiment, the anti-ILT4 antibody acidic variant is as identified by peak(s) eluted prior to the main peak according to a cation ion exchange method. In another embodiment, the anti-ILT4 antibody acidic variant is as identified by peak(s) eluted prior to the main peak according to a weak cation ion exchange method. In an ion exchange chromatography method, the “% acidic variant” refers to the total area of acidic species peaks divided by the total area of all peaks in the elution chromatogram. [0059] “Affinity” refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, “binding affinity” refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including KinExA and Biacore.
[0060] As used herein, the term “antibody” includes, but is not limited to, monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), fully human antibodies, and chimeric antibodies.
[0061] As used herein, unless otherwise indicated, “antigen-binding fragment” refers to antigen-binding fragments of antibodies, i. e. , antibody fragments that retain the ability to bind to the antigen bound by the full-length antibody, e.g., fragments that retain one or more CDR regions. Examples of antibody binding fragments include, but are not limited to. Fab, Fab’, F(ab’)2, Fv fragments and individual antibody heavy chains or light chains, and individual heavy chain or light chain variable regions.
[0062] As used herein, “basic variant” refers to the anti-ILT4 antibody that is more basic (e.g., as determined by cation exchange chromatography) than the anti-ILT4 antibody main species. Such basic variants are detected by various chromatography purification methods for separating molecule variants by charge, such as ion exchange, for example, cation exchange chromatography (e.g., the method described in Example 10 or WCX-10 HPLC (a weak cation exchange chromatography), optionally followed by mass spectroscopy. Generally, the basic species has a higher pH than the main species, and can have a more basic character due to modifications or differences from the main species. In one embodiment, the anti-ILT4 antibody basic species is as identified by peak(s) eluted after the mam peak according to a cation ion exchange method. In another embodiment, the anti-ILT4 antibody basic species is as identified by peak(s) eluted after the main peak according to a weak cation ion exchange method. In an ion exchange chromatography method, the “% basic species” refers to the total area of basic species peak(s) divided by the total area of all peaks in the elution chromatogram.
[0063] A “Fab fragment” is comprised of one light chain and the CHI and variable regions of one heavy chain. The heavy chain of a Fab molecule cannot form a disulfide bond with another heavy chain molecule. An “Fab fragment” can be the product of papain cleavage of an antibody.
[0064] An “Fc” region contains two heavy chain fragments comprising the CHI and CH2 domains of an antibody. The two heavy chain fragments are held together by two or more disulfide bonds and by hydrophobic interactions of the CH3 domains.
[0065] A “Fab’ fragment” contains one light chain and a portion or fragment of one heavy chain that contains the VH domain and the CHI domain and also the region between the CHI and CH2 domains, such that an interchain disulfide bond can be formed between the two heavy chains of two Fab’ fragments to form a F(ab’)2molecule.
[0066] A “F(ab’)2 fragment” contains two light chains and two heavy chains containing a portion of the constant region between the CHI and CH2 domains, such that an interchain disulfide bond is formed between the two heavy chains. A F(ab’)2 fragment thus is composed of two Fab’ fragments that are held together by a disulfide bond between the two heavy chains. An “F(ab’)2 fragment” can be the product of pepsin cleavage of an antibody.
[0067] The “Fv region” comprises the variable regions from both the heavy and light chains, but lacks the constant regions.
[0068] “Isolated antibody” refers to the purification status and in such context means the molecule is substantially free of other biological molecules such as nucleic acids, proteins, lipids, carbohydrates, or other material such as cellular debris and growth media. Generally, the term “isolated” is not intended to refer to a complete absence of such material or to an absence of water, buffers, or salts, unless they are present in amounts that substantially interfere with experimental or therapeutic use of the binding compound as described herein.
[0069] As used herein, “main species” or “main” refers to the anti-ILT4 antibody species identified as the majority of the antibody species in a mixture with one or more acidic or basic species thereof. Such main species are detected by various chromatography purification methods for separating molecule variants by charge, such as ion exchange, for example, cation exchange chromatography (e.g., the method described in Example 10) or WCX-10 HPLC (a weak cation exchange chromatography), optionally followed by mass spectroscopy. The mixture can be a result of for example, antibody preparations from mammalian cells and post-translational modifications thereof, upstream and downstream processing, or storage. In one embodiment, the main species is identified as the main peak according to a cation ion exchange method. In an ion exchange method, the “% main” refers to the total area of main peak divided by the total area of all peaks in the elution chromatogram. In one aspect of measuring the main species, acidic species or basic species, a Thermo Scientific ProPac WCX-10 column is used for the cation ion exchange method. In another embodiment, a Thermo Scientific ProPac WCX-10 column is used, with a Mobile Phase (A) 24 mM MES pH 6. 1 with 4% acetonitrile, and mobile phase (B) 20 mM sodium phosphate, 95 mM NaCl pH 8.0 with 4% acetonitrile, and a column temperature of 35 °C. In one embodiment, a non-linear gradient is used with: 22%-22%B for 0-0.6 min; 22%- 29%B for 0.6-15.0 min; 29%-70%B for 15.0-30.0 min; 70%-100%B for 30.0-30.5 min; and 100%-100%B from 30.5-33.0 min. In a further embodiment, the cation ion exchange method is described in Example 10.
[0070] The term “monoclonal antibody”, as used herein, refers to a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts. In contrast, conventional (polyclonal) antibody preparations typically include a multitude of different antibodies having different amino acid sequences in their variable domains that are often specific for different epitopes. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the invention may be made by the hybridoma method first described by Kohler et al. (1975) Nature 256: 495, or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567). The “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol. Biol. 222: 581-597, for example. See also Presta (2005) J. Allergy Clin. Immunol. 116:731.
[0071] The term “fully human antibody” or “human antibody” refers to an antibody that comprises human immunoglobulin protein sequences only. A fully human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell. Similarly, “mouse antibody” refers to an antibody that comprises mouse immunoglobulin sequences only. Alternatively, a fully human antibody may contain rat carbohydrate chains if produced in a rat, in a rat cell, or in a hybndoma derived from a rat cell. Similarly, “rat antibody” refers to an antibody that comprises rat immunoglobulin sequences only.
[0072] In general, the basic “antibody” structural unit comprises a tetramer. In a monospecific antibody, each tetramer includes two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa). The amino-terminal portion of each chain includes a “variable region” or “variable domain” of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy -terminal portion of the heavy chain may define a constant region primarily responsible for effector function.
[0073] Typically, human constant light chains are classified as kappa and lambda light chains. Furthermore, human constant heavy chains are typically classified as mu, delta, gamma, alpha, or epsilon, and define the antibody’s isotype as IgM, IgD, IgG, IgA, and IgE, respectively. Subtypes of these IgG include, for example, IgGl and IgG4. The invention includes anti-ILT4 antibodies and antigen-binding fragments comprising any of these light and/or heavy constant chains.
[0074] “Variable region,” “variable domain, “ “V region,” or “V chain” as used herein means the segment of IgG chains which is variable in sequence between different antibodies. A “variable region” of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination. The variable region of the heavy chain may be referred to as “VH.” The variable region of the light chain may be referred to as “VL.” Typically, the variable regions of both the heavy and light chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), which are located within relatively conserved framework regions (FR). The CDRs are usually aligned by the framework regions, enabling binding to a specific epitope. In general, from N -terminal to C- terminal, both light and heavy chains variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. The assignment of amino acids to each domain is, generally, in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al:, National Institutes of Health, Bethesda, Md.; 5th ed.; NIH Publ. No. 91-3242 (1991); Kabat (1978) Adv. Prot. Chem. 32: 1-75; Kabat, etal., (1977) J. Biol. Chem. 252:6609-6616; Chothia, etal., (1987) J Mol. Biol. 196:901-917 or Chothia, et al., (1989) Nature 342:878-883.
[0075] A “CDR” refers to one of three hypervariable regions (Hl, H2, or H3) within the nonframework region of the antibody VH P-sheet framework, or one of three hypervariable regions (LI, L2, or L3) within the non-framework region of the antibody VL P-sheet framework. Accordingly, CDRs are variable region sequences interspersed within the framework region sequences. CDR regions are well known to those skilled in the art and have been defined by, for example, Kabat as the regions of most hypervariability within the antibody variable domains. CDR region sequences also have been defined structurally by Chothia as those residues that are not part of the conserved -sheet framework, and thus are able to adapt to different conformation. Both terminologies are well recognized in the art. CDR region sequences have also been defined by AbM, Contact, and IMGT. The positions of CDRs within a canonical antibody variable region have been determined by comparison of numerous structures (Al-Lazikani et al., 1997, J. Mol. Biol. 273:927-48; Morea et al., 2000, Methods 20:267-79). Because the number of residues within a hypervariable region varies in different antibodies, additional residues relative to the canonical positions are conventionally numbered with a, b, c and so forth next to the residue number in the canonical variable region numbering scheme (Al-Lazikani et al., supra). Such nomenclature is similarly well known to those skilled in the art. Correspondence between the numbering system, including, for example, the Kabat numbering and the IMGT unique numbering system, is well known to one skilled in the art and shown below' in Table 1. In some embodiments, the CDRs are as defined by the Kabat numbering system. In other embodiments, the CDRs are as defined by the IMGT numbering system. In yet other embodiments, the CDRs are as defined by the AbM numbering system. In still other embodiments, the CDRs are as defined by the Chothia numbering system. In yet other embodiments, the CDRs are as defined by the Contact numbering system.
Table 1. Correspondence between the CDR Numbering Systems
Figure imgf000017_0001
[0076] Sequence identity refers to the degree to which the amino acids of two polypeptides are the same at equivalent positions when the two sequences are optimally aligned [0077] Sequence similarity includes identical residues and non-identical, biochemically related amino acids. Biochemically related amino acids that share similar properties and may be interchangeable are discussed above.
[0078] “Conservatively modified variants” or “conservative substitution” refers to substitutions of amino acids in a protein with other amino acids having similar characteristics (e.g., charge, side-chain size, hydrophobicity /hydrophilicity, backbone conformation and rigidity, etc.), such that the changes can frequently be made without altering the biological activity of the protein. Those of skill in this art recognize that, in general, single amino acid substitutions in non- essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)). In addition, substitutions of structurally or functionally similar amino acids are less likely to disrupt biological activity. Exemplary conservative substitutions are set forth in Table 2.
Table 2. Exemplary Conservative Amino Acid Substitutions
Figure imgf000018_0001
Figure imgf000019_0001
[0079] The term “epitope,” as used herein, refers to an area or region on an antigen to which an antibody or antigen-binding fragment binds. Binding of an antibody or antigen-binding fragment thereof disclosed herein to an epitope means that the antibody or antigen-binding fragment thereof binds to one or more amino acid residues within the epitope.
[0080] “Treat” or “treating” a cancer or an infectious condition as used herein means to administer a formulation of the invention to a subject having an immune condition or cancerous condition, or diagnosed with a cancer or pathogenic infection (e.g., viral, bacterial, fungal), to achieve at least one positive therapeutic effect, such as for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral organs, or reduced rate of tumor metastasis or tumor growth. “Treatment” may include one or more of the following: inducing/increasing an antitumor immune response, stimulating an immune response to a pathogen, toxin, and/or self-antigen, stimulating an immune response to a viral infection, decreasing the number of one or more tumor markers, inhibiting the growth or survival of tumor cells, eliminating or reducing the size of one or more cancerous lesions or tumors, decreasing the level of one or more tumor markers, ameliorating, reducing the severity or duration of cancer, prolonging the survival of a patient relative to the expected survival in a similar untreated patient. [0081] The term “cancer”, “cancerous”, or “malignant” refers to or describes the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include, but are not limited to, carcinoma, lymphoma, leukemia, blastoma, and sarcoma. More particular examples of such cancers include squamous cell carcinoma, myeloma, small-cell lung cancer, non-small cell lung cancer, glioma, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, gastrointestinal (tract) cancer, renal cancer, ovarian cancer, liver cancer, lymphoblastic leukemia, lymphocytic leukemia, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, melanoma, chondrosarcoma, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, brain cancer, stomach cancer, bladder cancer, hepatoma, breast cancer, colon carcinoma, and head and neck cancer.
[0082] The term “patient” (alternatively referred to as “subject” or “individual” herein) refers to a mammal (e.g., rat, mouse, dog, cat, rabbit) capable of being treated with the formulations of the invention, most preferably a human. In some embodiments, the patient is an adult patient. In other embodiments, the patient is a pediatnc patient. A patient “in need of treatment” is an individual diagnosed with, suspected of having, or predisposed to a disease or disorder in which a formulation of the invention is intended to treat or ameliorate (e.g., an ILT4-related disease such as cancer), or a patient for whom prevention of such a disorder is desired.
[0083] The term “therapeutically effective amount” or “effective amount” means an amount whereby sufficient therapeutic composition or formulation is introduced to a patient to treat a diseased or condition. One skilled in the art recognizes that this level may vary according to the patient’s characteristics such as age, weight, etc.
[0084] The term “about”, when modifying the quantity (e.g., mM, or M) of a substance or composition, the percentage (v/v or w/v) of a formulation component, the pH of a solution/formulation, or the value of a parameter characterizing a step in a method, or the like refers to variation in the numerical quantity that can occur, for example, through typical measuring, handling, and sampling procedures involved in the preparation, characterization and/or use of the substance or composition; through instrumental error in these procedures; through differences in the manufacture, source, or purity of the ingredients employed to make or use the compositions or carry out the procedures; and the like. In certain embodiments, “about” can mean a variation of ± 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, or 10%.
[0085] As used herein, “x% (w/v)” is equivalent to x g/100 ml (for example 5% w/v equals 50 mg/ml).
[0086] The phrase “consists essentially of,” or variations such as “consist essentially of’ or “consisting essentially of,” as used throughout the specification and claims, indicate the inclusion of any recited elements or group of elements, and the optional inclusion of other elements, of similar or different nature than the recited elements, that do not materially change the basic or novel properties of the specified dosage regimen, method, or composition. As a non-limiting example, a binding compound that consists essentially of a recited amino acid sequence may also include one or more amino acids, including substitutions of one or more amino acid residues, that do not materially affect the properties of the binding compound.
[0087] “Comprising” or variations such as “comprise” or “comprises” are used throughout the specification and claims in an inclusive sense, i.e., to specify the presence of the stated features but not to preclude the presence or addition of further features that may materially enhance the operation or utility of any of the embodiments of the invention, unless the context requires otherwise due to express language or necessary implication. [0088] The term “buffer” encompasses those agents which maintain the solution pH of the formulations in an acceptable range, or, for lyophilized formulations of the invention, provide an acceptable solution pH prior to lyophilization.
[0089] The terms “lyophilization,” “lyophilized,” and “freeze-dried” refer to a process by which the material to be dried is first frozen and then the ice or frozen solvent is removed by sublimation in a vacuum environment. Excipients may be included in pre-lyophilized formulations to enhance stability of the lyophilized product upon storage.
[0090] The term “pharmaceutical formulation” refers to preparations which are in such a form as to permit the active ingredients to be effective, and which contains no additional components which are toxic to the subjects to which the formulation would be administered. The term “formulation” and “pharmaceutical formulation” are used interchangeably throughout.
[0091] “Pharmaceutically acceptable” refers to excipients (vehicles, additives) and compositions that can reasonably be administered to a subject to provide an effective dose of the active ingredient employed and that are “generally regarded as safe,” e.g., that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction, such as gastric upset and the like, when administered to a human. In another embodiment, this tenn refers to molecular entities and compositions approved by a regulatory agency of the federal or a state government or listed in the U.S. Pharmacopeia or another generally recognized pharmacopeia for use in animals, and more particularly in humans.
[0092] A “reconstituted” formulation is one that has been prepared by dissolving a lyophilized protein formulation in a diluent such that the protein is dispersed in the reconstituted formulation. The reconstituted formulation is suitable for administration, e.g., parenteral administration), and may optionally be suitable for subcutaneous administration.
[0093] A “stable” formulation is one in which the protein therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage or under stress conditions. Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs. (1991) and Jones, A. Adv. Drug Delivery Rev. 10:29-90 (1993). Stability can be measured at a selected temperature for a selected time period. For example, in one embodiment, a stable formulation is a formulation with no significant changes observed at a refrigerated temperature (2-8° C) for at least 6 months. In one embodiment, a stable formulation is a formulation with no significant changes observed at a refrigerated temperature (2-8° C) for at least 12 months. In another embodiment, a stable formulation is a formulation with no significant changes observed at a refrigerated temperature (2-8° C) for at least 24 months. In another embodiment, stable formulation is a formulation with no significant changes observed at room temperature (23-27°C) for at least 3 months. In another embodiment, stable formulation is a formulation with no significant changes observed at room temperature (23-27°C) for at least 6 months. In another embodiment, stable formulation is a formulation with no significant changes observed at room temperature (23-27°C) for at least 12 months. In another embodiment, stable formulation is a formulation with no significant changes observed at room temperature (23-27°C) for at least 18 months. The criteria for stability for an antibody formulation are as follows. Typically, no more than 10%, preferably 5%, of antibody monomer is degraded as measured by SEC-HPLC. Typically, the formulation is colorless, or clear to slightly opalescent by visual analysis. Typically, the concentration, pH, and osmolality of the formulation have no more than +/-10% change. Potency is typically within 60-140%, preferably 80-120% of the control or reference. Typically, no more than 10%, preferably 5% of clipping of the antibody is observed, i.e., % low molecular weight species as determined, for example, by HP-SEC. Typically, no more than 10%, preferably no more than 5% of aggregation of the antibody is observed, i.e., % high molecular weight species as determined, for example, by HP-SEC.
[0094] An antibody “retains its physical stability” in a pharmaceutical fomrulation, if it shows no significant increase of aggregation, precipitation, and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering, size exclusion chromatography (SEC) and dynamic light scattering. The changes of protein conformation can be evaluated by fluorescence spectroscopy, which determines the protein tertiary structure, and by FTIR spectroscopy, which determines the protein secondary structure.
[0095] An antibody “retains its chemical stability” in a pharmaceutical formulation, if it shows no significant chemical alteration. Chemical stability can be assessed by detecting and quantifying chemically altered forms of the protein. Degradation processes that often alter the protein chemical structure include hydrolysis or clipping (evaluated by methods such as size exclusion chromatography and SDS-PAGE), oxidation (evaluated by methods such as by peptide mapping in conjunction with mass spectroscopy or MALDI/TOF/MS), deamidation (evaluated by methods such as ion-exchange chromatography, capillary isoelectric focusing, peptide mapping, isoaspartic acid measurement), and isomerization (evaluated by measuring the isoaspartic acid content, peptide mapping, etc.).
