WO2015118175A2 - TARGETED TGFβ INHIBITION - Google Patents

TARGETED TGFβ INHIBITION Download PDF

Info

Publication number
WO2015118175A2
WO2015118175A2 PCT/EP2015/052781 EP2015052781W WO2015118175A2 WO 2015118175 A2 WO2015118175 A2 WO 2015118175A2 EP 2015052781 W EP2015052781 W EP 2015052781W WO 2015118175 A2 WO2015118175 A2 WO 2015118175A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
tgfp
tumor
group
protein
Prior art date
Application number
PCT/EP2015/052781
Other languages
French (fr)
Other versions
WO2015118175A3 (en
Inventor
Kin-Ming Lo
Original Assignee
Merck Patent Gmbh
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
Priority to SI201531639T priority Critical patent/SI3105246T1/en
Priority to PE2020002093A priority patent/PE20210168A1/en
Application filed by Merck Patent Gmbh filed Critical Merck Patent Gmbh
Priority to AU2015213988A priority patent/AU2015213988B2/en
Priority to LTEP15717097.8T priority patent/LT3105246T/en
Priority to DK15717097.8T priority patent/DK3105246T3/en
Priority to CA2934979A priority patent/CA2934979A1/en
Priority to RS20210783A priority patent/RS62038B1/en
Priority to ES15717097T priority patent/ES2876430T3/en
Priority to PL15717097T priority patent/PL3105246T3/en
Priority to MX2016010067A priority patent/MX2016010067A/en
Priority to SG11201606577YA priority patent/SG11201606577YA/en
Priority to NZ721364A priority patent/NZ721364A/en
Priority to CN201580007865.3A priority patent/CN106103488B/en
Priority to KR1020167024679A priority patent/KR102363008B1/en
Priority to RU2016135062A priority patent/RU2752424C2/en
Priority to JP2016546949A priority patent/JP6731346B2/en
Priority to EP15717097.8A priority patent/EP3105246B1/en
Priority to BR112016014952A priority patent/BR112016014952A2/en
Priority to EP21165709.3A priority patent/EP3889172B1/en
Publication of WO2015118175A2 publication Critical patent/WO2015118175A2/en
Publication of WO2015118175A3 publication Critical patent/WO2015118175A3/en
Priority to IL246968A priority patent/IL246968B/en
Priority to PH12016501549A priority patent/PH12016501549A1/en
Priority to AU2019246876A priority patent/AU2019246876B2/en
Priority to IL281565A priority patent/IL281565B/en
Priority to HRP20210977TT priority patent/HRP20210977T1/en
Priority to CY20211100564T priority patent/CY1124408T1/en
Priority to AU2022200365A priority patent/AU2022200365A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/495Transforming growth factor [TGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/04Drugs for disorders of the respiratory system for throat disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • 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
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies

Abstract

This invention relates generally to bifunctional molecules including (a) a TGFβRII or fragment thereof capable of binding TGFβ and (b) an antibody, or antigen binding fragment thereof, that binds to an immune checkpoint protein, such as Programmed Death Ligand 1 (PD-L1), uses of such molecules (e.g., for treating cancer), and methods of making such molecules.

