EP3571227A1 - Anticorps anti-tgf-bêta et leur utilisation - Google Patents

Anticorps anti-tgf-bêta et leur utilisation

Info

Publication number
EP3571227A1
EP3571227A1 EP18711394.9A EP18711394A EP3571227A1 EP 3571227 A1 EP3571227 A1 EP 3571227A1 EP 18711394 A EP18711394 A EP 18711394A EP 3571227 A1 EP3571227 A1 EP 3571227A1
Authority
EP
European Patent Office
Prior art keywords
antibody
cancer
seq
fragment
tgf
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
EP18711394.9A
Other languages
German (de)
English (en)
Inventor
Gary Shapiro
Kevin BROWER
Patrick Finn
Richard C. GREGORY
Rao KODURI
Feng Liu
Natalia MALKOVA
Parminder MANKOO
Jack R. POLLARD
Huawei Qiu
Joachim Theilhaber
Christopher Winter
Marcella Yu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sanofi SA
Original Assignee
Sanofi SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sanofi SA filed Critical Sanofi SA
Publication of EP3571227A1 publication Critical patent/EP3571227A1/fr
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • C07K16/2818Immunoglobulins [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 CD28 or CD152
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • TGF- ⁇ Transforming growth factor beta
  • TGF- ⁇ is a cytokine that controls many key cellular functions including proliferation, differentiation, survival, migration, and epithelial mesenchymal transition. It regulates diverse biologic processes, such as extracellular matrix formation, wound healing, embryonic development, bone development, hematopoiesis, immune and inflammatory responses, and malignant transformation. Deregulation of TGF- ⁇ leads to pathological conditions, e.g., birth defects, cancer, chronic inflammation, and autoimmune and fibrotic diseases.
  • TGF- ⁇ has three known isoforms - TGF- ⁇ , 2, and 3. All three isoforms are initially translated as a pro-peptide. After cleavage, the mature C-terminal end remains associated with the N-terminus (called the latency associated peptide or LAP), forming the small latent complex (SLC), which is secreted from the cell. The inability of the SLC to bind to TGF- ⁇ receptor II (TGF ⁇ RII) prevents receptor engagement.
  • TGF ⁇ RII TGF- ⁇ receptor II
  • Activation by dissociation of the N- and C-termini occurs by one of several mechanisms, including proteolytic cleavage, acidic pH, or integrin structural alterations (Connolly et al., Int J Biol Sci (2012) 8(7):964-78).
  • TGF- ⁇ , 2, and 3 are pleiotropic in their function and expressed in different patterns across cell and tissue types. They have similar in vitro activities, but individual knockouts in specific cell types suggest non-identical roles in vivo despite their shared ability to bind to the same receptor (Akhurst et al., Nat Rev Drug Discov (2012)
  • TGF- ⁇ signaling is also modulated by other pathways, including WNT, Hedgehog, Notch, INF, TNF, and RAS.
  • TGF- ⁇ pan-TGF ⁇ -specific therapeutic antibodies safe for human patients.
  • TGF- ⁇ is highly conserved among species.
  • production of antibodies to human TGF- ⁇ in animals such as mice is a challenging task.
  • the present invention provides improved monoclonal antibodies that bind specifically to human TGF- ⁇ , TGF ⁇ 2, and TGF ⁇ 3 (i.e., pan-TGF ⁇ -specific). These antibodies are less prone to form half antibody (i.e. , a dimeric complex having one heavy chain and one light chain) during manufacturing. They also have superior pharmacokinetic profiles such as an increased half-life and thus may confer improved clinical benefits to patients.
  • TGF- ⁇ inhibition such as that effected by the antibodies and antigen-binding fragments of the present invention, alleviates the immunosuppressive microenvironment in tumors and
  • PD-1 programmed cell death protein 1
  • PD-L1 PD-1 ligands 1
  • PD-L2 PD-1 ligands 1
  • PD-L2 PD-1 ligands 1
  • PD-L2 PD-1 ligands 1
  • PD-L2 PD-1 ligands 1
  • PD-L2 PD-1 ligands 1
  • PD-L2 PD-1 ligands 1
  • PD-L2 PD-L2
  • the present invention provides an isolated monoclonal antibody that binds specifically to human TGF- ⁇ , TGF-P2, and TGF-P3, comprising the heavy chain complementarity-determining regions (CDR) 1 -3 in SEQ ID NO: 1 and the light chain CDR1 -3 in SEQ ID NO:2, wherein the antibody comprises a human lgG 4 constant region having a mutation at position 228 (EU numbering).
  • the mutation is a serine-to-proline mutation (S228P).
  • the antibody comprises a heavy chain variable domain (V H ) amino acid sequence corresponding to residues 1 -120 of SEQ ID NO: 1 and a light chain variable domain(V L ) amino acid sequence corresponding to residues 1 -108 of SEQ ID NO:2.
  • the antibody comprises a heavy chain amino acid sequence set forth in SEQ ID NO: 1 (with or without the C-terminal lysine) and a light chain amino acid sequence set forth in SEQ ID NO:2.
  • the invention also features an F(ab') 2 antigen-binding fragment of the above antibody.
  • the antibody or fragment of the present invention has an increased half-life, an increased exposure, or both, as compared to
  • fresolimumab For example, the increase is a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% or more increase.
  • the exposure of a drug such as an antibody or fragment of the present invention is a function of the concentration of the drug in the body with respect to time. The concentration of the drug in the body often is indicated by the level of the drug in the blood, plasma, or serum. Half-life and exposure (bio- exposure) of a drug can be measured by well-known methods, as illustrated in Example 7 below.
  • the invention further provides a composition comprising the antibody of the present invention, wherein the composition comprises less than 1 % of half antibody.
  • the half antibody formation may be determined through purity analysis of monoclonal antibody preparations by using, for example, SDS-capillary electrophoresis under non- reducing conditions or non-reducing SDS-PAGE analysis, followed by densitometry, or RP-HPLC (Angal et al., Mol Immunol (1993) 30(1 ): 105-8; Bloom et al., Protein Science (1997) 6:407-415; Schuurman et al., (2001 ) 38(1 ): 1 -8; and Solanos et al., Anal Chem (2006) 78:6583-94).
  • this composition is a pharmaceutical composition comprising also a pharmaceutically acceptable excipient.
  • the invention provides a method of inhibiting TGF- ⁇ signal transduction in a patient (human) in need thereof, comprising administering to the patient a therapeutic amount of the antibody or fragment of the present invention.
  • the patient has an immune-mediated disease (e.g., scleroderma), a fibrotic condition (e.g., a renal fibrotic condition such as focal segmental
  • FSGS glomerulosclerosis
  • lung fibrotic condition such as idiopathic pulmonary fibrosis
  • birth or bone defect e.g., osteogenesis imperfecta
  • the patient has cancer.
  • the antibody or fragment used in the method inhibits the differentiation of CD4 + T cells into inducible regulatory T cells (iTreg).
  • the antibody or fragment may alleviate the immunosuppressive tumor microenvironment. This action of the antibody or fragment helps activate the immune system and potentiates the efficacy of immunotherapy.
  • Efficacy of the treatment methods described herein may be indicated by, for example, one or more of the following in the patient (e.g., in the tumor tissue of the patient): (1 ) an increase in MIP2 and/or KC/GRO levels, (2) activation or infiltration to the tumor tissue of CD8 + T cells such as INF-y-positive CD8 + T cells, and (3) an increase in clustering of natural killer (NK) cells.
  • NK natural killer
  • the present invention further provides a method of treating cancer in a patient (human), comprising administering to the patient (1 ) a therapeutic effective amount of the antibody or fragment of the present invention, and (2) a therapeutic effective amount of an inhibitor of an immune checkpoint protein.
  • These two agents can be administered concurrently (e.g., in a single composition or in separate compositions), or sequentially in either order.
  • the two agents can, for example, be administered on the same day.
  • therapeutic agent (1 ) is administered to the patient before (e.g., one or more days before) therapeutic agent (2).
  • the immune checkpoint protein is PD-1 , PD-L1 , or PD- L2.
  • the inhibitor of the immune checkpoint protein is an anti- PD-1 antibody.
  • the anti-PD-1 antibody comprises (1 ) the heavy chain CDR1 -3 in SEQ ID NO:5 and the light chain CDR1 -3 in SEQ ID NO:6, (2) a V H amino acid sequence corresponding to residues 1 -1 17 of SEQ ID NO:5 and a V L amino acid sequence corresponding to residues 1 -107 of SEQ ID NO:6, or (3) a heavy chain amino acid sequence set forth in SEQ ID NO:5 (with or without the C-terminal lysine) and a light chain amino acid sequence set forth in SEQ ID NO:6.
  • the method comprises administering to the cancer patient an anti-TGF- ⁇ antibody comprising a heavy chain amino acid sequence set forth in SEQ ID NO:1 (with or without the C-terminal lysine) and a light chain amino acid sequence set forth in SEQ ID NO:2, and an anti-PD-1 antibody comprising a heavy chain amino acid sequence set forth in SEQ ID NO:5 (with or without the C-terminal lysine) and a light chain amino acid sequence set forth in SEQ ID NO:6.
  • the patient is refractory to anti-PD-1 antibody monotherapy.
  • the patient may have advanced or metastatic melanoma, or cutaneous squamous cell carcinoma.
  • the anti-TGF- ⁇ antibody and the anti-PD-1 antibody are administered every 2 weeks or every 3 weeks to the patient.
  • the two agents are administered respectively at a dose of 0.01 -40 (e.g., 0.02-20, 0.05-15, or 0.05-20) mg/kg body weight.
  • the present invention also provides a method of increasing an immune response in a patient in need thereof, comprising administering to the patient an immune checkpoint inhibitor and the antibody or fragment of the present invention.
  • the immune checkpoint inhibitor is an anti-PD-1 antibody, such as one comprising (1 ) the HCDR1 -3 in SEQ ID NO:5 and the LCDR1 -3 in SEQ ID NO:6; (2) a VH and a VL corresponding to residues 1 -1 17 in SEQ ID NO:5 and residues 1 -107 in SEQ ID NO:6, respectively; or (3) a heavy chain having the amino acid sequence of SEQ ID NO:5 (with or without the C-terminal lysine) and a light chain having the amino acid sequence of SEQ ID NO:6.
  • the methods of the present invention can be used to treat a variety of cancers, including, without limitation, melanoma (e.g., metastatic or advanced), lung cancer (e.g., non-small cell lung cancer), cutaneous squamous cell carcinoma, colorectal cancer, breast cancer, ovarian cancer, fallopian cancer, uterine cancer, head and neck cancer (e.g., head and neck squamous cell carcinoma), liver cancer (e.g., hepatocarcinoma), urothelial cancer, and kidney cancer (e.g., renal cell carcinoma).
  • the patient has a mesenchymal tumor or a mesenchymal subtype of a solid tumor.