[0096] An antibody “retains its biological activity” in a pharmaceutical formulation, if the biological activity of the antibody at a given time is within a predetermined range of the biological activity exhibited at the time the pharmaceutical formulation was prepared. The biological activity of an antibody can be determined, for example, by an antigen binding assay. [0097] The term “isotonic” means that the formulation of interest has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic pressure from about 270-328 mOsm. Slightly hypotonic pressure is 250-269 and slightly hypertonic pressure is 328-350 mOsm. Osmotic pressure can be measured, for example, using a vapor pressure or ice-freezing type osmometer.
[0098] Anti-ILT4 Antibodies and Antigen-Binding Fragments Thereof
[0099] The formulations disclosed herein can be used with any antibodies and antigen-binding fragments thereof (e.g., fully human antibodies) that bind to ILT4. In one embodiment, the anti- ILT4 antibody or antigen-binding fragment thereof is an antagonist. In another embodiment, the anti-ILT4 antibody or antigen-binding fragment thereof is an anti-ILT4 antibody or antigenbinding fragment thereof described herein (e.g., 1E1, 2A6, 3G7, or 2C1).
[0100] In one aspect, the formulations can be used with anti-ILT4 antibodies and antigenbinding fragments thereof as set forth herein having one or more of the properties set forth below:
• binds human ILT4 at one or more amino acid residues in LYREKKSASW (SEQ ID NO:59), TRIRPEL (SEQ ID NO:60), NGQF (SEQ ID NO:61), and/or HTGRYGCQ (SEQ ID NO:62), and/or protects TYREKKSASW (SEQ ID NO:59), TRIRPET (SEQ ID NO: 60), NGQF (SEQ ID NO: 1), and/or HTGRYGCQ (SEQ ID NO: 62) from deuterium (e.g., D2O) exchange, e.g, as detennined by hydrogen-deuterium exchange mass spectrometry;
• binds human ITT4 at domain 1 (see Wilcox et al. BMC Structural Biology 2:6 (2002));
• binds human ILT4 extracellular domain or TM form of ILT4 expressed on a cell surface, e.g., a pre-B cell, Chinese hamster ovary cell, U937 cell, or Jurkat JE6 cell.
• calculated pl -7.29 (e.g., 7.29 or 7.30);
• experimentally determined pl - 7.2;
• is characterized by a thermogram having Tm onset > 60°C, Tml- 65.2°C and Tm2 ~78.8°C;
• binds human ILT4 with a KD of about 1.7 X 10'8 M (e.g., as determined by surface plasmon resonance, e.g., binding of anti-ILT4 to polyhistidine tagged human ILT4);
• Ka=5.5 X 105 M'1 s'1 (e.g., as determined by surface plasmon resonance, e.g., binding of anti-ILT4 to polyhistidine tagged human ILT4); • Kd=9X10'3 s'1(e.g., as determined by surface plasmon resonance, e.g., binding of anti- ILT4 to polyhistidine tagged human ILT4);
• blocks binding of HLA-G (e.g., Fc fused HLA-G) to human ILT4 (e.g., ILT4 on mouse 3A9 T cells transfected with and expressing ILT4), e.g., with an IC50 of about 0.25 micrograms/ml (+0.06 micrograms/ml), e.g., as determined by surface plasmon resonance;
• blocks binding of HL A- A, HLA-B (e.g., fluorochrome labeled dexamers of HL A- A, such as HLA*A2:01 or HLA-B such as HLA*B7:02), and/or HLA-F (e.g, fluorochrome labeled tetramers of HLA-F) to ILT4 (e.g, ILT4 on mouse 3A9 T cells transfected with and expressing ILT4), e.g., as determined by surface plasmon resonance;
• blocks ILT4 (e.g., ILT4 on mouse 3A9 T cells transfected with and expressing ILT4), binding to ANGPTL1, ANGPTL4, and/or ANGPTL7 (e.g, biotinylated ANGPTL proteins), e.g, as determined by surface plasmon resonance;
• does not bind to ILT2, ILT3, ILT5, LILRB5, LILRA1, LILRA2, ILT7, ILT8, and/or ILT11;
• reverses ILT4-mediated suppression of IL2 in ILT4 transfected 3A9 cells, e.g. , with an EC50 of 0.43 micrograms/ml (±0.14 micrograms/ml);
• rescues ILT4:HLA-G induced suppression of mast cell degranulation (e.g., In the presence of plate-bound HLA-G tetramer), for example, wherein the mast cells express ILT4 and CD200RLa and are stimulated, for example, with antibody -mediated crosslinking of CD200RLa;
• enhances lipopolysaccharide (LPS)-induced expression of proinflammatory myeloid cytokines, for example, GM-CSF and/or TNF alpha, from a peripheral blood mononuclear cell (PBMC);
• enhances anti-CD3-induced expression of pro-inflammatory my eloid cytokines for example, GM-CSF and/or TNF alpha, from a peripheral blood mononuclear cell (PBMC);
• inhibits tumor growth in humans or, for example, in other mammals such as mice (e.g., Immuno-deficient NSG mice) which were reconstituted with human hematopoietic stem cells, for example, which harbor peripheral human CD45+ immune cells, for example, wherein the tumor is a human skin melanoma tumor such as from the cell line SKMEL5;
• relieves MDSC-mediated tumor tolerance in the body of a subject (e.g. , human subject) with a tumor;
• does not bind to cynomolgus monkey ILT4 and/or mouse pirB; • stains CD14+ human monocytes and/or CD11B+ human granulocytes; and/or
• binds to one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9 or all 10) of the human ILT4 haplotypes.
[0101] Antibody 1E1 (Q1E) heavy chain (IgG4)
Heavy chain
EVQLQOWGAGLLKPSETLSLTCAVYGGSFSGYYWSWIROPPGKGLEWIGEINHSGSTNY
NPSLKSRVTISVDTSKNOFSLKLSSVTAADTAVYYCARLPTRWVTTRYFDLWGRGTLVT
VSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL
QSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGG
PSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQF
NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQ
EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDK
SRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK
(SEQ ID NO:1; variable domain underscored; CDRs double underscored)
Heavy chain variable domain
EVQLQQWGAGLLKPSETLSLTCAVYGGSFSGYYWSWIRQPPGKGLEWIGEINHSGSTNY
NPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARLPTRWVTTRYFDLWGRGTLVT vss
(SEQ ID NO: 63)
[0102] Antibody 1E1 (Q1E, S54A) heavy chain (IgG4)
Heavy chain
EVQLQOWGAGLLKPSETLSLTCAVYGGSFSGYYWSWIROPPGKGLEWIGEINHAGSTNY
NPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARLPTRWVTTRYFDLWGRGTLVT
VSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL
QSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGG
PSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQF
NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQ
EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDK
SRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK
(SEQ ID NO:2; variable domain underscored; CDRs double underscored)
Heavy chain variable domain
EVQLQQWGAGLLKPSETLSLTCAVYGGSFSGYYWSWIRQPPGKGLEWIGEINHAGSTNY
NPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARLPTRWVTTRYFDLWGRGTLVT VSS (SEQ 1D NO:57)
[0103] Antibody 1E1 heavy chain (IgGl)
Heavy chain
QVQLQOWGAGLLKPSETLSLTCAVYGGSFSGYYWSWIROPPGKGLEWIGEINHSGSTNY
NPSLKSRVTISVDTSKNOFSLKLSSVTAADTAVYYCARLPTRWVTTRYFDLWGRGTLVT
VSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVL
QSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPEL
LGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPRE
EQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLP
PSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV
DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
(SEQ ID NO:44; variable domain underscored; CDRs double underscored)
Heavy chain variable domain
QVQLQQWGAGLLKPSETLSLTCAVYGGSFSGYYWSWIRQPPGKGLEWIGEINHSGSTNY
NPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARLPTRWVTTRYFDLWGRGTLVT vss
(SEQ ID NO: 69)
[0104] 1E1 heavy chain CDRs
CDR-H1: GYYWS (SEQ ID NO: 16)
CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17) (e.g.,
EINHSGSTNYNPSLKS (SEQ ID NO:47) or EINHAGSTNYNPSLKS(SEQ ID NO:48))
CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18)
[0105] Antibody 1E1 (Q1E) light chain (lambda)
Light chain
ESVLTQPPSVSGAPGORVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNSNRPSGV
PDRFSVSKSGASASLAITGLQAEDEADYYCOSFDNSLSAYVFGGGTOLTVLGQPKAAPS
VTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNKYA
ASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
(SEQ ID NO:3; variable domain underscored; CDRs double underscored)
Light chain variable domain
ESVLTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNSNRPSGV
PDRFSVSKSGASASLAITGLQAEDEADYYCQSFDNSLSAYVFGGGTQLTVL
(SEQ ID NO: 70) [0106] Antibody 1E1 (Q1E, S54A) light chain (lambda)
Light chain
ESVLTQPPSVSGAPGORVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNANRPSGV
PDRFSVSKSGASASLAITGLOAEDEADYYCOSFDNSLSAYVFGGGTOLTVLGOPKAAPS
VTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNKYA
ASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
(SEQ ID NO:4; variable domain underscored; CDRs double underscored)
Light chain variable domain
ESVLTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNANRPSGV
PDRFSVSKSGASASLAITGLQAEDEADYYCQSFDNSLSAYVFGGGTQLTVL
(SEQ ID NO:71)
[0107] Antibody 1E1 (Q1E, N53Q) light chain (lambda)
Light chain
ESVLTQPPSVSGAPGORVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGOSNRPSGV
PDRFSVSKSGASASLAITGLQAEDEADYYCOSFDNSLSAYVFGGGTQLTVLGQPKAAPS
VTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNKYA
ASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
(SEQ ID NO:5; variable domain underscored; CDRs double underscored)
Light chain variable domain
ESVLTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGQSNRPSGV
PDRFSVSKSGASASLAITGLQAEDEADYYCQSFDNSLSAYVFGGGTQLTVL
(SEQ ID NO: 72)
[0108] Antibody 1E1 (Q1E, N53E) light chain (lambda)
Light chain
ESVLTQPPSVSGAPGORVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGESNRPSGV
PDRFSVSKSGASASLAITGLQAEDEADYYCOSFDNSLSAYVFGGGTQLTVLGQPKAAPS
VTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNKYA
ASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
(SEQ ID NO: 6; variable domain underscored; CDRs double underscored)
[0109] Light chain variable domain
ESVLTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGESNRPSGV
PDRFSVSKSGASASLAITGLQAEDEADYYCQSFDNSLSAYVFGGGTQLTVL
(SEQ ID NO: 73) [0110] Antibody 1E1 (Q1E, N53D) light chain (lambda)
Light chain
ESVLTQPPSVSGAPGORVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGDSNRPSGV PDRFSVSKSGASASLAITGLOAEDEADYYCOSFDNSLSAYVFGGGTOLTVLGQPKAAPS VTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNKYA ASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
(SEQ ID NO: 7; variable domain underscored; CDRs double underscored)
Light chain variable domain
ESVLTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGDSNRPSGV
PDRFSVSKSGASASLAITGLQAEDEADYYCQSFDNSLSAYVFGGGTQLTVL (SEQ ID NO:58)
[0111] Antibody 1E1 light chain (lambda)
Light chain
OSVLTOPPSVSGAPGORVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNSNRPSGV PDRFSVSKSGASASLAITGLQAEDEADYYCOSFDNSLSAYVFGGGTQLTVLGQPKAAPS VTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNKYA ASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
(SEQ ID NO:45; variable domain underscored; CDRs double underscored)
Light chain variable domain
QSVLTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNSNRPSGV
PDRFSVSKSGASASLAITGLQAEDEADYYCQSFDNSLSAYVFGGGTQLTVL
(SEQ ID NO: 77)
[0112] 1E1 light chain CDRs
CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19)
CDR-L2: GX1X2NRPS; wherein Xi is N,Q,E or D and X2 is S or A (SEQ ID NO: 20) (e.g., GNSNRPS (SEQ ID NO:49), GQSNRPS (SEQ ID NO:50), GESNRPS (SEQ ID NO:51), GDSNRPS (SEQ ID NO:52), GNANRPS (SEQ ID NO: 53), GQANRPS (SEQ ID NO:54), GEANRPS (SEQ ID NO:55), or GDANRPS (SEQ ID NO:56))
CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21)
[0113] Antibodies and antigen-binding fragments thereof including the 1E1 heavy and light chain CDRs or the 1E1 VH and VL or the 1E1 heavy chain and light chain (or a variant thereof, e.g., as set forth herein) may be referred to as “1E1.”
[0114] Antibody 2A6 (Q1E) heavy' chain (IgG4) Heavy chain
EVQLVOSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMGGIIPIFGTAN
YAOKFOGRVTITADESTSTAYMELSSLRSEDTAVYYCARYFDSSGWYKGGAFDIWGQG
TMVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF
PAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPE
FLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPR
EEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTL
PPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLT
VDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK
(SEQ ID NO: 8; variable domain underscored; CDRs double underscored)
Heavy chain variable domain
EVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMGGIIPIFGTAN
YAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARYFDSSGWYKGGAFDIWGQG
TMVTVSS
(SEQ ID NO: 64)
[0115] Antibody 2A6 (Q1E, S102A, M119L) heavy chain (IgG4)
Heavy chain
EVQLVOSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGOGLEWMGGIIPIFGTAN
YAOKFOGRVTITADESTSTAYMELSSLRSEDTAVYYCARYFDASGWYKGGAFDIWGOG
TLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF
PAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPE
FLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPR
EEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKT1SKAKGQPREPQVYTL
PPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLT
VDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK
(SEQ ID NO: 9; variable domain underscored; CDRs double underscored)
Heavy chain variable domain
EVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMGGIIPIFGTAN
YAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARYFDASGWYKGGAFDIWGQG
TLVTVSS
(SEQ ID NO: 65)
[0116] Antibody 2A6 (Q1E, D101S, M119L) heavy chain (IgG4) Heavy chain EVQLVOSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMGGIIPIFGTAN YAOKFOGRVTITADESTSTAYMELSSLRSEDTAVYYCARYFSSSGWYKGGAFDIWGQG TLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF PAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPE FLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPR EEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTL PPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLT VDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK
(SEQ ID NO: 10; variable domain underscored; CDRs double underscored)
Heavy chain variable domain
EVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMGGIIPIFGTAN YAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARYFSSSGWYKGGAFDIWGQG TLVTVSS
(SEQ ID NO: 66)
[0117] The formulations disclosed herein can be used with antibodies comprising SEQ ID NO: 8, 9, 10, 64, 65, or 66, and antigen-binding fragments thereof, wherein residue 1 of SEQ ID NO: 8, 9, 10, 64, 65, or 66 is Q instead of E.
[0118] 2A6 heavy chain CDRs
CDR-H1: SYAIS (SEQ ID NO:22)
CDR-H2: GIIPIFGTANYAQKFQG (SEQ ID NO:23)
CDR-H3: YFX1X2SGWYKGGAFDI; wherein Xi is D or S and X2 is S or A (SEQ ID NO:24) (e.g., YFDSSGWYKGGAFD1 (SEQ ID NO:91), YFSSSGWYKGGAFD1 (SEQ ID NO:92), YFDASGWYKGGAFDI (SEQ ID NO:93), or YFSASGWYKGGAFDI (SEQ ID NO:94)) [0119] Antibody 2A6 light chain (lambda)
Light chain
OSVLTQPSSLSASPGASASLTCTLRSGINVDTYRIHWYOQKPGSPPQYLLRYKSDSDKHO GSGVPSRFSGSKDPSANAGILLISGLQSEDEADYYCAIWYSSTWVFGGGTOLTVLGQPKA APSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNK YAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
(SEQ ID NO: 11; variable domain underscored; CDRs double underscored) Light chain variable domain
QSVLTQPSSLSASPGASASLTCTLRSGINVDTYRIHWYQQKPGSPPQYLLRYKSDSDKHQ GSGVPSRFSGSKDPSANAG1EL1SGEQSEDEADYYCA1WYSSTWVFGGGTQLTVL
(SEQ ID NO: 74)
[0120] The formulations disclosed herein can be used with antibodies comprising SEQ ID
NO: 11 or 74 and antigen-binding fragments thereof, wherein residue 1 of SEQ ID NO: 11 or 74 is E instead of Q.
[0121] 2A6 light chain CDRs
CDR-L1: TLRSGINVDTYRIH (SEQ ID NO:25)
CDR-L2: YKSDSDKHQGS (SEQ ID NO:26)
CDR-L3: AIWYSSTWV (SEQ ID NO: 27)
[0122] Antibodies and antigen-binding fragments thereof including the 2A6 heavy and light chain CDRs or the 2A6 VH and VL or the 2A6 heavy chain and light chain (or a variant thereof, e.g., as set forth herein) may be referred to as “2A6.”
[0123] Antibody 3G7 (Q1E) heavy' chain (IgG4)
Heavy chain
EVQLVESGGGVVQPGRSLRLSCAASGFTFSSYAMHWVROAPGKGLEWVAVISYDGSNK
YYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARVGEWIOLWSPFDYWGOG TLyTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF PAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPE
FLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPR EEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTL PPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLT
VDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK
(SEQ ID NO: 12; variable domain underscored; CDRs double underscored).
Heavy chain variable domain
EVQLVESGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVAVISYDGSNK YYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARVGEWIQLWSPFDYWGQG TLVTVSS
(SEQ ID NO: 67)
[0124] The formulations disclosed herein can be used with antibodies comprising SEQ ID
NO: 12 or 67 and antigen-binding fragments thereof wherein residue 1 of SEQ ID NO: 12 or 67 is Q instead of E.