Description

TARGETED TGFp INHIBITION
FIELD OF THE INVENTION
[0001] This invention relates generally to bifunctional molecules including (a) a TGFpRII or fragment thereof capable of binding TGFP and (b) an antibody, or antigen binding fragment thereof, that binds to an immune checkpoint protein, such as Programmed Death Ligand 1 (PD- Ll), uses of such molecules (e.g., for treating cancer), and methods of making such molecules.
BACKGROUND [0002] In cancer treatment, it has long been recognized that chemotherapy is associated with high toxicity and can lead to emergence of resistant cancer cell variants. Even with targeted therapy against overexpressed or activated oncoproteins important for tumor survival and growth, cancer cells invariably mutate and adapt to reduce dependency on the targeted pathway, such as by utilizing a redundant pathway. Cancer immunotherapy is a new paradigm in cancer treatment that instead of targeting cancer cells, focuses on the activation of the immune system. Its principle is to rearm the host's immune response, especially the adaptive T cell response, to provide immune surveillance to kill the cancer cells, in particular, the minimal residual disease that has escaped other forms of treatment, hence achieving long-lasting protective immunity. [0003] FDA approval of the anti-CTLA-4 antibody ipilimumab for the treatment of melanoma in 2011 ushered in a new era of cancer immunotherapy. The demonstration that anti- PD-1 or anti-PD-Ll therapy induced durable responses in melanoma, kidney, and lung cancer in clinical trials further signify its coming of age (Pardoll, D.M., Nat Immunol. 2012; 13:1 129- 32). However, ipilimumab therapy is limited by its toxicity profile, presumably because anti- CTLA-4 treatment, by interfering with the primary T cell inhibitory checkpoint, can lead to the generation of new autoreactive T cells. While inhibiting the PD-Ll/PD-1 interaction results in dis-inhibiting existing chronic immune responses in exhausted T cells that are mostly antiviral or anticancer in nature (Wherry, E.J., Nat Immunol. 2011; 12:492-9), anti-PD-1 therapy can nevertheless sometimes result in potentially fatal lung-related autoimmune adverse events. Despite the promising clinical activities of anti-PDl and anti-PD-Ll so far, increasing the therapeutic index, either by increasing therapeutic activity or decreasing toxicity, or both, remains a central goal in the development of immunotherapeutics. SUMMARY OF THE INVENTION
[0004] The present invention is based on the discovery that a bifunctional protein containing at least portion of TGFp Receptor II (TGFpRII) that is capable of binding TGFp and antibody or antigen-binding fragment that binds to an immune checkpoint protein such as human protein Programmed Death Ligand 1 (PD-L1) can be an effective anti-tumor and anti-cancer therapeutic. The protein can exhibit a synergistic effect in cancer treatment, as compared to the effect of administering the two agents separately.
[0005] Accordingly, in a first aspect, the present invention features a protein including (a) human TGFpRII, or a fragment thereof capable of binding TGF (e.g., a soluble fragment); and (b) an antibody, or an antigen-binding fragment thereof, that binds PD-L1 (e.g., any of the antibodies or antibody fragments described herein).
[0006] In a related aspect, the invention features a polypeptide including (a) at least a variable domain of a heavy chain of an antibody that binds PD-L1 (e.g., amino acids 1-120 of SEQ ID NO: 2); and (b) human TGFPRII, or a soluble fragment thereof capable of binding TGFp (e.g., a human TGFpRII extra-cellular domain (ECD), amino acids 24-159 of SEQ ID NO: 9, or any of those described herein). The polypeptide may further include an amino acid linker connecting the C-terminus of the variable domain to the N-terminus of the human TGF RII or soluble fragment thereof capable of binding TGFp. The polypeptide may include the amino acid sequence of SEQ ID NO: 3 or an amino acid sequence substantially identical to SEQ ID NO: 3. The antibody fragment may be an scFv, Fab, F(ab')2, or Fv fragment. [0007] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 2 and human TGFpRII. The antibody may optionally include a modified constant region (e.g., any described herein, including a C- terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0008] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 2 and a fragment of human TGFpRII capable of binding TGFp (e.g., a soluble fragment). The antibody may optionally include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0009] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 2 and a human TGF RII ECD. The antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0010] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes amino acids 1-120 of SEQ ID NO: 2 and human
TGF RII. The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain). [0011] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes amino acids 1-120 of SEQ ID NO: 2 and a fragment of human TGF RII capable of binding TGFP (e.g., a soluble fragment). The antibody may include a modified constant region (e.g., any described herein, including a C-terminal
Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala- Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0012] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes amino acids 1-120 of SEQ ID NO: 2 and a human TGF RII ECD. The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0013] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 2 and human TGFpRII. The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0014] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 2 and a fragment of human TGFpRII capable of binding TGFp (e.g., a soluble fragment). The antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain). [0015] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 2 and a human TGF RII ECD. The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu- Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0016] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 12 and human TGFpRII. The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala- Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0017] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 12 and a fragment of human TGFpRII capable of binding TGFp (e.g., a soluble fragment). The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0018] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 12 and a human TGFpRII ECD. The antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0019] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 12 and human TGFpRII. The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0020] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 12 and a fragment of human TGFpRII capable of binding TGF (e.g., a soluble fragment). The antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0021] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes the hypervariable regions present in SEQ ID NO: 12 and a human TGFpRII ECD. The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu- Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0022] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 14 and human TGFpRII. The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala- Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0023] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 14 and a fragment of human TGFpRII capable of binding TGFp (e.g., a soluble fragment). The antibody may include a modified constant region (e.g., any described herein, including a C-terminal Lys-→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0024] In certain embodiments, the protein or polypeptide includes an antibody or antigen- binding fragment thereof that includes SEQ ID NO: 14 and a human TGFpRII ECD. The antibody may include a modified constant region (e.g., any described herein, including a C- terminal Lys→Ala substitution, a mutation of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) sequence to Ala-Thr-Ala-Thr (SEQ ID NO: 20), or a hybrid constant region including an IgGl hinge region and an IgG2 CH2 domain).
[0025] The invention also features a nucleic acid that includes a nucleotide sequence that encodes a polypeptide described above. In certain embodiments, the nucleic acid further includes a second nucleotide sequence encoding at least a variable domain of a light chain of an antibody which, when combined with the polypeptide, forms an antigen-binding site that binds PD-Ll (e.g., including amino acids 1-110 of SEQ ID NO: 1). The second nucleotide sequence may encode the amino acid sequence of SEQ ID NO: 1 (secreted anti-PD-Ll lambda light chain) or an amino acid sequence substantially identical to SEQ ID NO: 1. The invention also features a cell including any of the nucleic acids described above.
[0026] The invention also features a method of producing a protein including (a) the extracellular domain of the human TGFPRII, or a fragment thereof capable of binding TGFP (e.g., a soluble fragment), and (b) an antibody, or an antigen-binding fragment thereof, that binds human PD-Ll . The method includes maintaining a cell described under conditions that permit expression of the protein. The method may further include harvesting the protein.
[0027] The invention also features a protein including the polypeptide described above and at least a variable domain of a light chain of an antibody which, when combined with the polypeptide, forms an antigen-binding site that binds PD-Ll. The protein may include (a) two polypeptides, each having an amino acid sequence consisting of the amino acid sequence of SEQ ID NO: 3, and (b) two additional polypeptides each having an amino acid sequence consisting of the amino acid sequence of SEQ ID NO: 1.
[0028] The invention also features a protein described above for use in therapy. The therapy may include administration of radiation or administration of a chemotherapeutic, a biologic, or a vaccine. [0029] The invention also features a protein described above for use in promoting local depletion of TGFp at a tumor.
[0030] The invention also features a protein described above for use in inhibiting SMAD3 phosphorylation in a cell (e.g., a tumor cell or an immune cell). [0031] The invention also features a protein described above for use in treating cancer or for use in inhibiting tumor growth. The cancer or tumor may be selected from the group consisting of colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, myeloma, lymphoma, leukemia, thyroid, endometrial, uterine, bladder, neuroendocrine, head and neck, liver, nasopharyngeal, testicular, small cell lung cancer, non-small cell lung cancer, melanoma, basal cell skin cancer, squamous cell skin cancer, dermatofibrosarcoma protuberans, Merkel cell carcinoma, glioblastoma, glioma, sarcoma, mesothelioma, and myelodisplastic syndromes. The use may further include administration of radiation or administration of a chemotherapeutic, a biologic, or a vaccine.
[0032] The invention also features a method of promoting local depletion of TGFp. The method includes administering a protein described above, where the protein binds TGFp in solution, binds PD-Ll on a cell surface, and carries the bound TGFp into the cell (e.g., a cancer cell).
[0033] The invention also features a method of inhibiting SMAD3 phosphorylation in a cell (e.g., a cancer cell or an immune cell), the method including exposing the cell in the tumor microenvironment to a protein described above.
[0034] The invention also features a method of inhibiting tumor growth or treating cancer. The method includes exposing the tumor to a protein described above. The method may further include exposing the tumor to radiation or to a chemotherapeutic, a biologic, or a vaccine. In certain embodiments, the tumor or cancer is selected from the group consisting of colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, myeloma, lymphoma, leukemia, thyroid, endometrial, uterine, bladder, neuroendocrine, head and neck, liver, nasopharyngeal, testicular, small cell lung cancer, non-small cell lung cancer, melanoma, basal cell skin cancer, squamous cell skin cancer, dermatofibrosarcoma protuberans, Merkel cell carcinoma, glioblastoma, glioma, sarcoma, mesothelioma, and myelodisplastic syndromes. [0035] By "TGFpRII" or "TGF Receptor Π" is meant a polypeptide having the wild-type human TGFp Receptor Type 2 Isoform A sequence (e.g., the amino acid sequence of NCBI Reference Sequence (RefSeq) Accession No. NP_001020018 (SEQ ID NO: 8)), or a polypeptide having the wild-type human TGFP Receptor Type 2 Isoform B sequence (e.g., the amino acid sequence of NCBI RefSeq Accession No. NP 003233 (SEQ ID NO: 9)) or having a sequence substantially identical the amino acid sequence of SEQ ID NO: 8 or of SEQ ID NO: 9. The TGF RII may retain at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99% of the TGFP-binding activity of the wild-type sequence. The polypeptide of expressed TGFpRII lacks the signal sequence.
[0036] By a "fragment of TGFpRII capable of binding TGF " is meant any portion of NCBI RefSeq Accession No. P_001020018 (SEQ ID NO: 8) or of NCBI RefSeq Accession No. NP_003233 (SEQ ID NO: 9), or a sequence substantially identical to SEQ ID NO: 8 or SEQ ID NO: 9 that is at least 20 (e.g., at least 30, 40, 50, 60, 70, 80, 90, 100, 1 10, 120, 130, 140, 150, 160, 175, or 200) amino acids in length that retains at least some of the TGFp-binding activity (e.g., at least 0.1%, 0.5%, 1%, 5%, 10%, 25%, 35%, 50%, 75%, 90%, 95%, or 99%) of the wild-type receptor or of the corresponding wild-type fragment. Typically such fragment is a soluble fragment. An exemplary such fragment is a TGFpRII extra-cellular domain having the sequence of SEQ ID NO: 10.
[0037] By "substantially identical" is meant a polypeptide exhibiting at least 50%, desirably 60%, 70%, 75%, or 80%, more desirably 85%, 90%, or 95%, and most desirably 99% amino acid sequence identity to a reference amino acid sequence. The length of comparison sequences will generally be at least 10 amino acids, desirably at least 15 contiguous amino acids, more desirably at least 20, 25, 50, 75, 90, 100, 150, 200, 250, 300, or 350 contiguous amino acids, and most desirably the full-length amino acid sequence.
[0038] By "patient" is meant either a human or non-human animal (e.g., a mammal).
[0039] By "treating" a disease, disorder, or condition (e.g., a cancer) in a patient is meant reducing at least one symptom of the disease, disorder, or condition by administrating a therapeutic agent to the patient.
[0040] By "cancer" is meant a collection of cells multiplying in an abnormal manner.
[0041] Other embodiments and details of the invention are presented herein below. BRIEF DESCRIPTION OF THE DRAWINGS
[0042] FIG. 1A is a schematic drawing of an anti-PD-Ll /TGFp Trap molecule comprising one anti-PD-Ll antibody fused to two extracellular domain (ECD) of TGFP Receptor II via a (Gly4Ser)4Gly linker. FIG. IB is a photograph of a sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of anti-PD-Ll/TGFp Trap under non-reducing and reducing conditions.
[0043] FIG. 2 is photograph of an SDS-PAGE gel showing analysis of extent of clipping of anti-PD-Ll/TGF Trap expressed by clone 02B15 at various population doubling levels. Anti- PD-Ll /TGFp Trap from clone 02B15 after a single protein A chromatography step was analyzed by SDS-PAGE under reducing conditions. Lanes 1 and 10, See Blue Plus 2 MW Standard; lane 2, purified anti-PD-Ll /TGFp Trap reference; lane 3, clone 02B15 at PDLO; lane 4, clone 02B15 at PDL30; lane 5, clone 02B 15 at PDL60; and lane 6, clone 02B15 at PDL90. (PDL, population doubling level).
[0044] FIG. 3 is a graph showing FACS analysis of anti-PD-Ll /TGFp Trap binding to HEK cells transfected to express human PD-L1. [0045] FIG. 4 is a graph showing the ability of anti-PD-L 1/TGFP Trap to inhibit TGFp- induced phosphorylation of SMAD3 using a pSMAD3-luciferase reporter cell line (filled circle: anti-PD-Ll ; X: anti-PD-Ll (mut); filled square: anti-PD-Ll /TGFp Trap; filled triangle: anti-PD-Ll(mut)/TGFp Trap; +: anti-TGFp antibody 1D1 1 ; star: TGFp RII-Fc).
[0046] FIGS. 5A and 5B are graphs showing pharmacokinetics of intravenously administered anti-PD-Ll /TGFp Trap and related proteins in mice.
[0047] FIG. 6 A is a graph showing PD-L1 target-mediated endocytosis of anti-PD-Ll/ TGF Trap. FIG. 6B is a graph showing PD-L1 target-mediated endocytosis of anti-PD-Ll . FIG. 6C is a graph showing percent internalization of anti-PD-Ll/ TGFp Trap and anti-PD-Ll bound on HEK/PD-L 1 cells. [0048] FIGS. 7A-7C are graphs showing anti-tumor efficacy of anti-PD-Ll /TGFp Trap and related proteins in the EMT-6 breast carcinoma subcutaneous model (Example 7). FIG. 7A shows tumor growth curves of average tumor volumes of surviving mice in different treatment groups (star: Group 1 : filled circle: Group 2; filled triangle: Group 3; filled square: Group 4; open square: Group 5; filled square/dashed line: Group 6; filled square/stippled line: Group 7). FIG. 7B shows tumor growth curves of individual tumor volumes in different treatment groups. FIG. 7C is a Kaplan-Meier plot of percent survival in different treatment groups (symbols as in 7A).
[0049] FIG. 8 is a graph showing anti-tumor efficacy of anti-PD-Ll /TGFp Trap and related proteins in the MC38 colorectal carcinoma subcutaneous tumor model (Example 8; star: Group 1; filled circle: Group 2; filled circle/dashed line: Group 3; filled triangle: Group 4; filled triangle/dashed line: Group 5; filled square: Group 6; filled square/dashed line: Group 7).
[0050] FIG. 9 is a graph showing anti-tumor efficacy of anti-PDLl/TGFp Trap and related proteins in an orthotopic EMT-6 breast cancer model (Example 9; star: Group 1; filled circle/dashed line: Group 2; filled triangle: Group 3; filled triangle/dashed line: Group 4; filled diamond: Group 5).
[0051] FIG. 10 is a graph showing anti-tumor efficacy of anti-PDLl/TGFp Trap and related proteins in an intramuscular MC38 colorectal carcinoma model (Example 10; star: Group 1; filled circle: Group 2; filled circle/dashed line: Group 3: filled diamond/dashed line: Group 4; filled square: Group 5; filled square/dashed line: Group 6; filled diamond: Group 7).
[0052] Fig. 11 is a graph showing anti-tumor efficacy of anti-PD-Ll/TGF-β Trap and the combination of anti-PD-Ll and TGF Trap control administered to give equivalent in vivo exposure in an orthotopic EMT-6 breast tumor model (Example 11 ; star: Group 1 ; filled square: Group 2; open square: Group 3; filled diamond: Group 4; open diamond: Group 5). [0053] FIGS. 12A-12C are graphs showing anti-tumor efficacy of anti-PD-Ll /TGF-p Trap and the combination of anti-PD-Ll and TGF Trap control administered to give equivalent in vivo exposure in an intramuscular MC38 colorectal carcinoma model (Example 12). FIG. 12 A shows tumor growth curves of mice treated with both intermediate and low doses of the proteins (star: Group 1 ; filled squares: Group 2; open squares: Group 3; filled diamonds: Group 4; open diamonds Group 5). FIG. 12B (star: Group I; filled square: Group 2; filled diamond: Group 4; *: p < 0.0001 compared to Group 1 ; **: p < 0.0001 compared to Group 2) and 12C (star: Group 1 ; filled square: Group3; filled diamond: Group 5; *: p < 0.0001 compared to Group 1 ; **: p < 0.0001 compared to Group 3) show statistical analysis of tumor growth curves of mice treated with intermediate and low doses of the proteins, respectively
[0054] FIGS. 13 A-13B are graphs showing anti-tumor efficacy of anti-PD-L 1 (YW)/TGF-p Trap and related proteins in an orthotopic EMT-6 breast tumor model (Example 13 ; star: Group 1; filled circle: Group 2; filled triangle: Group 3; filled square: Group 4; filled diamond: Group 5). FIG. 13A shows tumor growth curves of mice in different treatment groups. FIG. 13B is a Kaplan-Meier plot of percent survival in different treatment groups.
[0055] FIGS. 14A-14B are graphs showing anti-tumor efficacy of anti-PD-Ll(YW)/TGF-p Trap and related proteins based on (A) tumor volumes and (B) tumor weights, in an intramuscular MC38 colorectal carcinoma model (Example 14; star: Group 1 ; filled circle: Group 2; filled triangle: Group 3; filled square: Group 4; filled diamond: Group 5).
[0056] FIG. 15 is a graph comparing the anti-tumor efficacy of an anti-PD-1 antibody treatment with and without TGFp Trap control in an orthotopic EMT-6 breast tumor model (Example 15; star: Group 1 ; filled square: Group 2; filled inverted triangle: Group 3; open inverted triangle: Group 4).
[0057] FIG. 16 is a graph comparing the anti -tumor efficacy of an anti-PD-1 antibody treatment with and without TGFp Trap control in an intramuscular MC38 colorectal tumor model (Example 16; star: Group 1 ; filled square: Group 2; filled inverted triangle: Group 3; open inverted triangle: Group 4).
[0058] FIG. 17 is a graph comparing the anti-tumor efficacy of an anti-LAG3 or anti-T!M3 antibody treatment with and without TGFP Trap control in an orthotopic EMT-6 breast tumor model (Example 17; star: Group 1; filled square: Group 2; filled triangle: Group 3; filled inverted triangle: Group 4; open triangle: Group 5; open inverted triangle: Group 6). [0059] FIG. 18 is a graph comparing the anti-tumor efficacy of an anti-LAG3 or anti-TIM3 antibody treatment with and without TGFp Trap control in an intramuscular MC38 colorectal tumor model (Example 18; star: Group 1; filled square: Group 2; filled triangle: Group 3; filled inverted triangle: Group 4; open triangle: Group 5; open inverted triangle: Group 6).
DETAILED DESCRIPTION
[0060] The current invention permits localized reduction in TGF in a tumor
microenvironment by capturing the TGFp using a soluble cytokine receptor (TGFpRII) tethered to an antibody moiety targeting a cellular immune checkpoint receptor found on the exterior surface of certain tumor cells or immune cells. An example of an antibody moiety of the invention to an immune checkpoint protein is anti-PD-Ll. This bifunctional molecule, sometimes referred to in this document as an "antibody-cytokine trap," is effective precisely because the anti -receptor antibody and cytokine trap are physically linked. The resulting advantage (over, for example, administration of the antibody and the receptor as separate molecules) is partly because cytokines function predominantly in the local environment through autocrine and paracrine functions. The antibody moiety directs the cytokine trap to the tumor microenvironment where it can be most effective, by neutralizing the local
immunosuppressive autocrine or paracrine effects. Furthermore, in cases where the target of the antibody is internalized upon antibody binding, an effective mechanism for clearance of the cytokine/cytokine receptor complex is provided. Antibody-mediated target internalization has been shown for PD-L1. This is a distinct advantage over using an anti-TGFp antibody because first, an anti-TGFp antibody might not be completely neutralizing; and second, the antibody can act as a carrier extending the half-life of the cytokine, and antibody/cytokine complexes often act as a circulating sink that builds up and ultimately dissociates to release the cytokine back in circulation (Montero-Julian et al, Blood. 1995; 85:917-24). The use of a cytokine trap to neutralize the ligand can also be a better strategy than blockading the receptor with an antibody, as in the case of CSF-1. Because CSF-1 is cleared from the circulation by receptor- mediated endocytosis, an anti-CSF-1 receptor antibody blockade caused a significant increase in circulating CSF-1 concentration (Hume et al, Blood. 2012;119: 1810-20)
[0061] Indeed, as described below, treatment with the anti-PD-Ll /TGFp Trap elicits a synergistic anti-tumor effect due to the simultaneous blockade of the interaction between PD- Ll on tumor cells and PD-1 on immune cells, and the neutralization of TGFP in the tumor microenvironment. As demonstrated in the following examples, anti-PDLl/TGFp Trap has efficacy superior to that of the single agent anti-PD-Ll or TGFP Trap control. Without being bound by theory, this presumably is due to a synergistic effect obtained from simultaneous blocking the two major immune escape mechanisms, and in addition, the targeted depletion of the TGFp in the tumor microenvironment by a single molecular entity. This depletion is achieved by (1) anti-PD-Ll targeting of tumor cells; (2) binding of the TGFp
autocrine/paracrine in the tumor microenvironment by the TGFP Trap; and (3) destruction of the bound TGFp through the PD-L1 receptor-mediated endocytosis. The aforementioned mechanisms of action cannot be achieved by the combination therapy of the two single agents anti-PD-Ll and TGFp Trap. Furthermore, the TGFpPJI fused to the C-terminus of Fc
(fragment of crystallization of IgG) was several-fold more potent than the TGFpRII-Fc that places the TGFpRII at the N-terminus of Fc (see Example 3). The superb efficacy obtained with anti-PDLl/TGFp Trap also allays some concerns that the TGFpRII does not trap TGFP2. As pointed out by Yang et al. , Trends Immunol. 2010; 31 :220-227, although some tumor types do secrete TGFP2 initially, as the tumor progresses, the TGFp in the tumor microenvironment is predominantly secreted by myeloid-derived suppressor cells, which secrete TGFpi . In addition to showing great promise as an effective immuno-oncology therapeutic, treatment with soluble TGFPRII can potentially reduce the cardiotoxicity concerns of TGFP targeting therapies, especially the TGFpRI kinase inhibitors. This is because of the important roles TGFP2 plays in embryonic development of the heart as well as in repair of myocardial damage after ischemia and reperfusion injury (Roberts et ah, J Clin Invest. 1992; 90:2056-62).
TGFB as a cancer target
[0062] TGFP had been a somewhat questionable target in cancer immunotherapy because of its paradoxical roles as the molecular Jekyll and Hyde of cancer (Bierie et ah, Nat Rev Cancer. 2006; 6:506-20). Like some other cytokines, TGFp activity is developmental stage and context dependent. Indeed TGFp can act as either a tumor promoter or a tumor suppressor, affecting tumor initiation, progression and metastasis. The mechanisms underlying this dual role of TGFp remain unclear (Yang et ah, Trends Immunol. 2010; 31 :220-227). Although it has been postulated that Smad-dependent signaling mediates the growth inhibition of TGFp signaling, while the Smad independent pathways contribute to its tumor-promoting effect, there are also data showing that the Smad-dependent pathways are involved in tumor progression (Yang et ah, Cancer Res. 2008; 68:9107-11).
[0063] Both the TGFp ligand and the receptor have been studied intensively as therapeutic targets. There are three ligand iso forms, TGFpi, 2 and 3, all of which exist as homodimers. There are also three TGFP receptors (TGFpR), which are called TGFpR type I, II and III (Lopez-Casillas et ah, J Cell Biol. 1994; 124:557-68). TGFpRI is the signaling chain and cannot bind ligand. TGFPRII binds the ligand TGFpi and 3, but not TGFp2, with high affinity. The TGFpRII/TGFp complex recruits TGFpRI to form the signaling complex (Won et ah, Cancer Res. 1999; 59:1273-7). TGFpRIII is a positive regulator of TGFp binding to its signaling receptors and binds all 3 TGF isoforms with high affinity. On the cell surface, the TGFp/TGFpRJII complex binds TGFpRII and then recruits TGFpRI, which displaces
TGFpRIII to form the signaling complex.
[0064J Although the three different TGFp isoforms all signal through the same receptor, they are known to have differential expression patterns and non-overlapping functions in vivo. The three different TGF-p isoform knockout mice have distinct phenotypes, indicating numerous non-compensated functions (Bujak et ah, Cardiovasc Res. 2007; 74: 184-95). While TGF i null mice have hematopoiesis and vasculogenesis defects and TGFp3 null mice display pulmonary development and defective palatogenesis, TGFp2 null mice show various developmental abnormalities, the most prominent being multiple cardiac deformities (Bartram et al, Circulation. 2001; 103:2745-52; Yamagishi et al, Anat Rec. 2012; 295:257-67).
Furthermore, TGFP is implicated to play a major role in the repair of myocardial damage after ischemia and reperfusion injury. In an adult heart, cardiomyocytes secrete TGFp, which acts as an autocrine to maintain the spontaneous beating rate. Importantly, 70-85% of the TGFP secreted by cardiomyocytes is TGFP2 (Roberts et al, J Clin Invest. 1992; 90:2056-62). In summary, given the predominant roles of TGFpi and TGFP2 in the tumor microenvironment and cardiac physiology, respectively, a therapeutic agent that neutralizes TGFpi but not TGFP2 could provide an optimal therapeutic index by minimizing the cardiotoxicity without compromising the anti-tumor activity. This is consistent with the findings by the present inventors, who observed a lack of toxicity, including cardiotoxicity, for anti-PD-Ll/TGFp Trap in monkeys.
[0065] Therapeutic approaches to neutralize TGFP include using the extracellular domains of TGFp receptors as soluble receptor traps and neutralizing antibodies. Of the receptor trap approach, soluble TGFpRIII may seem the obvious choice since it binds all the three TGFp ligands. However, TGFpRIII, which occurs naturally as a 280-330 kD glucosaminoglycan (GAG)-glycoprotein, with extracellular domain of 762 amino acid residues, is a very complex protein for biotherapeutic development. The soluble TGFpRIII devoid of GAG could be produced in insect cells and shown to be a potent TGFp neutralizing agent (Vilchis-Landeros et al, Biochem J 355:215, 2001). The two separate binding domains (the endoglin-related and the uromodulin-related) of TGFpRIII could be independently expressed, but they were shown to have affinities 20 to 100 times lower than that of the soluble TGFPRIII, and much diminished neutralizing activity (Mendoza et al, Biochemistry. 2009; 48: 11755-65). On the other hand, the extracellular domain of TGFpRII is only 136 amino acid residues in length and can be produced as a glycosylated protein of 25-35 kD. The recombinant soluble TGFPRII was further shown to bind TGFpi with a D of 200 pM, which is fairly similar to the KD of 50 pM for the full length TGFpRII on cells (Lin et al, J Biol Chem. 1995; 270:2747-54). Soluble TGFpRII- Fc was tested as an anti-cancer agent and was shown to inhibit established murine malignant mesothelioma growth in a tumor model (Suzuki et al, Clin Cancer Res. 2004; 10:5907-18). Since TGFpRII does not bind TGFp2, and TGFpRIII binds TGFpi and 3 with lower affinity than TGFpRII, a fusion protein of the endoglin domain of TGFpRIII and extracellular domain of TGFpRII was produced in bacteria and was shown to inhibit the signaling of TGFpi and 2 in cell based assays more effectively than either TGFpRII or RIII (Verona et al, Protein Eng Des Sel. 2008; 21 :463-73). Despite some encouraging anti-tumor activities in tumor models, to our knowledge no TGFp receptor trap recombinant proteins have been tested in the clinic.