  • Examples of such a solid tumor include those in the colon (e.g., colorectal cancer), ovary, head and neck (e.g., head and neck squamous cell carcinoma), liver (e.g., hepatocellular carcinoma), and the urothelial system.
  • the cancer including a mesenchymal tumor, may be characterized by overexpression of one or more of ACTA2 (smooth muscle a2 actin), VIM (vimentin), MGP (Matrix Gla Protein), ZWINT (ZW10 Interacting Kinetochore Protein), and ZEB2 (Zinc finger E-box-binding homeobox 2).
  • ACTA2 smooth muscle a2 actin
  • VIM vimentin
  • MGP Microx Gla Protein
  • ZWINT ZW10 Interacting Kinetochore Protein
  • ZEB2 Zinc finger E-box-binding homeobox 2
  • the expression levels of such biomarkers may be determined, for example, at the mRNA level or protein level in a biological sample from the patient, such as a tumor biopsy or circulating tumor cells.
  • the present invention also provides the aforementioned antibodies, fragments, or compositions for use in treating the conditions described herein, as well as the use of the aforementioned antibodies, fragments, or compositions in the manufacture of medicaments for treatment of the conditions described herein.
  • nucleic acid expression vectors encoding the heavy or light chain, or both, of an antibody of the invention; host cells comprising the heavy chain and light chain coding sequences for the antibody; and methods of making the antibody using the host cells comprising the steps of culturing the host cells in appropriate culture medium to allow expression of the antibody genes and then harvesting the antibody.
  • FIGS. 1A-E are graphs showing the effects of Ab1 , fresolimumab, and 1 D1 1 on the proliferation of mink lung (Mv 1 Lu) cells treated with 1 ng/ml of human TGF- ⁇ (A), human TGF-P2 (B), human TGF-P3 (C), murine TGF- ⁇ (D), or murine TGF-P2 (E)
  • the antibody concentration is in pg/ml.
  • FIG. 2 is a bar graph showing the effect of 50 g/ml Ab1 on human inducible regulatory T cell (iTreg) differentiation. Stimulation provided to the T cells was anti-CD3 and anti-CD28 antibodies plus IL-2.
  • FIG. 3 is a bar graph showing the effect of Ab1 on human inducible regulatory T cell (iTreg) differentiation in human CD4+ T cell cultures treated with 2 ng/ml human TGF- ⁇ . Stimulation provided to the T cells was anti-CD3 and anti-CD28 antibodies plus IL-2.
  • FIG. 4 is a bar graph showing the effect of Ab1 (30 g/ml) and human TGF- ⁇ 1 (18 ng/ml) on NFATc-driven luciferase expression on Jurkat T cells following T cell stimulation and anti-PD-1 treatment.
  • FIG. 5 is a graph showing the median tumor volumes with the median absolute deviation (MAD) in the indicated treatment groups using the C57BL/6 MC38 colon mouse model.
  • Vehicle PBS.
  • Anti-PD-1 x-anti-mPD-1 Mab (see Detailed Description below).
  • Isotype control of Ab1 anti-HEL hlgG4.
  • FIG. 6 is a scatter dot graph showing tumor volume changes from baseline on day 27 of indicated treatments using the C57BL/6 MC38 colon mouse model.
  • Control PBS.
  • Anti-PD-1 RPM1 14 mlgG1 x-anti-mPD-1 Mab.
  • FIGS. 7A-F are graphs showing the tumor volumes over time for each indicated treatment group using the C57BL/6 MC38 colon mouse model. Each line in the graphs represents one animal, "mpk”: mg/kg. "Ab1 Isotype Ctrl”: anti-HEL hlgG4. aPD1 : x-anti-mPD-1 Mab.
  • FIG. 8 is a graph showing the effects of Ab1 on active TGF- ⁇ concentration in LoVo tumor lysates.
  • FIG. 9A is a graph showing the serum concentration of Ab1 and fresolimumab over time in five groups of rats given a single dose of either antibody at 5 mg/kg.
  • Groups (Gr.) 1 -3 were given three different batches (B1 , B2, and B3) of fresolimumab. Groups 4 and 5 were given two different batches (B1 and B2) of Ab1 .
  • FIG. 9B is a graph showing the serum concentration of Ab1 and fresolimumab over time in monkeys given a single dose of either antibody at 1 mg/kg.
  • FIG. 9C is a graph showing the serum concentration of Ab1 and
  • fresolimumab over time in monkeys given five weekly doses of Ab1 at 1 mg/kg per dose or biweekly doses of fresolimumab at 1 mg/kg per dose for the indicated duration of studies.
  • FIG. 9D is a graph showing the serum concentration of Ab1 and
  • fresolimumab over time in monkeys given a single dose of either antibody at 10 mg/kg.
  • FIG. 9E is a graph showing the serum concentration of Ab1 and fresolimumab over time in monkeys given five weekly doses of Ab1 at 10 mg/kg per dose or biweekly doses of fresolimumab at 10 mg/kg per dose for the indicated duration of studies.
  • FIG. 10A is a graph showing changes in the levels of TGF- ⁇ in MC38 tumors following treatment with Ab1 (+/- anti-PD1 ).
  • FIG. 10B is a graph showing changes in the levels of MIP-2 in MC38 tumors following treatment with Ab1 (+/- anti-PD1 ).
  • FIG. 10C is a graph showing changes in the levels of KC/GRO in MC38 tumors following treatment with Ab1 (+/- anti-PD1 ).
  • FIG. 1 1 A are graphs quantifying CellTrace Violet staining and IFN- ⁇ staining of CD8 pos cells.
  • FIG. 1 1 B is a graph showing that Ab1 restored both proliferation and IFN- ⁇ production in TGFp-treated CD8 + T Cells.
  • FIG. 12A is a graph showing relative abundance of CD8 + T cells (log2- transformed) across the compendium of syngeneic mouse tumor models for colon cancer, leukemia, lung cancer, lymphoma, breast cancer, melanoma, mesothelioma and renal cancer.
  • FIG. 12B is a graph showing TGFp pathway activation across the
  • the present invention features improved pan-TGF-p-specific monoclonal antibodies that are less prone to form half antibody while also having superior pharmacokinetic profiles such as higher exposure in the body than prior known antibodies.
  • the present antibodies are collectively called "Ab1 and related antibodies” and share the common structure characteristics that they have the heavy chain CDR (HCDR) 1 -3 in SEQ ID NO: 1 and the light chain CDR (LCDR) 1 -3 in SEQ ID N0:2, and have a human lgG 4 constant region where residue 228 (EU numbering) in the hinge region has been mutated from serine to proline.
  • P228 is in box and boldface in the sequence of SEQ ID NO: 1 shown below.
  • Antibody Ab1 has an estimated molecular weight of 144 KD when un- glycosylated. Its heavy and light amino acid sequences are SEQ ID NOs: 1 and 2, respectively. These two sequences are shown below. Variable domains are italicized. CDRs are shown in boxes. The glycosylation site in the constant domain of the heavy chain is in boldface (N297).
  • the antibodies of the present invention do not have the C-terminal lysine in the heavy chain.
  • the C- terminal lysine may be removed during manufacture or by recombinant technology (i.e., the coding sequence of the heavy chain does not include a codon for the C-terminal terminal lysine).
  • antibodies comprising the heavy chain amino acid sequence of SEQ ID NO: 1 without the C-terminal lysine are contemplated within the invention.
  • Ab1 and related antibodies bind specifically to human TGF- ⁇ , - ⁇ 2, and - ⁇ 3.
  • the binding has a K D less than 10 "7 M, such as less than 10 "8 M (e.g., 1 -5 nM), as determined by, e.g., surface plasmon resonance (see, e.g., Example 1 below), or Bio-Layer Interferometry.
  • Ab1 and related antibodies also may have a strong TGF- ⁇ neutralizing potency when assayed in a mink lung epithelial cell assay (see, e.g., Example 2 below), or an EC50 of about 0.05 to 1 pg/ml as determined in an A549 cell IL-1 1 induction assay (see, e.g., Example 6 in PCT Publication WO 2006/086469, whose disclosure is incorporated by reference herein in its entirety).
  • fresolimumab may form as much as 6-18% half antibody (i.e., a dimer having a heavy chain and a light chain, rather than a tetramer having two heavy chains complexed with two light chains) under non-reducing denaturing conditions.
  • Ab1 yields substantially less half antibody ( ⁇ 1 %).
  • Ab1 and related antibodies give rise to purer drug product during manufacturing.
  • Ab1 and related antibodies may have improved pharmacokinetic (PK) profiles over fresolimumab. They may have linear PK behavior with a much longer half-life and a lower elimination rate than fresolimumab, leading to about 1 .7-folder higher exposure in vivo than fresolimumab.
  • PK pharmacokinetic
  • Ab1 in rats, Ab1 have been shown to have an average half-life of 7.1 days, compared to fresolimumab's 4.3 days, and an elimination rate (CL) of 0.30 ml/hr/kg, compared to fresolimumab's 0.51 ml/hr/kg
  • the constant domain of Ab1 and related antibodies can be further modified as needed, for example, at Kabat residue L248 (e.g., by introducing the mutation L248E), to reduce any undesired effector function of the molecule.
  • antibody refers to a tetrameric protein comprising two heavy (H) chains (about 50-70 kDa) and two light (L) chains (about 25 kDa) inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable domain (V H ) and a heavy chain constant region (C H ).
  • Each light chain is composed of a light chain variable domain (V L ) and a light chain constant region (C L ).
  • V H and V L domains can be subdivided further into regions of hypervariability, called “complementarity determining regions” (CDRs), interspersed with regions that are more conserved, called “framework regions” (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • Each V H or V L is composed of three CDRs and four FRs, arranged from amino-terminus to carboxyl-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, and FR4.
  • human antibody refers to an antibody in which the variable domain and constant region sequences are derived from human sequences.
  • the term encompasses antibodies with sequences derived from human genes, but those sequences have been modified, e.g., to decrease immunogenicity, increase affinity, and increase stability.
  • the term encompasses antibodies produced recombinantly in nonhuman cells, which may impart glycosylation not typical of human cells.
  • chimeric antibody refers to an antibody that comprises sequences from two different animal species.
  • a chimeric antibody may contain V H and V L of a murine antibody (i.e. , an antibody encoded by murine antibody genes such as an antibody obtained from an immunized mouse using the hybridoma technology) linked to the constant regions of an antibody from another species (e.g., human, rabbit, or rat).
  • an antigen-binding fragment of an antibody refers to a fragment of an antibody that retains the ability to specifically bind to an antigen.
  • an antigen-binding fragment of the present invention is an F(ab') 2 fragment, which is a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region (Fab is a monovalent antibody fragment consisting of the V L , V H , C L and C H i domains).
  • an antigen-binding fragment of the present invention may also comprise a CH2 or CH3 domain.