[0125] 3G7 heavy chain CDRs
CDR-H1: SYAMH (SEQ ID NO:28) CDR-H2: V1SYDGSNKYYADSVKG (SEQ ID NO:29)
CDR-H3: VGEWIQLWSPFDY (SEQ ID NO:30)
[0126] Antibody 3G7 light chain (kappa)
Light chain
DIOMTQSP S S VS AS VGDRVTITCRAS OGIS S WL AWYQOKPGKAPKFLIYAAS SLOS GVP S KFSGSGSGTDFTLTISSLQPEDFATYYCOOYNSYPPTFGGGTKVEIKRtVAAPSVFTFPPSD EQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTL SKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
(SEQ ID NO: 13; variable domain underscored; CDRs double underscored)
Light chain variable domain
DIQMTQSPSSVSASVGDRVTITCRASQGISSWLAWYQQKPGKAPKFLIYAASSLQSGVPS
KFSGSGSGTDFTLTISSLQPEDFATYYCQQYNSYPPTFGGGTKVEIK
(SEQ ID NO: 75)
[0127] 3G7 light chain CDRs
CDR-L1 : RASQGISSWLA (SEQ ID NO:31)
CDR-L2: AASSLQS (SEQ ID NO:32)
CDR-L3: QQYNSYPPT (SEQ ID NO:33)
[0128] Antibodies and antigen-binding fragments thereof including the 3G7 heavy and light chain CDRs or the 3G7 VH and VL or the 3G7 heavy chain and light chain (or a variant thereof, e.g., as set forth herein) may be referred to as “3G7.”
[0129] Antibody 2C1 (Q1E) heavy chain (IgG4)
Heavy chain
EVQLVOSGAEVKKPGASVKVSCKVSGYTLTELSMHWVROAPGKGLEWMGGFDPEDGE TIYAOKFOGRVTMTEDTSTDTAYMELSSLRSEDTAVYYCARAGPLYTIFGVVIIPDNWFD PWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTS GVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPP CPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNA KTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREP QVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFF
LYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK
(SEQ ID NO: 14; variable domain underscored; CDRs double underscored)
Heavy chain variable domain EVQLVQSGAEVKKPGASVKVSCKVSGYTLTELSMHWVRQAPGKGLEWMGGFDPEDGE
T1YAQKFQGRVTMTEDTSTDTAYMELSSLRSEDTAVYYCARAGPLYTIFGVVIIPDNWFD PWGQGTLVTVSS
(SEQ ID NO: 68)
[0130] The formulations disclosed herein can be used with antibodies comprising SEQ ID
NO: 14 or 68 and antigen-binding fragments thereof, wherein residue 1 of SEQ ID NO: 14 or 68 is Q instead of E.
[0131] 2C1 heavy chain CDRs
CDR-H1: ELSMH (SEQ ID NO:34)
CDR-H2: GFDPEDGETIYAQKFQG (SEQ ID NO 35)
CDR-H3: AGPLYTIFGVVIIPDNWFDP (SEQ ID NO:36)
[0132] Antibody 2C1 light chain (Q1E) (lambda)
Light chain
ESVLTQPPSVSGAPGORVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNSNRPSGV
PDRFSGSKSGTSASLAITGLQAEDEADYYCOSYDSSLSGSGVVFGGGTOLIILGQPKAAPS
VTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQSNNKYA
ASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS
(SEQ ID NO: 15; variable domain underscored; CDRs double underscored)
Light chain variable domain
ESVLTQPPSVSGAPGQRVTISCTGSSSNIGAGYDVHWYQQLPGTAPKLLIYGNSNRPSGV
PDRFSGSKSGTSASLAITGLQAEDEADYYCQSYDSSLSGSGVVFGGGTQLIIL
(SEQ ID NO: 76)
[0133] The formulations disclosed herein can be used with antibodies comprising SEQ ID
NO: 15 or 76 and antigen-binding fragments thereof, wherein residue 1 of SEQ ID NO: 15 or 76 is Q instead of E.
2C1 light chain CDRs
CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO:37)
CDR-L2: GNSNRPS (SEQ ID NO:38)
CDR-L3: QSYDSSLSGSGVV (SEQ ID NO:39)
[0134] Antibodies and antigen-binding fragments thereof including the 2C1 heavy and light chain CDRs or the 2C1 VH and VL or the 2C1 heavy chain and light chain (or a variant thereof, e.g., as set forth herein) may be referred to as “2C1.”
[0135] In various embodiments of the antibody or antigen-binding fragment thereof, a C- terminal lysine of a heavy chain immunoglobulin is absent.
[0136] Thus, in some embodiments, the antibody or antigen-binding fragment thereof comprises a light chain immunoglobulin, a heavy chain immunoglobulin, or both a light and heavy chain immunoglobulin, wherein the light chain immunoglobulin comprises the amino acid sequence set forth in SEQ ID NO:3, 4, 5, 6,7, 11, 13, 15, or 45; and/or the heavy chain immunoglobulin comprises the amino acid sequence set forth in SEQ ID NO: 1, 2, 8, 9, 10, 12, 14, 44, 79, 80, 81, 82, 83, 84, 85, or 86.
[0137] In certain embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 1 or 79; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 3.
[0138] In some embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 4.
[0139] In other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 5.
[0140] In yet other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 6.
[0141] In still other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 7.
[0142] In certain embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 2 or 80; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 3.
[0143] In some embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 8 or 82; and a light chain immunoglobulin comprising the ammo acid sequence set forth in SEQ ID NO: 11.
[0144] In other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 9 or 83; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 11.
[0145] In yet other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 10 or 84; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 11.
[0146] In still other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 12 or 85; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 13.
[0147] In yet still embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 14 or 86; and a light chain immunoglobulin comprising the amino acid sequence set forth in SEQ ID NO: 15.
[0148] In certain other embodiments, the antibody or antigen-binding fragment thereof comprises a light chain immunoglobulin, a heavy chain immunoglobulin, or both a light and heavy chain immunoglobulin, wherein the light chain variable domain comprises the amino acid sequence set forth in SEQ ID NO:70, 71, 72, 73, 58, 74, 75, 76, or 77, and/or the heavy chain variable domain compnses the ammo acid sequence set forth in SEQ ID NO:63, 57, 64, 65, 66, 67, 68, or 69.
[0149] In certain embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:63; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:70.
[0150] In some embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:71.
[0151] In other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain vanable domain comprising the ammo acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:72.
[0152] In yet other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:73.
[0153] In still other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58.
[0154] In certain embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:70.
[0155] In some embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:64; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:74.
[0156] In other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:65; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:74.
[0157] In yet other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:66; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO: 74.
[0158] In still other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:67; and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:75.
[0159] In yet still other embodiments, the antibody or antigen-binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:68; and a light chain variable domain comprising the ammo acid sequence set forth in SEQ ID NO:76.
[0160] In a further embodiment, the antibody or antigen-binding fragment thereof that binds ILT4 comprises an immunoglobulin light chain variable (VL) domain comprising a CDR-L1, CDR-L2 and CDR-L3 of 1E1 (e.g, SEQ ID NOs: 19-21); and an immunoglobulin heavy chain variable (VH) domain comprising a CDR-H1, CDR-H2 and CDR-H3 of 1E1 (e.g, SEQ ID NOs: 16-18).
[0161] In a further embodiment, the antibody or antigen-binding fragment thereof that binds ILT4 comprises an immunoglobulin light chain variable (VL) domain comprising a CDR-L1, CDR-L2 and CDR-L3 of 2A6 (e.g, SEQ ID NOs: 25-27); and an immunoglobulin heavy chain variable (VH) domain comprising a CDR-H1, CDR-H2 and CDR-H3 of 2A6 (e.g., SEQ ID NOs: 22-24).
[0162] In a further embodiment, the antibody or antigen-binding fragment thereof that binds ILT4 comprises an immunoglobulin light chain variable (VL) domain comprising a CDR-L1, CDR-L2 and CDR-L3 of 3G7 (e.g, SEQ ID NOs: 31-33); and an immunoglobulin heavy chain variable (VH) domain comprising a CDR-H1, CDR-H2 and CDR-H3 of 3G7 (e.g, SEQ ID NOs: 28-30).
[0163] In a further embodiment, the antibody or antigen-binding fragment thereof that binds ILT4 comprises an immunoglobulin light chain variable (VL) domain comprising a CDR-L1, CDR-L2 and CDR-L3 of 2C1 (e.g, SEQ ID NOs: 37-39); and an immunoglobulin heavy' chain variable (VH) domain comprising a CDR-H1, CDR-H2 and CDR-H3 of 2C1 (e.g., SEQ ID NOs: 34-36).
[0164] In one embodiment, the antibody or antigen-binding fragment comprises a VH domain comprising CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GNSNRPS(SEQ ID NO: 49), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0165] In another embodiment, the antibody or antigen-binding fragment comprises a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GQSNRPS(SEQ ID NO: 50), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0166] In yet another embodiment, the antibody or antigen-binding fragment thereof comprises a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GESNRPS(SEQ ID NO: 51), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21) [0167] In still another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GDSNRPS(SEQ ID NO: 52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0168] In one embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GNANRPS(SEQ ID NO: 53), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0169] In another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2:
EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GQANRPS(SEQ ID NO: 54), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0170] In yet another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2:
EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GEANRPS(SEQ ID NO: 55), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0171] In still another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO: 47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GDANRPS(SEQ ID NO: 56), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0172] In one embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GNSNRPS(SEQ ID NO: 49), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21). [0173] In another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2:
EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GQSNRPS(SEQ ID NO: 50), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0174] In yet another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising:CDR-Hl: GYYWS (SEQ ID NO: 16), CDR-H2:
EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GESNRPS(SEQ ID NO: 51), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21) [0175] In still another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GDSNRPS(SEQ ID NO: 52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0176] In one embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GNANRPS(SEQ ID NO: 53), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0177] In another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2:
EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GQANRPS(SEQ ID NO: 54), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0178] In yet another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2:
EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GEANRPS(SEQ ID NO: 55), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0179] In still another embodiment, the antibody or antigen-binding fragment thereof comprises: a VH domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: E1NHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: EPTRWVTTRYFDL (SEQ ID NO: 18); and/or, a VL domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR- L2: GDANRPS(SEQ ID NO: 56), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0180] The formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the VL domain of antibody 1E1 (e.g, SEQ ID NO:70, 71, 72, 73, 58 or 77) and/or the VH domain of antibody 1E1 (e.g., SEQ ID NO:63, 57 or 69).
[0181] The formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the VL domain of antibody 2A6 (e.g, SEQ ID NO:74) and/or the VH domain of antibody 2A6 (e.g., SEQ ID NO:64, 65 or 66).
[0182] The formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the VL domain of antibody 3G7 (e.g., SEQ ID NO:75) and/or the VH domain of antibody 3G7 (e.g., SEQ ID NO:67).
[0183] The formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the VL domain of antibody 2C1 (e.g., SEQ ID NO:76) and/or the VH domain of antibody 2C1 (e.g., SEQ ID NO:68).
[0184] The formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 1E1 (e.g., SEQ ID NO:3, 4, 5, 6, 7 or 45) and/or the heavy chain immunoglobulin of antibody 1E1 (e.g., SEQ ID NO: I, 2, 44, 79, 80, or 81).
[0185] The formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 2A6 (e.g. , SEQ ID NO: 11) and/or the heavy chain immunoglobulin of antibody 2A6 (e.g. , SEQ ID NO:8, 9, 10, 82, 83, or 84).
[0186] The formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 3G7 (e.g, SEQ ID NO: 13) and/or the heavy chain immunoglobulin of antibody 3G7 (e.g., SEQ ID NO: 12 or 85).
[0187] The formulations disclosed herein can be used with an antibody or antigen-binding fragment thereof that binds ILT4 and comprises the light chain immunoglobulin of antibody 2C1 (e.g, SEQ ID NO: 15) and/or the heavy chain immunoglobulin of antibody 2C1 (e.g., SEQ ID NO: 14 or 86).
[0188] The formulations disclosed herein can be used with an antibody that consists of two heavy chains and two light chains, wherein each light chain comprises the VL or light chain immunoglobulin of antibody 1E1, 2A6, 3G7, or 2C1, and each heavy chain comprises the VH or heavy chain immunoglobulin of antibody 1E1, 2A6, 3G7, or 2C1.
[0189] In one embodiment, the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO:58 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO:57.
[0190] In another embodiment, the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO:58 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO:57, wherein the light chain further comprises the amino acid sequence set forth in SEQ ID NO: 90.
[0191] In yet another embodiment, the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO:58 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO:57, wherein the heavy chain further comprises the amino acid sequence set forth in SEQ ID NO: 89.
[0192] In still another embodiment, the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO:58 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO: 57, wherein the light chain further comprises the amino acid sequence set forth in SEQ ID NO:90 and the heavy chain further comprises the amino acid sequence set forth in SEQ ID NO: 89.
[0193] In one embodiment, the antibody consists of two heavy chains and two light chains, wherein each light chain comprises the amino acid sequence set forth in SEQ ID NO: 7 and each heavy chain comprises the amino acid sequence set forth in SEQ ID NO:2.
[0194] In another embodiment, the antibody consists of two heavy chains and two light chains, wherein each light chain consists of the amino acid sequence set forth in SEQ ID NO:7 and each heavy chain consists of the amino acid sequence set forth in SEQ ID NO:2.
[0195] In an embodiment, the antibody or antigen-binding fragment thereof comprises a VL (with or without signal sequence), e.g., the VL in any of SEQ ID NO:58 or 70-77, having up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more conservative or non-conservative amino acid substitutions; and/or a VH (with or without signal sequence), e.g., the VH in any of SEQ ID NO:57 or 63-69, having up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more conservative or non-conservative amino acid substitutions, while still binding to ILT4.
[0196] The formulations disclosed herein can be used with polypeptides comprising the amino acid sequences disclosed herein, e.g., SEQ ID NOs: 1-39, 44, 45, 47-58, 63-77, or 79-86, as well as polypeptides comprising such amino acid sequences with up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 20 or more conservative or non-conservative amino acid substitutions therein.
[0197] In certain embodiments, the antibody or antigen-binding fragment thereof comprises a light chain immunoglobulin, a heavy chain immunoglobulin, or both a light and heavy chain immunoglobulin, wherein the light chain immunoglobulin has at least 90% amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO:3, 4, 5, 6,7, 11, 13, 15, or 45, and/or the heavy chain immunoglobulin has at least 90% amino acid sequence identity' to the amino acid sequence set forth in SEQ ID NO: 1, 2, 8, 9, 10, 12, 14, 44, 79, 80, 81, 82, 83, 84, 85, or 86.
[0198] In some embodiments, the antibody or antigen-binding fragment thereof comprises a light chain immunoglobulin, a heavy' chain immunoglobulin, or both a light and heavy chain immunoglobulin, wherein the light chain immunoglobulin comprises a light chain variable domain having at least 90% amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO:70, 71, 72, 73, 58, 74, 75, 76, or 77, and/or the heavy chain immunoglobulin comprises a heavy chain variable domain having at least 90% amino acid sequence identity to the amino acid sequence set forth in SEQ ID NO:63, 57, 64, 65, 66, 67, 68, or 69.
[0199] In an embodiment, an immunoglobulin heavy chain of an anti-ILT4 antibody or antigenbinding fragment of the invention is operably linked to a signal sequence, e.g., comprising the amino acid sequence MEWSWVFLFFLSVTTGVHS (SEQ ID NO:41) and/or an immunoglobulin light chain of an anti-ILT4 antibody or antigen-binding fragment of the invention is operably linked to a signal sequence, e.g., comprising the amino acid sequence MSVPTQVLGLLLLWLTDARC (SEQ ID NO:42).
[0200] In an embodiment, an N-terminal glutamine (Q) of an immunoglobulin chain set forth herein (e.g., heavy and/or light) is replaced with a pyroglutamic acid. In one embodiment, an N- terminal Q of a heavy chain immunoglobulin is replaced with a pyroglutamic acid. In another embodiment, an N-terminal Q of a light chain immunoglobulin is replaced with a pyroglutamic acid. In yet another embodiment, an N-terminal Q of a heavy chain immunoglobulin and an N- terminal Q of a heavy chain immunoglobulin are replaced with a pyroglutamic acid.
[0201] The formulations disclosed herein can also be used with antibodies or antigen-binding fragments that bind to the same epitope of ILT4 (e.g., human ILT4) as any anti-ILT4 antibody or antigen-binding fragment thereof disclosed herein (e.g., 1E1, 2A6, 3G7 or 2C1). In one embodiment, the epitope is LYREKKSASW (SEQ ID NO:59). In another embodiment, the epitope is TRIRPEL (SEQ ID NO:60). In yet another embodiment, the epitope is NGQF (SEQ ID NO:61). In still another embodiment, the epitope is HTGRYGCQ (SEQ ID NO:62). In certain embodiments, the antibody or antigen-binding fragment thereof binds to the same epitope of human ILT4 as an antibody or antigen-binding fragment thereof comprising the heavy chain and light chain amino acid sequences set forth in SEQ ID NOs: 1 and 3; 2 and 4; 2 and 5; 2 and 6; 2 and 7; 2 and 3; 8 and 11; 9 and 11; 10 and 11; 12 and 13; 14 and 15; 79 and 3; 80 and 4; 80 and 5; 80 and 6; 80 and 7; 80 and 3; 82 and 11; 83 and 11; 84 and 11; 85 and 13; and 86 and 15; respectively. In some embodiments, the antibody or antigen-binding fragment thereof binds to the same epitope of human ILT4 as an antibody or antigen-binding fragment thereof comprising the heavy chain variable domain and light chain variable domain amino acid sequences set forth in SEQ ID NOs:63 and 70; 57 and 71; 57 and 72; 57 and 73; 57 and 58; 57 and 70; 64 and 74; 65 and 74; 66 and 74; 67 and 75; 68 and 76; respectively.