[0066] Still another approach to neutralize all three isoforms of the TGFp ligands is to screen for a pan-neutralizing anti-TGFp antibody, or an anti-receptor antibody that blocks the receptor from binding to TGFpi, 2 and 3. GC1008, a human antibody specific for all isoforms of TGFp, was in a Phase I/II study in patients with advanced malignant melanoma or renal cell carcinoma (Morris et al, J Clin Oncol 2008; 26:9028 (Meeting abstract)). Although the treatment was found to be safe and well tolerated, only limited clinical efficacy was observed, and hence it was difficult to interpret the importance of anti-TGFp therapy without further characterization of the immunological effects (Flavell et al, Nat Rev Immunol. 2010; 10:554- 67). There were also TGFP-isoform-specific antibodies tested in the clinic. Metelimumab, an antibody specific for TGFpi was tested in Phase 2 clinical trial as a treatment to prevent excessive post-operative scarring for glaucoma surgery; and Lerdelimumab, an antibody specific for TGFP2, was found to be safe but ineffective at improving scarring after eye surgery in a Phase 3 study (Khaw et al , Ophthalmology 2007; 114: 1822-1830). Anti-TGFpRII antibodies that block the receptor from binding to all three TGFP isoforms, such as the anti- human TGFPRII antibody TRl and anti-mouse TGFpRII antibody MTl, have also shown some therapeutic efficacy against primary tumor growth and metastasis in mouse models (Zhong et al, Clin Cancer Res. 2010; 16: 1191-205). To date, the vast majority of the studies on TGFp targeted anticancer treatment, including small molecule inhibitors of TGFp signaling that often are quite toxic, are mostly in the preclinical stage and the anti-tumor efficacy obtained has been limited (Calone et al, Exp Oncol. 2012; 34:9-16; Connolly et al, Int J Biol Sci. 2012; 8:964- 78).
[0067] The antibody-TGFp trap of the invention is a bifunctional protein containing at least portion of a human TGFP Receptor II (TGFpRII) that is capable of binding TGFp. In one embodiment, the TGF trap polypeptide is a soluble portion of the human TGF Receptor Type 2 Isoform A (SEQ ID NO: 8) that is capable of binding TGFp. In a further embodiment, TGFp trap polypeptide contains at least amino acids 73-184 of SEQ ID NO:8. In yet a further embodiment, the TGFp trap polypeptide contains amino acids 24-184 of SEQ ID NO:8. In another embodiment, the TGFp trap polypeptide is a soluble portion of the human TGFp Receptor Type 2 Isoform B (SEQ ID NO: 9) that is capable of binding TGFp. In a further embodiment, TGFp trap polypeptide contains at least amino acids 48-159 of SEQ ID NO:9. In yet a further embodiment, the TGFP trap polypeptide contains amino acids 24-159 of SEQ ID NO:9. In yet a further embodiment, the TGFp trap polypeptide contains amino acids 24-105 of SEQ ID NO:9.
Immune Checkpoint Pis-inhibition [0068] The approach of targeting T cell inhibition checkpoints for dis-inhibition with therapeutic antibodies is an area of intense investigation (for a review, see Pardoll, Nat Rev Cancer. 2012; 12:253-264). In one approach, the antibody moiety or antigen binding fragment thereof targets T cell inhibition checkpoint receptor proteins on the T cell, such as, for example: CTLA-4, PD-1, BTLA, LAG-3, TIM-3, and LAIR1. In another approach, the antibody moiety targets the counter-receptors on antigen presenting cells and tumor cells (which co-opt some of these counter-receptors for their own immune evasion), such as, for example: PD-Ll (B7-H1), B7-DC, HVEM, TIM-4, B7-H3, or B7-H4.
[0069] The invention contemplates antibody TGFp traps that target, through their antibody moiety or antigen binding fragment thereof, T cell inhibition checkpoints for dis-inhibition. To that end the present inventors have tested the anti-tumor efficacy of combining a TGFp trap with antibodies targeting various T cell inhibition checkpoint receptor proteins, such as anti- PD-1, anti-PD-Ll, anti -TIM-3 and anti-LAG3. The experimental results are further detailed in Examples 7-18. The present inventors found that combining a TGFP trap with an anti-PD-Ll antibody exhibited remarkable anti-tumor activity beyond what was observed with the monotherapies. In contrast, none of the other combinations with antibodies to the targets listed above showed any superior efficacy. In particular, one may have expected that a combination treatment of a TGFp trap with an anti-PD-1 antibody would demonstrate similar activity to the one observed with anti-PD-Ll, as PD-1 / PD-Ll are cognate receptors that bind to each other to effect the immune checkpoint inhibition. However, this is not what the present inventors have found.
Anti-PD-Ll Antibodies
[0070] The invention can include any anti-PD-Ll antibody, or antigen-binding fragment thereof, described in the art. Anti-PD-Ll antibodies are commercially available, for example, the 29E2A3 antibody (Biolegend, Cat. No. 329701). Antibodies can be monoclonal, chimeric, humanized, or human. Antibody fragments include Fab, F(ab')2, scFv and Fv fragments, which are described in further detail below. [0071 ] Exemplary antibodies are described in PCT Publication WO 2013/079174. These antibodies can include a heavy chain variable region polypeptide including an HVR-Hl , HVR- Hl, and HVR-H3 sequence, where:
(a) the HVR-Hl sequence is X1YX2MX3; (b) the HVR-H2 sequence is SIYPSGGX4TFYADX5VKG;
(c) the HVR-H3 sequence is IKLGTVTTVX6Y; further where: Xi is , R, T, Q, G, A, W, M, I, or S; X2 is V, R, K, L, M, or I; X3 is H, T, N, Q, A, V, Y, W, F, or M; X4 is F or I; X5 is S or T; X6 is E or D.
[0072] In a one embodiment, Xi is M, I, or S; X2 is R, , L, M, or I; X3 is F or M; X4 is F or I; X5 is S or T; X6 is E or D.
[0073] In another embodiment Xi is M, I, or S; X2 is L, M, or I; X3 is F or M; X4 is I; X5 is S or T; X6 is D.
[0074] In still another embodiment, Xi is S; X2 is I; X3 is M; X4 is I; X5 is T; X6 is D.
[0075] In another aspect, the polypeptide further includes variable region heavy chain framework sequences juxtaposed between the HVRs according to the formula: (HC-FRl )- (HVR-Hl )-(HC-FR2)-(HVR-H2)-(HC-FR3)-(HVR-H3)-(HC-FR4).
[0076] In yet another aspect, the framework sequences are derived from human consensus framework sequences or human germline framework sequences.
[0077] In a still further aspect, at least one of the framework sequences is the following: HC-FRl is EVQLLESGGGLVQPGGSLRLSCAASGFTFS;
HC-FR2 is W VRQ APGKGLE WVS ;
HC-FR3 is RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR;
HC-FR4 is WGQGTLVTVSS.
[0078] In a still further aspect, the heavy chain polypeptide is further combined with a variable region light chain including an HVR-L1 , HVR-L2, and HVR-L3, where: (a) the HVR-L1 sequence is TGTX7X8DVGX9YNYVS;
(b) the HVR-L2 sequence is X10VX11X12RPS;
(c) the HVR-L3 sequence is SSX13TX14X15X16X17RV; further where: X7 is N or S; Xs is T, R, or S; X9 is A or G; Xio is E or D; Xn is I, N or S; X12 is D, H or N; X13 is F or Y; Xn is N or S; X15 is R, T or S; Xi6 is G or S; X17 is I or T.
[0079] In another embodiment, X7 is N or S; Xs is T, R, or S; X9 is A or G; Xio is E or D; X11 is N or S; X12 is N; X13 is F or Y; XH is S; Xis is S; Xi6 is G or S; X17 is T.
[0080] In still another embodiment, X7 is S; Xs is S; X9 is G; Xio is D; X11 is S; Xi2 is N; Xi3 is Y; XH is S; Xis is S; Xi6 is S; X17 is T. [0081] In a still further aspect, the light chain further includes variable region light chain framework sequences juxtaposed between the HVRs according to the formula: (LC- FR1MHVR-L1)-(LC-FR2)-(HVR-L2)-(LC-FR3)-(HVR-L3)-(LC-FR4).
[0082] In a still further aspect, the light chain framework sequences are derived from human consensus framework sequences or human germline framework sequences. [0083] In a still further aspect, the light chain framework sequences are lambda light chain sequences.
[0084] In a still further aspect, at least one of the framework sequence is the following:
LC-FR1 is QSALTQPASVSGSPGQSITISC;
LC-FR2 is WYQQHPGKAP LMIY; LC-FR3 is GVSNRFSGSKSGNTASLTISGLQAEDEADYYC;
LC-FR4 is FGTGTKVTVL.
[0085] In another embodiment, the invention provides an anti-PD-Ll antibody or antigen binding fragment including a heavy chain and a light chain variable region sequence, where:
(a) the heavy chain includes an HVR-Hl, HVR-H2, and HVR-H3, wherein further: (i) the HVR-Hl sequence is X1YX2MX3; (ii) the HVR-H2 sequence is
SIYPSGGX4TFYADXsVKG; (iii) the HVR-H3 sequence is IKLGTVTTVX6Y, and; (b) the light chain includes an HVR-L1, HVR-L2, and HVR-L3, wherein further: (iv) the HVR-L1 sequence is TGTX7X8DVGX9YNYVS; (v) the HVR-L2 sequence is
X10VX11X12RPS; (vi) the HVR-L3 sequence is SSXi3TXi4Xi5Xi6XnRV; wherein: Xi is K, R, T, Q, G, A, W, M, I, or S; X2 is V, R, K, L, M, or I; X3 is H, T, N, Q, A, V, Y, W, F, or M; X4 is F or I; X5 is S or T; X6 is E or D; X7 is N or S; Xs is T, R, or S; X9 is A or G; Xio is E or D; X11 is I, N, or S; X12 is D, H, or N; Xi3 is F or Y; X14 is N or S; X15 is R, T, or S; Xi6 is G or S; Xi7 is I or T.
[0086] In one embodiment, Xi is M, I, or S; X2 is R, K, L, M, or I; X is F or M; X4 is F or I; X is S or T; X6 is E or D; X7 is N or S; Xs is T, R, or S; X9 is A or G; Xio is E or D; Xnis N or S; X12 is N; X13 is F or Y; Xu is S; X15 is S; Xie is G or S; X17 is T.
[0087] In another embodiment, Xi is M, I, or S; X2 is L, M, or I; X3 is F or M; X4 is I; Xs is S or T; X6 is D; X7 is N or S; Xs is T, R, or S; X9 is A or G; Xio is E or D; Xi 1 is N or S; X12 is N; Xn is F or Y; Xi4 is S; Xis is S; Xi6 is G or S; Xn is T.
[0088] In still another embodiment, Xi is S; X2 is I; X3 is M; X4 is I; X5 is T; X6 is D; X7 is S; Xs is S; X9 is G; Xio is D; X11 is S; X12 is N; X13 is Y; Xi4 is S; Xis is S; Xie is S; X17 is T.
[0089] In a further aspect, the heavy chain variable region includes one or more framework sequences juxtaposed between the HVRs as: (HC-FR1)-(HVR-H1)-(HC-FR2)-(HVR-H2)-(HC- FR3)-(HVR-H3)-(HC-FR4), and the light chain variable regions include one or more framework sequences juxtaposed between the HVRs as: (LC-FR1 MHVR-Ll )-(LC-FR2)- (HVR-L2)-(LC-FR3)-(HVR-L3)-(LC-FR4).
[0090] In a still further aspect, the framework sequences are derived from human consensus framework sequences or human germline sequences.
[0091] In a still further aspect, one or more of the heavy chain framework sequences is the following: HC-FR1 is EVQLLESGGGLVQPGGSLRLSCAASGFTFS;
HC-FR2 is WVRQAPGKGLEWVS;
HC-FR3 is RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR;
HC-FR4 is WGQGTLVTVSS. [0092] In a still further aspect, the light chain framework sequences are lambda light chain sequences.
[0093] In a still further aspect, one or more of the light chain framework sequences is the following: LC-FR1 is QSALTQPASVSGSPGQSITISC;
LC-FR2 is WYQQHPGKAPKLMIY;
LC-FR3 is GVSNRFSGSKSGNTASLTISGLQAEDEADYYC;
LC-FR4 is FGTGTKVTVL.
[0094] In a still further aspect, the heavy chain variable region polypeptide, antibody, or antibody fragment further includes at least a CHI domain.
[0095] In a more specific aspect, the heavy chain variable region polypeptide, antibody, or antibody fragment further includes a CHI, a CH2, and a CH3 domain.
[0096] In a still further aspect, the variable region light chain, antibody, or antibody fragment further includes a CL domain. [0097] In a still further aspect, the antibody further includes a CHI, a CH2, a CH3, and a CL domain.
[0098] In a still further specific aspect, the antibody further includes a human or murine constant region.
[0099[ In a still further aspect, the human constant region is selected from the group consisting of lgGl , IgG2, IgG2, IgG3, IgG4.
[00100] In a still further specific aspect, the human or murine constant region is lgGl.
[00101] In yet another embodiment, the invention features an anti-PD-Ll antibody including a heavy chain and a light chain variable region sequence, where:
(a) the heavy chain includes an HVR-Hl, an HVR-H2, and an HVR-H3, having at least 80% overall sequence identity to SYIMM, SIYPSGGITFYADTVKG, and IKLGTVTTVDY, respectively, and (b) the light chain includes an HVR-Ll, an HVR-L2, and an HVR-L3, having at least 80% overall sequence identity to TGTS SD VGGYNY VS , DVSNRPS, and SSYTSSSTRV, respectively.
[00102] In a specific aspect, the sequence identity is 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
[00103J In yet another embodiment, the invention features an anti-PD-Ll antibody including a heavy chain and a light chain variable region sequence, where:
(a) the heavy chain includes an HVR-Hl, an HVR-H2, and an HVR-H3, having at least 80% overall sequence identity to MYMMM, SIYPSGGITFYADS VKG, and IKLGTVTTVDY, respectively, and
(b) the light chain includes an HVR-Ll, an HVR-L2, and an HVR-L3, having at least 80% overall sequence identity to TGTSSDVGAYNYVS, DVSNRPS, and SSYTSSSTRV, respectively.
[00104] In a specific aspect, the sequence identity is 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
[00105] In a still further aspect, in the antibody or antibody fragment according to the invention, as compared to the sequences of HVR-Hl, HVR-H2, and HVR-H3, at least those amino acids remain unchanged that are highlighted by underlining as follows:
(a) in HVR-Hl SYIMM,
(b) in HVR-H2 SIYPSGGITFYADTVKG,
(c) in HVR-H3 IKLGTVTTVDY; and further where, as compared to the sequences of HVR-Ll, HVR-L2, and HVR-L3 at least those amino acids remain unchanged that are highlighted by underlining as follows:
(a) HVR-Ll TGTSSD VGGYNY VS
(b) HVR-L2 DVSNRPS
(c) HVR-L3 SSYTSSSTRV. [00106] In another aspect, the heavy chain variable region includes one or more framework sequences juxtaposed between the HVRs as: (HC-FR1)-(HVR-H1)-(HC-FR2)-(HVR-H2)-(HC- FR3)-(HVR-H3)-(HC-FR4), and the light chain variable regions include one or more framework sequences juxtaposed between the HVRs as: (LC-FR1)-(HVR-L1)-(LC-FR2)- (HVR-L2)-(LC-FR3)-(HVR-L3)-(LC-FR4).
[00107] In yet another aspect, the framework sequences are derived from human germline sequences.
[00108] In a still further aspect, one or more of the heavy chain framework sequences is the following: HC-FR1 is EVQLLESGGGLVQPGGSLRLSCAASGFTFS;
HC-FR2 is WVRQ APGKGLE WVS ;
HC-FR3 is RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR;
HC-FR4 is WGQGTLVTVSS.
[00109] In a still further aspect, the light chain framework sequences are derived from a lambda light chain sequence.
[00110] In a still further aspect, one or more of the light chain framework sequences is the following:
LC-FR1 is QSALTQPASVSGSPGQSITISC;
LC-FR2 is WYQQHPGKAPKLMIY; LC-FR3 is GVSNRFSGSKSGNTASLTISGLQAEDEADYYC;
LC-FR4 is FGTGTKVTVL.
[00111] In a still further specific aspect, the antibody further includes a human or murine constant region.
[00112] In a still further aspect, the human constant region is selected from the group consisting of IgG 1 , IgG2, IgG2, IgG3 , IgG4. [00113] In a still further embodiment, the invention features an anti-PD-Ll antibody including a heavy chain and a light chain variable region sequence, where:
(a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence: EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYIMMVWRQAPGKGLEWVSSIYPSGGITF YADWKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR1KLGTVTTVDYWGQGTLV TVSS, and
(b) the light chain sequence has at least 85% sequence identity to the light chain sequence: QS ALTQPAS VSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSN RPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTRVFGTGTKVTVL.
[00114] In a specific aspect, the sequence identity is 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
[00115] In a still further embodiment, the invention provides for an anti-PD-Ll antibody including a heavy chain and a light chain variable region sequence, where:
(a) the heavy chain sequence has at least 85 % sequence identity to the heavy chain sequence:
EVQLLESGGGLVQPGGSLRLSCAASGFTFSMYMMMWVRQAPGKGLEVWSSIYPSGGI TFYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCARIKLGTVTTVDYWG QGTLVTVSS, and
(b) the light chain sequence has at least 85% sequence identity to the light chain sequence:
QSALTQPASVSGSPGQSITISCTGTSSDVGAYNYVSWYQQHPGKAPKLMIYDVSNR PSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTRVFGTGT VTVL. [00116] In a specific aspect, the sequence identity is 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%.
[00117] In another embodiment the antibody binds to human, mouse, or cynomolgus monkey PD-Ll . In a specific aspect the antibody is capable of blocking the interaction between human, mouse, or cynomolgus monkey PD-Ll and the respective human, mouse, or cynomolgus monkey PD-1 receptors.
[00118] In another embodiment, the antibody binds to human PD-Ll with a KD of 5xl0"9 M or less, preferably with a KD of 2xl0"9 M or less, and even more preferred with a KD of lxlO"9 M or less.
[00119] In yet another embodiment, the invention relates to an anti-PD-Ll antibody or antigen binding fragment thereof which binds to a functional epitope including residues Y56 and D61 of human PD-Ll.
[00120] In a specific aspect, the functional epitope further includes E58, E60, Q66, Rl 13, and Ml 15 of human PD-Ll.
[00121] In a more specific aspect, the antibody binds to a conformational epitope, including residues 54-66 and 112-122 of human PD-Ll.
[00122] In a further embodiment, the invention is related to an anti-PD-Ll antibody, or antigen binding fragment thereof, which cross-competes for binding to PD-Ll with an antibody according to the invention as described herein.
[00123] In a still further embodiment, the invention features proteins and polypeptides including any of the above described anti-PD-Ll antibodies in combination with at least one pharmaceutically acceptable carrier.
[00124] In a still further embodiment, the invention features an isolated nucleic acid encoding a polypeptide, or light chain or a heavy chain variable region sequence of an anti-PD-Ll antibody, or antigen binding fragment thereof, as described herein. In a still further
embodiment, the invention provides for an isolated nucleic acid encoding a light chain or a heavy chain variable region sequence of an anti-PD-Ll antibody, wherein:
(a) the heavy chain includes an HVR-H1, an HVR-H2, and an HVR-H3 sequence having at least 80% sequence identity to SYIMM, SIYPSGGITFYADTVKG, and
IKLGTVTTVDY, respectively, or
(b) the light chain includes an HVR-L1, an HVR-L2, and an HVR-L3 sequence having at least 80% sequence identity to TGTSSDVGGYNYVS, DVSNRPS, and SSYTSSSTRV, respectively. [00125] In a specific aspect, the sequence identity is 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
[00126] In a further aspect, the nucleic acid sequence for the heavy chain is: atggagttgc ctgttaggct gttggtgctg atgttctgga ttcctgctag ctccagcgag 60
gtgcagctgc tggaatccgg cggaggactg gtgcagcctg gcggctccct gagactgtct 120
tgcgccgcct ccggcttcac cttctccagc tacatcatga tgtgggtgcg acaggcccct 180
ggcaagggcc tggaatgggt gtcctccatc tacccctccg gcggcatcac cttctacgcc 240
gacaccgtga agggccggtt caccatctcc cgggacaact ccaagaacac cctgtacctg 300
cagatgaact ccctgcgggc cgaggacacc gccgtgtact actgcgcccg gatcaagctg 360
ggcaccgtga ccaccgtgga ctactggggc cagggcaccc tggtgacagt gtcctccgcc 420
tccaccaagg gcccatcggt cttccccctg gcaccctcct ccaagagcac ctctgggggc 480
acagcggccc tgggctgcct ggtcaaggac tacttccccg aaccggtgac ggtgtcgtgg 540
aactcaggcg ccctgaccag cggcgtgcac accttcccgg ctgtcctaca gtcctcagga 600
ctctactccc tcagcagcgt ggtgaccgtg ccctccagca gcttgggcac ccagacctac 660
atctgcaacg tgaatcacaa gcccagcaac accaaggtgg acaagaaagt tgagcccaaa 720
tcttgtgaca aaactcacac atgcccaccg tgcccagcac ctgaactcct ggggggaccg 780
tcagtcttcc tcttcccccc aaaacccaag gacaccctca tgatctcccg gacccctgag 840
gtcacatgcg tggtggtgga cgtgagccac gaagaccctg aggtcaagtt caactggtac 900
gtggacggcg tggaggtgca taatgccaag acaaagccgc gggaggagca gtacaacagc 960
acgtaccgtg tggtcagcgt cctcaccgtc ctgcaccagg actggctgaa tggcaaggag 1020
tacaagtgca aggtctccaa caaagccctc ccagccccca tcgagaaaac catctccaaa 1080
gccaaagggc agccccgaga accacaggtg tacaccctgc ccccatcacg ggatgagctg 1140
accaagaacc aggtcagcct gacctgcctg gtcaaaggct tctatcccag cgacatcgcc 1200
gtggagtggg agagcaatgg gcagccggag aacaactaca agaccacgcc tcccgtgctg 1260
gactccgacg gctccttctt cctctatagc aagctcaccg tggacaagag caggtggcag 1320
caggggaacg tcttctcatg ctccgtgatg catgaggctc tgcacaacca ctacacgcag 1380
aagagcctct ccctgtcccc gggtaaa 1407 and the nucleic acid sequence for the light chain is: atggagttgc ctgttaggct gttggtgctg atgttctgga ttcctgcttc cttaagccag 60 tccgccctga cccagcctgc ctccgtgtct ggctcccctg gccagtccat caccatcagc 120
tgcaccggca cctccagcga cgtgggcggc tacaactacg tgtcctggta tcagcagcac 180
cccggcaagg cccccaagct gatgatctac gacgtgtcca accggccctc cggcgtgtcc 240
aacagattct ccggctccaa gtccggcaac accgcctccc tgaccatcag cggactgcag 300
gcagaggacg aggccgacta ctactgctcc tcctacacct cctccagcac cagagtgttc 360
ggcaccggca caaaagtgac cgtgctgggc cagcccaagg ccaacccaac cgtgacactg 420
ttccccccat cctccgagga actgcaggcc aacaaggcca ccctggtctg cctgatctca 480
gatttctatc caggcgccgt gaccgtggcc tggaaggctg atggctcccc agtgaaggcc 540
ggcgtggaaa ccaccaagcc ctccaagcag tccaacaaca aatacgccgc ctcctcctac 600
ctgtccctga cccccgagca gtggaagtcc caccggtcct acagctgcca ggtcacacac 660
gagggctcca ccgtggaaaa gaccgtcgcc cccaccgagt gctca 705
[00127] Further exemplary anti-PD-Ll antibodies that can be used in an anti-PD-Ll TGFp Trap are described in US patent application publication US 2010/0203056. In one embodiment of the invention, the antibody moiety is YW243.55S70. In another embodiment of the invention, the antibody moiety is MPDL3280A.
[00128] In a further embodiment, the invention features an anti-PD-Ll antibody moiety including a heavy chain and a light chain variable region sequence, where:
(a) the heavy chain sequence has at least 85% sequence identity to the heavy chain sequence: EVQLVESGGGLVQPGGSLRLSCAASGFTFSDSWIHWVRQAPGKGLEWVAWISPYGGSTYY
ADSVKGRFTISADTSKNTAYLQMNSLRAEDTAVYYCARRHWPGGFDYWGQGTLVTVSS (SEQ ID NO: 12), and
(b) the light chain sequence has at least 85% sequence identity to the light chain sequence: DIQMTQSPSSLSASVGDRVTITCRASQDVSTAVAWYQQKPGKAPKLLIYSASFLYSGVPSRFS GSGSGTDFTLTISSLQPEDFATYYCQQYLYHPATFGQGTKVEIKR (SEQ ID NO: 13).
[00129] In a specific aspect, the sequence identity is 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
[00130] In a further embodiment, the invention features an anti-PD-Ll antibody moiety including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain variable region sequence is:
EVQLVESGGGLVQPGGSLRLSCAASGFTFSDSWI H VRQAPGKGLE VAWI SPYGGSTYY ADSVKGRFTI SADTSKNTAYLQMNSLRAEDTAVYYCARRHWPGGFDYWGQGTLVTVS S (SEQ ID NO: 12), and (b) the light chain variable region sequence is:
DIQMTQS PSSLSASVGDRVT ITCRASQDVSTAVAWYQQKPGKAPKLLIYSASFLYSGVPSRFS GSGSGTDFTLT I S SLQPEDFATYYCQQYLYHPATFGQGTKVE IKR (SEQ ID NO:13).
[00131] In a further embodiment, the invention features an anti-PD-L 1 antibody moiety including a heavy chain and a light chain variable region sequence, where: (a) the heavy chain variable region sequence is:
EVQLVESGGGLVQPGGSLRLSCAASGFTFSDS I HWVRQAPGKGLEWVAWI SPYGGSTYYADS VKGRF I SAD SKNTAYLQMNSLRAEDTAVYYCARRHWPGGFDYWGQGTLVTVS (SEQ ID NO: 14), and
(b) the light chain variable region sequence is: DIQMTQS PSSLSASVGDRVTITCRASQDVSTAVAWYQQKPGKAPKLLIYSASFLYSGVPSRFS GSGSGTDFTLTI S SLQPEDFATYYCQQYLYHPATFGQGTKVE IKR (SEQ ID NO:13).
[00132j Yet further exemplary anti-PD-L 1 antibodies that can be used in an anti-PD- Ll/TGFp Trap are described in US patent publication US 7,943,743.
[00133] In one embodiment of the invention, the anti-PD-L 1 antibody is MDX-1 105. [00134] In yet a further embodiment, the anti-PD-Ll antibody is MEDI-4736.
Constant region
[00135] The proteins and peptides of the invention can include a constant region of an immunoglobulin or a fragment, analog, variant, mutant, or derivative of the constant region. In preferred embodiments, the constant region is derived from a human immunoglobulin heavy chain, for example, IgGl , IgG2, IgG3, IgG4, or other classes. In one embodiment, the constant region includes a CH2 domain. In another embodiment, the constant region includes CH2 and CH3 domains or includes hinge-CH2-CH3. Alternatively, the constant region can include all or a portion of the hinge region, the CH2 domain and/or the CH3 domain.
[00136] In one embodiment, the constant region contains a mutation that reduces affinity for an Fc receptor or reduces Fc effector function. For example, the constant region can contain a mutation that eliminates the glycosylation site within the constant region of an IgG heavy chain. In some embodiments, the constant region contains mutations, deletions, or insertions at an amino acid position corresponding to Leu234, Leu235, Gly236, Gly237, Asn297, or Pro331 of IgGl (amino acids are numbered according to EU nomenclature). In a particular
embodiment, the constant region contains a mutation at an amino acid position corresponding to Asn297 of IgGl . In alternative embodiments, the constant region contains mutations, deletions, or insertions at an amino acid position corresponding to Leu281, Leu282, Gly283, Gly284, Asn344, or Pro378 of IgGl .
[00137] In some embodiments, the constant region contains a CH2 domain derived from a human IgG2 or IgG4 heavy chain. Preferably, the CH2 domain contains a mutation that eliminates the glycosylation site within the CH2 domain. In one embodiment, the mutation alters the asparagine within the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence within the CH2 domain of the IgG2 or lgG4 heavy chain. Preferably, the mutation changes the asparagine to a glutamine. Alternatively, the mutation alters both the phenylalanine and the asparagine within the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence. In one embodiment, the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence is replaced with a Gln-Ala-Gln-Ser (SEQ ID NO: 16) amino acid sequence. The asparagine within the Gln-Phe- Asn-Ser (SEQ ID NO: 15) amino acid sequence corresponds to Asn297 of IgGl.
[00138] In another embodiment, the constant region includes a CH2 domain and at least a portion of a hinge region. The hinge region can be derived from an immunoglobulin heavy chain, e.g., IgGl, IgG2, IgG3, IgG4, or other classes. Preferably, the hinge region is derived from human IgGl, IgG2, IgG3, IgG4, or other suitable classes. More preferably the hinge region is derived from a human IgGl heavy chain. In one embodiment the cysteine in the Pro- Lys-Ser-Cys-Asp-Lys (SEQ ID NO: 17) amino acid sequence of the IgGl hinge region is altered. In a preferred embodiment the Pro-Lys-Ser-Cys-Asp-Lys (SEQ ID NO: 17) amino acid sequence is replaced with a Pro-Lys-Ser-Ser-Asp-Lys (SEQ ID NO: 18) amino acid sequence. In one embodiment, the constant region includes a CH2 domain derived from a first antibody isotype and a hinge region derived from a second antibody isotype. In a specific embodiment, the CH2 domain is derived from a human IgG2 or IgG4 heavy chain, while the hinge region is derived from an altered human IgGl heavy chain.
[00139] The alteration of amino acids near the junction of the Fc portion and the non-Fc portion can dramatically increase the serum half-life of the Fc fusion protein (PCT publication WO 01/58957, the disclosure of which is hereby incorporated by reference). Accordingly, the junction region of a protein or polypeptide of the present invention can contain alterations that, relative to the naturally-occurring sequences of an immunoglobulin heavy chain and
erythropoietin, preferably lie within about 10 amino acids of the junction point. These amino acid changes can cause an increase in hydrophobicity. In one embodiment, the constant region is derived from an IgG sequence in which the C-terminal lysine residue is replaced. Preferably, the C-terminal lysine of an IgG sequence is replaced with a non-lysine amino acid, such as alanine or leucine, to further increase serum half-life. In another embodiment, the constant region is derived from an IgG sequence in which the Leu-Ser-Leu-Ser (SEQ ID NO: 19) amino acid sequence near the C-terminus of the constant region is altered to eliminate potential junctional T-cell epitopes. For example, in one embodiment, the Leu-Ser-Leu-Ser amino acid sequence is replaced with an Ala-Thr-Ala-Thr (SEQ ID NO: 20) amino acid sequence. In other embodiments, the amino acids within the Leu-Ser-Leu-Ser (SEQ ID NO: 19) segment are replaced with other amino acids such as glycine or proline. Detailed methods of generating amino acid substitutions of the Leu-Ser-Leu-Ser (SEQ ID NO: 19) segment near the C- terminus of an IgGl, IgG2, IgG3, IgG4, or other immunoglobulin class molecule have been described in U.S. Patent Publication No. 2003/0166877, the disclosure of which is hereby incorporated by reference.
[00140] Suitable hinge regions for the present invention can be derived from IgGl, IgG2, IgG3, IgG4, and other immunoglobulin classes. The IgGl hinge region has three cysteines, two of which are involved in disulfide bonds between the two heavy chains of the immunoglobulin. These same cysteines permit efficient and consistent disulfide bonding formation between Fc portions. Therefore, a preferred hinge region of the present invention is derived from IgGl, more preferably from human IgGl. In some embodiments, the first cysteine within the human IgGl hinge region is mutated to another amino acid, preferably serine. The IgG2 isotype hinge region has four disulfide bonds that tend to promote oligomerization and possibly incorrect disulfide bonding during secretion in recombinant systems. A suitable hinge region can be derived from an IgG2 hinge; the first two cysteines are each preferably mutated to another amino acid. The hinge region of IgG4 is known to form interchain disulfide bonds inefficiently. However, a suitable hinge region for the present invention can be derived from the IgG4 hinge region, preferably containing a mutation that enhances correct formation of disulfide bonds between heavy chain-derived moieties (Angal S, et al. (1993) Mol. Immunol., 30: 105-8).
[00141] In accordance with the present invention, the constant region can contain CH2 and/or CH3 domains and a hinge region that are derived from different antibody isotypes, i.e., a hybrid constant region. For example, in one embodiment, the constant region contains CH2 and/or CH3 domains derived from IgG2 or IgG4 and a mutant hinge region derived from IgGl.
Alternatively, a mutant hinge region from another IgG subclass is used in a hybrid constant region. For example, a mutant form of the IgG4 hinge that allows efficient disulfide bonding between the two heavy chains can be used. A mutant hinge can also be derived from an IgG2 hinge in which the first two cysteines are each mutated to another amino acid. Assembly of such hybrid constant regions has been described in U.S. Patent Publication No. 2003/0044423, the disclosure of which is hereby incorporated by reference.
[00142] In accordance with the present invention, the constant region can contain one or more mutations described herein. The combinations of mutations in the Fc portion can have additive or synergistic effects on the prolonged serum half-life and increased in vivo potency of the bifunctional molecule. Thus, in one exemplary embodiment, the constant region can contain (i) a region derived from an IgG sequence in which the Leu-Ser-Leu-Ser (SEQ ID NO: 19) amino acid sequence is replaced with an Ala-Thr-Ala-Thr (SEQ ID NO: 20) amino acid sequence; (ii) a C-terminal alanine residue instead of lysine; (iii) a CH2 domain and a hinge region that are derived from different antibody isotypes, for example, an IgG2 CH2 domain and an altered IgGl hinge region; and (iv) a mutation that eliminates the glycosylation site within the IgG2-derived CH2 domain, for example, a Gln-Ala-Gln-Ser (SEQ ID NO: 16) amino acid sequence instead of the Gln-Phe-Asn-Ser (SEQ ID NO: 15) amino acid sequence within the IgG2-derived CH2 domain.
Antibody fragments