  • isolated protein refers to a protein, polypeptide or antibody that by virtue of its origin or source of derivation (1 ) is not associated with naturally associated components that accompany it in its native state, (2) is substantially free of other proteins from the same species, (3) is expressed by a cell from a different species, or (4) does not occur in nature.
  • isolated polypeptide that is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be "isolated" from its naturally associated
  • a protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
  • TGF- ⁇ receptor is widely expressed on immune cells, leading to broad effects of TGF- ⁇ in both the innate and adaptive immune system.
  • TGF- ⁇ has been linked to many diseased conditions, for example, birth defects, cancer, chronic inflammation, autoimmunity, and fibrotic diseases.
  • a therapeutic amount of Ab1 or a related antibody may be used to treat these conditions.
  • a "therapeutically effective" amount refers to the amount of Ab1 , a related antibody, or another therapeutic agent referred to herein, that relieves one or more symptoms of the treated condition. This amount may vary based on the condition or patient being treated, and can be
  • the Ab1 or related antibody may be administered at 40, 20, or 15 mg/kg or less (such as 14, 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 mg/kg).
  • the doses may be 0.01 , 0.02, 0.03, 0.04, 0.05, 0.1 , 0.2, 0.3, 0.4, or 0.5 mg/kg.
  • the dosing frequency may be, for example, daily, every two, three, four, or five days, weekly, biweekly, or triweekly, monthly, or bimonthly.
  • the antibody may be administered intravenously (e.g., intravenous infusion over 0.5-8 hours), subcutaneously, topically, or any other route of administration that is appropriate for the condition and the drug formulation.
  • Ab1 and related antibodies are derived from human antibody genes and thus have low immunogenicity in humans. Toxicology studies of Ab1 are detailed in
  • Example 8 Certain cardiac and pulmonary side effects were observed in rats. Thus, patients may be monitored for adverse events when treating patients with Ab1 or a related antibody.
  • efficacy of the antibodies of the invention can be indicated by one or more of the following in the patient (e.g., in an affected tissue such as tumor tissue in the patient): (1 ) a decrease in the level or activity of TGF- ⁇ , (2) an increase in MIP2 and/or KC/GRO levels, (3) activation or infiltration to the tumor tissue of CD8 + T cells such as INF-y-positive CD8 + T cells, and (4) an increase in clustering of natural killer (NK) cells.
  • Conditions that can be treated by Ab1 and related antibodies may include, without limitation, bone defects (e.g., osteogenesis imperfecta), glomerulonephritis, neural or dermal scarring, lung or pulmonary fibrosis (e.g., idiopathic pulmonary fibrosis), radiation-induced fibrosis, hepatic fibrosis, myelofibrosis, scleroderma, immune-mediated diseases (including rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, Sjogren's syndrome, Berger's disease, and transplant rejection), and Dupuytren's contracture.
  • bone defects e.g., osteogenesis imperfecta
  • glomerulonephritis glomerulonephritis
  • neural or dermal scarring e.g., glomerulonephritis, neural or dermal scarring
  • lung or pulmonary fibrosis e.g., idiopathic pulmonary fibros
  • FSGS focal segmental glomerulosclerosis
  • diabetic type I and type II
  • radiational nephropathy obstructive nephropathy
  • diffuse systemic sclerosis hereditary renal disease (e.g., polycystic kidney disease, medullary sponge kidney, horseshoe kidney), glomerulonephritis, nephrosclerosis, nephrocalcinosis, , systemic or glomerular hypertension, tubulointerstitial nephropathy, renal tubular acidosis, renal tuberculosis, and renal infarction.
  • FSGS focal segmental glomerulosclerosis
  • diabetic type I and type II
  • radiational nephropathy obstructive nephropathy
  • diffuse systemic sclerosis hereditary renal disease (e.g., polycystic kidney disease, medullary sponge kidney, horseshoe kidney), glomerulonephritis, nephrosclerosis, nephrocalcinosis
  • renin inhibitors include but not limited to: renin inhibitors, angiotensin-converting enzyme (ACE) inhibitors, Ang II receptor antagonists (also known as "Ang II receptor blockers”), and aldosterone antagonists.
  • ACE angiotensin-converting enzyme
  • Ang II receptor antagonists also known as "Ang II receptor blockers”
  • aldosterone antagonists See, e.g., WO 2004/098637, whose disclosure is incorporated by reference herein in its entirety.
  • Ab1 and related antibodies are useful to treat diseases and conditions associated with the deposition of ECM, such as systemic sclerosis, postoperative adhesions, keloid and hypertrophic scarring, proliferative vitreoretinopathy, glaucoma drainage surgery, corneal injury, cataract, Peyronie's disease, adult respiratory distress syndrome, cirrhosis of the liver, post myocardial infarction scarring, post angioplasty restenosis, scarring after subarachnoid hemorrhage, fibrosis after laminectomy, fibrosis after tendon and other repairs, biliary cirrhosis (including sclerosing cholangitis), pericarditis, pleurisy, tracheostomy, penetrating CNS injury, eosinophilic myalgic syndrome, vascular restenosis, veno-occlusive disease, pancreatitis and psoriatic arthropathy.
  • diseases and conditions associated with the deposition of ECM such
  • Ab1 and related antibodies further are useful in conditions where promotion of re-epithelialization is beneficial.
  • Such conditions include but are not limited to diseases of the skin, such as venous ulcers, ischemic ulcers (pressure sores), diabetic ulcers, graft sites, graft donor sites, abrasions and burns, diseases of the bronchial epithelium, such as asthma, ARDS, diseases of the intestinal epithelium, such as mucositis associated with cytotoxic treatment, esophageal ulcers (reflex disease), gastroesophageal reflux disease, stomach ulcers, small intestinal and large intestinal lesions (inflammatory bowel disease).
  • diseases of the skin such as venous ulcers, ischemic ulcers (pressure sores), diabetic ulcers, graft sites, graft donor sites, abrasions and burns
  • diseases of the bronchial epithelium such as asthma, ARDS
  • diseases of the intestinal epithelium such as mucositis associated with
  • Ab1 and related antibodies are in conditions in which endothelial cell proliferation is desirable, for example, in stabilizing atherosclerotic plaques, promoting healing of vascular anastomoses, or in conditions in which inhibition of smooth muscle cell proliferation is desirable, such as in arterial disease, restenosis and asthma.
  • Ab1 and related antibodies also are useful to enhance the immune response to macrophage-mediated infections such as those caused by Leishmania spp.,
  • Ab1 and related antibodies also are useful for the prevention and/or treatment of ophthalmological conditions such as glaucoma and scarring after trabeculectomy.
  • TGF- ⁇ regulates several biological processes, including cell proliferation, epithelial-mesenchymal transition (EMT), matrix remodeling, angiogenesis, and immune functions. Each of these processes contributes to tumor progression.
  • EMT epithelial-mesenchymal transition
  • matrix remodeling matrix remodeling
  • angiogenesis angiogenesis
  • Ab1 and related antibodies are useful in the treatment of hyperproliferative diseases, such as cancers including but not limited to skin cancer (e.g., melanoma, including unresectable or metastatic melanoma, cutaneous squamous cell carcinoma, and keratoacanthoma), lung cancer (e.g., non-small cell lung cancer), esophageal cancer, stomach cancer, colorectal cancer, pancreatic cancer, liver cancer (e.g., hepatocellular carcinoma), primary peritoneal cancer, bladder cancer, renal cancer or kidney cancer (e.g., renal cell carcinoma), urothelial carcinoma, breast cancer, ovarian cancer, fallopian cancer, cervical cancer, uterine cancer, prostate cancer, testicular cancer, head and neck cancer (e.g., head and neck squamous cell carcinoma), brain cancer, glioblastoma, glioma, mesothelioma, leukemia, and lymphoma.
  • skin cancer e.g
  • Ab1 and related antibodies are useful in treating cancers in patients for whom a prior therapy based on an anti-PD-1 , anti-PD-L1 or anti- PD-L2 therapeutic agent has failed or is expected to fail, i.e., patients who are or expected to be non-responders to an anti-PD-1 , anti-PD-L1 , or anti-PD-L2 therapy.
  • Ab1 and related antibodies are useful in the treatment of cancers in patients who have relapsed from a prior anti-PD-1 , anti-PD-L1 , or anti-PD-L2 therapy.
  • the term "expected" means that a skilled person in the medical art may anticipate, without administering a therapy, whether a patient will be a responder or a non-responder and whether the therapy will fail or will not be effective, based on his/her general medical knowledge and the specific conditions of the patient.
  • the cancers are mesenchymal subtypes of solid tumors, including, without limitation, mesenchymal colorectal cancer, mesenchymal ovarian cancer, mesenchymal lung cancer, mesenchymal head cancer and
  • EMT Epithelial mesenchymal transition
  • Ab1 and related antibodies are useful in treating patients with advanced solid tumors.
  • Ab1 and related antibodies can also be used in the treatment of
  • hematopoietic disorders or malignancies such as multiple myeloma, myelodysplastic syndrome (MDS), Hodgkin lymphoma, non-Hodgkin lymphoma, and leukemia, as well as various sarcomas such as Kaposi's Sarcoma.
  • MDS myelodysplastic syndrome
  • Hodgkin lymphoma Hodgkin lymphoma
  • non-Hodgkin lymphoma non-Hodgkin lymphoma
  • leukemia as well as various sarcomas such as Kaposi's Sarcoma.
  • Ab1 and related antibodies can also be useful to inhibit cyclosporine- mediated malignancy or cancer progression (e.g., metastases).
  • treatment includes any medical intervention resulting in the slowing of cancer growth, delay in cancer progression or recurrence, or reduction in cancer metastases, as well as partial remission of the cancer in order to prolong life expectancy of a patient.
  • TGF- ⁇ suppresses almost all aspects of the anti-tumor immune response.
  • the cytokine promotes iTreg differentiation and reduces cytotoxic (CD8 + ) cell proliferation and infiltration.
  • Inhibition of TGF- ⁇ by Ab1 or a related antibody will alleviate the immunosuppressive tumor microenvironment, as described above, to bring positive outcomes to cancer patients.
  • Ab1 and related antibodies can allow checkpoint modulators, such as anti-PD-1 antibody, to better induce immune
  • Ab1 and related antibodies can also be used in conjunction with other cancer therapies such as chemotherapy (e.g., platinum- or taxoid-based therapy), radiation therapy, and therapies that target cancer antigens or oncogenic drivers.
  • chemotherapy e.g., platinum- or taxoid-based therapy
  • radiation therapy e.g., platinum- or taxoid-based therapy
  • therapies that target cancer antigens or oncogenic drivers e.g., cancer antigens or oncogenic drivers.