[0202] The formulations disclosed herein can be used with antibodies and antigen-binding fragments that cross-block the binding of any anti-ILT4 antibody or antigen-binding fragment thereof disclosed herein (e.g., 1E1, 2A6, 3G7 or 2C1) to ILT4 (e.g., human ILT4) or compete with any anti-ILT4 antibody or antigen-binding fragment thereof disclosed herein (e.g, 1E1, 2A6, 3G7 or 2C1) to ILT4 (e.g., human ILT4). The cross-blocking antibodies and antigenbinding fragments thereof discussed herein can be identified based on their ability to block any of the antibodies or fragments specifically set forth herein from binding to ILT4, in binding assays (e.g., bio-layer interferometry (BLI; for example FORTEBIO OCTET binding assay; Pall ForteBio Corp; Menlo Park, CA), surface plasmon resonance (SPR), BIACore, ELISA, flow cytometry). For example, in an embodiment of the invention, when using BLI, the tip of a fiberoptic probe is coated wdth ligand (e.g., ILT4) and acts as the biosensor wherein binding of anti- ILT4 antibody or antigen-binding fragment to the ILT4 alters the interference pattern of white light reflected from the probe layer bound to 1LT4 and an internal reference layer. The shift is indicative of ILT4/anti-ILT4 binding. In an embodiment of the invention, the ILT4 coated tip is immersed in a solution of analyte containing antibody or antigen-binding fragment, e.g., in the well of either a 96- or 384-well plate. In an embodiment of the invention, the plate is shaken during reading to create orbital flow. To read the assay, white light is directed down the length of the fiber. As mentioned above, interference between light reflecting off the reference layer and immobilized surfaces containing ILT4 of the tip creates a distinctive pattern of light returning up the fiber. As molecules bind to the immobilized sensor surface, that pattern changes in proportion to the extent of binding. For example, assays can be used in which an ILT4 (e.g., human ILT4) protein is immobilized on a BLI probe or plate, a reference anti-ILT4 antibody or fragment binds to ILT4 (e.g., at saturating concentration) and a test anti-ILT4 antibody or fragment is added. The ability of the test antibody to compete with the reference antibody for 1LT4 binding is then determined. In the BLI format, light interference of the ILT4 complex is monitored to determine if the test antibody effectively competes with the reference antibody, e.g., nanometers of light wavelength shift over time is monitored wherein a shift indicates additional binding of the test antibody and a lack of cross-blocking. In an embodiment of the invention, in the BLI format, cross-blocking is qualitatively deemed to have occurred between the antibodies if no additional binding of test antibody is observed. In an embodiment of the invention, as a control, crossblocking of the reference antibody with itself is confirmed; wherein the assay is determined to be operating correctly if the reference antibody can cross-block itself from ILT4 binding. The ability of a test antibody to inhibit the binding of the anti-ILT4 antibody or fragment 1E1, 2A6, 3G7 or 2C1, to ILT4 (e.g., human ILT4) demonstrates that the test antibody can cross-block the antibody or fragment for binding to ILT4 (e.g., human ILT4) and thus, may, in some cases, bind to the same epitope on ILT4 (e.g, human ILT4) as 1E1, 2A6, 3G7 and/or 2C1. As stated above, antibodies and fragments that bind to the same epitope as any of the anti-ILT4 antibodies or fragments of the invention also form part of the invention. In an embodiment of the invention, BLI is conducted in a sandwich format wherein a reference anti- ILT4 antibody or antigenbinding fragment is immobilized to the probe and then bound with ILT4. Test anti- ILT4 antibody or antigen-binding fragment is then tested for the ability to block binding of the references antibody or fragment.
[0203] In certain embodiments, the antibody or antigen-binding fragment thereof competes for binding to human ILT4 with an antibody or fragment comprising the heavy chain and light chain amino acid sequences set forth in SEQ ID NOs: 1 and 3; 2 and 4; 2 and 5; 2 and 6; 2 and 7; 2 and 3; 8 and 11; 9 and 11; 10 and 11; 12 and 13; 14 and 15; 79 and 3; 80 and 4; 80 and 5; 80 and 6; 80 and 7; 80 and 3; 82 and 11; 83 and 11; 84 and 11; 85 and 13; and 86 and 15; respectively. In some embodiments, the antibody or antigen-binding fragment thereof competes for binding to human ILT4 with an antibody or fragment comprising the heavy chain variable domain and light chain variable domain amino acid sequences set forth in SEQ ID NOs:63 and 70; 57 and 71; 57 and 72; 57 and 73; 57 and 58; 57 and 70; 64 and 74; 65 and 74; 66 and 74; 67 and 75; 68 and 76; respectively.
[0204] The formulations disclosed herein can be used with anti-ILT4 antibodies and antigenbinding fragments thereof comprising N-linked glycans that are typically added to immunoglobulins produced in Chinese hamster ovary cells (CHO N-linked glycans) or in engineered yeast cells (engineered yeast N-linked glycans), such as, for example, Pichict pastoris. For example, in an embodiment, the anti-lLT4 antibodies and antigen-binding fragments thereof comprise one or more of the “engineered yeast N-linked glycans” or “CHO N-linked glycans” (e.g, GO and/or GO-F and/or G1 and/or Gl-F and/or G2-F and/or Man5). In an embodiment, the anti-ILT4 antibodies and antigen-binding fragments thereof comprise the engineered yeast N- linked glycans, i.e., GO and/or G1 and/or G2, optionally, further including Man5. In an embodiment, the anti-ILT4 antibodies and antigen-binding fragments thereof comprise the CHO N-linked glycans, i.e., GO-F, Gl-F and G2-F, optionally, further including GO and/or G1 and/or G2 and/or Man5. In an embodiment, about 80% to about 95% (e.g., about 80-90%, about 85%, about 90% or about 95%) of all N-linked glycans on the anti-ILT4 antibodies and antigenbinding fragments thereof are engineered yeast N-linked glycans or CHO N-linked glycans. See Nett et al. Yeast. 28(3): 237-252 (2011); Hamilton et al. Science. 313(5792): 1441-1443 (2006); Hamilton et al. Curr Opin Biotechnol. 18(5): 387-392 (2007). For example, in an embodiment, an engineered yeast cell is GFI5.0 or YGLY8316 or strains set forth in U.S. Patent No. 7,795,002 or Zha et al. Methods Mol Biol. 988:31-43 (2013). See also international patent application publication no. WO2013/066765.
[0205] The anti-ILT4 antibodies and antigen-binding fragments thereof that can be used with various formulations disclosed herein (e.g, 1E1, 2A6, 3G7 and/or 2C1) can also be engineered to include modifications within the Fc region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or effector function (e.g., antigen-dependent cellular cytotoxicity). Furthermore, the antibodies and antigenbinding fragments thereof that can be used with various formulations disclosed herein (e.g., 1E1, 2A6, 3G7 and/or 2C1) can be chemically modified (e.g., one or more chemical moieties can be attached to the antibody, such as a labeled antibody or an antibody-drug conjugate) or be modified to alter its glycosylation, again to alter one or more functional properties of the antibody.
[0206] In some embodiments of the various formulations disclosed herein, the antibody or antigen-binding fragment is a monoclonal antibody.
Pharmaceutical Formulations
[0207] The pharmaceutical formulations described herein can retain physical, chemical, and/or biological stability of an anti-ILT4 antibody (e.g., 1E1, 2A6, 3G7, or 2C1) or antigen-binding fragment thereof during storage (e g, at about 5°C for at least 6 months, 12 months, 24 months, or 36 months, at -20°C for at least 6 months, 12 months, 24 months, or 36 months, or at -70°C for at least 6 months, 12 months, 24 months, or 36 months) and/or under various stress conditions (e.g, agitation, freeze and thaw cycles, or light exposure).
[0208] In one aspect, provided herein are various formulations of an anti-ILT4 monoclonal antibody (e.g., 1E1, 2A6, 3G7, or 2C1) or antigen-binding fragment thereof, comprising (i) an anti-ILT4 antibody or antigen-binding fragment thereof (e.g., 1E1, 2A6, 3G7, or 2C1); (ii) a buffer (e.g., L-histidine buffer or acetate buffer); (iii) a non-reducing sugar (e.g., sucrose); (iv) a non-ionic surfactant (e.g., PS-80); and (v) an antioxidant (e.g., L-methionine).
[0209] Buffers that can be used in the pharmaceutical formulations disclosed herein include, but are not limited to, succinate (sodium or potassium), L-histidine, phosphate (sodium or potassium), Tris (tris (hydroxymethyl) aminomethane), diethanolamine, citrate (sodium), acetate (sodium) and the like. In some embodiments of the formulation, the buffer is present in the formulation at a concentration of about 1-20 mM (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 mM).
[0210] The buffer of various formulations described herein has a pH in the range of from about 4.5 to about 7.0, from about 4.5 to about 6.8, from about 5.0 to about 6.8, from about 5.0 to about 6.5, from 5.0 to about 6.0, from about 5.5 to about 6.0. In arriving at the exemplary formulation, L-histidine and acetate buffers in the pH range of 5.0-6.8 were explored for suitability. When a range of pH values is recited, such as “a pH from about pH 5.5 to about 6.0,” the range is intended to be inclusive of the recited values. For example, a range from about 5.0 to about 6.0 includes 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, and 6.0. In the event where the anti-ILT4 formulation described herein is a lyophilized formulation, the pH refers to the pH after reconstitution of the lyophilized formulation. pH is typically measured at 25 °C using a standard glass bulb pH meter. As used herein, a solution comprising “histidine buffer at pH X” refers to a solution at pH X and comprising the histidine buffer, i.e., the pH is intended to refer to the pH of the solution.
[0211] In some embodiments, the anti-ILT4 formulation comprises a non-reducing sugar. As used herein, “non-reducing sugar” is a sugar not capable of acting as a reducing agent because it does not contain or cannot be converted to contain a free aldehyde group or a free ketone group. Examples of non-reducing sugars include, but are not limited to, disaccharides such as sucrose and trehalose. In an embodiment of the invention, the non-reducing sugar is present in an amount of from about 1% (w/v) to about 10% (w/v) (about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10% (w/v)). In another embodiment, the non-reducing sugar is present in an amount from about 6% (w/v) to about 8% (w/v) (about 6, 7, or 8% (w/v)). In a further embodiment, the non-reducing sugar is present in an amount of about 6% (w/v). In a further embodiment, the non-reducing sugar is present in an amount of about 7% (w/v). In a further embodiment, the non-reducing sugar is present in an amount of about 8% (w/v). In one embodiment, the non-reducing sugar is sucrose, trehalose, or raffinose. In a further embodiment, the sucrose is present at about 6% (w/v) to about 8% (w/v). In one embodiment, the sucrose is present at about 6% (w/v). In one embodiment, the sucrose is present at about 7% (w/v). In one embodiment, the sucrose is present at about 8% (w/v).
[0212] In certain embodiments, the anti-ILT4 formulations also comprise a surfactant. As used herein, a surfactant is a surface active agent that is amphipathic in nature. Surfactants can be added to the formulations herein to provide stability, reduce and/or prevent aggregation, or to prevent and/or inhibit protein damage during processing conditions such as purification, filtration, freeze-drying, transportation, storage, and delivery. In some embodiments, a surfactant can be useful for providing additional stability to the active ingredient(s).
[0213] Non-ionic surfactants that can be used in the formulations of the invention include, but are not limited to, polyoxyethylene sorbitan fatty acid esters (Polysorbates, sold under the trade name Tween® (Uniquema Americas LLC, Wilmington, DE)) including Polysorbate-20 (PS-20, polyoxyethylene sorbitan monolaurate), Polysorbate-40 (PS-40, polyoxyethylene sorbitan monopalmitate), Polysorbate-60 (PS-60, polyoxyethylene sorbitan monostearate), and Polysorbate-80 (PS-80, polyoxyethylene sorbitan monooleate); polyoxyethylene alkyl ethers such as Brij® 58 (Uniquema Americas LLC, Wilmington, DE) and Brij® 35; pol oxamers (e.g., poloxamer 188); Triton® X-100 (Union Carbide Corp., Houston, TX) and Triton® X-114; NP40; Span 20, Span 40, Span 60, Span 65, Span 80 and Span 85; copolymers of ethylene and propylene glycol (e.g., the pluronic® series of nonionic surfactants such as pluronic® F68, pluronic® 10R5, pluronic® F108, pluronic® F127, pluronic® F38, pluronic® L44, pluronic® L62 (BASF Corp., Ludwigshafen. Germany); and sodium dodecyl sulfate (SDS). In one embodiment, the non-ionic surfactant is PS-80 or PS-20. In one embodiment, the non-ionic surfactant is PS-20. In another embodiment, the non-ionic surfactant is PS-80.
[0214] The amount of non-ionic surfactant to be included in the formulations is an amount sufficient to perform the desired function, i.e., a minimal amount necessary to stabilize the active pharmaceutical ingredient (i.e., the anti-ILT4 antibody or antigen-binding fragment thereof e.g., 1E1, 2A6, 3G7, or 2C1)) in the formulation. All percentages for the non-ionic surfactant are listed as % (w/v). Typically, the surfactant is present in a concentration of from about 0.008% to about 0.1% (w/v). In some embodiments of this aspect of the invention, the surfactant is present in the formulation in an amount from about 0.01% to about 0.1%, from about 0.01% to about 0.09%, from about 0.01% to about 0.08%, from about 0.01% to about 0.07%, from about 0.01% to about 0.06%, from about 0.01% to about 0.05%, from about 0.01% to about 0.04%, from about 0.01% to about 0.03%, from about 0.01% to about 0.02%, from about 0.015% to about 0.04%, from about 0.015% to about 0.03%, from about 0.015% to about 0.02%, from about 0.02% to about 0.04%, from about 0.02% to about 0.035%, or from about 0.02% to about 0.03%. In alternative embodiments, the surfactant is present in an amount of about 0.01%, about 0.015%, about 0.02%, about 0.025%, about 0.03%, about 0.035%, or about 0.04%.
[0215] In specific embodiments, the formulations described herein comprise about 0.01% to about 0.04% (w/v) PS-80. In one embodiment, the formulations described herein comprise PS-80 in an amount of about 0.01%. In one embodiment, the amount of PS-80 is about 0.015%. In another embodiment, the amount of PS-80 is about 0.02%. In a further embodiment, the amount of PS-80 is about 0.025%. In another embodiment, the amount of PS-80 is about 0.03%. In a further embodiment, the amount of PS-80 is about 0.035%. In another embodiment, the amount of PS-80 is about 0.04%. In a further embodiment, the amount of PS-80 is about 0.045%.
[0216] The formulations described herein also comprise methionine, or pharmaceutically acceptable salt thereof as an antioxidant. In one embodiment, the methionine is L-methionine. In another embodiment, the methionine is a pharmaceutically acceptable salt of L-methionine, such as, for example, methionine HC1. In an embodiment of the invention, methionine is present in the formulation at a concentration of about 1-20 mM (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, and 20 mM). In another embodiment, the methionine is present from about 5 mM to about 10 mM (5, 6, 7, 8, 9, and 10 mM). In another embodiment, the methionine is present at about 10 mM.
[0217] Thus, in one aspect, provided herein is a pharmaceutical formulation for an anti-lLT4 antibody or antigen-binding fragment thereof, comprising: (i) about 10 mg/mL to about 200 mg/rnL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM of a buffer; (iii) about 6% to about 8% weight /volume (w/v) of a non-reducing sugar; (iv) about 0.01 % to about 0.10% (w/v) of anon-ionic surfactant; and (v) about 1 mM to about 20 mM of an anti-oxidant.
[0218] In certain embodiments, the buffer is a selected from the group consisting of L-histidine buffer, acetate buffer, and citrate buffer. In one embodiment, the buffer is L-histidine buffer. In another embodiment, the buffer is acetate buffer. In yet another embodiment, the buffer is citrate buffer.
[0219] In some embodiments, the non-reducing sugar is sucrose. [0220] In certain embodiments, the non-ionic surfactant is PS-80 or PS-20. In one embodiment, the non-ionic surfactant is PS-80. In another embodiment, the non-ionic surfactant is PS-20.
[0221] In some embodiments, the anti-oxidant is L-methionine.
[0222] Thus, in another aspect, provided herein is a pharmaceutical formulation for an anti- ILT4 antibody or antigen-binding fragment thereof, comprising: (i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM L-histidine buffer; (iii) about 6% to about 8% (w/v) sucrose; (iv) about 0.01 % to about 0.10% (w/v) PS-80; and (v) about 1 mM to about 20 mM L-methionine.
[0223] In some embodiments, the formulation comprises about 8 mM to about 12 mM L- histidine buffer.
[0224] In certain embodiments, the formulation comprises about 5 mM to about 10 mM L- methionine.
[0225] In other embodiments, the formulation comprises about 0.01 % to about 0.05% (w/v) PS-80.
[0226] In yet other embodiments, the formulation comprises about 10 mg/mL to about 150 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof. In still other embodiments, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL, about 12.5 mg/mL, about 15 mg/mL, about 25 mg/mL, about 50 mg/mL, about 75 mg/mL, about 100 mg/mL, about 125 mg/mL, or about 150 mg/mL. In one embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL. In another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 12.5 mg/mL. In yet another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 15 mg/mL. In still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 25 mg/mL. In one embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 50 mg/mL. In another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 75 mg/mL. In yet another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 100 mg/mL. In still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 125 mg/mL. In yet still another embodiment, the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 150 mg/mL.
[0227] Thus, in one specific embodiment, the formulation comprises about 25 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0228] In another specific embodiment, the formulation comprises about 50 mg/mL of the anti- ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0229] In yet another specific embodiment, the formulation comprises about 75 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0230] In still another specific embodiment, the formulation comprises about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0231] In yet still another specific embodiment, the formulation comprises about 125 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
[0232] In certain embodiments of various formulations provided herein, the formulation has a pH of about 5.0 - about 6.8. In some embodiments, the formulation has a pH of about 5.5 - about 6.0. In other embodiments, the formulation has a pH of about 5.5, about 5.6, about 5.7, about 5.8, about 5.9, or about 6.0. In one embodiment, the formulation has a pH of about 5.5. In another embodiment, the formulation has a pH of about 5.6. In yet another embodiment, the formulation has a pH of about 5.7. In still another embodiment, the formulation has a pH of about 5.8. In another embodiment, the formulation has a pH of about 5.9. In yet still another embodiment, the formulation has a pH of about 6.0.
[0233] Thus, in one specific embodiment, the pharmaceutical formulation for an anti-ILT4 antibody or antigen-binding fragment thereof compnses: (i) about 50 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer, pH about 5.5; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) polysorbate 80; and (v) about 10 mM L-methionine.
[0234] In certain embodiments of the various formulations disclosed herein, the anti-ILT4 antibody or antigen-binding fragment thereof is an anti-ILT4 antibody or antigen-binding fragment thereof described herein (e.g. , 1E1, 2A6, 3G7, or 2C1). In some embodiments of the formulations, the anti-ILT4 antibody or antigen-binding fragment thereof is 1E1 or a variant thereof. In some embodiments of the formulations, the anti-ILT4 antibody or antigen-binding fragment thereof is 2A6 or a variant thereof. In some embodiments of the formulations, the anti- ILT4 antibody or antigen-binding fragment thereof is 3G7 or a variant thereof. In some embodiments of the formulations, the anti-lLT4 antibody or antigen-binding fragment thereof is 2C1 or a variant thereof.
[0235] In certain embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein Xi is N, Q, E or D and X2 is S or A (SEQ ID NO:20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO:21).