[00143] The proteins and polypeptides of the invention can also include antigen-binding fragments of antibodies. Exemplary antibody fragments include scFv, Fv, Fab, F(ab')2, and single domain VHH fragments such as those of camelid origin. [00144] Single-chain antibody fragments, also known as single-chain antibodies (scFvs), are recombinant polypeptides which typically bind antigens or receptors; these fragments contain at least one fragment of an antibody variable heavy-chain amino acid sequence (VH) tethered to at least one fragment of an antibody variable light-chain sequence (VL) with or without one or more interconnecting linkers. Such a linker may be a short, flexible peptide selected to assure that the proper three-dimensional folding of the VL and VH domains occurs once they are linked so as to maintain the target molecule binding-specificity of the whole antibody from which the single-chain antibody fragment is derived. Generally, the carboxyl terminus of the VL or VH sequence is covalently linked by such a peptide linker to the amino acid terminus of a complementary VL and VH sequence. Single-chain antibody fragments can be generated by molecular cloning, antibody phage display library or similar techniques. These proteins can be produced either in eukaryotic cells or prokaryotic cells, including bacteria. [00145] Single-chain antibody fragments contain amino acid sequences having at least one of the variable regions or CDRs of the whole antibodies described in this specification, but are lacking some or all of the constant domains of those antibodies. These constant domains are not necessary for antigen binding, but constitute a major portion of the structure of whole antibodies. Single-chain antibody fragments may therefore overcome some of the problems associated with the use of antibodies containing part or all of a constant domain. For example, single-chain antibody fragments tend to be free of undesired interactions between biological molecules and the heavy-chain constant region, or other unwanted biological activity.
Additionally, single-chain antibody fragments are considerably smaller than whole antibodies and may therefore have greater capillary permeability than whole antibodies, allowing single- chain antibody fragments to localize and bind to target antigen-binding sites more efficiently. Also, antibody fragments can be produced on a relatively large scale in prokaryotic cells, thus facilitating their production. Furthermore, the relatively small size of single-chain antibody fragments makes them less likely than whole antibodies to provoke an immune response in a recipient. [00146] Fragments of antibodies that have the same or comparable binding characteristics to those of the whole antibody may also be present. Such fragments may contain one or both Fab fragments or the F(ab')2 fragment. The antibody fragments may contain all six CDRs of the whole antibody, although fragments containing fewer than all of such regions, such as three, four or five CDRs, are also functional. Protein production
[00147] The antibody-cytokine trap proteins are generally produced recombinantly, using mammalian cells containing a nucleic acid engineered to express the protein. Although one example of a suitable cell line and protein production method is described in Examples 1 and 2, a wide variety of suitable vectors, cell lines and protein production methods have been used to produce antibody-based biopharmaceuticals and could be used in the synthesis of these antibody-cytokine trap proteins.. Therapeutic indications
[00148] The anti-PD-Ll/TGF Trap proteins described in the application can be used to treat cancer or reduce tumor growth in a patient. Exemplary cancers include colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, myeloma, lymphoma, leukemia, thyroid, endometrial, uterine, bladder, neuroendocrine, head and neck, liver, nasopharyngeal, testicular, small cell lung cancer, non-small cell lung cancer, melanoma, basal cell skin cancer, squamous cell skin cancer, dermatofibrosarcoma protuberans, Merkel cell carcinoma, glioblastoma, glioma, sarcoma, mesothelioma, and myelodisplastic syndromes.
[00149] The cancer or tumor to be treated with an anti-PD-Ll/ TGFp Trap may be selected based on the expression or elevated expression of PD-L1 and TGFP in the tumor, the correlation of their expression levels with prognosis or disease progression, and preclinical and clinical experience on the sensitivity of the tumor to treatments targeting PD-L1 and TGFp. Such cancers or tumors include but are not limited to colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, bladder, head and neck, liver, non-small cell lung cancer, melanoma, Merkel cell carcinoma, and mesothelioma. Pharmaceutical compositions
[00150] The present invention also features pharmaceutical compositions that contain a therapeutically effective amount of a protein described herein. The composition can be formulated for use in a variety of drug delivery systems. One or more physiologically acceptable excipients or carriers can also be included in the composition for proper
formulation. Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 17th ed., 1985. For a brief review of methods for drug delivery, see, e.g., Langer (Science 249:1527-1533, 1990).
[00151] The pharmaceutical compositions are intended for parenteral, intranasal, topical, oral, or local administration, such as by a transdermal means, for therapeutic treatment. The pharmaceutical compositions can be administered parenterally (e.g., by intravenous, intramuscular, or subcutaneous injection), or by oral ingestion, or by topical application or intraarticular injection at areas affected by the vascular or cancer condition. Additional routes of administration include intravascular, intra-arterial, intratumor, intraperitoneal,
intraventricular, intraepidural, as well as nasal, ophthalmic, intrascleral, intraorbital, rectal, topical, or aerosol inhalation administration. Thus, the invention provides compositions for parenteral administration that comprise the above mention agents dissolved or suspended in an acceptable carrier, preferably an aqueous carrier, e.g., water, buffered water, saline, PBS, and the like. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, detergents and the like. The invention also provides compositions for oral delivery, which may contain inert ingredients such as binders or fillers for the formulation of a tablet, a capsule, and the like. Furthermore, this invention provides compositions for local administration, which may contain inert ingredients such as solvents or emulsifiers for the formulation of a cream, an ointment, and the like.
[00152] These compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as-is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the preparations typically will be between 3 and 1 1 , more preferably between 5 and 9 or between 6 and 8, and most preferably between 7 and 8, such as 7 to 7.5. The resulting compositions in solid form may be packaged in multiple single dose units, each containing a fixed amount of the above-mentioned agent or agents, such as in a sealed package of tablets or capsules. The composition in solid form can also be packaged in a container for a flexible quantity, such as in a squeezable tube designed for a topically applicable cream or ointment.
[00153] The optimal dose of the antibody-TGFP trap is based on the percent receptor occupancy by the antibody moiety to achieve maximal therapeutic effect because the cytokine trap is used in a large excess. For example, the therapeutic dose for a monoclonal antibody targeting a cellular receptor is determined such that the trough level is around 10 to 100 μg/ml, i.e., 60 to 600 nM (for antibody with a dissociation constant (KD) of 6 nM, this trough level would ensure that between 90 to 99% of the target receptors on the cells are occupied by the antibody). This is in large excess of cytokines, which are typically present in pg to ng/ml in circulation.
[00154] The optimal dose of antibody-TGFP trap polypeptide of the invention will depend on the disease being treated, the severity of the disease, and the existence of side effects. The optimal dose can be determined by routine experimentation. For parenteral administration a dose between 0.1 mg/kg and 100 mg/kg, alternatively between 0.5 mg/kg and 50 mg/kg, alternatively, between 1 mg/kg and 25 mg/kg, alternatively between 2 mg/kg and 10 mg/kg, alternatively between 5 mg/kg and 10 mg/kg is administered and may be given, for example, once weekly, once every other week, once every third week, or once monthly per treatment cycle.
EXAMPLES
[00155] The invention now being generally described, will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the scope of the invention in any way.
EXAMPLE 1 - DNA construction and protein expression
[00156] Anti-PD-Ll /TGFp Trap is an anti-PD-Ll antibody-TGFp Receptor II fusion protein. The light chain of the molecule is identical to the light chain of the anti-PD-Ll antibody (SEQ ID NO: 1). The heavy chain of the molecule (SEQ ID NO:3) is a fusion protein comprising the heavy chain of the anti-PD-Ll antibody (SEQ ID NO: 2) genetically fused to via a flexible
(Gly4Ser)4Gly linker (SEQ ID NO:l 1) to the N-terminus of the soluble TGFp Receptor II (SEQ ID NO: 10). At the fusion junction, the C-terminal lysine residue of the antibody heavy chain was mutated to alanine to reduce proteolytic cleavage. For expression of anti-PD-Ll /TGFp Trap, the DNA encoding the anti-PD-Ll light chain (SEQ ID NO:4) and the DNA encoding the anti-PD-Ll /TGF Receptor II (SEQ ID NO:5) in either the same expression vector or separate expression vectors were used to transfect mammalian cells using standard protocols for transient or stable transfection. Conditioned culture media were harvested and the anti-PD- Ll /TGFp Trap fusion protein was purified by standard Protein A Sepharose chromatography. The purified protein comprising one anti-PD-Ll antibody and two soluble TGFB Receptor II molecules (FIG. 1A) has an estimated molecular weight (MW) of about 190 kilodaltons on size exclusion chromatography and SDS-polyacrylamide electrophoresis under non-reducing conditions. Under reducing conditions, the light and heavy chains have apparent MW of 28 and 75 kilodaltons, respectively (FIG. IB).
[00157] The anti-PD-Ll (mut)/TGFp Trap fusion protein, which contains an analogous heavy chain fusion polypeptide (SEQ ID NO:7) and a light chain with the mutations A31G, D52E, R99Y in the variable region that abrogate the binding to PD-L1 (SEQ ID NO:6), was similarly prepared. It was used in subsequent experiments as a TGFP Trap control.
EXAMPLE 2 - Production of anti-PD-Ll/TGFp Trap as a biotherapeutic
[00158] The anti-PD-Ll/TGFp Trap produced by transient transfection of human embryonic kidney 293 (HEK) cells was found to contain varying degrees of a clipped species, which appeared as a faint band with an apparent MW of about 60 kD on SDS-PAGE under reducing conditions (FIG. IB). This band was confirmed to be the heavy chain of the anti-PD-Ll/TGFp Trap cleaved at a site in the N-terminal portion of TGFpRII close to the fusion junction.
[00159] Stable clones expressing anti-PD-Ll/TGFP Trap were generated in the CHO-S host cell line, which was pre-adapted for growth in serum-free media in suspension culture. Cells were transfected with an expression vector containing a gene encoding the anti-PD-Ll- TGFpRII protein and a glutamine synthetase selection marker. Subsequent selection of stable integrants was made with L-methionine sulfoximine (MSX). Anti-PD-Ll/TGF Trap expressing cell lines were generated using a minipool approach, followed by the deposition of single cells into 384-well plates, using a Beckton-Dickinson fluorescence activated cell sorter (FACS Aria II). Growth, productivity, and protein quality were evaluated in a generic platform fed-batch assay. Based on these analyses, 14 clones were selected as lead candidates for further studies. A stability study with the clones was carried out to -90 PDL (population doubling level) from research cell banks established during scale up of clones. At the conclusion of mini- pool development it was discovered that the heavy chain-linker-TGFpRII subunit underwent clipping, as was seen in transient expression. All clones in the stability study produced the clipped species, although it was shown in the protein A-purified material that the percent clipped species relative to the intact subunit varied with each clone. In addition, an improved purification process consisting a protein A chromatography followed by strong cation exchange was developed to reduce co-purification of the clipped species. Even with the improved process, purified material with the required final levels of clipped species of <5% could only be achieved using clones producing low levels of clipping. Based on these combined analyses, clone 02B 15 was selected as the final candidate clone. Analysis of anti-PD-Ll/TGFp Trap expressed by this clone at zero PDL, thirty PDL, sixty PDL and ninety PDL shows that the percentage of clipping did not increase with population doubling levels (FIG. 2). EXAMPLE 3 - Fluorescence-activated cell sorting (FACS) analysis of binding of anti-PD- Ll/TGFp Trap and controls to human PD-Ll on cells
[00160] The binding of anti-PD-Ll antibody and fusion proteins on HEK cells stably transfected to express human PD-Ll was studied using the following procedure. [00161] The following exemplary procedure was used determine PD-Ll binding by FACS: a. 50 μΐ serial dilutions of test samples were set up in FACS buffer.
b. 50 μΐ of HEK cells stably transfected to express human PD-Ll at 5 xlO6 cells/ml were dispensed to the wells with test samples and mixed.
c. Plate(s) were incubated in the dark on ice for 1 hour.
d. Cells were pelleted at 300x g for 5 minutes.
e. Supernatant was decanted and cells were resuspended in 300 μΐ FACS buffer and re-pelleted at 300x g for 5.
f. Sample rinse was repeated.
g. Cells were resuspended in 100 μΐ FACS buffer containing DyLight 488 conjugated whole IgG Goat Anti-Human IgG, Fey (1:300 diluted). h. Plate(s) was incubated in the dark on ice for 45 minutes.
i. Cells were pelleted at 3 OOx g for 5.
j. Supernatant was decanted and cells were resuspended in 300 μΐ FACS buffer and re-pelleted at 3 OOx g for 5 minutes
k. Sample rinse was repeated and cells were finally resuspended in 200 μΐ FACS buffer.
1. Data was acquired on FACS Caliber and was analyzed using Microsoft Excel.
EC50 was calculated using non-linear regression (Sigmoidal dose-response) with Graphpad Prism5.
[00162] As shown in Fig. 3, FACS analysis showed that the anti-PD-Ll /TGFp Trap fusion protein retains similar binding affinity as the positive control anti-PD-Ll antibody on HEK cells stably transfected to express human PD-Ll (HEK/PD-Ll cells). The EC50's for anti-PD- Ll/TGF Trap and anti-PD-Ll are 0.1 16 μ^πιΐ (0.64 nM) and 0.061 μ^πιΐ (0.41 nM), respectively. The observed MFI (mean fluorescent intensity) was specific to binding to human PD-Ll since no MFI was observed on the parental HEK cells that were not transfected. The anti-PD-Ll (mut)/TGF Trap negative control did not show any binding to the HEK cells stably transfected to express human PD-Ll.
EXAMPLE 4 - Determination of ability of anti-PD-Ll/TGFp Trap to inhibit TGFp induced phosphorylation of SMAD3
[00163] The ability of anti-PD-Ll/TGFp Trap to neutralize TGF was determined using 4T1 cells carrying a SMAD3-luciferase reporter. In the assay detailed below, inhibition of TGF - induced phosphorylation of SMAD3 was measured using a luciferase reporter under the control of the SMAD3 promoter.
[00164] An exemplary assay to evaluate potency to inhibit TGFp-induced reporter activity was performed as follows.
1. One day prior to the study, 4T1 cells carrying SMAD3 -luciferase reporter were fed.
2. On day 0, cells were plated in a Biocoat 96-well plate at a concentration of 5xl04 cells/well in 100 μΐ of fresh media and incubated overnight at 37°C and 5% CO2.
3. On day 1 :
i. 50 μΐ of fresh complete media containing indicated concentration of anti- PD-L1/TGFP trap samples to be tested or its controls was added to the wells and incubated for one hour. All samples were tested in triplicates.
ii. 50 μΐ of fresh complete media containing 20 ng/ml human TGFp was added to each well and samples were incubated overnight (final concentration in the well is 5 ng/ml).
4. On day 2:
i. 100 μΐ culture supernatant was removed and 100 μΐ fresh complete media, containing 150 μg/ml D-Luciferin was added, and samples were incubated for at least five minutes.
ii. Luminescence was measured using Envision 2104 plate reader by recording CPM.
5. Data was analyzed using MS Excel or Graphpad prism 5. Luciferase activity was recorded as CPM. Inhibitory Activity of (%) was calculated using the following equation:
Inhibition (%) = (1- CPM of sample / CPM max of anti-PD-Ll treated sample) X 100
6. Nonlinear regression fit was carried out using Sigmoidal dose-response (variable slope) of Graphpad prism 5. IC50 values were calculated.
[00165] FIG. 4 shows that anti-PD-L l/TGFp Trap inhibits TGFp-induced pSMAD3 reporter activity in a dose dependent manner. The fact that the anti-PD-L l(mut)/TGFp Trap control had comparable potency and IC50 (concentration required to inhibit 50% of the maximal activity) plus the fact that the anti-PD-Ll antibody had no effect showed that this inhibition of signaling is independent of anti-PD-Ll activity. Surprisingly, anti-PD-L l /TGFp Trap was several-fold more potent than TGFpRII-Fc (R&D Systems), which places the TGFPRII at the N-terminus instead of the C-terminus of the fusion protein. It is also noteworthy that anti-PD-Ll /TGFp Trap is significantly more potent than IDl 1 (GC1008), the anti-TGFp antibody that was tested in patients with advanced malignant melanoma or renal cell carcinoma (Morris et ah, J Clin Oncol 2008; 26:9028 (Meeting abstract)). In this assay, IDl 1 and TGFpRII-Fc showed similar activity. EXAMPLE 5 - Pharmacokinetic (PK) analysis in mice
[00166] Eighteen male C57BL/6 mice, 5-6 weeks old, were randomly assigned to 3 groups (N=6/group), and each group received one of the three proteins (anti-PD-Ll/TGFp Trap, anti- PD-Ll (mut)/TGF Trap, and anti-PD-Ll). Mouse body weight was recorded before dosing. After a brief warm-up under a heating lamp, each mouse received 120 μg of protein in 200 μΐ intravenously (IV) via the tail vein regardless of its body weight. Each group dosed with the same protein was further divided into 2 subgroups (n=3). Blood samples were alternately taken from each of two subgroups, i.e. one subgroup was withdrawn for blood samples at lh, 24h, 72h, and 168h, whereas another subgroup was for blood samples at 7h, 48h, 120h, and 240h. At each time point, approximate 50 μΐ of blood samples were collected from each mouse via tail vein using a heparinized micro glass capillary (100 μΐ in capacity). The blood sample was then transferred to a tube pre-coated with Li-Heparin and kept at 4 °C. Within 10 min of collection, the blood samples were spun at 14,000 rpm for 10 min. At least 20 μΐ of plasma sample was transferred into a new set of pre-labeled tubes and stored at -20 °C until the day of analysis. [00167] The ELISA to measure total human IgG used goat anti-Human IgG (H+L) (heavy and light chains) (Jackson ImmunoResearch Laboratories) coated wells for capture and peroxidase-AffiniPure mouse anti-Human IgG, F(ab')2 (Jackson ImmunoResearch
Laboratories) for detection. The ELISA to measure fully functional anti-PD-Ll antibody and/or fusion protein used PD-Ll-Fc (extracellular domain of human PD-L1 fused to Fc) coated wells (coated at 1.25 μg/ml) for capture and peroxidase-AffiniPure mouse anti-Human IgG, F(ab')2 for detection. The ELISA to measure fully functional anti-PD-Ll and intact TGFpRII used PD- Ll-Fc coated wells for capture and biotinylated anti -human TGF RII (R&D Systems) for detection.
[00168] FIG. 5A shows that the anti-PD-Ll /TGFp Trap fusion protein had a PK profile very similar to that of the anti-PD-Ll antibody. For example, as measured by the total human IgG ELISA, the serum concentrations at the 168 hr time point of anti-PD-Ll /TGFp Trap and anti- PD-Ll were 16.8 and 16.2 μg/ml, respectively, and the respective area under the curve (AUC) from 0 to 168 hr were 4102 and 3841 hr^g/ml. Similarly, when the serum concentrations were measured by the total functional anti-PD-Ll ELISA, the serum concentrations at the 168 hr time point of anti-PD-Ll /TGFp Trap and anti-PD-Ll were 9.5 and 1 1.1 μg/ml, respectively, and the respective AUC from 0 to 168 hr were 3562 and 3086 hr^g/ml. The serum
concentration of intact anti-PD-Ll/TGFp Trap fusion protein was determined by the ELISA, which detects fully functional anti-PD-Ll and the fused TGF RII. In this case, the serum concentration of anti-PD-Ll/TGFP Trap was 5.9 μg/ml at the 168 hr time point and the AUC (0 to 168 hr) was 2656 hr-^g/ml, which were somewhat lower than those from the fully functional anti-PD-Ll ELISA, presumably due to degradation of the TGFpRII moiety after receptor- mediated endocytosis. Antibody binding to PD-Ll has been shown to result in PD-Ll -mediated endocytosis, and an antibody-X fusion protein is known to undergo degradation of the X moiety after receptor-mediated endocytosis (Gillies et ah, Clin Cancer Res. 2002; 8:210-6). This is supported by the finding in FIG. 5 that when the antibody moiety does not bind PD-Ll, as in the anti-PD-Ll (mut)/TGFp Trap control, the exposure is about 3 times higher, with a serum concentration of 53 μg/ml at the 168 hr time point and AUC(0 to 168 hr) of 9585 hr- μg/ml, suggesting that at least part of the clearance is receptor-mediated.
[00169] In order to confirm the ~3-fold difference in exposure between anti-PD-Ll /TGFp Trap and anti-PD-Ll (mut)/TGFp Trap, the pharmacokinetics experiment was repeated and the concentrations of the intact fusion proteins in the serum samples were determined. Mice (B6.129S2 female mice, 8 wks old, Jackson Lab) were injected with anti-PD-Ll /TGFp Trap or anti-PD-Ll (mut)/TGFp Trap (164 μg/mouse). The serum concentrations of the two fusion proteins were measured by an ELISA using anti-human IgG Fab (Jackson Immunoresearch, West Grove, PA) for capture and biotinylated anti-human TGFpRII (R&D Systems,
Minneapolis, MN) and peroxidase-conjugated streptavidin (Zymed/ThermoFisher Scientific, Grand Island, NY) to detect intact anti-PD-Ll /TGFP Trap proteins. The serum concentrations of the intact fusion proteins at various time points were shown in the Table below and plotted in FIG. 5B. The total area under the curve (AUC) up to 336 hr is 1 1781 hr^g/ml for anti-PD- Ll/TGFP Trap and 35575 hr^g/ml for anti-PD-Ll (mut)/TGFp Trap (Table 1), therefore confirming the three-fold higher exposure of the Trap control molecule.
Table 1. Exposures of anti-PD-Ll /TGFP Trap and the anti-PD-Ll (mut)/TGFp Trap control as determined by the area under the curve (AUC) in the pharmacokinetics graph in Fig. 5B.
AUC (h* g/ml)
Time (h) Anti-PD-Ll/TGF3 Trap Anti-PD-Ll(mut)/TGF3 Trap
7 72 173
24 1161 2789
48 1306 3511
72 1113 2968
120 2327 5192
168 2014 5225
240 2159 7530
336 1629 8188 total 1 11781 | 35575 |
EXAMPLE 6 - PD-L1 target-mediated endocytosis of anti-PD-Ll/ TGFp Trap
[00170] Receptor-mediated endocytosis was studied using the Alexa Fluor 488 quenching techniques according to manufacturer's protocol (Life Technologies, Carlsbad, CA). Briefly, HEK cells expressing PD-L1 (HEK/PD-L1 cells) were incubated with 10 μg/ml Alexa Fluor 488-conjugated anti-PD-Ll /TGFp Trap on ice for about 1 hr and washed 4 times with cold media. Washed cells were then pulsed at 37 °C for 0.25, 0.5, 0.75, 1, 1.5, 2, 3 and 4 hr to allow internalization. Cell samples at each time point were then divided into two portions. One portion was incubated on ice and total fluorescence from the Alexa Fluor 488-conjugated anti- PD-Ll/TGFP Trap bound on the cell surface and internalized was measured; the other portion was incubated with anti-Alexa Fluor 488 at 4 °C for about an hour and the non-quenchable fluorescence from the internalized Alexa Fluor 488-conjugated anti-PD-Ll /TGFp Trap was measured. A graph showing a time course of the non-quenchable and total mean fluorescence intensity (MFI) of anti-PD-Ll /TGFP Trap at 37 °C is shown in FIG. 6A. The receptor-mediated internalization kinetics is very similar to that of the anti-PD-Ll antibody, which is shown in FIG. 6B. The percentage of receptor-mediated internalization of anti-PD-Ll /TGFp Trap and anti-PD-Ll on HEK/PD-L1 cells at various time points at 37 °C is shown in FIG. 6C, using the following formula to account for the fact that the quenching by the anti-Alexa Fluor 488 is not 100%: Internalized fluorescence = Total MFI - (Total MFI - Non-quenchable MFI)/Quenching efficiency
EXAMPLE 7 - Anti-PD-Ll/TGFp Trap demonstrated a superior anti-tumor effect that is synergistic of anti-PD-Ll and TGFp Trap activities in the EMT-6 (breast carcinoma) subcutaneous model [00171] 8-12 week old female Jh (Igh-T-111101™) Balb/C mice (Taconic Farms, Hudson, NY) were inoculated with 0.5 x 106 viable EMT6 cells in 0.1 ml PBS on the right flanks
subcutaneously. About five days later, when tumors reached an average size of 20-30 mm3, mice were sorted into groups (N=10) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections was initiated (Day 0). Group 1 received 400 μg of isotype antibody control three times weekly (or "eod" (every other day); Group 2 received 400 μg of anti-PD-Ll antibody three times weekly; Group 3 received 164 μg of anti-PD- Ll(mut)/TGFp Trap three times weekly; Group 4 received 492 μ of anti-PD-Ll /TGFp Trap three times weekly; Group 5 received 492 μg of anti-PD-L l /TGF Trap twice weekly
(equimolar to 400 μ§ of anti-PD-Ll antibody); Group 6 received 164 μg of anti-PD-Ll /TGFp Trap three times weekly; and Group 7 received 55 μg of anti-PD-Ll /TGFp Trap three times weekly. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula: tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Anti-tumor efficacy was reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00172] All the treatments were well tolerated. The inhibition of tumor growth by the various treatments is shown in FIG. 7A, which showed the average tumor volumes of the surviving mice, and FIG. 7B, which showed the individual tumor volume of the surviving mice, noting that mice with tumors over 2500 mm3 had to be euthanized. Anti-PD-Ll /TGF Trap demonstrated potent anti-tumor efficacy, achieving T/C ratios of 0.30, 0.40, and 0.44 for the high (492 μg, Group 4), medium (164 μg, Group 6), and low (55 μg, Group 7) dose groups, respectively on Day 28.). While the anti-PD-Ll antibody (Group 2, T/C =0.87, p>0.05, on Day 16, the last day for which the average tumor volume of all the mice were available, i.e., before mice with tumors over 2500 mm3 were euthanized) or the TGF Trap control (Group 2, T/C =0.97 on Day 16, p>0.05) alone had marginal efficacy in this model, combining the two agents in a single molecule resulted in profound synergistic anti-tumor effect. This is evident in the median survival times observed for the 492 μg dose (58 and greater than 80 days, respectively, for three times weekly dosing and twice weekly dosing) and 164 μg dose (35 days) of the fusion protein (log rank test: p<0.0001) (FIG. 7C). Importantly, anti-PD-Ll /TGFp Trap at the medium dose of 164 μg (Group 6), with a median survival of 35 days, was far more efficacious than the same dose of anti-PD-Ll (mut)/TGFP Trap (Group 3) or three times the equivalent dose of anti-PD-Ll (Group 2), both of which yielded a median survival of 22 days, respectively (log rank test: pO.0001). This synergistic anti-tumor activity is especially striking because the exposure of the TGF Trap moiety of the 164 μg dose of PD-Ll(mut)/TGFp Trap should be about 3 times higher than that of the 164 μg dose of PD-Ll/TGFp Trap due to receptor- mediated clearance of the latter (see Examples 5 and 6). It is remarkable that tumors in mice which received the high dose of anti-PD-Ll /TGFp Trap continued to regress after dosing was stopped on Day 18 (3 of 10 from Group 4 and 6 of 10 from Group 5 with complete regressions at day 78), demonstrating the long-lasting immunologic anti-tumor effect of targeting the two immunosuppressive mechanisms simultaneously (FIG. 7C). It is also noteworthy that the efficacy for Group 4 is not any better than that of Group 5, suggesting that the dose of 492 μg administered twice weekly was near the saturating dose, or was a more optimal dosing regimen than the 492 μg administered three times weekly. [00173] The protective effect of the anti-tumor immunity elicited by the anti-PD-Ll /TGFp Trap treatment was evident when the mice with tumors in complete regression were challenged with 25,000 viable EMT6 cells injected subcutaneously. While all ten naive mice in a control group developed tumors to an average tumor volume of 726 mm3 by Day 18 post challenge, none of the eleven mice previously treated with PD-Ll/TGFp Trap (three from Group 4, six from Group 5, and one each from Groups 6 and 7) showed any sign of tumor growth.
EXAMPLE 8 - Anti-PD-Ll/TGF-β Trap showed profound synergistic anti-tumor activity in the MC38 (colorectal carcinoma) subcutaneous tumor model.
[00174] 8-12 week old female B6.129S2-IghmtmlCg J mice (Jackson Laboratory, Bar Harbor, ME) were injected with 0.5xl06 viable MC38 tumor cells in 0.1 ml PBS subcutaneously into the right flank. About eight days later, when average tumor size reached about 80-100 mm3, mice were sorted into groups (N=10) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections was initiated (Day 0). Group 1 received 400 μg of isotype antibody control; Group 2 received 400 g of anti-PD-Ll antibody; Group 3 received 133 μg of anti-PD-Ll antibody; Group 4 received 492 μg of anti-PD-Ll (mut)/TGFp Trap; Group 5 received 164 μg of anti-PD-Ll (mut)/TGFp Trap; Group 6 received 492 μg of anti-PD- Ll /TGFp Trap; and Group 7 received 164 μg of anti-PD-Ll /TGF Trap. The treatment was administered three times weekly for two weeks. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula: tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Anti-tumor efficacy was reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00175] All the treatments were well tolerated. The inhibition of tumor growth by the various treatments is shown in FIG. 8. On day 19 of the study, anti-PD-Ll/TGFP Trap demonstrated potent dose-dependent anti-tumor efficacy, achieving T/C ratios of 0.18 (pO.001) and 0.38
(pO.001) for the high (492 μg, Group 6) and low (164 μg, Group 7) dose groups, respectively. On the other hand, neither anti-PD-Ll or anti-PD-Ll (mut)/TGF Trap showed any anti-tumor activity at all. Therefore, a profound syngergistic anti-tumor activity was obtained when the anti-PD-Ll antibody and the TGFP Trap moiety were combined into one molecule to target these two immunosuppressive mechanisms simultaneously.
EXAMPLE 9 - Anti-PDLl/TGFp Trap was effective in the EMT-6 orthotopic model of metastatic breast cancer.
[00176] 8-12 week old female Jh (Igh-.FmlDhu) Balb/C mice (Taconic Farms, Hudson, NY) were inoculated with 0.25x106 viable EMT6 cells in 0.1 ml PBS into the right mammary pad. About a week later, when average tumor size reached about 50 mm3, mice were sorted into groups (N=10) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections was initiated (Day 0). Group 1 received 133 μg of isotype antibody control; Group 2 received 133 μg of anti-PD-Ll antibody; Group 3 received 164 μg of anti-PD- Ll (mut)/TGFp Trap; Group 4 received 164 μg of anti-PD-Ll /TGFp Trap; and Group 5 received a combination of 133 μg of anti-PD-Ll and 164 μg of anti-PD-Ll (mut)/TGFp Trap. Treatment was repeated on Days 0, 2, 4, 7, 9, 11 (i.e. 3 times weekly for two weeks). Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula: tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Anti-tumor efficacy was reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00177] All treatments were well tolerated. The inhibition of tumor growth by the various treatments is shown in FIG. 9. Anti-PD-Ll /TGFp Trap demonstrated potent anti-tumor efficacy, achieving T/C ratio of 0.03 (p<0.001) on Day 21. On the other hand, equimolar doses of anti-PD-Ll or anti-PD-Ll (mut)/TGFp Trap were less efficacious, giving T/C ratios of 0.31 (p<0.001 vs. Group 1 ; p<0.001 vs. Group 4) and 0.68 (pO.001 vs. Group 1; p<0.001 vs. Group 4), respectively. The combination therapy of equimolar doses of anti-PD-Ll and anti-PD- Ll (mut)/TGFp Trap achieved almost identical anti-tumor efficacy as the fusion protein, although the exposure of the TGF Trap of the fusion protein (Group 4) was estimated to be about 3-fold lower than that of the anti-PD-Ll (mut)/TGFp Trap in the combination (Group 5) based on pharmacokinetics analysis (see Example 5). It is also remarkable that the tumors in Groups 4 and 5 continued to regress after the last day of dosing, e.g., average tumor size decreased from 212 mm3 on Day 1 1, the last day of dosing, to 26 mm3 on Day 24 for anti-PD- LI /TGF Trap treatment, demonstrating the long-lasting immunologic anti-tumor effect of targeting the two immunosuppressive mechanisms simultaneously.