  • Cancers that can be treated by a combination involving Ab1 or a related antibody and an immune checkpoint inhibitor such as an anti-PD-1 antibody include the cancers listed in the above subsection.
  • the cancers are refractory to a prior anti-PD-1 , anti- PD-L1 , or anti-PD-L2 therapy, such as advanced or metastatic melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck squamous cell carcinoma, and Hodgkin Lymphoma.
  • Refractory patients are patients whose disease progresses as confirmed, e.g., radiologically within 12 weeks of commencing treatment without any evidence of a response.
  • Ab1 or a related antibody can be used in conjunction with another cancer therapy such as anti-PD-1 therapy to treat mesenchymal cancers such as colorectal cancer, non-small cell lung cancer, ovarian cancer, bladder cancer, head and neck squamous cell carcinoma, renal cell carcinoma, hepatocellular carcinoma, and cutaneous squamous cell carcinoma. See also discussions above.
  • anti-PD-1 antibodies examples include nivolumab, pembrolizumab,
  • pidilizumab, MEDI0608 (formerly AMP-514; see, e.g., WO 2012/145493 and U.S. Patent 9,205, 148), PDR001 (see, e.g., WO 2015/1 12900), PF-06801591 (see, e.g., WO
  • the anti-PD-1 antibodies include those disclosed in WO 2015/1 12800 (such as those referred to as H1 M7789N, H1 M7799N, H1 M7800N, H2M7780N, H2M7788N,
  • the disclosure of WO 2015/1 12800 is incorporated by reference herein in its entirety.
  • the antibodies disclosed in WO 2015/1 12800 and related antibodies including antibodies and antigen-binding fragments having the CDRs, V H and V L sequences, or heavy and light chain sequences disclosed in that PCT
  • a useful anti-PD-1 antibody may comprise the heavy and light chain amino acid sequences shown below as SEQ ID NOs:5 and 6, respectively; the V H and V L sequences in SEQ ID NOs:5 and 6 (shown in italics); or one or more (e.g., all six) CDRs in SEQ ID NOs:5 and 6 (shown in boxes).
  • the antibodies of the present invention do not have the C-terminal lysine in the heavy chain.
  • the C- terminal lysine may be removed during manufacture or by recombinant technology (i.e., the coding sequence of the heavy chain does not include a codon for the C-terminal terminal lysine).
  • antibodies comprising the heavy chain amino acid sequence of SEQ ID NO: 5 without the C-terminal lysine are contemplated within the invention.
  • an anti-TGF- ⁇ antibody or fragment of the present invention can also be used in conjunction with an antibody against an
  • immunomodulatory antigen such as PD-L1 and CTLA-4.
  • PD-L1 and CTLA-4 immunomodulatory antigen
  • exemplary anti-PD-L1 antibodies are atezolizumab, avelumab, durvalumab, LY3300054 and BMS-936559.
  • Exemplary anti-CTLA-4 antibodies are ipilimumab or tremelimumab.
  • Efficacy of Ab1 and related antibodies can be determined by biomarkers or target occupancy.
  • target occupancy can be assayed by evaluating levels of active TGF- ⁇ in biopsies using a Meso Scale Discovery (MSD) assay.
  • MSD Meso Scale Discovery
  • target engagement can be assayed by evaluating the effect of decreased circulating TGF- ⁇ on peripheral blood mononuclear cells such as
  • T cells lymphocytes
  • B cells NK cells
  • monocytes lymphocytes
  • increased proliferation of circulating CD8 + T cells can be evaluated using CD45 + RO + CCR7 + CD28 + Ki67 + as markers in flow cytometry.
  • Activation of circulating NK cells can be evaluated using CD3 " CD56 high dim CD16 + or CD137 + as markers in flow cytometry.
  • Ki- 67, PD-1 , and ICOS can be used as PD markers associated with T cell activation.
  • Immune modulation upon treatment by Ab1 or a related antibody can be assayed by evaluating changes of infiltrating immune cells and immune markers by multiplex immunohistochemistry (IHC) assays using, e.g., the NeoGenomics platform.
  • IHC immunohistochemistry
  • NeoGenomic's MultiOmyx TIL Panel stains for a panel of immune markers, allowing for quantitative determination of density and localization of various immune cells.
  • the immune markers may indicate differentiation of iTreg; infiltration and proliferation of CD8 + T cells; and generation of IFN ⁇ by CD8 + T cells.
  • Ab1 has been shown to inhibit CD4 + T cells' differentiation into iTreg (see, e.g., Example 3, infra), and to increase CD8 + T cell proliferation and their generation of IFN ⁇ (as shown in a mixed lymphocyte reaction assay; data not shown).
  • efficacy of treatment by Ab1 or a related antibody can be indicated by inhibition of iTreg, induction of CD8 + T cell proliferation and infiltration to tumor or other diseased tissues, increased IFN ⁇ production, and/or an increased ratio of CD8 + T cells to Treg cells.
  • Immune modulation upon treatment by Ab1 or a related antibody also can be assayed in peripheral blood by methylation-PCR based quantitative immune cell counting of CD8 + T cells, Treg cells, NK cells, and other immune cells.
  • the treatment efficacy may manifest clinically as a delay or reversal in disease progression such as tumor progression.
  • the Ab1 and related antibodies, as well as antibodies targeting other co- targets such as PD-1 , PD-L1 or PD-L2, can be made by methods well established in the art.
  • DNA sequences encoding the heavy and light chains of the antibodies can be inserted into expression vectors such that the genes are operatively linked to necessary expression control sequences such as transcriptional and translational control sequences.
  • Expression vectors include plasm ids, retroviruses, adenoviruses, adeno- associated viruses (AAV), plant viruses such as cauliflower mosaic virus, tobacco mosaic virus, cosmids, YACs, EBV derived episomes, and the like.
  • the antibody light chain coding sequence and the antibody heavy chain coding sequence can be inserted into separate vectors, and may be operatively linked to the same or different expression control sequences (e.g., promoters).
  • both coding sequences are inserted into the same expression vector and may be operatively linked to the same expression control sequences (e.g., a common promoter), to separate identical expression control sequences (e.g., promoters), or to different expression control sequences (e.g., promoters).
  • the antibody coding sequences may be inserted into the expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present).
  • the recombinant expression vectors may carry regulatory sequences that control the expression of the antibody chain genes in a host cell.
  • regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from retroviral LTRs, cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)), polyoma and strong mammalian promoters such as native immunoglobulin and actin promoters.
  • CMV cytomegalovirus
  • SV40 Simian Virus 40
  • AdMLP adenovirus major late promoter
  • polyoma e.g., the adenovirus major late promoter (AdMLP)
  • AdMLP aden
  • the recombinant expression vectors of the invention may carry additional sequences, such as sequences that regulate replication of the vector in host cells (e.g., origins of replication) and selectable marker genes.
  • the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced.
  • Selectable marker genes may include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification), the neo gene (for G418 selection), and the glutamate
  • the expression vectors encoding the antibodies of the present invention are introduced to host cells for expression.
  • the host cells are cultured under conditions suitable for expression of the antibody, which is then harvested and isolated.
  • Host cells include mammalian, plant, bacterial or yeast host cell.
  • Mammalian cell lines available as hosts for expression are well known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC).
  • CHO Chinese hamster ovary
  • NS0 cells NS0 cells
  • SP2 cells HEK-293T cells
  • 293 Freestyle cells Invitrogen
  • NIH-3T3 cells HeLa cells
  • BHK baby hamster kidney
  • COS African green monkey kidney cells
  • human hepatocellular carcinoma cells e.g., Hep G2
  • A549 cells and a number of other cell lines.
  • Cell lines may be selected based on their expression levels.
  • Other cell lines that may be used are insect cell lines, such as Sf9 or Sf21 cells.
  • expression of antibodies can be enhanced using a number of known techniques.
  • the glutamine synthetase gene expression system (the GS system) is a common approach for enhancing expression under certain conditions.
  • Tissue culture media for the host cells may include, or be free of, animal- derived components (ADC), such as bovine serum albumin.
  • ADC animal- derived components
  • bovine serum albumin a component that is preferred for human safety.
  • Tissue culture can be performed using the fed-batch method, a continuous perfusion method, or any other method appropriate for the host cells and the desired yield.
  • the antibody of the invention can be formulated for suitable storage stability.
  • the antibody can be lyophilized or stored or reconstituted for use using pharmaceutically acceptable excipients.
  • the two or more therapeutic agents such as antibodies can be co-formulated, e.g., mixed and provided in a single composition.
  • excipient or “carrier” is used herein to describe any ingredient other than the compound(s) of the invention.
  • carrier is used herein to describe any ingredient other than the compound(s) of the invention.
  • excipient(s) will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
  • “Pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • pharmaceutically acceptable excipients are water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride will be included in the composition. Additional examples of
  • a pharmaceutical composition of the invention may be prepared, packaged, or sold in bulk, as a single unit dose, or as a plurality of single unit doses.
  • a "unit dose" is a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject or a convenient fraction of such a dosage such as, for example, one-half or one- third of such a dosage.
  • compositions of the invention are typically suitable for parenteral administration.
  • parenteral administration of a
  • pharmaceutical composition includes any route of administration characterized by physical breaching of a tissue of a subject and administration of the pharmaceutical composition through the breach in the tissue, thus generally resulting in the direct administration into the blood stream, into muscle, or into an internal organ.
  • Parenteral administration thus includes, but is not limited to, administration of a pharmaceutical composition by injection of the composition, by application of the composition through a surgical incision, by application of the composition through a tissue-penetrating nonsurgical wound, and the like.