[0236] In some embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO:48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO:18); and a light chain variable domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GDSNRPS(SEQ ID NO:52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO:21).
[0237] In other embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58.
[0238] In yet other embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:2 and a light chain comprising the amino acid sequence set forth in SEQ ID NOY.
[0239] In still other embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 80 and a light chain comprising the amino acid sequence set forth in SEQ ID NOY.
[0240] In certain embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:58.
[0241] In some embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO:2 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
[0242] In other embodiments of the various formulations provided herein, the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO: 80 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
[0243] Thus, in some embodiments, provided herein is a pharmaceutical formulation for an anti-ILT4 antibody or antigen-binding fragment thereof, comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L-histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein Xi is N, Q, E or D and X2 is S or A (SEQ ID NO:20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO:21).
[0244] In certain embodiments, provided herein is a pharmaceutical formulation for an anti- ILT4 antibody or antigen-binding fragment thereof, comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: E1NHAGSTNYNPSLKS (SEQ ID NO:48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO:18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GDSNRPS(SEQ ID NO:52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO:21).
[0245] In other embodiments, provided herein is a pharmaceutical formulation for an anti-ILT4 antibody or antigen-binding fragment thereof, comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58. [0246] In yet other embodiments, provided herein is a pharmaceutical formulation for an anti- ILT4 antibody or antigen-binding fragment thereof, comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:2 and a light chain comprising the amino acid sequence set forth in SEQ ID NO:7.
[0247] In still other embodiments, provided herein is a pharmaceutical formulation for an anti- ILT4 antibody or antigen-binding fragment thereof, comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 80 and a light chain comprising the amino acid sequence set forth in SEQ ID NO:7.
[0248] In some embodiments, provided herein is a pharmaceutical formulation of an anti-ILT4 antibody or antigen-binding fragment thereof, comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:58.
[0249] In some embodiments, provided herein is a pharmaceutical formulation of an anti-lLT4 antibody or antigen-binding fragment thereof, comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO:2 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
[0250] In some embodiments, provided herein is a pharmaceutical formulation of an anti-ILT4 antibody or antigen-binding fragment thereof, comprising: (i) about 25 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L- histidine buffer; (iii) about 7% (w/v) sucrose; (iv) about 0.025% (w/v) PS-80; and (v) about 10 mM L-methionine; wherein the anti-lLT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO: 80 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
[0251] In one embodiment, the formulation is a liquid formulation. In another embodiment, the liquid formulation is stored at about 3°C - 5°C. In yet another embodiment, the liquid formulation is frozen to at least -70°C or below. In still another embodiment, the liquid formulation is a reconstituted solution from a lyophilized formulation.
[0252] In certain embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, (i) the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultra-performance size exclusion chromatography; (ii) the turbidity of the formulation is at most about 0.135 as measured by OD350-500; (iii) the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, and/or the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography; (iv) the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging; and/or (v) the % oxidation of one or more amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
[0253] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultra-performance size exclusion chromatography.
[0254] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the turbidity of the formulation is at most about 0.135 as measured by OD350-500.
[0255] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, as determined by high performance ion-exchange chromatography. In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, as determined by high performance ion-exchange chromatography. In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography. In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, and the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography.
[0256] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging.
[0257] In some embodiments, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of one or more amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of one amino acid residue selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of two amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of three amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-lLT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of four amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping. In one embodiment, after the formulation is stored at about 3°C to about 5°C for 6 months, the % oxidation of all five amino acid residues from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
[0258] The formulations described herein can be made as a liquid formulation. A liquid antibody formulation can be made by taking the drug substance (e.g., anti-lLT4 monoclonal antibody) which is in liquid form (e.g., anti-ILT4 monoclonal antibody in an aqueous pharmaceutical formulation) and buffer exchanging it into the desired buffer as the last step of the purification process. There is no lyophilization step in this embodiment. The drug substance in the final buffer is concentrated to a desired concentration. Excipients such as sucrose and PS 80 are added to the drug substance and it is diluted using the appropriate buffer to final protein concentration. The final formulated drug substance is filtered using 0.22pm filters and filled into a final container (e.g., glass vials).
[0259] The formulations described herein can also be reconstituted from lyophilized formulations. Lyophilized formulations of therapeutic proteins provide several advantages. Lyophilized formulations in general offer better chemical stability than solution formulations, and thus increased half-life. A lyophilized formulation may also be reconstituted at different concentrations depending on clinical factors, such as route of administration or dosing. For example, a lyophilized formulation may be reconstituted at a high concentration (i.e., in a small volume) if necessary for subcutaneous administration, or at a lower concentration if administered intravenously. High concentrations may also be necessary if high dosing is required for a particular subject, particularly if administered subcutaneously where injection volume must be minimized. One such lyophilized antibody formulation is disclosed at U.S. Pat. No. 6,267,958, which is hereby incorporated by reference in its entirety. Lyophilized formulations of another therapeutic protein are disclosed at U.S. Pat. No. 7,247,707, which is hereby incorporated by reference in its entirety.
[0260] Typically, the lyophilized formulation is prepared in anticipation of reconstitution at high concentration of drug product (DP, in an exemplary embodiment anti-lLT4 antibody or antigen-binding fragment thereof), i.e., in anticipation of reconstitution in a low volume of water. Subsequent dilution with water or isotonic buffer can then readily be used to dilute the DP to a lower concentration. Typically, excipients are included in a lyophilized formulation of the invention at levels that will result in a roughly isotonic formulation when reconstituted at high DP concentration, e.g., for subcutaneous administration. Reconstitution in a larger volume of water to give a lower DP concentration will necessarily reduce the tonicity of the reconstituted solution, but such reduction may be of little significance in non-subcutaneous, e.g., intravenous, administration. If isotonicity is desired at lower DP concentration, the lyophilized powder may be reconstituted in the standard low volume of water and then further diluted with isotonic diluent, such as 0.9% sodium chloride. [0261] The lyophilized formulations of the invention are formed by lyophilization (freeze- drying) of a pre-lyophilization solution. Freeze-drying is accomplished by freezing the formulation and subsequently subliming water at a temperature suitable for primary drying. Under this condition, the product temperature is below the eutectic point or the collapse temperature of the formulation. Typically, the shelf temperature for the primary drying will range from about -30 to 25°C (provided the product remains frozen during primary drying) at a suitable pressure, ranging typically from about 50 to 250 mTorr. The formulation, size and type of the container holding the sample (e.g., glass vial) and the volume of liquid will dictate the time required for drying, which can range from a few hours to several days (e.g., 40-60 hrs). A secondary drying stage may be carried out at about 0-40°C, depending primarily on the type and size of container and the ty pe of protein employed. The secondary drying time is dictated by the desired residual moisture level in the product and typically takes at least about 5 hours.
Typically , the moisture content of a lyophilized formulation is less than about 5%, and preferably less than about 3%. The pressure may be the same as that employed during the primary drying step. Freeze-drying conditions can be varied depending on the formulation and vial size.
[0262] In some instances, it may be desirable to lyophilize the protein formulation in the container in which reconstitution of the protein is to be carried out in order to avoid a transfer step. The container in this instance may, for example, be a 3, 5, 10, 20, 50 or 100 cc vial.
[0263] The lyophilized formulations are reconstituted prior to administration. The protein may be reconstituted at a concentration of about 10, 15, 20, 25, 30, 40, 50, 60, 75, 80, 90 or 100 mg/mL or higher concentrations such as 150 mg/mL, 200 mg/mL, 250 mg/mL, or 300 mg/mL up to about 500 mg/mL. In one embodiment, the protein concentration after reconstitution is about 10-300 mg/mL. In one embodiment, the protein concentration after reconstitution is about 20-250 mg/mL. In one embodiment, the protein concentration after reconstitution is about 150-250 mg/mL. In one embodiment, the protein concentration after reconstitution is about 180-220 mg/mL. In one embodiment, the protein concentration after reconstitution is about 50-150 mg/mL. In one embodiment, the protein concentration after reconstitution is about 150 mg/mL. In one embodiment, the protein concentration after reconstitution is about 125 mg/mL. In one embodiment, the protein concentration after reconstitution is about 100 mg/mL. In one embodiment, the protein concentration after reconstitution is about 75 mg/mL. In one embodiment, the protein concentration after reconstitution is about 50 mg/mL. In one embodiment, the protein concentration after reconstitution is about 25 mg/mL. High protein concentrations are particularly useful where subcutaneous delivery of the reconstituted formulation is intended. However, for other routes of administration, such as intravenous administration, lower concentrations of the protein may be desired (e.g., from about 5-50 mg/rnL).
[0264] Reconstitution generally takes place at a temperature of about 25 °C to ensure complete hydration, although other temperatures may be employed as desired. The time required for reconstitution will depend, e.g., on the type of diluent, amount of excipient(s) and protein. Exemplary diluents include sterile water, bacteriostatic water for injection (BWFI), a pH buffered solution (e.g., phosphate-buffered saline), sterile saline solution, Ringer’s solution or dextrose solution.
Methods of Use
[0265] In another aspect, provided herein is a method of treating cancer in a subject, comprising administering a therapeutically effective amount of any formulation described herein to the subject. In some specific embodiments of this method, the formulation is administered to the subject via intravenous administration. In other embodiments, the formulation is administered to the subject by subcutaneous administration. In one embodiment, the invention comprises a method of treating cancer in a human patient comprising administering any formulation described herein to the patient.
[0266] In any of the methods of the invention, the cancer can be selected from the group consisting of: melanoma, lung cancer, head and neck cancer, bladder cancer, breast cancer, gastrointestinal cancer, multiple myeloma, hepatocellular cancer, lymphoma, renal cancer, mesothelioma, ovarian cancer, esophageal cancer, anal cancer, bi 1 i ary tract cancer, colorectal cancer, cervical cancer, thyroid cancer, salivary cancer, prostate cancer (e.g., hormone refractory prostate adenocarcinoma), pancreatic cancer, colon cancer, esophageal cancer, liver cancer, thyroid cancer, glioblastoma, glioma, and other neoplastic malignancies.
[0267] In some embodiments the lung cancer in non-small cell lung cancer.
[0268] In alternate embodiments, the lung cancer is small-cell lung cancer.
[0269] In some embodiments, the lymphoma is Hodgkin lymphoma.
[0270] In other embodiments, the lymphoma is non-Hodgkin lymphoma. In particular embodiments, the lymphoma is mediastinal large B-cell lymphoma.
[0271] In some embodiments, the breast cancer is triple negative breast cancer.
[0272] In further embodiments, the breast cancer is ER+/HER2- breast cancer.
[0273] In some embodiments, the bladder cancer is urothelial cancer. [0274] In some embodiments, the head and neck cancer is nasopharyngeal cancer. In some embodiments, the cancer is thyroid cancer. In other embodiments, the cancer is salivary cancer. In other embodiments, the cancer is squamous cell carcinoma of the head and neck.
[0275] In some embodiments, the cancer is a solid tumor with a high level of microsatellite instability (MSI-H).
[0276] In some embodiments, the cancer is a solid tumor with a high mutational burden.
[0277] In some embodiments, the cancer is metastatic colorectal cancer with high levels of microsatellite instability (MSI-H).
[0278] In some embodiments, the cancer is selected from the group consisting of colorectal cancer, esophageal cancer, melanoma, non-small cell lung cancer, ovarian cancer, renal cell cancer and small cell lung cancer.
[0279] In other embodiments of the above treatment methods, the cancer is a hematological malignancy. In certain embodiments, the Heme malignancy is acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), diffuse large B-cell lymphoma (DLBCL), EBV-positive DLBCL, primary mediastinal large B-cell lymphoma, T-cell/histiocyte-rich large B-cell lymphoma, follicular lymphoma, Hodgkin’s lymphoma (HL), mantle cell lymphoma (MCL), multiple myeloma (MM), myeloid cell leukemia-1 protein (Mcl-1), myelodysplastic syndrome (MDS), non-Hodgkin lymphoma (NHL), or small lymphocytic lymphoma (SLL).
[0280] Malignancies that demonstrate improved disease-free and overall survival in relation to the presence of tumor-infdtrating lymphocytes in biopsy or surgical material, e.g., melanoma, colorectal, liver, kidney, stomach/esophageal, breast, pancreas, and ovarian cancer are encompassed in the methods and treatments described herein. Such cancer subtypes are known to be susceptible to immune control by T lymphocytes. Additionally, included are refractory or recurrent malignancies whose growth may be inhibited using the antibodies described herein.
[0281] Additional cancers that can benefit from treatment with the formulations described herein include those associated with persistent infection with viruses such as human immunodeficiency viruses, hepatitis viruses class A, B and C, Epstein Barr virus, human papilloma viruses that are known to be causally related to for instance Kaposi’s sarcoma, liver cancer, nasopharyngeal cancer, lymphoma, cervical, vulval, anal, penile and oral cancers.
[0282] The formulations can also be used to prevent or treat infections and infectious disease. Thus, the invention provides a method for treating chronic infection in a mammalian subject comprising administering an effective amount of a formulation of the invention to the subject. In some specific embodiments of this method, the formulation is administered to the subject via intravenous administration. In other embodiments, the formulation is administered to the subject by subcutaneous administration.
[0283] These agents can be used alone, or in combination with vaccines, to stimulate the immune response to pathogens, toxins, and self-antigens. The antibodies or antigen-binding fragment thereof can be used to stimulate immune response to viruses infectious to humans, including but not limited to: human immunodeficiency viruses, hepatitis viruses class A, B and C, Epstein Barr virus, human cytomegalovirus, human papilloma viruses, and herpes viruses. Antagonist anti-PD-1 antibodies or antibody fragments can be used to stimulate immune response to infection with bacterial or fungal parasites, and other pathogens. Viral infections with hepatitis B and C and HIV are among those considered to be chronic viral infections.
[0284] The formulations of the invention may be administered to a patient in combination with one or more “additional therapeutic agents”. The additional therapeutic agent may be a biotherapeutic agent (including but not limited to antibodies to VEGF, EGFR, Her2/neu, VEGF receptors, other grow th factor receptors, CD20, CD40, CD-40L, OX-40, 4-1BB, and ICOS), an immunogenic agent (for example, attenuated cancerous cells, tumor antigens, antigen presenting cells such as dendritic cells pulsed with tumor derived antigen or nucleic acids, immune stimulating cytokines (for example, IL-2, IFNa2, GM-CSF), and cells transfected with genes encoding immune stimulating cy tokines such as but not limited to GM-CSF).
[0285] As noted above, in some embodiments of the methods of the invention, the method further comprises administering an additional therapeutic agent. In particular embodiments, the additional therapeutic agent is an anti-PD-1 antibody or antigen-binding fragment thereof, an anti-PD-Ll antibody or antigen binding fragment thereof, an anti-LAG3 antibody or antigen binding fragment thereof, an anti-TIGIT antibody or antigen binding fragment thereof, an anti- GITR antibody or antigen-binding fragment thereof, an anti-CTL4 antibody or antigen-binding fragment thereof, an anti-CD27 antibody or antigen-binding fragment thereof. In one embodiment, the additional therapeutic agent is a Newcastle disease viral vector expressing IL- 12. In a further embodiment, the additional therapeutic agent is dinaciclib. In still further embodiments, the additional therapeutic agent is a STING agonist.
[0286] Suitable routes of administration may, for example, include parenteral delivery, including intramuscular, subcutaneous, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal. Drugs can be administered in a variety of conventional ways, such as intraperitoneal, parenteral, intraarterial or intravenous injection. Modes of administration in which the volume of solution must be limited (e.g., subcutaneous administration) require a lyophilized formulation to enable reconstitution at high concentration.
[0287] Selecting a dosage of the additional therapeutic agent depends on several factors, including the serum or tissue turnover rate of the entity, the level of symptoms, the immunogenicity of the entity, and the accessibility of the target cells, tissue or organ in the individual being treated. The dosage of the additional therapeutic agent should be an amount that provides an acceptable level of side effects. Accordingly, the dose amount and dosing frequency of each additional therapeutic agent (e.g., biotherapeutic or chemotherapeutic agent) will depend in part on the particular therapeutic agent, the severity of the cancer being treated, and patient characteristics. Guidance in selecting appropriate doses of antibodies, cytokines, and small molecules are available. See, e.g., Wawrzynczak (1996) Antibody Therapy, Bios Scientific Pub. Ltd, Oxfordshire, UK; Kresina (ed.) (1991) Monoclonal Antibodies, Cytokines and Arthritis, Marcel Dekker, New York, NY; Bach (ed.) (1993) Monoclonal Antibodies and Peptide Therapy in Autoimmune Diseases, Marcel Dekker, New Y ork, NY ; Baert et al. (2003) New Engl. J. Med. 348:601-608; Milgrom et al. (1999) New Engl. J. Med. 341 : 1966-1973; Slamon eNr/. (2001) New Engl. J. Med. 344:783-792; Beniaminovitz et al. (2000) New Engl. J. Med. 342:613-619; Ghosh et al. (2003) New Engl. J. Med. 348:24-32; Lipsky et al. (2000) New Engl. J. Med. 343: 1594-1602; Physicians’ Desk Reference 2003 (Physicians’ Desk Reference, 57th Ed);
Medical Economics Company; ISBN: 1563634457; 57th edition (November 2002). Determination of the appropriate dosage regimen may be made by the clinician, e.g., using parameters or factors known or suspected in the art to affect treatment or predicted to affect treatment, and will depend, for example, the patient’s clinical history (e.g., previous therapy), the ty pe and stage of the cancer to be treated and biomarkers of response to one or more of the therapeutic agents in the combination therapy.
[0288] Various literature references are available to facilitate selection of pharmaceutically acceptable carriers or excipients for the additional therapeutic agent. See, e.g., Remington ’s Pharmaceutical Sciences and U.S. Pharmacopeia: National Formulary, Mack Publishing Company, Easton, PA (1984); Hardman et al. (2001) Goodman and Gilman ’s The Pharmacological Basis of Therapeutics , McGraw-Hill, New York, NY; Gennaro (2000) Remington: The Science and Practice of Pharmacy, Lippincott, Williams, and Wilkins, New York, NY; Avis et al. (eds.) (1993) Pharmaceutical Dosage Forms: Parenteral Medications, Marcel Dekker, NY; Lieberman, et al. (eds.) (1990) Pharmaceutical Dosage Forms: Tablets, Marcel Dekker, NY; Lieberman et al. (eds.) (1990) Pharmaceutical Dosage Forms: Disperse Systems, Marcel Dekker, NY; Weiner and Kotkoskie (2000) Excipient Toxicity and Safety, Marcel Dekker, Inc., New York, NY.