EXAMPLE 10 - Anti-PD-Ll/TGFp Trap has better anti-tumor efficacy than the combination of anti-PD-Ll and TGFp Trap in an intramuscular MC38 colorectal carcinoma model.
[00178] 8- 12 week old female B6.129S2-Ighmtmlc¾n/J mice (Jackson Laboratory, Bar Harbor, ME) were injected with 0.5xl06 viable MC38 tumor cells in 0.1 ml PBS intramuscularly in the right thigh. About a week later, when average tumor size reaches about 50 mm3, mice were sorted into groups (N=8) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections was initiated (Day 0) and repeated again two days later
(Day 2). Group 1 received 400 μg of isotype antibody control; Group 2 received 400 μg of anti- PD-Ll antibody; Group 3 received 133 μg of anti-PD-Ll antibody; Group 4 received 164 μg of anti-PD-Ll (mut)/TGFp Trap; Group 5 received 492 μg of anti-PD-Ll /TGFp Trap; Group 6 received 164 μg of anti-PD-Ll /TGFp Trap; and Group 7 received a combination of 133 μg of anti-PD-Ll and 164 μg of anti-PD-Ll (mut)/TGFp Trap. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula: tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Anti-tumor efficacy was reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00179] All the treatments were well tolerated. The inhibition of tumor growth by the various treatments is shown in FIG. 10. Anti -PD-Ll /TGFp Trap demonstrated very potent anti-tumor efficacy, achieving T/C ratios of 0.024 (p<0.001) and 0.052 (pO.001) for the high (492 μg, Group 5) and low (164 μg, Group 6) dose groups, respectively, on Day 15. On the other hand, equimolar doses of anti-PD-Ll were less efficacious, giving T/C ratios of 0.59 (p<0.001) and 0.45 (p<0.001) for the high (400 μg, Group 2) and low (133 μg, Group 3) dose groups, respectively. Anti-PD-Ll (mut)/TGFp Trap at 164 μg (Group 4) was completely ineffective, and it should be pointed out that although this dose is equimolar with the low dose anti-PD- Ll/TGFp Trap group (Group 6), the exposure of the TGFP Trap should be fairly similar to that of the high dose anti-PD-Ll/TGFp Trap group (Group 5) because of the differences in pharmacokinetics (see Example 5). Therefore, the data demonstrated that anti-PD-Ll/TGFp Trap had potent synergistic anti-tumor activity in this model. It is especially noteworthy that, anti-PD-Ll/TGFp Trap was more efficacious than the combination therapy of equimolar doses of anti-PD-Ll and anti-PD-Ll (mut)/TGFp Trap, which had a T/C ratio of 0.16 (pO.001 vs. Group 1 and p>0.05 vs. Group 6) despite a higher TGFp Trap exposure of about threefold (see Example 5). In addition, anti-PD-Ll /TGFp Trap treatment resulted in 4 out of 10 mice with complete tumor regression, while the combination of anti-PD-Ll and the Trap control induced complete regression in only 2 out of 10 mice (data not shown). It is also remarkable that the tumors in the mice treated with anti-PD-Ll /TGFp Trap continued to regress after the last day of dosing on day 2, and stayed completely regressed thereafter (until at least Day 102), demonstrating the profound and long-lasting immunologic anti-tumor effect of this fusion protein. Without being bound by theory, the data supports a mechanism in which the anti-PD- LI /TGFP Trap fusion protein not only exploits the synergistic effect of blocking the two major immune escape pathways, but is superior to the combination therapy due to the targeting of the tumor microenvironment by a single molecular entity. Many immunosuppressive cytokines secreted by tumor cells or subverted immune cells (e.g. tumor associated macrophages, myeloid-derived suppressor cells) have autocrines or paracrine functions. Therefore, anti-PD- L 1/TGFp Trap has the capability to deliver the TGFp Trap to the tumor microenvironment via binding to PD-L1+ tumor cells, where the Trap neutralizes the locally secreted TGFp. In addition, instead of acting just like a sink for bound TGFP that accumulates in circulation, anti- PD-Ll/TGFp Trap bound TGFp could be effectively destroyed through the PD-L1 receptor- mediated endocytosis (Examples 5 and 6). EXAMPLE 11 - Treatment with anti-PDLl/TGFp Trap or the combination of anti-PD- Ll and TGFp Trap control at equivalent exposure in the EMT-6 orthotopic model of metastatic breast cancer.
[00180] At equimolar doses, anti-PDLl/TGFp Trap had similar efficacy as the combination of anti-PD-Ll and TGFp Trap control in the orthotopic EMT-6 breast cancer model (Example 9). In the following study the efficacy of anti-PDLl/TGFp Trap or the combination of anti-PD- Ll and TGFp Trap control administered for equivalent exposure was tested.
[00181] 8-12 week old female Jh (Igh- 1101™) Balb/C mice (Taconic Farms, Hudson, NY) were inoculated with 0.25xl06 viable EMT6 cells in 0.1 ml PBS into the right mammary pad. About a week later, when average tumor size reached about 80 mm3, mice were sorted into groups (N=12) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections was initiated on Day 0 and repeated 7 days later. Group 1 received 133 μg of isotype antibody control; Group 2 received 164 μg of anti-PD-Ll /TGFp Trap; Group 3 received 55 μg of anti-PD-Ll /TGFp Trap; Group 4 received a combination of 133 of anti- PD-L1 and 55 μg of anti-PD-Ll (mut)/TGFp Trap; and Group 5 received a combination of 44.3 μg of anti-PD-Ll and 18.3 μg of anti-PD-Ll (mut)/TGFp Trap. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula: tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Antitumor efficacy is reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00182] All the treatments were well tolerated. Anti-PD-Ll /TGF Trap and the combination therapy demonstrated potent anti-tumor efficacy at both dose levels tested.
EXAMPLE 12 - Anti-PD-Ll/TGF-β Trap has better antitumor efficacy than the combination of anti-PD-Ll and TGFP Trap administered to give equivalent exposure in an intramuscular MC38 colorectal carcinoma model.
[00183] The results in Example 10 suggested that at equimolar doses the anti-PD-Ll/TGF-β Trap has better antitumor efficacy than the combination of anti-PD-Ll and TGF Trap control even though the in vivo exposure of anti-PD-Ll (mut)/TGF Trap control is about 3 times that of anti-PD-Ll /TGFP Trap (Example 5). In a follow-up study the anti-tumor efficacy of anti- PD-Ll/TGF Trap and the combination of anti-PD-Ll and anti-PD-Ll (mut)/TGFp Trap based on equal exposure was compared. Lower doses than in Example 10 were administered to avoid dosing near saturating levels.
[00184] 8- 12 week old female B6.129S2-Ighmtmlc¾7J mice (Jackson Laboratory, Bar Harbor, ME) were injected with 0.5xl06 viable MC38 tumor cells in 0.1 ml PBS intramuscularly in the right thigh. A week later, when average tumor size reached about 200 mm3, mice were sorted into groups (N=12) so that the average tumor sizes of all groups were similar. Treatment by intravenous injections was initiated (Day 0) and repeated again on Day 4. Group 1 received 133 μg of isotype antibody control; Group 2 received 164 μg of anti-PD-Ll/TGF Trap; Group 3 received 55 μg of anti-PD-Ll /TGFp Trap; Group 4 received a combination of 133 μg of anti- PD-Ll and 55 μg of anti-PD-Ll (mut)/TGFP Trap; and Group 5 received a combination of 44.3 μg of anti-PD-Ll and 1 .3 μg of anti-PD-Ll (mut)/TGFp Trap. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula: tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Anti- tumor efficacy is reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00185] All the treatments were well tolerated. Anti-PD-Ll/TGF Trap demonstrated very potent anti-tumor efficacy, achieving T/C ratios of 0.13 (pO.001) and 0.19 (pO.001) for the intermediate (164 μg, Group 2, called intermediate dose relative to the high dose of 492 μg that seemed to be saturating in Example 10) and low (55 μg, Group 3) dose groups, respectively, on Day 9. On the other hand, the combination of anti-PD-Ll and anti-PD-Ll (mut)/TGFp Trap were less efficacious, giving T/C ratios of 0.34 (p<0.001) and 0.37 (pO.001) for the intermediate (Group 4) and low (Group 5) dose groups, respectively (Fig. 12A or Table). It is especially noteworthy that when administered to give equivalent in vivo exposure of the anti- PD-Ll antibody and the TGFp Trap component, anti-PD-Ll /TGFP Trap was significantly more efficacious than the combination therapy of anti-PD-Ll and anti-PD-Ll (muf)/TGFp Trap at both dose levels (at the intermediate dose, T/C of 0.13 for anti-PD-Ll /TGFp Trap vs. 0.34 for the combination pO.0001 (Fig. 12B); at the low dose, T/C of 0.19 for anti-PD-Ll /TGFp Trap vs. 0.37 for the combination p<0.0001 (Fig. 12C)).
EXAMPLE 13 - Anti-PD-Ll(YW)/TGFp Trap has superior anti-tumor effect that is synergistic of anti-PD-Ll and TGFp Trap activities in the EMT-6 (breast carcinoma) orthotopic model. [00186] YW243.55S70 is a human antibody that recognizes both human and murine PD-Ll (US Patent Application Publication No. US2010/0203056 Al). Its variable region sequence of the heavy chain (VH) and variable region sequence of the light chain (VL) (provided as SEQ ID NO: 14 and SEQ ID NO: 13, respectively) were used to replace the corresponding variable region sequences of the anti-PD-Ll /TGFp Trap described in Example 1 to give anti-PD- Ll(YW)/TGFp Trap by standard molecular biology techniques. After construction of the DNA coding for anti-PD-Ll (YW)/TGFp Trap, the antibody fusion protein was expressed as described in Example 1. The anti-PD-Ll antibody YW243.55S70 is similarly expressed for comparison of efficacy in murine tumor models.
[00187] 8-12 week old female Jh (Igh-JtmlDhu) Balb/C mice (Taconic Farms, Hudson, NY) were inoculated with 0.25x106 viable EMT6 cells in 0.1 ml PBS into the right mammary pad. About a week later, when average tumor size reached about 50-100 mm3, mice were sorted into groups (N=10) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections was initiated (Day 0). Group 1 received 133 μ of isotype antibody control; Group 2 received 133 g of anti-PD-Ll(YW) antibody; Group 3 received 164 μg of anti-PD-Ll(mut)/TGF Trap; Group 4 received 164 μg of anti-PD-Ll(YW)/TGFp Trap; and Group 5 received a combination of 133 μg of anti-PD-Ll(YW) and 164 μg of anti-PD- LI (mut)/TGFp Trap. Treatment was repeated on Days 4 and 7. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Antitumor efficacy is reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00188] All the treatments were well tolerated. The inhibition of tumor growth by the various treatments is shown in FIG. 13 A, which showed the average tumor volumes of the mice on Day 17, the last day for which the average tumor volume of all the mice were available, i.e., before mice with tumors over 2500 mm3 were euthanized. Anti-PD-Ll(YW)/TGFp Trap demonstrated potent anti-tumor efficacy, achieving a T/C ratio of 0.25 (p<0.0001) that is slightly better than that of the combination treatment in Group 5 (T/C=0.31, pO.0001), but superior to that of the anti-PD-Ll(YW) antibody in Group 2 (T/C=0.57, p<0.0001) and the TGF Trap control in Group 3 (T/C=0.66, p<0.0001). The synergistic anti-tumor effect of the antibody fusion protein also resulted in prolonged survival of the treated mice, as shown in FIG. 13B. The anti-PD- Ll/TGFp Trap treated group had a median survival time of 65 days, which was significantly better than that of the anti-PD-Ll(YW) antibody treated group (24 days) or the TGF Trap control treated group (21 days). It also compares favorably with the median survival time of 53.5 days for the combination treatment group. Despite dosing stopped after day 7, the continual tumor growth inhibition and the prolonged survival of the anti-PD-Ll(YW)/TGFp Trap treated mice demonstrate the long-lasting immunologic anti-tumor effect resulting from dual blockade of the two major immunosuppressive pathways.
EXAMPLE 14 - Anti-PD-Ll(YW)/TGF-p Trap has superior anti-tumor effect that is synergistic of anti-PD-Ll and TGFp Trap activities in the MC38 (colorectal carcinoma) intramuscular tumor model
[00189] 8-12 week old female B6.129S2-Ighmtm,Cgn/J mice (Jackson Laboratory, Bar Harbor, ME) were injected with 0.5xl06 viable MC38 tumor cells in 0.1 ml PBS intramuscularly in the right thigh. About a week later, when average tumor size reaches about 150-200 mm3, mice were sorted into groups (N=10) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections was initiated (Day 0) and repeated again four days later (Day 4). Group 1 received 133 μg of isotype antibody control; Group 2 received 133 μg of anti- PD-Ll(YW) antibody; Group 3 received 164 μg of anti-PD-Ll (mut)/TGFp Trap; Group 4 received 164 μg of anti-PD-Ll(YW)/TGFp Trap; and Group 5 received a combination of 133 μg of anti-PD-Ll(YW) and 164 μg of anti-PD-L l(mut)/TGFp Trap. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Anti-tumor efficacy was reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00190] All the treatments were well tolerated. The inhibition of tumor growth by the various treatments is shown in FIG. 14 A, which showed the average tumor volumes of the mice on Day 10, the last day for which the average tumor volume of all the mice were available. Anti-PD- Ll (YW)/TGFP Trap demonstrated very potent anti-tumor efficacy, achieving a T/C ratio of 0.14 (p<0.0001) that is slightly better than that of the combination treatment in Group 5 (T/C=0.19, pO.0001), but superior to that of the anti-PD-Ll(YW) antibody in Group 2
(T/C=0.34, pO.0001) and the TGFp Trap control in Group 3 (T/C=0.99, p<0.0001), which had no activity in this model. The anti-tumor efficacy of anti-PD-L 1(YW)/TGFP Trap was further confirmed by tumor weight measurements taken on Day 11. By this time, the isotype control group had to be euthanized because the tumors had grown beyond 2500 mm3. Therefore, the experiment was terminated and all the groups were euthanized and the tumor weights determined. The individual tumor weights are shown in FIG. 14B. The analysis of tumor weights confirmed that anti-PD-L 1 (YW)/TGFp Trap therapy significantly inhibited MC38 tumor growth (T/C=0.13; pO.0001). The efficacy of anti-PD-L l(YW)/TGFp Trap was significantly better than that observed with anti-PD-Ll (T/C=0.37; p=0.003) or the TGFp Trap control (T/C=1.0, pO.0001). The anti-tumor efficacy of anti-PD-L l(YW)/TGFp Trap, based on the tumor weight analysis, was not statistically better than the mice treated with the combination of anti-PD-Ll and the TGFp Trap control (T/C=0.17; p=0.96). EXAMPLE 15 - Combination treatment of Anti-PD-1 and TGFp Trap do not provide any additive anti-tumor effect in an EMT-6 (breast carcinoma) orthotopic model
[00191] In this study we tested if the combination treatment of anti-PD-1 and TGFp Trap provides any additive anti-tumor effect in the EMT-6 orthotopic model. CT-01 1, also known as pidiluzumab, is a humanized anti-human PD1 antibody that was tested in the clinic for treatment of hematological malignancies (Berger et al, Clin Cancer Res. 2008; 14:3044-3051). It also recognizes murine PD-1 and has shown anti-tumor activity that synergizes with cyclophosphamide and vaccine treatment in syngeneic tumor models (Mkrtichyan et al., Eur J Immunol. 201 1; 41 :2977-86). The VH and VL sequences of CT-011 were used to produce a recombinant antibody with human IgGl /kappa constant regions by standard molecular biology techniques.
[00192] 8-12 week old female Jh (Igh-JtmlDhu) Balb/C mice (Taconic Farms, Hudson, NY) were inoculated with 0.25xl06 viable EMT6 cells in 0.1 ml PBS into the right mammary pad. About a week later, when average tumor size reached about 100 mm3, mice were sorted into groups (N=l 0) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections was initiated (Day 0). Group 1 received 364 μg of isotype antibody control; Group 2 received 164 μg of anti-PD-Ll(mut)/TGFP Trap, which served as the TGF Trap control; Group 3 received 200 μg of anti-PD-1 (CT-011); and Group 4 received a combination of 200 μg of anti-PD-1 (CT-01 1) and 164 g of anti-PD-Ll(mut)/TGFp Trap control. Treatment was repeated on Days 2, 4, 7, 9, and 11, i.e. 3 times weekly for two weeks. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Anti-tumor efficacy was reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00193] All the treatments were well tolerated. Anti-PD- 1 (CT-011 ) showed very modest anti-tumor efficacy (T/C = 0.87, p>0.05) in this model, while its combination with the TGFp Trap control had the same efficacy as the TGFp Trap control alone (FIG.15). EXAMPLE 16 - Combination treatment of Anti-PD-1 and TGFp Trap do not provide any additive anti-tumor effect in an MC38 (colorectal carcinoma) intramuscular tumor model.
[00194] In this study we tested if the combination treatment of anti-PD-1 and TGFP Trap provides any additive anti-tumor effect in the intramuscular MC38 colorectal tumor model. 8- 12 week old female B6.129S2-IghmtmlCg7J mice (Jackson Laboratory, Bar Harbor, ME) were injected with 0.5xl06 viable MC38 tumor cells in 0.1 mL PBS intramuscularly in the right thigh. About a week later, when average tumor size reaches about 190 mm3, mice are sorted into groups (N=10) so that the average tumor sizes of all groups are similar, and treatment by intravenous injections is initiated (Day 0). Group 1 received 364 μg of isotype antibody control on Days 0, 2, 4, and 7; Group 2 received 164 μg of the anti-PD-Ll(mut)/TGFp Trap control on Days 0 and 2; Group 3 received 200 μg of anti-PD-1 (CT-011) on Days 0, 2, 4, and 7; and Group 4 received a combination of 200 μg of anti-PD-1 (CT-01 1) on Days 0, 2, 4, and 7, and 164 μg of anti-PD-Ll(mut)/TGFp Trap control on Days 0 and 2. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Antitumor efficacy was reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00195] All the treatments were well tolerated. Anti-PD- 1 (CT-011 ) showed very modest anti-tumor efficacy (T/C = 0.87, p>0.05), while the anti-PD-Ll(mut)/TGFp Trap control had no efficacy in this model, as seen in previous examples. The combination of anti-PD-1 (CT-01 1) with the TGF Trap control had no efficacy at all (FIG.15). EXAMPLE 17 - Combination treatment of TGFp Trap with either anti-LAG3 or anti- TIM-3 do not provide any additive anti-tumor effect in an EMT-6 (breast carcinoma) orthotopic model
[00196] In this study we tested if the combination treatment of TGFp Trap with either anti- LAG3 or anti-TIM3 provides any additive anti-tumor effect in the orthotopic EMT-6 breast tumor model. The anti-LAG3 antibody used is a rat IgGl monoclonal anti-murine LAG3 antibody C9B7W (BioXcell, Beverly, MA), which was shown to synergize with anti-murine PD-1 treatment in syngeneic tumor models (Woo et al, Cancer Res, 2011; 72:917-27). The anti- TIM-3 antibody used is a rat IgG2a monoclonal anti-murine TIM3 antibody RMT3-23
(BioXcell, Beverly, MA), which also was shown to synergize with anti-murine PD-1 treatment in syngeneic tumor models, although its efficacy as a single agent was relatively modest (Ngiow et al, Cancer Res, 201 1 ; 71 :3540-51). [00197] 8-12 week old female Jh (Igh-JtmlDhu) Balb/C mice (Taconic Farms, Hudson, NY) were inoculated with 0.25x106 viable EMT6 cells in 0.1 ml PBS into the right mammary pad. About a week later, when average tumor size reached about 1 10 mm3, mice were sorted into groups (N=9) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections was initiated (Day 0). Group 1 received 133 μg of isotype antibody control; Group 2 received 164 μg of the anti-PD-Ll(mut)/TGFP Trap control; Group 3 received 200 μg of anti-LAG3; Group 4 received 250 μg of anti-TIM3; Group 5 received a combination of 200 μg of anti-LAG3 and 164 μg of anti-PD-Ll(mut)/TGFp Trap control; and Group 6 received a combination of 250 μg of anti-TIM3 and 164 μg of anti-PD-Ll (mut)/TGFp Trap control. Treatment was repeated on Days 2, 4, 7, 9, and I I , i.e. 3 times weekly for two weeks. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Anti-tumor efficacy was reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00198] As observed previously, the anti-PD-Ll(mut)/TGFp Trap control (Group 2) showed very modest efficacy in this EMT-6 model. Anti-T1M3 (Group 4) as a single agent showed a similarly modest efficacy as the Trap control, and in combination therapy with the Trap control (Group 6) showed no additive effect. Anti-LAG3 either as a single agent (Group 3) or in combination therapy with the Trap control (Group 5) did not show any efficacy.
EXAMPLE 18 - Combination treatment of TGFp Trap with either anti-LAG3 or anti- TIM-3 do not provide any additive anti-tumor effect in an MC38 (colorectal carcinoma) intramuscular tumor model
[00199] In this study we tested if the combination treatment of TGFP Trap with either anti- LAG3 (C9B7W) or anti-TIM3 (RMT3-23) provides any additive anti-tumor effect in the intramuscular MC38 colorectal tumor model.
[00200] 8-12 week old female B6.129S2-IghmtmlCg J mice (Jackson Laboratory, Bar Harbor, ME) were injected with 0.5xl06 viable MC38 tumor cells in 0.1 mL PBS intramuscularly in the right thigh. About a week later, when average tumor size reaches about 50 mm3, mice were sorted into groups (N=8) so that the average tumor sizes of all groups were similar, and treatment by intravenous injections is initiated (Day 0). Group 1 received 133 μg of isotype antibody control; Group 2 received 164 μg of the anti-PD-Ll(mut)/TGFp Trap control; Group 3 received 200 μg of anti-LAG3; Group 4 received 250 μg of anti-TIM3; Group 5 received a combination of 200 μg of anti-LAG3 and 164 g of anti-PD-Ll(mut)/TGFp Trap control; and Group 6 received a combination of 250 μg of anti-TIM3 and 164 μg of anti-PD-Ll(mut)/TGFp Trap control. Treatment was repeated on Days 2, 4, 7, 9, 1 1, 15 and 18. Body weights were measured twice weekly to monitor toxicity. Tumor volumes were determined at different time points using the formula tumor volume (mm3) = length x width x height x 0.5236. Any mice with tumors over 2500 mm3 were sacrificed following the institute's animal health protocol. Anti-tumor efficacy was reported as a T/C ratio, where T and C are the average tumor volumes of the group treated with antibody or fusion protein, and the group treated with the isotype control, respectively.
[00201] As observed previously, the anti-PD-Ll(mut)/TGFp Trap control (Group 2) did not have any efficacy in this MC38 model. Anti-LAG3 as a single agent (Group 3) showed a moderate efficacy, achieving a T/C of 0.66 (p<0.05). However, combination with the Trap control (Group 5) did not improve its efficacy. Anti-TIM3 either as a single agent (Group 4) or in combination therapy with the Trap control (Group 6) did not show any efficacy.
SEQUENCES SEQ ID NO: 1
Peptide sequence of the secreted anti-PD-Ll lambda light chain QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSNRPSGVSNR FSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTRVFGTGTKVTVLGQPKANPTVTLFPPSS EELQANKATLVCLISDFYPGAVTVAWKADGSPVKAGVETTKPSKQSNNKYAASSYLSLTPEQW KSHRSYSCQVTHEGSTVEKTVAPTECS
SEQ ID NO: 2
Peptide sequence of the secreted H chain of anti-PDLl
EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYIMMWVRQAPGKGLEWVSSIYPSGGITFYADT VKGRFTISRD SKNTLYLQMNSLRAEDTAVYYCARIKLGTVTTVDY GQGTLVTVSSASTKGP SVFPLAPSS STSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVV TVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKD TL ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQD WLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ KSLSLSPGK
SEQ ID NO: 3
Peptide sequence of the secreted H chain of anti-PDLl /TGFP Trap
EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYIMMWVRQAPGKGLEWVSSIYPSGGITFYADT VKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARIKLGTVTTVDYWGQGTLVTVSSAST GP SVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVV TVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKD TLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNA TKPREEQYNSTYRVVSVLTVLHQD WLNGKEYKCKVSN ALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ KSLSLSPGAGGGGSGGGGSGGGGSGGGGSGIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDV RFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASP KCIM EKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPD
SEQ ID NO: 4
DNA sequence from the translation initiation codon to the translation stop codon of the anti- PD-L1 lambda light chain (the leader sequence preceding the VL is the signal peptide from urokinase plasminogen activator) atgagggccctgctggctagactgctgctgtgcgtgctggtcgtgtccgacagcaagggcCAG TCCGCCCTGACCCAGCCTGCCTCCGTGTCTGGCTCCCCTGGCCAGTCCATCACCATCAGCTGC ACCGGCACCTCCAGCGACGTGGGCGGCTACAACTACGTGTCCTGGTATCAGCAGCACCCCGGC AAGGCCCCCAAGCTGATGATCTACGACGTGTCCAACCGGCCCTCCGGCGTGTCCAACAGATTC TCCGGCTCCAAGTCCGGCAACACCGCCTCCCTGACCATCAGCGGACTGCAGGCAGAGGACGAG GCCGACTACTACTGCTCCTCCTACACCTCCTCCAGCACCAGAGTGTTCGGCACCGGCACAAAA GTGACCGTGCTGggccagcccaaggccaacccaaccgtgacactgttccccccatcctccgag gaactgcaggccaacaaggccaccctggtctgcctgatctcagatttctatccaggcgccgtg accgtggcctggaaggctgatggctccccagtgaaggccggcgtggaaaccaccaagccctcc aagcagtccaacaacaaatacgccgcctcctcctacctgtccctgacccccgagcagtggaag tcccaccggtcctacagctgccaggtcacacacgagggctccaccgtggaaaagaccgtcgcc cccaccgagtgctcaTGA SEQ ID NO: 5
DNA sequence from the translation initiation codon to the translation stop codon (mVK SP leader: small underlined; VH: capitals; IgGlni3 with K to A mutation: small letters; (G4S)x4-G linker: bold capital letters; TGF RII: bold underlined small letters; two stop codons: bold underlined capital letters) atggaaacagacaccctgctgctgtgggtgctgctgctgtgggtgcccggctccacaggcGAG GTGCAGCTGCTGGAATCCGGCGGAGGACTGGTGCAGCCTGGCGGCTCCCTGAGACTGTCTTGC GCCGCCTCCGGCTTCACCTTCTCCAGCTACATCATGATGTGGGTGCGACAGGCCCCTGGCAAG GGCCTGGAATGGGTGTCCTCCATCTACCCCTCCGGCGGCATCACCTTCTACGCCGACACCGTG AAGGGCCGGTTCACCATCTCCCGGGACAACTCCAAGAACACCCTGTACCTGCAGATGAACTCC CTGCGGGCCGAGGACACCGCCGTGTACTACTGCGCCCGGATCAAGCTGGGCACCGTGACCACC GTGGACTACTGGGGCCAGGGCACCCTGGTGACAGTGTCCTCCgctagcaccaagggcccatcg gtcttccccctggcaccctcctccaagagcacctctgggggcacagcggccctgggctgcctg gtcaaggactacttccccgaaccggtgacggtgtcgtggaactcaggcgccctgaccagcggc gtgcacaccttcccggctgtcctacagtcctcaggactctactccctcagcagcgtggtgacc gtgccctccagcagcttgggcacccagacctacatctgcaacgtgaatcacaagcccagcaac accaaggtggacaagagagttgagcccaaatcttgtgacaaaactcacacatgcccaccgtgc ccagcacctgaactcctggggggaccgtcagtcttcctcttccccccaaaacccaaggacacc ctcatgatctcccggacccctgaggtcacatgcgtggtggtggacgtgagccacgaagaccct gaggtcaagttcaactggtacgtggacggcgtggaggtgcataatgccaagacaaagccgcgg gaggagcagtacaacagcacgtaccgtgtggtcagcgtcctcaccgtcctgcaccaggactgg ctgaatggcaaggagtacaagtgcaaggtctccaacaaagccctcccagcccccatcgagaaa accatctccaaagccaaagggcagccccgagaaccacaggtgtacaccctgcccccatcccgg gaggagatgaccaagaaccaggtcagcctgacctgcctggtcaaaggcttctatcccagcgac atcgccgtggagtgggagagcaatgggcagccggagaacaactacaagaccacgcctcccgtg ctggactccgacggctccttcttcctctatagcaagctcaccgtggacaagagcaggtggcag caggggaacgtcttctcatgctccgtgatgcatgaggctctgcacaaccactacacgcagaag agcctctccctgtccccgggtgctGGCGGCGGAGGAAGCGGAGGAGGTGGCAGCGGTGGCGGT GGCTCCGGCGGAGGTGGCTCCGGAatccctccccacgtgcagaagtccgtgaacaacgacatg atcgtgaccgacaacaacggcgccgtgaagttccctcagctgtgcaagttctgcgacgtgagg ttcagcacctgcgacaaccagaagtcctgcatgagcaactgcagcatcacaagcatctgcgag aagccccaggaggtgtgtgtggccgtgtggaggaagaacgacgaaaacatcaccctcgagacc gtgtgccatgaccccaagctgccctaccacgacttcatcctggaagacgccgcctcccccaag tgcatcatgaaggagaagaagaagcccggcgagaccttcttcatgtgcagctgcagcagcgac gagtgcaatgacaacatcatctttaqcgaggagtacaacaccagcaaccccgacTGATAA
SEQ ID NO: 6
Polypeptide sequence of the secreted lambda light chain of anti-PD-Ll(mut)/ TGFp Trap, with mutations A31G,D52E,R99Y
QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYEVSNRPSGVSNR FSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTYVFGTGTKVTVLGQP ANPTVTLFPPSS EELQANKATLVCLISDFYPGAVTVAWKADGSPVKAGVETTKPSKQSNNKYAASSYLSLTPEQW KSHRSYSCQVTHEGSTVEKTVAPTECS SEQ ID NO: 7
Polypeptide sequence of the secreted heavy chain of anti-PD-Ll(mut)/ TGFp Trap
EVQLLESGGGLVQPGGSLRLSCAASGFTFSMYMMMWVRQAPGKGLE VSSIYPSGGITFYADS VKGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCARIKLGTVTTVDY GQGTLVTVSSAS KGP SVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVV TVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKD TLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQD WLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ KSLSLSPGAGGGGSGGGGSGGGGSGGGGSGIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDV RFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASP KCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPD
SEQ ID NO: 8
Human TGF RII Isoform A Precursor Polypeptide (NCBI RefSeq Accession No:
NPJ)01020018)
MGRGLLRGLWPLHIVLWTRIASTIPPHVQKSDVEMEAQKDEIICPSCNRTAHPLRHINNDMIV TDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVC HDPKLPYHDFILEDAASP CIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYN SNPDLLLVI FQVTGISLLPPLGVAISVIIIFYCYRVNRQQKLSSTWETGKTRKLMEFSEHCAIILEDDRSDI SSTCANNINHNTELLPIELDTLVGKGRFAEVYKAKLKQNTSEQFETVAVKIFPYEEYASWKTE KDIFSDINLKHENILQFLTAEER TELGKQYWLITAFHAKGNLQEYLTRHVISWEDLRKLGSS LARGIAHLHSDHTPCGRPKMPIVHRDLKSSNILVKNDLTCCLCDFGLSLRLDPTLSVDDLANS GQVGTARYMAPEVLESRMNLENVESFKQTDVYSMALVLWEMTSRCNAVGEVKDYEPPFGSKVR EHPCVESMKDNVLRDRGRPEIPSF LNHQGIQMVCETLTECWDHDPEARLTAQCVAERFSELE HLDRLSGRSCSEEKIPEDGSLNTTK
SEQ ID NO: 9
Human TGFpPJI Isoform B Precursor Polypeptide (NCBI RefSeq Accession No: NP__003233
MGRGLLRGLWPLHIVLWTRIASTIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDN QKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIM E KKPGETFFMCSCSSDECNDNIIFSEEYNTSNPDLLLVIFQVTGISLLPPLGVAISVII IFYCY RVNRQQKLSSTWETGKTRKLMEFSEHCAIILEDDRSDISSTCANNINHNTELLPIELDTLVGK GRFAEVYKAKLKQNTSEQFETVAVKIFPYEEYASWKTEKDIFSDINLKHENILQFLTAEERKT ELGKQYWLITAFHAKGNLQEYLTRHVISWEDLRKLGSSLARGIAHLHSDHTPCGRPK PIVHR DLKSSNILVKNDLTCCLCDFGLSLRLDPTLSVDDLANSGQVGTARYMAPEVLESRMNLENVES FKQTDVYSMALVL EMTSRCNAVGEVKDYEPPFGS VREHPCVES KDNVLRDRGRPEIPSF LNHQGIQMVCETLTECWDHDPEARLTAQCVAERFSELEHLDRLSGRSCSEEKIPEDGSLNTTK SEQ ID NO: 10
A Human TGFpRII Isoform B Extracellular Domain Polypeptide
IPPHVQKSVNNDMIVTDNNGAVKFPQLC FCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVA VWRKNDENI LE VCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIF SEEYNTSNPD
SEQ ID NO: 11
(Gly4Ser)4Gly linker
GGGGSGGGGSGGGGSGGGGSG SEQ ID NO: 12
Polypeptide sequence of the secreted heavy chain variable region of anti-PD-Ll antibody MPDL3280A
EVQLVESGGGLVQPGGSLRLSCAASGFTFSDSWIHWVRQAPGKGLEWVAWISPYGGSTYY ADSVKGRF ISADTSKNTAYLQMNSLRAEDTAVYYCARRHWPGGFDYWGQGTLVTVSS
SEQ ID NO: 13
Polypeptide sequence of the secreted light chain variable region of anti-PD-Ll antibody MPDL3280A and the anti-PD-Ll antibody YW243.55S70
DIQMTQSPSSLSASVGDRVTITCRASQDVSTAVAWYQQKPGKAPKLLIYSASFLYSGVPSRFS GSGSGTDFTLTISSLQPEDFATYYCQQYLYHPATFGQGTKVEIKR
SEQ ID NO: 14
Polypeptide sequence of the secreted heavy chain variable region of anti-PD-Ll antibody YW243.55S70
EVQLVESGGGLVQPGGSLRLSCAASGFTFSDSWIHWVRQAPGKGLEWVAWISPYGGSTYYADS VKGRFTISADTSKNTAYLQMNSLRAEDTAVYYCARRHWPGGFDYWGQGTLVTVSA
INCORPORATION BY REFERENCE
[00202] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes.
EQUIVALENTS
[00203] The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the invention described herein. Various structural elements of the different embodiments and various disclosed method steps may be utilized in various combinations and permutations, and all such variants are to be considered forms of the invention. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

Claims

WHAT IS CLAIMED IS:
1. A protein comprising: a) human TGFpRII, or a fragment thereof capable of binding TGFP; and b) an antibody, or an antigen-binding fragment thereof, that binds human protein Programmed Death Ligand 1 (PD-L1).
2. A polypeptide comprising: a) at least a variable domain of a heavy chain of an antibody that binds human protein Programmed Death Ligand 1 (PD-L1); and
b) human TGFpRII, or a fragment thereof capable of binding TGFp.
3. The polypeptide of claim 2, further comprising an amino acid linker connecting the C- terminus of the variable domain to the N-terminus of the human TGFpRII or fragment thereof.
4. The polypeptide of claim 3, comprising the amino acid sequence of SEQ ID NO: 3.
5. A nucleic acid comprising a nucleotide sequence encoding a polypeptide according to any one of claims 2-4.
6. The nucleic acid of claim 5, further comprising a second nucleotide sequence encoding at least a variable domain of a light chain of an antibody which, when combined with the polypeptide, forms an antigen-binding site that binds PD-L1.
7. The nucleic acid of claim 6, wherein the second nucleotide sequence encodes the amino acid sequence of SEQ ID NO: 1 (secreted anti-PD-Ll lambda light chain).
8. A cell comprising the nucleic acid of claim 5 and a second nucleic acid encoding at least a variable domain of a light chain of an antibody which, when combined with the polypeptide, forms an antigen-binding site that binds PD-L1.
9. The cell of claim 8, wherein the second nucleic acid encodes the amino acid sequence of SEQ ID NO: 1.
10. A cell comprising the nucleic acid of claim 6 or 7.
11. A method of producing a protein comprising a) TGF RII, or a fragment thereof capable of binding TGFp, and b) an antibody, or an antigen-binding fragment thereof, that binds human protein Programmed Death Ligand 1 (PD-Ll), the method comprising maintaining a cell according to any one of claims 8-10 under conditions permitting expression of the protein.
12. The method of claim 11, further comprising harvesting the protein.
13. A protein comprising : a polypeptide according to any one of claims 2-4; and
at least a variable domain of a light chain of an antibody which, when combined with the polypeptide, forms an antigen-binding site that binds PD-Ll .
14. The protein of claim 13, comprising two polypeptides each having an amino acid sequence consisting of the amino acid sequence of SEQ ID NO: 3, and two additional polypeptides each having an amino acid sequence consisting of the amino acid sequence of SEQ ID NO: 1.
15. A protein according to claim 1, 13, or 14, for use in therapy.
16. A protein according to claim 15, wherein the therapy comprises administration of radiation.
17. A protein according to claim 15 or 16, wherein the therapy comprises administration of a chemotherapeutic .
18. A protein according to claim 1, 13, or 14, for use in promoting local depletion of TGFp at a tumor.
19. A protein according to claim 1, 13, or 14, for use in inhibiting SMAD3 phosphorylation in a cell.
20. The protein of claim 19, wherein the cell is a tumor cell.
21. A protein according to claim 1. 13, or 14, for use in treating cancer.
22. A protein according to claim 1, 13, or 14, for use in inhibiting tumor growth.
23. A protein according to claim 21 or 22, wherein the cancer or tumor is selected from the group consisting of colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, myeloma, lymphoma, leukemia, thyroid, endometrial, uterine, bladder, neuroendocrine, head and neck, liver, nasopharyngeal, testicular, small cell lung cancer, non-small cell lung cancer, melanoma, basal cell skin cancer, squamous cell skin cancer, dermatofibrosarcoma protuberans, Merkel cell carcinoma, glioblastoma, glioma, sarcoma, mesothelioma, and myelodisplastic syndromes.
24. A protein according to any one of claims 21-23, wherein the use comprises administration of radiation.
25. A protein according to any one of claims 21-24, wherein the use comprises administration of a chemotherapeutic.
26. A method of promoting local depletion of TGFp, the method comprising administering a protein according to claim 1, 13, or 14, wherein the protein binds TGFP in solution, binds PD- Ll on a cell surface, and carries the bound TGFp into the cell.
27. A method of inhibiting SMAD3 phosphorylation in a cell, the method comprising exposing the cell to a protein according to claim 1, 13, or 14.
28. A method according to claim 26 or 27, wherein the cell is a cancer cell.
29. A method of inhibiting tumor growth, the method comprising exposing the tumor to a protein according to claim 1, 13, or 14.
30. The method of claim 29, further comprising exposing the tumor to radiation.
31. A method according to claim 29 or 30, further comprising exposing the tumor to a chemotherapeutic .
32. A method of treating cancer, the method comprising administering to a cancer patient a protein according to claim 1, 13, or 14.
33. The method of claim 32, further comprising exposing the cancer patient to radiation.
34. A method according to claim 32 or 33, further comprising administering a
chemotherapeutic.
35. A method according to any one of claims 29-34, wherein the tumor or cancer is selected from the group consisting of colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, myeloma, lymphoma, leukemia, thyroid, endometrial, uterine, bladder,
neuroendocrine, head and neck, liver, nasopharyngeal, testicular, small cell lung cancer, non- small cell lung cancer, melanoma, basal cell skin cancer, squamous cell skin cancer, dermatofibrosarcoma protuberans, Merkel cell carcinoma, glioblastoma, glioma, sarcoma, mesothelioma, and myelodisplastic syndromes.
PCT/EP2015/052781 2014-02-10 2015-02-10 TARGETED TGFβ INHIBITION WO2015118175A2 (en)

Priority Applications (26)

Application Number Priority Date Filing Date Title
RU2016135062A RU2752424C2 (en) 2014-02-10 2015-02-10 Targeted tgf beta inhibition
KR1020167024679A KR102363008B1 (en) 2014-02-10 2015-02-10 TARGETED TGFβ INHIBITION
AU2015213988A AU2015213988B2 (en) 2014-02-10 2015-02-10 Targeted TGFbeta inhibition
PE2020002093A PE20210168A1 (en) 2014-02-10 2015-02-10 DIRECTED INHIBITION OF THE TRANSFORMING GROWTH FACTOR ß (TGFß)
DK15717097.8T DK3105246T3 (en) 2014-02-10 2015-02-10 Targeted TGF-beta inhibition
CA2934979A CA2934979A1 (en) 2014-02-10 2015-02-10 Targeted tgf.beta. inhibition
RS20210783A RS62038B1 (en) 2014-02-10 2015-02-10 Targeted tgf beta inhibition
ES15717097T ES2876430T3 (en) 2014-02-10 2015-02-10 Targeted inhibition of TGFß
PL15717097T PL3105246T3 (en) 2014-02-10 2015-02-10 Targeted tgf beta inhibition
MX2016010067A MX2016010067A (en) 2014-02-10 2015-02-10 TARGETED TGFß INHIBITION.
JP2016546949A JP6731346B2 (en) 2014-02-10 2015-02-10 Targeted TGFβ inhibition
NZ721364A NZ721364A (en) 2014-02-10 2015-02-10 Targeted tgfβ inhibition
CN201580007865.3A CN106103488B (en) 2014-02-10 2015-02-10 Targeted TGF-beta inhibition
SI201531639T SI3105246T1 (en) 2014-02-10 2015-02-10 Targeted tgf beta inhibition
LTEP15717097.8T LT3105246T (en) 2014-02-10 2015-02-10 Targeted tgf beta inhibition
SG11201606577YA SG11201606577YA (en) 2014-02-10 2015-02-10 TARGETED TGFβ INHIBITION
EP15717097.8A EP3105246B1 (en) 2014-02-10 2015-02-10 Targeted tgf beta inhibition
BR112016014952A BR112016014952A2 (en) 2014-02-10 2015-02-10 TARGETED TGFBETA INHIBITION
EP21165709.3A EP3889172B1 (en) 2014-02-10 2015-02-10 Targeted tgf beta inhibition
IL246968A IL246968B (en) 2014-02-10 2016-07-27 Targeted tgf-beta inhibition
PH12016501549A PH12016501549A1 (en) 2014-02-10 2016-08-04 Tergeted tgf� inhibition
AU2019246876A AU2019246876B2 (en) 2014-02-10 2019-10-11 Targeted TGFß Inhibition
IL281565A IL281565B (en) 2014-02-10 2021-03-16 Targeted tgf-beta inhibition
HRP20210977TT HRP20210977T1 (en) 2014-02-10 2021-06-18 Targeted tgf beta inhibition
CY20211100564T CY1124408T1 (en) 2014-02-10 2021-06-23 TARGETED TGFβ INHIBITION
AU2022200365A AU2022200365A1 (en) 2014-02-10 2022-01-20 Targeted TGFß Inhibition

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461938048P 2014-02-10 2014-02-10
US61/938,048 2014-02-10

Publications (2)

Publication Number Publication Date
WO2015118175A2 true WO2015118175A2 (en) 2015-08-13
WO2015118175A3 WO2015118175A3 (en) 2015-10-08

Family

ID=52988007

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2015/052781 WO2015118175A2 (en) 2014-02-10 2015-02-10 TARGETED TGFβ INHIBITION

Country Status (27)

Country Link
US (4) US9676863B2 (en)
EP (2) EP3889172B1 (en)
JP (2) JP6731346B2 (en)
KR (1) KR102363008B1 (en)
CN (2) CN113549159A (en)
AU (3) AU2015213988B2 (en)
BR (1) BR112016014952A2 (en)
CA (1) CA2934979A1 (en)
CL (1) CL2016002017A1 (en)
CY (1) CY1124408T1 (en)
DK (1) DK3105246T3 (en)
ES (1) ES2876430T3 (en)
HR (1) HRP20210977T1 (en)
HU (1) HUE054873T2 (en)
IL (2) IL246968B (en)
LT (1) LT3105246T (en)
MX (2) MX2016010067A (en)
NZ (1) NZ721364A (en)
PE (2) PE20161096A1 (en)
PH (1) PH12016501549A1 (en)
PL (1) PL3105246T3 (en)
PT (1) PT3105246T (en)
RS (1) RS62038B1 (en)
RU (2) RU2021114500A (en)
SG (2) SG10202108879SA (en)
SI (1) SI3105246T1 (en)
WO (1) WO2015118175A2 (en)

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017201036A1 (en) * 2016-05-17 2017-11-23 Genentech, Inc. Stromal gene signatures for diagnosis and use in immunotherapy
WO2018029367A1 (en) * 2016-08-12 2018-02-15 Merck Patent Gmbh Combination therapy for cancer
US9988452B2 (en) 2014-10-14 2018-06-05 Novartis Ag Antibody molecules to PD-L1 and uses thereof
WO2018208720A1 (en) * 2017-05-09 2018-11-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Combination pdl1 and tgf-beta blockade in patients with hpv+ malignancies
WO2018205985A1 (en) 2017-05-12 2018-11-15 江苏恒瑞医药股份有限公司 FUSION PROTEIN CONTAINING TGF-β RECEPTOR AND MEDICINAL USES THEREOF
EP3352800A4 (en) * 2015-09-24 2019-03-27 The University of North Carolina at Chapel Hill Methods and compositions for reducing metastases
WO2019113464A1 (en) * 2017-12-08 2019-06-13 Elstar Therapeutics, Inc. Multispecific molecules and uses thereof
WO2019175243A1 (en) 2018-03-14 2019-09-19 Merck Patent Gmbh Compounds and uses thereof to treat tumors in a subject
WO2019211489A1 (en) 2018-05-04 2019-11-07 Merck Patent Gmbh COMBINED INHIBITION OF PD-1/PD-L1, TGFβ AND DNA-PK FOR THE TREATMENT OF CANCER
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
WO2019242505A1 (en) 2018-06-17 2019-12-26 上海健信生物医药科技有限公司 Antibody targeting cldn18.2, bispecific antibody, adc, and car, and applications thereof
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
WO2020093024A2 (en) 2018-11-01 2020-05-07 Merck Patent Gmbh Methods of administering anti-tim-3 antibodies
WO2020093023A1 (en) 2018-11-01 2020-05-07 Merck Patent Gmbh Anti-tim-3 antibodies
WO2020094122A1 (en) 2018-11-09 2020-05-14 江苏恒瑞医药股份有限公司 TGF-β RECEPTOR FUSION PROTEIN PHARMACEUTICAL COMPOSITION AND USE THEREOF
JP2020514290A (en) * 2017-01-07 2020-05-21 メルク パテント ゲーエムベーハー Dosing Schedule and Dosage Form for Targeted TGF-B Inhibition
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10851157B2 (en) 2019-07-01 2020-12-01 Gensun Biopharma, Inc. Antagonists targeting the TGF-β pathway
WO2021084124A1 (en) 2019-11-01 2021-05-06 Ares Trading S.A. COMBINED INHIBITION OF PD-1, TGFβ AND ATM TOGETHER WITH RADIOTHERAPY FOR THE TREATMENT OF CANCER
WO2021089704A1 (en) 2019-11-05 2021-05-14 Merck Patent Gmbh Combined inhibition of pd-1, tgfb and tigit for the treatment of cancer
EP3838260A1 (en) 2019-12-20 2021-06-23 Ares Trading S.A. Igg:tgf rii fusion protein composition
WO2021123432A1 (en) 2019-12-20 2021-06-24 Ares Trading S.A. Igg:tgfbetarii fusion protein composition
CN113271962A (en) * 2018-08-22 2021-08-17 默克专利有限公司 Treatment of triple negative breast cancer with targeted TGF-beta inhibition
WO2021169986A1 (en) * 2020-02-25 2021-09-02 Wuxi Biologics (Shanghai) Co. Ltd. A bifunctional fusion protein and uses thereof
WO2021185337A1 (en) 2020-03-20 2021-09-23 荣昌生物制药(烟台)股份有限公司 Bispecific fusion protein and application thereof
WO2021209458A1 (en) 2020-04-14 2021-10-21 Ares Trading S.A. Combination treatment of cancer
WO2021209358A1 (en) 2020-04-14 2021-10-21 Glaxosmithkline Intellectual Property Development Limited Combination treatment for cancer based upon an icos antibody and a pd-l1 antibody tgf-beta-receptor fusion protein
WO2022017487A1 (en) 2020-07-24 2022-01-27 迈威(上海)生物科技股份有限公司 TGF-β RII MUTANT AND FUSION PROTEIN THEREOF
WO2021263169A3 (en) * 2020-06-26 2022-02-03 Sorrento Therapeutics, Inc. Oncolytic herpes simplex viruses (hsv) expressing immunomodulatory fusion proteins
WO2022063114A1 (en) * 2020-09-23 2022-03-31 海正生物制药有限公司 Tgfbr2-ecd mutant, and fusion protein comprising same and use thereof
CN114349867A (en) * 2020-10-14 2022-04-15 广东菲鹏制药股份有限公司 Fusion protein and application thereof
EP3820496A4 (en) * 2018-07-09 2022-04-20 Precigen, Inc. Fusion constructs and methods of using thereof
WO2022090527A1 (en) 2020-11-02 2022-05-05 Ares Trading S.A. Combination treatment of cancer
WO2022090529A1 (en) 2020-11-02 2022-05-05 Ares Trading S.A. Combination treatment of cancer
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US11370819B2 (en) 2014-02-10 2022-06-28 Merck Patent Gmbh Targeted TGFβ inhibition
RU2776204C1 (en) * 2017-05-12 2022-07-14 Цзянсу Хэнжуй Медицин Ко., Лтд. Fused protein containing tgf-beta receptor, and its pharmaceutical use
WO2022233718A2 (en) 2021-05-03 2022-11-10 Merck Patent Gmbh Her2 targeting fc antigen binding fragment-drug conjugates
WO2022248380A1 (en) 2021-05-25 2022-12-01 Merck Patent Gmbh Egfr targeting fc antigen binding fragment-drug conjugates
WO2022258622A1 (en) 2021-06-07 2022-12-15 Ares Trading S.A. Combination treatment of cancer
US11583593B2 (en) 2016-01-14 2023-02-21 Synthis Therapeutics, Inc. Antibody-ALK5 inhibitor conjugates and their uses
EP4073124A4 (en) * 2019-12-11 2024-01-24 Wuxi Biologics Ireland Ltd Bi-functional antibody against pd-l1 and tgfbeta
EP4107190A4 (en) * 2020-02-18 2024-03-20 Nanjing Genscript Biotech Co Ltd Fusion proteins and uses thereof

Families Citing this family (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3036262A4 (en) 2013-08-22 2017-03-01 Acceleron Pharma Inc. Tgf-beta receptor type ii variants and uses thereof
CA2978942A1 (en) 2015-03-13 2016-09-22 Cytomx Therapeutics, Inc. Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof
CN108136001B (en) * 2015-04-03 2022-07-29 佐马技术有限公司 Treatment of cancer using TGF-beta inhibitors and PD-1 inhibitors
DK3303394T3 (en) 2015-05-29 2020-07-06 Agenus Inc ANTI-CTLA-4 ANTIBODIES AND PROCEDURES FOR USE THEREOF
KR20180035884A (en) 2015-08-04 2018-04-06 악셀레론 파마 인코포레이티드 Methods for treating myeloproliferative disorders
WO2017020291A1 (en) * 2015-08-06 2017-02-09 Wuxi Biologics (Shanghai) Co. Ltd. Novel anti-pd-l1 antibodies
US10894823B2 (en) 2016-03-24 2021-01-19 Gensun Biopharma Inc. Trispecific inhibitors for cancer treatment
SG11201810023QA (en) 2016-05-27 2018-12-28 Agenus Inc Anti-tim-3 antibodies and methods of use thereof
KR102617453B1 (en) 2016-09-19 2023-12-26 현대모비스 주식회사 Apparatus and method for recognizing supplementarty traffic signs
US10906957B2 (en) 2016-09-27 2021-02-02 Epicentrx, Inc. Immunomodulatory fusion proteins
CN117586403A (en) 2016-10-11 2024-02-23 艾吉纳斯公司 anti-LAG-3 antibodies and methods of use thereof
EA201991383A1 (en) 2016-12-07 2019-12-30 Эйдженус Инк. ANTIBODIES AGAINST CTLA-4 AND WAYS OF THEIR APPLICATION
MA50948A (en) 2016-12-07 2020-10-14 Agenus Inc ANTIBODIES AND METHODS OF USING THE SAME
US10537637B2 (en) 2017-01-05 2020-01-21 Gensun Biopharma Inc. Checkpoint regulator antagonists
EP3568474A1 (en) * 2017-01-10 2019-11-20 Intrexon Corporation Modulating expression of polypeptides via new gene switch expression systems
GB201700567D0 (en) * 2017-01-12 2017-03-01 Genagon Therapeutics Ab Therapeutic agents
CN110198954A (en) 2017-01-13 2019-09-03 艾吉纳斯公司 T cell receptor and its application method in conjunction with NY-ESO-1
MX2019010392A (en) 2017-03-02 2019-12-02 Nat Res Council Canada Tgf-b-receptor ectodomain fusion molecules and uses thereof.
US20200048352A1 (en) * 2017-03-30 2020-02-13 Merck Patent Gmbh Combination of an anti-pd-l1 antibody and a dna-pk inhibitor for the treatment of cancer
EP3609921A2 (en) 2017-04-13 2020-02-19 Agenus Inc. Anti-cd137 antibodies and methods of use thereof
EP3618863B1 (en) 2017-05-01 2023-07-26 Agenus Inc. Anti-tigit antibodies and methods of use thereof
KR20240023201A (en) * 2017-05-04 2024-02-20 악셀레론 파마 인코포레이티드 Tgf-beta receptor type ii fusion proteins and uses thereof
EP3630836A1 (en) 2017-05-31 2020-04-08 Elstar Therapeutics, Inc. Multispecific molecules that bind to myeloproliferative leukemia (mpl) protein and uses thereof
WO2018222949A1 (en) 2017-06-01 2018-12-06 Cytomx Therapeutics, Inc. Activatable anti-pdl1 antibodies, and methods of use thereof
JP7387585B2 (en) 2017-09-04 2023-11-28 アジェナス インコーポレイテッド T-cell receptor that binds mixed lineage leukemia (MLL)-specific phosphopeptide and methods of use thereof
MX2020003672A (en) 2017-09-27 2020-08-03 Epicentrx Inc Immunomodulatory fusion proteins.
JP2021502066A (en) * 2017-11-06 2021-01-28 ジェネンテック, インコーポレイテッド Cancer diagnosis and therapy
WO2019173692A2 (en) 2018-03-09 2019-09-12 Agenus Inc. Anti-cd73 antibodies and methods of use thereof
WO2019178362A1 (en) 2018-03-14 2019-09-19 Elstar Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
WO2019178364A2 (en) 2018-03-14 2019-09-19 Elstar Therapeutics, Inc. Multifunctional molecules and uses thereof
AU2019261451A1 (en) 2018-04-26 2020-12-03 Agenus Inc. Heat shock protein-binding peptide compositions and methods of use thereof
US20210380663A1 (en) * 2018-05-03 2021-12-09 Acceleron Pharma Inc. NOVEL BINDERS OF TGFBeta-SUPERFAMILY LIGANDS AND USES THEREOF
US20220372107A1 (en) * 2018-05-03 2022-11-24 Acceleron Pharma Inc. MULTISPECIFIC BINDERS OF TGFBeta-SUPERFAMILY LIGANDS AND USES THEREOF
CN112118858A (en) * 2018-05-15 2020-12-22 默克专利有限公司 Targeted TGF-BETA inhibited dosing regimens for cancer treatment in untreated subjects
CA3100544A1 (en) 2018-05-17 2019-11-21 Bolt Biotherapeutics, Inc. Immunoconjugates
MX2020013535A (en) * 2018-06-13 2021-02-26 Merck Patent Gmbh Treatment of stage iii nsclc and mitigation of pathological conditions associated with the treatment.
AU2019288765A1 (en) * 2018-06-22 2021-01-07 Merck Patent Gmbh Dosing regimens for targeted TGF-β inhibition for use in treating biliary tract cancer
BR112020026862A2 (en) 2018-06-29 2021-04-20 Gensun Biopharma, Inc. antitumor antagonists of immunological checkpoint regulators
CN112638374A (en) * 2018-07-02 2021-04-09 默克专利有限公司 Combination therapy with targeted TGF-beta inhibition for the treatment of advanced non-small cell lung cancer
EP3818083A2 (en) 2018-07-03 2021-05-12 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
AU2019346645A1 (en) 2018-09-27 2021-04-29 Marengo Therapeutics, Inc. CSF1R/CCR2 multispecific antibodies
SG11202105975SA (en) * 2018-12-06 2021-07-29 Guangdong Tcrcure Biopharma Technology Co Ltd Combinational tcr-t cell therapy targeting tumor antigens, tgf-beta, and immune checkpoints
WO2020172571A1 (en) 2019-02-21 2020-08-27 Elstar Therapeutics, Inc. Multifunctional molecules that bind to t cell related cancer cells and uses thereof
GB2599227B (en) 2019-02-21 2024-05-01 Marengo Therapeutics Inc Multifunctional molecules that bind to T cells and uses thereof to treat autoimmune disorders
GB2597851A (en) 2019-02-21 2022-02-09 Marengo Therapeutics Inc Antibody molecules that bind to NKP30 and uses thereof
WO2020190734A1 (en) 2019-03-15 2020-09-24 Bolt Biotherapeutics, Inc. Immunoconjugates targeting pd-l1
CN114206926B (en) * 2019-06-10 2022-06-17 山东博安生物技术股份有限公司 anti-PDL 1 and TGF beta bifunctional fusion protein and application thereof
CA3142887A1 (en) 2019-06-13 2020-12-17 Bolt Biotherapeutics, Inc. Aminobenzazepine compounds, immunoconjugates, and uses thereof
CN110218260A (en) * 2019-06-24 2019-09-10 王跃驹 Application of the plant as host in expression M7842
US20210107969A1 (en) * 2019-07-09 2021-04-15 The Johns Hopkins University Molecules, compositions and methods for treatment of cancer
CA3149810A1 (en) * 2019-08-15 2021-02-18 Nantbio, Inc. Tgf-beta trap
BR112022003740A2 (en) 2019-08-30 2022-05-31 Agenus Inc Anti-cd96 antibodies and methods of using them
CN114072428B (en) 2019-08-30 2024-02-13 正大天晴药业集团南京顺欣制药有限公司 Fusion protein targeting PD-L1 and TGF-beta and application thereof
WO2021046112A1 (en) 2019-09-03 2021-03-11 Bolt Biotherapeutics, Inc. Aminoquinoline compounds, immunoconjugates, and uses thereof
WO2021046347A1 (en) 2019-09-04 2021-03-11 Bolt Biotherapeutics, Inc. Immunoconjugate synthesis method
CN112442132A (en) * 2019-09-05 2021-03-05 复旦大学 Recombinant difunctional fusion protein targeting tumor and application thereof
JP2022550420A (en) * 2019-09-30 2022-12-01 ハーバー・バイオメド・(シャンハイ)・カンパニー・リミテッド ANTI-PD-L1 ANTIGEN-BINDING PROTEIN AND APPLICATION THEREOF
BR112022006001A2 (en) 2019-09-30 2022-07-12 Bolt Biotherapeutics Inc IMMUNOCONJUGATE, 8-AMIDO-2-AMINOBENZAZEPINE-BINDING COMPOUND, 5-AMINO-PYRAZOAZEPINE-BINDING COMPOUND, AMINOQUINOLINE-BINDING COMPOUND, USE OF AN IMMUNOCONJUGATE AND METHODS TO TREAT CANCER AND TO PREPARE AN IMMUNOCONJUGATE
JP2022553046A (en) * 2019-10-21 2022-12-21 ナンチン リーズ バイオラブス カンパニー,リミティド Recombinant proteins targeting PD-1 and TGFβ
KR20220088901A (en) 2019-10-25 2022-06-28 볼트 바이오테라퓨틱스 인코퍼레이티드 Thienoazepine immunoconjugates, and uses thereof
WO2021093760A1 (en) * 2019-11-12 2021-05-20 江苏恒瑞医药股份有限公司 TGF-β RECEPTOR-CONTAINING FUSION PROTEIN AND PHARMACEUTICAL USE THEREOF
CN114746451A (en) * 2019-11-27 2022-07-12 上海岸迈生物科技有限公司 TGF-beta/PD-L1 bispecific binding proteins
CN113004414B (en) * 2019-12-20 2022-09-06 广东菲鹏制药股份有限公司 Bifunctional antibody for resisting PD1 and TGF beta, preparation method thereof and pharmaceutical composition containing same
CN113121699A (en) * 2019-12-30 2021-07-16 国典(北京)医药科技有限公司 PTTRAP1 bispecific fusion protein and application thereof
WO2021138407A2 (en) 2020-01-03 2021-07-08 Marengo Therapeutics, Inc. Multifunctional molecules that bind to cd33 and uses thereof
EP4093510A1 (en) 2020-01-21 2022-11-30 Bolt Biotherapeutics, Inc. Anti-pd-l1 antibodies
WO2021150702A1 (en) 2020-01-21 2021-07-29 Bolt Biotherapeutics, Inc. Anti-pd-l1 antibodies
EP4139363A1 (en) 2020-04-24 2023-03-01 Marengo Therapeutics, Inc. Multifunctional molecules that bind to t cell related cancer cells and uses thereof
SG11202112792WA (en) 2020-04-28 2021-12-30 Univ Rockefeller Neutralizing anti-sars-cov-2 antibodies and methods of use thereof
KR20230005907A (en) 2020-05-01 2023-01-10 볼트 바이오테라퓨틱스 인코퍼레이티드 Anti-DECTIN-2 Antibodies
US20230293716A1 (en) 2020-05-08 2023-09-21 Bolt Biotherapeutics, Inc. Elastase-substrate, peptide linker immunoconjugates, and uses thereof
EP4188950A1 (en) * 2020-07-28 2023-06-07 Lepu Biopharma Co., Ltd. Bifunctional molecules targeting pd-l1 and tgf-beta
US11028174B1 (en) 2020-07-28 2021-06-08 Lepu Biopharma Co., Ltd. Bifunctional molecules targeting PD-L1 and TGF-β
WO2022042537A1 (en) * 2020-08-24 2022-03-03 江苏恒瑞医药股份有限公司 APPLICATION OF COMBINATION OF FUSION PROTEIN OF TGF-β RECEPTOR AND MULTI-TARGETED TYROSINE KINASE INHIBITOR IN PREPARING ANTI-TUMOR DRUG
WO2022046920A2 (en) 2020-08-26 2022-03-03 Marengo Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
GB2616128A (en) 2020-08-26 2023-08-30 Marengo Therapeutics Inc Antibody molecules that bind to NKp30 and uses thereof
CN116761818A (en) 2020-08-26 2023-09-15 马伦戈治疗公司 Method for detecting TRBC1 or TRBC2
WO2022042715A1 (en) * 2020-08-31 2022-03-03 杭州九源基因工程有限公司 BIFUNCTIONAL FUSION PROTEIN TARGETING PD-L1 AND TGFβ, PREPARATION METHOD THEREFOR, AND APPLICATION THEREOF
WO2022057061A1 (en) * 2020-09-16 2022-03-24 Suzhou Neologics Bioscience Co., Ltd. Pd-l1 antibodies, fusion proteins, and uses thereof
CN117157314A (en) * 2020-09-16 2023-12-01 苏州逻晟生物医药有限公司 PD-L1 antibodies, fusion proteins and uses thereof
CN116323657B (en) * 2020-09-24 2023-12-08 上海齐鲁制药研究中心有限公司 Bifunctional molecule for simultaneously targeting PD-L1 and TGF beta and medical application thereof
CN114573701A (en) * 2020-12-02 2022-06-03 上海华奥泰生物药业股份有限公司 anti-PD-L1/TGF-beta bifunctional antibody and application thereof
CN116723866A (en) 2020-12-11 2023-09-08 博尔特生物治疗药物有限公司 anti-PD-L1 immunoconjugates and uses thereof
US20220195066A1 (en) 2020-12-11 2022-06-23 Bolt Biotherapeutics, Inc. Anti-cea immunoconjugates, and uses thereof
JP2024501453A (en) 2020-12-11 2024-01-12 ボルト バイオセラピューティクス、インコーポレーテッド Anti-CEA immunoconjugates and their uses
CN116917334A (en) 2021-02-22 2023-10-20 浙江道尔生物科技有限公司 Multi-domain fusion protein with anticancer activity
AU2022230795A1 (en) 2021-03-02 2023-09-21 Glaxosmithkline Intellectual Property Development Limited Substituted pyridines as dnmt1 inhibitors
WO2022204536A1 (en) 2021-03-26 2022-09-29 Bolt Biotherapeutics, Inc. 2-amino-4-carboxamide-benzazepine immunoconjugates, and uses thereof
IL306114A (en) 2021-03-26 2023-11-01 Bolt Biotherapeutics Inc 2-amino-4-carboxamide-benzazepine immunoconjugates, and uses thereof
WO2023027561A1 (en) * 2021-08-27 2023-03-02 세라노틱스(주) Bispecific molecule specifically binding to b7-h3 and tgfβ and uses thereof
US11945856B2 (en) 2022-01-28 2024-04-02 35Pharma Inc. Activin receptor type IIB variants and uses thereof
CN114990129B (en) * 2022-05-11 2023-02-03 北京贝来生物科技有限公司 Preparation and application of mesenchymal stem cells expressing alpha PDL1: fc fusion protein

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001058957A2 (en) 2000-02-11 2001-08-16 Lexigen Pharmaceuticals Corp. Enhancing the circulating half-life of antibody-based fusion proteins
US20030044423A1 (en) 2001-03-07 2003-03-06 Lexigen Pharmaceuticals Corp. Expression technology for proteins containing a hybrid isotype antibody moiety
US20030166877A1 (en) 2001-03-30 2003-09-04 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
US20100203056A1 (en) 2008-12-09 2010-08-12 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5785682A (en) 1995-03-22 1998-07-28 Abbott Laboratories Pre-filled syringe drug delivery system
AU737106B2 (en) * 1997-04-18 2001-08-09 Biogen, Inc. Type II TGF-beta receptor/immunoglobulin constant region fusion proteins
JP2005112812A (en) * 2003-10-09 2005-04-28 Zeria Pharmaceut Co Ltd Antibody specific to phosphorylation site and screening method using the same
MXPA06008157A (en) 2003-12-24 2007-09-07 Johnson & Johnson Treatment of malignant gliomas with tgf-beta inhibitors.
US7670595B2 (en) 2004-06-28 2010-03-02 Merck Patent Gmbh Fc-interferon-beta fusion proteins
US7795389B2 (en) 2004-09-28 2010-09-14 The Board Of Regents Of The University Of Texas System Antagonizing TGF-beta activity with various ectodomains TGF-beta receptors used in combination or as fusion proteins
CN101541832B (en) * 2006-09-07 2014-11-12 克鲁塞尔荷兰公司 Human binding molecules capable of neutralizing influenza virus H5N1 and uses thereof
WO2008157367A1 (en) 2007-06-15 2008-12-24 Genzyme Corporation Fusion proteins containing two tgf-beta binding domains of tgf-beta type ii receptor
US20140127200A1 (en) 2008-01-03 2014-05-08 The Scripps Research Institute Multispecific Antibody Targeting and Multivalency Through Modular Recognition Domains
WO2009152610A1 (en) 2008-06-20 2009-12-23 The Royal Institution For The Advancement Of Learning/Mcgill University Interleukin-2/soluble tgf-beta type ii receptor b conjugates and methods and uses thereof
MX2011000039A (en) 2008-07-02 2011-05-31 Emergent Product Dev Seattle Tgf-b antagonist multi-target binding proteins.
FR2944448B1 (en) * 2008-12-23 2012-01-13 Adocia STABLE PHARMACEUTICAL COMPOSITION COMPRISING AT LEAST ONE MONODONAL ANTIBODY AND AT LEAST ONE AMPHIPHILIC POLYSACHARIDE COMPRISING SUBSTITUENTS DERIVED FROM HYDROFOB ALCOHOLS OR HYDROPHOBIC AMINES.
JP5873003B2 (en) 2009-03-18 2016-03-01 アーメイゲン・テクノロジーズ・インコーポレイテッドArmagen Technologies, Inc. Compositions and methods for blood brain barrier delivery of IgG decoy receptor fusion proteins
CN102753580A (en) 2009-10-28 2012-10-24 亚培生物医疗股份有限公司 Anti-egfr antibodies and their uses
KR101573109B1 (en) 2009-11-24 2015-12-01 메디뮨 리미티드 Targeted binding agents against b7-h1
SI2519543T1 (en) * 2009-12-29 2016-08-31 Emergent Product Development Seattle, Llc Heterodimer binding proteins and uses thereof
US20130129723A1 (en) * 2009-12-29 2013-05-23 Emergent Product Development Seattle, Llc Heterodimer Binding Proteins and Uses Thereof
EP3064509A3 (en) 2010-02-23 2016-11-02 F. Hoffmann-La Roche AG Anti-angiogenesis therapy for the treatment of ovarian cancer
IL300733A (en) 2010-03-05 2023-04-01 Univ Johns Hopkins Compositions and methods for targeted immunomodulatory antibodies and fusion proteins
MY193562A (en) * 2011-08-01 2022-10-19 Genentech Inc Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
EP3489254B9 (en) 2012-04-30 2022-12-21 Biocon Limited Targeted/immunomodulatory fusion proteins and methods for making same
MY186099A (en) 2012-05-31 2021-06-22 Genentech Inc Methods of treating cancer using pd-l1 axis binding antagonists and vegf antagonists
WO2014193898A1 (en) 2013-05-31 2014-12-04 Merck Sharp & Dohme Corp. Combination therapies for cancer
AU2014364606A1 (en) 2013-12-17 2016-07-07 Genentech, Inc. Combination therapy comprising OX40 binding agonists and PD-1 axis binding antagonists
RU2021114500A (en) 2014-02-10 2021-06-07 Мерк Патент Гмбх TARGETED TGFβ INHIBITION
GB201403875D0 (en) 2014-03-05 2014-04-16 Cantargia Ab Novel antibodies and uses thereof
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
CN107108748A (en) 2014-08-15 2017-08-29 默克专利有限公司 SIRP alpha immunization Immunoglobulin fusion albumen

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001058957A2 (en) 2000-02-11 2001-08-16 Lexigen Pharmaceuticals Corp. Enhancing the circulating half-life of antibody-based fusion proteins
US20030044423A1 (en) 2001-03-07 2003-03-06 Lexigen Pharmaceuticals Corp. Expression technology for proteins containing a hybrid isotype antibody moiety
US20030166877A1 (en) 2001-03-30 2003-09-04 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US20100203056A1 (en) 2008-12-09 2010-08-12 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof

Non-Patent Citations (34)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
ANGAL S ET AL., MOL. IMMUNOL., vol. 30, 1993, pages 105 - 8
BARTRAM, CIRCULATION, vol. 103, 2001, pages 2745 - 52
BERGER ET AL., CLIN CANCER RES., vol. 14, 2008, pages 3044 - 3051
BIERIE ET AL., NAT REV CANCER, vol. 6, 2006, pages 506 - 20
BUJAK ET AL., CARDIOVASC RES., vol. 74, 2007, pages 184 - 95
CALONE ET AL., EXP ONCOL., vol. 34, 2012, pages 9 - 16
CONNOLLY ET AL., INT J BIOL SCI., vol. 8, 2012, pages 964 - 78
FLAVELL ET AL., NAT REV IMMUNOL., vol. 10, 2010, pages 554 - 67
GILLIES ET AL., CLIN CANCER RES., vol. 8, 2002, pages 210 - 6
HUME, BLOOD, vol. 119, 2012, pages 1810 - 20
KHAW ET AL., OPHTHALMOLOGY, vol. 114, 2007, pages 1822 - 1830
L6PEZ-CASILLAS ET AL., J CELL BIOL., vol. 124, 1994, pages 557 - 68
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LIN, J BIOL CHEM., vol. 270, 1995, pages 2747 - 54
MENDOZA ET AL., BIOCHEMISTRY, vol. 48, 2009, pages 11755 - 65
MKRTICHYAN ET AL., EUR J IMMUNOL., vol. 41, 2011, pages 2977 - 86
MONTERO-JULIAN ET AL., BLOOD, vol. 85, 1995, pages 917 - 24
MORRIS ET AL., J CLIN ONCOL, vol. 26, 2008, pages 9028
NGIOW ET AL., CANCER RES, vol. 71, 2011, pages 3540 - 51
PARDOLL, D.M., NAT IMMUNOL., vol. 13, 2012, pages 1129 - 32
PARDOLL, NAT REV CANCER, vol. 12, 2012, pages 253 - 264
ROBERTS ET AL., J CLIN INVEST., vol. 90, 1992, pages 2056 - 62
SUZUKI ET AL., CLIN CANCER RES., vol. 10, 2004, pages 5907 - 18
VERONA ET AL., PROTEIN ENG DES SEL., vol. 21, 2008, pages 463 - 73
VILCHIS-LANDEROS ET AL., BIOCHEM J, vol. 355, 2001, pages 215
WHERRY, E.J., NAT IMMUNOL., vol. 12, 2011, pages 492 - 9
WON ET AL., CANCER RES., vol. 59, 1999, pages 1273 - 7
WOO ET AL., CANCER RES, vol. 72, 2011, pages 917 - 27
YAMAGISHI, ANAT REC., vol. 295, 2012, pages 257 - 67
YANG ET AL., CANCER RES., vol. 68, 2008, pages 9107 - 11
YANG ET AL., TRENDS IMMUNOL, vol. 31, 2010, pages 220 - 227
YANG ET AL., TRENDS IMMUNOL., vol. 31, 2010, pages 220 - 227
ZHONG ET AL., CLIN CANCER RES., vol. 16, 2010, pages 1191 - 205

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11708412B2 (en) 2013-09-26 2023-07-25 Novartis Ag Methods for treating hematologic cancers
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US11827704B2 (en) 2014-01-24 2023-11-28 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11155620B2 (en) 2014-01-31 2021-10-26 Novartis Ag Method of detecting TIM-3 using antibody molecules to TIM-3
US10981990B2 (en) 2014-01-31 2021-04-20 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11370819B2 (en) 2014-02-10 2022-06-28 Merck Patent Gmbh Targeted TGFβ inhibition
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US9988452B2 (en) 2014-10-14 2018-06-05 Novartis Ag Antibody molecules to PD-L1 and uses thereof
US10851165B2 (en) 2014-10-14 2020-12-01 Novartis Ag Antibody molecules to PD-L1 and methods of treating cancer
US11219694B2 (en) 2015-09-24 2022-01-11 The University Of North Carolina At Chapel Hill Methods and compositions for reducing metastases
EP3352800A4 (en) * 2015-09-24 2019-03-27 The University of North Carolina at Chapel Hill Methods and compositions for reducing metastases
US11583593B2 (en) 2016-01-14 2023-02-21 Synthis Therapeutics, Inc. Antibody-ALK5 inhibitor conjugates and their uses
CN109312408B (en) * 2016-05-17 2022-12-23 豪夫迈·罗氏有限公司 Stromal gene signatures for diagnosis and for use in immunotherapy
WO2017201036A1 (en) * 2016-05-17 2017-11-23 Genentech, Inc. Stromal gene signatures for diagnosis and use in immunotherapy
JP2019523218A (en) * 2016-05-17 2019-08-22 ジェネンテック, インコーポレイテッド Stromal gene signature for diagnosis and use in immunotherapy
JP7359547B2 (en) 2016-05-17 2023-10-11 ジェネンテック, インコーポレイテッド Stromal gene signatures for diagnosis and use in immunotherapy
CN109312408A (en) * 2016-05-17 2019-02-05 豪夫迈·罗氏有限公司 For diagnosing and signing for matrix gene used in immunotherapy
JP2019527706A (en) * 2016-08-12 2019-10-03 メルク パテント ゲーエムベーハー Cancer combination therapy
CN109641963A (en) * 2016-08-12 2019-04-16 默克专利有限公司 The combination therapy of cancer
WO2018029367A1 (en) * 2016-08-12 2018-02-15 Merck Patent Gmbh Combination therapy for cancer
JP2020514290A (en) * 2017-01-07 2020-05-21 メルク パテント ゲーエムベーハー Dosing Schedule and Dosage Form for Targeted TGF-B Inhibition
US11440963B2 (en) 2017-05-09 2022-09-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Combination PDL1 and TGF-beta blockade in patients with HPV+ malignancies
WO2018208720A1 (en) * 2017-05-09 2018-11-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Combination pdl1 and tgf-beta blockade in patients with hpv+ malignancies
KR102629503B1 (en) * 2017-05-12 2024-01-24 지앙수 헨그루이 파마슈티컬스 컴퍼니 리미티드 Fusion protein containing TGF-β receptor and pharmaceutical use thereof
EP3623389A4 (en) * 2017-05-12 2021-01-20 Jiangsu Hengrui Medicine Co., Ltd. Fusion protein containing tgf- receptor and medicinal uses thereof
US11274142B2 (en) 2017-05-12 2022-03-15 Jiangsu Hengrui Medicine Co., Ltd. Fusion protein containing TGF-β receptor and medicinal uses thereof
WO2018205985A1 (en) 2017-05-12 2018-11-15 江苏恒瑞医药股份有限公司 FUSION PROTEIN CONTAINING TGF-β RECEPTOR AND MEDICINAL USES THEREOF
RU2776204C1 (en) * 2017-05-12 2022-07-14 Цзянсу Хэнжуй Медицин Ко., Лтд. Fused protein containing tgf-beta receptor, and its pharmaceutical use
KR20200004801A (en) * 2017-05-12 2020-01-14 지앙수 헨그루이 메디슨 컴퍼니 리미티드 Fusion Proteins Containing TVF-β Receptors and Their Pharmaceutical Uses
WO2019113464A1 (en) * 2017-12-08 2019-06-13 Elstar Therapeutics, Inc. Multispecific molecules and uses thereof
WO2019175243A1 (en) 2018-03-14 2019-09-19 Merck Patent Gmbh Compounds and uses thereof to treat tumors in a subject
WO2019211489A1 (en) 2018-05-04 2019-11-07 Merck Patent Gmbh COMBINED INHIBITION OF PD-1/PD-L1, TGFβ AND DNA-PK FOR THE TREATMENT OF CANCER
WO2019242505A1 (en) 2018-06-17 2019-12-26 上海健信生物医药科技有限公司 Antibody targeting cldn18.2, bispecific antibody, adc, and car, and applications thereof
US11912763B2 (en) 2018-06-17 2024-02-27 L & L Biopharma Co., Ltd. Antibody targeting CLDN18.2, bispecific antibody, ADC, and CAR, and applications thereof
US11535669B2 (en) 2018-07-09 2022-12-27 Precigen, Inc. Fusion constructs and methods of using thereof
EP3820496A4 (en) * 2018-07-09 2022-04-20 Precigen, Inc. Fusion constructs and methods of using thereof
EP3840776A4 (en) * 2018-08-22 2022-09-28 Merck Patent GmbH Treatment of triple negative breast cancer with targeted tgf-b inhibition
CN113271962A (en) * 2018-08-22 2021-08-17 默克专利有限公司 Treatment of triple negative breast cancer with targeted TGF-beta inhibition
WO2020093024A3 (en) * 2018-11-01 2020-06-04 Merck Patent Gmbh Methods of administering anti-tim-3 antibodies
WO2020093024A2 (en) 2018-11-01 2020-05-07 Merck Patent Gmbh Methods of administering anti-tim-3 antibodies
WO2020093023A1 (en) 2018-11-01 2020-05-07 Merck Patent Gmbh Anti-tim-3 antibodies
EP3878461A4 (en) * 2018-11-09 2022-08-17 Jiangsu Hengrui Medicine Co., Ltd. TGF-ß RECEPTOR FUSION PROTEIN PHARMACEUTICAL COMPOSITION AND USE THEREOF
WO2020094122A1 (en) 2018-11-09 2020-05-14 江苏恒瑞医药股份有限公司 TGF-β RECEPTOR FUSION PROTEIN PHARMACEUTICAL COMPOSITION AND USE THEREOF
WO2020263312A1 (en) * 2019-06-28 2020-12-30 Gensun Biopharma, Inc. ANTITUMOR ANTAGONIST CONSISTING OF A MUTATED TGFβ1 - RII EXTRACELLULAR DOMAIN AND AN IMMUNOGLOBULIN SCAFFOLD
JP2022540571A (en) * 2019-06-28 2022-09-16 ゲンスン バイオファーマ、インコーポレーテッド An anti-tumor antagonist composed of a mutated TGFβ1-RII extracellular domain and an immunoglobulin scaffold
US10851157B2 (en) 2019-07-01 2020-12-01 Gensun Biopharma, Inc. Antagonists targeting the TGF-β pathway
WO2021084124A1 (en) 2019-11-01 2021-05-06 Ares Trading S.A. COMBINED INHIBITION OF PD-1, TGFβ AND ATM TOGETHER WITH RADIOTHERAPY FOR THE TREATMENT OF CANCER
WO2021089704A1 (en) 2019-11-05 2021-05-14 Merck Patent Gmbh Combined inhibition of pd-1, tgfb and tigit for the treatment of cancer
EP4073124A4 (en) * 2019-12-11 2024-01-24 Wuxi Biologics Ireland Ltd Bi-functional antibody against pd-l1 and tgfbeta
EP3838260A1 (en) 2019-12-20 2021-06-23 Ares Trading S.A. Igg:tgf rii fusion protein composition
WO2021123432A1 (en) 2019-12-20 2021-06-24 Ares Trading S.A. Igg:tgfbetarii fusion protein composition
EP4107190A4 (en) * 2020-02-18 2024-03-20 Nanjing Genscript Biotech Co Ltd Fusion proteins and uses thereof
EP4093779A4 (en) * 2020-02-25 2024-03-06 Wuxi Biologics Ireland Ltd A bifunctional fusion protein and uses thereof
WO2021169986A1 (en) * 2020-02-25 2021-09-02 Wuxi Biologics (Shanghai) Co. Ltd. A bifunctional fusion protein and uses thereof
WO2021185337A1 (en) 2020-03-20 2021-09-23 荣昌生物制药(烟台)股份有限公司 Bispecific fusion protein and application thereof
WO2021209358A1 (en) 2020-04-14 2021-10-21 Glaxosmithkline Intellectual Property Development Limited Combination treatment for cancer based upon an icos antibody and a pd-l1 antibody tgf-beta-receptor fusion protein
WO2021209458A1 (en) 2020-04-14 2021-10-21 Ares Trading S.A. Combination treatment of cancer
WO2021263169A3 (en) * 2020-06-26 2022-02-03 Sorrento Therapeutics, Inc. Oncolytic herpes simplex viruses (hsv) expressing immunomodulatory fusion proteins
WO2022017487A1 (en) 2020-07-24 2022-01-27 迈威(上海)生物科技股份有限公司 TGF-β RII MUTANT AND FUSION PROTEIN THEREOF
WO2022063114A1 (en) * 2020-09-23 2022-03-31 海正生物制药有限公司 Tgfbr2-ecd mutant, and fusion protein comprising same and use thereof
CN114349867A (en) * 2020-10-14 2022-04-15 广东菲鹏制药股份有限公司 Fusion protein and application thereof
WO2022090529A1 (en) 2020-11-02 2022-05-05 Ares Trading S.A. Combination treatment of cancer
WO2022090527A1 (en) 2020-11-02 2022-05-05 Ares Trading S.A. Combination treatment of cancer
WO2022233718A2 (en) 2021-05-03 2022-11-10 Merck Patent Gmbh Her2 targeting fc antigen binding fragment-drug conjugates
WO2022248380A1 (en) 2021-05-25 2022-12-01 Merck Patent Gmbh Egfr targeting fc antigen binding fragment-drug conjugates
WO2022258622A1 (en) 2021-06-07 2022-12-15 Ares Trading S.A. Combination treatment of cancer

Also Published As

Publication number Publication date
JP2017506217A (en) 2017-03-02
IL281565A (en) 2021-05-31
EP3889172A1 (en) 2021-10-06
SG10202108879SA (en) 2021-09-29
CL2016002017A1 (en) 2017-06-09
HRP20210977T1 (en) 2021-09-17
AU2015213988A1 (en) 2016-07-07
IL246968A0 (en) 2016-09-29
CY1124408T1 (en) 2022-07-22
US20150225483A1 (en) 2015-08-13
BR112016014952A2 (en) 2017-09-19
EP3105246B1 (en) 2021-03-31
SG11201606577YA (en) 2016-09-29
JP6731346B2 (en) 2020-07-29
ES2876430T3 (en) 2021-11-12
US20230056881A1 (en) 2023-02-23
HUE054873T2 (en) 2021-10-28
MX2021003685A (en) 2021-06-04
RU2016135062A3 (en) 2018-12-12
RU2752424C2 (en) 2021-07-28
LT3105246T (en) 2021-08-25
KR20160119197A (en) 2016-10-12
CA2934979A1 (en) 2015-08-13
NZ721364A (en) 2023-04-28
AU2015213988B2 (en) 2019-07-11
IL281565B (en) 2021-12-01
CN106103488B (en) 2021-07-30
PH12016501549A1 (en) 2016-10-03
PE20210168A1 (en) 2021-01-28
PL3105246T3 (en) 2021-11-29
JP2020174674A (en) 2020-10-29
PE20161096A1 (en) 2016-10-22
PT3105246T (en) 2021-06-28
SI3105246T1 (en) 2021-11-30
AU2019246876B2 (en) 2021-10-21
CN113549159A (en) 2021-10-26
KR102363008B1 (en) 2022-02-16
RU2021114500A (en) 2021-06-07
AU2019246876A1 (en) 2019-10-31
US20200055949A1 (en) 2020-02-20
DK3105246T3 (en) 2021-06-14
EP3889172B1 (en) 2024-05-01
MX2016010067A (en) 2016-10-07
WO2015118175A3 (en) 2015-10-08
IL246968B (en) 2021-04-29
RU2016135062A (en) 2018-03-15
CN106103488A (en) 2016-11-09
AU2022200365A1 (en) 2022-02-17
US20180002436A1 (en) 2018-01-04
RS62038B1 (en) 2021-07-30
EP3105246A2 (en) 2016-12-21
US11370819B2 (en) 2022-06-28
US9676863B2 (en) 2017-06-13

Similar Documents

Publication Publication Date Title
US11370819B2 (en) Targeted TGFβ inhibition
US20220017621A1 (en) Combination Therapy for Cancer
RU2750675C1 (en) Antibodies to pd-1 and compositions
US20190330375A1 (en) Dosing regimens and dosage forms for targeted tgf-b inhibition
KR102526945B1 (en) Humanized antibodies against cd269(bcma)
IL293751A (en) Heavy chain antibodies binding to cd38
US20240025967A1 (en) Recombinant protein targeting pd-1 and tgfbeta
CN114867751A (en) 4-1BB and OX40 binding proteins and related compositions and methods, anti-4-1 BB antibodies, anti-OX 40 antibodies

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: 15717097

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2934979

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 122020025158

Country of ref document: BR

REEP Request for entry into the european phase

Ref document number: 2015717097

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015717097

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016014952

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2015213988

Country of ref document: AU

Date of ref document: 20150210

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2016546949

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 001267-2016

Country of ref document: PE

WWE Wipo information: entry into national phase

Ref document number: 246968

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/010067

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 12016501549

Country of ref document: PH

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

Ref document number: 15717097

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20167024679

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A201609356

Country of ref document: UA

ENP Entry into the national phase

Ref document number: 2016135062

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016014952

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160624