  • parenteral administration is contemplated to include, but is not limited to, subcutaneous, intraperitoneal, intramuscular, intrasternal, intravenous, intraarterial, intrathecal, intraventricular, intraurethral, intracranial, intratumoral, and intrasynovial injection or infusions; and kidney dialytic infusion techniques. Regional perfusion is also contemplated. Preferred embodiments may include the intravenous and the subcutaneous routes.
  • Formulations of a pharmaceutical composition suitable for parenteral administration typically comprise the active ingredient combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline. Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration. Injectable formulations may be prepared, packaged, or sold in unit dosage form, such as in ampoules or in multi dose containers containing a preservative. Formulations for parenteral administration include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and the like. Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents.
  • a pharmaceutically acceptable carrier such as sterile water or sterile isotonic saline.
  • Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration.
  • injectable formulations may be prepared, packaged, or sold in unit dosage form
  • the active ingredient is provided in dry (i.e., powder or granular) form for reconstitution with a suitable vehicle (e.g., sterile pyrogen free water) prior to parenteral administration of the reconstituted composition.
  • a suitable vehicle e.g., sterile pyrogen free water
  • Parenteral formulations also include aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (e.g., with a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
  • exemplary parenteral administration forms include solutions or suspensions in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • Other parentally-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form, or in a liposomal preparation.
  • Formulations for parenteral administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed- sustained- pulsed-, controlled- , targeted-, and programmed-release.
  • CDR complementarity-determining regions
  • the antibody of embodiment 1 wherein the antibody comprises a heavy chain variable domain (V H ) amino acid sequence corresponding to residues 1 -120 of SEQ ID NO: 1 and a light chain variable domain(V L ) amino acid sequence corresponding to residues 1 -108 of SEQ ID NO:2.
  • V H heavy chain variable domain
  • V L light chain variable domain
  • the antibody comprises a heavy chain amino acid sequence set forth in SEQ ID NO:1 (with or without the C-terminal lysine) and a light chain amino acid sequence set forth in SEQ ID NO:2.
  • a) inhibits the differentiation of CD4 + T cells into inducible regulatory T cells (iTreg);
  • NK natural killer
  • compositions comprising the antibody or fragment of any one of embodiments 1 -6, wherein the composition comprises less than 1 % of half antibody.
  • a method of inhibiting TGF- ⁇ signal transduction in a patient in need thereof comprising administering to the patient a therapeutic amount of the antibody or fragment of any one of embodiments 1 -6.
  • the cancer is selected from the group consisting of melanoma, lung cancer, cutaneous squamous cell carcinoma, colorectal cancer, breast cancer, ovarian cancer, head and neck cancer, hepatocellular carcinoma, urothelial cancer, and renal cell carcinoma.
  • a method of treating cancer in a patient comprising administering to the patient (1 ) the antibody or fragment of any one of embodiments 1 -6, and (2) an inhibitor of an immune checkpoint protein.
  • the anti-PD-1 antibody comprises the heavy chain CDR1 -3 in SEQ ID NO:5 and the light chain CDR1 -3 in SEQ ID NO:6.
  • the anti-PD-1 antibody comprises a V H amino acid sequence corresponding to residues 1 -1 17 of SEQ ID NO:5 and a V L amino acid sequence corresponding to residues 1 -107 of SEQ ID NO:6.
  • the anti-PD-1 antibody comprises a heavy chain amino acid sequence set forth in SEQ ID NO:5 (with or without the C- terminal lysine) and a light chain amino acid sequence set forth in SEQ ID NO:6.
  • anti-TGF- ⁇ antibody comprises a heavy chain amino acid sequence set forth in SEQ ID NO: 1 (with or without the C-terminal lysine) and a light chain amino acid sequence set forth in SEQ ID NO:2.
  • ACTA2 overexpression of one or more of ACTA2, VIM, MGP, and ZWINT.
  • the cancer is selected from the group consisting of melanoma, lung cancer, cutaneous squamous cell carcinoma, colorectal cancer, breast cancer, ovarian cancer, head and neck cancer, hepatocellular carcinoma, urothelial cancer, and renal cell carcinoma.
  • a method of increasing an immune response in a patient in need thereof comprising administering to the patient an immune checkpoint inhibitor and the antibody or fragment of any one of embodiments 1 -6.
  • anti-TGF- ⁇ antibody comprises a heavy chain amino acid sequence set forth in SEQ ID NO: 1 (with or without the C-terminal lysine) and a light chain amino acid sequence set forth in SEQ ID NO:2.
  • ACTA2 overexpression of one or more of ACTA2, VIM, MGP, and ZWINT.
  • An isolated nucleic acid molecule comprising a nucleotide sequence encoding the heavy chain, the light chain, or both, of the antibody or fragment of any one of embodiments 1 -6.
  • a host cell comprising the expression vector of embodiment 43.
  • a host cell comprising first and second nucleotide sequences encoding the heavy chain and light chain, respectively, of the antibody or antigen-binding fragment
  • a method of producing a pharmaceutical composition comprising:
  • therapeutic agent is an immune checkpoint inhibitor described herein.
  • CM5 carboxy-methylated
  • Ab1 at a series of concentrations (1 .1 1 , 3.33, 10, and 30 nM) was injected onto immobilized recombinant TGF- ⁇ to measure the binding interaction in real time.
  • the TGF- ⁇ homodimers were immobilized at a low density to reduce avidity effects. Injections were performed in triplicate and the binding assay was repeated three times.
  • the data from the kinetic experiments were processed using the Biacore T200 Biaevaluation v2.0 software.
  • the recombinant proteins were either produced internally (human TGF- ⁇ , 2 and 3) or acquired from R&D Systems (murine TGF- ⁇ and 2).
  • Table 1 shows the amino acid sequence homology of the three active TGF- ⁇ isoforms between rhesus monkeys, mice, or rats and humans (homology is reported as the percentage of conserved amino acids over total amino acids).
  • Table 1 Homology of TGF- ⁇ Active Isoforms to Humans
  • the k a , k d , and K D values of Ab1 as determined by the above method are presented in Table 2 below.
  • the K D values of Ab1 for human TGF- ⁇ , 2, and 3 were determined to be 1.48, 3.00, and 1.65 nM, respectively.
  • the K D values of Ab1 for murine/rat TGF- ⁇ and 2 were determined to be 2.80 and 1.88 nM, respectively. These binding properties are similar to those of fresolimumab.
  • TGF- ⁇ Protocols "Transforming Growth Factor- ⁇ Protocols.”
  • Ab1 , fresolimumab, and 1 D1 1 a murine anti- TGF- ⁇ antibody, whose heavy and light chain sequences are disclosed herein as SEQ ID NOs:9 and 10.
  • the recombinant TGF- ⁇ proteins were either produced internally (human TGF- ⁇ , 2 and 3) or acquired from R&D Systems (murine TGF- ⁇ and 2).
  • Treg Regulatory T cells
  • CD4 + T cells without the addition of exogenous TGF- ⁇ were treated with 50 pg/ml of isotype control (human lgG4, kappa anti-hen egg lysozyme (HEL) antibody, Crown Biosience), Ab1 or fresolimumab in the presence or absence of stimulation (anti-CD3, anti-CD28, and IL-2) for 6 days followed by flow cytometric analysis.
  • isotype control human lgG4, kappa anti-hen egg lysozyme (HEL) antibody, Crown Biosience
  • Ab1 or fresolimumab in the presence or absence of stimulation anti-CD3, anti-CD28, and IL-2
  • Mean percent and standard deviation of the CD25 + FOXP3 + population was calculated from the parent population (lymphocytes/live/single cells/CD4 + CD127 " ) in triplicate.
  • Stimulation of total human CD4 + T cells with anti-CD3, anti-CD28, and IL-2 increased the percentage of FOXP3 + CD25 + (iTreg) in the culture from 0% to 15%.
  • Treatment with 50 pg/ml Ab1 or 50 pg/ml fresolimumab decreased the percentage of iTreg to a similar extent (8% and 7% respectively; FIG. 2).
  • treatment with an isotype human lgG 4 (hlgG 4 ) control had minimal effect on iTreg differentiation (20% iTreg) (FIG. 2).
  • CD4 + T cells isolated from a second healthy normal volunteer generated similar results.
  • TGF- ⁇ 2 ng/ml
  • Treatment with increasing concentrations of Ab1 decreased the percentage of iTreg in a concomitant fashion from 55% to 15% at 200 pg/ml and 43% at 6.25 pg/ml.
  • Treatment with fresolimumab decreased the percentage of iTreg to a similar extent as Ab1 (from 55% to 16% at 200 pg/ml and 32% at 6.25 pg/ml).
  • a cell assay system purchased from Promega for this study.
  • This system comprises two cell types: 1 ) Jurkat T cells expressing human PD-1 and a luciferase reporter driven by an NFAT response element, and 2) CHO-K1 cells expressing human PD-L1 and an engineered cell surface protein designed to activate cognate T cell receptor in an antigen-independent manner.
  • CHO-K1 cells Upon co-culture the Jurkat T cells interact with the CHO-K1 cells, causing T cell receptor stimulation and NFATc translocation into the nucleus where it drives luciferase expression.
  • tyrosine-protein phosphatase non-receptor 1 1 SHP2
  • Blockade of PD-1 signaling relieves the SHP2- dependent suppression and thus allows for maximal luciferase expression.
  • the system thus provides a functional method for determining the effect of TGF- ⁇ on T cell signaling and the impact of Ab1 on anti-PD-1 treatment of the T cells.
  • TGF- ⁇ was purchased from R&D Systems.
  • Isotype control antibody for Ab1 (anti-HEL hlgG 4 ) was purchased from Crown Bioscience (Cat#C0004-5).
  • Mouse anti-hPD-1 IgG and its isotype control antibody were purchased from BioLegend (Cat#329912). Fourteen replicates were analyzed for each sample.
  • the RLU values are also presented in Table 3 below.
  • Ab1 and an anti-mouse PD-1 (mPD-1 ) monoclonal antibody (mAb) as single agents and in combination were evaluated in C57BL/6 female mice.
  • Ab1 (10, 20, and 50 mg/kg) or an isotype control Ab (anti-HEL hlgG 4 purchased from Crown Bioscience; used at 10 and 20 mg/kg) were administered IV every three days (Q3D) as single agents or in combination with the anti-PD1 Mab at 5 mg/kg IV twice weekly.
  • the anti-PD1 Ab used in this study was, designated as
  • anti-mPD-1 Mab "antimPD1_hyb_RMP1 14_mlgG1 LCfullrat” (or x-anti-mPD-1 Mab). It was a chimeric rat anti-mPD-1 antibody generated by replacing the rat Fc region of rat lgG 2a clone RMP1 - 14 (BioXcell, Cat. #BE0146) with a mouse Igd Fc region. The heavy and light amino sequences of this chimeric antibody are shown in SEQ ID NOs:7 and 8. Tolerability was assessed by measuring animal body weights and clinical observations. At the end of the three week treatment, four hours after the last treatment, terminal sampling was performed and tissues (heart, kidney, liver, lung, and spleen) were fixed in