[0289] A pharmaceutical antibody formulation can be administered by continuous infusion, or by doses at intervals of, e.g., one day, 1-7 times per week, one week, two weeks, three weeks, monthly, bimonthly, etc. A preferred dose protocol is one involving the maximal dose or dose frequency that avoids significant undesirable side effects. A total weekly dose is generally at least 0.05 pg/kg, 0.2 pg/kg, 0.5 pg/kg, 1 pg/kg, 10 pg/kg, 100 pg/kg, 0.2 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 10 mg/kg, 25 mg/kg, 50 mg/kg body weight or more. See, e.g., Yang et al. (2003) New Engl. J. Med. 349:427-434; Herold et al. (2002) New Engl. J. Med. 346: 1692-1698; Liu et al. (1999) Neurol. Neurosurg. Psych. 67:451-456; Portielji et al. (20003) Cancer Immunol. Immunother. 52: 133-144. The desired dose of a small molecule therapeutic, e.g., a peptide mimetic, natural product, or organic chemical, is about the same as for an antibody or polypeptide, on a moles/kg basis.
[0290] Embodiments of the invention also include one or more of the biological formulations described herein (i) for use in, (ii) for use as a medicament or composition for, or (iii) for use in the preparation of a medicament for: (a) therapy (e.g., of the human body); (b) medicine; (c) induction of or increasing of an antitumor immune response; (d) decreasing the number of one or more tumor markers in a patient; (e) halting or delaying the growth of a tumor or a blood cancer; (1) halting or delaying the progression of a PD-1 -related disease or an ILT4-related disease; (g) stabilization of a PD-1 -related disease or an ILT4-related disease; (h) inhibiting the grow th or survival of tumor cells; (i) eliminating or reducing the size of one or more cancerous lesions or tumors; (j) reduction of the progression, onset or severity of a PD-l-related disease or an ILT4- related disease; (k) reducing the seventy or duration of the clinical symptoms of a PD-l-related or ILT4- related disease; (1) prolonging the survival of a patient relative to the expected survival in a similar untreated patient; (m) inducing complete or partial remission of a cancerous condition or other PD-l-related or ILT4- related disease; (n) treatment of cancer; or (o) treatment of chronic infections.
General Methods
[0291] Standard methods in molecular biology' are described in Sambrook, Fritsch and Maniatis (1982 & 1989 2nd Edition, 2001 3rd Edition) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY; Sambrook and Russell (2001) Molecular Cloning, 3rd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY; Wu (1993) Recombinant DNA, Vol. 217, Academic Press, San Diego, CA. Standard methods also appear in Ausbel, et al. (2001) Current Protocols in Molecular Biology, Vols.1-4, John Wiley and Sons, Inc. New York, NY, which describes cloning in bacterial cells and DNA mutagenesis (Vol. 1), cloning in mammalian cells and yeast (Vol. 2), glycoconjugates and protein expression (Vol. 3), and bioinformatics (Vol. 4).
[0292] Methods for protein purification including immunoprecipitation, chromatography, electrophoresis, centrifugation, and crystallization are described (Coligan, et al. (2000) Current Protocols in Protein Science, Vol. 1, John Wiley and Sons, Inc., New York). Chemical analysis, chemical modification, post-translational modification, production of fusion proteins, glycosylation of proteins are described (see, e.g., Coligan, et al. (2000) Current Protocols in Protein Science, Vol. 2, John Wiley and Sons, Inc., New York; Ausubel, et al. (2001) Current Protocols inMolecular Biology, Vol. 3, John Wiley and Sons, Inc., NY, NY, pp. 16.0.5-16.22.17; Sigma-Aldrich, Co. (2001) Products for Life Science Research, St. Louis, MO; pp. 45-89;
Amersham Pharmacia Biotech (2001) BioDirectory, Piscataway, N.J., pp. 384-391). Production, purification, and fragmentation of polyclonal and monoclonal antibodies are described (Coligan, et al. (2001) Current Protocols in Immunology, Vol. 1, John Wiley and Sons, Inc., New York; Harlow and Lane (1999) Using Antibodies , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY; Harlow and Lane, supra). Standard techniques for characterizing ligand/receptor interactions are available (see, e.g., Coligan, et al. (2001) Current Protocols in Immunology, Vol. 4, John Wiley, Inc., New York).
[0293] Monoclonal, polyclonal, and humanized antibodies can be prepared (see, e.g., Sheperd and Dean (eds.) (2000) Monoclonal Antibodies, Oxford Univ. Press, New York, NY;
Kontermann and Dubel (eds.) (2001) Antibody Engineering, Springer-Verlag, New York; Harlow and Lane (1988) Antibodies A Laboratory Manual, Cold Spring Harbor Laboratory' Press, Cold Spring Harbor, NY, pp. 139-243; Carpenter, et al. (2000) J. Immunol. 165:6205; He, et al. (1998) J. Immunol. 160:1029; Tang et al. (1999) J. Biol. Chem. 274:27371-27378; Baca et a/. (1997) J. Biol. Chem. 272:10678-10684; Chothia ef a/. (1989) Nature 342:877-883; Foote and Winter (1992) J. Mol. Biol. 224:487-499; U.S. Pat. No. 6,329,511).
[0294] An alternative to humanization is to use human antibody libraries displayed on phage or human antibody libraries in transgenic mice (Vaughan et al. (1996) Nature Biotechnol. 14:309- 314; Barbas (1995) Nature Medicine 1:837-839; Mendez et al. (1997) Nature Genetics 15:146- 156; Hoogenboom and Chames (2000) Immunol. Today 21.Ml-MT, Barbas et al. (2001) Phage Display: A Laboratory Manual, Cold Spring Harbor Laboratory' Press, Cold Spring Harbor, New York; Kay et al. (1996) Phage Display of Peptides and Proteins: A Laboratory Manual, Academic Press, San Diego, CA; de Bruin et al. (1999) Nature Biotechnol. 17:397-399).
[0295] Purification of antigen is not necessary for the generation of antibodies. Animals can be immunized with cells bearing the antigen of interest. Splenocytes can then be isolated from the immunized animals, and the splenocytes can fused with a myeloma cell line to produce a hybridoma (see, e.g., Meyaard et al. (1997) Immunity 7:283-290; Wright et al. (2000) Immunity 13:233-242; Preston et al., supra,' Kaithamana et al. (1999) J. Immunol. 163:5157-5164).
[0296] Antibodies can be conjugated, e.g., to small drug molecules, enzymes, liposomes, polyethylene glycol (PEG). Antibodies are useful for therapeutic, diagnostic, kit or other purposes, and include antibodies coupled, e.g., to dyes, radioisotopes, enzymes, or metals, e.g., colloidal gold (see, e.g., Le Doussal et al. (1991) J. Immunol. 146: 169-175; Gibellini et al.
(1998) J. Immunol. 160:3891-3898; Hsing and Bishop (1999) J. Immunol. 162:2804-2811; Everts et al. (2002) J. Immunol. 168:883-889).
[0297] Methods for flow cytometry, including fluorescence activated cell sorting (FACS), are available (see, e.g., Owens, et al. (1994) Flow Cytometry Principles for Clinical Laboratory Practice, John Wiley and Sons, Hoboken, NJ; Givan (2001) Flow Cytometry, 2nd ed:, Wiley-Liss, Hoboken, NJ; Shapiro (2003) Practical Flow Cytometry, John Wiley and Sons, Hoboken, NJ). Fluorescent reagents suitable for modifying nucleic acids, including nucleic acid primers and probes, polypeptides, and antibodies, for use, e.g, as diagnostic reagents, are available (Molecular Probesy (2003) Catalogue, Molecular Probes, Inc., Eugene, OR; Sigma- Aldrich (2003) Catalogue, St. Louis, MO).
[0298] Standard methods of histology of the immune system are described (see, e.g., Muller- Harmelink (ed.) (1986) Human Thymus: Histopathology and Pathology, Springer Verlag, New York, NY; Hiatt, et al. (2000) Color Atlas of Histology, Lippincott, Williams, and Wilkins, Phila, PA; Louis, et al. (2002) Basic Histology: Text and Atlas, McGraw-Hill, New York, NY).
[0299] Software packages and databases for determining, e.g., antigenic fragments, leader sequences, protein folding, functional domains, glycosylation sites, and sequence alignments, are available (see, e.g, GenBank, Vector NTI® Suite (Informax, Inc, Bethesda, MD); GCG Wisconsin Package (Accelrys, Inc., San Diego, CA); DeCypher® (TimeLogic Corp., Crystal Bay, Nevada); Menne, et al. (2000) Bioinformatics 16: 741-742; Menne, et al. (2000) Bioinformatics Applications Note 16:741-742; Wren, et al. (2002) Comput. Methods Programs Biomed. 68: 177-181; von Heijne (1983) Eur. J. Biochem. 133: 17-21; von Heijne (1986) Nucleic Acids Res. 14:4683-4690).
Analytical Methods [0300] Analytical methods suitable for evaluating the product stability include size exclusion chromatography (SEC), dynamic light scattering test (DLS), differential scanning calorimetry (DSC), iso-asp quantification, potency, UV at 340 nm, UV spectroscopy, and FTIR. SEC (J. Pharm. Scien., 83: 1645-1650, (1994); Pharm. Res., 11 :485 (1994); J. Pharm. Bio. Anal., 15: 1928 (1997); J. Pharm. Bio. Anal., 14: 1133-1140 (1986)) measures percent monomer in the product and gives information of the amount of soluble aggregates. DSC (Pharm. Res., 15:200 (1998); Pharm. Res., 9: 109 (1982)) gives information of protein denaturation temperature and glass transition temperature. DLS (American Lab., November (1991)) measures mean diffusion coefficient, and gives information of the amount of soluble and insoluble aggregates. UV at 340 nm measures scattered light intensity at 340 nm and gives information about the amounts of soluble and insoluble aggregates. UV spectroscopy measures absorbance at 278 nm and gives information of protein concentration. FTIR (Eur. J. Pharm. Biopharm., 45:231 (1998); Pharm. Res., 12: 1250 (1995); J. Pharm. Scien., 85: 1290 (1996); J. Pharm. Scien., 87:1069 (1998)) measures IR spectrum in the amide one region, and gives information of protein secondary structure.
[0301] The iso-asp content in the samples is measured using the Isoquant Isoaspartate Detection System (Promega). The kit uses the enzyme Protein Isoaspartyl Methyltransferase (PIMT) to specifically detect the presence of isoaspartic acid residues in a target protein. PIMT catalyzes the transfer of a methyl group from S-adenosyl-L-methionine to isoaspartic acid at the .alpha. -carboxyl position, generating S-adenosyl-L-homocysteine (SAH) in the process. This is a relatively small molecule, and can usually be isolated and quantitated by reverse phase HPLC using the SAH HPLC standards provided in the kit.
[0302] The potency or bioidentity of an antibody can be measured by its ability to bind to its antigen. The specific binding of an antibody to its antigen can be quantitated by any method known to those skilled in the art, for example, an immunoassay, such as ELISA (enzyme-linked immunosorbent assay).
[0303] All publications mentioned herein are incorporated by reference for the purpose of describing and disclosing methodologies and materials that might be used in connection with the invention.
[0304] Having described different embodiments of the invention herein with reference to the accompanying drawings, it is to be understood that the invention is not limited to those precise embodiments, and that various changes and modifications may be effected therein by one skilled in the art without departing from the scope or spirit of the invention as defined in the appended claims. [0305] Embodiment 1 provides a formulation of an anti-human immunoglobulin-like transcnpt 4 (anti-ILT4) antibody or antigen-binding fragment thereof, comprising: (i) about 10 mg/rnL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM of a buffer; (iii) about 6% to about 8% weight /volume (w/v) of a nonreducing sugar; (iv) about 0.01 % to about 0. 10% (w/v) of a non-ionic surfactant; and (v) about 1 mM to about 20 mM of an anti-oxidant, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21), wherein the buffer is an L-histidine buffer, an acetate buffer, or a citrate buffer, wherein the non-reducing sugar is a disaccharide, wherein the non- iomc surfactant is polysorbate 20 or polysorbate 80, and wherein the antioxidant is methionine. [0306] Embodiment 2 provides the formulation of embodiment 1, wherein the buffer is an L- histidine buffer.
[0307] Embodiment 3 provides the formulation of embodiment 1 or 2, wherein the nonreducing sugar is sucrose.
[0308] Embodiment 4 provides the formulation of any one of embodiments 1-3, wherein the non-ionic surfactant is polysorbate 80.
[0309] Embodiment 5 provides the formulation of any one of embodiments 1-4, wherein the anti-oxidant is L-methionine.
[0310] Embodiment 6 provides formulation of an anti-human immunoglobulin-like transcript 4 (“anti-ILT4”) antibody or antigen-binding fragment thereof, comprising: (i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 5 mM to about 20 mM L-histidine buffer; (iii) about 6% to about 8% weight/volume (w/v) sucrose; (iv) about 0.01 % to about 0. 10% (w/v) polysorbate 80; and (v) about 1 mM to about 20 mM L- methionine, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21). [0311] Embodiment 7 provides the formulation of embodiment 6, comprising about 8 mM to about 12 mM L-histidine buffer.
[0312] Embodiment 8 provides the formulation of embodiment 6 or 7, comprising about 5 mM to about 10 mM L-methionine.
[0313] Embodiment 9 provides the formulation of any of embodiments 6-8, comprising about 0.01 % to about 0.05% (w/v) polysorbate 80.
[0314] Embodiment 10 provides the formulation of any of embodiments 1-9, comprising about 10 mg/mL to about 150 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof.
[0315] Embodiment 11 provides the formulation of embodiment 10, wherein the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL, about 12.5 mg/mL, about 25 mg/mL, about 50 mg/mL, about 75 mg/mL, about 100 mg/mL, about 125 mg/mL, or about 150 mg/mL.
[0316] Embodiment 12 provides the formulation of any of embodiments 1-11, comprising about 25 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L- histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
[0317] Embodiment 13 provides the formulation of any of embodiments 1-11, comprising about 50 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L- histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
[0318] Embodiment 14 provides the formulation of any of embodiments 1-11, comprising about 75 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L- histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
[0319] Embodiment 15 provides the formulation of any of embodiments 1-11, comprising about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
[0320] Embodiment 16 provides the formulation of any of embodiments 1-11, comprising about 125 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L- methionine.
[0321] Embodiment 17 provides the formulation of any one of embodiments 1-16, wherein the formulation has a pH of about 5.0 to about 6.8.
[0322] Embodiment 18 provides the formulation of any one of embodiments 1-16, wherein the formulation has a pH of about 5.5 to about 6.0.
[0323] Embodiment 19 provides the formulation of any one of embodiments 1-16, wherein the formulation has a pH of about 5.5.
[0324] Embodiment 20 provides the formulation of an anti-human immunoglobulin-like transcript 4 (“anti-ILT4”) antibody or antigen-binding fragment thereof, comprising: (i) about 50 mg/rnL to about 100 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof; (ii) about 10 mM L-histidine buffer, pH about 5.5; (iii) about 7% weight/volume (w/v) sucrose; (iv) about 0.025% (w/v) polysorbate 80; and (v) about 10 mM L-methionme, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0325] Embodiment 21 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO:47), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0326] Embodiment 22 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1 : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1 : TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GDSNRPS(SEQ ID NO: 52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
[0327] Embodiment 23 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising the ammo acid sequence set forth in SEQ ID NO:57 and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58. [0328] Embodiment 24 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:2 and a light chain comprising the amino acid sequence set forth in SEQ ID NO:7.
[0329] Embodiment 25 provides the formulation of any embodiments of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 80 and a light chain comprising the amino acid sequence set forth in SEQ ID NO:7.
[0330] Embodiment 26 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO: 57 and a light chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:58.
[0331] Embodiment 27 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO:2 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
[0332] Embodiment 28 provides the formulation of any one of embodiments 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO: 80 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
[0333] Embodiment 29 provides the formulation of any one of embodiments 1-28, wherein the formulation is a liquid formulation stored at about 3°C to about 5°C, is frozen to -20°C or below, is frozen to -70°C or below, or is a reconstituted solution from a lyophilized formulation.
[0334] Embodiment 30 provides the formulation of any one of embodiments 1-27, wherein after storage at about 3°C to about 5°C for 6 months: (i) the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultraperformance size exclusion chromatography; (ii) the turbidity of the formulation is at most about 0. 135 as measured by OD350-500; (iii) the % main peak of the anti-ILT4 antibody or antigenbinding fragment thereof is at least about 63%, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, and/or the basic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography; (iv) the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging; and/or (v) the % oxidation of one or more amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO: 2 or 80 is less than about 4%, as determined by reduced peptide mapping.
[0335] Embodiment 31 provides the formulation of any of embodiments 1-30, wherein the anti- ILT4 antibody or antigen-binding fragment thereof is a monoclonal antibody.
[0336] Embodiment 32 provides a method of treating a cancer in a human patient in need thereof, the method comprising administering an effective amount of the formulation of any one of embodiment 1-31.
[0337] Embodiment 33 provides the method of embodiment 32, wherein the cancer is selected from the group consisting of colorectal cancer, esophageal cancer, melanoma, non-small cell lung cancer, ovarian cancer, renal cell cancer and small cell lung cancer.
[0338] Embodiment 34 provides the use of the formulation of any one of embodiments 1-31 for preparing a medicament for treating a cancer.
[0339] Embodiment 35 provides the use of the formulation of any one of embodiments 1-31 for the treatment of cancer in a human patient.
[0340] Embodiment 36 provides the use of embodiment 34, wherein the cancer is selected from the group consisting of colorectal cancer, esophageal cancer, melanoma, non-small cell lung cancer, ovarian cancer, renal cell cancer and small cell lung cancer.
EXAMPLE 1
Anti-ILT4 Pre-formulation Analysis
[0341] Table 3 summarizes the pre-formulation developability plan. A fully human anti-ILT4 monoclonal antibody, 1E1 with a human lambda light chain constant domain and a human IgG4- S228P heavy chain constant domain (“Antibody 1”), was tested. All formulations were prepared in 96-well Axygen Polypropylene deep well plates (Product # P-DW-l l-C) using the JANUS automated liquid handling system equipped with the Parisian pipetting arm. There was 500 pL aliquot in each well with a final concentration of 1 mg/mL Antibody 1.
Table 3. Anti-ILT4 Pre-formulation Developability Plan
Figure imgf000070_0001
Figure imgf000071_0001
[0342] A 200 pL aliquot was removed from the plate for initial sample analysis (20 pL for Differential Scanning Fluorimetry (DSF) thermal transition analysis and 180 pL for sub-visible particle analysis using Guava® easyCyte flow cytometer). The remaining 300 pL sample in the 96-well Axygen plate was covered and tightly sealed with a Nunc™ microplate sealing cap mat (Product # 276000). The plate was double-packed in SCS Dri-Shield® 2000 Series Moisture Barrier Bags (Product # 700810) to prevent evaporation. The sealed plate was subjected to stress for 10 days at 50°C in a thermal chamber. Protein concentration was monitored before and after thermal stress to account for any increase in protein concentration due to evaporation. The increase in protein concentration across the plate was less than 5%. All the standard precautions were observed during handling of protein solution (e g., equilibration of the solution to room temperature, minimal shaking and exposure to room light).
[0343] After the thermal stress, the plate was equilibrated at room temperature for 30 minutes. The plate was then carefully opened in a laminar hood. Using 200 pL multi-channel pipette, the stressed sample was mixed up to 5 times. Then 200 pL of sample was removed and dispensed to a 96-well costar clear plate (Product # 3635) and the absorbance was measured at wavelengths 280, 320, 350 and 500 nm. Following UV analysis, 180 pL of the 200 pL aliquot was carefully transferred to a Coming® 96-well clear V-bottom microplate (Product #3894) for sub-visible particle analysis. The remaining 20 pL was utilized for quantitative protein purity analysis with reduced high-throughput CE-SDS using Caliper’s LabChip GXII system. On the same day, the remaining 100 pL stressed sample in the Axygen deep well plate was transferred to ABgene 96- well PCR plate (Product # AB-2800) along with initial (unstressed) sample for UP-SEC and cIEF analysis.
Thermal stability by DSF
[0344] The thermal stability of the Antibody 1 formulation was evaluated by DSF under different buffer, pH, and ionic strength conditions. Tml (first unfolding midpoint) for majority of the tested conditions fell in the range from 60-66°C (data not shown). Overall, the results from this assay demonstrated that there are no liabilities in terms of thermal stability.
Soluble aggregates by UP-SEC
[0345] UP-SEC analysis of stressed samples of Antibody 1 across different formulation conditions showed similar colloidal stability profile (data not shown). Antibody 1 formulations showed resistance to fragmentation and >3% soluble aggregation in formulations with pH < 5.5 (data not shown).
Sub-visible particles by Guava® easyCyte Flow Cytometer
[0346] Sub-visible particle (from 0.22 microns to 10 microns) testing for Antibody 1 was carried out by Guava® easyCyte flow cytometer. Antibody 1 showed no significant formation of sub-visible aggregates post-stress in majority of formulation compositions (data not shown). Fragmentation by CE-SDS
[0347] To obtain comprehensive understanding of proteolytic cleavage of the heavy or light chains of the anti-ILT4 monoclonal antibody, the stressed samples were also analyzed by reducing sodium dodecyl sulfate capillary electrophoresis (R-CE-SDS). Fragmentation of Antibody 1 was minimal in 10 mM Histidine with pH in the range of 5.6-6.8 or 10 niM Acetate with pH in the range of 5.0-5.6.
Chemical stability by cIEF
[0348] The chemical stability of the Antibody 1 formulation was evaluated by cIEF under different buffer, pH, and ionic strength conditions. cIEF profile showed a decrease in relative main peak area and significant increase in the relative peak area of the acidic variants after 10 days at 50°C. Addition of salt did not result in any significant improvement in the stability of the protein across studied compositions. The formulation that imparted the maximum stability to Antibody 1 was identified as 10 mM Histidine in the pH range of 5.5 -6.0 or 10 mM Acetate at pH 5.6.
[0349] The stressed samples were also analyzed by UV/Vis spectrophotometry for turbidity (A350-500) to evaluate colloidal instability . The results did not trend as a function of any evaluated parameter.
[0350] The above results showed that the Antibody 1 formulation has favorable colloidal (as measured by turbidity and sub-visible particulate analysis) and thermal stability profiles, and that the unformulated antibody exhibited chemical instability and aggregation propensity. As a result, a buffer of 10 mM Histidine or 10 mM Acetate within a pH range of about 5.5-6.0 was selected for further formulation development. EXAMPLE 2
Anti-ILT4 Formulation Early Feasibility Assessment
[0351] Five anti-ILT4 monoclonal antibody formulations were prepared at 50 mg/mL concentration of Antibody 1 in the following matrix: 10 mM acetate pH 5.4, 10 mM acetate pH 5.8, 10 mM L-histidine pH 5.4, 10 mM L-histidine pH 5.8, and 10 mM L-histidine pH 5.8 with 9% (w/v) sucrose. The concentrateability of the Antibody 1 formulation was further assessed by concentrating Antibody 1 to 100 mg/mL in each of the five formulation matrixes. The formulations were tested for visual appearance, turbidity (OD350-500), viscosity, concentration, and pH. The self-associating property of the Antibody 1 in each of the five formulations was determined from the diffusion interaction parameter (ko) and relative solubility value.
[0352] The data are summarized in Table 4. The data suggest better colloidal stability (OD350- 500) of the Antibody 1 at low pH (5.4) than at high pH (5.8) in both 10 mM acetate and 10 mM histidine buffers with improved stability in the presence of 9% sucrose as stabilizer (10 mM histidine buffer, pH 5.8). Overall, the ko values were negative, suggesting the inherent property of the antibody to self-associate. The ko values were less negative at low pH (5.4) for both buffers, suggesting lower self-association at that pH. The presence of sucrose of stabilizer was found to lower the crowing effect in 10 mM histidine buffer at pH 5.8. Overall, the relative solubility of the Antibody 1 in 10 mM acetate buffer (pH 5.4 and pH 5.8) and in 10 mM histidine buffer (pH 5.4) was comparable. The solubility was relatively lower at higher pH (5.8) in 10 mM histidine buffer. There was no impact of pH or formulation buffer on the viscosity of the five formulations (similar values) at 100 mg/mL concentration, demonstrating ability of the molecule to concentrate beyond 50 mg/mL.
Figure imgf000074_0001
Figure imgf000074_0005
Figure imgf000074_0002
Figure imgf000074_0003
Figure imgf000074_0004
EXAMPLE 3
Anti-ILT4 Formulation Forced Degradation Studies
[0353] A forced degradation study was conducted on formulations including Antibody 1. The tested Antibody 1 formulation contained 50 mg/mL antibody, 10 mM L-histidine buffer at pH 6.0, 7%(w/v) sucrose, and 0.02%(w/v) PS-80. The Antibody 1 formulation was exposed to various stress conditions, including heat (50°C, up to 7 days), high pH (pH 10, 25°C, up to 7 days), low pH (pH 3.5, 25°C, up to 7 days), light (25°C, up to 2X ICH scale), or peroxide (0.1% tBHP, 25°C, up to 24 hours).
[0354] The stressed samples were visually inspected for color and particulate, and tested by UV, UP-SEC, NR-CE-SDS, R-CE-SDS, HP-IEX, DLS and LC-MS (intact and reduced mass, and reduced peptide mapping).
[0355] The major degradations (>10% change at the end point of a stress when comparing with the initial point) include increase of acidic variants upon heat (50°C), high pH (pH 10.0) and light exposure stresses; increases of deamidation and isomerization upon high pH stress; increases of HMW species, basic variants, Trp oxidation upon light exposure (2X) stress; and increase of Met oxidation upon light exposure and peroxide (0.1% tBHP) stresses. There was no major degradation pathway upon low pH (pH 3.5) stress.
[0356] The minor degradations (< 10% change at the end point of a stress when comparing with the initial point) include increase of HMW species upon heat, high pH, and low pH stresses; increase of LMW species upon heat, high pH, low pH, and light exposure stresses; increase of turbidity upon light exposure stress; and increase of basic variants upon peroxide stress.
[0357] The results of the light stress study are summarized in Table 5 below. The detailed results of the other stress studies are not shown.
Table 5. Analytical Summary of Light Stressed Anti-ILT4 Formulation
Figure imgf000075_0001
Figure imgf000076_0001
[0358] Impact on potency was detected by ELISA upon light exposure at 2X ICH scale. The GeoMean values were 106 and 28 for dark control and 2X exposure stressed sample, respectively, suggesting that light exposure decreases the potency of Antibody 1.
EXAMPLE 4
Anti-ILT4 Formulation Early Stability Studies
[0359] Early feasibility data demonstrated preference for lower pH (5.4+0.2) based on better colloidal, biophysical and chemical stability (Example 2). Based on the forced degradation studies (Example 3), light oxidation was known to be one of the primary liabilities of the anti- 1LT4 monoclonal antibody. Four different formulations for Antibody 1 were staged on early thermal stability at 50 mg/mL antibody concentration at target pH of 5.5. Two formulation matrixes consisted of 10 mM histidine (with or without 5 rnM L-methionine as an antioxidant) and the other two consisted of 10 mM acetate (with or without 5 mM L-methionine as an antioxidant); each of the four formulations were prepared with 7% (w/v) sucrose as a stabilizer and 0.025% w/v polysorbate 80 (PS-80) as a surfactant at pH 5.5. The 10 mM L-histidine buffer consists of 0.29 mg/mL L-histidine and 1.71 mg/mL L-histidine monohydrochloride monohydrate to obtain target formulation pH of 5.5. The 10 mM L-methionine in the formulation is equivalent to 1.49 mg/mL L-methionine whereas the 7% (w/v) sucrose and 0.025% (w/v) polysorbate 80 are equivalent to 70 mg/mL sucrose and 0.25 mg/mL PS-80 in the formulation, respectively. The formulations were staged up to 6 months on stability under 1CH conditions: 5°C (5°C±3°C), 25°C (25°C, 60% RH), and 40°C (40°C, 75% RH). The stability plan is provided in Table 6 below. The four formulations were tested for visual appearance, pH, protein concentration (UVzso), and colloidal stability by turbidity (OD350-500) measurements, and purity by ultra-performance size exclusion chromatography (UP-SEC), charge variants by high performance ion-exchange chromatography (HP-IEX), subvisible particle count by microflow imaging (MFI) and % change in oxidation.
Table 6. Early Stability Plan
Figure imgf000077_0001
[0360] The early stability data for the four anti-ILT4 monoclonal antibody formulations are summarized in Tables 7-10. The stability trends for the four formulations are provided in FIGS. 1A-1I and FIGS. 2A-2I. As seen in these figures, Antibody 1 showed similar stability profiles in both acetate and L-histidine buffers at pH 5.5 in the presence of 7% (w/v) sucrose as a stabilizer and 0.025% (w/v) polysorbate 80 as a surfactant under all three stability conditions (5°C, 25°C, and 40°C). The degradation rate was high at accelerated condition of 25°C and even higher at stressed condition of 40°C for all assays. The Antibody 1 formulations were stable up to 6 months at 5°C storage as per the stability plan. The presence of 5 mM L-methionine as an antioxidant in the formulation was found to improve the colloidal stability (OD350-500) as well as reduce soluble aggregates (%HMW) to some extent under 25°C accelerated as well as 40°C stressed conditions for the anti-ILT4 monoclonal antibody formulations in the two buffer matrixes (FIGS. 1 A-1I). There was no impact of 5 mM L-methionine on the charge profile by HP-IEX (FIGS. 2A-2I). L-histidine was chosen as the lead buffer and acetate was chosen as back-up. As seen in reduced peptide mapping results of Tables 8-1 1 , the presence of 5 mM L- methionine in either acetate or L-histidine formulations was found to reduce the oxidation levels of the methionine residues (M359, M253, and M429) and to some extent of the tryptophan residues (W102, and W7) upon thermal stress, especially for the L-histidine formulation. Table 7. Early Stability (L-Histidine Formulation pH 5.5)
Figure imgf000078_0001
Table 8. Early Stability (L-Histidine + L-Methionine Formulation pH 5.5)
Figure imgf000079_0001
Table 9. Early Stability (Acetate Formulation pH 5.5)
Figure imgf000080_0001
Table 10. Early Stability (Acetate + L-Methionine Formulation pH 5.5)
Figure imgf000081_0001
EXAMPLE 5
Anti-ILT4 Formulation pH Range Stability Study
[0361] The pH ranging studies were conducted to determine the Antibody 1 drug product stability ±0.5 pH units from the target formulation pH of 5.5, i.e., at pH 5.0 or at pH 6.0, and to help define pH specifications for formal stability. Four Antibody 1 pH ranging formulations were staged up to 6 months on stability under ICH conditions: 5°C (5°C 3°C), 25°C (25°C, 60% RH), and 40°C (40°C, 75% RH). Two formulations were prepared in 10 mM histidine buffer, with 7% (w/v) sucrose as a stabilizer and 0.025% (w/v) polysorbate 80 as a surfactant; one formulation at pH 5.0 and the other at pH 6.0. Two formulations were prepared in 10 mM acetate buffer, with 7% (w/v) sucrose as a stabilizer and 0.025% (w/v) polysorbate 80 as a surfactant; one formulation at pH 5.0 and the other at pH 6.0. The purified Antibody Iwas dialyzed against each of the buffers, the pH of the final formulation was measured following the dialysis, and no pH adjustments were made after this step. The final formulated drug substance was filtered to obtain drug product bulk which were subsequently filled into 2 mL glass vials, rubber stoppered, and sealed. The stability plan was the same as shown in Table 6. The four formulations were tested for visual appearance, pH, protein concentration (UV280), colloidal stability by turbidity (OD350- 500) measurements, purity by UP-SEC, charge variants by HP-IEX, and subvisible particle count by MFI.
[0362] The data are summarized in Tables 11-14. The stability trends for the four Antibody IpH ranging formulations are provided in FIGS. 3A-3I and FIGS. 4A-4I, and the data from early stability study at pH 5.5 is included to enable comparison. As seen in these figures, there was no significant change in any of the extreme pH conditions (pH 5.0 and pH 6.0) in comparison with the target pH 5.5 for the anti-lLT4 monoclonal antibody formulations in either acetate or histidine buffer matrix at 5°C storage. The anti-ILT4 monoclonal antibody was found to be stable within ± 0.5 units of the target pH 5.5. At accelerated condition of 25°C and at stressed condition of 40°C, the anti-ILT4 monoclonal antibody showed better colloidal (OD350-500), physical (%HMW and %monomer) and chemical stability at the lower pH (5.0), followed by the target pH (5.5) and the higher pH (6.0). Based on these data set, pH 5.5 was selected as the anti-ILT4 monoclonal antibody formulation pH. Table 11. pH Ranging Studies (L-Histidine Formulation pH 5.0)
Figure imgf000083_0001
Table 12. pH Ranging Studies (L-Histidine Formulation pH 6.0)
Figure imgf000084_0001
Table 13. pH Ranging Studies (Acetate Formulation pH 5.0)
Figure imgf000085_0001
Table 14. pH Ranging Studies (Acetate Formulation pH 6.0)
Figure imgf000086_0001
EXAMPLE 6
Anti-ILT4 Formulation Surfactant Ranging Study
[0363] Surfactant ranging studies of the anti-ILT4 monoclonal antibody formulations including Antibody 1 were carried out using polysorbate 80 (PS-80) to analyze the sensitivity of the antibody to agitation stress in the presence or absence of the surfactant. The anti-ILT4 monoclonal antibody formulations consisted of 50 mg/mL Antibody 1 in 10 rnM histidine buffer at pH 5.5 and 7% (w/v) sucrose. Five different formulations were prepared, each covering a series of polysorbate 80, i.e., 0 % (w/v) or 0 mg/mL PS-80, 0.01% (w/v) or 0.1 mg/mL PS-80, 0.025% (w/v) or 0.25 mg/mL PS-80, 0.05% (w/v) or 0.5 mg/mL PS-80, and 0.1% (w/v) or 1 mg/mL PS-80. The 2 mL glass vials were filled with 2.2 mL of the anti-ILT4 filtered bulk drug product, rubber stoppered, and sealed. The anti-ILT4 drug product vials were agitated at 300 rpm on Thermo Scientific Shaker up to 3 days or up to 7 days placed horizontally (at ambient conditions). The same five anti-ILT4 formulations (PS-80 ranging) were placed up to 7 days at ambient room temperature without shaking and were used as respective study controls to eliminate temperature effect. All the vials (stressed and ambient control) were covered with aluminum foil to eliminate light effect. The samples were analyzed for visual appearance, protein concentration (UV280), turbidity (OD350-500), subvisible particulates by MFI, purity by UP-SEC, and charge variants by HP-IEX.
[0364] The data are summarized in Figures 7 and 8. The stability trends for the five anti-ILT4 monoclonal antibody surfactant ranging formulations are provided in FIGS. 5A-5G. As seen in these figures, the absence of surfactant resulted in generation of visible particles from 3 days of agitation up to 7 days of agitation with increasing amounts. Few visible particles were also seen in the 7 days ambient control sample. There was an increase in turbidity (OD350-500), an increase in subvisible particle counts (> 2 pm, > 5 pm, > 10 pm and > 25 pm), as well as an increase in soluble aggregate levels (%HMW). In the presence of 0.1 mg/mL [0.01% (w/v)], 0.25 mg/mL [0.025 % (w/v)], 0.5 mg/mL [0.5 % (w/v)] or 1.0 mg/mL [0.1% (w/v)] surfactant, there was not a significant difference in visual appearance, turbidity (OD350-500) or soluble aggregate levels (%HMW). Up to 7 days of agitation, a certain degree of increase in subvisible particles was seen with > 0.5 mg/mL PS-80 concentration.
[0365] The data suggest that the surfactant plays a crucial role in prevention of agitation- induced aggregates. After addition of the surfactant, all the stability-indicating attributes became comparable. Overall, 0.25 mg/mL PS-80 appeared to minimize particle content, compared to 0.5 mg/mL or 1 mg/mL PS-80. Therefore, adding a higher level of surfactant did not yield any significant advantage. As a result, a PS-80 concentration of 0.25 mg/mL was selected for the L- histidine formulation containing 50 mg/mL of anti-ILT4 monoclonal antibody.
EXAMPLE 7
Anti-ILT4 Formulation Light Stress Study Using 10 mM L-Methionine
[0366] During the forced degradation studies, the major degradation pathway of the anti-ILT4 monoclonal antibody was confirmed as light-induced oxidation, impacting potency of the anti- ILT4 monoclonal antibody (Example 3). The early stability data (Example 4) suggested the benefit of 5 mM L-methionine in reducing oxidation of L-methionine as well as of L-tryptophan residues upon thermal stress. In an attempt to further explore the impact of higher (10 mM) L- methionine content in the anti-ILT4 monoclonal antibody L-histidine formulation, the light stress studies were conducted.
[0367] Based on the early stability data, 10 mM L-histidine at pH 5.5 was finalized as the buffer-pH matrix for formulation development activities. Two L-histidine formulations were tested. One formulation (H) contained 50 mg/mL of the anti-ILT4 monoclonal antibody, 10 mM L-histidine at pH 5.5, 7% (w/v) sucrose, and 0.025% (w/v) polysorbate 80; the other formulation (H+M) contained 50 mg/mL of the anti-ILT4 monoclonal antibody, 10 mM L-histidine at pH 5.5, 10 mM L-methionine, 7% (w/v) sucrose, and 0.025% (w/v) polysorbate 80. A third formulation (HC) consisted of 50 mg/mL of the anti-ILT4 monoclonal antibody, 10 mM L-histidine at pH 5.5, 7% (w/v) sucrose, and 0.025% (w/v) polysorbate 80 sucrose to serve as dark control (nonlight exposed) under light stress (combination of ultraviolet light-UV and cool white light-CWL or visible light) at 0.2 ICH, 0.5 ICH or 1 ICH in a Caron Photostability Chamber at ambient room temperature condition. The dark control samples were covered with aluminum foil and placed in the same chamber as the light exposed samples to eliminate temperature effect. The samples were exposed to a cumulative amount of light exposure and were removed as exposure limits were completed. The samples were analyzed for visual appearance, protein concentration (UVzso), turbidity (OD350-500), purity by UP-SEC, charge variants by HP-IEX, oxidation levels by reduced peptide mapping (the anti-ILT4 monoclonal antibody oxidation hotspots M359, W7, M253, W102, M429), as well as potency by Binding ELISA.
[0368] As seen in FIGS. 6A-6K and Figure 9, 10 mM L-methionine was found to improve the colloidal stability (OD350-500), reduce soluble aggregate levels, maintain the monomer content (% monomer), minimize change in charged species, and minimize oxidation of L-methionine as well as L-tryptophan residues (to some extent) in the anti-ILT4 monoclonal antibody. Moreover, the potency was found to improve in the presence of 10 mM L-methionine upon exposure to extreme light conditions (1 1CH) as shown in Figure 9. Thus, the lead anti-lLT4 monoclonal antibody formulation was selected as follows: 50 mg/mL anti-ILT4 antibody, 0.29 mg/mL L-histidine, 1.71 mg/mL L-histidine monohydrochloride monohydrate, 1.49 mg/mL L-methionine, 70 mg/mL sucrose, and 0.25 mg/mL polysorbate 80.
EXAMPLE 8
Anti-ILT4 Formulation Freeze-thaw Study
[0369] The stability of the lead anti-ILT4 monoclonal antibody formulation (50 mg/mL anti- ILT4, 0.29 mg/mL L-histidine, 1.71 mg/mL L-histidine monohydrochloride monohydrate, 1.49 mg/mL L-methionine, 70 mg/mL sucrose, 0.25 mg/mL polysorbate 80, pH 5.5) was assessed under freeze-thaw stress. A 2.2 mL volume was used to fill the anti-ILT4 drug product in 2 mL Type 1 glass vial, rubber stoppered, and sealed. The drug product vials were exposed up to five freeze-thaw cycles (i.e., IX F/T, 3X F/T, and 5X F/T); each cycle consisting of freezing at -80°C following by thawing at 25°C (25°C, 60% RH chamber) with 5°C control. The samples were analyzed for visual appearance, protein concentration (UV280), turbidity (OD350-500), purity by UP-SEC, and charge variants by HP-IEX.
[0370] The lead anti-ILT4 L-histidine formulation did not show any change up to 5X freezethaw as shown in Table 15. The visual appearance, turbidity (OD350-500), subvisible particulate counts, and biochemical profiles (UP-SEC and HP-IEX) were comparable to up to 5X freezethaw stress. Overall, freeze-thaw cycling had no measurable impact on aggregation or biochemical properties of the lead anti-ILT4 formulation.
EXAMPLE 9
Anti-ILT4 Formulation Properties
[0371] The anti-lLT4 monoclonal antibody formulation properties, such as density, viscosity, osmolality, and apparent glass transition temperature (Tg ’), were determined.
[0372] Apparent glass transition temperature (Tg )
[0373] Modulated differential scanning microcalorimetry (mDSC) was used for determination of Tg . The following parameters were used during the run: temperature equilibrated at -90.0°C; modulation of ± 0.5°C every 60 seconds, isothermal for 5.0 mins with ramp up of 2.0°C/min to 25.0°C.
Osmolality
[0374] The Advanced Instruments, Inc. Osmometer was calibrated using two calibration standards (100 mOsm/kg and 500 mOsm/kg) provided by Advanced Instruments, Inc., which were measured to be 100 mOsm/kg and 500 mOsm/kg, respectively (within the acceptable limits). 250 pL of sample was taken in the sample dispenser and the osmolality was measured in triplicate.
Density
[0375] Anton Paar Densitometer was used for density measurement. The oscillating U-tube of the densitometer was rinsed with water followed by ethanol and air-dried using in-built pump. The air density in the oscillating U-tube was measured to be 0.00126 g/cm3 at 20°C prior to sample density measurement. For density measurement, 1 mL of the sample was filled into a sterile 1 mL syringe. The sample was checked for air bubbles and gently inserted via sample port into the oscillating U-tube. The densities were measured at 5°C, 20°C, and 30°C.
Viscosity
[0376] Rheosense mVROC viscometer was used for viscosity measurement. The water bath was stabilized at 20°C and a system check was performed using aquet solution. About 400 pL of sample was taken into a clean, 0.5 mL syringe and locked in the system for measurement. The viscosity was measured at 20°C using flow-rate of 50 pL/min with measurement time of 60 seconds. Viscosity values with slope fit Rsqrd value of at least 0.98 were used for recording.
[0377] The properties of the anti-ILT4 monoclonal antibody formulation, 100 mg/vial (50 mg/mL), such as apparent glass transition temperature (Tg’), osmolality, density, viscosity and extractable volume, are summarized in Table 16.
Table 16. Anti-ILT4 Monoclonal Antibody Formulation Properties
Figure imgf000090_0001
EXAMPLE 10: Ion Exchange (1EX) method to measure acidic species of anti-lLT4 antibodies [0378] For the IEX method, using a Waters Alliance LC system (Milford, MA, U.S.A.), the Thermo Scientific’s ProPac WCX-10 (p/n: 054993, particle size 10 um, diameter 4 mm, length 250 mm) was chosen with a loading of 80 pg sample. Mobile Phase (A) 24 mM MES pH 6. 1 with 4% acetonitrile, and mobile phase (B) 20 mM sodium phosphate, 95 mM NaCl pH 8.0 with 4% acetonitrile was used as a non-linear, sigmoidal shape, pH gradient, and the separation was monitored over 34 min with a flow rate of 0.5 rnL min-1, with the column temperature being 35 °C. The gradient used was: 22%-22%B for 0-0.6 min; 22%-29%B for 0.6-15.0 min; 29%- 70%B for 15.0-30.0 min; 70%-100%B for 30.0-30.5 min; and 100%-100%B from 30.5-33.0 mm. Mobile phase (C) 10 mM CHES pH 8.0, 40 mM Tns, 15 mM EDTA, 200 mM NaCl, and 4% acetonitrile was used to strip the column at 0.5 mL min-1 from 33. 1-34.0 min, followed by re-equilibration with 22%B from 34.5-44.5 min at 1.0 mL min-1. From 44.5-45 min, the flowrate was reduced to 0.5 mL min-1. The elution was monitored at 280 nm for the detection of peaks. The assay variability was determined to be within 1%.
Table 15. Drug Product Freeze- Thaw Study
Figure imgf000091_0001
Figure imgf000092_0001

Claims

WHAT IS CLAIMED IS:
1. A formulation of an anti -human immunoglobulin-like transcript 4 (anti-ILT4) antibody or antigen-binding fragment thereof, comprising:
(i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof;
(ii) about 5 mM to about 20 mM of a buffer;
(iii) about 6% to about 8% weight /volume (w/v) of a non-reducing sugar;
(iv) about 0.01 % to about 0.10% (w/v) of a non-ionic surfactant; and
(v) about 1 mM to about 20 mM of an anti-oxidant, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21), wherein the buffer is an L-histidine buffer, an acetate buffer, or a citrate buffer, wherein the non-reducing sugar is a disaccharide, wherein the non-ionic surfactant is polysorbate 20 or polysorbate 80, and wherein the antioxidant is methionine.
2. The formulation of claim 1, wherein the buffer is an L-histidine buffer.
3. The formulation of claim 1 or 2, wherein the non-reducing sugar is sucrose.
4. The formulation of any one of claims 1-3, wherein the non-ionic surfactant is polysorbate 80.
5. The formulation of any one of claims 1-4, wherein the anti-oxidant is L- methionine.
6. A formulation of an anti-human immunoglobulin-like transcript 4 (“anti-ILT4”) antibody or antigen-binding fragment thereof, comprising: (i) about 10 mg/mL to about 200 mg/mL of the anti-ILT4 antibody or antigenbinding fragment thereof;
(ii) about 5 rnM to about 20 mM L-histidine buffer;
(iii) about 6% to about 8% weight/volume (w/v) sucrose;
(iv) about 0.01 % to about 0.10% (w/v) polysorbate 80; and
(v) about 1 mM to about 20 mM L-methionine, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3:
LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
7. The formulation of claim 6, comprising about 8 mM to about 12 mM L-histidine buffer.
8. The formulation of claim 6 or 7, comprising about 5 mM to about 10 mM L- methionine.
9. The formulation of any of claims 6-8, comprising about 0.01 % to about 0.05% (w/v) polysorbate 80.
10. The formulation of any of claims 1-9, comprising about 10 mg/mL to about 150 mg/mL of the anti-ILT4 antibody or antigen-binding fragment thereof.
11. The formulation of claim 10, wherein the concentration of the anti-ILT4 antibody or antigen-binding fragment thereof is about 10 mg/mL, about 12.5 mg/mL, about 25 mg/mL, about 50 mg/mL, about 75 mg/mL, about 100 mg/mL, about 125 mg/mL, or about 150 mg/mL.
12. The formulation of any of claims 1-11, comprising about 25 mg/mL of the anti- ILT4 antibody or antigen-binding fragment thereof, about 10 rnM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
13. The formulation of any of claims 1-11, comprising about 50 mg/mL of the anti- ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
14. The formulation of any of claims 1-11, comprising about 75 mg/mL of the anti- ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
15. The formulation of any of claims 1-11, comprising about 100 mg/mL of the anti- ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
16. The formulation of any of claims 1-11, comprising about 125 mg/mL of the anti- ILT4 antibody or antigen-binding fragment thereof, about 10 mM L-histidine buffer, about 7% w/v sucrose, about 0.025% polysorbate 80, and about 10 mM L-methionine.
17. The formulation of any one of claims 1-16, wherein the formulation has a pH of about 5.0 to about 6.8.
18. The formulation of any one of claims 1-16, wherein the formulation has a pH of about 5.5 to about 6.0.
19. The formulation of any one of claims 1-16, wherein the formulation has a pH of about 5.5.
20. A formulation of an anti-human immunoglobulin-like transcript 4 (“anti-ILT4”) antibody or antigen-binding fragment thereof, comprising:
(i) about 50 mg/mL to about 100 mg/mL of the anti-ILT4 antibody or antigenbinding fragment thereof;
(ii) about 10 mM L-histidine buffer, pH about 5.5;
(iii) about 7% weight/volume (w/v) sucrose;
(iv) about 0.025% (w/v) polysorbate 80; and
(v) about 10 mM L-methionine, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR-H1: GYYWS (SEQ ID NO: 16), CDR-H2: EINHXGSTNYNPSLKS wherein X is S or A (SEQ ID NO: 17), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18), and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein XI is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
21. The formulation of any one of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR- Hl : GYYWS (SEQ ID NO: 16), CDR-H2: EINHSGSTNYNPSLKS (SEQ ID NO:47), and CDR- H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GX1X2NRPS, wherein Xi is N, Q, E or D and X2 is S or A (SEQ ID NO: 20), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
22. The formulation of any one of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising: CDR- Hl : GYYWS (SEQ ID NO: 16), CDR-H2: EINHAGSTNYNPSLKS (SEQ ID NO: 48), and CDR-H3: LPTRWVTTRYFDL (SEQ ID NO: 18); and a light chain variable domain comprising: CDR-L1: TGSSSNIGAGYDVH (SEQ ID NO: 19), CDR-L2: GDSNRPS(SEQ ID NO: 52), and CDR-L3: QSFDNSLSAYV (SEQ ID NO: 21).
23. The formulation of any one of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising the ammo acid sequence set forth in SEQ ID NO: 57 and a light chain variable domain comprising the amino acid sequence set forth in SEQ ID NO:58.
24. The formulation of any one of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:2 and a light chain comprising the amino acid sequence set forth in SEQ ID NO:7.
25. The formulation of any one of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO:80 and a light chain comprising the ammo acid sequence set forth in SEQ ID NO:7.
26. The formulation of any one of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO:57 and a light chain variable domain consisting of the amino acid sequence set forth in SEQ ID NO: 58.
27. The formulation of any one of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO:2 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
28. The formulation of any one of claims 1-20, wherein the anti-ILT4 antibody or antigen binding fragment thereof comprises a heavy chain consisting of the amino acid sequence set forth in SEQ ID NO: 80 and a light chain consisting of the amino acid sequence set forth in SEQ ID NO:7.
29. The formulation of any one of claims 1-28, wherein the formulation is a liquid formulation stored at about 3°C to about 5°C, is frozen to -20°C or below, is frozen to -70°C or below, or is a reconstituted solution from a lyophilized formulation.
30. The formulation of any one of claims 1-27, wherein after storage at about 3°C to about 5°C for 6 months:
(i) the % monomer of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 99% as determined by ultra-performance size exclusion chromatography;
(ii) the turbidity of the formulation is at most about 0. 135 as measured by OD350-500;
(iii) the % main peak of the anti-ILT4 antibody or antigen-binding fragment thereof is at least about 63%, the % acidic variant of the anti-ILT4 antibody or antigen-binding fragment thereof is at most about 23%, and/or the basic variant of the anti-ILT4 antibody or antigenbinding fragment thereof is at most about 14%, as determined by high performance ion-exchange chromatography;
(iv) the subvisible particle count of particles that are at least 2 pm in size is at most about 3500 as determined by microflow imaging; and/or
(v) the % oxidation of one or more amino acid residues selected from the group consisting of W7, W102, M253, M359, and M429 in the heavy chain of the anti-ILT4 antibody as set forth in SEQ ID NO:2 or 80 is less than about 4%, as determined by reduced peptide mapping.
31. The formulation of any of claims 1-30, wherein the anti-ILT4 antibody or antigenbinding fragment thereof is a monoclonal antibody.
32. A method of treating a cancer in a human patient in need thereof, the method comprising administering an effective amount of the formulation of any one of claims 1-31.
33. The method of claim 32, wherein the cancer is selected from the group consisting of colorectal cancer, esophageal cancer, melanoma, non-small cell lung cancer, ovarian cancer, renal cell cancer and small cell lung cancer.
34. Use of the formulation of any one of claims 1-31 for preparing a medicament for treating a cancer.
35. Use of the formulation of any one of claims 1-31 for the treatment of cancer in a human patient.
36. The use of claim 34, wherein the cancer is selected from the group consisting of colorectal cancer, esophageal cancer, melanoma, non-small cell lung cancer, ovarian cancer, renal cell cancer and small cell lung cancer.
PCT/US2023/019713 2022-04-29 2023-04-25 Pharmaceutical formulations of an anti-ilt4 antibody or antigen-binding fragment thererof and methods of use WO2023211873A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263336670P 2022-04-29 2022-04-29
US63/336,670 2022-04-29

Publications (1)

Publication Number Publication Date
WO2023211873A2 true WO2023211873A2 (en) 2023-11-02

Family

ID=88519545

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/019713 WO2023211873A2 (en) 2022-04-29 2023-04-25 Pharmaceutical formulations of an anti-ilt4 antibody or antigen-binding fragment thererof and methods of use

Country Status (1)

Country Link
WO (1) WO2023211873A2 (en)

Similar Documents

Publication Publication Date Title
JP7402693B2 (en) Stable formulations of anti-TIGIT antibodies alone and in combination with programmed death receptor 1 (PD-1) antibodies, and methods of use thereof
KR102624564B1 (en) Stable formulations of anti-CTLA4 antibodies alone and in combination with programmed death receptor 1 (PD-1) antibodies and methods of use thereof
US20240052036A1 (en) Formulations of anti-lag3 antibodies and co-formulations of anti-lag3 antibodies and anti-pd-1 antibodies
KR101782203B1 (en) Lyophilised and aqueous anti-cd40 antibody formulations
JP7475335B2 (en) CSF-1R antibody preparation
US20240075133A1 (en) Methods and compositions for reduction of immunogenicity
KR20190021373A (en) Anti-CD19 antibody preparation
KR20210096640A (en) Anti-PD-L1 Antibody Formulations
KR20230017223A (en) Methods of treating IgA nephropathy with APRIL binding antibodies
RU2745814C1 (en) Aqueous pharmaceutical composition of levilimab and the use thereof
WO2023211873A2 (en) Pharmaceutical formulations of an anti-ilt4 antibody or antigen-binding fragment thererof and methods of use
WO2023211868A1 (en) Stable formulations of anti-ilt4 antibodies or antigen-binding fragments thererof in combination with anti-pd-1 antibodies and methods of use thereof
AU2021272212A1 (en) Formulation comprising anti-il-23p19 antibody, method for preparing same and use thereof
WO2023163968A1 (en) Stable formulations of a multivalent vhh based cytotoxic t lymphocyte associated antigen 4 (ctla4) binder binding to human ctla4 and methods of use thereof
EA045933B1 (en) AQUEOUS PHARMACEUTICAL COMPOSITION OF LEVILIMAB AND ITS APPLICATION

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: 23797108

Country of ref document: EP

Kind code of ref document: A2