  • a dosage was considered excessively toxic if it produced 15% body weight loss during three consecutive days in an individual mouse, 20% body weight loss during one day, or 10% or more drug-related deaths, unless tumor-induced cachexia leading to body weight loss was observed in the control vehicle-treated group. Animal body weight included the tumor weight.
  • Histopathological analyses identified increased numbers of lymphocytes in the spleen (white pulp) of all treated groups including isotype control antibody treated groups, whatever the combination and without any dose-relationship. No other significant microscopic findings were observed. Two mice from the combination group, the isotype control Ab (10 mg/kg) and the anti-PD-1 Mab (5 mg/kg), were found dead after final administration on the last day of study. Histopathological analyses did not find any drug related causes of death.
  • mice Female C57BL/6 mice were obtained from Charles River Labs (Wilmington, MA, USA). Animals were allowed to acclimate for at least three days before the study enrollment. Mice were 1 1 weeks-old and weighted between 17.0 and 20.9 g at the beginning of the study. They had free access to food (Harlan 2916 rodent diet,
  • MC38 is a colon adenocarcinoma cell line.
  • the cells were obtained from the National Cancer Institute (Bethesda, MD, USA) and cultured in 5% C0 2 at 37°C in complete medium (CM), which included Roswell Park Memorial Institute medium
  • RPMI-1640 with L-glutamine (Gibco, Cat#1 1875), supplemented with 10% heat- inactivated fetal bovine serum (HI FBS) (Gibco, Cat# 10438026).
  • the cells were harvested and re-suspended in Dulbecco's phosphate-buffered saline (DPBS) (Gibco, Cat#14190), and 1 x 10 6 cells/200 ⁇ per mouse were subcutaneously (SC) implanted into the right flank of female C57BL/6 mice.
  • DPBS Dulbecco's phosphate-buffered saline
  • Ab1 was administered to the animals in an aqueous solution. It was 0.22 pm- filtered through PES and stored in sterile aliquots at 2 to 10°C. The antibody was given to the animals at 10 ml/kg intraperitoneally (IP), 25 mg/kg.
  • IP intraperitoneally
  • Anti-HEL hlgG 4 (Crown Bioscience) was used as an isotype control for Ab1 . This antibody was given to control animals by IP at 10 ml/kg by IP, 25 mg/kg.
  • the x-anti-mPD-1 Mab ⁇ supra was provided in DPBS (Gibco, Cat#14190- 094) and was given to the animals by IP at 10 ml/kg, 5 mg/kg.
  • mice On day 0, 60 animals were implanted with MC38 tumor cells. On day 8 post implantation, the mice, which had an average tumor size of 50-75 mm 3 , were pooled and randomly distributed to control and treatment groups (ten mice per group).
  • mice were euthanized when morbidity or weight loss >20% was observed. Tumors were measured with a caliper twice weekly until final sacrifice. When a tumor size reached approximately 2000 mm 3 or there were animal health issues (20% area of a tumor ulcerated), the animals would be euthanized and date of death recorded. Solid tumor volumes were estimated from two dimensional tumor measurements and calculated according to the following equation:
  • Tumor volume (mm 3 ) [length (mm) x width 2 (mm 2 )] / 2
  • the percent median regression for a group at a given day was then obtained by taking the median of the individual percent regression calculated for each animal of the group at this day.
  • the day of calculation was determined on the day when AT/AC (i.e., the ratio of medians of tumor volume changes from baseline between the treated and control groups) was calculated, except when median percent regression was not representative of the activity of the group. In that case, the day was determined on the first day when the median percent regression was maximal.
  • Regressions were defined as partial (PR) if the tumor volume decreased to 50% of the tumor volume at the start of treatment. Complete regression (CR) was considered to have been achieved when tumor volume was below 14 mm 3 or could not be recorded.
  • the primary efficacy end points were tumor volume changes from baseline as indicated by AT/AC, median percent regression, partial regression, and complete regression. Changes in tumor volume for each treated (T) and control (C) group were calculated for each animal every day by subtracting the tumor volume on the day of first treatment (staging day) from the tumor volume on the specified observation day. The median ⁇ was calculated for the treated group and the median AC was calculated for the control group. The ratio AT/AC is calculated and expressed as percentage:
  • a AT/AC ratio ⁇ 40% was considered therapeutically active.
  • a AT/AC ratio of 0% was considered as tumor stasis.
  • a AT/AC ratio ⁇ 0% was considered as tumor regression.
  • Percent tumor regression was defined as percentage of tumor volume decrease in the treated group on a specified observation day compared to the tumor volume at the beginning of the study (to). At a specific time point (t) and for each animal, percent regression was calculated using the following formula:
  • % regression (at t) [(volume ⁇ - volume t ) / volume ⁇ ] x 100
  • the median percent regression for a group on a given day was then calculated by taking the median of individual % regression values calculated for each animal in the group.
  • the day of calculation was determined by the day when AT/AC was calculated, except when the median percent regression was not representative of the activity of the group. In that case, the day was determined by the first day when the median percent regression was maximal.
  • Tables 5 and 6 and FIGS. 5-7 present additional data showing the activity of the antibodies alone or in combination on tumor volumes in the mouse model.
  • Tumor volume changes from baseline mm 3 M edian (nMAD), n, and p-value*
  • MC38 colon adenocarcinoma cells were cultured in RPMI-1640 supplemented with 10% FBS in a humidified 5% C0 2 incubator and then implanted subcutaneously (1X10 6 cells) into the flank of female C57/BI6J mice (Jackson Laboratory, Bar Harbor, ME). Once tumors reached an average size of 50-75 mm 3 , the mice were pooled and randomly distributed to control and treatment groups (10 mice per group). Tumor-bearing mice were then treated intraperitoneally with PBS, an lgG4 isotype control antibody (25 mg/kg). or Ab1 (1 , 10, and 25mg/kg) three times per week until each animal had received a total of 6 to 7 doses. Tumors were
  • Ab1 at a dose of 25 mg/kg Q3D and mouse a-PD-1 antibody at a dose of 5 mg/kg demonstrated partial activity with 2/8 and 4/8 complete regression, respectively, in MC38 tumor bearing mice.
  • the combination of Ab1 at 1 , 10, or 25 mg/kg Q3D and mouse a-PD-1 antibodies at 5 mg/kg Q3D was therapeutically active.
  • the effect of the combination of Ab1 at all tested doses and mouse a-PD-1 antibodies at 5 mg/kg Q3D was greater than the effect of each single agent with 5/8, 6/8 and 7/8 complete regression for 1 , 10, and 25 mg/kg of Ab1 , respectively.
  • Table 6A provides a summary of the results.
  • Intratumoral TGF- ⁇ levels were studied in a LoVo colorectal cancer subcutaneous xenograft-transplanted BALB/c mouse model.
  • the mice were injected with either Ab1 or isotype control Mab at 10, 25, or 50 mg/kg intravenously every 3 days for a total of eight IV administrations, beginning when the tumor volume was less than 100 mm 3 .
  • the lysates were cleared by centrifugation for 10 min at 20,000 x g in an Eppendorf 5417C centrifuge at 4°C. The supernatants were transferred into clean chilled Eppendorf tubes and further cleared by centrifugation for another 20 min as described above. After that, the supernatants were transferred into a plastic 96-weel storage block, snap frozen in liquid nitrogen, and stored at -80°C. [0150] The next day the samples were thawed at room temperature and placed on ice. Protein concentration in the lysates was measured using a Bicinchoninic Acid (BCA) Protein Assay kit (Thermo 23225) according to the manufacturer's instructions. Lysates were normalized to a protein concentration of approximately 8 mg/ml by using MSD Tris Lysis buffer with protease and phosphatase inhibitors (see above), and aliquoted in plastic microtubes.
  • BCA Bicinchoninic Acid
  • TGF- ⁇ concentration in normalized tumor lysates was measured according to the manufacturer's instructions using a human TGF- ⁇ kit (MSD, K151 IUC-2) employing electrochemiluminescence assay.
  • MSD human TGF- ⁇ kit
  • Recombinant mouse TGF- ⁇ R&D Systems, Cat. #7666-MB-005
  • MSD Lysis Buffer was used as a calibrator.
  • Samples were loaded on the plate in duplicates.
  • Electrochemiluminescence signal was measured using MESO SECTOR S 600 plate reader (MSD), and the TGF- ⁇ 1 concentration in the samples was quantified using MSD Discovery Workbench software v4.0 based on the standard curve.
  • mice injected with isotype control had a median of 21 .4 pg/mg total protein, the corresponding levels in mice injected with Ab1 was undetectable (FIG. 8).
  • TGF- ⁇ levels ranged from about 7 to 25 pg/mg.
  • TGF- ⁇ levels ranged from about 1 pg/mg to as high as 43 pg/mg.
  • This Example describes the studies that characterized Abl 's pharmacokinetic (PK) profile and compared it to that of fresolimumab.
  • PK pharmacokinetic
  • Five groups of cannulated Sprague-Dawley rats were given a single dose of Ab1 or fresolimumab at 5 mg/kg intravenously. Each group had five females and five males.
  • Blood from the rats were collected at 0.25, 6, 24, 48, 72, 144, 192, and 240 hours post-dosing.
  • Ab1 and fresolimumab serum concentrations were determined by ELISA. Comparability was determined if the 90% confidence intervals for the AUC ratio (of test material to reference) was within the range of 80% to 125%.
  • Ab1 also had linear PK behavior in monkeys and displayed higher exposure after both single and repeated dosings than did fresolimumab at both 1 mg/kg and 10 mg/kg per dose.
  • Ab1 had a half-life of 13 days, whereas fresolimumab had a half-life of 4.5 days;
  • Ab1 had a CL of about 0.40 ml/hr/kg, whereas fresolimumab had a CL of 0.66 ml/hr/kg.
  • the monkey study also showed Ab1 had about 1 .7-folder higher exposure than
  • NOAEL no observed adverse effect level
  • STD10 a severely toxic dose that causes death or irreversible severe toxicity in 10% of animals
  • Toxicity included heart valve proliferation characterized by multiple thickened nodules; and abnormal lung conditions such as mixed cell alveolar exudate, mixed cell perivascular infiltrates, hypertrophy of the muscular arteries, hemorrhage, and/or increased lung weight.
  • NOAEL and HNSTD i.e. , the highest nonseverely toxic dose above which lethality, life-threatening toxicities, or irreversible toxicities occur
  • doses for monkeys in weekly repeat dosing for 5 weeks were found to be 10 mg/kg/dose (cf.
  • fresolimumab whose NOAEL in monkeys was shown to be 1 mg/kg when administered biweekly for 7 or 13 doses, or Q3D for 4 weeks). See also the data shown in Table 9 below.
  • LD Lethal Dose 50 mg/kg/dose 1 > 10 mg/kg/dose
  • HNLD Highest Non-Lethal dose
  • Ab1 can be administered to human patients safely at a dosage level of about 0.05 mg/kg to 0.5 mg/kg weekly or less frequently, e.g., biweekly.
  • mice deficient in the ⁇ 2- microglobulin gene and therefore lacking CD8 + cytotoxic T cell responses were implanted with B16-F10 in the footpad and treated as before.
  • 1 D1 1 had no effect on the number of metastases in the draining lymph node in these mice.
  • TGF- ⁇ mediated immune suppression in melanoma may contribute to innate resistance.
  • TGF- ⁇ induced gene expression changes were quenchable by 1 D1 1 treatment, confirming the specificity of TGF- ⁇ activation signature.
  • mesenchymal tumors e.g., CRC, HCC, head and neck squamous cell carcinoma, and ovarian cancer
  • mesenchymal tumors also were enriched for both TGF- ⁇ activation and predicted anti-PD-1 resistance.
  • This finding was consistent with the role of TGF- ⁇ signaling in EMT. Therefore, we arrived at our gateway indication 2: Mesenchymal tumors, especially those with immune infiltration, may benefit from anti-TGF- ⁇ and anti- PD-1 combination therapy.
  • Machine learning methods were used to identify from over 30 EMT marker genes a smaller number of genes, for example, ACTA2, VIM, MGP, ZEB2, and ZWINT, that could be used to select mesenchymal tumors.
  • TGF- ⁇ activation transcriptional signature and genes within the signature may serve as useful biomarkers for cancer patient selection at baseline for anti-TGF- ⁇ and anti-PD-1 antibody combination therapy.
  • MC38 tumor-bearing mice were treated with a single dose of either PBS or anti-PD-1 alone (5 mg/kg), or increasing doses of Ab1 (10, 25 or 50 mg/kg, i.p.) in combination with anti-PD-1 (5 mg/kg) when the volume of the tumors was from 61 to 1 10 mm 3 .
  • Tumors were collected 1 hour, 6 hours, 10 hours, 24 hours, 72 hours, and 168 hours after the treatment, snap frozen in 2 ml plastic tubes with 2.8 mm ceramic balls (Precyllys KT3961 -1007.2), and stored at -80°C. To prepare lysates, tumors were thawed at room temperature.
  • Tris Lysis buffer R60TX-2
  • 1x HaltTM protease and phosphatase inhibitor cocktail Thermo 78440
  • Precellys®24 Dual homogenizer Bertin Instruments
  • Lysates were cleared by centrifugation for 10 min at 20,000 x g in an Eppendorf 5417C centrifuge at 4°C.
  • Supernatants were transferred into clean chilled Eppendorf tubes and further cleared by centrifugation for another 30 min as described above.
  • MIP-2 CXCL2
  • KC/GRO CXCL1
  • MIP-2 and KC/GRO CXCL1
  • the levels of MIP-2 and KC/GRO were also evaluated in these same samples.
  • the intratumoral levels of MIP-2 were shown to increase by at least 4-fold in animals treated with Ab1 together with anti-PD-1 , compared to those in animals treated with either PBS or anti-PD-1 alone; and the elevation of MIP-2 levels was shown to persist for at least 168 hours (FIG. 10B).
  • the levels of KC/GRO were shown to increase, though at later time points of 72 and 168 hours as compared to those of MIP-2 (FIG. 10C).
  • the Ab1 and anti-PD-1 mAb combination induced a decrease in the levels of active TGF- ⁇ earlier than the increase in the levels of MIP-2 and KC/GRO.
  • TGF- ⁇ is known to impact the immune system by inhibiting the activities of different immune cell types.
  • TGF- ⁇ has been reported to inhibit natural killer (NK) cell activity and NK cell-mediated ADCC (Trotta et al., Journal of immunology (2008) 181 :3784-3792).
  • NK cells have recently been reported to form dense clusters as a mechanism to enhance their activity and activation via localization of IL-2 within these densely packed clusters (Kim et al., Scientific Reports (2017) 7:40623). Purified human NK cells cultured in vitro in the presence of IL2 are shown to form these densely packed clusters.
  • NK cells were freshly isolated from the blood of healthy donors by negative selection with the NK cell RosetteSep reagent according to the manufacturer's protocols (Stem Cell Technologies). NK cells were cultured at 1 .2x10 5 cells/well in IL-2 (100 lU/mL) supplemented Myelocult (Stem Cell Technologies) in round bottom assay plates (Costar). TGF- ⁇ was added to a final concentration of 0.1 , 1 or 10 ng/mL in the presence of either an irrelevant lgG4 or Ab1 at 100 pg/mL, as indicated. The cells were cultured for 72 hours and NK cell clustering was visualized by capturing images on a Nikon microscope.
  • TGF- ⁇ The addition of increasing doses of TGF- ⁇ was shown to inhibit NK cell clustering.
  • Ab1 but not an lgG4 control antibody, was added to the NK cell cultures, the NK cell clusters were shown to develop. This result demonstrates that TGF- ⁇ neutralization impacts NK cell activation, leading to increased activity and proliferation of NK cells to support the anti-tumor response of the immune system.
  • Example 13 Reversion of TGF- -Mediated Suppression of lFN- ⁇ Production in Proliferating CD8 + T cells by Ab1 Treatment
  • TGF- ⁇ has been reported to inhibit the activity of CD8 + T cells (Flavell et al., Nature Reviews Immunology (2010) 10:554- 567).
  • MLR mixed lymphocyte reaction
  • CD8 + cell proliferation and IFN- ⁇ production were first assessed in the presence of TGF- ⁇ .
  • CD3 + cells were isolated using EasySep T Cell enrichment kit (StemCell Technologies) from PBMCs fractionated from normal healthy donors following Ficoll gradient isolation.
  • the CD3 + cells were then labeled with CellTrace Violet according to the manufacturer's protocol (ThermoFisher).
  • An MLR assay was performed by mixing the labeled CD3 + cells (2X10 5 cells) with irradiated BLCL cells (Astarte Bio) (2X10 4 cells; 2 min) in RPMI supplemented with 10% FBS. TGF- ⁇ , lgG4 control antibody and/or Ab1 were added to the cultures, as indicated, and the cultures were incubated at 37°C with 5% C0 2 for 4 days. The cells were next stimulated for 4 hours in the presence of PMA cell stimulation cocktail (eBioscience) and protein transporter inhibitor cocktail (eBioscience).
  • Live cells were discriminated by staining with Zombie NIR viability dye (BioLegend) on ice and washed with FACs buffer. Cells were fixed with True-Nuclear buffer (BioLegend), washed, pelleted and resuspended in FACs buffer. Cells were prepared for flow cytometry by staining with BV650 Anti-huCD4, PERCP/Cy5.5 anti-huCD8, FITC anti-huCD3, and PE anti-hulFNy (BioLegend). Flow Cytometry was run on a BD Canto and results analyzed in FlowJo software and gating on live cells, singlets, and CD3 + cells.
  • Zombie NIR viability dye BioLegend
  • the percentage of I FNY + CD8 + T cells was quantitated by gating on the CD8 + cells that had proliferated based upon diminished CellTrace Violet staining and were positive for IFN- ⁇ staining. FMOs were run as controls for all antibody staining.
  • neutralization is able to impact the adaptive immune system by blocking the
  • FIG. 12A shows the relative abundance of CD8 + T cells (log2-transformed) across the compendium. Relative CD8 + T cell abundance was estimated using the unique marker gene CD8B, which has been shown to be a highly specific indicator of presence of CD8 T cells (Becht et al., Curr Opin Immunol (2016) 39:7-13; and Becht et al., Genome Biol (2016) 17:218). Each box plot summarizes the range of values across the biological replicates.
  • the MC38 model showed about 2-fold more CD8 + T cell infiltration than the EMT6 model (left and right boxes, respectively).
  • the A20 and EL4 lymphoma models displayed overall highest and lowest levels of CD8 + T cell infiltration, respectively, with negligible CD8 + T cells in EL4.
  • the MC38, MC38.ova, CT26, and L1210 murine cell lines exhibited the highest levels of the CD8 gene signature. Additionally, the EMT-6 breast cancer cell line was shown to have T cell infiltration close to baseline, which is consistent with recent reports that EMT6 tumors have an immune-excluded phenotype (S. Mariathasan et al. 2017, ESMO Immuno-Oncology Congress, Geneva, Geneva Switzerland).
  • FIG. 12B shows TGF- ⁇ pathway activation across the compendium.
  • a 170- gene transcriptional signature of TGFp pathway activation derived from in vitro stimulation of MCF7 cells by TGF- ⁇ and validated by comparison with several other TGF- ⁇ signatures, was used to assign a pathway activation score to each profile in the compendium. Scores were computed using "regulated gene set enrichment analysis” (rGSEA, Theilhaber et al. 2014), and expressed as log2 enrichments of signature genes against the gene background. While the MC38 models displayed average activation, the EMT6 model displayed very high TGF- ⁇ pathway activation (left and right boxes, respectively).
  • rGSEA regulated gene set enrichment analysis
  • EMT-6 breast cells (CRL-2755, ATCC) were cultured in RPMI-1640 supplemented with 10% FBS in a humidified 5% C0 2 incubator and then implanted subcutaneously (0.5X10 6 cells/mouse) into the flank of female BALB/c mice (Shanghai Lingchang Bio-Technology Co. Ltd, Shanghai, China). Once tumors reached an average size of 68-1 16 mm 3 , the mice were pooled and randomly distributed to control and treatment groups (10 mice per group).
  • Ab1 at a dose of 10 or 25 mg/kg Q3D and mouse a-PD-1 antibody at a dose of 5 mg/kg demonstrated partial activity with 1/10, 2/10 and 2/10 complete regression, respectively, in EMT-6 tumor-bearing mice.
  • the combination of Ab1 at a dose of 10 or 25 mg/kg Q3D and mouse a-PD-1 antibodies at 5 mg/kg Q3D was therapeutically active.
  • Table 10 is a summary of the results.
  • SEQ ID NO:3 (fresolimumab heavy chain, including leader sequence - residues 1 -19)
  • SEQ ID NO:4 (fresolimumab light chain, including leader sequence - residues 1 -19)
  • SEQ ID NO:6 anti-PD-1 Mab light chain
  • DIVMTQGTLP NPVPSGESVS ITCRSSKSLL YSDGKTYLNW YLQRPGQSPQ LLIYWMSTRA SGVSDRFSGS GSGTDFTLKI SGVEAEDVGI YYCQQGLEFP TFGGGTKLEL KRADAAPTVS IFPPSTEQLA TGGASVVCLM NNFYPRDISV KWKIDGTERR DGVLDSVTDQ DSKDSTYSMS STLSLTKADY ESHNLYTCEV VHKTSSSPVV KSFNRNEC
  • HVQLQQSGPE LVRPGASVKL SCKASGYIFI TYWMNWVKQR PGQGLEWIGQ IFPASGSTNY NEMFEGKATL TVDTSSSTAY MQLSSLTSED SAVYYCARGD GNYALDAMDY WGQGTSVTVS SAKTTPPSVY PLAPGSAAQT NSMVTLGCLV KGYFPEPVTV TWNSGSLSSG VHTFPAVLQS DLYTLSSSVT VPSSTWPSQT VTCNVAHPAS STKVDKKIVP RDCGCKPCIC TVPEVSSVFI FPPKPKDVLT ITLTPKVTCV VVDISKDDPE VQFSWFVDDV EVHTAQTKPR EEQFNSTFRS VSELPIMHQD WLNGKEFKCR VNSAAFPAPI EKTISKTKGR PKAPQVYTIP PPKEQMAKDK VSLTCMITDF FPEDITVEWQ WNGQPAENYK NTQPIMDT
  • NIVLTQSPAS LAVSLGQRAT ISCRASESVD SYGNSFMHWY QQKSGQPPKL LIYLASNLES GVPARFSGSG SRTDFTL ID PVEADDAATY YCQQNNEDPL TFGAGTKLEL KRADAAPTVS IFPPSSEQLT SGGASVVCFL NNFYPKDINV KWKIDGSERQ NGVLNSWTDQ DSKDSTYSMS STLTLTKDEY ERHNSYTCEA THKTSTSPIV KSFNRNEC

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne un anticorps pan-TGF-β amélioré pour le traitement d'états pathologiques qui sont induits par le TGF-β, notamment des maladies auto-immunes, des états pathologiques fibrotiques et des cancers. L'invention concerne également des procédés et des utilisations de l'anticorps conjointement avec d'autres agents immunomodulateurs, tels qu'un anticorps anti-PD-1.
EP18711394.9A 2017-01-20 2018-01-19 Anticorps anti-tgf-bêta et leur utilisation Pending EP3571227A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP17305061 2017-01-20
PCT/IB2018/000088 WO2018134681A1 (fr) 2017-01-20 2018-01-19 Anticorps anti-tgf-bêta et leur utilisation

Publications (1)

Publication Number Publication Date
EP3571227A1 true EP3571227A1 (fr) 2019-11-27

Family

ID=58018036

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18711394.9A Pending EP3571227A1 (fr) 2017-01-20 2018-01-19 Anticorps anti-tgf-bêta et leur utilisation

Country Status (3)

Country Link
EP (1) EP3571227A1 (fr)
EA (1) EA201991729A1 (fr)
WO (1) WO2018134681A1 (fr)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201610044D0 (en) 2016-06-08 2016-07-20 Ucb Biopharma Sprl Antibodies
CN112703038A (zh) 2018-07-10 2021-04-23 赛诺菲 靶向CD38和TGF-β的抗癌组合疗法
WO2020160291A2 (fr) 2019-01-30 2020-08-06 Scholar Rock, Inc. INHIBITEURS SPÉCIFIQUES DU COMPLEXE LTBP DE TGFβ ET LEURS UTILISATIONS
AU2021205440A1 (en) 2020-01-11 2022-09-01 Scholar Rock,Inc. TGF-beta inhibitors and use thereof
MX2022008609A (es) 2020-01-11 2022-11-10 Scholar Rock Inc Inhibidores de tgf¿ y uso de los mismos.
WO2022204581A2 (fr) 2021-03-26 2022-09-29 Scholar Rock, Inc. Inhibiteurs de tgf-bêta et leur utilisation
TW202304979A (zh) * 2021-04-07 2023-02-01 瑞士商諾華公司 抗TGFβ抗體及其他治療劑用於治療增殖性疾病之用途
WO2022256723A2 (fr) 2021-06-03 2022-12-08 Scholar Rock, Inc. Inhibiteurs de tgf-bêta et leur utilisation thérapeutique
EP4370148A1 (fr) 2021-07-14 2024-05-22 Scholar Rock, Inc. Inhibiteurs spécifiques du complexe ltbp de tgf bêta1 et leurs utilisations

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004098637A1 (fr) 2003-04-30 2004-11-18 Genzyme Corporation Antagonistes du tgf-beta combine avec des antagonistes du systeme renine-angiotensine-aldosterone pour le traitement de l'insuffisance renale
CN104151427B (zh) 2005-02-08 2017-09-08 根茨美公司 针对TGFβ的抗体
RU2625034C2 (ru) 2011-04-20 2017-07-11 МЕДИММЬЮН, ЭлЭлСи Антитела и другие молекулы, которые связывают в7-н1 и pd-1
DK3312195T3 (da) * 2013-03-20 2020-01-06 Genzyme Corp Fremgangsmåder til behandling af osteogenesis imperfecta
HRP20221262T1 (hr) 2013-09-13 2022-12-09 Beigene Switzerland Gmbh Protutijela anti-pd1 i njihova uporaba kao terapeutskih i dijagnostičkih sredstava
TWI681969B (zh) 2014-01-23 2020-01-11 美商再生元醫藥公司 針對pd-1的人類抗體
JOP20200094A1 (ar) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc جزيئات جسم مضاد لـ pd-1 واستخداماتها
MA40035A (fr) * 2014-10-14 2016-04-21 Dana Farber Cancer Inst Inc Molécules d'anticorps de pd-l1 et leurs utilisations
TWI595006B (zh) 2014-12-09 2017-08-11 禮納特神經系統科學公司 抗pd-1抗體類和使用彼等之方法
JP6901400B2 (ja) * 2015-04-03 2021-07-14 ゾーマ テクノロジー リミテッド TGF−β及びPD−1の阻害物質を使用する癌の治療法

Also Published As

Publication number Publication date
WO2018134681A1 (fr) 2018-07-26
WO2018134681A8 (fr) 2019-10-10
EA201991729A1 (ru) 2019-12-30

Similar Documents

Publication Publication Date Title
US11242384B2 (en) Anti-TGF-beta antibodies and their use
WO2018134681A1 (fr) Anticorps anti-tgf-bêta et leur utilisation
US20230052212A1 (en) Fgfr2 inhibitors alone or in combination with immune stimulating agents in cancer treatment
US20230340613A1 (en) Methods for monitoring and treating cancer
CA2966507A1 (fr) Procede et biomarqueurs permettant de predire l'efficacite et d'evaluer un traitement par l'agoniste ox40
CA2988420A1 (fr) Methodes destinees a traiter le cancer a l'aide d'anticorps anti-ox40 et d'antagonistes se liant a l'axe pd-1
KR20170004006A (ko) 암을 치료하기 위한 항-ccr4 항체 및 4-1bb 효능제의 조합
CA2934028A1 (fr) Polytherapie comprenant des agonistes se liant a ox40 et des antagonistes se liant a l'axe pd-1
KR20160108566A (ko) 암을 치료하기 위한 pd-1 길항제 및 vegfr 억제제의 조합
CA3032437A1 (fr) Anticorps anti-siglec-7 pour le traitement du cancer
JP2019515670A (ja) がんをモニタリングし治療するための方法
CA3107773A1 (fr) Anticorps agonistes anti-cd226
JP2023502023A (ja) Cd200受容体アンタゴニスト結合分子
US20220378909A1 (en) Methods of Treating Cancer with Anti-PD-1 Antibodies
EP4087873A1 (fr) Anticorps anti-galectine-9 et ses utilisations
KR102568885B1 (ko) 항-tgf-베타 항체 및 이의 용도
NZ796440A (en) Anti-TGF-beta antibodies and their use
EA041300B1 (ru) Антитела к tgf-бета и их применение

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20190819

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RIN1 Information on inventor provided before grant (corrected)

Inventor name: FINN, PATRICK

Inventor name: YU, MARCELLA

Inventor name: KODURI, RAO

Inventor name: QIU, HUAWEI

Inventor name: MANKOO, PARMINDER

Inventor name: WINTER, CHRISTOPHER

Inventor name: SHAPIRO, GARY

Inventor name: POLLARD, JACK, R.

Inventor name: MALKOVA, NATALIA

Inventor name: BROWER, KEVIN

Inventor name: LIU, FENG

Inventor name: THEILHABER, JOACHIM

Inventor name: GREGORY, RICHARD, C.

RAV Requested validation state of the european patent: fee paid

Extension state: TN

Effective date: 20190819

Extension state: MA

Effective date: 20190819

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40009678

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20200831

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS