NZ764590B2 - Anti-galectin-9 antibodies and uses thereof - Google Patents

Anti-galectin-9 antibodies and uses thereof Download PDF

Info

Publication number
NZ764590B2
NZ764590B2 NZ764590A NZ76459018A NZ764590B2 NZ 764590 B2 NZ764590 B2 NZ 764590B2 NZ 764590 A NZ764590 A NZ 764590A NZ 76459018 A NZ76459018 A NZ 76459018A NZ 764590 B2 NZ764590 B2 NZ 764590B2
Authority
NZ
New Zealand
Prior art keywords
seq
antibody
galectin
chain variable
cdr2
Prior art date
Application number
NZ764590A
Other versions
NZ764590A (en
Inventor
Joseph Bolen
Linxiao Chen
Eric Elenko
Aleksandra Filipovic
Akiko Koide
Shohei Koide
George Miller
Original Assignee
New York University
PureTech Health LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by New York University, PureTech Health LLC filed Critical New York University
Priority claimed from PCT/US2018/058028 external-priority patent/WO2019084553A1/en
Publication of NZ764590A publication Critical patent/NZ764590A/en
Publication of NZ764590B2 publication Critical patent/NZ764590B2/en

Links

Classifications

    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation
    • 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
    • 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
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]

Abstract

Disclosed herein are anti-Galectin-9 antibodies and methods of using such for inhibiting a signaling pathway mediated by Galectin-9 or eliminating pathologic cells expressing Galectin-9. Such anti-Galectin-9 antibodies may also be used to diagnose and/or to treat diseases associated with Galectin-9, such as autoimmune diseases and solid tumors. such as autoimmune diseases and solid tumors.

Description

ANTI-GALECTIN-9 ANTIBODIES AND USES THEREOF BACKGROUND OF INVENTION Immune checkpoint blockade has demonstrated unprecedented success in the past few years as cancer ent. Often antibodies are used to block immune inhibitory pathways, such as the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed death I (PD-I) pathways. While therapies targeting those two pathways have shown success in treating l cancer types, anti-CTLA-4 and anti-PD-I therapies have a response rate of 10 to 60% of treated IO patients, depending on cancer type, and have not yet shown the y to exceed a response rate of 60%, even when used in combination (Kyvistborg et al., Enhancing ses to cancer immunotherapy; Science. 2018 Feb 2;359(6375):516-517). Additionally, a large number of cancer types are refractory to these therapies. As part of efforts to improve existing immunotherapies in the clinic, the field has started to focus on the role of abnormalities in interferon signaling and upregulation of alternative checkpoints as potential causes for the limitation of current therapies. One such ial alternate oint is T-cell globulin mucin-3 (Tim-3) /Galectin-9 (e.g., reviewed in Yang and Hung; The role of T-cell immunoglobulin mucin-3 and its ligand galectin-9 in antitumor immunity and cancer immunotherapy; Cancer biology and cancer ent; Oct 2017, Vol.60 No.IO: 064, and references therein).
Galectin-9 is a tandem-repeat lectin consisting of two carbohydrate recognition domains (CRDs) and was discovered and described for the first time in 1997 in patients suffering from Hodgkin's ma (HL) (Tureci et al., J. Biol. Chem. 1997, 272, 6416-6422). Three isoforms exist, and can be located within the cell or extracellularly. Elevated Galectin-9 levels have been in observed a wide range of cancers, including melanoma, Hodgkin's lymphoma, hepatocellular, pancreatic, gastric, colon and clear cell renal cell cancers (Wdowiak et al. Int. J. Mol. Sci. 2018, 19, 210). In renal cancer, patients with high Galectin-9 expression showed more advanced progression of the disease with larger tumor size and necrosis (Kawashima et al.; Bru Int. 2014;113:320-332). In melanoma-a cancer considered as one of the most lethal cancers due to its aggressive metastasis and resistance to therapy - Galectin-9 was expressed in 57% of tumors and was significantly increased in the plasma of patients with advanced melanoma ed to y controls (Enninga et al., Melanoma Res. 2016 Oct; 26(5): 429-441). A number of studies have shown utility for Gal-9 as a stic marker, and more recently as a potential - I - new drug target (Enninga et al., 2016, Kawashima et al. BJU Int 2014, 113: 320—332, ita et al., Int J Cancer. 2002 Jun 20,99(6):809-16, and references therein). Galectin-9 has been described to play an important role in in a number of cellular processes such as on, cancer cell aggregation, sis, and chemotaXis. Recent studies have shown a role for Galectin-9 in immune modulation in support of the tumor, e.g., h negative regulation of Th1 type responses, Th2 polarization and polarization of macrophages to the M2 phenotype. This work also includes studies that have shown that Galectin-9 participates in direct inactivation of T cells through interactions with the T-cell immunoglobulin and mucin n 3 (T11V1-3) receptor (Dardalhon et al., JImmunol, 2010, 185, 1383-1392, Sanchez-Fueyo et al., Nat Immunol, 2003, 4, 1093-1101). Galectin-9 has also been found to play a role in polarizing T cell entiation into tumor suppressive phenotypes), as well as promoting tolerogenic macrophage programming and adaptive immune suppression (Daley et al., Nat Med, 2017, 23, 556-567). In mouse models of pancreatic ductal adenocarcinoma (PDA), blockade of the checkpoint ction between Galectin-9 and the receptor Dectin-1 found on innate immune cells in the tumor microenvironment (TME) has been shown to increase anti-tumor immune responses in the TME and to slow tumor progression (Daley et al., NaZMed, 2017, 23, 556-567). Galectin-9 also has been found to bind to CD206, a surface marker ofM2 type macrophages, resulting in a reduced secretion of CVL22 (MDC), a macrophage derived chemokine which has been associated with longer survival and lower recurrence risk in lung cancer (Enninga et al, J Pathol. 2018 Aug,245(4):468-477).
Accordingly, modulating the ty of Galectin-9 and/or one or more of its ors may e a novel cancer therapy approach, alone or in combination with eXisting therapies. bed herein are novel human antibodies which bind to human Galectin-9 and their therapeutic use in the treatment of cancer.
SUMMARY OF INVENTION The present disclosure is based, at least in part, on the development of anti-Galectin-9 antibodies that potently suppress signaling triggered by Galectin-9. Such antibodies are capable of suppressing Galectin-9 signaling and/or eliminating Galectin-9 positive pathologic cells, thereby benef1ting treatment of diseases associated with in-9.
Accordingly, one aspect of the present disclosure provides an isolated anti-Galectin-9 dy, which binds to an epitope in a carbohydrate recognition domain (CRD) of a Galectin-9 polypeptide, for example, a human Galectin-9 polypeptide. In some embodiments, the anti- in-9 antibody described herein may bind to both a human Galactin-9 polypeptide and a non-human Galactin-9 polypeptide (e.g., a mouse Galactin-9, a rat Galactin-9, or a primate in-9). In some ments, the anti-Galactin-9 antibody binds exclusively to one of the Galectin-9 CRDs. In some embodiments, the anti-Galactin-9 antibody binds to both of the in-9 CRDs, e.g., with similar or ent affinities. In some embodiments, the anti- Galectin-9 antibody disclosed herein binds an epitope within the CRDl region. In some embodiments, the anti-Galectin-9 antibody disclosed herein binds an epitope within the CRDl region, which CRDl region may have the amino acid sequence of SEQ ID NO: 3. In some embodiments, the anti-Galectin-9 antibody disclosed herein binds an epitope within the CRDl region having the amino acid sequence of SEQ ID NO: 3. In some embodiments, the anti- Galectin-9 antibody binds to the same epitope as a reference antibody selected from the group ting of G9.l-1, G9.l-2, G9.l-3, G9.l-4, G9.l-5, , G9.l-7, G9.1-8, G9.1-8m1, G9.l- 8m2, G9.l-8m3, G9.1-8m4, m5, G9.1-8m6, G9.1-8m7, G9.1-8m8, G9.l-8m9, G9.l- 8m10, G9.1-8m11, G9.1-8m12, G9.l-8m13, G9.l-8m14, G9.l-9, G9.l-10, and G9.l-11 antibodies, and/or competes against the reference antibody from binding to the CRDl region. In some embodiments, the anti-Galectin-9 antibody binds to the same epitope as antibody G9. 1-8 or antibody G9. 1-8m13 and/or competes against antibody G9. 1-8 or antibody G9.l-8m13 from binding to the CRDl region.
In some embodiments, the anti-Galectin-9 antibody disclosed herein is an antibody 2O selected from the group consisting of , G9.l-2, G9.l-3, G9.l-4, G9.l-5, G9.l-6, G9.l-7, G9.1-8, G9.1-8m1, G9.1-8m2, G9.1-8m3,G9.l-8m4,G9.1-8m5,G9.1-8m6,G9.1-8m7,G9.1- 8m8,G9.l-8m9,G9.l-8m10,G9.l-8m11,G9.l-8m12,G9.1-8m13,G9.l-8m14,G9.1-9,G9.1- , and G9.l-11 antibodies. In some embodiments, the anti-Galectin-9 antibody is G9.l-8 antibody. In some embodiments, the dy is G9. 1-8m13 antibody. In some examples, the anti-Galectin-9 antibody may se the same heavy chain mentarity determining regions (CDRs) and the same light chain CDRs as the reference antibody, e.g., any of the nce antibodies provided herein. In one specific example, the anti-Galectin-9 antibody comprises the same heavy chain variable region and the same light chain variable region as the reference antibody, e.g., any of the reference antibodies provided above and elsewhere . 3O In some embodiments, the anti-Galectin-9 antibody has a VL sequence comprising SEQ ID NO: 21 or ting essentially of SEQ ID NO: 21 or consisting of SEQ ID NO: 21. In some embodiments, the anti-Galectin-9 antibody has a VH sequence comprising SEQ ID NO: 86 or consisting ially of SEQ ID NO: 86 or consisting of SEQ ID NO: 86. In some embodiments, the anti-Galectin-9antibody has a VL sequence comprising SEQ ID NO: 21 and a VH sequence comprising SEQ ID NO: 86. In some embodiments, the alectin-9antibody has a VL sequence consisting essentially of SEQ ID NO: 21 and a VH sequence consisting essentially of SEQ ID NO: 86. In some embodiments, the anti-Galectin-9antibody has a VL sequence consisting of SEQ ID NO: 21 and a VH sequence consisting of SEQ ID NO: 86.
In some embodiments, the anti-Galectin antibody has a VH sequence comprising SEQ ID NO: 22 or consisting essentially of SEQ ID NO: 22 or consisting of SEQ ID NO: 22. In some embodiments, the anti-Galectin-9 antibody has a VL sequence sing SEQ ID NO: 21 and a VH sequence comprising SEQ ID NO: 22. In some embodiments, the anti-Galectin-9antibody has a VL sequence consisting essentially of SEQ ID NO: 21 and a VH sequence consisting essentially of SEQ ID NO: 22. In some embodiments, the anti-Galectin-9antibody has a VL ce consisting of SEQ ID NO: 21 and a VH sequence consisting of SEQ ID NO: 22.
In some embodiments, the anti-Galectin-9 antibody has a VL sequence sing one or more of the sequences set forth in SEQ ID NOs: 328, 329, and 337. In some embodiments, the anti-Galectin-9 antibody has a VH sequence comprising one or more of the sequences set forth in SEQ ID NOs: 361, 364, 374, 366, and 383. In some ments, the anti-Galectin-9 dy has a VL ce comprising one or more of the sequences set forth in SEQ ID NOs: 328, 329, and 337, and a VH sequence comprising one or more of the sequences set forth in SEQ ID NOs: 361, 364, and 374. In some embodiments, the anti-Galectin-9 antibody has a VL sequence sing one or more of the sequences set forth in SEQ ID NOs: 328, 329, and 337, and a VH 2O sequence comprising one or more of the sequences set forth in SEQ ID NOs: 361, 366, and 383.
In some embodiments, the anti-Galectin-9 antibody disclosed herein binds an epitope within the Galectin-9 CRD2 region. In some embodiments, the anti-Galectin-9 dy disclosed herein binds an epitope within the Galectin-9 CRD2 region, which CRD2 region may have the amino acid sequence of SEQ ID NO: 4. In some embodiments, the anti-Galectin-9 antibody disclosed herein binds an e within the CRD2 region having the amino acid sequence of SEQ ID NO: 4. In some embodiments, the anti-Galectin-9 antibody binds an epitope within the Galectin-9 CRD2 region that comprises a phan residue corresponding with residue W309 of SEQ ID NO: 1. In some embodiments, the anti-Galectin-9 antibody binds an epitope within the in-9 CRD2 region that does not comprise one or more residues 3O corresponding with R253, R271, Y330, R334, R341 and Y236 of SEQ ID NO: 1. In some embodiments, the anti-Galectin-9 dy may bind an epitope within the Galectin-9 CRD2 region that comprises a tryptophan residue corresponding with residue W3 09 of SEQ ID NO: 1 and additionally does not comprise one or more residues corresponding to R253, R271, Y330, R334, R341 and Y236 of SEQ ID NO: 1. In some embodiments, the anti-Galectin-9 antibody binds to the same epitope as a reference antibody selected from the group consisting of G9.2-l, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, 2, 3, G9.2-l4, 5, G9.2-l6, G9.2-l7, G9.2-l7mut6, G9.2-l8, G9.2-l9, 0, G9.2- 21, G9.2-22, G9.2-23, 4, 5, G9.2-26, and G9.2-low y binder antibodies, and/or competes against the reference antibody from binding to the CRD2 region. In some embodiments, the anti-Galectin-9 antibody binds to the same e as antibody G9.2-l7 or antibody G9.2-l7mut6 and/or competes against antibody 7 or antibody G92-l7mut6 from binding to the CRD2 region. In some embodiments, the anti-Galectin-9 antibody is an antibody selected from the group consisting of G9.2-l, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12, 3, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-l7mut6, G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, and G9.2-26 antibodies. In some embodiments, the anti-Galectin-9 antibody is G9.2-l7 antibody or G9.2-l7mut6 antibody. In some embodiments, the anti-Galectin-9 antibody has a VL sequence comprising SEQ ID NO: 54 or consisting essentially of SEQ ID NO: 54 or consisting of SEQ ID NO: 54. In some embodiments, the anti-Galectin-9 antibody has a VH sequence sing SEQ ID NO: 55 or consisting essentially of SEQ ID NO: 55 or ting of SEQ ID NO: 55. In some embodiments, the anti-Galectin-9antibody has a VL sequence comprising SEQ ID NO: 54 and a VH sequence comprising SEQ ID NO: 55. In some embodiments, the anti-Galectin-9antibody has a VL sequence consisting essentially of SEQ ID NO: 54 and a VH sequence consisting 2O ially of SEQ ID NO: 55. In some ments, the anti-Galectin-9antibody has a VL ce consisting of SEQ ID NO: 54 and a VH sequence consisting of SEQ ID NO: 55. In some embodiments, the antibody has a VH sequence comprising SEQ ID NO: 56. In some embodiments, the antibody has a VH sequence consisting ially of SEQ ID NO: 56 or consisting of SEQ ID NO: 56. In some embodiments, the isolated antibody has a VL sequence comprising SEQ ID NO: 54 and a VH sequence comprising SEQ ID NO: 56. In some embodiments, the isolated dy has a VL sequence consisting essentially of SEQ ID NO: 54 and a VH sequence consisting essentially of SEQ ID NO: 56. In some embodiments, the isolated antibody has a VL sequence consisting of SEQ ID NO: 54 and a VH sequence consisting of SEQ ID NO: 56. 3O In some embodiments, the anti-Galectin-9 antibody has a VL sequence comprising one or more of the sequences set forth in SEQ ID NOs: 328, 329, and 352. In some embodiments, the anti-Galectin-9 antibody has a VH sequence comprising one or more of the sequences set forth in SEQ ID NOs: 361, 388, 406, and 407. In some embodiments, the anti-Galectin-9 antibody has a VL sequence comprising one or more of the sequences set forth in SEQ ID NOs: 328, 329, and 352, and a VH sequence comprising one or more of the sequences set forth in SEQ ID NOs: 361, 388, and 406. In some embodiments, the anti-Galectin-9 antibody has a VL sequence comprising one or more of the sequences set forth in SEQ ID NOs: 328, 329, and 352, and a VH sequence comprising one or more of the ces set forth in SEQ ID NOs: 361, 388, and 407.
In some examples, the anti-Galectin-9 antibody may comprise the same heavy chain complementarity determining regions (CDRs) and the same light chain CDRs as the reference antibody, e.g., any of the reference antibodies provided herein. In one specific e, the anti-Galectin-9 antibody comprises the same heavy chain variable region and the same light chain le region as a nce antibody, e.g., any of the reference antibodies provided herein. In some embodiments, the anti-Galectin-9 antibody comprises a heavy chain complementarity determining region 1 (CDRl), a heavy chain mentary determining region 2 (CDR2), and a heavy chain complementary determining region 3 (CDR3), which collectively are at least 90% (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) identical to the heavy chain CDRs of a reference antibody, e.g., any of the reference dies provided herein. In some embodiments, the anti-Galectin-9 antibody comprises a light chain CDRl, a light chain CDR2, and a light chain CDR3, which tively are at least 90% (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) cal to the light chain CDRs of a reference antibody, e.g., any of the reference antibodies provided herein.
In some embodiments, the anti-Galectin-9 antibody comprises both a heavy chain complementarity determining region 1 (CDRl), a heavy chain complementary determining region 2 (CDR2), and a heavy chain complementary determining region 3 (CDR3), which tively are at least 90% (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) identical to the heavy chain CDRs of a reference antibody, e.g., any of the reference antibodies provided herein and a light chain CDRl, a light chain CDR2, and a light chain CDR3, which collectively are at least 90% (e.g., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%) identical to the light chain CDRs of a reference antibody, e.g., any of the reference antibodies provided herein. In some examples, the anti-Galectin-9 antibody may comprise the same heavy chain CDRs and the same light chain CDRs as the reference antibodies noted above. In one specific example, the alectin-9 antibody may comprise the same heavy chain variable region and the same light chain variable region as of a reference antibody, e.g., any of the reference antibodies provided herein. In some embodiments, the exemplary isolated anti- in 9 antibodies which bind to CRDl include G9.1-1, G9.1-2, G9.1-3, G9.1-4, , G9.1-6, G9.1-7, G9.1-8, G9.1-9, G9.1-10, G9.1-11, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1- 8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G9.1-8m8, G9.1-8m9, G9.1-8m10, G9.1-8m11, G9.1- 8m12, G9.1-8m13, and G9.1-8m14. In some embodiments, the exemplary isolated alectin 9 antibodies which bind to CRD2 include , G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, 7mut6 , G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder.
In some embodiments, the isolated anti-Galectin 9 antibodies, or antigen binding portion f, se heavy and light chain variable regions, wherein the light chain variable region comprises an amino acid ce selected from SEQ ID NO: 29, 13, 34, 36, 38, 40, 42, 44, 46, 48, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71. In some embodiments, the light chain le regions consists of an amino acid ce selected from SEQ ID NO: 29, 13, 34, 36, 38, 40, 42, 44, 46, 48, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71. In some embodiments, the ed anti- Galectin 9 antibodies, or antigen binding portions f, comprise heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence selected from SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73. In some embodiments, the heavy chain variable regions ts of an amino acid sequence selected from SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73.
In some embodiments, the isolated anti-Galectin 9 antibodies, or antigen binding portion thereof, se heavy and light chain variable regions, wherein the light chain variable region 2O comprises an amino acid sequence selected from SEQ ID NO: 29, 13, 34, 36, 38, 40, 42, 44, 46, 48, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71, and the heavy chain variable region comprises an amino acid sequence selected from SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73. In some embodiments, the isolated alectin 9 dies, or antigen binding portion thereof, comprise heavy and light chain variable regions, wherein the light chain variable region consists of an amino acid sequence selected from SEQ ID NO: 29, 13, 34, 36, 38, 40, 42, 44, 46, 48, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71, and the heavy chain variable region consists of an amino acid sequence selected from SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73. 3O In some embodiments, the isolated anti-Galectin 9 antibodies, or antigen binding portions thereof, comprise heavy and light chain variable regions, wherein the light chain variable region comprises an amino acid sequence selected from SEQ ID NO: 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, and 27. In some embodiments, the light chain variable regions consist of an amino acid sequence selected from SEQ ID NO: 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, and 27. In some embodiments, the isolated anti-Galectin 9 antibodies, or antigen binding portions f, comprise heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence ed from SEQ ID NO: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26,28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87. In some embodiments, the heavy chain variable regions consist of an amino acid sequence selected from SEQ ID NO: 8, , 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87.
Accordingly, in some embodiments, provided herein are isolated anti-Galectin 9 antibodies, or antigen binding portions f, comprising heavy and light chain variable regions, wherein the light chain le region comprises an amino acid sequence selected from SEQ ID NO: 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, and 27 and the heavy chain variable region ses an amino acid sequence ed from SEQ ID NO: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87. In some embodiments, the light chain variable regions consists of an amino acid sequence selected from SEQ ID NO: 7, 9, 11, 13, 15, 17, 19, 21, 23, , and 27, and the heavy chain variable regions consists of an amino acid sequence selected from SEQ ID NO: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87.
In some embodiments, any of the anti-Galectin-9 antibody disclosed herein may comprise a heavy chain variable domain (VH) that is at least 85% identical to the VH of a reference antibody disclosed herein. Alternatively or in addition, the anti-Galectin-9 dy 2O may comprise a light chain variable domain (VL) that is at least 85% identical to the VL of the reference dy.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region comprising SEQ ID NO: 54. In some embodiments, the anti-Galectin-9 dy or antigen binding portion f comprises a VH region comprising SEQ ID NO: 55.
In some embodiments, the anti-Galectin-9 antibody comprises a VL region consisting of SEQ ID NO: 54. In some embodiments, the alectin-9 antibody or antigen binding portion thereof comprises a VH region consisting of SEQ ID NO: 55. In some embodiments, the anti- Galectin-9 antibody comprises a VL and VH region comprising SEQ ID NO: 54 and 55, respectively. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL and VH region consisting of SEQ ID NO: 54 and 55, respectively. In some embodiments, the anti-Galectin-9 antibody is clone 92-17. In some embodiments, the alectin-9 antibody or antigen binding portion thereof comprises a VH region comprising SEQ ID NO: 56. In some embodiments, the alectin-9 antibody comprises a VL and VH region comprising SEQ ID NO: 54 and 56, respectively. In some specific ments, the anti-Galectin-9 antibody or antigen binding n f comprises a VL and VH region consisting of SEQ ID NO: 54 and 56, respectively. In some embodiments, the alectin-9 antibody is clone 9.2-17 mut6.
In some embodiments, the anti-Galectin-9 antibody comprises a VL region comprising SEQ ID NO: 21. In some embodiments, the anti-Galectin-9 antibody comprises a VL region consisting of SEQ ID NO: 21. In some embodiments, the anti-Galectin-9 antibody comprises a VH region comprising SEQ ID NO: 86. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VH region consisting of SEQ ID NO: 86. In some embodiments, the anti-Galectin-9 antibody comprises a VL and VH region comprising SEQ ID NO: 21 and 86, respectively. In some embodiments, the anti-Galectin-9 antibody comprises a VL and VH region consisting of SEQ ID NO: 21 and 86, respectively, In some embodiments, the anti-Galectin-9 antibody is clone G9.1-8m13. In some embodiments, the anti-Galectin-9 antibody ses a VH region comprising SEQ ID NO: 22. In some embodiments, the anti- in-9 antibody or antigen binding portion thereof comprises a VH region consisting of SEQ ID NO: 22. In some embodiments, the alectin-9 antibody comprises a VL and VH region comprising SEQ ID NO: 21 and 22, respectively. In some embodiments, the anti-Galectin-9 antibody comprises a VL and VH region consisting of SEQ ID NO: 21 and 22, respectively. In some embodiments, the anti-Galectin-9 antibody is clone G9.1-8.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion f 2O comprises a VL region which has the same amino acid ce as the VL region of antibody 9.1-8m13 (SEQ ID NO: 21). In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VH region which has the same amino acid ce as the VH region of antibody 13 (SEQ ID NO: 86). In some embodiments, the anti-Galectin-9 antibody comprises VL and VH s which have the same amino acid sequences as the VL and VH regions of antibody 9.1-8m13 (SEQ ID NO: 21 and 86, respectively).
In some embodiments, the alectin-9 antibody or antigen binding portion thereof comprises a VL region which has the same amino acid sequence as the VL region of dy 9.2-17 (SEQ ID NO: 54). In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VH region which has the same amino acid sequence as the VH region of antibody 9.2-17 (SEQ ID NO: 55). In some embodiments, the anti-Galectin-9 antibody comprises VL and VH regions which have the same amino acid sequences as the VL and VH regions of 92-17 (SEQ ID NO: 54 and 55, respectively).
In some embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence identity to a VL region set forth in SEQ ID NO: 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, and 27. In some embodiments, the anti-Galectin-9 antibody comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH region set forth in SEQ ID NOs: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87.
In some embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence ty to a VL region set forth in SEQ ID NOs: 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, and 27 and a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH region set forth in SEQ ID NOs: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87.
In some embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to a VL region set forth in SEQ ID NOs: 13, 29, 34, 36, 38, 40, 42, 44, 46, 48, 29, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71. In some ments, the anti-Galectin-9 antibody comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH region set forth in SEQ ID NOs: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73.
In some embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL region set forth in SEQ ID NOs: 13,29, 34, 36, 38, 40, 42, 44, 46, 48, 29, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71 and a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH region set forth in SEQ ID NOs: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73.
In some ments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL region set forth in SEQ ID NO: 21.
In some ments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH region set forth in SEQ ID NO: 86. In some embodiments, the alectin-9 antibody or antigen binding portion thereof comprises a VL and VH region that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL and VH regions set forth in SEQ ID NO: 21 and 86, respectively.
In some specific embodiments, the anti-Galectin-9 antibody or antigen binding fragment thereof comprises a VL that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to a VL region set forth in SEQ ID NO: 54. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding fragment thereof comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH region set forth in SEQ ID NO: 55. In some specific embodiments, the anti-Galectin-9 antibody or antigen g fragment thereof comprises a VL and/or VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to a VL and/or VH region set forth in SEQ ID NO: 54 and 55, respectively. In some c embodiments, the anti-Galectin-9 dy comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL region of G9. l-8ml3. In some specific ments, the anti-Galectin-9 antibody comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the VH region of G9.l-8ml3. In some specific embodiments, the anti-Galectin-9 dy comprises VL and VH regions that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to VL and VH regions of G9. l-8ml3. In some specific embodiments, the anti-Galectin-9 antibody ses a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to the VL region of G9.2-l7. In some specific embodiments, the anti-Galectin-9 antibody comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH region of 7. In some specific embodiments, the anti- Galectin-9 antibody comprises VL and VH regions that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to VL and VH regions of G9.2-l7.
Accordingly, in some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise (a) VL CDRl amino acid ce set forth in SEQ ID NO: 328, (b) VL CDR2 amino acid sequence set forth in SEQ ID NO: 329, (c) VL CDR3 amino acid sequence selected from SEQ ID NO: 0, (d) VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 427, 428, 431, 435, 436, 437, (d) VH CDR2s amino acid sequence selected from SEQ ID NO: 362, 363, 387-3 89 and 446-466,(e) VH CDR3 amino acid sequence selected from SEQ ID NO: 390-417. Accordingly, in some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise (a) VL CDRl amino acid sequence set forth in SEQ ID NO: 328, (b) VL CDR2 amino acid ce set forth in SEQ ID NO: 329, (c) VL CDR3 amino acid sequence selected from SEQ ID NO: 330-340, (d) VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 424-434, (e) VH CDR2 amino acid sequence selected from SEQ ID NO: 362-366 and 43 8-445, (f) VH CDR3 amino acid sequence selected from SEQ ID NO: 367-3 86.
In some ments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl ses SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR2 comprises XIX2X3X4XSSX6X7XSSYADSVKG (SEQ ID NO: 467), in which X1 = Y or S, X2=IorS,X3=YorS,X4=PorS,X5=YorS,X6=GorS,X7=YorS,andX8=TorS. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR3 comprises X1SX2X3X4X5X6X7X8X9XIOKXI1X12X13GMDY(SEQ ID NO: 468), in which X1 = Y or S, X2 = T, S, or absent, X3 = Y, S, or absent, X4 = S or , X5 = W, S, or , X6 = S or absent, X7 = G, S, or absent, X8 = G, T, S, or absent, X9 = I, Y, S, or absent, X10 = G, S, or Y, X11= W or S, X12 = V or S, and X13 = W or S. In some embodiments, the alectin-9 antibody or 2O binding n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl consists of SEQ ID NO: 328. In some ments, the alectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR2 consists of X1X2X3X4X5SX6X7XSSYADSVKG (SEQ ID NO: 467), inwhichX1=YorS, X2=IorS, X3=Yor S, X4=P orS, X5=YorS, X6=GorS, X7 = Y or S, and X8 = T or S. In some embodiments, the anti-Galectin-9 dy or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR3 consists of X1SX2X3X4X5X6X7X8X9X10KX11X12X13GMDY (SEQ ID NO: 468), in which X1 = Y or S, X2 = T, S, or absent, X3 = Y, S, or absent, X4 = S or absent, X5 = W, S, or absent, X6 = S or absent, X7 = G, S, or absent, X8 = G, T, S, or absent, X9 = I, Y, S, or absent, X10 = G, S, or Y, X11 = W or S, X12 = V or S, and X13 = W or S.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl comprises SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR2 comprises SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR3 comprises SEQ ID NO: 337. In some embodiments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl consists of SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR2 consists of SEQ ID NO: 329. In some ments, the anti-Galectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the light chain variable region CDR3 consists of SEQ ID NO: 337. In some embodiments, the anti-Galectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the light chain le region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 337, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, respectively. In some embodiments, the antibody ses the same VL CDRs as G9. l-8ml3. In some ments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the heavy chain le region CDRl comprises SEQ ID NO: 361. In some embodiments, the alectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein 2O the heavy chain variable region CDR2 comprises SEQ ID NO: 366. In some embodiments, the alectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDR3 region comprises SEQ ID NO: 383. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain le s, wherein the heavy chain variable region CDRl consists of SEQ ID NO: 361. In some embodiments, the anti-Galectin-9 antibody or g n thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDR2 consists of SEQ ID NO: 366. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDR3 region consists of SEQ ID NO: 383. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 361, 366, and 383, tively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 366, and 383, respectively. In some embodiments, the anti-Galectin-9 antibody comprises the same VH CDRs as G9. l-8ml3. In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 366, and 383, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 s consist of SEQ ID NO: 328, 329, and 337, respectively, and SEQ ID NO: 361, 366, and 383, respectively. In one specific embodiment, the anti-Galectin-9 antibody comprises the same VL and VH CDRs as G9.l-8ml3.
In some ments, the anti-Galectin-9 dy or binding portion f comprises heavy and light chain variable regions, wherein the light chain variable region CDRl comprises SEQ ID NO: 328. In some embodiments, the alectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR2 comprises SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR3 comprises SEQ ID NO: 352. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl ts of SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR2 consists of SEQ ID NO: 329. In some 2O ments, the anti-Galectin-9 dy or binding portion thereof consists of heavy and light chain variable regions, wherein the light chain variable region CDR3 comprises SEQ ID NO: 352. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable s, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 352, tively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 352, respectively. In some embodiments, the anti-Galectin-9 dy comprises the same VL CDRs as G9.2-l7. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 361, 388, and 406, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 388, and 406, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-l7. In some embodiments, the anti- Galectin-9 antibody or binding n thereof comprises heavy and light chain le regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 352, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 388, and 406, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 s consist of SEQ ID NO: 328, 329, and 352, respectively, and SEQ ID NO: 361, 388, and 406, respectively. In one specific embodiment, the alectin-9 antibody comprises the same VL and VH CDRs as G9.2-l7.
Accordingly, in some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise (a) VL CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl amino acid sequence set forth in SEQ ID NO: 328, (b) VL CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the VL CDR2 amino acid sequence set forth in SEQ ID NO: 329, (c) VL CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to a VL CDR3 amino acid sequence selected from SEQ ID NO: 330-340, (d) VH CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH CDRl amino acid sequence set forth in SEQ ID NO: SEQ ID NO: 361, 427, 428, 431, 435, 436, 437, (e) VH CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to a VH CDR2 amino acid sequence selected from SEQ ID NO: 362-366 and 43 8-445, (f) VH CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) ce identity to a VH CDR3 amino acid sequence ed from SEQ ID NO: 367-3 86. Accordingly, in some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise (a) VL CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the VL CDRl amino acid sequence set forth in SEQ ID NO: 328, (b) VL CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDR2 amino acid sequence set forth in SEQ ID NO: 329, (c) VL CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDR3 amino acid ce selected from SEQ ID NO: 341-360, (d) VH CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 424-434, (d) VH CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to a VH CDR2 amino acid sequence selected from SEQ ID NO: 362, 363, 387-3 89 and 446-466,(e) VH CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH CDR3 amino acid sequence selected from SEQ ID NO: 390-417.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 328, 329, and 337, respectively. In some embodiments, the dy VL CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl, CDR2, and CDR3 amino acid sequences of G9. 1- 8ml3. In some embodiments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, n the heavy chain variable region CDRl, CDR2, and CDR3 amino acid ces have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 361, 366, and 383, respectively. In some embodiments, the antibody VH CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH CDRl, CDR2, and CDR3 amino acid sequences of G9. 1- 8ml3. In one specific ment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain le region CDRl, CDR2, and CDR3 amino acid ces set forth in SEQ ID NO: 361, 366, and 383, respectively. In one specific embodiment, the antibody VL CDRl, CDR2, and CDR3 and VH CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl, CDR2, and CDR3 and VH CDRl, CDR2, and CDR3 amino acid sequences as G9. . In some embodiments, the anti-Galectin-9 dy or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 328, 329, and 352, respectively. In some embodiments, the antibody VL CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl, CDR2, and CDR3 amino acid sequences -l7. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain le region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 361, 388, and 406, respectively. In some embodiments, the antibody VH CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to the VH CDRl, CDR2, and CDR3 amino acid sequences of G9.2-l7. In some embodiments, the alectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in comprise SEQ ID NO: 328, 329, and 352, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain le region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 361, 388, and 406, respectively. In one specific embodiment, the antibody VL CDRl, CDR2, and CDR3 and VH CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl, CDR2, and CDR3 and VH CDRl, CDR2, and CDR3 amino acid sequences of G9.2-l7.
In some embodiments of any of the anti-Galectin dies provided herein, the heavy chain constant region of the anti-Galectin-9 antibody is from a human IgG (a gamma heavy chain) of any IgG subfamily as described herein, e.g., IgGl or IgG4.
In some embodiments, the amino acid sequences of exemplary anti-Galectin antibody light chains pond to sequences set forth in SEQ ID NO: 88-98 and SEQ ID NO: 99-115.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain ce of SEQ ID NO: 108. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 95 (or their variable regions). In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 108 (or their variable regions). In some embodiments, the amino acid sequences of exemplary anti-Galectin antibody heavy chains correspond to sequences set forth in SEQ ID NO: 116-140, 169-193, 222-246, 275-299 (anti-Galectin-9 antibodies binding to CRDl) and SEQ ID NO: 141-168, 194-220, 4, 300-327 (anti- Galectin-9 antibodies binding to CRD2). In some embodiments, the heavy chain constant region of the anti-Galectin-9 antibody is from a human IgG1. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 136. In some embodiments, the IgGl is a mutant with minimal Fc receptor ment. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 189. In some embodiments, the heavy chain constant region of the anti-Galectin-9 antibody is from a human IgG4. In some embodiments, the anti- Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 242. In some embodiments, the IgG4 is IgG4 ge mutant. In some embodiments, the anti-Galectin-9 antibodies or n-binding n thereof comprise a heavy chain ce of SEQ ID NO: 295.
In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 157. In some embodiments, the anti-Galectin- 9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 210. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 263. In some embodiments, the anti- Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 316.
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 136 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent n) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 189 (or its variable region). In some ments, heavy chains of anti-Galectin-9 antibodies se an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 242 (or its variable region). In some embodiments, heavy chains of anti- Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence ty to the heavy chain sequence set forth in SEQ ID NO: 295 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 157 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 210 (or its variable ). In some ments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 263 (or its variable region). In some ments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to the heavy chain sequence set forth in SEQ ID NO: 316 (or its variable ). In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 95 and a heavy chain sequence of SEQ ID NO: 136. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a light chain sequence of SEQ ID NO: 95 and a heavy chain sequence of SEQ ID NO: 189. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 95 and a heavy chain sequence of SEQ ID NO: 242.
In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 95 and a heavy chain sequence of SEQ ID NO: 295. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 108 and a heavy chain sequence of SEQ ID NO: 157.
In some ments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 108 and a heavy chain sequence of SEQ ID NO: 210.
In some ments, the anti-Galectin-9 antibodies or antigen-binding n thereof se a light chain sequence of SEQ ID NO: 108 and a heavy chain sequence of SEQ ID NO: 263.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 108 and a heavy chain sequence of SEQ ID NO: 316.
In one embodiment, the anti-Galectin-9 antibody comprises a light chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 95 and a heavy chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to SEQ ID NO: 136. In one embodiment, the anti-Galectin-9 antibody comprises a light chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 95 and a heavy chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to SEQ ID NO: 189. In one embodiment, the anti-Galectin-9 antibody comprises a light chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 95 and a heavy chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to SEQ ID NO: 242. In one embodiment, the anti-Galectin-9 antibody comprises a light chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to SEQ ID NO: 95 and a heavy chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 295.
In one embodiment, the alectin-9 antibody comprises a light chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 108 and a heavy chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to f SEQ ID NO: 157 .
In one embodiment, the anti-Galectin-9 antibody comprises a light chain amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 108 and a heavy chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to SEQ ID NO: 210 . In one embodiment, the anti- Galectin-9 antibody comprises a light chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 108 and a heavy chain amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 263. In one ment, the anti-Galectin-9 antibody comprises a light chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 108 and a heavy chain amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 316.
Any of the anti-Galectin-9 antibodies provided herein may comprise a heavy chain variable region framework of VH 3-48, and/or a light chain variable region framework of VK l- 39. In some embodiments, any of the VH and/or VL frameworks described herein are ne VH and/or VL genes. In some embodiments, the anti-Galectin-9 antibodies described herein is a full-length antibody (e.g., an IgG molecule) or an antigen-binding fragment thereof. In some examples, the antibody is a Fab or a single-chain antibody. In any ces, the dy can be a human antibody or a humanized antibody.
In another aspect, the present disclosure provides an isolated nucleic acid or set of nucleic acids which encode or collectively encode any of the anti-Galectin-9 antibodies sed herein. In some instances, the heavy chain and light chain of the antibody are encoded by two separate nucleic acid les (a set of nucleic acids). In other instances, the heavy chain and light chain of the antibody are encoded by one nucleic acid molecule, which may be in multicistronic format, or under the control of distinct promoters. In some embodiments, the nucleic acid or set of nucleic acids are located on one or two vectors. In some es, the one or two vectors can be one or two expression vectors. Further, the present disclosure provides a host cell comprising any of the isolated c acid or set of nucleic acids coding for the anti- Galectin-9 antibodies described herein.
Also provided herein is a method for producing the anti-Galectin-9 antibody, comprising culturing the host cell bed herein under suitable conditions ng for expressing of the antibody, and harvesting the antibody thus produced from the cell e (e.g., from the culture medium).
Further, the present disclosure provides a pharmaceutical composition, comprising any of the anti-Galectin-9 antibodies or a nucleic acid(s) encoding such, and a pharmaceutically acceptable carrier.
In yet r aspect, the present disclosure features a method of inhibiting Galectin mediated cell signaling in a subject, the method comprising administering to a subject in need f an effective amount of an anti-Galectin-9 antibody or a pharmaceutical composition sing an anti-Galectin-9 antibody. In some embodiments, the anti-Galectin-9 antibody is any of the alectin-9 antibodies disclosed herein or a pharmaceutical composition comprising such. In some embodiments, the subject in need thereof is a human patient having, suspected of having, or at risk for having, an autoimmune disease, a solid cancer, a microbial disease, a hematological malignancy, or an allergic disorder. Exemplary autoimmune diseases include, but are not limited to, a rheumatoid condition (e.g., rheumatoid tis), an autoimmune respiratory disease, an autoimmune metabolic and/or endocrine disorder (e.g., type I diabetes), or a f1brotic condition. Exemplary solid tumors include, but are not limited to, atic ductal adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, cholangiocarcinoma, breast cancer, small cell and non small cell lung cancer, upper and lower gastrointestinal malignancies, gastric cancer, squamous cell head and neck cancer, urinary , hepatocellular carcinoma, ovarian cancer, sarcomas, mesothelioma, glioblastoma, esophageal cancer, bladder cancer, urothelial cancer, renal cancer, al and endometrial cancer. Exemplary logical malignancies include, but are not limited to, acute lymphoblastic leukemia, chronic lymphocytic leukemia, lymphomas, multiple myeloma, and acute myelogenous leukemia, c myelogenous leukemia, myelodysplastic syndromes, or roliferative neoplasms and other myeloproliferative and ysplastic disorders. In some examples, the effective amount of the pharmaceutical composition is sufficient to block interaction between in-9 and Dectin-l. In some embodiments, the effective amount of the pharmaceutical composition is sufficient to block interaction between Galectin-9 and CD206.
Alternatively, or in addition, but not limited to, the effective amount of the pharmaceutical composition is sufficient to block interaction between Galactin-9 and Tim-3.
Further, the present disclosure provides a method for modifying, eliminating and/or reducing pathologic cells expressing Galectin-9 (e.g., via antibody-dependent cell cytotoxicity or ADCC), the method comprising administering to a subject having pathologic cells expressing Galectin-9 an effective amount of an anti-Galectin-9 dy, such as any of the anti-Galectin-9 antibodies described herein, or a ceutical composition thereof. In some embodiments, the subject is a human patient having cancer cells expressing Galectin-9 and/or pathologic immune cells expressing Galectin-9. In some embodiments, the effective amount of the ceutical composition is sufficient to te antibody-dependent cell cytotoxicity (ADCC) and/or block against pathologic cells expressing Galectin-9.
Any of the treatment methods described herein may further comprise administering to the subject an inhibitor of a checkpoint molecule, an tor of a co-stimulatory receptor, or an inhibitor of an innate immune cell target. Examples of checkpoint molecules include, but are not limited to, PD-l, PD-Ll, PD-L2, CTLA-4, LAG3, THVI-3 and A2aR. Examples of co- stimulatory receptors include, but are not limited to, 0X40, GITR, CD137, CD40, CD27, and ICOS. es of innate immune cell s include, but are not limited to, KIR, NKG2A, CD96, TLR, and IDO.
The present disclosure also provides pharmaceutical compositions for use in treating a disease associated with Galectin-9 (e.g., those described herein), wherein the pharmaceutical composition comprises an anti-Galectin-9 antibody, such as any of the alectin-9 antibodies bed , or a c acid(s) encoding such antibody, and a pharmaceutically acceptable carrier. Also, the present disclosure provides uses of the anti-Galectin-9 antibodies or the encoding nucleic acids for cturing a medicament for use in treating the target diseases as described herein.
The details of one or more embodiments of the invention are set forth in the description below. Other features or advantages of the present invention will be apparent from the following drawing and detailed description of several ments, and also from the appended claims. 2O BRIEF DESCRIPTION OF DRAWINGS The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present disclosure, which can be better understood by reference to the drawing in combination with the detailed description of specific embodiments presented herein.
Figs. 1A-IB e charts showing a binding characterization of Fabs for Galectin-9 CRD2 using phage ELISAs. Fig. 1A: binding to human and mouse Galectin-9 shown by phage ELISA. Fig. 1B: affinity of Fabs clones to Galectin-9 CRD2 determined by competition phage ELISA.
Figs. 2A-ZB include charts g a binding terization of Fabs for Galectin-9 CRDl using phage ELISA. Fig. 2A: binding of Fab clones to human and mouse Galectin-9 CRDl shown by phage ELISA. Fig. 2B: affinity of Fabs clones to Galectin-9 CRDl determined by competition phage ELISA.
Figs. 3A-3B e charts g epitope binning of G.9-2 Fab clones (binding to CRD2) using competition phage ELISA. Fig. 3A: mouse Galectin-9 CRD2-coated wells pre- incubated with d G9.2-l or G9.2-3 Fabs prior to on of phage-displayed Galectin-9 CRD2 binding Fab clones. Fig. 3B: human Galectin-9 CRD2-coated wells pre-incubated with purified G9.2-15 or G9.2-l7 Fabs prior to addition of phage-displayed Galectin-9 CRD2 binding Fab clones.
Fig. 4 includes ms showing the affinity of purified G9.2 Fabs to Galectin-9 CRD2, characterized using a bead-based binding assay. The curves show the best fit of the one-to-one binding model. Top left: G9.2-l Fab. Top right: G9.2-3 Fab. Bottom left: G9.2-15 Fab.
Bottom right: G9.2-l7 Fab. Apparent Kd values are shown in the table.
Fig. 5 includes ms showing the affinity of purified G9.l Fabs to Galectin-9 CRDl, characterized using a bead-based binding assay. Experiments were performed in the same manner as in Fig. 4. Top left: G9.l-6 Fab. Top right: G9.l-5 Fab. Bottom left: G9.l-8 Fab.
Bottom right: G9. l-ll Fab. Apparent Kd values are shown in the table.
Fig. 6 includes diagrams showing a surface plasmon resonance analysis of Fab G9.2-15 and Fab G92. 17 binding to CRD2 of human (top) and mouse (bottom) in-9. The binding and dissociation phases of the experiments are marked in the top panels. Left: G9.2-15 Fab.
Right: G9.2-l7 Fab.
Fig. 7 includes diagrams showing an SPR analysis of G9.2-l7 human IgG4 binding to CRD2 of human (top) and mouse (bottom) in-9. The gray lines show the sensorgrams for the nding negative control, G9.2-iso human IgG4. 2O Fig. 8 includes diagrams showing the staining of cell line samples with Fabs for Galectin-9 CRD2. Histograms for flow cytometry data are shown. Top left: G9.2-l Fab. Top right: G9.2-3 Fab. Bottom left: G9.2-15 Fab. Bottom right: 7 Fab.
Fig. 9 is a chart showing the inhibitory effects of G9.2-l7 and G9. 1-8 on Galectin-9 mediated activation of Dectin-l signaling.
Figs. 10A-10B include diagrams showing epitope mapping of G.9-2. 17 on human Galectin-9 CRD2 by systematic mutagenesis. Fig. 10A: A diagram showing the binding activity of G9.2-l7 to in-9 CRD2 mutants as determined by phage ELISA. The ion in ELISA signal indicates a site on the Galectin-9 CRD2 that is critical to G9.2-l7 binding. Fig. 10B: a diagram depicting the location of W309 as mapped on the crystal structure of human 3O in-9 CRD2 (PDB ID 3NV2), which is opposite to the g site of the sugar ligand as mapped on the crystal structure (W3 09 corresponds with W277 in UniProt ID 000182-2; PDB ID 3NV2).
Fig. 11 contains charts showing size-exclusion chromatography es of Fab G9.2-l7 (top), Fab G9.2-l7mut6 (middle) and Fab G9.2-Iso (bottom). Purified Fab samples were run on TOSOH TSK ist G2WXL Column in PBS and detected using absorbance at 280 nm.
Fig. 12 contains charts showing surface plasmon resonance analyses of Fab G9.2-l7 (top) and Fab G92. l7mut6 (bottom) binding to the CRD2 of human (left) and mouse (right) Galectin-9. Human or mouse Galectin-9 CRD2 was immobilized on an Avicap chip preloaded with neutravidin on a Pall ForteBio r instrument. Fab samples were then flowed using the OneStep method. The binding and iation phases of the experiments are marked in the top panels.
Fig. 13 is a graph showing a g characterization of G92 Fab clone for wild-type in-9 CRD2 or the W3 039K mutant using phage ELISA. Binding of Fab clones to human Galectin-9 CRD2 assayed using phage ELISA. Either biotinylated wild type human Galectin-9 CRD2, the W309K Galectin-9 CRD2 mutant, or Galectin-9 CRD2 pre-incubated with G9.2-l7 IgG was immobilized to neutravidin-coated wells and incubated with individual phage-displayed Fab clones.
Fig. 14 is a Kaplan—Meier plot showing that blocking Galectin-9 results in significant extension of al in animal models of pancreatic cancer (KPC mice).
Fig. 15 is a photograph of mouse tumors showing that blocking galectin-9 and anti-PDl generates a superior response. 2O Fig. 16 is a bar graph showing the tumor mass of mice treated with G9.2-l7 mIgGl.
Mice (n=lO/group) with orthotopically implanted KPC tumors were treated with commercial isotype (200ug) or commercial dGal9 (200ug) mAb or G9.2-Iso mIgGl (200ug) or G9.2-l7 mIgGl at two doses (200ug or 400ug) once weekly for three weeks. Tumors were removed and weighed, and subsequently processed and d for flow cytometry.
Figs. 1 7 depicts a bar graph showing tumor weight of mice treated with G9.2-l7 mIgG2a alone or in combination with dPDl mAb. Mice (n=lO/group) with orthotopically implanted KPC tumors were treated with commercial dPD-l (200ug) mAb or G9.2-l7 mIg2a (200ug), or a combination of G9.2-l7 and dPD-l, or matched e once weekly for three weeks. Tumors were removed and weighed and subsequently processed and stained for flow cytometry. Each point represents one mouse; >“p<0.05; >“"‘p<0.01; >“""l‘p<0.001; >“""k’l‘IKOOOOl; by unpaired Student’s .
Figs. 18A-18C depict graphs showing binding of purified G9.l-8ml-5 mIgGl to human Galectin-9 CRDl as characterized using a bead-based binding assay.
Figs. 19A-19G depict graphs g binding of purified G9. l-8m6-ll Fabs to human Galectin-9 CRDl as characterized using a bead-based binding assay.
Figs. 20A-20C depict graphs showing the y of purified G9.l-8m8, 9, and 11 mIgG2a antibodies to human Galectin-9 CRDl as characterized using a ased binding assay.
Figs. 21A-21D depict graphs showing binding of purified G9. l-8ml l-l4 Fabs to human Galectin-9 CRDl as characterized using a bead-based binding assay.
Figs. 22A-22D depict graphs showing binding of purified G9. l-8ml2-l4 mIgG2a antibodies to human in-9 CRDl as characterized using a bead-based binding assay.
Figs. 23A and 23B depict graphs showing the results of an apoptosis assay demonstrating that Gal-9 antibodies inhibit in-9 induced apoptosis of Jurkat cells. Jurkat cells were treated with or without in-9 (280 nM), G9.2-l7 IgG (1 uM), and/or G9. l-8ml3 IgG (1 uM) for 6 hours (Fig. 23A).Cells were then stained with annexin-V and PI followed by flow cytometry analysis. AnneXinV positive cells represent cells in both early and late stage apoptosis. Bars represent e of three replicates, represented as individual data points.
Statistical analysis performed by unpaired Student’s I—test. (*p<0.05, >“"‘p<0.01, >“""l‘p<0.001, ****p<0.000l).
Fig. 24 depicts a graph showing the readout of assays demonstrating anti-Galectin-9 antibodies disclosed herein disrupt the interaction between Galectin-9 and CD206. Fig. 24A 2O depicts a graph showing an ELISA measuring the interaction between lized human Galectin-9 and soluble CD206 in the absence and presence of the addition of G9. l-8ml3, or 7 antibody. Isotype antibody wells serves as control. Galectin-9 coated wells were incubated with CD206 with or t G9. l-8ml3, G9.2-l7, a combination of both antibodies, or an isotype. (Experiments performed in cate, 05, >“*p<O.Ol, >“""“p<0.001, >“""“"‘p<0.0001, by ed Student’s t-test). These results te that both G9. l-8ml3 and G9.2-l7 antibodies inhibit the interaction between Galectin-9 and CD206 and their effects are additive.
Fig. 25 depicts a line graph showing binding of purified G9.l-8ml2-l4 mIgG2a antibodies to human Galectin-9 CRD2 as compared to G9. 18 (WT) as characterized using a bead-based binding assay.
Figs. 26A and 26B depict bar graphs showing INF-alpha (Fig. 26A) and IFNgamma (Fig. 26B) expression in CD3+ T cells in pancreatic arcinoma primary tumor sample patient-derived organotypic tumor spheroids (PDOTs) treated with 92-17 IgG4 (100 nM) as compared to isotype control (100 nM).
Figs. 27A - 27C depict bar graphs showing CD44 (Fig. 27A), TNF-alpha (Fig. 27B) and IFNgamma (Fig. 27C) sion in CD3+ T cells in atic adenocarcinoma y tumor sample patient-derived organotypic tumor spheroids (PDOTS) treated with 92-17 IgGl (100 nM) or 92-17 IgG4 (100 nM) as compared to IgGl or IgG4 isotype control (100 nM).
Figs. 28A — 28F depict bar graphs showing immune profile expression in a Gall Bladder Cancer tumor sample (PDOTS) treated with 92-17 IgG4 (100 nM) as compared to IgG4 isotype control (100 nM); CD44 in CD3+ T cells (Fig. 28A), TNF-alpha in CD3+ T cells (Fig. 28B), CD44 in CD4+ T cells (Fig. 28C), TNF-alpha in CD4+ T cells (Fig. 28D), CD44 in CD8+ T cells (Fig. 28E), pha in CD8+ T cells (Fig. 28F).
Figs. 29A - 29C depict bar graphs showing CD44 (Fig. 29A), TNF-alpha (Fig. 29B) and IFNgamma (Fig. 29C) expression in CD3+ T cells in a sample of liver metastasis from a colorectal cancer patient (PDOTs) treated with 92-17 IgGl (100 nM) or 92-17 IgG4 (100 nM) as ed to IgGl (100 nM) or untreated control (Utx).
Fig. 30 depicts a line graph showing the effect of 92-17 in a Bl6FlO aneous syngeneic model. Tumors were engrafted subcutaneously and treated with G9.2-l7 IgGl mouse mAb. Animals were dosed on day 0 and day 4 intravenously (i.v.) unless otherwise specified in the legend.
Fig. 31 depicts a line graph showing the effect of 92-17 in a Bl6FlO subcutaneous syngeneic model. Tumors were engrafted subcutaneously and treated with G9.2-l7 IgG2a 2O mouse mAb. Animals were dosed on day 0 and once every 4 days thereafter until the end of the experiment. mAbs were administered i.v. unless otherwise specified in the legend.
Fig. 32 depicts a graph showing a cell based binding assay CRL-2l34 cell lines were incubated with a biotinylated Fab, and bound Fab was detected using neutravidin conjugated with DyLight 650. Samples were then analyzed using flow cytometry. Strong s were observed for the in-9 antibody 92-17, but not for the isotype controls. The KD (nM) values for the Gal-9 antibodies in the two formats were as follows: G9.2-l7 hIgGl: 0.41:0.07; G9.2-l7 mIgGl: 2.91:0.66.
Fig. 33A and 33B depict graphs showing a thermal stability determination of anti- Galectin-9 antibodies. The first derivative of the fluorescence emission plotted as a function of temperature T). The g temperature is represented as the temperature at which a peak is observed. Thermal transition was determined using change in binding of fluorophor SYPRO Orange (ThermoFisher) using a real-time PCR ment with a heating rate of 1°C per minute, essentially following a method as described in Vedadi et al., Chemical screening methods to identify s that promote protein stability, protein crystallization, and structure determination; Proc Natl Acad Sci U S A. 2006 Oct 24,103(43):15835-40.
ED DESCRIPTION OF INVENTION Provided herein are dies capable of binding to Galectin-9 (e.g., human, mouse, or both). In some embodiments, the anti-Galectin-9 antibodies bind to one or more epitopes in the CRDl and/or CRD2 domains. Such anti-Galectin-9 antibodies are capable of ssing the signaling mediated by Galectin-9 (e.g., the signaling pathway mediated by in-9/Dectin-l or Galectin-9/Tim-3) or eliminating ogic cells expressing Galectin-9 via, e.g., ADCC.
Accordingly, the anti-Galectin-9 antibodies described herein can be used for inhibiting any of the Galectin-9 signaling and/or eliminating in-9 positive pathologic cells, thereby benefiting treatment of diseases associated with Galectin-9, for example, mune diseases, solid tumors, allergic disorders, or hematological disorders such as hematological malignancies.
Galectin-9, a tandem-repeat lectin, is a beta-galactoside-binding protein, which has been shown to have a role in modulating cell-cell and cell-matrix interactions. It is found to be strongly overexpressed in n’s disease tissue and in other ogic states. It may also be found circulating in the tumor microenvironment (TME).
Galectin-9 is found to interact with Dectin-l, an innate immune receptor which is highly expressed on macrophages in PDA, as well as on cancer cells (Daley D, et al. Dectin 1 activation on macrophages by galectin 9 promotes pancreatic carcinoma and peritumoral immune nce, Nat Med. 2017,23(5):556-6). Regardless of the source of Galectin-9, disruption of its interaction with Dectin-l has been shown to lead to the reprogramming of CD4+ and CD8+ cells into indispensable mediators of umor immunity. Thus, Galectin-9 serves as a valuable therapeutic target for blocking the signaling mediated by Dectin-l. Accordingly, in some embodiments, the anti-Galectin-9 antibodies describe herein t the interaction between Galectin-9 and Dectin-l.
Galectin-9 is also found to interact with THVI-3, a type I cell e glycoprotein expressed on the surface of leukemic stem cells in all varieties of acute myeloid leukemia (except for M3 (acute promyelocytic leukemia)), but not expressed in normal human hematopoietic stem cells (HSCs). TIM-3 signaling resulting from Galectin-9 ligation has been found to have a pleiotropic effect on immune cells, inducing apoptosis in Th1 cells (Zhu et al., Nat Immunol, 2005, 6: 1245-1252) and stimulating the secretion of tumor necrosis factor-0t (TNF-d), leading to the maturation of monocytes into tic cells, resulting in inflammation by innate immunity (Kuchroo et al., Nat Rev Immunol, 2008, 8:577-5 80). Further Galectin- 9/TIM-3 signaling has been found to co-activate NF-KB and B-catenin signaling, two pathways that promote LSC self-renewal (Kikushige et al., Cell Stem Cell, 2015, 17(3):341-3 52). An anti- Galectin-9 antibody that interferes with Galectin-9/T11Vl-3 binding could have a therapeutic effect, especially with respect to leukemia and other hematological malignancies. ingly, in some embodiments, the anti-Galectin-9 antibodies described herein disrupt the interaction between Galectin-9 and T11V1-3.
Galectin-9 is also found to interact with CD206, a mannose receptor highly expressed on M2 polarized macrophages, thereby promoting tumor survival (Enninga et al., positive myeloid cells bind galectin-9 and promote a tumor-supportive microenvironment. J Pathol. 2018 Aug,245(4):468-477). Tumor-associated macrophages expressing CD206 are ors of tumor immunosuppression, angiogenesis, metastasis, and e (see, e.g., Scodeller et al., Precision Targeting of Tumor Macrophages with a CD206 Binding Peptide. M1 and M2 had been described as the functional states of macrophages, Sci Rep. 2017 Nov 7,7(1): 14655, and references therein). Specifically, M1 (also termed classically activated macrophages) are d by Thl-related cytokines and bacterial products, eXpress high levels of IL-12, and are cidal. By contrast, M2 (so-called atively activated hages) are ted by Th2-related factors, eXpress high level of anti-inflammatory cytokines, such as 1L-10, and facilitate tumor progression s and Mantovani, Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm, Nat Immunol. 2010 Oct, 11(10):889-96). The pro- tumoral effects of M2 include the promotion of angiogenesis, ement of invasion and metastasis, and the protection of the tumor cells from chemotherapy-induced apoptosis (Hu et al., Functional icance of hages in pancreatic cancer biology,Tumour Biol. 2015 Dec, 36(12): 9119—9126, and references therein). Tumor-associated macrophages are thought be of M2-like phenotype and have a protumor role. Galectin-9 has been shown to mediate myeloid cell differentiation toward an M2 ype (Enninga et al., Galectin-9 modulates immunity by promoting Th2/MZ entiation and impacts survival in patients with metastatic melanoma, Melanoma Res. 2016 Oct,26(5):429-41). It is possible that Galectin-9 binding CD206 may result in reprogramming TAMs towards the M2 phenotype, similar to what has been usly shown for Dectin. Without wishing to be bound by theory, blocking the interaction of Galectin-9 with CD206 may provide one mechamism by which an anti-Galectin antibody, e.g., es described herein in Table 1 and Table 2, such as antibody 9.1-8m13 and/or antibody 9.2-17, can be therapeutically benef1cial. Accordingly, in some embodiments, the anti-Galectin-9 dies described herein disrupt the interaction between Galectin-9 and CD206.
Galectin-9 has also been shown to interact with protein disulf1de isomerase (PDI) and 4- lBB (Bi S, et al. in-9 binding to cell e protein disulf1de ase regulates the redox nment to enhance T-cell migration and HIV entry; Proc Natl Acad Sci U S A. 201 1,108(26):10650-5, Madireddi et al. Galectin-9 controls the therapeutic activity of 4-lBB- targeting antibodies. J Exp Med. 2014,211(7):l433-48).
Anti-Galectin-9 dies can serve as therapeutic agents for treating diseases associated with Galectin-9 (e.g., those in which a Galectin-9 signaling plays a role). Without being bound by theory, an anti-Galectin-9 antibody may block a signaling pathway mediated by Galectin-9. For example, the antibody may interfere with the interaction between Galectin-9 and its binding partner (e.g., Dectin-l, TIM-3 or CD206), thereby blocking the signaling triggered by the Galectin-9/Ligand ction. Alternatively, or in addition, an anti-Galectin-9 antibody may also exert its therapeutic effect by inducing blockade and/or cytotoxicity, for example, ADCC, CDC, or ADCP against pathologic cells that express Galectin-9. A pathologic cell refers to a cell that contributes to the initiation and/or development of a disease, either directly or indirectly.
Accordingly, described herein are anti-Galectin-9 antibodies and therapeutic uses thereof for treating diseases associated with Galectin-9.
Antibodies Binding to Galectin-9 2O The present sure provides antibodies that bind Galectin-9, for example, human and/or mouse Galectin-9.
In some instances, the alectin antibody described herein binds to an epitope in a carbohydrate recognition domain (CRD) of in-9, e.g., CRDl or CRD2. . In some instances, the anti-Galectin antibody may bind to CRDl and CRD2. Galectin-9 is a protein well known in the art. For example, NCBI GenBank Accession Nos. BAB83625.1 and NP_O34838.2 provide information for human and mouse Galectin-l, respectively. Provided herein are exemplary human and mouse in-9 polypeptides, Human galectin-9 (isoform 1, aka “long,”) is provided as SEQ ID NO: 1, human CRDl and CRD2 are provided herein as SEQ ID NO: 3 and SEQ ID NO: 4, respectively, mouse galectin-9 (isoform 1, aka “long,”) is 3O provided as SEQ ID NO: 2, human and mouse CRDl and CRD2 are provided herein as SEQ ID NO: 5 and SEQ ID NO: 6, respectively.
The CRDl domain of human in-9 (SEQ ID NO: 3) encompasses residues 1-148 of SEQ ID NO: 1, and the CRD2 domain (SEQ ID NO: 4) spans residues 218-355 of SEQ ID NO: 1. Similarly, the CRDl domain of murine in-9 (SEQ ID NO: 5) spans residues 1-147 of SEQ ID NO:2, and the CRD2 domain (SEQ ID NO: 6) spans residues 226-353 of SEQ ID NO: Galectin-9 polypeptides from other species are known in the art and can be obtained from publicly available gene database, for example, GenBank, using either the human ce or the mouse sequence as a query. The CRDl and CRD2 domains of a Galectin-9 ptide can be identified by aligning the sequence of that in-9 polypeptide with that of the human or mouse Galectin-9 as described herein.
The antibodies described herein bind Galectin-9 or a fragment thereof (e.g., CRDl or CRD2). As used herein, the term “anti-Galectin-9 antibody” refers to any antibody capable of binding to a Galectin-9 polypeptide, which can be of a suitable source, for example, human or a non-human mammal (e. g., mouse, rat, rabbit, primate such as monkey, etc.). In some ments, the anti-Galectin-9 antibody can be used therapeutically to suppress the bioactivity of Galectin-9. In some embodiments, the anti-Galectin-9 dy may be used in research or may be used in diagnostic/prognostic methods, e.g., for the detection of cells expressing Galectin-9 in an assessment of treatment eligibility and/or efficacy. Alternatively, or in addition, an anti-Galectin-9 antibody may block the interaction between Galactin-9 and its ligand (e. g., Dectin-l, THVI-3), thereby suppressing the signaling pathway triggered by, for example, the Galactin-9/Dectin-l or Galectin-9/TIM-3 interaction. An anti-Galectin-9 antibody may also elicit the death of cells expressing Galectin-9, for example, through an dy- ent cellular cytotoxicity (ADCC) mechanism.
An dy changeably used in plural form) is an globulin le capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, ptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule. As used herein, the term “antibody”, e.g., anti-Galectin-9 antibody, encompasses not only intact (e.g., full-length) polyclonal or monoclonal antibodies, but also antigen-binding fragments thereof (such as Fab, Fab', F(ab')2, Fv), single chain (scFv), mutants thereof, fusion ns sing an antibody portion, humanized antibodies, chimeric antibodies, diabodies, nanobodies, linear antibodies, single chain dies, multispecific antibodies (e.g., bispecific antibodies) and any other d configuration of the 3O immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies. An antibody, e.g., anti-Galectin-9 antibody, includes an antibody of any class, such as IgD, IgE, IgG, IgA, or IgM (or sub-class f), and the antibody need not be of any particular class. Depending on the antibody amino acid sequence of the constant domain of its heavy chains, immunoglobulins can be assigned to different classes.
There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2. The chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
A typical antibody molecule comprises a heavy chain variable region (VH) and a light chain le region (VL), which are usually involved in antigen g. The VH and VL regions can be further subdivided into regions of hypervariability, also known as “complementarity determining regions” (“CDR”), interspersed with s that are more conserved, which are known as “framework regions” (“FR”). Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to y-terminus in the following order: FRl, CDRl, FR2, CDR2, FR3, CDR3, FR4. The extent of the framework region and CDRs can be precisely identified using methodology known in the art, for example, by the Kabat definition, the Chothia definition, the AbM def1nition, and/or the contact definition, all of which are well known in the art. See, e.g., Kabat, EA, et a]. (1991) Sequences of Proteins ofImmunological Interest, Fifth Edition, US. ment of Health and Human Services, NIH Publication No. 91-3242, Chothia et al., (1989) Nature 342:877, Chothia, C. et al. (1987) J. Mol. Biol. 196:901-917, Al-lazikani et al (1997) J. Molec. Biol. 273:927-948, and o, J. Mol. it. -143 (2004). See also hgmp.mrc.ac.uk and bioinf.org.uk/abs).
The anti-Galectin-9 antibody described herein may be a full-length antibody, which contains two heavy chains and two light chains, each including a variable domain and a constant domain. Alternatively, the anti-Galectin-9 antibody can be an antigen-binding fragment of a full-length antibody. Examples of binding fragments encompassed within the term “antigen- binding fragment” of a full length dy include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains, (ii) a F(ab')2 fragment, a bivalent fragment ing two Fab fragments linked by a disulfide bridge at the hinge region, (iii) a Fd fragment consisting of the VH and CH1 domains, (iv) a Fv fragment consisting of the VL and VH s of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341 :544-546), which consists of a VH domain, and (vi) an ed mentarity determining region (CDR) that retains functionality. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent les known as single chain Fv (scFv). See e.g., Bird el al. (1988) Science 242:423-426, and Huston el al. (1988) Proc. Natl. Acad. Sci. USA 85:5879- 5883.
The anti-Galectin-9 dy as described herein, e.g., in Table 1 and/or Table 2, can bind and inhibit (e.g., reduce or eliminate) the activity of Galectin-9. In some embodiments, the anti-Galectin-9 antibody as described herein can bind and inhibit the activity of Galectin-9 by at least 30% (e.g., 31%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any ent therein). The apparent inhibition constant (KialDlD or K1,app), which provides a measure of inhibitor potency, is related to the concentration of tor required to reduce enzyme activity and is not dependent on enzyme concentrations. The inhibitory activity of an alectin-9 antibody described herein can be determined by routine methods known in the art.
The K1,aplD value of an antibody may be determined by measuring the inhibitory effect of different concentrations of the antibody on the extent of the reaction (e.g., enzyme activity), fitting the change in pseudo-first order rate constant (v) as a function of inhibitor tration to the modified on equation (Equation 1) yields an estimate of the apparent Ki value. For a itive inhibitor, the Ki?1le can be obtained from the y-intercept extracted from a linear regression analysis of a plot of Krapp versus substrate concentration.
([El [1] KIWN ([El [1] Kiapp)2+4[El-Kapp v : A - (Equation 1) Where A is equivalent to vo/E, the initial velocity (v0) of the enzymatic reaction in the absence of inhibitor (1) divided by the total enzyme concentration (E). In some embodiments, the anti-Galectin-9 dy described herein may have a Kiapp value of 1000, 900, 800, 700, 600, 500, 400, 300, 200, 100, 50, 40, 30, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5 pM or less for the target antigen or antigen epitope. In some ments, the anti-Galectin-9 antibody may have a lower Ki?1le for a first target (e.g., the CRD2 of Galectin-9) ve to a second target (e.g., CRDl of Galectin-9). Differences in Kiapp (e.g., for specificity or other comparisons) can be at least 1.5, 2, 3, 4, 5, 10, 15,20, 37.5, 50, 70, 80, 91, 100, 500, 1000, ,000 or 105 fold. In some examples, the anti-Galectin-9 antibody inhibits a first antigen (e.g., a first protein in a first conformation or mimic thereof) greater ve to a second antigen (e.g., the same first protein in a second conformation or mimic thereof; or a second n). In some embodiments, any of the anti-Galectin-9 antibodies may be further aff1nity matured to reduce the Ki?1le of the antibody to the target antigen or antigenic epitope thereof.
In some embodiments, the anti-Galectin-9 antibody suppresses the -l signaling, e.g., in tumor inf11trating immune cells, such as macrophages. In some embodiments, the anti- Galectin-9 antibody suppresses the -l signaling triggered by in-9 by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or r, including any increment therein). Such inhibitory activity can be determined by conventional methods or the assays described herein, for example, Example 2. Alternatively or in addition, the anti-Galectin-9 antibody may suppress the T cell immunoglobulin mucin-3 (TIM-3) signaling initiated by in-9. In some embodiments, the anti-Galectin-9 antibody suppresses the T cell globulin mucin-3 (TIM-3) signaling, e.g., in tumor inflltrating immune cells, e.g., in some embodiments by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any ent therein). Such inhibitory activity can be determined by conventional methods or the assays described herein, for example, e 2.
In some embodiments, the anti-Galectin-9 antibody suppresses the CD206 signaling, e.g., in tumor inf11trating immune cells. In some embodiments, the anti-Galectin-9 antibody suppresses the CD206 signaling red by Galectin-9 by at least 30% (e.g., 3 1%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein). Such inhibitory activity can be determined by conventional methods or the assays bed herein, for example, Example 13. In some embodiments, the anti-Galectin-9 antibody blocks or prevents binding of Galectin-9 to CD206 by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, ing any increment therein). Such inhibitory activity can be determined by conventional s or the assays described herein, for example, Example 13.
In some embodiments, any of the anti-Galectin-9 antibodies described herein induce cell cytotoxicity, such as ADCC, in target cells expressing Galectin-9, e.g., wherein the target cells are cancer cells or immune suppressive immune cells. In some embodiments, the anti-Galectin-9 antibody induces apoptosis in immune cells, such as T cells, or cancer cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or r, including any increment n).
Such inhibitory activity can be determined by conventional methods or the assays described herein, for example, Example 14. In some embodiments, any of the anti-Galectin-9 antibodies described herein induce cell cytotoxicity such as complement-dependent cytotoxicity (CDC) against target cells expressing Galectin-9.
Antibody-dependent cell-mediated phagocytosis (ADCP) is an important mechanism of action for antibodies that mediate part or all of their action though phagocytosis. In that case, dies mediate uptake of specific antigens by antigen ting cells. ADCP can be mediated by monocytes, macrophages, neutrophils, and dendritic cells, through FcyRIIa, FcyRI, and FcyRIIIa, of which FcyRIIa (CD32a) on hages represent the predominant y.
In some embodiments, any of the anti-Galectin-9 antibodies described herein induce cell phagocytosis of target cells, e.g., cancer cells or immune suppressive immune cells sing Galectin-9 (ADCP). In some embodiments, the anti-Galectin-9 antibody increases ytosis of target cells, e.g., cancer cells or immune suppressive immune cells, by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any ent therein).
In some ments, any of the anti-Galectin-9 antibodies described herein induce cell cytotoxicity such as complement-dependent cytotoxicity (CDC) against target cells, e.g., cancer cells or immune suppressive immune cells. In some embodiments, the anti-Galectin-9 antibody increases CDC against target cells by at least 30% (e.g., 3 1%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, any of the anti-Galectin-9 antibodies described herein induce T cell activation, e.g., in tumor infiltrating T cells, i.e., suppress Galectin-9 mediated inhibition of T cell activation, either directly or indirectly. In some ments, the anti-Galectin-9 dy promotes T cell activation by at least 30% (e.g., 3 1%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein). T cell activation can be determined by conventional methods or the assays described herein, for example, Example 6 (e.g., measurement of CD44, 0X40, ma, PD-l). In some embodiments, the anti- Galectin-9 antibody promotes CD4+ cell activation by at least 30% (e.g., 3 1%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein). In a miting example, the anti-Galectin antibody induces CD44 expression in CD4+ cells. In some embodiments, the anti-Galectin-9 antibody increases CD44 expression in CD4+ cells by at least % (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein). In a non-limiting example, the anti-Galectin dy induces IFNgamma expression in CD4+ cells. In some embodiments, the anti-Galectin-9 antibody increases IFNgamma expression in CD4+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein). In a non-limiting e, the anti-Galectin antibody induces TNFalpha expression in CD4+ cells. In some embodiments, the anti-Galectin-9 antibody increases TNFalpha expression in CD4+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the anti-Galectin-9 antibody promotes CD8+ cell activation by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater), including any increment therein). In a non-limiting example, the anti-Galectin antibody induces CD44 expression in CD8-- cells. In some embodiments, the anti-Galectin-9 antibody increases CD44 expression in CD8-- cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein). In a non-limiting example, the anti-Galectin antibody induces IFNgamma expression in CD8+ cells. In some embodiments, the alectin- 9 antibody increases IFNgamma expression in CD8+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein). In a non- ng example, the anti-Galectin antibody induces TNFalpha expression in CD8+ cells. In some embodiments, the anti-Galectin-9 antibody increases TNFalpha expression in CD8+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
The antibodies described herein can be murine, rat, human, or any other origin (including chimeric or humanized antibodies). Such dies are non-naturally occurring, 1'.e., would not be produced in an animal t human act (e.g., immunizing such an animal with a d n or fragment thereof or isolated from antibody libraries).
Any of the dies described herein, e.g., alectin-9 antibody, can be either monoclonal or polyclonal. A “monoclonal antibody” refers to a homogenous antibody population and a “polyclonal dy” refers to a heterogeneous antibody population. These two terms do not limit the source of an antibody or the manner in which it is made.
In some embodiments, the anti-Galectin-9 antibody is a humanized antibody. In some embodiments, the anti-Galectin-9 dy is a zed antibody having one of more of the elements or characteristics described below or elsewhere herein. Humanized antibodies refer to forms of non-human (e.g., murine) antibodies that are specific chimeric immunoglobulins, globulin chains, or antigen-binding fragments thereof that contain minimal sequence derived from non-human immunoglobulin. In general, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a CDR of the ent are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specif1city, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. rmore, the humanized antibody may comprise es that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance. In some instances, the humanized antibody may comprise substantially all of at least one, and typically two, variable domains, in which all or ntially all of the CDR regions correspond to those of a man immunoglobulin and all or ntially all of the FR s are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
Antibodies may have Fc regions d as described in WO 99/58572. Other forms of humanized antibodies have one or more CDRs (one, two, three, four, five, or siX) which are altered with respect to the original antibody, which are also termed one or more CDRs “derived from” one or more CDRs from the original antibody. Humanized antibodies may also involve affinity maturation.
Methods for constructing zed dies are also well known in the art. See, e. g., Queen el al, Proc. Natl. Acad. Sci. USA, 29-10033 . In one example, variable regions of VH and VL of a parent non-human antibody are subjected to three-dimensional molecular modeling analysis following methods known in the art. Next, framework amino acid residues predicted to be important for the formation of the correct CDR structures are identified using the same molecular modeling analysis. In parallel, human VH and VL chains having amino acid sequences that are homologous to those of the parent non-human antibody are identified from any antibody gene database using the parent VH and VL sequences as search queries.
Human VH and VL acceptor genes are then selected.
The CDR regions within the selected human acceptor genes can be replaced with the CDR regions from the parent non-human antibody or onal variants thereof. When necessary, residues within the framework regions of the parent chain that are predicted to be important in interacting with the CDR regions can be used to substitute for the corresponding residues in the human or genes.
In some embodiments, the anti-Galectin-9 antibody is a chimeric antibody. In some embodiments, the anti-Galectin-9 antibody is a chimeric dy which may include a heavy constant region and a light constant region from a human dy. Chimeric dies refer to antibodies having a variable region or part of variable region from a first species and a constant region from a second species. Typically, in these chimeric antibodies, the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals (e.g., a non-human mammal such as mouse, rabbit, and rat), while the constant portions are homologous to the sequences in antibodies derived from r mammal such as human. In some embodiments, amino acid modifications can be made in the variable region and/or the constant region.
In some embodiments, the anti-Galectin-9 antibodies described herein specifically bind to the corresponding target antigen or an epitope thereof, e.g., in-9 antigen or epitope. An antibody that “specifically binds” to an n or an epitope is a term well understood in the art.
A molecule is said to exhibit “specific g” if it reacts more frequently, more y, with greater duration and/or with greater affinity with a particular target antigen than it does with alternative targets. An antibody “specifically binds” to a target antigen or epitope if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances. For example, an dy that specifically (or entially) binds to an antigen (Galectin-9) or an antigenic epitope n is an antibody that binds this target antigen with r affinity, avidity, more readily, and/or with greater duration than it binds to other antigens or other epitopes in the same n. It is also understood with this definition that, for example, an antibody that specifically binds to a first target antigen may or may not specifically or preferentially bind to a second target antigen. As such, “specific binding” or “preferential binding” does not necessarily e (although it can include) ive binding. In some examples, an antibody that “specifically binds” to a target antigen or an epitope thereof may not bind to other antigens or other epitopes in the same antigen (1'. e., only baseline binding ty can be detected in a conventional method). In some embodiments, the anti-Galectin-9 antibodies described herein specifically bind to Galectin-9. In some embodiments, the anti- Galectin-9 antibodies described herein specifically bind to the CRD2 of Galectin-9. In some embodiments, the anti-Galectin-9 antibodies described herein specifically bind to the CRDl of Galectin-9. Alternatively, or in addition, the anti-Galectin-9 dy described herein specifically binds human Galectin-9 or a nt thereof as relative to the mouse counterpart, or vice versa (e.g., having a binding affinity at least 10-fold higher to one antigen than the other as determined in the same assay under the same assay conditions).
In some embodiments, the anti-Galectin -9 antibody binds only to CRDl (and not CRD2), for e, meaningful binding to CRD2 or binding to CRD2 is not detectable by a routine assay . In some embodiments, the anti-Galectin -9 or a fragment thereof binds only to CRD2 (and not CRDl). In some embodiments, certain antibodies described herein may bind to both CRDl and CRD2. In some embodiments, certain antibodies or fragments thereof described herein may bind to both CRDl and CRD2, but with a lower affinity to CRD2. In some embodiments, certain antibodies or fragments thereof described herein may bind to both CRDl and CRD2, but with a lower affinity to CRDl.
In some embodiments, the effect of a CRDl g Gal-9 antibody and a CRD2 binding Gal-9 antibody may be additive. In some embodiments, the effect of a CRDl binding Gal-9 antibody and a CRD2 binding Gal-9 antibody may be synergistic. In some embodiments, a “cocktail” i.e., a mixture of two or more dies may be used in a ition. Such compositions may comprise one or more antibodies that bind to CRDl described herein and one or more antibodies that bind to CRD2 described herein. In a non-limiting example, an antibody sing the variable region of clone 9.1-8ml3 (e.g., SEQ ID NO: 21 (light chain and SEQ ID NO: 86) can be combined with an antibody comprising the variable region of clone 9.2-17 (SEQ ID NO: 54 (light chain and SEQ ID NO: 55) in a composition. Antibodies may be mixed in equimolar amounts or in other ratios, as determined optimal for performance.
In some embodiments, an antibody might bind to both CRDl and CRD2. In other ces, the anti-Galectin-9 antibody described herein may cross-react to human and a non- human Galectin-9 (e.g., mouse), e.g., the difference in binding affinity to the human and the non-human Galectin-9 is less than 5-fold, e.g., less than 2-fold, or ntially similar.
In some embodiments, an anti-Galectin-9 antibody as described herein has a le binding affinity for the target antigen (e.g., Galectin-9) or antigenic epitopes thereof. As used herein, “binding affinity” refers to the apparent association constant or KA. The KA is the reciprocal of the dissociation constant (KB). The anti-Galectin-9 antibody described herein may have a binding affinity (KB) of at least 10'5, 10'6, 10'7, 10'8, 10'9, 10'10 M, or lower for the target antigen or antigenic epitope. An increased binding y corresponds to a sed KD.
Higher affinity binding of an antibody for a first antigen relative to a second antigen can be 2O indicated by a higher KA (or a smaller numerical value KD) for g the first antigen than the KA (or numerical value KD) for binding the second antigen. In such cases, the antibody has specificity for the first antigen (e.g., a first protein in a first conformation or mimic thereof) relative to the second antigen (e.g., the same first protein in a second conformation or mimic thereof, or a second protein). In some embodiments, the anti-Galectin-9 antibodies described herein have a higher binding affinity (a higher KA or smaller KD) to the CRDl of Galectin-9 as compared to the binding affinity to the CRD2 of in-9. In some embodiments, the anti- Galectin-9 antibodies described herein have a higher binding affinity (a higher KA or smaller KD) to the CRD2 of in-9 as compared to the binding affinity to the CRDl of Galectin-9.
Differences in binding affinity (e.g., for specificity or other comparisons) can be at least 1.5, 2, 3, 4, 5, 10, 15, 20, 37.5, 50, 70, 80, 91, 100, 500, 1000, 10,000 or 105 fold. In some embodiments, any of the anti-Galectin-9 antibodies may be further affinity matured to increase the binding affinity of the antibody to the target antigen or antigenic epitope f.
Binding affinity (or binding specificity) can be determined by a variety of methods ing brium dialysis, equilibrium binding, gel ion, ELISA, surface plasmon resonance, or spectroscopy (e.g., using a fluorescence assay). Exemplary conditions for evaluating binding affinity are in HBS-P buffer (10 mM HEPES pH7.4, 150 mM NaCl, 0.005% (v/v) Surfactant P20).
These techniques can be used to e the concentration of bound binding protein as a function of target protein concentration. Under certain conditions, the fractional concentration of bound binding protein ([Bound]/[Total]) is generally related to the concentration of total target protein ([Target]) by the following equation: [Bound]/[Total] = t]/(Kd+[Target]) It is not always necessary to make an exact determination of KA, though, since sometimes it is sufficient to obtain a quantitative measurement of y, e.g., determined using a method such as ELISA or FACS analysis, is tional to KA, and thus can be used for comparisons, such as determining whether a higher affinity is, e.g., 2-fold higher, to obtain a qualitative measurement of y, or to obtain an inference of affinity, e.g., by activity in a functional assay, e.g., an in vitro or in vivo assay. In some cases, the in vitro binding assay is indicative of in vivo activity. In other cases, the in vitro binding assay is not necessarily indicative of in vivo activity. In some cases tight binding is beneficial, but in other cases tight g may not be as desirable in vivo, and an antibody with lower binding affinity may be more desirable. A number of exemplary anti-Galectin-9 antibodies (specific to CRDl or CRD2) are provided herein.
Exemplary antibody clones (reference antibodies) of the disclosure binding to CRDl include G9.l-1,G9.1-2,G9.l-3,G9.l-4,G9.l-5,G9.1-6,G9.1-7,G9.l-8,G9.1-9,G9.1-10, 1, ml, m2, G9.l-8m3, G9.l-8m4, G9.l-8m5, m6,G9.1-8m7, G9.l- 8m8, G9.l-8m9, G9.l-8m10, G9.l-8m11, G9.l-8m12, G9.l-8m13, and G9.l-8m14.
Exemplary antibody clones (reference antibodies) of the disclosure binding to CRD2 include , G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12,G9.2-13,G9.2-14,G9.2-15,G9.2-16,G9.2-17,G9.2-17mut6,G9.2-18,G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder.
Variable regions Exemplary anti-Galectin-9 antibodies bed herein binding to CRDl are antibodies, e.g., onal, recombinant, and/or human antibodies, having the CDR and/or variable region sequences of antibodies G9.l-l, G9.l-2, G9.l-3, G9.l-4, G9.l-5, , G9.l-7, G9.l-8, G9.l- 9,G9.1-10,G9.l-11,G9.l-8m1,G9.1-8m2,G9.l-8m3,G9.l-8m4,G9.l-8m5,G9.1-8m6,G9.1- 8m7, G9.l-8m8, G9.l-8m9, G9.l-8mlO, G9.l-8mll, G9.l-8m12, G9.l-8ml3, and G9.l-8ml4.
Exemplary anti-Galectin-9 antibodies described herein binding to CRD2 are antibodies, e.g., monoclonal, recombinant, and/or human dies, having the CDR and/or variable region sequences of antibodies G9.2-l, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2- 9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-l7mut6, G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, 6, and G9.2- low y binder. Exemplary sequences and SEQ ID NOs are listed in Table l and 2. CDRs determined using the Kabat methodology are shown in boldface. Table 3 presents the CDRs, determined with Kabat methodology, of ed . Herein the terms “m” and “mut”, e.g., “9.1-8m” and “9.1-8mut” are used interchangeably. For example, the “G9.l-8ml”, “G9. l-8m2” “G9.l-8m3”, 8m4”, “G9.l-8m5”, 8m6”, “G9.l-8m7”, 8m8”, “G9.l-8m9”, “G9.l-8mlO”, “G9.l-8ml l”, “G9.l-8m12”, “G9.l-8ml3”, and 8ml4” are used interchangeably with “G9. l-8mutl”, “G9.l-8mut2”, “G9. l-8mut3”, “G9.l-8mut4”, “G9.l- 8mut5”, “G9.l-8mut6”, “G9.l-8mut7”, “G9.l-8mut8”, “G9.l-8mut9”, “G9.l-8muth”, “G9.l- 8mutl 1”, “G9. l-8mut12”, “G9.l-8mutl3”, and “G9. l-8mutl4, tively.
Table 1. Antibodies directed against CRDl LC/ IgG CDR CDR CDR VR HC 4 Clone Sequencc 1 2 3 1G1 mut SEQ ID NO: VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-1 SGSRSGTDFTLTISSLQPEDFAT YYCQQSWVGSLITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTFSSSSIHWVRQAP GKGLEWVASIYSSYGYTYYA G9.1-1 DSVKGRFTISADTSKNTAYLQ 275 MNSLRAEDTAVYYCARYYW GWSQNQGFWWYGLDYWGQ GTLVTvss VLzDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP LIYSASSLYSGVPSRF G9.1-2 SGSRSGTDFTLTISSLQPEDFAT YYCQQWQWGYSLVTFGQGT KVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTISSSSIHWVRQAP GKGLEWVASISSYYGSTYYAD G9.1-2 276 SVKGRFTISADTSKNTAYLQM NSLRAEDTAVYYCARSWSSSF WYNWALDYWGQGTLVTVSS VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-3 h—* h—* 328 329 332 90 SGSRSGTDFTLTISSLQPEDFAT YYCQQSWYSNKPITFGQGTK VEIKR VHIEVQLVESGGGLVQPGGSLR LSCAASGFTIYSSSIHWVRQAP GKGLEWVAYIYSSSGYTSYAD G9.1-3 SVKGRFTISADTSKNTAYLQM fi—* [\J 436 363 369 118 171 224 277 NSLRAEDTAVYYCARYSHSSL YYSWIWALDYWGQGTLVTVS VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-4 fi—* U.) 328 329 333 SGSRSGTDFTLTISSLQPEDFAT 91 -9 91 YYCQQSSSSLITFGQGTKVEIK R -9 VHIEVQLVESGGGLVQPGGSLR GFTIYYSSIHWVRQA WVASISSSSGSTSYA G9.1-4 DSVKGRFTISADTSKNTAYLQ ._1 .p 437 440 370 119 172 225 278 MNSLRAEDTAVYYCARSYRP YSSYYWGMDYWGQGTLVTV VLIDIQMTQSPSSLSASVGDRV SQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-5 fi—* U1 328 329 334 92 \O [\J 92 92 SGSRSGTDFTLTISSLQPEDFAT YYCQQYYGWFYPVTFGQGT KVEIKR VHIEVQLVESGGGLVQPGGSLR LSCAASGFTIYYSSIHWVRQA PGKGLEWVASISSSYGSTYYA G9.1-5 DSVKGRFTISADTSKNTAYLQ H O\ 437 441 371 120 173 226 279 MNSLRAEDTAVYYCARSVSW YPYYYYYGYGSGLDYWGQG TLVTVSS MTQSPSSLSASVGDRV SQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-6 H \l 328 329 335 -9 O U.) -9 93 SGSRSGTDFTLTISSLQPEDFAT YYCQQYHSSLFTFGQGTKVEI VHIEVQLVESGGGLVQPGGSLR LSCAASGFTLSSSSIHWVRQA PGKGLEWVASIYSSYGSTSYA G9.1-6 DSVKGRFTISADTSKNTAYLQ fi—* 00 427 442 372 121 174 227 280 MNSLRAEDTAVYYCARSSHW YMYWSYWGWYIGMDYWGQ GTLVTVSS VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-7 fi—* \O 328 329 336 94 94 94 94 SGSRSGTDFTLTISSLQPEDFAT YYCQQYPGYRGLITFGQGTK VEIKR VH:EVQLVESGGGLVQPGGSLR G9.1-7 NO L») 0\ fi—* 443 373 122 175 228 281 LSCAASGFTVSSSSIHWVRQA I J;N I PGKGLEWVASISSYYGYTYYA DSVKGRFTISADTSKNTAYLQ MNSLRAEDTAVYYCARSYSY KYYTMDYWGQGTL VTVSS VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-8 [\J H 328 329 337 95 95 95 95 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VH:EVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYPYSGYTSY G9.1-8 ADSVKGRFTISADTSKNTAYL [\J [\J 361 364 374 123 176 229 282 QMNSLRAEDTAVYYCARYST YSWGGIGKWVWGMDYWGQ GTLVTVSS VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-9 [\J U.) 328 329 338 96 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYFHKIPITFGQGTKV EIKR VHIEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYSSSGYTSYA G9.1-9 DSVKGRFTISADTSKNTAYLQ N.p 361 363 384 138 191 244 297 MNSLRAEDTAVYYCARYSSY FAMDYWGQGTLVT VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-10 U] 328 329 339 97 \O \l 97 97 SGSRSGTDFTLTISSLQPEDFAT YYCQQWYWYYPVTFGQGTK VEIKR VHIEVQLVESGGGLVQPGGSLR LSCAASGFTVSYSSIHWVRQA PGKGLEWVASIYSYYGSTYYA G9.1-10 FTISADTSKNTAYLQ [\J C\ 429 444 385 139 192 245 298 MNSLRAEDTAVYYCARGHYQ EGRKSGFSYWSPALDYWGQ GTLVTVSS VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP GKAPKLLIYSASSLYSGVPSRF G9.1-11 [\J \l 328 329 340 98 SGSRSGTDFTLTISSLQPEDFAT YYCQQTYWGLITFGQGTKVE VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVYSSSIHWVRQA PGKGLEWVASIYSYYGYTSYA 1 DSVKGRFTISADTSKNTAYLQ [\J 00 428 445 386 140 193 246 299 MNSLRAEDTAVYYCARSTEG YDRWGYYSSYWSSGLDYWG QGTLVTVSS MTQSPSSLSASVGDRV G9.1- TITCRASQSVSSAVAWYQQKP 21 328 329 337 95 95 95 95 GKAPKLLIYSASSLYSGVPSRF I J; b.) I SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVASSSSSSGYTSYA G91- DSVKGRFTISADTSKNTAYLQ 74 361 365 374 124 177 230 283 MNSLRAEDTAVYYCARYSTY SWGGIGKWVWGMDYWGQG TLVTVSS VLzDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- GKAPKLLIYSASSLYSGVPSRF [\J 1—4 328 329 337 95 -9 95 95 8m2 SGSRSGTDFTLTISSLQPEDFAT YYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYPYSSSSSYA G91- DSVKGRFTISADTSKNTAYLQ \l U1 361 366 374 125 178 231 284 MNSLRAEDTAVYYCARYSTY SWGGIGKWVWGMDYWGQG TLVTVSS VLzDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- GKAPKLLIYSASSLYSGVPSRF M1-1 328 329 337 —-—9 5 9 95 8m3 TDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYPYSGYTSY G91- ADSVKGRFTISADTSKNTAYL 0\ 361 364 375 126 179 232 285 QMNSLRAEDTAVYYCARfl SWGGIGKWVWGMDYWGQG TLVTVSS VLzDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- GKAPKLLIYSASSLYSGVPSRF M1-1 328 329 337 —9 \0 U1 —9 95 8m4 TDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA WVAYIYPYSGYTSY G91- ADSVKGRFTISADTSKNTAYL \l \l 361 364 376 127 180 233 286 QMNSLRAEDTAVYYCARY_ST SKWVWGMDYWGQG TLVTVSS VLzDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- GKAPKLLIYSASSLYSGVPSRF [\J 1—4 328 329 337 95 95 95 95 8m5 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA G91- PGKGLEWVAYIYPYSGYTSY \l 00 361 364 377 128 181 234 287 ADSVKGRFTISADTSKNTAYL QMNSLRAEDTAVYYCARYST I J;J; I YSWGGIGSSSSSMDYWGQGT LVTVSS VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- GKAPKLLIYSASSLYSGVPSRF 21 328 329 337 \0 U1 \0 U] 95 95 8m6 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA WVAYIYPYSGYTSY G91- ADSVKGRFTISADTSKNTAYL \l \0 361 364 378 129 182 235 288 QMNSLRAEDTAVYYCARY_ST YSSSSSKWVWGMDYWGQGT LVTVSS VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- LIYSASSLYSGVPSRF [\J H 328 329 337 —9 \0 U1 —9 95 8m7 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYPYSGYTSY G91- ADSVKGRFTISADTSKNTAYL 361 364 379 130 183 236 289 QMNSLRAEDTAVYYCARY_ST YSSSSKWVWGMDYWGQGTL VTVSS VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- GKAPKLLIYSASSLYSGVPSRF [\J H 328 329 f337 —-—9 5 9 95 8m8 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR GFTVSSSSIHWVRQA PGKGLEWVAYIYPYSGYTSY G91- ADSVKGRFTISADTSKNTAYL fi—* 361 364 380 131 184 237 290 QMNSLRAEDTAVYYCARY_ST YSSSKWVWGMDYWGQGTLV TVSS MTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- GKAPKLLIYSASSLYSGVPSRF [\J fi—* 328 329 337 95 9 95 95 8m9 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR - VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYPYSGYTSY G91- ADSVKGRFTISADTSKNTAYL 00 [\J 361 364 383 132 185 238 291 QMNSLRAEDTAVYYCARY_ST YSSKWVWGMDYWGQGTLVT VLIDIQMTQSPSSLSASVGDRV SQSVSSAVAWYQQKP G9.1- GKAPKLLIYSASSLYSGVPSRF [\J H 328 329 337 95 95 95 95 8m10 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR I 4; kit I VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYPYSGYTSY G91- ADSVKGRFTISADTSKNTAYL 292 8m10 QMNSLRAEDTAVYYCARY_ST YSKWVWGMDYWGQGTLVT VLzDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- LIYSASSLYSGVPSRF 8m11 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYPYSGYTSY G91- ADSVKGRFTISADTSKNTAYL 293 8m11 QMNSLRAEDTAVYYCARY_ST YKWVWGMDYWGQGTLVTV MTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- GKAPKLLIYSASSLYSGVPSRF 8m12 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR — VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYPYSSSSSYA G91- DSVKGRFTISADTSKNTAYLQ 294 8m12 EDTAVYYCARYSTY SSSKWVWGMDYWGQGTLVT VLzDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- LIYSASSLYSGVPSRF 8m13 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR — VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA PGKGLEWVAYIYPYSSSSSYA G91- DSVKGRFTISADTSKNTAYLQ 295 8m13 EDTAVYYCARYSTY SSKWVWGMDYWGQGTLVTV VLIDIQMTQSPSSLSASVGDRV TITCRASQSVSSAVAWYQQKP G91- GKAPKLLIYSASSLYSGVPSRF 8m14 SGSRSGTDFTLTISSLQPEDFAT YYCQQSYYDSNPITFGQGTKV EIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQA G91- PGKGLEWVAYIYPYSSSSSYA 8m14 DSVKGRFTISADTSKNTAYLQ MNSLRAEDTAVYYCARYSTY KWVWGMDYWGQGTLVTVSS Table 2. Antibodies directed against CRDZ LC IgGl CDR CDR CDR IgG4 VR /HC LAL IgG4 Clone Sequence 1 2 3 mut I G1 A —SEQIDNO: VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—1 APKLLIYSASSLYSGVPSRFSGS 341 RSGTDFTLTISSLQPEDFATYYC QQYKSKYPFTFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTLYSSSIHWVRQAP GKGLEWVASIYSSSGYTYYAD G9.2-1 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARTYTWKSS WSYQTGYGLDYWGQGTLVTV VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—2 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSSSLITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTFSSSSIHWVRQAP GKGLEWVASISPYYGSTYYAD G9.2-2 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARAVYYYVY NRSWYWWSGGFDYWGQGTL VTVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—3 YSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSSSLITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTFSSSSIHWVRQAP VASISSSSGSTSYADS G9.2-3 ISADTSKNTAYLQMNS LRAEDTAVYYCARPAYSYPYY YFHYGAMDYWGQGTLVTVSS MTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—4 APKLLIYSASSLYSGVPSRFSGS TLTISSLQPEDFATYYC QQSSSSLITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTFSSSSIHWVRQAP GKGLEWVASIYPSYGYTSYAD G9.2-4 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARAWYHHE YWGHYSGMDYWGQGTLVTVS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—5 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSWGLITFGQGTKVEIKR VH:EVQLVESGGGLVQPGGSLR LSCAASGFTFSSSSIHWVRQAP GKGLEWVASIYSSYGSTYYAD G9.2-5 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARSGYSHPY YSYYSGMDYWGQGTLVTVSS VL:DIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2-6 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQFWGSKLFTFGQGTKVEIKR VH:EVQLVESGGGLVQPGGSLR LSCAASGFTFSSSSIHWVRQAP GKGLEWVASIYSYSGYTYYAD G9.2-6 TISADTSKNTAYLQMN SLRAEDTAVYYCARTYMAGY KYYFISGYGFDYWGQGTLVTV VL:DIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK YSASSLYSGVPSRFSGS G9.2-7 RSGTDFTLTISSLQPEDFATYYC QQMYYPGYLITFGQGTKVEIK VH:EVQLVESGGGLVQPGGSLR LSCAASGFTFSYSSIHWVRQAP GKGLEWVASIYPSYGYTYYAD G9.2-7 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARYWDYGW MDYWGQGTLVTVSS VL:DIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK APKLLIYSASSLYSGVPSRFSGS G9.2-8 RSGTDFTLTISSLQPEDFATYYC QQDRWWSALTFGQGTKVEIK LVESGGGLVQPGGSLR LSCAASGFTFSYSSIHWVRQAP GKGLEWVASIYSYSGYTSYAD G9.2-8 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARYMENWE WPYHSAMDYWGQGTLVTVSS VL:DIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2-9 YSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSYGSWYPITFGQGTKVEIKR VH:EVQLVESGGGLVQPGGSLR LSCAASGFTFYSSSIHWVRQAP GKGLEWVASIYSSYGSTYYAD G9.2-9 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARSWWYPY WQYYPGGWHSSGFDYWGQG TLVTVSS VL:DIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—10 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQGWYASPITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTFYSSSIHWVRQAP VAYISPSSGYTSYADS G9.2—10 ISADTSKNTAYLQMNS LRAEDTAVYYCARYTMTYQY YPSGAMDYWGQGTLVTVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2-11 YSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQYSSHKYPFTFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTIYSSYIHWVRQAP GKGLEWVASIYSSSGYTYYAD G9.2-11 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARSYIYYMW QYNYGMSGYGLDYWGQGTLV TVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK YSASSLYSGVPSRFSGS G9.2—12 RSGTDFTLTISSLQPEDFATYYC QQWVYPGSLITFGQGTKVEIK VHzEVQLVESGGGLVQPGGSLR LSCAASGFTLSYSSIHWVRQAP GKGLEWVASISSSYGYTYYAD G9.2—12 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARHSPYYLH SWWWSGLDYWGQGTLVTVSS MTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2-13 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQYKSKYPFTFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTLYYSSIHWVRQAP GKGLEWVASISPSYGSTSYADS G9.2-13 VKGRFTISADTSKNTAYLQMNS LRAEDTAVYYCARHSWYYPY YYYALDYWGQGTLVTVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—14 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSSSLITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR GFTVSSSSIHWVRQAP GKGLEWVASISSSSGYTYYADS G9.2—14 VKGRFTISADTSKNTAYLQMNS LRAEDTAVYYCARYWSYPYVY FLAFDYWGQGTLVTVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—15 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSWGLITFGQGTKVEIKR G9.2—15 VKGRFTISADTSKNTAYLQMNS LRAEDTAVYYCARNVENYPY WAWPWGYYGAIDYWGQGTL VTVSS VLIDIQMTQSPSSLSASVGDRVTI SVSSAVAWYQQKPGK G9.2—16 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSSSLITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQAP GKGLEWVASIYSSSGYTYYAD G9.2—16 SVKGRFTISADTSKNTAYLQMN TAVYYCARTYKWSYY TGYGFDYWGQGTLVTVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—17 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSTDPITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQAP GKGLEWVAYISSSSGYTYYAD G9.2—17 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARYWSYPSW WPYRGMDYWGQGTLVTVSS VLIDIQMTQSPSSLSASVGDRVTI SVSSAVAWYQQKPGK G92- APKLLIYSASSLYSGVPSRFSGS 17mut6 RSGTDFTLTISSLQPEDFATYYC PITFGQGTKVEIKR G92- VHzEVQLVESGGGLVQPGGSLR 1 7mut6 LSCAASGFTVSSSSIHWVRQAP (mutatio GKGLEWVAYISSSSGYTYYAD n is SVKGRFTISADTSKNTAYLQMN underlin SLRAEDTAVYYCARYWSYPSW ed) SPYRGMDYWGQGTLVTVSS VLzDIQMTQSPSSLSASVGDRVTI SVSSAVAWYQQKPGK G9.2—18 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSSSLITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSYSIHWVRQAP GKGLEWVAYIYSSSGYTSYAD G9.2—18 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARVGYYYPY LYLGDGLDYWGQGTLVTVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2—19 YSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSQYDLITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSSYSIHWVRQAP GKGLEWVASISSSSGSTSYADS G9.2—19 VKGRFTISADTSKNTAYLQMNS LRAEDTAVYYCARNAWHYEPS YWYGNYATYGFDYWGQGTLV TVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2-20 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSTDPITFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVSYSSIHWVRQAP GKGLEWVASISSSSSSTYYADS G9.2—20 VKGRFTISADTSKNTAYLQMNS LRAEDTAVYYCARGQQYYPD QYWGLDYWGQGTLVTVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2-21 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSSSSSLFTFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR GFTVSYSSIHWVRQAP GKGLEWVASIYSSSGYTYYAD G9.2-21 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARTYYTYFD YYGFDYWGQGTLV TVSS MTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQRWYPGDLITFGQGTKVEIK VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVYSSSIHWVRQAP GKGLEWVASISSSYGYTSYADS G9.2—22 VKGRFTISADTSKNTAYLQMNS LRAEDTAVYYCARDYYNYMSS YWWYSALDYWGQGTLVTVSS VLzDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2-23 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSYFPSLVTFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVYSSSIHWVRQAP VASIYPYYGYTSYAD G9.2-23 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARKIFWPVS WMWQGYYPALDYWGQGTLV TVSS VLIDIQMTQSPSSLSASVGDRVTI TCRASQSVSSAVAWYQQKPGK G9.2-24 APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQWSQSPV'IFGQGTKVEIKR VHzEVQLVESGGGLVQPGGSLR LSCAASGFTVYSSSIHWVRQAP GKGLEWVASIYSSYGYTSYAD G9.2—24 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARSYSSETHY GWAMDYWGQGTLVTVSS VLIDIQMTQSPSSLSASVGDRVTI G9 2—25‘ TCRASQSVSSAVAWYQQKPGK APKLLIYSASSLYSGVPSRFSGS RSGTDFTLTISSLQPEDFATYYC QQSYVYYPFTFGQGTKVEIKR VHIEVQLVESGGGLVQPGGSLR LSCAASGFTLSSSSIHWVRQAP GKGLEWVASIYSSYGSTSYADS G9.2-25 VKGRFTISADTSKNTAYLQMNS LRAEDTAVYYCARQYYTYFE YALDYWGQGTLVT VLzDIQMTQSPSSLSASVGDRVTI SVSSAVAWYQQKPGK APKLLIYSASSLYSGVPSRFSGS G9.2—26 RSGTDFTLTISSLQPEDFATYYC QQGGWYYGPITFGQGTKVEIK VHIEVQLVESGGGLVQPGGSLR LSCAASGFTVSSSSIHWVRQAP GKGLEWVAYISSYSGSTYYAD G9.2—26 SVKGRFTISADTSKNTAYLQMN SLRAEDTAVYYCARSSALYWM DFSYSALDYWGQGTLVTVSS VLzDIQMTQSPSSLSASVGDRVTI G92- TCRASQSVSSAVAWYQQKPGK APKLLIYSASSLYSGVPSRFSGS affinity RSGTDFTLTISSLQPEDFATYYC binder QQSSTDPITFGQGTKVEIKR VHIEVQLVESGGGLVQPGGSLR G9.2- LSCAASGFTVSSSSIHWVRQAP low GKGLEWVAYISSSSGYTYYAD affinity SVKGRFTISADTSKNTAYLQMN binder SLRAEDTAVYYCARSSSSSSSSS SSSSDYWGQGTLVTVSS Table 3. Selected Antibody CDR ces ——Seuence G9.1-8 RASQSVSSAVA 691-81111 G9.1-8m2 G9.1-8m3 G9.1-8m4 G9.1-8m5 G9.1-8m6 G9.1-8m8 G9.1-8m9 G9.1-8m10 —VHCDR3 YSTYSKWVWGMDY G9.1-8m12 G9.1-8m13 G9.1-8m14 G9.2-17 G9.2-17m6 Such CRDl and CRD2 binding anti-Galectin-9 antibodies are isolated and urally characterized as described herein. The disclosure also contemplates antibodies having at least 80% identity (e.g., at least 85%, at least 90%, at least 95%, or at least 99% identity) to their variable region or CDR sequences. The VL amino acid sequences of G9.2-l, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, , G9.2-8, G9.2-9, G9.2-10, G9.2-11, 2, G9.2-13, G9.2-14, G9.2-15,G9.2-16,G9.2-17,G9.2-17mut6,G9.2-18,G9.2-19,G9.2-20,G9.2-21,G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, G9.2-low affinity binder are set forth in SEQ ID NO: 29, 13, 34, 36, 38, 40, 42, 44, 46, 48, 29, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71. The VH amino acid sequences of G9.2-1, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, 7, G9.2-17mut6, 8, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, G9.2-low aff1nity binder are set forth in SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73. Accordingly, provided herein are isolated anti-Galectin-9 antibodies, or antigen binding n thereof, comprising heavy and light chain variable s, wherein the light chain variable region comprises an amino acid sequence selected from SEQ ID NO: 29, 13, 34, 36, 38, 40, 42, 44, 46, 48, 29, 34, 54, 58, 61, 63 7 65, 73, 67, 69, and 71. In some embodiments, the light chain variable regions consists of an amino acid sequence selected from SEQ ID NO: 29, 13, 34, 36, 38, 40, 42, 44, 46, 48, 29, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71. Also provided are isolated anti-Galectin-9 antibodies, or antigen binding portions f, comprising heavy and light chain variable regions, wherein the heavy chain variable region comprises an amino acid sequence selected from SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73. In some embodiments, the heavy chain variable regions consists of an amino acid sequence selected from SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53 7 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73. Accordingly, provided herein are isolated anti- Galectin-9 antibodies, or antigen binding portion thereof, comprising heavy and light chain 2O variable regions, wherein the light chain variable region comprises an amino acid sequence selected from SEQ ID NO: 29, 13, 34, 36, 38, 40, 42, 44, 46, 48, 29, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71, and the heavy chain variable region comprises an amino acid sequence ed from SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73. Accordingly, provided herein are isolated anti-Galectin-9 antibodies, or n binding portion f, comprising heavy and light chain variable regions, wherein the light chain variable region consists of an amino acid sequence selected from SEQ ID NO: 29, 13, 34, 36, 38, 40, 42, 44, 46, 48, 29, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71, and the heavy chain variable region consists of an amino acid ce selected from SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 3O 64, 66, 68, 70, 72 and 73.
In some embodiments, the anti-Galectin-9 antibody comprises a VL region having the sequence of SEQ ID NO: 54. In some embodiments, the anti-Galectin-9 antibody comprises a VH region having the ce of SEQ ID NO: 55. In some embodiments, the anti-Galectin-9 antibody comprises a VH region having the sequence of SEQ ID NO: 56. In some ments, the anti-Galectin-9 antibody comprises a VL region having the sequence of SEQ ID NO: 54 and a VH region having the sequence of SEQ ID NO: 55. In some embodiments, the anti-Galectin-9 antibody comprises a VL region having the ce of SEQ ID NO: 54 and a VH region having the sequence of SEQ ID NO: 56.
The VL amino acid sequences of G9.l-l, G9.1-2, G9.1-3, G9.1-4, G9.1-5, G9.1-6, G9.1-7, G9.1-8, G9.1-9, 0, G9.1-11, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.1- 8m5, G9.1-8m6,G9.1-8m7, G9.1-8m8, G9.1-8m9, G9.1-8m10, G9.1-8m11, G9.1-8m12, G9.1- 8ml3, G9.1-8m14 are set forth in SEQ ID NO: 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, and 27, respectively. The VH amino acid sequences of G9.l-l, G9.1-2, G9.1-3, G9.1-4, G9.1-5, G9.1-6, G9.1-7, G9.1-8, G9.1-9, G9.1-10, 1, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.1- 8m5, G9.1-8m6,G9.1-8m7, G9.1-8m8, G9.1-8m9, G9.1-8m10, G9.1-8m11, G9.1-8m12, G9.1- 8ml3, G9.1-8m14 are set forth in SEQ ID NO: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87. Accordingly, provided herein are isolated anti- Galectin-9 antibodies, or antigen binding portions thereof, comprising heavy and light chain variable regions, wherein the light chain variable region ses an amino acid sequence selected from SEQ ID NO: 7, 9, ll, 13, 15, l7, 19, 21, 23, 25, and 27. In some embodiments, the light chain variable region consists of an amino acid sequence selected from SEQ ID NO: 7, 9, ll, 13, 15, l7, 19, 21, 23, 25, and 27. Also provided are ed anti-Galectin-9 antibodies, or antigen binding ns thereof, comprising heavy and light chain variable regions, wherein 2O the heavy chain variable region comprises an amino acid sequence selected from SEQ ID NO: 8, , 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87. In some embodiments, the heavy chain variable regions consists of an amino acid sequence selected from SEQ ID NO: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87. ingly, ed herein are ed anti-Galectin-9 antibodies, or antigen binding portions thereof, comprising heavy and light chain variable regions, wherein the light chain variable region comprises an amino acid sequence ed from SEQ ID NO: 7, 9, ll, 13, 15, l7, 19, 21, 23, 25, and 27 and the heavy chain variable region comprises an amino acid sequence selected from SEQ ID NO: 8, 10, 12, l4, l6, 18, 20, 22, 24, 26,28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87. In some embodiments, the light 3O chain variable s consists of an amino acid sequence selected from SEQ ID NO: 7, 9, ll, 13, 15, l7, 19, 21, 23, 25, and 27, and the heavy chain variable regions consists of an amino acid sequence selected from SEQ ID NO: 8, 10, 12, l4, l6, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87.
In some embodiments, the alectin-9 dy comprises a VL region having the sequence of SEQ ID NO: 21. In some embodiments, the anti-Galectin-9 antibody comprises a VH region haVing the sequence of SEQ ID NO: 22. In some embodiments, the anti-Galectin-9 antibody comprises a VH region haVing the sequence of SEQ ID NO: 86. In some embodiments, the alectin-9 antibody comprises a VL region haVing the sequence of SEQ ID NO: 21 and a VH region haVing the sequence of SEQ ID NO: 22. In some embodiments, the anti-Galectin-9 antibody ses a VL region haVing the sequence of SEQ ID NO: 21 and a VH region haVing the sequence of SEQ ID NO: 86.
In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises any of SEQ ID NO: 7-87. In some specific embodiments, the anti-Galectin-9 antibody comprises one or more ces of any sequence(s) selected from SEQ ID NO: 7-87 and any combination(s) thereof.
In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises any of SEQ ID NOs: 7-28 and 74-87. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises one or more ces of any sequence(s) selected from SEQ ID NO: 7-28 and 74-87 and any combination(s) thereof.
In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises any of SEQ ID NOs: 13, 29-73. In some specific embodiments, the anti- Galectin-9 antibody or antigen binding portion thereof comprises one or more sequences of any 2O sequence(s) ed from SEQ ID NO: 13, 29-73 and any combination(s) thereof. In some specific embodiments, the alectin-9 antibody or antigen binding portion thereof comprises any of SEQ ID NOs: 54, 55, or 54 and 56. In some c embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises one or more sequences of any sequence(s) selected from SEQ ID NO: 54, 55, or 54 and 56 and any combination(s) thereof.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion f comprises a VL region comprising SEQ ID NO: 54. In some embodiments, the anti-Galectin-9 dy or antigen binding portion thereof comprises a VH region comprising SEQ ID NO: 55.
In some embodiments, the anti-Galectin-9 antibody ses a VL region consisting of SEQ ID NO: 54. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion f comprises a VH region consisting of SEQ ID NO: 55. In some embodiments, the anti- Galectin-9 antibody comprises a VL and VH region comprising SEQ ID NO: 54 and 55. In some specific embodiments, the anti-Galectin-9 antibody or n binding portion f comprises a VL and VH region consisting of SEQ ID NO: 54 and 55.
In some ments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region comprising SEQ ID NO: 21. In some embodiments, the alectin-9 dy or antigen binding portion thereof comprises a VL region consisting of SEQ ID NO: 21. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VH region comprising SEQ ID NO: 86. In some embodiments, the anti-Galectin-9 dy or antigen binding portion thereof comprises a VH region consisting of SEQ ID NO: 86. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL and VH region comprising SEQ ID NO: 21 and 86. In some embodiments, the alectin-9 antibody or antigen binding portion f comprises a VL and VH region consisting of SEQ ID NO: 21 and 86.
In some c embodiments, the anti-Galectin-9 antibody or antigen binding portion f comprises a VL or VH region comprising any of of SEQ ID NOs: 21, 22 and 74-87. In some c embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL or VH region consisting of SEQ ID NOs: 21, 22 and 74-87. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL and/or VH region comprising sequence(s) selected from SEQ ID NO: 21, 22 and 74-87 and any combination(s) f. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL and/or VH region consisting of sequence(s) selected from SEQ ID NO: 21, 22 and 74-87 and any ation(s) thereof. 2O In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 7 and SEQ ID NO: 8. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region sing SEQ ID NO: 9 and SEQ ID NO: 10. In some embodiments, the anti-Galectin-9 antibody or antigen g portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 11 and SEQ ID NO: 12. In some embodiments, the anti- Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 13 and SEQ ID NO: 14. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 15 and SEQ ID NO: 16. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 17 and SEQ ID NO: 18. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region sing SEQ ID NO: 19 and SEQ ID NO: 20. In some embodiments, the anti-Galectin-9 dy or antigen binding n thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 22. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 23 and SEQ ID NO: 24. In some ments, the anti-Galectin-9 antibody or n binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 25 and SEQ ID NO: 26. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 27 and SEQ ID NO: 28. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 74. In some embodiments, the anti- in-9 antibody or antigen binding n thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 75. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 76. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion f comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 77. In some embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 78. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 79. In some ments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region sing SEQ ID NO: 21 and SEQ ID NO: 80. In 2O some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 81. In some embodiments, the anti-Galectin-9 dy or antigen binding n thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 82. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof ses a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 83. In some embodiments, the anti- Galectin-9 antibody or n binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 84. In some embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL region and a VH region comprising SEQ ID NO: 108 and SEQ ID NO: 85. In some embodiments, the anti-Galectin-9 antibody or 3O antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 21 and SEQ ID NO: 86. In some embodiments, the anti-Galectin-9 antibody or antigen binding n f ses a VL region and a VH region comprising SEQ ID NO: 29 and SEQ ID NO: 30. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 13 and SEQ ID NO: 31. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 13 and SEQ ID NO: 32. In some embodiments, the alectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 13 and SEQ ID NO: 33. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 34 and SEQ ID NO: 35. In some embodiments7 the anti-Galectin-9 antibody or antigen g n thereof ses a VL region and a VH region comprising SEQ ID NO: 36 and SEQ ID NO: 37. In some embodiments, the anti- in-9 antibody or antigen binding n thereof comprises a VL region and a VH region comprising SEQ ID NO: 38 and SEQ ID NO: 39. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region sing SEQ ID NO: 40 and SEQ ID NO: 41. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 42 and SEQ ID NO: 43. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 44 and SEQ ID NO: 45. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion f comprises a VL region and a VH region comprising SEQ ID NO: 46 and SEQ ID NO: 47. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 48 and SEQ ID NO: 49. In 2O some embodiments, the anti-Galectin-9 antibody or antigen binding portion f comprises a VL region and a VH region comprising SEQ ID NO: 29 and SEQ ID NO: 50. In some ments, the anti-Galectin-9 dy or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 13 and SEQ ID NO: 51. In some embodiments7 the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 34 and SEQ ID NO: 52. In some embodiments, the anti- Galectin-9 dy or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 13 and SEQ ID NO: 53. In some embodiments, the anti-Galectin-9 dy or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 54 and SEQ ID NO: 55. In some embodiments, the anti-Galectin-9 antibody or 3O antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 13 and SEQ ID NO: 57. In some embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 58 and SEQ ID NO: 59. In some embodiments, the alectin-9 antibody or n binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 54 and SEQ ID NO: 60. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 61 and SEQ ID NO: 62. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 63 and SEQ ID NO: 64. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 65 and SEQ ID NO: 66. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 54 and SEQ ID NO: 56. In some embodiments, the anti- Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 67 and SEQ ID NO: 68. In some embodiments, the anti-Galectin-9 antibody or antigen g portion f comprises a VL region and a VH region comprising SEQ ID NO: 69 and SEQ ID NO: 70. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 71 and SEQ ID NO: 72. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region comprising SEQ ID NO: 54 and SEQ ID NO: 73. In some specific ments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 7 and SEQ ID NO: 8. In some ic embodiments, the alectin-9 dy or antigen g portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 9 and SEQ 2O ID NO: 10. In some specific embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 11 and SEQ ID NO: 12. In some specific ments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region ting of SEQ ID NO: 13 and SEQ ID NO: 14. In some ic embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 15 and SEQ ID NO: 16. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 17 and SEQ ID NO: 18. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 19 and SEQ 3O ID NO: 20. In some specific ments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 22. In some ic embodiments, the anti-Galectin-9 antibody or antigen binding portion f comprises a VL region and a VH region consisting of SEQ ID NO: 23 and SEQ ID NO: 24. In some specific embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises a VL region and a VH region ting of SEQ ID NO: 25 and SEQ ID NO: 26. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL region and a VH region consisting of SEQ ID NO: 27 and SEQ ID NO: 28. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 74. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 75. In some specific embodiments, the anti-Galectin-9 antibody or antigen g portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 76. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 77. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region ting of SEQ ID NO: 21 and SEQ ID NO: 78. In some specific ments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 79. In some ic embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof ses a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 80. In some specific embodiments, the alectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ 2O ID NO: 81. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 82. In some specific embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 83. In some ic embodiments, the anti-Galectin-9 dy or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 21 and SEQ ID NO: 84. In some specific ments, the anti-Galectin-9 dy or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 108 and SEQ ID NO: 85. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL region and a VH region ting of SEQ ID NO: 21 and SEQ 3O ID NO: 86. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL region and a VH region consisting of SEQ ID NO: 29 and SEQ ID NO: 30. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 13 and SEQ ID NO: 31. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 13 and SEQ ID NO: 32. In some specific embodiments, the anti-Galectin-9 antibody or n g portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 13 and SEQ ID NO: 33. In some specific embodiments, the anti-Galectin-9 dy or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 34 and SEQ ID NO: 35. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL region and a VH region consisting of SEQ ID NO: 36 and SEQ ID NO: 37. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 38 and SEQ ID NO: 39. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region ting of SEQ ID NO: 40 and SEQ ID NO: 41. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 42 and SEQ ID NO: 43. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 44 and SEQ ID NO: 45. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises a VL region and a VH region consisting of SEQ ID NO: 46 and SEQ ID NO: 47. In some specific ments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 48 and SEQ 2O ID NO: 49. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 29 and SEQ ID NO: 50. In some specific embodiments, the anti-Galectin-9 antibody or antigen g portion thereof comprises a VL region and a VH region ting of SEQ ID NO: 13 and SEQ ID NO: 51. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 34 and SEQ ID NO: 52. In some specific embodiments, the anti-Galectin-9 antibody or antigen g portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 13 and SEQ ID NO: 53. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 54 and SEQ 3O ID NO: 55. In some specific ments, the anti-Galectin-9 dy or antigen g portion thereof comprises a VL region and a VH region ting of SEQ ID NO: 13 and SEQ ID NO: 57. In some specific embodiments, the anti-Galectin-9 antibody or antigen g portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 58 and SEQ ID NO: 59. In some specific embodiments, the anti-Galectin-9 dy or antigen binding portion thereof ses a VL region and a VH region consisting of SEQ ID NO: 54 and SEQ ID NO: 60. In some specific embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 61 and SEQ ID NO: 62. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof ses a VL region and a VH region consisting of SEQ ID NO: 63 and SEQ ID NO: 64. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 65 and SEQ ID NO: 66. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 54 and SEQ ID NO: 56. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof ses a VL region and a VH region consisting of SEQ ID NO: 67 and SEQ ID NO: 68. In some specific embodiments, the anti-Galectin-9 dy or antigen binding portion thereof comprises a VL region and a VH region ting of SEQ ID NO: 69 and SEQ ID NO: 70. In some specific embodiments, the anti-Galectin-9 dy or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 71 and SEQ ID NO: 72. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises a VL region and a VH region consisting of SEQ ID NO: 54 and SEQ ID NO: 73.
In some embodiments, the alectin-9 antibody comprises sequence haVing at least 2O 80% (e.g., 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% and any incremental percent therein) sequence ty with any of the anti-Galectin-9 antibodies described in the previous paragraphs. In some ments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL region set forth in SEQ ID NOs: 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, and 27. In some embodiments, the anti-Galectin-9 antibody comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to a VH region set forth in SEQ ID NOs: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87. In some embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and 3O any increment therein) ce identity to a VL region set forth in SEQ ID NOs: 7, 9, 11, 13, , 17, 19, 21, 23, 25, and 27 and a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH region set forth in SEQ ID NOs: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87. In some embodiments, the anti-Galectin-9 antibody comprises a VL or VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL or VH region set forth in SEQ ID NOs: 7-288 and 74-87. In some specific embodiments, the anti-Galectin-9 antibody comprises a VL or VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL or VH region set forth in SEQ ID NO: 7-288 and 74-87and any ation(s) thereof.
In some embodiments, the anti-Galectin-9 dy comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL region of an antibody selected from G9. l-l, G9.l-2, G9.l-3, G9.l-4, G9. 1-5, G9.l-6,G9.1-7,G9.l-8,G9.1-9,G9.1-10,G9.l-11,G9.l-8m1,G9.l-8m2,G9.1-8m3,G9.1- 8m4, G9.l-8m5, G9.l-8m6,G9.1-8m7, G9.l-8m8, G9.l-8m9, G9.l-8m10, G9.l-8m11, G9.l- 8m12, G9.l-8ml3, and G9. l-8ml4. In some embodiments, the anti-Galectin-9 antibody comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH region of an dy selected from G9. l-l, ,G9.1-3,G9.l-4,G9.1-5,G9.l-6,G9.1-7,G9.l-8,G9.l-9,G9.l-10,G9.l-11,G9.1-8m1, m2, G9.l-8m3, m4, G9.l-8m5, G9.l-8m6,G9.1-8m7, G9.l-8m8, G9.l-8m9, G9.l- 8m10, G9.l-8ml l, G9.l-8m12, G9.l-8ml3, and G9.l-8ml4. In some embodiments, the anti- Galectin-9 antibody comprises a VL and a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL or VH region of an antibody selected from G9.l-l, G9.l-2, G9.l-3, G9.l-4, G9.l-5, G9.l-6, G9.l-7, G9.l-8, G9.l-9, 0, G9.l-11, G9.l-8ml, G9.l-8m2, G9.l-8m3, G9.l-8m4, G9.l-8m5, G9.l- 8m6,G9.1-8m7, G9.l-8m8, G9.l-8m9, G9.l-8m10, G9.l-8m11, G9.l-8m12, G9.l-8m13, and G9. l-8ml4.
In some specific embodiments, the anti-Galectin-9 antibody comprises a VL or VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL or VH region set forth in any of SEQ ID NOs: 21, 22 and 74- 87. In some c embodiments, the anti-Galectin-9 antibody comprises a VL or VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence identity to a VL or VH region set forth in SEQ ID NO: 21, 22 and 74-87and any combination(s) thereof.
In some specific embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence identity to the VL region of G9. l-8ml3. In some embodiments, the alectin-9 dy comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence identity to the VH region of G9.1-8m13. In some embodiments, the anti-Galectin-9 antibody comprises a VL and a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL or VH region of G9. 1-8m13.
In some embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL region set forth in SEQ ID NO: 21 .In some embodiments, the anti-Galectin-9 dy or antigen binding portion f comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH region set forth in SEQ ID NO: 86. In some ments, the anti-Galectin-9 antibody or n binding portion thereof comprises a VL and VH region that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to the VL and VH s set forth in SEQ ID NO: 21 and 86.
In some embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL region set forth in SEQ ID NO: 13, 29, 34, 36, 38, 40, 42, 44, 46, 48, 29, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71. In some embodiments, the anti-Galectin-9 antibody comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to a VH region set forth in SEQ ID NO: 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73. In some embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL region set forth in SEQ ID NO: 13,29, 34, 36, 38, 40, 42, 44, 46, 48, 29, 34, 54, 58, 61, 63, 65, 73, 67, 69, and 71 and a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH region set forth in SEQ ID NO: , 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 and 73. In some specific embodiments, the anti-Galectin-9 antibody comprises a VL or VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL or VH region set forth in SEQ ID NO: 29-75 and 77-85. In some c embodiments, the anti-Galectin-9 antibody comprises a VL or VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL or VH region set forth in SEQ ID NO: 13, 29-73 and any combination(s) thereof. In some specific embodiments, the anti-Galectin-9 antibody comprises a VL or VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL or VH region set forth in any of SEQ ID NOs: 54, 55, and 56.
In some embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to a VL region of an antibody selected from , G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, , G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2- 16, G9.2-17, G9.2-l7mut6 , G9.2-18, G9.2-19, 0, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder. In some embodiments, the anti-Galectin-9 dy comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence identity to a VH region of an antibody selected from G9.2-1, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12,G9.2-13,G9.2-14,G9.2-15,G9.2-16,G9.2-17,G9.2-17mut6,G9.2-18,G9.2-19, 0, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder.
In some embodiments, the anti-Galectin-9 antibody comprises VL and VH regions that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to VL and VH s of an antibody selected from G9.2-l, G9.2-2, , G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2- ,G9.2-16,G9.2-17,G9.2-17mut6,G9.2-18,G9.2-19,G9.2-20,G9.2-21,G9.2-22,G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low y binder.
In some embodiments, the anti-Galectin-9 antibody comprises a heavy chain CDR haVing at least 80% (e.g., 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% and any incremental percent therein) sequence identity with a ce selected from any of SEQ ID NO: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 31, 32, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 52, 53, 55, 56, 57, 59, 60, 62, 64, 66, 68, 70, 72 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, and 87.
Alternatively or in addition, the anti-Galectin-9 antibody ses a light chain CDR haVing at least 80% (e.g., 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% and any incremental percent therein) sequence identity with a sequence ed from any of SEQ ID NO: 7, 9, ll, 13, 15, 17, 19, 21, 23, 25, 27, 29, 34, 36, 38, 40, 42, 44, 46, 48, 54, 58, 61, 63, 65, 67, 69, 71, and 73.
In some specific embodiments, the anti-Galectin-9 antibody comprises a VL region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL region of G9.2-l7. In some specific embodiments, the anti-Galectin- 9 antibody comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the VH region of G9.2-l7. In some specific embodiments, the anti-Galectin-9 antibody comprises VL and VH regions that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to VL and VH regions of G9.2-l7.
In some specific embodiments, the anti-Galectin-9 antibody or antigen binding fragment thereof comprises a VL that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VL region set forth in SEQ ID NO: 54. In some specific embodiments, the anti-Galectin-9 antibody or antigen binding fragment thereof comprises a VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH region set forth in SEQ ID NO: 55. In some specific embodiments, the anti-Galectin-9 antibody or antigen g fragment thereof ses a VL and/or VH region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to a VL and/or VH region set forth in SEQ ID NO: 54 and 55.
Complementarity Determining Regions (CDRS) Anti-Galectin-9 antibodies, e.g., g to CRDl, can comprise the light and heavy chain CDRls, CDR2s and CDR3s of G9.l-l, G9.l-2, G9.l-3, G9.l-4, G9.l-5, G9.l-6, G9.l-7, G9.l-8, G9.l-9, G9.l-10, G9.l-ll, G9.l-8ml, G9.l-8m2, G9.l-8m3, G9.l-8m4, G9.l-8m5, G9.l-8m6,G9.1-8m7,G9.l-8m8,G9.l-8m9,G9.l-8m10, G9.l-8mll,G9.l-8m12,G9.l-8ml3, and G9.l-8ml4, or combinations thereof. The amino acid sequence of the VL CDRls of G9. l-l, G9.l-2, , G9.l-4, G9.l-5, , G9.l-7, G9.l-8, G9.l-9, G9.l-10, G9.l-ll, G9.l-8ml, G9.l-8m2, G9.l-8m3, G9.l-8m4, G9.l-8m5, G9.l-8m6,G9.l-8m7, m8, G9.l-8m9, G9.l- 8m10, G9.l-8mll, G9.l-8m12, G9.l-8ml3, and G9.l-8ml4 is set forth in SEQ ID NO: 328.
The amino acid ce ofthe VL CDR2s of G9.l-l, G9.l-2, G9.l-3, G9.l-4, G9.l-5, G9.l-6, G9.l-7, , G9.l-9, 0, G9.l-ll, G9.l-8ml, G9.l-8m2, G9.l-8m3, G9.l-8m4, G9.l- 8m5, G9.l-8m6,G9.l-8m7, G9.l-8m8, G9.l-8m9, G9.l-8m10, G9.l-8mll, G9.l-8m12, G9.l- 8ml3, and G9.l-8ml4 is set forth in SEQ ID NO: 329. The amino acid sequences of the VL CDR3s of G9.l-l, G9.l-2, G9.l-3, G9.l-4, G9.l-5, G9.l-6, G9.l-7, , G9.l-9, G9.l-10, G9.l-ll,G9.l-8ml,G9.l-8m2,G9.l-8m3,G9.l-8m4,G9.l-8m5,G9.l-8m6,G9.1-8m7,G9.l- 8m8, G9.l-8m9, G9.l-8m10, G9.l-8mll, G9.l-8m12, G9.l-8ml3, and G9.l-8ml4 are set forth in SEQ ID NO: 330-340. The amino acid sequences ofthe VH CDRls of G9.l-l, G9.l-2, G9. 1- 3,G9.l-4,G9.l-5,G9.l-6,G9.1-7,G9.1-8,G9.l-9,G9.l-10,G9.l-ll,G9.l-8ml,G9.l-8m2, G9.l-8m3, G9.l-8m4, G9.l-8m5, m6,G9.l-8m7, G9.l-8m8, G9.l-8m9, G9.l-8m10, G9.l-8mll, G9.l-8m12, ml3, and G9.l-8ml4 are set forth in SEQ ID NO: 361, 427, 428, 431, 435, 436, 437. The amino acid sequences ofthe VH CDR2s of G9.1-1, G9.1-2, G9.1- 3, G9.1-4, G9.1-5, G9.1-6, G9.1-7, G9.1-8, G9.1-9, G9.1-10, G9.1-11, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G9.1-8m8, m9, G9.1-8m10, G9.1-8m11, G9.1-8m12, G9.1-8m13, and G9.1-8m14 are set forth in SEQ ID NO: 6, and 438-445. The amino acid sequences of the VH CDR3s of G9.1-1, G9.1-2, G9.1-3, G9.1-4, G9.1- , G9.1-6, G9.1-7, G9.1-8, G9.1-9, 0, G9.1-11, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1- 8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G9.1-8m8, G9.1-8m9, G9.1-8m10, G9.1-8m11, G9.1- 8m12, G9.1-8m13, and G9.1-8m14 are set forth in SEQ ID NO: 367-386.
In some embodiments, the anti-Galectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody comprises a VL CDR2 having the ce of SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 antibody ses a VL CDR3 having a sequence selected from any of SEQ ID NOs: 330-340.
In some embodiments, the anti-Galectin-9 dy comprises a VL CDR3 having the sequence of SEQ ID NO: 337. In some embodiments, the anti-Galectin-9 antibody ses a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, and a VL CDR3 having a sequence selected from any of SEQ ID NOs: 330-340. In some embodiments, the anti-Galectin-9 antibody ses a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, and a VL CDR3 having the sequence of SEQ ID NO: 337. In some embodiments, the anti-Galectin-9 dy ses a 2O VH CDRl having a sequence selected from any of SEQ ID NOs: 361, 427, 428, 431, 435, 436, and 437. In some embodiments, the anti-Galectin-9 antibody ses a VH CDRl having the sequence of SEQ ID NO: 361. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDR2 having a sequence selected from any of SEQ ID NOs: 362-366, and 43 8-445. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDR2 having a sequence selected from SEQ ID NO: 364 or 366. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDR3 having a sequence selected from any of SEQ ID NOs: 367-3 86. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDR3 having the sequence of SEQ ID NO: 374 or 383. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDRl having a sequence ed from any of SEQ ID NOs: 361, 427, 428, 431, 43 5, 436, and 437, a VH CDR2 having a sequence selected from any of SEQ ID NOs: 362-366 and 43 8-445, and a VH CDR3 having a sequence selected from any of SEQ ID NOs: 367-3 86. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDRl having the sequence of SEQ ID NO: 361, a VH CDR2 having the ce of SEQ ID NO: 364, and a VH CDR3 having the sequence of SEQ ID NO: 374. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDRl having the sequence of SEQ ID NO: 361, a VH CDR2 having the sequence of SEQ ID NO: 366, and a VH CDR3 having the sequence of SEQ ID NO: 383. In some embodiments, the anti-Galectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, a VL CDR3 having a sequence selected from any of SEQ ID NOs: 0, a VH CDRl having a sequence selected from any of SEQ ID NOs: 361, 427, 428, 431, 435, 436, and 437, a VH CDR2 having a sequence selected from any of SEQ ID NOs: 362-366 and 43 8-445, and a VH CDR3 having a sequence ed from any of SEQ ID NOs: 367-3 86. In some ments, the alectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, a VL CDR3 having the sequence of SEQ ID NO: 337, a VH CDRl having the sequence of SEQ ID NO: 361, a VH CDR2 having the sequence of SEQ ID NO: 364, and a VH CDR3 having the ce of SEQ ID NO: 374. In some embodiments, the anti-Galectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, a VL CDR3 having the ce of SEQ ID NO: 337, a VH CDRl having the sequence of SEQ ID NO: 361, a VH CDR2 having the sequence of SEQ ID NO: 366, and a VH CDR3 having the sequence of SEQ ID NO: 383. In any of these embodiments, the anti-Galectin-9 antibody binds to CRDl.
In some embodiments, the anti-Galectin-9 antibodies, e.g., binding to CRD2, comprise the light and heavy chain CDRls, CDR2s and CDR3s of G9.2-1, G9.2-2, G9.2-3, G9.2-4, G9.2- 2O 5, G9.2-6, G9.2-7, G9.2-8, , G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-17mut6 , G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and ow affinity , or combinations thereof. The amino acid sequence of the VL CDRls of , G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-17mut6 , G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, 4, 5, G9.2-26, and G9.2-low affinity binder is set forth in SEQ ID NO: 328. The amino acid sequence ofthe VL CDR2s of G9.2-1, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, , G9.2-9, 0, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-17mut6, G9.2-18, G9.2-19, G9.2-20, 1, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2- 3O low affinity binder is set forth in SEQ ID NO: 329. The amino acid sequences of the VL CDR3s of G9.2-1, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-17mut6 , G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder are set forth in SEQ ID NO: 341-360. The amino acid sequences of the VH CDRl of G9.2-1, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, 0, G9.2-11, 2, G9.2-l3, G9.2-l4, G9.2-15, G9.2-l6, G9.2-l7, G9.2-l7mut6, G9.2-l8, G9.2-l9, 0, G9.2- 21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low y binder are set forth in SEQ ID NO: 361, 424-434. The amino acid sequences of the VH CDR2s of G9.2-l, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-l4, G9.2-15, G9.2-l6, G9.2-l7, G9.2-l7mut6, 8, G9.2-l9, G9.2-20, G9.2-2l, G9.2- 22, 3, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder are set forth in SEQ ID NO: 362, 363, 387-3 89 and 446-466. The amino acid ces of the VH CDR3s of G9.2-l, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, G9.2-12, G9.2-l3, G9.2-l4, 5, G9.2-l6, G9.2-l7, G9.2-l7mut6, G9.2-l8, G9.2-l9, G9.2-20, G9.2- 21, G9.2-22, 3, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder are set forth in SEQ ID NO: 7.
In some embodiments, the alectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328. In some ments, the anti-Galectin-9 antibody comprises a VL CDR2 having the sequence of SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 antibody comprises a VL CDR3 having a ce selected from any of SEQ ID NOs: 341-360.
In some embodiments, the anti-Galectin-9 antibody comprises a VL CDR3 having the sequence of SEQ ID NO: 352. In some embodiments, the anti-Galectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, 2O and a VL CDR3 having a sequence selected from any of SEQ ID NOs: 341-360. In some embodiments, the anti-Galectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, and a VL CDR3 having the sequence of SEQ ID NO: 352. In some ments, the anti-Galectin-9 antibody ses a VH CDRl having a sequence selected from any of SEQ ID NOs: 361, and 424-434. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDRl having the sequence of SEQ ID NO: 361. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDR2 having a sequence selected from any of SEQ ID NOs: 362, 363, 387-3 89 and 446-466. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDR2 having the sequence of SEQ ID NO: 388. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDR3 having 3O a sequence selected from any of SEQ ID NOs: 7. In some embodiments, the anti- Galectin-9 antibody comprises a VH CDR3 having the sequence of SEQ ID NO: 406 or 407. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDRl having a sequence selected from any of SEQ ID NOs: 361, and 424-434, a VH CDR2 having a sequence selected from any of SEQ ID NOs: 362, 363, 387-3 89 and 446-466, and a VH CDR3 having a sequence selected from any of SEQ ID NOS: 390-417. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDRl having the sequence of SEQ ID NO: 361, a VH CDR2 having the sequence of SEQ ID NO: 388, and a VH CDR3 having the sequence of SEQ ID NO: 406. In some embodiments, the anti-Galectin-9 antibody comprises a VH CDRl having the sequence of SEQ ID NO: 361, a VH CDR2 having the sequence of SEQ ID NO: 388, and a VH CDR3 having the ce of SEQ ID NO: 407. In some embodiments, the anti-Galectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, a VL CDR3 having a sequence selected from any of SEQ ID NOs: 341-360, a VH CDRl having a sequence selected from any of SEQ ID NOs: 361, and 424- 434, a VH CDR2 having a sequence selected from any of SEQ ID NOs: 362, 363, 387-3 89 and 446-466, and a VH CDR3 having a sequence selected from any of SEQ ID NOs: 390-417. In some embodiments, the anti-Galectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, a VL CDR3 having the sequence of SEQ ID NO: 352, a VH CDRl having the sequence of SEQ ID NO: 361, a VH CDR2 having the ce of SEQ ID NO: 388, and a VH CDR3 having the sequence of SEQ ID NO: 406. In some embodiments, the anti-Galectin-9 antibody comprises a VL CDRl having the sequence of SEQ ID NO: 328, a VL CDR2 having the sequence of SEQ ID NO: 329, a VL CDR3 having the sequence of SEQ ID NO: 352, a VH CDRl having the sequence of SEQ ID NO: 361, a VH CDR2 having the sequence of SEQ ID NO: 388, and a VH CDR3 having the 2O sequence of SEQ ID NO: 407. In any of these embodiments, the anti-Galectin-9 antibody binds to CRDl.
Because Galectin-9 binding specificity is dictated essentially by the CDRl, 2 and 3 regions, the VH CDRl, 2 and 3 ces and the VL CDRl, 2 and 3 ces sed above, can be mixed and matched to generate new Galectin-9 binding antibodies, as long as each resulting new antibody has a VL CDRl, 2 and 3 and a VH CDRl, 2 and 3. Such antibodies resulting from a new combination of CDRs described herein can be tested using the g assays described herein. In some embodiments, the CDRl, CDR2 and/or CDR3 sequence from a particular VH or VL ce is replaced with a structurally similar CDR sequence(s). Novel VH and VL sequences can be created by substituting one or more VH and/or VL CDR sequence(s) with structurally similar sequences from the CDR sequences sed herein, according to methods known in the art.
Accordingly, in some embodiments, anti-Galectin-9 antibodies or antigen binding ns thereof comprise (a) VL CDRl amino acid sequence set forth in SEQ ID NO: 328, (b) VL CDR2 amino acid sequence set forth in SEQ ID NO: 329, (c) VL CDR3 amino acid sequence selected from SEQ ID NO: 330-340 and 0 (d) VH CDRl amino acid sequence set forth in SEQ ID NO: SEQ ID NO: 361, 427, 428, 431, 435, 436, 437, and SEQ ID NO: 361, 4 (e) VH CDR2 amino acid sequence ed from SEQ ID NO: 362-366 and 43 8-445, 362, 363, and 387-3 89 and 446-466, (f) VH CDR3 amino acid sequence selected from SEQ ID NO: 367-386 and 390-417.
In some embodiments, anti-Galectin-9 antibodies or n binding portions thereof comprise a VL CDRl amino acid sequence set forth in SEQ ID NO: 328. In some embodiments, alectin-9 antibodies or antigen binding ns thereof comprise a VL CDR2 amino acid sequence set forth in SEQ ID NO: 329. In some embodiments, anti-Galectin-9 IO antibodies or antigen binding portions thereof comprise a VL CDR3 amino acid sequence selected from SEQ ID NO: 330-340. In some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise a VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 427, 428, 431, 435, 436, 437. In some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof se a VH CDR2 amino acid sequence selected from SEQ ID NO: 362-366, and 43 8-445. In some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise a VH CDR3 amino acid ce ed from SEQ ID NO: 367-3 86. Accordingly, in some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise (a) VL CDRl amino acid sequence set forth in SEQ ID NO: 328, (b) VL CDR2 amino acid sequence set forth in SEQ ID NO: 329, (c) VL CDR3 amino acid 2O sequence selected from SEQ ID NO: 330-340, (d) VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 427, 428, 431, 43 5, 436, 437, (e) VH CDR2 amino acid sequence selected from SEQ ID NO: 6 and 43 8-445, (f) VH CDR3 amino acid ce selected from SEQ ID NO: 367-386.
In some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise a VL CDRl amino acid sequence set forth in SEQ ID NO: 328. In some embodiments, anti-Galectin-9 antibodies or n binding portions thereof comprise a VL CDR2 amino acid sequence set forth in SEQ ID NO: 329. In some embodiments, alectin-9 antibodies or antigen binding portions thereof comprise a VL CDR3 amino acid sequence selected from SEQ ID NO: 341-360. In some embodiments, anti-Galectin-9 antibodies or antigen g portions thereof comprise a VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 424-434. In some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise a VH CDR2 amino acid sequence selected from SEQ ID NO: 362, 363, 387-3 89 and 446-466. In some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise a VH CDR3 amino acid ce selected from SEQ ID NO: 390-417.
Accordingly, in some ments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise (a) VL CDRl amino acid sequence set forth in SEQ ID NO: 328; (b) VL CDR2 amino acid sequence set forth in SEQ ID NO: 329; (c) VL CDR3 amino acid sequence selected from SEQ ID NO: 34l-360;(d) VH CDRl amino acid sequence set forth in SEQ ID NO: 361; (e) VH CDR2s amino acid sequence selected from SEQ ID NO: 362; 363; 387-3 89 and 446-466;(f) VH CDR3 amino acid sequence selected from SEQ ID NO: 390-417. 9.1 Antibody Clones and d CDRS Clone 9.1-derived Light Chain Variable Regions In some embodiments; the anti-Galectin-9 antibody or antigen binding n thereof comprises heavy and light chain variable regions; wherein the light chain le region CDRl; CDR2; and CDR3 comprise SEQ ID NO: 328; 329; and 330; respectively. In some embodiments; the light chain variable region CDRl; CDR2; and CDR3 consist of SEQ ID NO: 328; 329; and 330; respectively. In some embodiments; the antibody ses the same VL CDRs as G9.l-l.
In some ments; the alectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions; wherein the light chain variable region CDRl; CDR2; and CDR3 comprise SEQ ID NO: 328; 329; and 331; respectively. In some embodiments; the light chain variable region CDRl; CDR2; and CDR3 consist of SEQ ID NO: 2O 328; 329; and 331; respectively. In some embodiments; the antibody comprises the same VL CDRs as G9. 1-2.
In some embodiments; the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions; wherein the light chain variable region CDRl; CDR2; and CDR3 comprise SEQ ID NO: 328; 329; and 332; respectively. In some embodiments; the light chain variable region CDRl; CDR2; and CDR3 t of SEQ ID NO: 328; 329; and 332; respectively. In some embodiments; the antibody comprises the same VL CDRs as G9. 1-3.
In some embodiments; the anti-Galectin-9 antibody or n binding portion thereof ses heavy and light chain variable regions; wherein the light chain variable region CDRl; CDR2; and CDR3 comprise SEQ ID NO: 328; 329; and 333; respectively. In some embodiments; the light chain le region CDRl; CDR2; and CDR3 consist of SEQ ID NO: 328; 329; and 333; respectively. In some embodiments; the antibody comprises the same VL CDRs as G9. 1-4.
In some embodiments, the anti-Galectin-9 dy or antigen g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 334, tively. In some embodiments, the light chain le region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 328, 329, and 334, respectively. In some ments, the antibody comprises the same VL CDRs as G9. 1-5.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 335, respectively. In some ments, the light chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 328, 329, and 335, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9. 1-6.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 336, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 328, 329, and 336, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9. 1-7.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof 2O comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 328, 329, and 337, tively. In some embodiments, the antibody comprises the same VL CDRs as G9. 1-8.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, n the light chain variable region CDRl, CDR2, and CDR3 se SEQ ID NO: 328, 329, and 338, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 328, 329, and 338, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9. 19 In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof ses heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 339, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 328, 329, and 339, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.l-lO.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 340, respectively. In some ments, the light chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 328, 329, and 340, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.l-ll.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable s, wherein the light chain variable region CDRl comprises SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR2 comprises SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR3 comprises SEQ ID NO: 337. In some ments, the anti-Galectin-9 dy or antigen binding portion f comprises heavy and light chain variable regions, wherein the light chain variable region CDRl consists of SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody or antigen g portion thereof ses heavy and light chain variable regions, n the light chain variable region 2O CDR2 consists of SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable s, wherein the light chain variable region CDR3 consists of SEQ ID NO: 337. In some embodiments, the antibody comprises the same VL CDRs as 9.1-8, G9.l-8ml, G9.l-8m2, G9.l-8m3 G9.l-8m4 , , G9.l-8m5 G9.l-8m6 G9.l-8m8 G9.l-8m9 G9.l- , , G9.l-8m7, , , G9.l-8m10, G9.l-8mll, 8m12, G9.l-8ml3, or G9.l-8ml4. In some embodiments, the antibody comprises the same VL CDRs as G9. l-8ml2. In some embodiments, the antibody ses the same VL CDRs as G9. l-8ml3.
In some embodiments, the alectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl comprises SEQ ID NO: 328. In some ments, the anti-Galectin-9 dy or binding portion f comprises heavy and light chain variable regions, wherein the light chain le region CDR2 comprises X1X2X3X4X5SX6X7XSSYADSVKG (SEQ ID NO: 467), in whichX1=YorS,X2=IorS,X3=YorS,X4=PorS,X5=YorS,X6=GorS,X7=YorS, and X8 = T or S. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable s, wherein the light chain variable region CDR3 comprises X1SX2X3X4X5X6X7X8X9X10KX11X12X13GMDY (SEQ ID NO: 468), in which X1 = Y or S, X2 = T, S, or , X3 = Y, S, or absent, X4 = S or absent, X5 = W, S, or absent, X6 = S or absent, X7 = G, S, or absent, X8 = G, T, S, or absent, X9 = I, Y, S, or absent, X10 = G, S, or Y, X11 = W or S, X12 = V or S, and X13 = W or S. In some examples, the anti-Galectin-9 antibody contains G at X7, Y at X8, and/or T at X9 in the heavy chain CDR2 domain. atively, or in addition, the anti-Galectin-9 antibody contains deletions at one or more of X4 — X7 in the heavy chain CDR3 domain. In other examples, the anti-Galectin-9 antibody contains S at one or more of X6 — X8 in the heavy chain CDR2 domain. Alternatively or in addition, the anti-Galectin-9 antibody contains deletions at one or more of X5 — X7 in the heavy chain CDR3 domain. In a further example, the anti-Galectin-9 antibody contains S at one or more of X6 — X8 in the heavy chain CDR2 domain. Alternatively or in on, the anti-Galectin-9 antibody contains ons at one or more of X3 — X9 and/or X10 is Y in the heavy chain CDR3 domain.
Clone 9. l-derived Heavy Chain le Regions In some embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises heavy and light chain variable s, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 431, 438, and 367, tively. In some ments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 431, 43 8, and 367, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.l-l.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 435, 439, and 368, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 43 5, 439, and 368, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9. 1-2.
In some embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 436, 363, and 369, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 436, 363, and 369, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9. 1-3.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 437, 440, and 370, respectively. In some ments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 437, 440, and 370, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-4.
In some embodiments, the anti-Galectin-9 dy or antigen g portion f comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 437, 441, and 371, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 437, 441, and 371, tively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-5.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain le region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 427, 442, and 372, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 427, 442, and 372, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-6.
In some embodiments, the anti-Galectin-9 antibody or antigen g portion thereof 2O comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 443, and 373, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 443, and 373, respectively. In some embodiments, the antibody comprises the same VH CDRs as .
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable s, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 374, respectively. In some embodiments, the heavy chain le region CDRl, CDR2, and CDR3 t of SEQ ID NO: 361, 364, and 374, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 363, and 384, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 363, and 384, respectively. In some embodiments, the antibody ses the same VH CDRs as G9. 1-9.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 429, 444, and 385, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 429, 444, and 385, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9. l- . In some embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises heavy and light chain variable s, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 428, 445, and 386, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 445, and 386, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.l-ll.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain le regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 365, and 374, respectively. In some ments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 365, and 374, respectively. In some embodiments, the dy comprises the same VH CDRs as G9. l-8ml. 2O In some embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 366, and 374, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 366, and 374, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9. l-8m2.
In some embodiments, the anti-Galectin-9 antibody or antigen g portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 375, respectively. In some ments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 364, and 375, respectively. In some embodiments, the antibody ses the same VH CDRs as G9. l-8m3.
In some embodiments, the anti-Galectin-9 antibody or antigen g portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 376, respectively. In some ments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 364, and 376, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8m4.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 377, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 364, and 377, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8m5.
In some embodiments, the anti-Galectin-9 antibody or antigen binding n thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 378, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 t of SEQ ID NO: 361, 364, and 378, respectively. In some embodiments, the antibody ses the same VH CDRs as G9.1-8m6.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof ses heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 379, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 364, and 379, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8m7.
In some embodiments, the anti-Galectin-9 antibody or n binding portion thereof comprises heavy and light chain le regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 380, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 364, and 380, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8m8.
In some embodiments, the anti-Galectin-9 dy or n binding portion thereof comprises heavy and light chain variable s, wherein the heavy chain variable region 3O CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 383, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 364, and 383, tively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8m9.
In some embodiments, the anti-Galectin-9 antibody or n binding n thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 381, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 t of SEQ ID NO: 361, 364, and 381, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8m10.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion f comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 382, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 364, and 382, tively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8m11.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 366, and 380, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 366, and 380, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8m12.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof 2O comprises heavy and light chain variable s, wherein the heavy chain variable region CDRl ses SEQ ID NO: 361. In some ments, the anti-Galectin-9 antibody or antigen binding portion thereof ses heavy and light chain variable regions, wherein the heavy chain variable region CDR2 ses SEQ ID NO: 366. In some embodiments, the anti- Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDR3 region comprises SEQ ID NO: 383. In some embodiments, the anti-Galectin-9 dy or antigen binding portion f comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl consists of SEQ ID NO: 361. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDR2 consists of SEQ ID NO: 366. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, n the heavy chain variable region CDR3 region consists of SEQ ID NO: 383. In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 366, and 383, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 366, and 383, respectively. In some ments, the antibody comprises the same VH CDRs as G9. 1-8m13.
In some embodiments, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 366, and 382, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 361, 366, and 382, tively. In some embodiments, the antibody comprises the same VH CDRs as G9.1-8m14.
Clone 9.1-9.1 Heavy and Light Chain Variable Regions In one specific embodiment, the anti-Galectin-9 antibody or antigen binding portion thereof comprises heavy and light chain le regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 330, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 431, 438, and 367, respectively. In some embodiments, the light and heavy chain le region CDRl, CDR2, and CDR3 consist of SEQ ID NO: 328, 329, and 330 and SEQ ID NO: 431, 438, and 367, respectively. In some ments, the antibody comprises the same VL and VH CDRs as G9. 1-1.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof 2O comprises heavy and light chain variable regions, n: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 331, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 435, 439, and 368, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 331 and SEQ ID NO: 435, 439, and 368. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. 1-2. In one specific embodiment, the anti-Galectin-9 dy or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 332, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 436, 363, and 369, respectively. In 3O some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 332 and SEQ ID NO: 436, 363, and 369. In one specific ment, the antibody comprises the same VL and VH CDRs as G9. 1-3.
In one c embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 333, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 437, 440, and 370, respectively. In some embodiments, the light and heavy chain le region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 333, and SEQ ID NO: 437, 440, and 370.
In one specific embodiment, the antibody ses the same VL and VH CDRs as G9. 1-4.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 334, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 437, 441, and 371, tively. In some embodiments, the light and heavy chain le region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 334, and SEQ ID NO: 437, 441, and 371.
In one specific embodiment, the antibody ses the same VL and VH CDRs as G9. 1-5.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 335, tively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 427, 442, and 372, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and 2O CDR3 regions consist of SEQ ID NO: 328, 329, and 335, and SEQ ID NO: 427, 442, and 372.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. 16 In one c embodiment, the anti-Galectin-9 dy or binding n thereof comprises heavy and light chain variable s, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 336, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 443, and 373, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 336, and SEQ ID NO: 361, 443, and 373.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. 1-7.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 374. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 374. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. 18 In one specific embodiment, the anti-Galectin-9 antibody or binding portion f ses heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 338, tively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 363, and 384. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 338, and SEQ ID NO: 361, 363, and 384. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. 1-9.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain le region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 339, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 429, 444, and 385. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 339, and SEQ ID NO: 429, 444, and 385. In one specific embodiment, the dy comprises the same VL and VH CDRs as G9. 1-10.
In one c embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain le region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 340, respectively, and the heavy 2O chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 428, 445, and 386. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 340, and SEQ ID NO: 428, 445, and 386. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. 1-1 1.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain le region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 365, and 374. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 s consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 365, and 374. In one c 3O embodiment, the antibody comprises the same VL and VH CDRs as G9. l-8ml.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain le region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 366, and 374. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 s consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 366, and 374. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. l-8m2.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, n: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 375. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions t of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 375. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. l-8m3.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 376. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 376. In one specific ment, the antibody comprises the same VL and VH CDRs as G9. l-8m4.
In one specific embodiment, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region 2O CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 377. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 377. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. l-8m5.
In one specific embodiment, the anti-Galectin-9 antibody or binding n f ses heavy and light chain variable regions, n: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain le region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 378. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 378. In one c embodiment, the antibody comprises the same VL and VH CDRs as G9. l-8m6.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 379. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 379. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.1-8m7.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain le regions, wherein: the light chain le region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 380. In some ments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 380. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.1-8m8.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 se SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 se SEQ ID NO: 361, 364, and 383. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 383. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.1-8m9.
In one specific ment, the anti-Galectin-9 antibody or binding portion thereof 2O comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, tively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 381. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 s consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 381. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.1-8m10.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 364, and 382. In some ments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 364, and 382. In one c embodiment, the antibody comprises the same VL and VH CDRs as G9.1-8m11.
In one specific embodiment, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain le regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 se SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 366, and 380. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 366, and 380. In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. l-8ml2.
In one specific embodiment, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein: the light chain le region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 se SEQ ID NO: 361, 366, and 383. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 366, and 383.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9. . In one specific embodiment, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain le regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 366, and 382, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 337, and SEQ ID NO: 361, 366, and 382. In one specific embodiment, the antibody comprises the same VL and VH CDRs 2O as G9.l-8ml4.
Sequence Identity In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise light chain CDRs that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity, individually or collectively, as compared with the corresponding VL CDRs of an exemplary antibody described herein.
Alternatively or in on, the anti-Galectin-9 antibody (e.g., specific to CRDl or CRD2) may comprise heavy chain CDRs that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity, individually or collectively, as compared with the VH CDRs as an exemplary antibody described herein. 3O In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise light chain CDRs that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity, individually or tively, as compared with the corresponding VL CDRs of an antibody or antigen binding portion thereof selected from G9.1-1, G9.1-2, G9.1-3, G9.1-4, , G9.1-6, G9.1-7, G9.1-8, G9.1-9, G9.1- , G9.1-11, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G9.1-8m8, G9.1-8m9, G9.1-8m10, G9.1-8mll, G9.1-8ml2, G9.1-8ml3, and G9.1-8ml4. In some embodiments, the anti-Galectin-9 antibody (e.g., c to CRDl and/or CRD2) may comprise heavy chain CDRs that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity, individually or collectively, as compared with the corresponding VH CDRs of an antibody or antigen g n thereof selected from G9.1-l, G9.1-2, G9.1-3, G9.1-4, G9.1-5, G9.1-6, , G9.1-8, G9.1-9, G9.1-10, 1, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, m5, G9.1-8m6,G9.1-8m7, m8, G9.1-8m9, G9.1- 8m10, G9.1-8m11, G9.1-8m12, G9.1-8m13, and G9.1-8m14.
In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise light chain CDRs and heavy chain CDRs that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity, individually or collectively, as compared with the corresponding VL CDRs and VH CDRs of an antibody or antigen binding portion f selected from G9.1-l, G9.1-2, G9.1-3, G9.1-4, G9.1- , G9.1-6, G9.1-7, G9.1-8, G9.1-9, G9.1-10, G9.1-11, G9.1-8m1, m2, G9.1-8m3, G9.1- 8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G9.1-8m8, G9.1-8m9, m10, G9.1-8m11, G9.1- 8ml2, G9.1-8ml3, and G9.1-8ml4.
In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise a VL CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to VL CDRl amino acid sequence set forth in SEQ ID NO: 374. In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise a VL CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDR2 amino acid ce set forth in SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise a VL CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to a VL CDR3 amino acid sequence selected from SEQ ID NO: 0.
In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise a VH CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 427, 428, 431, 435, 436, 437. In some embodiments, the anti-Galectin-9 antibody (e.g., c to CRDl and/or CRD2) may comprise a VH CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH CDR2 amino acid sequence selected from SEQ ID NO: 362-366 and 43 8-445. In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise a VH CDR3 amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH CDR3 amino acid sequence selected from SEQ ID NO: 367-3 86.
Accordingly, in some ments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise (a) VL CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl amino acid sequence set forth in SEQ ID NO: 328, (b) VL CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the VL CDR2 amino acid sequence set forth in SEQ ID NO: 329, (c) VL CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to a VL CDR3 amino acid sequence selected from SEQ ID NO: 330-340, (d) VH CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 427, 428, 431, 435, 436, 437, (e) VH CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH CDR2 amino acid sequence selected from SEQ ID NO: 362- 366 and 43 8-445, (f) VH CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to a VH CDR3 amino acid sequence ed from SEQ ID NO: 367-3 86.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 328, 329, and 337, tively. In some embodiments, the antibody VL CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to the VL CDRl, CDR2, and CDR3 amino acid ces of G91- 8ml3. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 amino acid ces have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 361, 366, and 383. In some embodiments, the antibody VH CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH CDRl, CDR2, and CDR3 amino acid sequences of G9. l-8ml3. In one specific embodiment, the anti-Galectin-9 dy or binding n thereof comprises heavy and light chain variable regions, wherein: the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain le region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 328, 329, and 337, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 361, 366, and 383. In one specific embodiment, the dy VL CDRl, CDR2, and CDR3 and VH CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl, CDR2, and CDR3 and VH CDRl, CDR2, and CDR3 amino acid sequences as ml3. 9.2 Antibody Clones and Related CDRS Clone 9.2-derived Light Chain Variable Region 2O In some embodiments, the anti-Galectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 341, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 341, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-l.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain le region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 333, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ 3O ID NO: 328, 329, and 333, tively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-2.
In some embodiments, the anti-Galectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 333, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 333, respectively. In some embodiments, the antibody comprises the same VL CDRs as .
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable s, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 342, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 342, respectively. In some embodiments, the dy comprises the same VL CDRs as G9.2-4.
In some embodiments, the anti-Galectin-9 antibody or g n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 343, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 343, respectively. In some embodiments, the antibody ses the same VL CDRs as G9.2-5.
In some embodiments, the alectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 344, respectively. In some 2O embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 344, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-6.
In some embodiments, the anti-Galectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 345, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 s consist of SEQ ID NO: 328, 329, and 345, tively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-7. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 346, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 346. In some embodiments, the antibody comprises the same VL CDRs as G9.2-8.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 347, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 s consist of SEQ ID NO: 328, 329, and 347, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-9.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof ses heavy and light chain variable regions, wherein the light chain le region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 348, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions t of SEQ ID NO: 328, 329, and 348, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-lO.
In some embodiments, the anti-Galectin-9 antibody or g n thereof comprises heavy and light chain variable regions, n the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 349, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions t of SEQ ID NO: 328, 329, and 349, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-ll.
In some embodiments, the alectin-9 antibody or binding portion thereof 2O comprises heavy and light chain le regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 350, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 350, tively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-12.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 341, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 341. In some ments, the antibody comprises the same VL CDRs as G9.2-l3.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 333, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 333, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-l4.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 343, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 343, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-15.
In some ments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 333, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 333. In some embodiments, the antibody comprises the same VL CDRs as G9.2-l6.
In some embodiments, the anti-Galectin-9 dy or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl comprises SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the light chain le region CDR2 comprises SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 dy or binding 2O portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR3 comprises SEQ ID NO: 352. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof ses heavy and light chain variable regions, wherein the light chain variable region CDRl consists of SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDR2 consists of SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 dy or g portion thereof consists of heavy and light chain variable s, wherein the light chain variable region CDR3 comprises SEQ ID NO: 352. In some embodiments, the anti-Galectin-9 antibody or g n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 352, tively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 352, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-l7.
In some embodiments, the anti-Galectin-9 dy or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 352, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 352, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-l7mut6.
In some ments, the alectin-9 dy or binding portion thereof ses heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 s comprise SEQ ID NO: 328, 329, and 333, respectively. In some ments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 333, respectively. In some embodiments, the dy comprises the same VL CDRs as G9.2-18.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 354, tively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 354. In some embodiments, the antibody comprises the same VL CDRs as G9.2-l9.
In some embodiments, the anti-Galectin-9 dy or binding portion thereof 2O comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 352, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 352, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-20.
In some ments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 355, respectively. In some ments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 355, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-21.
In some embodiments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 356, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 356, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-22.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof ses heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 357, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 357, respectively. In some embodiments, the antibody ses the same VL CDRs as G9.2-23.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof ses heavy and light chain variable regions, n the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 358, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 358, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-24.
In some ments, the anti-Galectin-9 dy or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 359, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 359, respectively. In some embodiments, the dy comprises the same 2O VL CDRs as 5.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 360, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions t of SEQ ID NO: 328, 329, and 360, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-26.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof ses heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 s comprise SEQ ID NO: 328, 329, and 352, respectively. In some embodiments, the light chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 352, respectively. In some embodiments, the antibody comprises the same VL CDRs as G9.2-low affinity binder.
Clone 9.2-derived Heavy Chain Variable Region In some embodiments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, n the heavy chain variable region CDRl, CDR2, and CDR3 s comprise SEQ ID NO: 424, 446, and 390, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 424, 446, and 390, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-l.
In some ments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 431, 447, and 391, respectively. In some ments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 431, 447, and 391, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-2.
In some ments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions se SEQ ID NO: 431, 448, and 392, tively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 431, 448, and 392. In some embodiments, the antibody comprises the same VH CDRs as G9.2-3. 2O In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, n the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 431, 449, and 393, tively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 431, 449, and 393, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-4.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 431, 450, and 394, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 s consist of SEQ 3O ID NO: 431, 450, and 394, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-5.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 43 l, 451, and 395, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 431, 452, and 395. In some embodiments, the antibody comprises the same VH CDRs as G9.2-6.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 425, 453, and 396, respectively. In some embodiments, the heavy chain le region CDRl, CDR2, and CDR3 s consist of SEQ ID NO: 425, 453, and 396, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-7.
In some ments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 425, 453, and 397, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 425, 453, and 397, tively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-8.
In some embodiments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 426, 454, and 398, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of 2O SEQ ID NO: 426, 454, and 398, respectively. In some embodiments, the dy comprises the same VH CDRs as G9.2-9.
In some embodiments, the alectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 426, 387, and 399, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 426, 387, and 399, respectively. In some embodiments, the dy comprises the same VH CDRs as G9.2-lO.
In some ments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 432, 455, and 400, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 432, 455, and 400, respectively. In some embodiments, the antibody ses the same VH CDRs as G9.2-ll.
In some ments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 433, 456, and 401, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 433, 456, and 401, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-12.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain le s, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 434, 362, and 402, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 434, 362, and 402, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-l3.
In some embodiments, the alectin-9 antibody or binding portion thereof comprises heavy and light chain variable s, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 361, 457, and 403, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 457, and 403, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-l4.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 361, 458, and 404, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 458, and 404, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-15.
In some embodiments, the anti-Galectin-9 dy or binding portion thereof comprises heavy and light chain variable s, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 361, 459, and 405, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 459, and 405, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-l6.
In some embodiments, the alectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain le region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 361, 388, and 406, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 388, and 406. In some embodiments, the antibody ses the same VH CDRs as G9.2-17.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 s comprise SEQ ID NO: 361, 388, and 407, respectively. In some embodiments, the heavy chain le region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 388, and 407, respectively. In some embodiments, the antibody comprises the same VH CDRs as 7mut6.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 430, 363, and 408, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 430, 363, and 408, respectively. In some embodiments, the antibody comprises the same VH CDRs as 8.
In some embodiments, the anti-Galectin-9 dy or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 430, 460, and 409, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 430, 460, and 409, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-19.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 429, 461, and 410, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 429, 461, and 410, respectively. In some embodiments, the antibody ses the same VH CDRs as G9.2-20.
In some embodiments, the anti-Galectin-9 antibody or g portion f comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 429, 462, and 411, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 429, 462, and 411, tively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-21.
In some embodiments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 428, 463, and 412, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 428, 463, and 412, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-22.
In some ments, the anti-Galectin-9 antibody or binding portion thereof ses heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 428, 464, and 413, respectively. In some ments, the heavy chain variable region CDRl, CDR2, and CDR3 regions t of SEQ ID NO: 428, 464, and 413, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-23.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain le regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 428, 465, and 414, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 428, 465, and 414, respectively. In some embodiments, the antibody ses the same VH CDRs as G9.2-24.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 427, 466, and 415, respectively. In some 2O embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 427, 466, and 415. In some embodiments, the antibody comprises the same VH CDRs as G9.2-25.
In some ments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain le region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 361, 389, and 416, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 389, and 416, respectively. In some embodiments, the antibody comprises the same VH CDRs as G9.2-26.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain le regions, n the heavy chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 361, 388, and 417, respectively. In some embodiments, the heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 361, 388, and 417, respectively. In some embodiments, the antibody ses the same VH CDRs as ow aff1nity binder.
Clone 9.2-derived Heavy and light chain variable regions In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions se SEQ ID NO: 328, 329, and 341, respectively, and the heavy chain le region CDRl, CDR2, and CDR3 se SEQ ID NO: 424, 446, and 390 respectively. In some ments, the light and heavy chain variable region CDRl, CDR2, and CDR3 s consist of SEQ ID NO: 328, 329, and 341, and SEQ ID NO: 424, 446, and 390.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-1.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 333, tively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 431, 447, and 391, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 333, and SEQ ID NO: 431, 447, and 391.
In one specific ment, the antibody comprises the same VL and VH CDRs as G9.2-2.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 333, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 431, 448, and 392, 2O respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 333, and SEQ ID NO: 431, 448, and 392.
In one specific ment, the antibody comprises the same VL and VH CDRs as .
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 342, tively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 431, 449, and 393, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 342, and SEQ ID NO: 431, 449, and 393.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-4. 3O In some embodiments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 343, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 431, 450, and 394, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 343, and SEQ ID NO: 431, 450, and 394.
In one c embodiment, the antibody comprises the same VL and VH CDRs as G9.2-5.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, n the light chain variable region CDRl, CDR2, and CDR3 s comprise SEQ ID NO: 328, 329, and 344, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 431, 451, and 395, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 344, and SEQ ID NO: 431, 451, and 395.
In one c embodiment, the antibody comprises the same VL and VH CDRs as G9.2-6.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 345, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 425, 452, and 396, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 345, and SEQ ID NO: 425, 452, and 396.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-7.
In some ments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 346, respectively, and the heavy chain 2O variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 245, 453, and 397, respectively. In some ments, the light and heavy chain le region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 346, and SEQ ID NO: 245, 453, and 397.In one c ment, the antibody comprises the same VL and VH CDRs as G9.2-8.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 347, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 426, 454, and 398, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions t of SEQ ID NO: 328, 329, and 347, and SEQ ID NO: 426, 454, and 398. 3O In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-9.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable s, wherein the light chain variable region CDRl, CDR2, and CDR3 s comprise SEQ ID NO: 328, 329, and 348, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 426, 387, and 399, -lO2- respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 348, and SEQ ID NO: 426, 387, and 399.
In one specific embodiment, the dy comprises the same VL and VH CDRs as G9.2-lO.
In some embodiments, the anti-Galectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 349, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 432, 455, and 400, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 349, and SEQ ID NO: 432, 455, and 400.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-l 1.
In some ments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions se SEQ ID NO: 328, 329, and 350, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 se SEQ ID NO: 433, 456, and 401, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 350, and SEQ ID NO: 433, 456, and 401.
In one specific embodiment, the antibody ses the same VL and VH CDRs as G9.2-l2.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, 2O and CDR3 regions comprise SEQ ID NO: 328, 329, and 341, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 434, 362, and 402, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 341, and SEQ ID NO: 434, 362, and 402.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-l3.
In some embodiments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 333, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 se SEQ ID NO: 361, 457, and 403, respectively. In some embodiments, the light and heavy chain le region CDRl, CDR2, and CDR3 regions t of SEQ ID NO: 328, 329, and 333, and SEQ ID NO: 361, 457, and 403.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-l4.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof ses heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 343, respectively, and the heavy chain -lO3- variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 458, and 404, respectively. In some embodiments, the light and heavy chain variable region CDRI, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 343, and SEQ ID NO: 361, 458, and 404.
In one c embodiment, the antibody comprises the same VL and VH CDRs as G9.2-15.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain le regions, wherein the light chain variable region CDRI, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 333, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 459, and 405, respectively. In some ments, the light and heavy chain variable region CDRI, CDR2, and CDR3 regions t of SEQ ID NO: 328, 329, and 333, and SEQ ID NO: 361, 459, and 405.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-16.
In some embodiments, the anti-Galectin-9 antibody or binding portion f comprises heavy and light chain variable regions, wherein the light chain variable region CDRI, CDR2, and CDR3 s comprise SEQ ID NO: 328, 329, and 352, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 388, and 406, respectively. In some embodiments, the light and heavy chain variable region CDRI, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 352, and SEQ ID NO: 361, 388, and 406.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-17.
In some embodiments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRI, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 352, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 388, and 404, respectively. In some embodiments, the light and heavy chain variable region CDRI, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 352, and SEQ ID NO: 361, 388, and 404.
In one specific embodiment, the antibody ses the same VL and VH CDRs as G9.2- l7mut6.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof ses heavy and light chain variable regions, n the light chain variable region CDRI, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 333, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 430, 363, and 408, respectively. In some embodiments, the light and heavy chain variable region CDRI, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 333, and SEQ ID NO: 430, 363, and 408.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-18. —104— In some embodiments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 354, respectively, and the heavy chain le region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 430, 460, and 409, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 354, and SEQ ID NO: 430, 460, and 409.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-l9.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 352, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 429, 461, and 410, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 352, and SEQ ID NO: 429, 461, and 410.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-20.
In some embodiments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 s se SEQ ID NO: 328, 329, and 355, tively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 429, 462, and 41 1, tively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and 2O CDR3 regions consist of SEQ ID NO: 328, 329, and 355, and SEQ ID NO: 429, 462, and 411.
In one specific embodiment, the dy ses the same VL and VH CDRs as G9.2-2l.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 356, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 428, 463, and 412, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions t of SEQ ID NO: 328, 329, and 356, and SEQ ID NO: 428, 463, and 412.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-22.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 357, respectively, and the heavy chain le region CDRl, CDR2, and CDR3 se SEQ ID NO: 428, 464, and 413, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and -lO5- CDR3 regions consist of SEQ ID NO: 328, 329, and 357, and SEQ ID NO: 428, 464, and 413.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-23.
In some embodiments, the alectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain le region CDRl, CDR2, and CDR3 s comprise SEQ ID NO: 328, 329, and 358, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 428, 465, and 414, respectively. In some embodiments, the light and heavy chain le region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 358, and SEQ ID NO: 428, 465, and 4l4.In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-24.
In some embodiments, the anti-Galectin-9 antibody or g portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 359, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 427, 466, and 415, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 359, and SEQ ID NO: 427, 466, and 415.
In one specific embodiment, the dy ses the same VL and VH CDRs as G9.2-25.
In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 360, tively, and the heavy chain 2O variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 389, and 416, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 360, and SEQ ID NO: 361, 389, and 416.
In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-26.
In some embodiments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 regions comprise SEQ ID NO: 328, 329, and 352, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 comprise SEQ ID NO: 361, 388, and 417, respectively. In some embodiments, the light and heavy chain variable region CDRl, CDR2, and CDR3 regions consist of SEQ ID NO: 328, 329, and 352, and SEQ ID NO: 361, 388, and 417. 3O In one specific embodiment, the antibody comprises the same VL and VH CDRs as G9.2-low y binder. -lO6- Sequence Identity In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise light chain CDRs that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity, individually or collectively, as compared with the corresponding VL CDRs of an antibody or antigen g portion thereof selected from G9.2-1, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2- ,G9.2-11,G9.2-12,G9.2-13,G9.2-14,G9.2-15,G9.2-16,G9.2-17,G9.2-17mut6,G9.2-18, 9, 0, G9.2-21, 2, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder. In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise heavy chain CDRs that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity, individually or collectively, as ed with the ponding VH CDRs of an dy or antigen binding portion thereof selected from G9.2-1, G9.2-2, G9.2-3, , G9.2-5, , G9.2-7, G9.2-8, G9.2-9, G9.2- 2-11,G9.2-12,G9.2-13,G9.2-14,G9.2-15,G9.2-16,G9.2-17,G9.2-17mut6,G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder.
In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise light chain CDRs and heavy chain CDRs that have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty, individually or collectively, as compared with the ponding VL CDRs and VH CDRs of an antibody or antigen binding portion thereof selected from G9.2-l, G9.2-2, G9.2-3, G9.2-4, G9.2- , G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, G9.2-11, 2, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-l7mut6 , G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and ow affinity binder.
In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise a VL CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl amino acid sequence set forth in SEQ ID NO: 328. In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise a VL CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDR2 amino acid sequence set forth in SEQ ID NO: 329. In some embodiments, the anti-Galectin-9 dy (e.g., specific to CRDl and/or CRD2) may comprise a VL CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, -lO7- 99% and any increment therein) sequence ty to a VL CDR3 amino acid sequence selected from SEQ ID NO: 341-360.
In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise a VH CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 424-434. In some embodiments, the anti- in-9 antibody (e.g., specific to CRDl and/or CRD2) may se a VH CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH CDR2 amino acid sequence selected from SEQ ID NO: 362, 363, 387-3 89 and 446-466. In some embodiments, the anti-Galectin-9 antibody (e.g., specific to CRDl and/or CRD2) may comprise a VH CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to a VH CDR3 amino acid sequence ed from SEQ ID NO: 390-417.
Accordingly, in some embodiments, anti-Galectin-9 antibodies or antigen binding portions thereof comprise (a) VL CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the VL CDRl amino acid sequence set forth in SEQ ID NO: 328, (b) VL CDR2 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDR2 amino acid ce set forth in SEQ ID NO: 329, (c) VL CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDR3 amino acid sequence selected from SEQ ID NO: 341-360, (d) VH CDRl amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VH CDRl amino acid sequence set forth in SEQ ID NO: 361, 424-434, (d) VH CDR2 amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to a VH CDR2 amino acid sequence selected from SEQ ID NO: 362, 363, 387-3 89 and 446-466,(e) VH CDR3 amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to a VH CDR3 amino acid sequence selected from SEQ ID NO: 390-417.
In some ments, the anti-Galectin-9 antibody or binding n thereof comprises heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 328, 329, and 352, -lO8- tively. In some ments, the dy VL CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl, CDR2, and CDR3 amino acid sequences of G9.2- l7. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof comprises heavy and light chain variable regions, wherein the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 361, 388, and 406, respectively. In some embodiments, the antibody VH CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to the VH CDRl, CDR2, and CDR3 amino acid sequences of G9.2- l7. In some embodiments, the anti-Galectin-9 antibody or binding portion thereof ses heavy and light chain variable regions, wherein the light chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain le region CDRl, CDR2, and CDR3 amino acid ces set forth in comprise SEQ ID NO: 328, 329, and 352, respectively, and the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to the heavy chain variable region CDRl, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NO: 361, 388, and 406, respectively. In one specific embodiment, the antibody VL CDRl, CDR2, and CDR3 and VH CDRl, CDR2, and CDR3 amino acid sequences have at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the VL CDRl, CDR2, and CDR3 and VH CDRl, CDR2, and CDR3 amino acid sequences of G9.2-l7.
Epilopes and Constant Regions In some embodiments, the anti-Galectin-9 antibodies described herein bind to the same e as any of the exemplary dies described herein (e.g., antibody comprising any of SEQ ID NO: 7-87 or the CDRs thereof) or competes against the exemplary antibody from binding to the Galectin-9 antigen. An “epitope” refers to the site on a target antigen that is ized and bound by an antibody. The site can be entirely composed of amino acid components, entirely composed of chemical modifications of amino acids of the protein (e.g., glycosyl moieties), or composed of combinations f. Overlapping epitopes include at least one common amino acid residue. An epitope can be linear, which is typically 6-15 amino acids -lO9- in length. Alternatively, the epitope can be conformational. The epitope to which an antibody binds can be determined by routine technology, for example, the epitope mapping method (see, e. g., descriptions below). An antibody that binds the same epitope as an exemplary antibody described herein may bind to exactly the same epitope or a ntially overlapping epitope (e.g., containing less than 3 non-overlapping amino acid residue, less than 2 non-overlapping amino acid residues, or only 1 non-overlapping amino acid residue) as the ary antibody.
Whether two antibodies e t each other from binding to the e antigen can be determined by a competition assay, which is well known in the art.
In some embodiments, the anti-Galectin-9 antibody may bind to an epitope at least a segment of which is in CRDl of a galectin-9 protein (e.g., a human galectin-9 or a mouse galectin-9). In some embodiments, the antibody may bind an epitope which is entirely within the CRDl of the Galectin-9 protein. In some embodiments, the antibody may bind an epitope which is partially within the CRDl of the Galectin-9 protein. In some embodiments, the e to which the anti-Galectin antibody binds is a linear epitope. In some embodiments, the epitope to which the anti-Galectin antibody binds is a conformational epitope.
In some ments, the anti-Galectin-9 antibody may bind an epitope at least a segment of which is in CRD2 of a Galectin-9 protein (e.g., a human galectin-9 or a mouse galectin-9). In some ments, the anti-Galectin-9 antibody may bind an epitope which is entirely within the CRD2 of the Galectin-9 protein. In some specific embodiments in which the 2O anti-Galectin-9 antibody binds an epitope partially or entirely within CDR2, the antibody binds an epitope comprising at least residue W309. In some specific embodiments, in which the anti- Galectin-9 antibody binds an epitope partially or entirely within CDR2, the epitope to which the anti-Galectin-9 antibody binds does not contain one or more of R253, R271, Y330, R334, R341 and Y236 of SEQ ID NO: 1. In some embodiments, the epitope to which the anti-Galectin antibody binds is a linear e encompassing e W3 09. In some ments, the epitope to which the anti-Galectin antibody binds is a conformational epitope comprising W3 09.
In some examples, the anti-Galectin-9 antibody comprises the same VH and/or VL CDRs as an exemplary antibody described . Two antibodies having the same VH and/or VL CDRs means that their CDRs are cal when ined by the same approach (e.g., the Kabat approach or the Chothia approach as known in the art). Such anti-Galectin-9 antibodies may have the same VH, the same VL, or both as compared to an exemplary antibody described herein.
Two heavy chain variable regions (or two light chain variable regions) having the same CDRs means that the CDRs in the two heavy chain variable regions (or light chain variable -llO- regions) as determined by the same numbering scheme are identical. Exemplary numbering schemes for determining antibody CDRs include the “Kabat” numbering scheme (Kabat et al. (1991), 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md), the “Chothia” numbering scheme (Al-Lazikani et al., (1997) J1Vfl3 273,927-948), the “Contact” numbering scheme (MacCallum et al., J. Mol. Biol. 262:732-745 (1996)), the “IMGT” numbering scheme (Lefranc M P et al., Dev Comp Immunol, 2003 January, 27(1):55-77), and the “AHo” numbering scheme (Honegger A and Pluckthun A, J Mol Biol, 2001 Jun. 8, 309(3):657-70). As known to those skilled in the art, the CDR s of the exemplary antipKal and anti-FXII antibodies identified herein are determined by the ia” ing scheme, which is used as an example.
Also within the scope of the present disclosure are onal variants of any of the exemplary anti-Galectin-9 antibodies as disclosed herein. Such functional variants are substantially similar to the exemplary antibody, both structurally and functionally. A functional variant comprises substantially the same VH and VL CDRs as the exemplary antibody. For example, it may comprise only up to 5 (e.g., 4, 3, 2, or 1) amino acid residue variations in the total CDR regions of the antibody and binds the same epitope of Galectin-9 with ntially r aff1nity (e.g., having a K13 value in the same order). Alternatively or in addition, the amino acid residue ions are conservative amino acid residue tutions. As used herein, a “conservative amino acid substitution” refers to an amino acid substitution that does not alter the relative charge or size characteristics of the protein in which the amino acid substitution is made. ts can be prepared according to methods for altering ptide sequence known to one of ordinary skill in the art such as are found in references which compile such s, e. g. Molecular Cloning: A Laboratory Manual, J. Sambrook, et al., eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1989, or Current Protocols in Molecular Biology, F.M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York.
Conservative substitutions of amino acids include substitutions made amongst amino acids within the following groups: (a) M, I, L, V, (b) F, Y, W, (c) K, R, H, (d) A, G, (e) S, T, (f) Q, N, and (g) E, D.
The nt identity” of two amino acid sequences is determined using the algorithm of Karlin and Altschul Proc. Natl. Acad. Sci. USA 87:2264-68, 1990, modified as in Karlin and Altschul Proc. Natl. Acad. Sci. USA 90:5873-77, 1993. Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. J. Mol. Biol. 215:403-10, 1990. BLAST n searches can be med with the XBLAST program, score=50, wordlength=3 to obtain amino acid ces homologous to the protein molecules of interest. -lll- Where gaps exist between two sequences, Gapped BLAST can be utilized as described in Altschul et al., Nucleic Acids Res. 25(17):3389-3402, 1997. When utilizing BLAST and Gapped BLAST ms, the default parameters of the tive programs (e.g., XBLAST and NBLAST) can be used. The anti-Galectin-9 antibody may comprise a heavy chain le region framework derived from a subclass of germline VH fragment. Such germline VH regions are well known in the art. See, e.g., the HVIGT database (www.imgt.org) or at www.vbase2.org/vbstat.php. Examples include the IGHVl subfamily (e.g., IGHVl-2, 3, IGHVl-8, IGHVl-lS, 24, IGHVl-45, IGHVl-46, IGHVl-58, and IGHVl-69), the IGHV2 subfamily (e.g., IGHV2-5, IGHV2-26, and IGHV2-70), the IGHV3 subfamily (e.g., IGHV3-7, IGHV3-9, l l, l3, IGHV3-15, IGHV3-20, IGHV3-21, IGHV3-23, IGHV3-30, IGHV3-33, IGHV3-43, IGHV3-48, IGHV3-49, IGHV3-53, IGHV3-64, IGHV3-66, IGHV3-72, and IGHV3-73, IGHV3-74), the IGHV4 subfamily (e.g., IGHV4-4, IGHV4-28, IGHV4-31, IGHV4-34, IGHV4-39, IGHV4-59, IGHV4-61, and IGHV4-B), the IGHV subfamily (e.g., IGHVS-Sl, or IGHV6-l), and the IGHV7 subfamily (e.g., IGHV7l).
Alternatively or in addition, the anti-Galectin-9 dy may comprise a light chain variable region that contains a framework derived from a germline VK nt. Examples include an IGKVl framework (e.g., IGKVl-OS, IGKVl-lZ, IGKVl-27, IGKVl-33, or IGKVl- 39), an IGKV2 framework (e.g., 28), an IGKV3 framework (e.g., IGKV3-l l, IGKV3- , or IGKV3 -20), and an IGKV4 framework (e.g., l). In other instances, the anti- Galectin-9 antibody may comprise a light chain variable region that contains a framework derived from a germline V?» nt. es include an IGM framework (e.g., IGKVl-3 6, IGKVl-40, IGKVl-44, IGKVl-47, IGKVl-Sl), an IGKZ framework (e.g., IGKV2-8, IGKV2-l l, IGKV2-l4, IGKV2-18, IGKV2-23,), an IGK3 framework (e. g., IGKV3-l, IGKV3-9, 10, IGKV3-12, IGKV3-l6, IGKV3-l9, IGKV3-21, IGKV3-25, IGKV3-27,), an IG7t4 framework (e. g., 3, 60, IGKV4-69,), an IGXS framework (e.g., IGKV5-39, IGKV5-45,), an IGX6 framework (e.g., IGXV6-57,), an IGM framework (e.g., IGKV7-43, 46, ), an IGKS framework (e.g., IGKV8-6l), an IGK9 framework (e.g., IGXV9-49), or an IGMO framework (e.g., Iva10—54).
In some embodiments, the heavy chain of any of the anti-Galectin-9 antibodies as described herein may further comprise a heavy chain constant region (CH) or a portion thereof (e.g., CH1, CH2, CH3, or a combination thereof). The heavy chain constant region can be of any suitable origin, e.g., human, mouse, rat, or rabbit. In one specific example, the heavy chain constant region is from a human IgG (a gamma heavy chain) of any IgG subfamily as described herein.
In some embodiments, the heavy chain constant region of the antibodies described herein may comprise a single domain (e.g., CH1, CH2, or CH3) or a combination of any of the single s, of a constant region (e.g., SEQ ID NO: 419-423). In some embodiments, the light chain constant region of the antibodies described herein may comprise a single domain (e.g., CL), of a constant region (e.g., SEQ ID NO: 418). Exemplary light and heavy chain sequences are listed below. The hIgGl LALA sequence includes two mutations, L234A and L23 5A, which suppress chR binding as well as a P329G mutation to h complement Clq g, thus hing all immune effector functions. These mutations are underlined and bolded in the sequences listed below. The hIgG4 Fab Arm Exchange Mutant ce includes a mutation to suppress Fab Arm Exchange (S228P), ined and bolded. The light chain sequence for G9.2-l7 is identical among all G9.l7 constructs. Similarly, the light chain sequence for G9. l-8ml3 is identical among all G9.l-8ml3 constructs. Bolded residues are the VH and VL regions. A IL2 signal sequence (MYRMQLLSCIALSLALVTNS, SEQ ID NO: 29) is located N—terminally of the variable region. It is used in sion vectors, which is cleaved during secretion and thus not in the mature antibody molecule. The mature n (after secretion) starts with "EVQ" for the heavy chain and "DEVI" for the light chain.
Exemplary Heavy and Light Chain ces 2O G9.2—17 hIgGl Heavy Chain (SEQ ID NO: 157) EVQLVESGGGLVQPGGSLRLSCAASGFTVSSSSIHWVRQAPGKGLEWVAYISSSSGYTYYADSVKGRFTI SADTSKNTAYLQMNSLRAEDTAVYYCARYWSYPSWWPYRGMDYWGQGTLVTVSSASTKGPSVFPLAPSS< STSGGTAALGCLV<DYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVW H<PSNT<VD<KVEP<SCD<T{TCPPCPAPELLGGPSVFLFPPKP{DTLM SRTPfiVTCVVVDVSHfiDPfiV KFNWYVDGVEVHWA<T A352W RfifiQYNSTYRVVSVLTVL{QDWLNGKIYKCKVSNKALPAP:|4 fiKT SKA<GQP RTPQVYTTPPSR'fiWT_J. QVSLTCLVKGEYPSD AVfiWfiSNGQPIWNYKTTPPVLDSDGSFFLYS{LTVD:|4 KSRWQQGNVFSCSVWHL‘J LIWHYTQKSLSLSPGK* __Light Chain (SEQ ID NO: 108) 3O DIQMTQSPSSLSASVGDRVTITCRASQSVSSAVAWYQQKPGKAPKLLIYSASSLYSGVPSRFSGSRSGTD FTLTISSLQPEDFATYYCQQSSTDPITFGQGTKVEIKRTVAAPSVE"EPPSDEQLKSGTASVVCLLNNFY PREAKVQWKVDNALQSGNSQfiSVTfiQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFN RGEC* G9.2—17 hIgGl LALA Heavy Chain (SEQ ID NO: 210) EVQLVESGGGLVQPGGSLRLSCAASGFTVSSSSIHWVRQAPGKGLEWVAYISSSSGYTYYADSVKGRFTI SADTSKNTAYLQMNSLRAEDTAVYYCARYWSYPSWWPYRGMDYWGQGTLVTVSSASTKGPSVFPLAPSS< STSGGTAALGCLV<DYFPIEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVW H<PSNT<VD<KVEP<SCD<T{TCPPCPAPEAAGGPSVFLFPPKP{DTTM SRTP*VTCVVVDVSHfi3PfiV 4O KFNWYVDGVEVHWA<T A352W RnflQYNSTYRVVSVLTVL{QDWLNGKIYKCKVSNKALEAP:|4 flKT SKA<GQP RTPQVYTTPPSR'fiWT QVSLTCLVKGEYPSD AVfiWfiSNGQPIWNYKTTPPVLDSDGSFFLYS{LTVD:| _J. 4 KSRWQQGNVFSCSVWHL‘J LdWHYTQKSLSLSPGK* -ll3- Light Chain (SEQ ID NO: 108) DIQMTQSPSSLSASVGDRVTITCRASQSVSSAVAWYQQKPGKAPKLLIYSASSLYSGVPSRFSGSRSGTD FTLTISSLQPEDFATYYCQQSSTDPITFGQGTKVEIKRTVAAPSVELEPPSDLQLKSGTASVVCLLNNFY PRLAKVQWKVDNALQSGNSQLSVTLQDSKDSTYSLSSTLTLSKADYLKHKVYACLVTHQGLSSPVTKSFN RGLC* G9.2—17 hIgG4 Heavy Chain (SEQ ID NO: 263) SGGGLVQPGGSLRLSCAASGFTVSSSSIHWVRQAPGKGLEWVAYISSSSGYTYYADSVKGRFTI SADTSKNTAYLQMNSLRAEDTAVYYCARYWSYPSWWPYRGMDYWGQGTLVTVSSASTKGPSVFPLAPCSR AALGCLV<DYFPLPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGT{TYTCNVID <VD<RVLS{YGPPCPSCPAPILFLGGPSVFLFPPKP{DTLM TCVVVDVSQLDPL'VQt'W WYVDGVLV{WAKT<PRL'Qt'NSTYRVVSVLTVL{QDWLNGKIYKCKVSNKGLPSS:|L LKT SKA<GQPRLP QVYTT.PPSQ'L'MT<WQVSLTCLVKGE'YPSD AVLWLSNGQPIWNYKTTPPVLDSDGSFFLYSRLTVDKSR:|_; L WQLGNVFSCSVM{LAL{WHYTQKSLSLSPGK* Light Chain (SEQ ID NO: 108) DIQMTQSPSSLSASVGDRVTITCRASQSVSSAVAWYQQKPGKAPKLLIYSASSLYSGVPSRFSGSRSGTD FTLTISSLQPEDFATYYCQQSSTDPITFGQGTKVEIKRTVAAPSVt'LEPPSDLQLKSGTASVVCLLNNFY PRLAKVQWKVDNALQSGNSQ*‘SVTLQDSKDSTYSLSSTLTLSKADYLKHKVYACILVTHQGLSSPVTKSFN RGLC* G9.2—17 hIgG4 Fab Arm Exchange mut Heavy Chain (SEQ ID NO: 316) EVQLVESGGGLVQPGGSLRLSCAASGFTVSSSSIHWVRQAPGKGLEWVAXISSSSGYTYYADSVK GRFTISADTSKNTAYLQMNSLRAEDTAVYYCARYWSYPSWWPYRGMDYWGQGTLVTVSSASTKGPSVFPL APCSKSTSIESTAALGCLV DYFPLPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGT{TY TCNVIDHKPSNT<VD<KVL AMA{YGPPCPPCPAPLFLGGPSVFLFPPKP{DTTIM SRTP*VTCVVVDVSQLDP LVQFWWYVIDGVLViWAKT 'NSTYRVVSVLTVL{QDWLNGKIYKCKVSNKGLPSS:|L LKT SKA<G QPRILPQVYTTPPSQLLMT 2QVSLTCLVKGE'YPSD AVLWLSNGQPIWNYKTTPPVLDSDGSFFLYSKLT:|L 3O VDKSKWQLGNVFSCSVMiIL‘JALiWHYTQKSLSLSPGK* Light Chain (SEQ ID NO: 108) DIQMTQSPSSLSASVGDRVTITCRASQSVSSAVAWYQQKPGKAPKLLIYSASSLYSGVPSRFSGSRSGTD FTLTISSLQPEDFATYYCQQSSTDPITFGQGTKVEIKRTVAAPSVt'LEPPSDLQLKSGTASVVCLLNNFY PRLAKVQWKVDNALQSGNSQ*SVTLQDSKDSTYSLSSTLTLSKADYLKHKVYACLVTHQGLSSPVTKSFN RGLC* G9.1—8m13 hIgGl Heavy Chain (SEQ ID NO: 136) EVQLVESGGGLVQPGGSLRLSCAASGFTVSSSSIHWVRQAPGKGLEWVAYIYPYSSSSSYADSVKGRFTI 40 SADTSKNTAYLQMNSLRAEDTAVYYCARYSTYSSKWVWGMDYWGQGTLVTVSSASTKGPSVFPLAPSS{S TSGGTAALGCLV{DYFPLPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTY:CNVWH {PSNT<VD<KVLP<SCD<T{TCPPCPAPLLLGGPSVFLFPPKP{DTLM SRTPLVTCVVVDVSHLDPLVK FNWYVDGVLVHWA<T AAPRLLQYNSTYRVVSVLTVL{QDWLNGKIYKCKVSNKALPAP:|L LKT SKA<GQPR LPQVYTTPPSR'LWT WQVSLTCLVKGEYPSD AVLWLSNGQPIWNYKTTPPVLDSDGSFFLYS{LTVDK:|_; L 45 SRWQQGNVFSCSVWHIAL{WHYTQKSLSLSPGK*L‘J Light Chain (SEQ ID NO: 108) DIQMTQSPSSLSASVGDRVTITCRASQSVSSAVAWYQQKPGKAPKLLIYSASSLYSGVPSRFSGSRSGTD FTLTISSLQPEDFATYYCQQSYYDSNPITFGQGTKVEIKRTVAAPSVt'LEPPSDLQLKSGTASVVCLLNN 50 FYPRLAKVQWKVDNALQSGNSQ*SVTLQDSKDSTYSLSSTLTLSKADYLKHKVYACLVTHQGLSSPVTKS FNRGLC* —114— G9.1—8m13 hIgGl LALA Heavy Chain (SEQ ID NO: 189) EVQLVESGGGLVQPGGSLRLSCAASGFTVSSSSIHWVRQAPGKGLEWVAYIYPYSSSSSYADSVKGRFTI SADTSKNTAYLQMNSLRAEDTAVYYCARYSTYSSKWVWGMDYWGQGTLVTVSSASTKGPSVFPLAPSS{S TSGGTAALGCLV{DYFPLPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTY:CNVWH {PSNT<VD<KVLP<SCD<T{TCPPCPAPLAAGGPSVFLFPPKP{DTLM SRTPLVTCVVVDVSHLDPLVK FNWYVDGVLVHWA<T AAPRLLQYNSTYRVVSVLTVL{QDWLNGKIYKCKVSNKALEAP:|L LKT SKA<GQPR LPQVYTTPPSR'LWT WQVSLTCLVKGEYPSD AVLWLSNGQPIWNYKTTPPVLDSDGSFFLYS{LTVDK:|_J. L SRWQQGNVFSCSVWHIALiWHYTQKSLSLSPGK*L‘J IO __Light Chain (SEQ ID NO: 108) DIQMTQSPSSLSASVGDRVTITCRASQSVSSAVAWYQQKPGKAPKLLIYSASSLYSGVPSRFSGSRSGTD FTLTISSLQPEDFATYYCQQSYYDSNPITFGQGTKVEIKRTVAAPSVE'LEPPSDLQLKSGTASVVCLLNN FYPRLAKVQWKVDNALQSGNSQLSVTLQDSKDSTYSLSSTLTLSKADYLKHKVYACLVTHQGLSSPVTKS FNRGLC* G9.1—8m13 hIgG4 Heavy Chain (SEQ ID NO: 242) EVQLVESGGGLVQPGGSLRLSCAASGFTVSSSSIHWVRQAPGKGLEWVAXIYPYSSSSSYADSVKGRFTI NTAYLQMNSLRAEDTAVYYCARYSTYSSKWVWGMDYWGQGTLVTVSSASTKGPSVFPLAPCSKS TSLSTAALGCLV{DYFPLPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGT{TYTCNVDH KPSNT<VD<KVLS{YGPPCPSCPAPILFLGGPSVFLFPPKP{DTLM SRTPLVTCVVVDVSQLDPL'VQE'WW YVDGVLViWAKT<PRL'Qt'NSTYRVVSVLTVL{QDWLNGKIYKCKVSNKGLPSS:|L LKT SKA<GQPRLPQ VYTTPPSQLLMT 3:2L‘J QVSLTCLVKGE'YPSD NGQPIWNYKTTPPVLDSDGSFFLYSKLTVDKSKW:|L QLGNVFSCSVMiI LiWHYTQKSLSLSPGK* Light Chain (SEQ ID NO: 108) DIQMTQSPSSLSASVGDRVTITCRASQSVSSAVAWYQQKPGKAPKLLIYSASSLYSGVPSRFSGSRSGTD FTLTISSLQPEDFATYYCQQSYYDSNPITFGQGTKVEIKRTVAAPSVELEPPSDLQLKSGTASVVCLLNN FYPRLAKVQWKVDNALQSGNSQLSVTLQDSKDSTYSLSSTLTLSKADYLKHKVYACLVTHQGLSSPVTKS FNRGLC* G9.1—8m13 hIgG4 Fab Arm Exchange mut Heavy Chain (SEQ ID NO: 295) EVQLVESGGGLVQPGGSLRLSCAASGFTVSSSSIHWVRQAPGKGLEWVAYIYPYSSSSSYADSVKGRFTI SADTSKNTAYLQMNSLRAEDTAVYYCARYSTYSSKWVWGMDYWGQGTLVTVSSASTKGPSVFPLAPCSRS ALGCLV{DYFPLPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGT{TYTCNVDH KPSNT<VD<RVLS{YGPPCPPCPAPLFLGGPSVFLFPPKP{DTTM SRTPLVTCVVVDVSQLDPL'VQE'WW YVDGVLViWAKT<PRLQt'NSTYRVVSVLTVL{QDWLNGKLYKCKVSNKGLPSS LKT SKA<GQPRLPQ VYTTPPSQLLMT QVSLTCLVKGE'YPSD AVLWLSNGQPLWNYKTTPPVLDSDGSFFLYSRLTVDKSRW QLGNVFSCSVMII3:2L‘J LiWHYTQKSLSLSPGK* Light Chain (SEQ ID NO: 108) DIQMTQSPSSLSASVGDRVTITCRASQSVSSAVAWYQQKPGKAPKLLIYSASSLYSGVPSRFSGSRSGTD SLQPEDFATYYCQQSYYDSNPITFGQGTKVEIKRTVAAPSVE'LEPPSDLQLKSGTASVVCLLNN FYPRLAKVQWKVDNALQSGNSQLSVTLQDSKDSTYSLSSTLTLSKADYLKHKVYACLVTHQGLSSPVTKS 45 FNRGLC In some embodiments, the alectin 9 antibody has a light chain comprising, consisting essentially of, or consisting of SEQ ID NO: 108. In some embodiments, the anti- Galectin 9 antibody has a heavy chain comprising, ting essentially of, or consisting of any 50 one of the sequences selected from the group consisting of SEQ ID NO: 295, 242, 189, 157, -llS- 210, 263, 316, and 136. In some embodiments, the alectin 9 dy has a light chain comprising, consisting essentially of, or consisting of SEQ ID NO: 108 and a heavy chain comprising, consisting essentially of, or consisting of any one of the sequences selected from the group consisting of SEQ ID NO: 295, 242, 189, 157, 210, 263, 316, and 136. In some embodiments, the anti-Galectin 9 antibody has a light chain comprising SEQ ID NO: 108 and a heavy chain comprising any one of the sequences selected from the group consisting of SEQ ID NO: 295, 242, 189, 157, 210, 263, 316, and 136. In some embodiments, the anti-Galectin 9 antibody has a light chain consisting essentially of SEQ ID NO: 108 and a heavy chain consisting essentially of any one of the sequences selected from the group consisting of SEQ ID NO: 295, 242, 189, 157, 210, 263, 316, and 136. In some embodiments, the anti-Galectin 9 antibody has a light chain consisting of SEQ ID NO: 108 and a heavy chain consisting of any one of the sequences selected from the group consisting of SEQ ID NO: 295, 242, 189, 157, 210,263, 316, and 136.
In some embodiments, the constant region is from human IgG4. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a heavy chain IgG4 constant region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence ty to SEQ ID NO: 423. In one embodiment, the constant region of the anti- Galectin-9 antibody comprises a heavy chain IgG4 constant region comprising SEQ ID NO: 423. In one embodiment, the constant region of the anti-Galectin-9 dy ses a heavy chain IgG4 constant region consisting of SEQ ID NO: 423. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a heavy chain IgG4 constant region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence identity to SEQ ID NO: 421. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a heavy chain IgG4 constant region comprising SEQ ID NO: 421. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a heavy chain IgG4 constant region consisting of SEQ ID NO: 421.
In some embodiments, the constant region of the anti-Galectin-9 antibody ses a light chain IgG4 constant region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to SEQ ID NO: 418. In one embodiment, the constant region of the alectin-9 antibody comprises a light chain IgG4 nt region comprising SEQ ID No: 418. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a light chain IgG4 constant region consisting of SEQ ID NO: 418. In some embodiments, the constant region is from a human IgG1. In some embodiments, the constant region of the anti-Galectin-9 antibody ses a heavy chain IgGl constant region that has at -ll6- least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 419. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a heavy chain IgGl constant region comprising SEQ ID NO: 419. In one embodiment, the nt region of the anti-Galectin-9 antibody comprises a heavy chain IgG4 constant region consisting of SEQ ID NO: 419. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a light chain IgGl constant region comprising SEQ ID NO: 418. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a light chain IgG4 constant region consisting of SEQ ID NO: 418.
In some embodiments, the anti-Galectin-9 antibody comprises a modified constant region. In some embodiments, the alectin-9 antibody se a modified constant region that is immunologically inert, e.g., does not trigger complement mediated lysis, or does not stimulate antibody-dependent cell mediated cytotoxicity . ADCC activity can be assessed using methods disclosed in US. Pat. No. 5,500,362. In other embodiments, the constant region is d as described in Eur. J. Immunol. (1999) 29:2613-2624, PCT Application No. PCT/GB99/0144l, and/or UK Patent Application No. 98099518. In some embodiments, the IgG4 constant region is a mutant with reduced heavy chain ge. In some embodiments, the constant region is from a human IgG4 Fab Arm Exchange mutant S229P. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a heavy chain IgG4 constant region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to SEQ ID NO: 422. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a heavy chain IgG4 constant region sing SEQ ID NO: 422. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a heavy chain IgG4 constant region consisting of SEQ ID NO: 422. In some embodiments, the constant region of the anti-Galectin-9 antibody comprises a light chain IgG4 constant region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to SEQ ID NO: 418. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a light chain IgG4 constant region comprising SEQ ID NO: 418. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a light chain IgG4 constant region consisting of SEQ ID NO: 418. In some embodiments, the IgG is a mutant with minimal Fc receptor engagement. In one example, the constant region is from a human IgG1 LALA. In one embodiment, the constant region of the anti-Galectin-9 antibody ses a heavy chain IgGl nt region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence ty to SEQ ID NO: 420. In one embodiment, the constant region of the anti-Galectin-9 antibody -ll7- comprises a heavy chain IgGl constant region comprising SEQ ID NO: 420. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a heavy chain IgGl constant region consisting of SEQ ID NO: 420. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a light chain IgGl constant region that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to SEQ ID NO: 418. In one embodiment, the constant region of the anti-Galectin-9 antibody ses a light chain IgGl constant region comprising SEQ ID NO: 418. In one embodiment, the constant region of the anti-Galectin-9 antibody comprises a light chain IgG4 nt region consisting of SEQ ID NO: 418.
In some embodiments, the anti-Galectin -9 antibody has chains corresponding to SEQ ID NO: 88-98 (anti-Galectin-9 antibodies g to CRDl) and SEQ ID NO: 99-115 (anti- Galectin-9 antibodies binding to CRD2) for the light , The amino acid sequences of exemplary heavy chains pond to SEQ ID NO: 116-140 (hIgGl), 169-193 (hIgGl LALA), 222-246 (hIgG4), 275-299 (hIgG4 exchange mut) (anti-Galectin-9 antibodies binding to CRDl) and SEQ ID NO: 141-168 ), 194-221(hIgGl LALA), 247—274 (hIgG4), 300—327 (hIgG4 exchange mut) (anti-Galectin-9 antibodies g to CRD2) for the heavy chains. IgG LALA, IgG4 exchange mut are located in the heavy , accordingly the light chains are the same for all IgG1 and IgG4 ces disclosed herein. In some embodiments, the amino acid sequences of exemplary anti-Galectin antibody light chains correspond to ces set forth in SEQ ID NO: 88-98 and SEQ ID NO: 99-1 15.
Clone 9.l-derived Light Chains In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to any of the light chains set forth herein (or their variable regions), (e.g., light chain sequences set forth in SEQ ID NO: 88-98. In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to any of the light chains set forth herein, (e.g., light chain sequences set forth in SEQ ID NO: 88-98. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 88-98. In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 88-98. -ll8- In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 88. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 89. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 90. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a light chain sequence of SEQ ID NO: 91. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 92. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain ce of SEQ ID NO: 93. In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain ce of SEQ ID NO: 94. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof se a light chain sequence of SEQ ID NO: 95. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a light chain sequence of SEQ ID NO: 96. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain ce of SEQ ID NO: 97. In some ments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a light chain sequence of SEQ ID NO: 98.
In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 95 (or their variable regions). In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 95. In some embodiments, light chains of alectin-9 dies comprise an amino acid sequence set forth in SEQ ID NO: 95 (or their variable regions). In some embodiments, light chains of anti-Galectin-9 antibodies consist of the amino acid sequence set forth in SEQ ID NO: 95.
Clone 9.l-derived Heavy chains In some embodiments, the amino acid sequences of exemplary anti-Galectin antibody heavy chains correspond to sequences set forth in SEQ ID NO: 116-140 (hIgGl), 169-193 (hIgGl LALA), 222-246 (hIgG4), 275-299 (hIgG4 exchange mut) (anti-Galectin-9 antibodies binding to CRDl).In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and -ll9- any ent therein) sequence identity to any of the heavy chains set forth herein (or their variable regions), e.g., sequences set forth in SEQ ID NO: 116-140, 169-193, 222-246, 275-299 (anti-Galectin-9 dies binding to CRDl). In some embodiments, heavy chains of anti- Galectin-9 antibodies t of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to any of the heavy chains set forth , e.g., sequences set forth in SEQ ID NO: 116-140, 3, 222-246, 275-299 (anti-Galectin-9 antibodies binding to CRDl).
In some embodiments, heavy chains of alectin-9 antibodies comprise an heavy chain amino acid sequence set forth in SEQ ID NO: 116-140, 169-193, 222-246, 275-299 (anti- Galectin-9 antibodies binding to CRDl). In some embodiments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 116-140, 169-193, 222- 246, 275-299 (anti-Galectin-9 antibodies binding to CRDl).
In some embodiments, the constant region is IgG1. In some embodiments, the anti- Galectin-9 antibodies or antigen-binding n thereof se a heavy chain sequence of SEQ ID NO: 116. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 117. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 118. In some embodiments, the anti-Galectin-9 antibodies or antigen- binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 119. In some 2O embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 120. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 121. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 122. In some ments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 123. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 124. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 125. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 126. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 127. In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 128. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 129. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 130. In some ments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 131. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 132. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 133. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 134. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 135. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 136. In some ments, the alectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 137. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 138. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 139. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 140.
In some embodiments, the constant region is IgGl LALA. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain ce of SEQ ID NO: 169. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 170. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 171. In some embodiments, the anti-Galectin-9 antibodies or antigen- binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 172. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 173. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 174. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 175. In some ments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 176. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 177. In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 178. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 179. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 180. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 181. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 182. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 183. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 184. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 185. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain ce of SEQ ID NO: 186. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 187. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 188. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 189. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 190. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 191. In some embodiments, the anti-Galectin-9 antibodies 2O or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 192. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 193.
In some embodiments, the constant region is IgG4. In some embodiments, the anti- Galectin-9 dies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 222. In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 223. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 224. In some embodiments, the anti-Galectin-9 antibodies or antigen- binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 225. In some 3O embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 226. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 227. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 228. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 229. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 230. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 231. In some embodiments, the alectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 232. In some ments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 233. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 234. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 235. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 236. In some embodiments, the anti-Galectin-9 dies or n-binding n f comprise a heavy chain sequence of SEQ ID NO: 237. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 238. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 29. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 240. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 241. In 2O some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 242. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 243. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 244. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 245. In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 246.
In some embodiments, the constant region is IgG4mut. In some embodiments, the anti- Galectin-9 antibodies or n-binding n thereof se a heavy chain sequence of SEQ ID NO: 275. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 276. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 277. In some embodiments, the anti-Galectin-9 dies or antigen- binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 278. In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 279. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 280. In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 281. In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 282. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 283. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 284. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 285. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 286. In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 287. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 288. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 289. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 290. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a 2O heavy chain sequence of SEQ ID NO: 291. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 292. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 293 In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain ce of SEQ ID NO: 294.In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 295. In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 296. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 297. In some embodiments, the anti-Galectin-9 antibodies 3O or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 298. In some embodiments, the alectin-9 antibodies or antigen-binding portion f se a heavy chain sequence of SEQ ID NO: 299.
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any —124— increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 136 (or its variable region). In some embodiments, heavy chains of alectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 136. In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 136 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 136.
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 189 (or its variable region). In some embodiments, heavy chains of alectin-9 antibodies consist of an amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to the heavy chain sequence set forth in SEQ ID NO: 189. In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 189 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies t of an amino acid sequence set forth in SEQ ID NO: 189.
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 242 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 dies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 242. In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 242 (or its variable ). In some embodiments, heavy chains of anti-Galectin-9 antibodies t of an amino acid sequence set forth in SEQ ID NO: 242.
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain ce set forth in SEQ ID NO: 295 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the heavy chain sequence set forth in SEQ ID -l25- NO: 295. In some embodiments, heavy chains of anti-Galectin-9 antibodies se an amino acid sequence set forth in SEQ ID NO: 295. In some embodiments, heavy chains of anti- Galectin-9 dies consist of an amino acid sequence set forth in SEQ ID NO: 295.
Clone 9.1 derived heavy and light chains A VH domain can comprise the amino acid sequence of any VH domain described herein fused to a human IgG, e.g., an IgGl, constant region, such as human IgG1 constant domain amino acid sequence, hIgG LALA, hIgG4, or IgG4mut .
In some embodiments, the amino acid sequences of exemplary anti-Galectin antibody light chains correspond to SEQ ID NO: 88 -98, orthe amino acid sequences of the ary anti-Galectin antibody heavy chains correspond to SEQ ID NO: 116-140, 3,222-246, 275-299.
In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 88 and a heavy chain having a sequence selected from of SEQ ID NO: 116, 169, 222, or 275.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 89 and a heavy chain having a sequence selected from of SEQ ID NO: 117, 170, 223, or 276.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof 2O comprise a light chain having the sequence of SEQ ID NO: 90 and a heavy chain having a sequence selected from of SEQ ID NO: 118, 171, 224, or 277.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 91 and a heavy chain having a sequence selected from of SEQ ID NO: 119, 172, 225, or 278.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 92 and a heavy chain having a sequence ed from of SEQ ID NO: 120, 173, 226, or 279.
In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the ce of SEQ ID NO: 93 and a heavy chain having a sequence selected from of SEQ ID NO: 121, 174, 227, or 280.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 94 and a heavy chain having a sequence selected from of SEQ ID NO: 122, 175, 228, or 281. -l26- In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 123, 176, 229, or 282.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the ce of SEQ ID NO: 96 and a heavy chain having a sequence selected from of SEQ ID NO: 138, 191, 244, or 297.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f se a light chain having the sequence of SEQ ID NO: 97 and a heavy chain having a sequence selected from of SEQ ID NO: 139, 192, 245, or 298.
In some embodiments, the anti-Galectin-9 dies or antigen-binding portion f se a light chain having the sequence of SEQ ID NO: 98 and a heavy chain having a sequence selected from of SEQ ID NO: 140, 193, 246, or 299.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 124, 177, 230, or 283.
In some ments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 125, 178, 231, or 284.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof 2O comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 126, 179, 232, or 285.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 127,180, 233, or 286, In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 128, 181, 234, or 287.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 129, 182, 235, or 288.
In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 130, 183, 236, or 289. -l27- In some embodiments, the alectin-9 antibodies or n-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 131, 184, 237, or 290.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a light chain having the ce of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 132, 185, 238, or 291.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 133, 186, 239, o r292.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 137, 187, 240, or 293.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 138, 188, 241, or 294.
In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a sequence selected from of SEQ ID NO: 139, 189, 242, or 295.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f 2O comprise a light chain having the sequence of SEQ ID NO: 95 and a heavy chain having a ce selected from of SEQ ID NO: 140, 190, 243, or 296.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a light chain sequence of SEQ ID NO: 95 and comprise a heavy chain sequence of SEQ ID NO: 136.In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 95 (or their le regions), and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 136 (or its variable ). In some ments, light chains of anti-Galectin-9 antibodies t of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 95 and heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity -l28- to the heavy chain sequence set forth in SEQ ID NO: 136. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 95 (or their variable regions) heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 136 (or its variable region). In some embodiments, light chains of anti-Galectin-9 antibodies consist of the amino acid sequence set forth in SEQ ID NO: 95 and heavy chains of anti-Galectin-9 dies consist of an amino acid sequence set forth in SEQ ID NO: 136.
In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 95 and comprise a heavy chain sequence of SEQ ID NO: 189. In some embodiments, light chains of anti-Galectin-9 dies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the light chain sequence set forth in SEQ ID NO: 95 (or their le regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 189 (or its variable region) and heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 189 . In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that 2O has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 95. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 95 (or their variable regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 189 (or its variable region). In some embodiments, light chains of anti-Galectin-9 antibodies consist of the amino acid sequence set forth in SEQ ID NO: 95 and heavy chains of anti-Galectin-9 antibodies se an amino acid sequence set forth in SEQ ID NO: 189.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 95 and se a heavy chain sequence of SEQ ID NO: 242. In some embodiments, light chains of anti-Galectin-9 dies se an amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 95 (or their variable regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment -l29- therein) sequence identity to the heavy chain ce set forth in SEQ ID NO: 242 (or its variable region). In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 95 and heavy chains of alectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain ce set forth in SEQ ID NO: 242. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 95 (or their variable regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 242 (or its variable region). In some embodiments, light chains of anti-Galectin-9 antibodies consist of the amino acid ce set forth in SEQ ID NO: 95 and heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 242.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 95 and comprise a heavy chain sequence of SEQ ID NO: 295.In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 95 (or their variable regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain ce set forth in SEQ ID NO: 295 (or its variable region). In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain ce set forth in SEQ ID NO: 95 and heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) ce identity to the heavy chain sequence set forth in SEQ ID NO: 295. In some ments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 95 (or their variable s) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid ce set forth in SEQ ID NO: 295. In some embodiments, light chains of anti-Galectin-9 antibodies consist of the amino acid sequence set forth in SEQ ID NO: 95 and heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 295. -l30- Clone 9.2-derived Light Chains In some ments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (6g, 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to any of the light chains set forth herein (or their variable regions), (e.g., light chain sequences set forth in SEQ ID NO: 99-115). In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (6g, 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to any of the light chains set forth herein, (e.g., light chain sequences set forth in SEQ ID NO: 99-115).
In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 99-115. In some ments, light chains of anti-Galectin-9 dies t of a sequence set forth in SEQ ID NO: 99-115.
In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a light chain sequence of SEQ ID NO: 99. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 100. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 101. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a light chain sequence of SEQ ID NO: 102. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 103. In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 104. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 105. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a light chain sequence of SEQ ID NO: 106. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a light chain sequence of SEQ ID NO: 107. In some embodiments, the alectin-9 dies or antigen-binding n thereof comprise a light chain sequence of SEQ ID NO: 108. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 109. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 110. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 111. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 112. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a light chain sequence of SEQ ID NO: 113. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 114. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 115. In some embodiments, light chains of anti- Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to the light chain sequence set forth in SEQ ID NO: 108 (or their variable regions). In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 108.
In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 108 (or their variable regions). In some embodiments, light chains of anti-Galectin-9 antibodies consist set forth in SEQ ID NO: 108.
Clone 9.2-derived Heavy Chains In some embodiments, the amino acid sequences of ary anti-Galectin antibody heavy chains pond to sequences set forth in SEQ ID NO: 141-168 ), 194- 221(hIgG1 LALA), 247-274 (hIgG4), 300-327 (hIgG4 ge mut) (anti-Galectin-9 antibodies binding to CRD2) for the heavy chains.
In some embodiments, heavy chains of alectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to any of the heavy chains set forth herein (or their variable regions), e.g., ces set forth in SEQ ID NO: 141-168, 194-220, 247-274, 300-327 (anti- Galectin-9 antibodies binding to CRD2). In some embodiments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to any of the heavy chains set forth herein, e.g., sequences set forth in SEQ ID NO: 141-168, 194-220, 247-274, 300-327 (anti- in-9 antibodies binding to CRD2).
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an heavy chain amino acid sequence set forth in SEQ ID NO: 141-168, 194-220, 247-274, 300-327 (anti- Galectin-9 antibodies binding to CRD2). In some ments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 8, 194-220, 247- 274, 300-327 (anti-Galectin-9 dies binding to CRD2).
In some embodiments, the constant region is IgGl. In some ments, the anti- in-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 141. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 142. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 143. In some embodiments, the anti-Galectin-9 antibodies or antigen- binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 144. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 145. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 146. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 147. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 148. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 149. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 150. In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 151. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 152. In 2O some embodiments, the anti-Galectin-9 antibodies or n-binding portion f comprise a heavy chain sequence of SEQ ID NO: 153. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 154. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 155. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 156. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 157. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 158. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 159. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 160. In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 161. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 162. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 163. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 164. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 165. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 166. In some embodiments, the alectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 167. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 168.
In some embodiments, the constant region is IgGl LALA. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 194. In some embodiments, the alectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 195. In some embodiments, the anti-Galectin-9 dies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 196. In some embodiments, the anti-Galectin-9 antibodies or antigen- g portion thereof comprise a heavy chain sequence of SEQ ID NO: 197. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 198. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 199. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 200. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 201. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 202. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 203. In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 304. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 205. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 206. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 207. In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 208. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 209. In —134— some embodiments, the alectin-9 dies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 210. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 211. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 212. In some ments, the anti-Galectin-9 dies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 213. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 214. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 215. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 216. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 217. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 218. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 219. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 220. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 221.
In some embodiments, the constant region is IgG4. In some embodiments, the anti- 2O Galectin-9 antibodies or n-binding portion thereof se a heavy chain sequence of SEQ ID NO: 247. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 248. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 249. In some embodiments, the anti-Galectin-9 antibodies or antigen- binding portion thereof comprise a heavy chain ce of SEQ ID NO: 250. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 251. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 252. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 253. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain ce of SEQ ID NO: 254. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 255. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 256. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 257. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 258. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 259. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 260. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 261. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 262. In some ments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 263. In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 264. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 265. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 266. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 267. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 268. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a 2O heavy chain sequence of SEQ ID NO: 269. In some embodiments, the anti-Galectin-9 dies or n-binding portion thereof comprise a heavy chain ce of SEQ ID NO: 270. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 271. In some embodiments, the alectin-9 antibodies or antigen-binding n thereof se a heavy chain sequence of SEQ ID NO: 272. In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 273. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a heavy chain sequence of SEQ ID NO: 274.
In some embodiments, the nt region is IgG4 mut. In some embodiments, the anti- Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 300. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 301. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 302. In some embodiments, the anti-Galectin-9 antibodies or antigen- binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 303. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 304. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 305. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 306. In some embodiments, the alectin- 9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 307. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a heavy chain sequence of SEQ ID NO: 308. In some embodiments, the anti- Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 309. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 310. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 311. In some embodiments, the anti-Galectin-9 antibodies or antigen- g portion thereof comprise a heavy chain sequence of SEQ ID NO: 312. In some ments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 313. In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 314. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 315. In some embodiments, the anti-Galectin- 9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 316. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a heavy chain sequence of SEQ ID NO: 317. In some embodiments, the anti- Galectin-9 dies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 318. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 319. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 320. In some embodiments, the anti-Galectin-9 antibodies or antigen- binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 321. In some embodiments, the anti-Galectin-9 dies or antigen-binding n thereof se a heavy chain sequence of SEQ ID NO: 322. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 323. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 324. In some embodiments, the anti-Galectin- 9 antibodies or n-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 325. In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a heavy chain sequence of SEQ ID NO: 326.
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 157 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain ce set forth in SEQ ID NO: 157.In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 157 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 157.
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 210 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to the heavy chain ce set forth in SEQ ID NO: 210. In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino 2O acid sequence set forth in SEQ ID NO: 210 (or its variable ). In some embodiments, heavy chains of anti-Galectin-9 antibodies t of an amino acid sequence set forth in SEQ ID NO: 2lO.In some ments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any ent therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 263 (or its variable ). In some embodiments, heavy chains of anti-Galectin-9 dies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 263.
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 263 (or its le region). In some embodiments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 263.
In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any -l38- increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 316 (or its variable region). In some embodiments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 316. In some embodiments, heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 316 (or its variable ). In some ments, heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 3 16.
Clone 9.2 Derived Heavy and Light Chains In some embodiments, the amino acid sequences of exemplary anti-Galectin antibody light chains correspond to SEQ ID NO: 99-108, and the amino acid sequences of the ary anti-Galectin antibody heavy chains correspond to SEQ ID NO: 141-168, 194-221,249-274, 300-327.
In some embodiments, the alectin-9 antibodies or antigen-binding n thereof comprise a light chain having the sequence of SEQ ID NO: 99 and a heavy chain having a sequence selected from SEQ ID NO: 141, 194, 247, or 300.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 91 and a heavy chain having a sequence selected from SEQ ID NO: 142, 195, 248, or 301.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 91 and a heavy chain having a sequence selected from SEQ ID NO: 143, 196, 249, or 302.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 91 and a heavy chain having a sequence selected from SEQ ID NO: 144, 197, 250, or 303.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding n thereof comprise a light chain having the sequence of SEQ ID NO: 100 and a heavy chain having a ce selected from SEQ ID NO: 145, 198, 251, or 304.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a light chain having the sequence of SEQ ID NO: 101 and a heavy chain having a sequence ed from SEQ ID NO: 146, 199, 252, or 305.
In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 102 and a heavy chain having a sequence selected from SEQ ID NO: 147, 200, 253, or 306.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 103 and a heavy chain having a sequence selected from SEQ ID NO: 148, 201, 254, or 307.
In some embodiments, the alectin-9 dies or antigen-binding portion thereof comprise a light chain having the ce of SEQ ID NO: 104 and a heavy chain having a sequence selected from SEQ ID NO: 149, 202, 255, or 308.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 105 and a heavy chain having a sequence selected from SEQ ID NO: 150, 203, 256, or 309.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 106 and a heavy chain having a ce selected from SEQ ID NO: 151, 204, 257, or 310.
In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a light chain having the ce of SEQ ID NO: 107 and a heavy chain having a sequence selected from SEQ ID NO: 152, 205, 258, or 311.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 99 and a heavy chain having a sequence selected from SEQ ID NO: 153, 206, 259, or 312.
In some embodiments, the alectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 91 and a heavy chain having a sequence selected from SEQ ID NO: 154, 207, 260, or 313.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 100 and a heavy chain having a sequence selected from SEQ ID NO: 155, 208, 261, or 314.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion f comprise a light chain having the sequence of SEQ ID NO: 91 and a heavy chain having a sequence selected from SEQ ID NO: 156, 209, 262, or 315.
In some embodiments, the anti-Galectin-9 antibodies or n-binding portion f comprise a light chain having the sequence of SEQ ID NO: 108 and a heavy chain having a sequence selected from SEQ ID NO: 157, 210, 263, or 316.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 108 and a heavy chain having a sequence selected from SEQ ID NO: 158, 211, 264, or 317. —140— In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 91 and a heavy chain having a sequence selected from SEQ ID NO: 159, 212, 265, or 318.
In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 109 and a heavy chain having a sequence selected from SEQ ID NO: 160, 213, 266, or 319.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 108 and a heavy chain having a sequence ed from SEQ ID NO: 161, 214, 267, or 320.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 110 and a heavy chain having a sequence ed from SEQ ID NO: 162, 215, 268, or 321.
In some embodiments, the alectin-9 dies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 111 and a heavy chain having a sequence selected from SEQ ID NO: 163, 216, 269, or 322.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 112 and a heavy chain having a sequence ed from SEQ ID NO: 164, 217, 270, or 323.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 113 and a heavy chain having a sequence selected from SEQ ID NO: 165, 218, 271, or 324.
In some embodiments, the anti-Galectin-9 dies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 114 and a heavy chain having a sequence selected from SEQ ID NO: 166, 219, 272, or 325.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the ce of SEQ ID NO: 115 and a heavy chain having a sequence selected from SEQ ID NO: 167, 220, 273, or 326.
In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain having the sequence of SEQ ID NO: 108 and a heavy chain having a sequence selected from SEQ ID NO: 168, 221, 274, or 327.
In some embodiments, the anti-Galectin-9 antibodies or n-binding portion thereof comprise a light chain sequence of SEQ ID NO: 108 and comprise a heavy chain sequence of SEQ ID NO: 157. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any —141— increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 108 (or their variable regions), and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to the heavy chain sequence set forth in SEQ ID NO: 157 (or its le ). In some ments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) ce identity to the light chain sequence set forth in SEQ ID NO: 108 and heavy chains of anti-Galectin-9 antibodies consist of an amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 157. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 108 (or their variable regions) heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 157 (or its variable region). In some ments, light chains of anti-Galectin-9 antibodies consist of the amino acid sequence set forth in SEQ ID NO: 108 and heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 157.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 108 and comprise a heavy chain sequence of SEQ ID NO: 210. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment n) sequence identity to the light chain sequence set forth in SEQ ID NO: 108 (or their variable s) and heavy chains of alectin-9 antibodies se an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 210 (or its variable ) and heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 210 . In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 108. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 108 (or their variable regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 210 (or its variable region). In some embodiments, light chains of anti-Galectin-9 antibodies consist of the amino —142— acid sequence set forth in SEQ ID NO: 108 and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 210.
In some embodiments, the anti-Galectin-9 antibodies or antigen-binding portion thereof se a light chain sequence of SEQ ID NO: 108 and comprise a heavy chain sequence of SEQ ID NO: 263. In some embodiments, light chains of anti-Galectin-9 dies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 108 (or their variable regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 263 (or its variable region). In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 108 and heavy chains of anti-Galectin-9 antibodies consist of an amino acid ce that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain ce set forth in SEQ ID NO: 263. In some embodiments, light chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 108 (or their variable regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 263 (or its variable ). In some embodiments, light chains of alectin-9 antibodies consist of the amino acid sequence set forth in SEQ ID NO: 108 and heavy chains of alectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 263.
In some ments, the anti-Galectin-9 antibodies or antigen-binding portion thereof comprise a light chain sequence of SEQ ID NO: 108 and comprise a heavy chain sequence of SEQ ID NO: 3 l6.In some ments, light chains of anti-Galectin-9 dies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 108 (or their variable regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence ty to the heavy chain sequence set forth in SEQ ID NO: 3 16 (or its variable region). In some embodiments, light chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the light chain sequence set forth in SEQ ID NO: 108 and heavy chains of anti-Galectin-9 antibodies consist of an amino acid sequence that has at —143— least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, 99% and any increment therein) sequence identity to the heavy chain sequence set forth in SEQ ID NO: 316. In some embodiments, light chains of alectin-9 antibodies comprise an amino acid ce set forth in SEQ ID NO: 108 (or their variable regions) and heavy chains of anti-Galectin-9 antibodies comprise an amino acid sequence set forth in SEQ ID NO: 316. In some embodiments, light chains of anti-Galectin- 9 antibodies consist of the amino acid ce set forth in SEQ ID NO: 108 and heavy chains of alectin-9 antibodies consist of an amino acid sequence set forth in SEQ ID NO: 316.
In some embodiments, the anti-Galectin-9 antibody comprises an IgGl heavy chain having the ce of SEQ ID NO: 157 and a light chain having the sequence of SEQ ID NO: 108. In some embodiments, the alectin-9 antibody comprises an IgGl heavy chain having the sequence of SEQ ID NO: 210 and a light chain having the sequence of SEQ ID NO: 108. In some embodiments, the anti-Galectin-9 antibody comprises an IgG4 heavy chain having the sequence of SEQ ID NO: 263 and a light chain having the ce of SEQ ID NO: 108. In some embodiments, the anti-Galectin-9 antibody ses an IgG4 heavy chain having the sequence of SEQ ID NO: 316 and a light chain having the sequence of SEQ ID NO: 108. In some embodiments, the anti-Galectin-9 antibody comprises an IgGl heavy chain having the sequence of SEQ ID NO: 136 and a light chain having the sequence of SEQ ID NO: 108. In some embodiments, the anti-Galectin-9 antibody comprises an IgGl heavy chain having the sequence of SEQ ID NO: 189 and a light chain having the sequence of SEQ ID NO: 108. In some embodiments, the anti-Galectin-9 antibody comprises an IgG4 heavy chain having the sequence of SEQ ID NO: 242 and a light chain having the sequence of SEQ ID NO: 108. In some embodiments, the anti-Galectin-9 antibody comprises an IgG4 heavy chain having the sequence of SEQ ID NO: 295 and a light chain having the sequence of SEQ ID NO: 108.
Antibody heavy and light chain constant regions are well known in the art, e.g., those provided in the IMGT database (www.imgt.org) or at www.vbase2.org/vbstat.php., both of which are incorporated by reference herein.
Preparation of Anti-Galectin-9 Antibodies Antibodies capable of binding Galectin-9 as described herein can be made by any method known in the art. See, for example, Harlow and Lane, (1998) dies: A Laboratory , Cold Spring Harbor Laboratory, New York.
In some embodiments, antibodies specific to a target antigen (e.g., Galectin-9 or a CRD thereof) are made by conventional hybridoma logy. The full-length target antigen or a fragment thereof, optionally coupled to a carrier protein such as KLH, can be used to immunize —144— a host animal for generating antibodies binding to that antigen. The route and le of immunization of the host animal are generally in keeping with established and conventional ques for antibody ation and production, as further described herein. General techniques for production of mouse, humanized, and human antibodies are known in the art and are described . It is contemplated that any mammalian subject including humans or antibody producing cells therefrom can be manipulated to serve as the basis for production of mammalian, including human hybridoma cell lines. Typically, the host animal is inoculated intraperitoneally, intramuscularly, orally, subcutaneously, intraplantar, and/or intradermally with an amount of gen, including as described herein.
Hybridomas can be prepared from the cytes and immortalized myeloma cells using the general somatic cell hybridization technique of Kohler, B. and Milstein, C. (1975) Nature 256:495-497 or as d by Buck, D. W., et al., In Vilro, 18:377-381 (1982).
Available myeloma lines, including, but not limited to, X63-Ag8.653 and those from the Salk Institute, Cell Distribution Center, San Diego, Calif., USA, may be used in the ization.
Generally, the technique involves fusing myeloma cells and lymphoid cells using a fusogen such as polyethylene , or by electrical means well known to those skilled in the art. After the fusion, the cells are separated from the fusion medium and grown in a ive growth medium, such as hypoxanthine-aminopterin-thymidine (HAT) medium, to eliminate unhybridized parent cells. Any of the media described herein, supplemented with or without serum, can be used for culturing hybridomas that secrete monoclonal antibodies. As another alternative to the cell fusion technique, EBV immortalized B cells may be used to e the anti-Galectin-9 monoclonal antibodies described herein. The hybridomas are ed and subcloned, if desired, and supematants are assayed for anti-immunogen ty by conventional assay procedures (e.g., radioimmunoassay, enzyme immunoassay, or fluorescence immunoassay).
Hybridomas that may be used as source of antibodies encompass all derivatives, progeny cells of the parent hybridomas that produce monoclonal antibodies capable of interfering with the Galectin-9 activity. Hybridomas that produce such antibodies may be grown in vitro or in vivo using known procedures. The monoclonal antibodies may be ed from the culture media or body fluids, by conventional immunoglobulin purif1cation procedures such as ammonium sulfate precipitation, gel electrophoresis, dialysis, chromatography, and ultraflltration, if desired. Undesired activity if present, can be removed, for e, by running the preparation over adsorbents made of the immunogen attached to a solid phase and eluting or releasing the desired antibodies off the immunogen. Immunization of a host animal with a target —145— antigen or a fragment containing the target amino acid sequence conjugated to a protein that is immunogenic in the s to be immunized, e.g., keyhole limpet anin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, for example maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N- hydroxysuccinimide (through lysine es), glutaraldehyde, succinic anhydride, SOCl, or R1N=C=NR, where R and R1 are different alkyl groups, can yield a population of antibodies (e.g., onal antibodies).
If desired, an antibody (monoclonal or polyclonal) of st (e.g., produced by a hybridoma) may be sequenced and the polynucleotide sequence may then be cloned into a vector for sion or propagation. The sequence encoding the dy of interest may be ined in vector in a host cell and the host cell can then be expanded and frozen for future use. In an alternative, the polynucleotide sequence may be used for genetic manipulation to "humanize" the antibody or to improve the affinity (aff1nity maturation), or other characteristics of the dy. For example, the nt region may be engineered to more resemble human constant regions to avoid immune response if the dy is used in clinical trials and treatments in humans. It may be desirable to genetically manipulate the antibody sequence to obtain greater affinity to the target antigen and greater efficacy in inhibiting the activity of Galectin-9. It will be apparent to one of skill in the art that one or more polynucleotide s can be made to the dy and still maintain its g specificity to the target antigen.
In other embodiments, fully human antibodies are ed using commercially available mice that have been engineered to express specific human immunoglobulin proteins. Transgenic animals that are designed to produce a more desirable (e.g., fully human antibodies) or more robust immune response may also be used for generation of humanized or human antibodies.
Examples of such technology are XenomouseRTM from Amgen, Inc. (Fremont, Calif.) and HuMAb-MouseRTM and TC MouseTM from Medarex, Inc. (Princeton, N.J.). In other embodiments, antibodies are made recombinantly by phage display or yeast technology. See, for example, US. Pat. Nos. 5,565,332, 5,580,717, 743, and 6,265,150, and Winter et al., (1994) Annu. Rev. Immunol. 12:433-455. In alternate embodiments, phage display technology (McCafferty et al., (1990) Nature 348:552-553) is used to produce human antibodies and 3O antibody nts in vilro, from immunoglobulin variable (V) domain gene repertoires from unimmunized donors.
In alternate embodiments, antibodies capable of binding to the target antigens as described herein are isolated from a suitable antibody library. Antibody libraries, which contain a plurality of antibody components, can be used to identify antibodies that bind to a specific target antigen (e.g., the CRDl or CRD2 of Galectin-9 in this case) following routine selection processes as known in the art. In the selection process, an antibody library can be probed with the target n or a fragment thereof and members of the library that are capable of g to the target antigen can be isolated, typically by retention on a t. Such screening process may be med by le rounds (e.g., including both positive and negative selections) to enrich the pool of dies capable of binding to the target antigen. Individual clones of the ed pool can then be isolated and further characterized to identify those having desired binding activity and ical activity. Sequences of the heavy chain and light chain le domains can also be determined via tional methodology. There are a number of e methods known in the art to identify and isolate antibodies capable of binding to the target antigens described herein, including phage display, yeast display, ribosomal display, or mammalian display technology.
As an example, phage displays typically use a covalent linkage to bind the protein (e.g., antibody) component to a bacteriophage coat protein. The e results from translation of a nucleic acid encoding the antibody ent fused to the coat protein. The linkage can include a flexible peptide , a protease site, or an amino acid orated as a result of suppression of a stop codon. Phage display is described, for example, in US. Pat. No. ,223,409, Smith (1985) Science 228:1315-1317, WO 92/18619, WO 91/17271, WO 92/20791, WO 92/15679, WO 93/01288, WO 92/01047, WO 92/09690, WO 90/02809, de Haard et al. (1999) J. Biol. Chem 274:18218-30, Hoogenboom et al. (1998) Immunotechnology 4:1-20, Hoogenboom et al. (2000) Immunol Today 2371-8 and Hoet et al. (2005) Nat Biotechnol. 23(3)344-8. Additional suitable methods are described in Miller et al., PloS One, 2012, 7, e43746, Fellouse et al., JMol Biol, 2007, 373, 924-940. Bacteriophage displaying the protein component can be grown and harvested using standard phage preparatory methods, e.g. PEG precipitation from growth media. After selection of individual y phages, the nucleic acid encoding the selected protein components can be isolated from cells infected with the selected phages or from the phage themselves, after amplification. Individual colonies or plaques can be selected, and then the nucleic acid may be isolated and sequenced.
Other display formats include ased display (see, e.g., WO 03/029456), protein- nucleic acid fusions (see, e.g., US. Pat. No. 6,207,446), ribosome display (See, e.g., Mattheakis et al. (1994) Proc. Natl. Acad. Sci. USA 91 :9022 and Hanes et al. (2000) Nat Biotechnol. 18: 1287-92, Hanes et al. (2000) Methods Enzymol. 328:404-30, and Schaff1tzel et al. (1999) J ImmunolMethods. 23 1(1-2): 1 19-3 5), and E. coli periplasmic display (JImmunolMethods. 2005 Nov 22,PMID: 16337958). —147— After display library members are isolated for binding to the target antigen, each isolated library member can be also tested for its ability to bind to a rget molecule to evaluate its binding specificity. Examples of non-target molecules include streptavidin on magnetic beads, blocking agents such as bovine serum n, non-fat bovine milk, soy protein, any ing or target immobilizing monoclonal antibody, or non-transfected cells which do not express the target. A high-throughput ELISA screen can be used to obtain the data, for e. The ELISA screen can also be used to obtain quantitative data for binding of each library member to the target as well as for cross species reactivity to related targets or subunits of the target antigen and also under different condition such as pH 6 or pH 7.5. The non-target and target binding data are compared (e.g., using a computer and re) to identify library members that specifically bind to the target.
After selecting candidate library s that bind to a target, each ate library member can be further analyzed, e.g., to further characterize its binding properties for the target, e. g., Galectin-9. Each ate library member can be ted to one or more secondary screening assays. The assay can be for a binding property, a catalytic property, an tory property, a physiological property (e.g., xicity, renal clearance, immunogenicity), a structural property (e.g., stability, conformation, erization state) or another functional property. The same assay can be used repeatedly, but with varying conditions, e.g., to determine pH, ionic, or thermal ivities.
As appropriate, the assays can use a display library member directly, a recombinant polypeptide produced from the c acid encoding the selected polypeptide, or a synthetic peptide synthesized based on the sequence of the selected polypeptide. In the case of selected Fabs, the Fabs can be evaluated or can be modified and produced as intact IgG proteins. ary assays for binding properties are described below.
Binding proteins can also be evaluated using an ELISA assay. For example, each protein is contacted to a microtitre plate whose bottom surface has been coated with the target, e. g., a ng amount of the target. The plate is washed with buffer to remove non-specifically bound polypeptides. Then the amount of the binding protein bound to the target on the plate is determined by probing the plate with an antibody that can recognize the binding protein, e.g., a tag or constant portion of the binding protein. The antibody is linked to a detection system (e.g., an enzyme such as alkaline phosphatase or horse radish peroxidase (HRP) which produces a colorimetric product when appropriate substrates are provided).
Alternatively, the ability of a binding protein described herein to bind a target antigen can be analyzed using a homogenous assay, i.e., after all components of the assay are added, additional fluid manipulations are not required. For example, fluorescence nce energy transfer (FRET) can be used as a homogenous assay (see, for example, cz et al., US.
Patent No. 5,631,169, Stavrianopoulos, et al., US. Patent No. 4,868,103). A fluorophore label on the first molecule (e.g., the molecule identified in the fraction) is selected such that its emitted fluorescent energy can be absorbed by a fluorescent label on a second molecule (e.g., the target) if the second molecule is in proximity to the first le. The fluorescent label on the second molecule fluoresces when it absorbs to the transferred energy. Since the efficiency of energy transfer between the labels is related to the distance separating the molecules, the spatial onship between the molecules can be assessed. In a situation in which binding occurs between the molecules, the fluorescent emission of the tor’ molecule label in the assay should be maximal. A binding event that is configured for monitoring by FRET can be conveniently measured through standard etric detection means, e.g., using a fluorimeter.
By titrating the amount of the first or second binding molecule, a binding curve can be generated to estimate the equilibrium g constant.
Surface plasmon resonance (SPR) can be used to analyze the interaction of a binding n and a target antigen. SPR or Biomolecular ction Analysis (BIA) detects biospecific interactions in real time, without labeling any of the interactants. Changes in the mass at the binding surface (indicative of a binding event) of the BIA chip result in alterations of the refractive index of light near the surface (the optical phenomenon of SPR). The changes in the refractivity generate a detectable signal, which are measured as an indication of real-time reactions between biological molecules. Methods for using SPR are described, for example, in US. Patent No. 5,641,640, Raether, 1988, Surface Plasmons er Verlag, der and czky, 1991, Anal. Chem. 63:2338-2345, Szabo et al., 1995, Curr. Opin. Struct. Biol. 5699-705 and on-line resources provide by BIAcore ational AB (Uppsala, Sweden).
Information from SPR can be used to provide an accurate and quantitative e of the equilibrium dissociation constant (KD), and kinetic parameters, including Ken and Koff, for the binding of a binding protein to a target. Such data can be used to compare different biomolecules. For e, selected proteins from an sion library can be compared to identify proteins that have high affinity for the target or that have a slow Koff. This information can also be used to develop structure-activity relationships (SAR). For example, the kinetic and equilibrium binding parameters of matured versions of a parent n can be compared to the parameters of the parent protein. t amino acids at given positions can be identified that correlate with particular binding parameters, e.g., high affinity and slow Koff. This information can be combined with structural modeling (e.g., using homology modeling, energy —149— minimization, or structure determination by x-ray crystallography or NMR). As a result, an understanding of the physical interaction between the protein and its target can be formulated and used to guide other design processes.
As a further example, cellular assays may be used. Binding ns can be screened for ability to bind to cells which transiently or stably s and display the target of interest on the cell surface. For example, in-9 binding proteins can be fluorescently labeled and binding to Galectin-9 in the presence or absence of nistic antibody can be detected by a change in fluorescence intensity using flow try e.g., a FACS machine.
Antigen-binding fragments of an intact antibody (full-length antibody) can be prepared via routine methods. For example, F(ab')2 fragments can be produced by pepsin digestion of an antibody molecule, and Fab fragments that can be generated by reducing the disulfide bridges of F(ab')2 fragments.
Genetically ered antibodies, such as zed antibodies, chimeric antibodies, single-chain antibodies, and bi-specif1c antibodies, can be produced via, e.g., conventional recombinant technology. In one example, DNA encoding a monoclonal antibodies specific to a target antigen can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies). Once isolated, the DNA may be placed into one or more sion vectors, which are then transfected into host cells such as E. coli 2O cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not ise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the inant host cells. See, e.g., PCT Publication No. WO 87/04462. The DNA can then be ed, for example, by tuting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences, Morrison et al., (1984) Proc.
Nat. Acad. Sci. 81 :6851, or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. In that manner, genetically engineered antibodies, such as “chimeric” or “hybrid” antibodies, can be prepared that have the binding specificity of a target antigen.
Techniques developed for the production of “chimeric antibodies” are well known in the 3O art. See, e.g., Morrison et al. (1984) Proc. Natl. Acad. Sci. USA 81, 6851, Neuberger et al. (1984) Nature 312, 604, and Takeda et al. (1984) Nature 2.
Methods for constructing humanized dies are also well known in the art. See, e. g., Queen et al., Proc. Natl. Acad. Sci. USA, 86: 10029-10033 . In one example, variable regions of VH and VL of a parent man antibody are subjected to three-dimensional -l50- molecular modeling analysis ing methods known in the art. Next, framework amino acid es predicted to be important for the formation of the correct CDR structures are identified using the same molecular modeling analysis. In parallel, human VH and VL chains having amino acid sequences that are homologous to those of the parent non-human antibody are identified from any antibody gene database using the parent VH and VL sequences as search queries.
Human VH and VL or genes are then selected.
The CDR s within the selected human acceptor genes can be replaced with the CDR regions from the parent non-human antibody or functional variants thereof. When necessary, residues within the framework regions of the parent chain that are predicted to be important in interacting with the CDR regions (see above description) can be used to substitute for the corresponding es in the human acceptor genes.
A single-chain antibody can be prepared via recombinant technology by linking a nucleotide sequence coding for a heavy chain variable region and a nucleotide sequence coding for a light chain variable region. Preferably, a e linker is incorporated between the two variable regions. Alternatively, techniques described for the production of single chain dies (US. Patent Nos. 4,946,778 and 4,704,692) can be adapted to produce a phage or yeast scFv library and scFv clones specific to Galectin-9 can be identified from the y following routine procedures. ve clones can be subjected to further screening to identify those that t Galectin-9 activity.
Antibodies obtained following a method known in the art and described herein can be terized using methods well known in the art. For example, one method is to identify the epitope to which the antigen binds, or “epitope mapping.” There are many methods known in the art for mapping and characterizing the location of epitopes on proteins, including solving the crystal structure of an antibody-antigen complex, competition assays, gene fragment sion assays, and synthetic peptide-based assays, as described, for example, in Chapter 11 of Harlow and Lane, Using Antibodies, a Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1999. In an onal example, epitope mapping can be used to determine the ce, to which an antibody binds. The epitope can be a linear epitope, 1'.e., contained in a single stretch of amino acids, or a conformational epitope formed by a three-dimensional 3O interaction of amino acids that may not necessarily be contained in a single h (primary structure linear ce). Peptides of varying lengths (e.g., at least 4-6 amino acids long) can be isolated or synthesized (e.g., recombinantly) and used for binding assays with an antibody. In another example, the epitope to which the antibody binds can be determined in a systematic screening by using overlapping peptides derived from the target antigen sequence and determining binding by the antibody. According to the gene fragment sion assays, the open reading frame encoding the target antigen is fragmented either randomly or by specific genetic constructions and the reactivity of the expressed fragments of the n with the antibody to be tested is determined. The gene fragments may, for example, be produced by PCR and then transcribed and translated into protein in vitro, in the presence of radioactive amino acids. The binding of the dy to the radioactively labeled antigen fragments is then ined by immunoprecipitation and gel electrophoresis. Certain epitopes can also be identified by using large libraries of random peptide sequences yed on the surface of phage particles (phage libraries). Alternatively, a defined library of overlapping peptide fragments can be tested for binding to the test antibody in simple binding assays. In an additional example, mutagenesis of an antigen binding domain, domain swapping experiments and alanine scanning nesis can be performed to identify residues ed, suff1cient, and/or necessary for epitope binding. For example, domain swapping experiments can be med using a mutant of a target n in which various fragments of the Galectin-9 polypeptide have been replaced (swapped) with ces from a closely related, but antigenically distinct protein (such as another member of the B-galactoside-binding soluble lectin family). By assessing g of the antibody to the mutant Galectin-9, the importance of the particular antigen fragment to antibody binding can be assessed.
Alternatively, competition assays can be performed using other antibodies known to bind to the same antigen to determine whether an antibody binds to the same epitope as the other antibodies. Competition assays are well known to those of skill in the art.
In some examples, an anti-Galectin-9 antibody is prepared by recombinant technology as exemplified below.
Nucleic acids encoding the heavy and light chain of an anti-Galectin-9 antibody as described herein can be cloned into one sion vector, each tide sequence being in operable linkage to a le promoter. In one example, each of the tide sequences encoding the heavy chain and light chain is in operable linkage to a distinct promoter.
Alternatively, the nucleotide ces encoding the heavy chain and the light chain can be in operable e with a single promoter, such that both heavy and light chains are expressed 3O from the same promoter. When necessary, an internal ribosomal entry site (IRES) can be inserted between the heavy chain and light chain encoding sequences.
In some examples, the nucleotide sequences encoding the two chains of the antibody are cloned into two vectors, which can be introduced into the same or different cells. When the two chains are expressed in different cells, each of them can be isolated from the host cells expressing such and the isolated heavy chains and light chains can be mixed and ted under suitable conditions allowing for the formation of the antibody.
Generally, a nucleic acid sequence encoding one or all chains of an antibody can be cloned into a suitable expression vector in operable linkage with a suitable promoter using methods known in the art. For example, the nucleotide sequence and vector can be contacted, under suitable ions, with a restriction enzyme to create complementary ends on each molecule that can pair with each other and be joined together with a ligase. Alternatively, synthetic nucleic acid linkers can be ligated to the termini of a gene. These synthetic linkers contain nucleic acid sequences that correspond to a particular restriction site in the vector. The selection of expression vectors/promoter would depend on the type of host cells for use in producing the antibodies.
A variety of promoters can be used for expression of the antibodies described herein, including, but not limited to, cytomegalovirus (CMV) intermediate early promoter, a viral LTR such as the Rous a virus LTR, HIV-LTR, HTLV-l LTR, the simian virus 40 (SV40) early er, E. coli lac UV5 promoter, and the herpes x tk virus promoter.
Regulatable promoters can also be used. Such regulatable promoters include those using the lac repressor from E. coli as a transcription modulator to regulate transcription from lac or-bearing mammalian cell promoters [Brown, M. et al., Cell, 49:603-612 (1987)], those using the tetracycline repressor (tetR) [Gossen, M., and Bujard, H., Proc. Natl. Acad. Sci. USA 2O 89:5547-5551 (1992), Yao, F. et al., Human Gene y, 91939-1950 , Shockelt, P., et al., Proc. Natl. Acaa’. Sci. USA, 92:6522-6526 (1995)]. Other systems include FK506 dimer, VP16 or p65 using astradiol, RU486, diphenol murislerone, or rapamycin. Inducible systems are available from ogen, Clontech and Ariad.
Regulatable promoters that include a sor with the operon can be used. In one embodiment, the lac repressor from E. coli can function as a transcriptional tor to regulate ription from lac operator-bearing mammalian cell promoters (M. Brown et al., Cell, 49:603-612 (1987), Gossen and Buj ard (1992), M. Gossen et al., Nail. Acaa’. Sci. USA, 89:5547-5551 (1992)) combined the tetracycline repressor (tetR) with the transcription activator (VP 16) to create a tetR-mammalian cell ription activator fusion protein, tTa (tetR-VP 16), 3O with the tetO-bearing minimal promoter derived from the human cytomegalovirus (hCMV) major immediate-early promoter to create a tetR-tet operator system to control gene expression in mammalian cells. In one embodiment, a tetracycline inducible switch is used. The tetracycline repressor (tetR) alone, rather than the tetR-mammalian cell transcription factor fusion derivatives can function as potent modulator to regulate gene expression in -l53- mammalian cells when the ycline operator is properly oned downstream for the TATA element of the CMVIE promoter (Yao et al., Human Gene Therapy, 10(16): 399 (2003)). One particular advantage of this tetracycline ble switch is that it does not require the use of a tetracycline repressor-mammalian cells transactivator or repressor fusion protein, which in some instances can be toxic to cells (Gossen et al., Natl. Acad. Sci. USA, 89:5547-5551 (1992), Shockett et al., Proc. Natl. Acad. Sci. USA, 92:6522-6526 (1995)), to achieve its regulatable effects.
Additionally, the vector can contain, for example, some or all of the following: a selectable marker gene, such as the neomycin gene for selection of stable or transient transfectants in mammalian cells, enhancer/promoter sequences from the immediate early gene of human CMV for high levels of transcription, transcription termination and RNA processing s from SV40 for mRNA ity, SV40 polyoma origins of replication and ColEl for proper episomal replication, internal me g sites (IRESes), versatile multiple cloning sites, and T7 and SP6 RNA promoters for in vitro transcription of sense and antisense RNA.
Suitable vectors and methods for ing vectors containing transgenes are well known and available in the art.
Examples of polyadenylation signals useful to practice the methods described herein include, but are not limited to, human collagen I polyadenylation signal, human collagen II polyadenylation signal, and SV40 polyadenylation signal.
One or more vectors (e.g., expression s) comprising c acids encoding any of the antibodies may be introduced into suitable host cells for producing the antibodies. The host cells can be cultured under suitable conditions for expression of the antibody or any polypeptide chain thereof. Such antibodies or polypeptide chains thereof can be recovered by the cultured cells (e.g., from the cells or the culture supernatant) via a conventional method, e.g., affinity purification. If necessary, polypeptide chains of the antibody can be incubated under suitable conditions for a suitable period of time allowing for production of the antibody.
In some embodiments, s for preparing an antibody described herein involve a recombinant expression vector that encodes both the heavy chain and the light chain of an anti- Galectin-9 antibody, as also described herein. The recombinant expression vector can be introduced into a suitable host cell (e.g., a dhfr- CHO cell) by a conventional method, e.g., m phosphate-mediated ection. Positive transformant host cells can be selected and cultured under suitable conditions ng for the expression of the two polypeptide chains that form the antibody, which can be recovered from the cells or from the culture medium. When —154— necessary, the two chains recovered from the host cells can be incubated under suitable conditions allowing for the formation of the antibody.
In one example, two recombinant expression vectors are provided, one encoding the heavy chain of the anti-Galectin-9 antibody and the other encoding the light chain of the anti- Galectin-9 antibody. Both of the two recombinant expression vectors can be introduced into a suitable host cell (e. g., dhfr- CHO cell) by a tional method, e.g., calcium phosphate- mediated transfection. Alternatively, each of the expression vectors can be introduced into a suitable host cells. Positive transforrnants can be selected and cultured under suitable conditions allowing for the expression of the polypeptide chains of the antibody. When the two expression vectors are introduced into the same host cells, the antibody produced therein can be recovered from the host cells or from the culture . If necessary, the polypeptide chains can be recovered from the host cells or from the culture medium and then incubated under le conditions allowing for formation of the antibody. When the two expression vectors are introduced into different host cells, each of them can be recovered from the corresponding host cells or from the corresponding culture media. The two polypeptide chains can then be ted under suitable conditions for formation of the antibody. rd molecular biology ques are used to prepare the inant expression vector, transfect the host cells, select for transformants, culture the host cells and recovery of the antibodies from the culture . For example, some antibodies can be isolated by affinity chromatography with a Protein A or Protein G coupled .
Any of the nucleic acids encoding the heavy chain, the light chain, or both of an anti- Galectin-9 antibody as described herein, vectors (e.g., expression vectors) containing such, and host cells comprising the vectors are within the scope of the present disclosure.
Anti-Galectin-9 antibodies thus prepared can be can be terized using s known in the art, whereby ion, amelioration, or neutralization of Galectin-9 biological activity is detected and/or measured. For example, an ELISA-type assay may be suitable for qualitative or quantitative measurement of Galectin-9 tion of Dectin-l or TIM-3 signaling.
The bioactivity of an anti-Galectin-9 antibody can verified by incubating a candidate dy with Dectin-l and Galectin-9, and monitoring any one or more of the following 3O characteristics: (a) binding between Dectin-l and Galectin-9 and inhibition of the signaling transduction mediated by the binding, (b) preventing, ameliorating, or treating any aspect of a solid tumor, (c) blocking or decreasing Dectin-l activation, (d) inhibiting (reducing) sis, production or release of in-9. Alternatively, TIM-3 can be used to verify the bioactivity of an anti-Galectin-9 antibody using the protocol described above. Alternatively, CD206 can be used to verify the bioactivity of an anti-Galectin-9 antibody using the protocol described above.
Additional assays to ine bioactivity of an anti-Galectin-9 antibody include measurement of CD8+ and CD4+ (conventional) T-cell activation (in an in vitro or in vivo assay, e.g., by measuring inflammatory cytokine levels, e.g., IFNgamma, TNFalpha, CD44, ICOS granzymeB, Perforin, IL2 (upregulation), CD26L and IL-10 (downregulation)), measurement of reprogramming of macrophages (in vitro or in vivo), e.g., from the M2 to the M1 phenotype (e.g., increased MHCII, reduced CD206, increased TNF-alpha and iNOS).
Alternatively, levels of ADCC can be assessed, e.g., in an in vitro assay, as described .
Methods of Treatment The present sure provides pharmaceutical compositions comprising at least one anti-Galectin-9 antibody described herein or antigen binding fragment thereof and uses of such for inhibiting or ng a signaling mediated by Galectin-9 or eliminating or reducing Galectin-9 positive cells. Any of the anti-Galectin-9 antibodies described herein can be used in any of the s described herein. In some embodiments, the anti-Galentin-9 antibody is selected from G9.1-1, G9.1-2, G9.1-3, G9.1-4, G9.1-5, , G9.1-7, G9.1-8, G9.1-9, G91- , 1, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G9.1-8m8, m9, m10, G9.1-8m11, G9.1-8m12, G9.1-8m13, and G9.1-8m14, or 2O combinations thereof. In some embodiments, the anti-Galectin-9 antibody is selected from G9.2- 1, G9.2-2, , G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, , G9.2-10, G9.2-11, G9.2-12, 3,G9.2-14,G9.2-15,G9.2-16,G9.2-17,G9.2-17mut6,G9.2-18,G9.2-19,G9.2-20, G9.2-2l, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low aff1nity , or ations thereof. miting examples of such antibodies include for example antibody 92-17 or 9.1-8mutl3. Such antibodies can be used for treating diseases associated with Galectin-9. In some aspects, the invention provides methods of treating cancer. In some embodiments, the present disclosure methods for reducing, ameliorating, or eliminating one or more symptom(s) associated with cancer.
In some embodiments, the disclosure provides a method for ng cancer in a subject, the method comprising administering to a subject in need thereof an effective amount of an anti- Galectin-9 antibody described herein or antigen binding fragment thereof. In some embodiments, the disclosure provides a method for treating cancer in a subject, the method sing administering to a subject in need thereof an effective amount of a pharmaceutical composition comprising an anti-Galectin-9 antibody described herein or antigen binding fragment thereof. In some embodiments, the anti-Galectin-9 antibody is selected from the group ting of , G9.1-2, G9.1-3, G9.1-4, G9.1-5, G9.1-6, G9.1-7, G9.1-8, , G9.1-10, G9.1-11, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G9.1- 8m8, G9.l-8m9, G9.l-8mlO, G9.l-8mll, m12, G9.l-8ml3, and ml4 antibodies , or combinations thereof. In some embodiments, the anti-Galectin-9 antibody is selected from the group consisting of G9.2-l, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G92- -10,G9.2-11,G9.2-12,G9.2-13,G9.2-14,G9.2-15,G9.2-16,G9.2-17,G9.2-17mut6, G9.2-18, 9, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G92— low affinity binder antibodies, or combinations f. Non-limiting examples of such antibodies e for example antibody 9.2-17 or 9.1-8mutl3.
Given that pro-tumor action of Galectin-9 is mediated through interaction with immune cells (i.e., interactions with with lymphoid cells via TIM-3, CD44, and 4lBB, and with hages via dectin-l and CD206) and given that Galectin-9 is expressed in a large number of tumors, targeting Galectin-9, e.g., using a Galectin-9 binding antibody to inhibit interaction with its receptors, provides a therapeutic approach that can be applied across a variety of different tumor types.
In some embodiments, the cancer is selected from l cancer, adrenocortical carcinoma, anal cancer, appendix cancer, bile duct cancer, bladder cancer, bone cancer (e.g., Ewing sarcoma tumors, osteosarcoma, malignant fibrous histiocytoma), brain cancer (e.g., astrocytomas, brain stem glioma, craniopharyngioma, ependymoma), bronchial tumors, cholangiocarcinoma, cholangiosarcoma, central nervous system tumors, breast cancer, Castleman disease, cervical cancer, colon cancer, rectal cancer, colorectal cancer, endometrial cancer, esophageal cancer, eye cancer, gallbladder cancer, gastrointestinal cancer, intestinal carcinoid tumors, gastrointestinal stromal tumors, genitourinary cancers, gestational trophoblastic disease, heart cancer, Kaposi sarcoma, kidney cancer, laryngeal cancer, hypopharyngeal cancer, leukemia (e.g., acute lymphoblastic leukemia, acute myeloid leukemia, chronic cytic leukemia, c myelogenous leukemia), liver cancer, lung cancer (for example, non-small cell lung cancer, NSCLC, and small cell lung cancer, SCLC), lymphoma (e.g., AIDS-related lymphoma, Burkitt lymphoma, cutaneous T cell lymphoma, Hogkin 3O ma, Non-Hogkin lymphoma, primary l nervous system lymphoma), malignant elioma, multiple myeloma, myelodysplastic syndrome, nasal cavity cancer, sal sinus cancer, pancreatic duct adenocarcinoma (PDA) nasopharyngeal cancer, neuroblastoma, oral cavity , oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, pituitary tumors, prostate cancer, retinoblastoma, rhabdomyosarcoma, rhabdoid tumor, salivary gland cancer, sarcoma, skin cancer (e.g., basal cell carcinoma, melanoma), squamous cell head and neck cancer, small intestine cancer, stomach cancer, teratoid tumor, testicular cancer, throat cancer, thymus cancer, thyroid cancer, unusual childhood cancers, upper and lower gastrointestinal malignancies (including, but not limited to, geal, gastric, and hepatobiliary cancer), urethral , uterine cancer, uterine sarcoma, vaginal cancer, vulvar cancer, Waldenstrom macroglobulinemia, and Wilms tumor. In some embodiments, the cancer is selected from hematological ancies include acute lymphoblastic leukemia, chronic cytic leukemia, lymphomas, multiple myeloma, acute myelogenous leukemia, chronic myelogenous leukemia, myelodysplastic syndromes and the roliferative neoplasms, such as essential thrombocythemia, polycythemia vera, myelof1brosis, and gallbladder cancer (adenocarcinomas or squamous cell carcinoma). In some ments, the symptom(s) associated thereof include, but are not limited to, anemia, loss of appetite, irritation of bladder , bleeding and bruising (thrombocytopenia), changes in taste or smell, constipation, diarrhea, dry mouth, gia, edema, fatigue, hair loss (alopecia), infection, infertility, lymphedema, mouth sores, nausea, pain, peripheral neuropathy, tooth decay, urinary tract ions, and/or problems with memory and concentration. The method may comprise preparing a ceutical composition with an anti-Galectin-9 antibody described herein, and administering the pharmaceutical composition to a subject in a therapeutically effective amount.
In some embodiments, the disclosure provides a method for treating gall bladder cancer in a t, the method comprising administering to a t in need f an effective amount of an alectin-9 antibody described herein, e.g., in Table l or Table 2 herein, including but not limited to, 9.1-8ml3 and/or 92-17, or an antigen binding nt thereof.
In certain embodiments, administering the ceutical composition, e.g., one or more of the anti-Galectin-9 antibodies described herein, e.g., in Table 1 and/or Table 2,including, but not limited to, 92-17 and 9.1-8ml3, to the subject s cell proliferation, tumor growth, and/or tumor volume in a subject, or reduces the number of metastatic lesions over time. In some embodiments, administering the composition results in complete response, partial response, or stable disease.
Pancreatic ductal adenocarcinoma (PDA) is a devastating disease with few long-term survivors (Yadav et al., Gaslroenterolog, 2013, 144, 1252-1261). Inflammation is paramount in PDA progression as oncogenic mutations alone, in the absence of concomitant inflammation, are insufficient for tumorigenesis (Guerra et al., Cancer Cell, 2007, 11, 291-302). Innate and adaptive immunity cooperate to promote tumor progression in PDA. In particular, specif1c innate immune sub sets within the tumor microenvironment (TME) are apt at educating adaptive immune effector cells s a tumor-permissive phenotype. Antigen presenting cell (APC) populations, including M2-polarized tumor-associated macrophages (TAMs) and myeloid dendritic cells (DC), induce the generation of immune suppressive Th2 cells in favor of tumor- protective Th1 cells (Ochi et al., JofExp Med, 2012, 209, 1671-1687, Zhu et al., Cancer Res, 2014, 74, 5057-5069) . Similarly, it has been shown that myeloid derived suppressor cells (MDSC) negate anti-tumor CD8+ cytotoxic hocyte (CTL) responses in PDA and promote metastatic progression (Connolly et al., JLeak Biol, 2010, 87, 713-725, Pylayeva- Gupta et al., Cancer Cell, 2012, 21, 836-847, Bayne et al., Cancer Cell, 2012, 21, 822-835).
Recently, dectin-1 on hages was shown to bind galectin-9 in pancreatic ductal adenocarcinoma (PDA) (Daley et al., 2017). Removal of dectin-1 signaling (in Dectin-/- mice) resulted in a decrease in tumor infiltration of M2 type (suppressive CD206+) macrophages.
Additionally, antibody-based Galectin-9 neutralization only enhanced T cell activation in -1+/+ hosts, indicating that Galectin-9 exerts primary -suppressive effects specific to Dectin-1 signaling. Upon interruption of the Dectin-1—Galectin-9 axis, CD4+ and CD8+ T cells — which are dispensable to FDA progression in hosts with an intact signaling axis — became reprogrammed into indispensable mediators of anti-tumor immunity. Without g to be bound by , blocking GalectinDectin-1 signaling presents one exemplary mechanism (in on to TIM-3 and other signaling pathways) that could underlie a strong anti-tumor response a Galectin-9 targeting immunotherapy approach in PDA e.g., by administering an antibody that binds to Galectin-9, such as those described herein.
In some embodiments, the disclosure es a method for ng pancreatic ductal adenocarcinoma (FDA) in a t, the method comprising administering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody described herein or antigen binding fragment thereof. In some embodiments, the anti-Galentin-9 antibody is selected from the group consisting of G9.1-1, G9.1-2, G9.1-3, G9.1-4, G9.1-5, G9.1-6, , , G9.1-9, G9.1-10, G9.1-11, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G9.1- 8m8, G9.1-8m9, G9.1-8m10, G9.1-8m11, G9.1-8m12, m13, and G9.1-8m14 antibodies , and combinations thereof. In some embodiments, the anti-Galectin-9 antibody is selected from the group ting of G9.2-1, G9.2-2, G9.2-3, G9.2-4, G9.2-5, , G9.2-7, G9.2-8, G9.2- 9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-17mut6, G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2- low affinity binder antibodies and combinations thereof. Non-limiting examples of such antibodies include for example antibody 92-17 or 9.1-8mut13. In any of these methods of ent, the alectin-9 antibody is antibody 9.2-17 and/or antibody 9.1-8mut13.
In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the anti-Galectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2, and wherein the cancer is pancreatic ductal adenocarcinoma (PDA). In some embodiments, the disclosure provides the use of an anti- Galectin-9 dy as a medicament for the treatment of cancer, wherein the anti-Galectin-9 antibody is antibody 9.1-8m13 and/or 9.2-17, and wherein the cancer is pancreatic ductal arcinoma (PDA). Colorectal cancer (CRC), also known as bowel cancer, colon cancer, or rectal cancer, is any cancer affecting the colon and the rectum. CRC is known to be driven by genetic tions of tumor cells and is also influenced by host ctions. Recent reports have demonstrated a direct ation n the densities of certain T lymphocyte ulations and a favorable clinical outcome in CRC, supporting a major role of - mediated ty in repressing tumor progression of CRC.
Tim-3, as noted elsewhere herein, is an immune tory molecule, which triggers downstream cascade events upon stimulation by galectin-9 (Zhu C, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity, Nature immunology. 2005, 6: 1245— 1252). Tim-3 has been found to be a critical mediator in CRC progression (Yu et al,, Tim-3 is upregulated in human colorectal carcinoma and associated with tumor progression, Mol Med Rep. 2017 Feb, 15(2): 689—695). In this study, expression of Tim-3 was significantly associated with tumor size (P=0.007), tumor-node-metastasis staging (P<0.0001) and distant metastasis 2O (P<0.0001). Additionally, increased Tim-3 expression is associated with M2 macrophage polarization in colon cancer and promotes tumor growth. Blockade of the Tim-3 pathway inhibited both the polarization of tumor-supporting macrophages and colon cancer growth (Jiang et al., Tim-3 promotes tumor-promoting M2 macrophage polarization by binding to STATl and suppressing the STATl-miR—155 signaling axis, Oncoimmunology, 2016 Aug 3,5(9):e1211219). Given these findings and high expression of Galectin-9 observed in ctal cancers (Lahm et al., J. Cancer Res. Clin. Oncol.2001,127:375—3 86), modulating the Galectin- 9/TIM-3 axis by inhibiting the interaction between Galectin-9 and TIM-3, e.g., by administrating an antibody that binds to Galectin-9, is a pproach to treating such cancers in the clinic which may result in improved outcomes.
In some embodiments, the disclosure provides a method for treating colorectal cancer (CRC) in a subject, the method comprising stering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody described herein or antigen binding fragment f. In some embodiments, the alentin-9 antibody is selected from the group -l60- consisting of G9.1-1, G9.1-2, G9.1-3, G9.1-4, G9.1-5, G9.1-6, G9.1-7, G9.1-8, G9.1-9, G9.1-10, 1, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G91- 8m8, G9.1-8m9, m10, G9.1-8m11, G9.1-8m12, G9.1-8m13, and G9.1-8m14 antibodies, and combinations thereof. In some ments, the alectin-9 antibody is selected from the group consisting of G9.2-1, G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G92- 9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, 4, G9.2-15, G9.2-16, 7, G9.2-17mut6, G9.2-18, G9.2-l9, G9.2-20, G9.2-2l, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G92- low affinity binder antibodies, and combinations thereof. Non-limiting examples of such antibodies include for example dy 92-17 or 9.1-8mut13. In any of these methods of treatment, the anti-Galectin-9 antibody is antibody 9.2-17 and/or antibody 9.1-8mut13.
In some ments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the anti-Galectin-9 antibody is any of the dies described herein in Table 1 and/or Table 2, and wherein the cancer is colorectal cancer(CRC). In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the alectin-9 antibody is antibody 13 and/or 9.2-17, and wherein the cancer is colorectal cancer (CRC).
Melanoma is the deadliest form of skin cancer and has been sing in nce for the past 30 years, especially in young adults. Recent advances have resulted in the development of numerous immune-activating therapies that have greatly ed patient survival. 2O Accumulation of c ers, most frequently mutations in B-Raf and N—Ras, in the melanocyte are a hallmark of melanoma (Rodriguez-Cerdeira et al., Advances in Immunotherapy for Melanoma: A hensive Review, Mediators Inflamm. 2017, 2017: 3264217, and references n). However, the interaction between the microenvironment is necessary for these alterations to result in the transformation of a stic melanocyte into a melanoma cell. The microenvironment then also further promotes invasion and metastasis. New therapeutic strategies including CTLA-4, PD-l and PD-L1/2 blockers, have been developed and have dramatically improved outcomes for melanoma patients (Farkona et al., Cancer immunotherapy: the beginning of the end of ? BMC Med. 2016;14:73). However, these therapies depend on the presence of a functional immune system, which is suppressed in patients with advanced cancer, and new methods to reactivate this suppressed systemic immunity are needed to further improve outcomes for ma patients.
In patients with metastatic melanoma, high blood levels of galectin-9 are correlated with worse overall survival and a bias towards a Th2 inflammatory state supportive of tumor growth.
Additionally, galectin-9 is co-localized with the M2 macrophage population in metastatic -l6l- melanoma and e forms of galectin-9 in the blood correspond with poor survival (Enninga et al., Melanoma Res. 2016 Oct,26(5):429-4l). Association of Galectin-9 with M2 macrophages was found to be due to Galectin-9 ligation to CD206 on M2 macrophages, which resulted in pro- tumor phenotype in the local microenvironment. Accordingly, both Galectin-9/dectin-l and in-9/CD206 interactions may promote macrophage mediated immune ssive effects.
Without wishing to be bound by theory, these findings indicate that inhibiting Galectin-9/dectin- l and Galectin-9/CD206 interactions, e.g., by stering an antibody that binds to Galectin-9, may present a rationale for employing anti-Galectin-9 antibodies in a therapeutic approach in melanoma, which will lead to improved overall survival, in patients, including but not limited to those refractory to anti- CTLA-4, PD-l and PD-Ll/2 therapies.
In some embodiments, the disclosure provides a method for treating melanoma in a t, the method comprising administering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody described herein or antigen binding fragment thereof. In some embodiments, the anti-Galentin-9 antibody is selected from the group consisting of G9. l-l, G9.l-2,G9.1-3,G9.l-4,G9.1-5,G9.l-6,G9.1-7,G9.l-8,G9.l-9,G9.l-10,G9.l-11,G9.1-8m1, G9.l-8m2, G9.l-8m3, G9.l-8m4, G9.l-8m5, G9.l-8m6,G9.1-8m7, G9.l-8m8, m9, G9.l- 8mlO, G9.l-8ml l, G9.l-8m12, G9.l-8ml3, and G9.l-8ml4 dies, and combinations thereof. In some embodiments, the anti-Galectin-9 antibody is selected from the group consisting of G9.2-1, G9.2-2, G9.2-3, G9.2-4, , G9.2-6, G9.2-7, G9.2-8, , G9.2-10, 2O 1,G9.2-12,G9.2-13,G9.2-14,G9.2-15,G9.2-16,G9.2-17,G9.2-17mut6,G9.2-18, G9.2-19, 0, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low aff1nity binder antibodies, and ations thereof. Non-limiting examples of such antibodies include for example antibody 92-17 or 9. l-8mutl3. In any of these methods of treatment, the anti-Galectin-9 antibody is antibody 9.2-17 and/or antibody 9. l-8mutl3.
In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the alectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2, and wherein the cancer is melanoma. In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the anti-Galectin-9 dy is antibody 9. l- 3O 8ml3 and/or , and wherein the cancer is melanoma.
Cholangiocarcinoma (CCA) is an epithelial cancer that forms in the bile ducts and is the most common biliary malignancy and the second most common hepatic malignancy after hepatocellular oma and the overall incidence of cholangiocarcinoma has increased ssively worldwide over the past four decades. CCAs are classified into three subtypes -l62- based on their anatomic location, intrahepatic giocarcinoma (iCCA), perihilar CCA (pCCA), and distal CCA (dCCA) (see, e.g., Loeuillard et al., Animal models of cholangiocarcinoma, Biochim Biophys Acta Mol Basis Dis. 2018 Apr 5., and Rizvi et al., Cholangiocarcinoma — evolving concepts and therapeutic strategies; Nat Rev Clin Oncol. 2018 Feb, 15(2): 95—111).
In a restrospective immune profiling study in of 99 surgically resected iHCC, TIM positive staining of centrally located, tumor infiltrative lymphocytes was observed, at levels 3 times r than PD-l staining. Overall survival was cantly associated with lower numbers of TIM-3 tumor infiltrating lymphocytes (ascopubs.org/doi/abs/lO. l200/JCO.2018.36.15_suppl. 12049). Accordingly, reducing THVI-3 activity or signaling, e.g., by inhibiting the Gal-9/Tim-3 interaction in an immunotherapeutic approach, e.g., by administering an anti-Galectin-9 dy such as one or more of the anti- Galectin-9 antibodies described herein, may have a positive impact on overall survival.
In some embodiments, the disclosure provides a method for treating cholangiocarcinoma in a subject, the method comprising administering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody described herein or antigen binding fragment thereof. In some embodiments, the anti-Galentin-9 antibody is selected from the group consisting of G9. 1- 1,G9.l-2,G9.l-3,G9.1-4,G9.l-5,G9.1-6,G9.l-7,G9.l-8,G9.1-9,G9.l-10,G9.1-11,G9.1- 8ml, G9.l-8m2, G9.l-8m3, m4, G9.l-8m5, m6,G9.1-8m7, G9.l-8m8, G9.l-8m9, 2O G9.l-8m10, G9.l-8ml l, G9.l-8m12, G9.l-8ml3, and G9.l-8ml4 antibodies, and combinations thereof. In some embodiments, the anti-Galectin-9 antibody is selected from the group consisting of G9.2-1, G9.2-2, G9.2-3, G9.2-4, , G9.2-6, , G9.2-8, G9.2-9, G9.2-10, G9.2-11,G9.2-12,G9.2-13,G9.2-14,G9.2-15,G9.2-16,G9.2-17,G9.2-17mut6,G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder antibodies, and combinations thereof. Non-limiting examples of such antibodies e for example dy 92-17 or 9. l-8mutl3. In any of these s of ent, the anti-Galectin-9 antibody is antibody 9.2-17 and/or antibody 9. l-8mutl3.
In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the anti-Galectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2, and wherein the cancer is cholangiocarcinoma. In some embodiments, the disclosure provides the use of an alectin-9 dy as a medicament for the treatment of cancer, wherein the alectin-9 antibody is antibody l3 and/or 92-17, and n the cancer is cholangiocarcinoma. -l63- Renal Cell Carcinoma (RCC) has the highest mortality rate of the genitourinary cancers and the incidence of RCC has risen steadily, while the outcome s poor. Approximately 273,000 new cases of kidney cancer are diagnosed worldwide each year. About one third of patients with localized disease will suffer recurrence or metastasis. Once metastasis , malignancy metastasize, the 5-year survival for patients is less than 10 %. Clear-cell renal cell carcinoma (ccRCC) is the major histological subtype, which accounts for 80—90 % of all the RCCs. RCC is sensitive to immunotherapy and targeted therapy while highly resistant to both chemotherapy and radiation therapy.
In RCC patients, Gal-9 is expressed at much higher levels in cancerous lesions than the surrounding normal tissue, and patients with high Galectin-9 expression showed more advanced progression of the disease with larger tumor size and necrosis (Kawashima et al., BIU Int. 13:320—332). Gal-9 in tumor tissue of ccRCC patients was significantly positively associated with tumor size, Fuhrman grade, necrosis, and impaired al outcome ing poor survival and early recurrence (Fu et al., Galectin-9 predicts erative recurrence and survival of patients with clear-cell renal cell carcinoma,Tumour Biol. 2015 Aug,36(8):5791-9).
THVI-3 is also associated with poor prognosis in RCC, and own of TIM-3 suppresses the proliferation and invasion ty of ccRCC cell lines (Yuan et al., Prognostic implication of Tim-3 in clear cell renal cell carcinoma. Neoplasma. 2014,61 :35—40). Accordingly, the Gal- 9/T11VI-3 axis might play an important role in the development of renal cell carcinoma and administration of immunotherapeutic agents which inhibit Gal-9 binding to THVI-3, such as the anti-Galectin-9 antibodies described herein, e.g., in Table 1 and/or Table 2, may result in increased survival and lower reoccurrence in RCC.
In some embodiments, the sure es a method for treating renal cancer in a subject, the method comprising administering to a subject in need thereof an effective amount of an alectin-9 antibody described herein or antigen binding fragment thereof. In some embodiments, the anti-Galentin-9 dy is selected from the group consisting of G9.1-1, G9.1-2, G9.1-3, G9.1-4, G9.1-5, G9.1-6, G9.1-7, G9.1-8, G9.1-9, G9.1-10, G9.1-11, G9.1-8m1, G9.1-8m2, m3, G9.1-8m4, m5, G9.1-8m6,G9.1-8m7, G9.1-8m8, G9.1-8m9, G9.1- 8m10, G9.1-8m11, G9.1-8m12, G9.1-8m13, and G9.1-8m14 antibodies, and combinations thereof. In some embodiments, the anti-Galectin-9 antibody is selected from the group ting of , G9.2-2, , G9.2-4, G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2-9, G9.2-10, 1, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-17mut6 , G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low aff1nity binder antibodies, and combinations thereof. Non-limiting examples of such antibodies include for example dy 92-17 or antibody 9. l-8mutl3. In any of these methods of treatment, the anti-Galectin-9 antibody is antibody 9.2-17 and/or antibody 9. l-8mutl3.
In some embodiments, the disclosure es the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the anti-Galectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2, and wherein the cancer is renal cell carcinoma (RCC). In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the anti-Galectin-9 antibody is antibody 9.1-8ml3 and/or 9.2-17, and wherein the cancer is renal cell oma (RCC).
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer.
Hepatocellular oma occurs most often in people with chronic liver diseases, such as cirrhosis caused by hepatitis B or hepatitis C infection. HCC is usually accompanied by cirrhotic liver with extensive lymphocyte infiltration due to c viral ion. Many studies have trated that tumor-inflltrating effector CD8+ T cells and T helper l7 (Thl7) cells correlate with improved survival after surgical resection of tumors. r, tumor-inflltrating effector T cells fail to control tumor growth and metastasis Pang et al., The immunosuppressive tumor nvironment in hepatocellular carcinoma, Cancer Immunol Immunother 8:877- 886) The TIM-3/galectin-9 interaction contributes to immune dysfunction in human HCC (Li, 2O et al., Tim-3/galectin-9 signaling pathway es T-cell dysfunction and ts poor prognosis in patients with hepatitis B virus-associated hepatocellular carcinoma, Hepatology. 2012 (4): 1342-51). High Galectin-9 expression is found on myeloid APCs and high numbers of Tim-3+ T cells are found in HBV-associated HCC, and blocking Tim-3/galectin-9 signaling using TIM-3 antibodies recovers effector T-cell function in T cells isolated from human HCC. Thus, the targeting Tim-3/Galectin-9 axis, e.g., by administering anti-Galectin-9 antibodies, e.g., such as anti-Galectin-9 antibodies shown in Table l and Table 2 herein, including, but not limited to, antibody 9. l-8mutl3 and/or antibody 9.2-l7, constitutes a novel immune therapeutic strategy for ng ts with HBV-associated HCC.
In some embodiments, the disclosure provides a method for treating hepatocellular 3O carcinoma in a subject, the method sing administering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody described herein or antigen binding fragment -l65- thereof. In some embodiments, the anti-Galentin-9 dy is selected from the group consisting of G9.1-1, G9.1-2, G9.1-3, G9.1-4, , G9.1-6, G9.1-7, G9.1-8, G9.1-9, G9.1-10, G9.1-11, G9.1-8m1, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.1-8m5, G9.1-8m6,G9.1-8m7, G91- 8m8, G9.1-8m9, G9.1-8m10, G9.1-8m11, G9.1-8m12, G9.1-8m13, and G9.1-8m14 dies, and combinations f. In some embodiments, the alectin-9 antibody is selected from the group consisting of , G9.2-2, G9.2-3, , G9.2-5, G9.2-6, G9.2-7, G9.2-8, G9.2- 9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-17mut6, G9.2-18, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2- low ty binder antibodies, and combinations thereof. Non-limiting examples of such antibodies include for example antibody 92-17 or 9.1-8mut13. In any of these s of treatment, the anti-Galectin-9 antibody is antibody 9.2-17 and/or antibody 9.1-8mut13.Acute myeloid leukemia (AML) is the most common form of acute leukemia, with an incidence that increases with advanced age. Commonly of n etiology, AML can also occur as a result of exposure to genotoxic agents or following a previous hematologic disorder. AML is complex, with genetic, epigenetic, and phenotypic heterogeneity (Lowenberg and Rowe, Introduction to the review series on advances in acute d leukemia (AML), Blood 2016 127: 1).
In some embodiments, the disclosure provides the use of an alectin-9 antibody as a medicament for the treatment of cancer, wherein the anti-Galectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2, and wherein the cancer is hepatocellular 2O carcinoma (HCC). In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a ment for the treatment of cancer, wherein the anti-Galectin-9 antibody is antibody 9.1-8m13 and/or 9.2-17, and wherein the cancer is hepatocellular carcinoma (HCC).
Recent s t that the THVI-3/Gal-9 axis that TIM-3 and Gal-9 are connected to the establishment of AML. Malignant stem cells achieve dominant clonal selection through acquisition of multiple genetic abnormalities. These genetic abnormalities progressively accumulate in self-renewing hematopoietic stem cells (HSCs), and, as a consequence, these genetically ed preleukemic HSCs transform into leukemic stem cells (LSCs). As part of this process, preleukemic HSCs outgrow normal HSCs, and finally enew at a hematopoietic progenitor cell stage to become myeloid LSCs (Walter et al., Clonal architecture of secondary acute myeloid leukemia, N. Engl. J. Med, 366 (2012), pp. 1090-1098). ge et al., (Kikushige et al., A THVI-3/Gal-9 Autocrine Stimulatory Loop Drives Self-Renewal of Human Myeloid Leukemia Stem Cells and Leukemic Progression (Cell Stem Cell 17 , 3(2015), 341-352) observed that serum Galectin-9 levels were significantly elevated in AML patients and -l66- that the Tim3/Gal-9 axis stimulates an autocrine loop which functions to allow clonal dominancy and self-renewal of LSCs. Galmediated TIM-3 stimulation lead to the induction of LSC self renewal pathways. Of note, since significant lation of THVI-3 in HSC and HPC populations, as well as elevation of serum Gal-9, was observed in patients with preleukemic myeloid disorders, acquisition of Galectin-9 secretion likely occurs early during leukemia progression. Accordingly, targeting the Gal-9/TIM-3 aXis, e.g., through the administration of an anti-Galectin-9 antibody, such as one or more of the anti-Galectin-9 antibodies described herein, e.g., in Table 1 and/or Table 2, including dy 9.1-8mutl3 and/or antibody 9.2-17, may tute a novel approach to cancer stem cell therapy common to human myeloid malignancies, and moreover, such therapies may be useful not only to eradicate LSCs in AMLs, but also to prevent progression of preleukemic disorders into overt AML. Such preleukemic disorders include the CoxoC\‘C (RCMD) stage in\\ \ (MDS) or the CE: C\ ofmyeloproliferative neoplasms (MPN), including chronic myelogenous leukemia.
In some embodiments, the sure provides a method for treating a hematological malignancy in a subject, the method comprising administering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody bed herein or antigen binding fragment thereof. In some embodiments, the disclosure provides a method for treating acute lymphoblastic leukemia in a subject, the method comprising administering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody described herein or antigen binding fragment thereof. In some embodiments, the sure provides a method for treating acute myeloid ia in a subject, the method comprising administering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody described herein or antigen binding nt thereof.
In some embodiments, the disclosure provides a method for preventing progression of kemic ers into acute myeloid ia in a subject, the method comprising administering to a subject in need thereof an effective amount of an anti-Galectin-9 dy described herein or antigen binding fragment thereof. In some embodiments, the preleukemic disorders comprise RCMD stage in MDS or the C‘C‘C C‘\\ of MPN, including chronic enous leukemia. In some embodiments, the anti-Galentin-9 antibody is selected from the group ting of G9.l-l, G9.l-2, G9.l-3, G9.l-4, G9.l-5, , G9.l-7, G9.l-8, G9.l-9, G9.l-10, G9.l-11, G9.l-8ml, G9.l-8m2, G9.l-8m3, G9.l-8m4, G9.l-8m5, G9.l-8m6,G9.l- 8m7, G9.l-8m8, G9.l-8m9, G9.l-8m10, G9.l-8mll, G9.l-8m12, ml3, and G9.l-8ml4 -l67- antibodies, and combinations thereof. In some embodiments, the anti-Galectin-9 antibody is selected from the group consisting of , G9.2-2, G9.2-3, G9.2-4, , G9.2-6, G9.2-7, , G9.2-9, G9.2-10, G9.2-11, 2, G9.2-13, G9.2-14, G9.2-15, G9.2-16, G9.2-17, G9.2-l7mut6 , 8, G9.2-19, G9.2-20, G9.2-21, G9.2-22, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low affinity binder antibodies, and combinations f. Non-limiting es of such antibodies include for example antibody 92-17 or 9.1-8mutl3. In any of these methods of treatment, the anti-Galectin-9 antibody is antibody 9.2-17 and/or antibody 9. l- 8mutl3.
In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the ent of cancer, wherein the anti-Galectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2, and wherein the cancer is a hematological malignancy. In some embodiments, the sure es the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, n the anti-Galectin-9 antibody is antibody 9.1-8ml3 and/or 9.2-17, and wherein the cancer is hematological malignancy.
In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the anti-Galectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2, and wherein the cancer is AML. In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of , wherein the anti-Galectin-9 antibody is antibody 9.1-8ml3 and/or 2O 9.2-17, and wherein the cancer is AML.
In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, wherein the anti-Galectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2, and wherein the cancer is ALL. In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer, n the anti-Galectin-9 antibody is antibody 9.1-8ml3 and/or 9.2-17, and n the cancer is ALL. In any of the above-described methods, the ent method further comprises stering to the subject an inhibitor of a checkpoint molecule, an activator of a co-stimulatory receptor, and/or an inhibitor of an innate immune cell target. In some embodiments, the treatment method further comprises administering to the subject an 3O inhibitor of a checkpoint molecule. In some embodiments, the checkpoint molecule is selected from the group ting of PD-l, PD-Ll, PD-L2, CTLA-4, LAG3, TIM-3 and A2aR. In some ments, the treatment method further comprises administering to the subject an inhibitor of an activator of a co-stimulatory receptor, and/or an inhibitor of an innate immune cell target.
In some embodiments, the co-stimulatory receptor is selected from the group consisting of -l68- 0X40, GITR, CD137, CD40, CD27, and ICOS. In some embodiments, the treatment method further comprises administering to the subject an inhibitor of an innate immune cell target. In some embodiments, the innate immune cell target is selected from the group consisting of KIR, NKG2A, CD96, TLR, and IDO. In some embodiments, the anti-Galectin-9 antibody is selected from the group consisting of G9.1-1, G9.1-2, G9.1-3, G9.1-4, G9.1-5, G9.1-6, G9.1-7, , G9.1-9, G9.1-10, G9.1-11, G9.l-8ml, G9.1-8m2, G9.1-8m3, G9.1-8m4, G9.l-8m5, G91- 8m6,G9.1-8m7, G9.l-8m8, G9.1-8m9, G9.l-8m10, G9.l-8mll, G9.l-8m12, G9.l-8ml3, and G9. l-8ml4 antibodies, and ations thereof. In some embodiments, the anti-Galectin-9 antibody is selected from the group consisting of , G9.2-2, G9.2-3, G9.2-4, G9.2-5, G9.2- 6, G9.2-7, , G9.2-9, G9.2-10, G9.2-11, G9.2-12, G9.2-13, 4, G9.2-15, G9.2-16, G9.2-17, G9.2-l7mut6 , G9.2-18, G9.2-19, G9.2-20, G9.2-21, 2, G9.2-23, G9.2-24, G9.2-25, G9.2-26, and G9.2-low y binder antibodies and combinations thereof. Non- limiting es of such antibodies include for example antibody 92-17 or 9.1-8mutl3. In any of these s of treatment, the anti-Galectin-9 antibody is antibody 9.2-l7 and/or antibody 9.1-8mutl3. In some embodiments, the cancer is selected from pancreatic , e.g., pancreatic ductal adenocarcinoma, cholangiocarcinoma, hepatocellular carcinoma, colorectal cancer, ma, renal cell carcinoma, and acute myeloid leukemia.
In some ments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer in combination with a checkpoint tor molecule, 2O e.g., wherein the checkpoint inhibitor molecule is selected from the group consisting of PD-l, PD-Ll, PD-L2, CTLA-4, LAG3, TIM-3 and A2aR, wherein the anti-Galectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2. In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer in combination with a checkpoint molecule, wherein the checkpoint inhibitor le is selected from the group consisting of PD-l, PD-Ll, PD-L2, CTLA-4, LAG3, TIM-3 and A2aR, and wherein the anti-Galectin-9 dy is antibody 9. l-8ml3 and/or 92-17.
In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer in combination with a co-stimulatory molecule, e.g., wherein the co-stimulatory molecule is ed from the group consisting of 0X40, GITR, 3O CD137, CD40, CD27, and ICOS, and wherein the anti-Galectin-9 antibody is any of the antibodies described herein in Table 1 and/or Table 2. In some embodiments, the disclosure provides the use of an anti-Galectin-9 antibody as a medicament for the treatment of cancer in combination with a co-stimulatory, wherein the co-stimulatory molecule is selected from the group consisting of 0X40, GITR, CD137, CD40, CD27, and ICOS, n the anti-Galectin-9 -l69- antibody is antibody 9. l-8ml3 and/or 92-17. In some ments, the methods of the present disclosure may se anti-tumor ty (e.g., reduce cell proliferation, tumor growth, tumor volume, and/or tumor burden or load or reduce the number of metastatic lesions over time) by at least about 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or more as compared to levels prior to treatment or in a control subject. In some embodiments, reduction is measured by comparing cell proliferation, tumor growth, and/or tumor volume in a subject before and after administration of the pharmaceutical composition. In some embodiments, the method of treating or ameliorating a cancer in a subject allows one or more symptoms of the cancer to improve by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or more. Before, during, and after the administration of the pharmaceutical composition, cancerous cells and/or biomarkers in a subject may be measured in a biological sample, such as blood, serum, plasma, urine, peritoneal fluid, and/or a biopsy from a tissue or organ. In some embodiments, the methods may include administration of the compositions of the invention to reduce tumor , size, load or burden in a subject to an undetectable size, or to less than about 1%, 2%, 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, or 90% ofthe subject's tumor volume, size, load or burden prior to treatment. In other embodiments, the methods may e administration of the compositions of the invention to reduce the cell proliferation rate or tumor growth rate in a subject to an undetectable rate, or to less than about 1%, 2%, 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, or 90% ofthe rate prior to treatment. In other ments, the methods may include administration of the compositions of the invention to reduce the development of or the number or size of metastatic lesions in a t to an undetectable rate, or to less than about 1%, 2%, 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, or 90% of the rate prior to treatment.
In some embodiments, compositions are provided comprising one or more anti-Galectin- 9 dies, which may be used to treat, manage, ameliorate, and/or prevent cancer. In some embodiments, compositions of the sure comprise two or more anti-Galectin-9 antibodies, alone or in combination with prophylactic agents, therapeutic agents (e.g., chemotherapy or immunotherapy), and/or pharmaceutically able carriers and the use f are provided.
In some embodiments, the one or more antibodies bind to CRDl. In some embodiments, the one or more antibodies bind to CRD2. In some embodiments, the one or more dies bind to CRDl. In some embodiments, the compositions can comprise a combinattion of antibodies, some of which bind to CRDl, and some of which bind to CRD2. A non-limiting example of a ation is a combination comprising 9.2-17 and 9.l-8mutl. Antibodies can be combined in equimolar or non-equimolar amounts. -l70- Pharmaceutical Compositions The anti-Galectin-9 antibodies, as well as the encoding nucleic acids or nucleic acid sets, vectors comprising such, or host cells sing the s, as bed herein can be mixed with a pharmaceutically acceptable carrier ient) to form a pharmaceutical composition for use in treating a target disease. “Acceptable” means that the carrier must be compatible with the active ingredient of the composition (and preferably, capable of izing the active ient) and not deleterious to the subject to be treated. Pharmaceutically acceptable excipients (carriers) including buffers, which are well known in the art. See, e.g., Remington: The Science and Practice of Pharmacy 20th Ed. (2000) Lippincott Williams and Wilkins, Ed. K. E. Hoover.
The pharmaceutical compositions to be used in the present methods can comprise pharmaceutically acceptable carriers, excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions. (Remington: The Science and Practice of Pharmacy 20th Ed. (2000) Lippincott Williams and Wilkins, Ed. K. E. Hoover). Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations used, and may comprise buffers such as phosphate, e, and other c acids, idants including ascorbic acid and methionine, preservatives (such as octadecyldimethylbenzyl ammonium de, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, , butyl or benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol), low lar weight (less than about 10 residues) polypeptides, proteins, such as serum albumin, gelatin, or globulins, hydrophilic rs such as nylpyrrolidone, amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine, monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrans, chelating agents such as EDTA, sugars such as sucrose, mannitol, trehalose or sorbitol, orming counter-ions such as sodium, metal complexes (e.g.
Zn-protein complexes), and/or non-ionic surfactants such as TWEENTM, PLURONICSTM or polyethylene glycol (PEG). In some examples, the pharmaceutical composition described herein comprises liposomes containing the antibodies (or the ng nucleic acids) which can be prepared by methods known in the art, such as described in Epstein, et al., Proc. Natl. Acad.
Sci. USA 82:3688 (1985), Hwang, et al., Proc. Natl. Acad. Sci. USA 77:4030 (1980), and US.
Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in US. Pat. No. 5,013,556. Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and -l7l- PEG-derivatized phosphatidylethanolamine E). Liposomes are ed through filters of defined pore size to yield liposomes with the desired diameter.
The anti-Galectin-9 antibodies, or the encoding nucleic acid(s), may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial rization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly- (methylmethacylate) apsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are known in the art, see, e.g., Remington, The Science and Practice of Pharmacy 20th Ed. Mack Publishing (2000).
In other examples, the ceutical composition described herein can be formulated in sustained-release format. Suitable es of sustained-release preparations include semipermeable matrices of solid hobic polymers containing the antibody, which es are in the form of shaped es, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinyl alcohol)), polylactides (US. Pat. No. 919), copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, able lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid- glycolic acid copolymer and leuprolide acetate), e acetate isobutyrate, and poly-D-(-) hydroxybutyric acid.
The pharmaceutical compositions to be used for in vivo administration must be sterile.
This is readily accomplished by, for example, ion through sterile filtration membranes.
Therapeutic antibody compositions are lly placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
The ceutical compositions described herein can be in unit dosage forms such as tablets, pills, capsules, powders, granules, solutions or sions, or suppositories, for oral, parenteral or rectal administration, or administration by inhalation or insuffiation.
For preparing solid compositions such as tablets, the principal active ingredient can be mixed with a pharmaceutical carrier, e.g., conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, c acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g., water, to form a solid preformulation composition ning a homogeneous mixture of a compound of the present invention, or a non-toxic pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the -l72- composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention. The tablets or pills of the novel composition can be coated or otherwise nded to provide a dosage form affording the advantage of ged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer that serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate. Suitable e-active agents include, in particular, non-ionic agents, such as polyoxyethylenesorbitans (e.g., TweenTM 20, 40, 60, 80 or 85) and other sorbitans (e.g., SpanTM 20, 40, 60, 80 or 85). Compositions with a surface-active agent will conveniently se between 0.05 and 5% surface-active agent, and can be between 0.1 and 2.5%. It will be iated that other ingredients may be added, for example mannitol or other pharmaceutically able es, if necessary.
Suitable emulsions may be prepared using commercially available fat emulsions, such as IntralipidTM, LiposynTM, InfonutrolTM, LipofundinTM and LipiphysanTM. The active ingredient may be either dissolved in a pre-mixed on ition or alternatively it may be dissolved in an oil (e.g., soybean oil, safflower oil, cottonseed oil, sesame oil, corn oil or almond oil) and an emulsion formed upon mixing with a phospholipid (e. g. egg olipids, soybean phospholipids or soybean lecithin) and water. It will be appreciated that other ingredients may be added, for example glycerol or glucose, to adjust the tonicity of the emulsion. Suitable emulsions will typically contain up to 20% oil, for example, between 5 and 20%. The fat emulsion can comprise fat droplets between 0.1 and 10 .im, particularly 0.1 and 0.5 .im, and have a pH in the range of 5.5 to 8.0.
The emulsion compositions can be those prepared by mixing an antibody with IntralipidTM or the components thereof (soybean oil, egg phospholipids, glycerol and water). ceutical compositions for tion or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or c ts, or es thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as set out above. In some ments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. -l73- Compositions in preferably sterile pharmaceutically acceptable ts may be nebulized by use of gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face mask, tent or ittent positive pressure breathing machine. Solution, suspension or powder compositions may be administered, preferably orally or y, from devices which deliver the formulation in an appropriate manner.
Therapeutic Applications The present disclosure provides methods of eliminating pathologic cells expressing Galectin-9, the method comprising administering to a subject having pathologic cells expressing Galectin-9 an effective amount of a pharmaceutical composition comprising an anti-Galectin-9 antibody described herein. The present disclosure also provides methods of inhibiting Galectin- 9-mediated cell signaling in a subject, the method comprising stering to a subject in need thereof an effective amount of a ceutical composition comprising an anti-Galectin-9 antibody described herein.
To practice the methods disclosed herein, an effective amount of the pharmaceutical composition described herein can be administered to a subject (e.g., a human) in need of the treatment via a suitable route, systemically or y. In some embodiments, the anti-Galectin-9 antibodies are administered by intravenous administration, e.g., as a bolus or by continuous infusion over a period of time, by uscular, intraperitoneal, erebrospinal, subcutaneous, intra-arterial, intra-articular, intrasynovial, intrathecal, intratumoral, oral, inhalation or topical routes. Commercially available nebulizers for liquid formulations, including jet zers and ultrasonic nebulizers are useful for administration. Liquid formulations can be ly nebulized and lyophilized powder can be nebulized after reconstitution. atively, the antibodies as described herein can be aerosolized using a fluorocarbon formulation and a metered dose inhaler, or inhaled as a lyophilized and milled The subject to be treated by the methods described herein can be a mammal, more preferably a human. Mammals include, but are not limited to, farm s, sport animals, pets, 3O primates, , dogs, cats, mice and rats. A human subject who needs the treatment may be a human patient having, at risk for, or suspected of having a target disease/disorder, such as a solid tumor, hematological malignancy, mune disease (such as an allergic disorder), microbial e, and f1brotic condition. —174— Examples of solid tumor cancers include pancreatic duct adenocarcinoma (PDA), colorectal cancer (CRC), melanoma, cholangiocarcinoma, breast cancer, lung cancer (for example, non-small cell lung cancer, NSCLC, and small cell lung cancer, SCLC), upper and lower gastrointestinal malignancies (including, but not limited to, esophageal, gastric, and hepatobiliary cancer), squamous cell head and neck cancer, genitourinary cancers, ovarian cancer, and sarcomas. Hematological malignancies include acute lymphoblastic leukemia, chronic lymphocytic leukemia, lymphomas, multiple myeloma, acute myelogenous leukemia, chronic myelogenous leukemia, ysplastic syndromes and the myeloproliferative neoplasms, such as essential ocythemia, polycythemia vera and myelofibrosis. A subject haVing a solid tumor or a logical malignancy can be identified by routine medical examination, e.g., laboratory tests, organ functional tests, CT scans, or ultrasounds. In some embodiments, the subject to be d by the method described herein may be a human cancer patient who has undergone or is subjecting to an anti-cancer therapy, for example, chemotherapy, radiotherapy, immunotherapy, or y.
Examples of autoimmune diseases include rheumatoid ions, metabolic and endocrine conditions, as well as respiratory and allergic ions. A subject haVing an autoimmune disease can be identified by routine medical examination, e.g., with laboratory tests, such as antinuclear antibodies, anti-mitochondrial autoantibodies, anti-neutrophil cytoplasmic antibody, anti-phospholipid antibodies, anti-citrullinated peptide (anti-CCP), anti- toid factor, immunoglobulin A, C-reactive n test, complement test, erythrocyte sedimentation rate (ESR) test, blood ng prof1e, and protein electrophoresis/immunof1xation electrophoresis, among others. In some embodiments, the subject to be d by the method described herein may be a human t with an autoimmune disease who has one or is subjecting to an autoimmune disease treatment, for example, immunosuppressive mediation, hormone replacement therapy, blood transfusions, anti-inflammatory medication, and/or pain medication.
Microbial diseases can be caused by a variety of pathogens, including bacteria, fungi, protozoa and Viruses. Exemplary infectious bacteria include Streptococcus pyogenes, Streptococcus pneumoniae, Neisseria gonorrheae, ria meningitidis, Corynebacterium diphtheriae, idium botulinum, Clostridium perfringens, Clostria’ium , Hemophilus influenzae, ella pneumoniae, Klebsiella ozaenas, Klebsiella rhinoscleromotis, Staphylococcus , Vibrio colerae, Escherichia coli, Pseudomonas aeruginosa, Campylobacter (Vibrio) fetus, Aeromonas hydrophila, Bacillus aereus, dsiella tarda, Yersinia enterocolitica, Yersinia pestis, Yersinia pseudotuberculosis, Shigella dysenteriae, -l75- Snigellaflexneri, Shigella sonnei, Salmonella urium, Treponemapallidum, Treponema ue, Treponema caraleneum, Borrelia vinceniii, Borrelia rferi, Leplospira iclerohemorrhagiae, Mycobaclerium tuberculosis, Pneumocysiis carinii, Francisella lularensis, Brucella aborlus, Brucella suis, la meliiensis, Mycoplasma spp., Rickellsiaprowazeki, Rickellsia lsulsugumusni, and dia spp. Examples of pathologic fungi include ioides immiiis, Aspergillusfumigaius, Candida albicans, Blasiomyces dermaiilidis, Cryptococcus neoformans, and Hisioplasma capsulalum. Pathologic oa e Enlomoeba histolylica, Toxoplasma gondii, monas lenas, Trichomonas hominis, Trichomonas lis, Tryoanosoma gambiense, Trypanosoma rhodesiense, Trypanosoma cruzi, Leishmania donovani, ania lropica, Leishmania braziliensis, Pneumocyslis pneumonia, Plasmodium vivax, Plasmodiumfalciparum, and Plasmodium malaria. Examples of helminiths include Enierobius vermicularis, Trichuris Zrichiura, Ascaris lumbricoides, Tricninella spiralis, Sironggdoides siercoralis, Schisiosomajaponicum, Schisiosoma i, osoma haematobium, and hookworms. Viral infectious diseases include those caused by Adenovirus, Lassa fever virus (Arenavirus), Astrovirus, Hantavirus, Rift Valley Fever virus (Phlebovirus), Calicivirus, Ebola virus, Marburg Virus, Japanese encephalitis virus, Dengue virus, Yellow fever virus, Hepatitis C virus, Hepatitis G virus, Hepatitis B virus, tis D virus, Herpes simplex virus 1, Herpes simplex virus 2, Cytomegalovirus, Epstein Barr virus, Varicella Zoster Virus, Human Herpesvirus 7, Human Herpesvirus 8, Influenza virus, Parainfiuenza virus, Rubella virus, Mumps virus, Morbillivirus, Measles virus, Respiratory Syncytial virus, omaviruses, JC virus (Polyomavirus), BK virus (Polyomavirus), Parvovirus, Coxsackie virus (A and B), Hepatitis A virus, Polioviruses, Rhinoviruses, Reovirus, Rabies Virus (Lyssavirus), Human Immunodeficiency virus 1 and 2, and Human T-cell Leukemia virus. A subject having a microbial disease can be identified by routine medical examination, e.g., laboratory tests. For example, microscopy (e.g., Gram-positive and/or Gram- negative staining), sample culturing, biochemical tests (e.g., tests for metabolic and/or enzymatic products, such as fermentation products, acids, alcohol, or gases), and molecular diagnostics (e.g., PCR) may be used. In some ments, the subject to be treated by the method described herein may be a human infectious disease patient who has undergone or is 3O subjecting to an antimicrobial therapy, for example, immunotherapy.
Examples of fibrotic conditions include pulmonary fibrosis (e.g., cystic fibrosis, idiopathic pulmonary fibrosis), sis, y atresia, atrial fibrosis, endomyocardial fibrosis, glial scar, fibrosis, Crohn’s disease, ren’s contracture, keloid, mediastinal fibrosis, myelofibrosis, nephrogenic systemic fibrosis, progressive massive fibrosis, retroperitoneal -l76- fibrosis, and scleroderma/systemic sclerosis. A subject having a f1brotic condition can be identified by routine medical ation, e.g., laboratory tests, CT scans, X-rays, echocardiograms, or ultrasounds. In some embodiments, the subject to be treated by the method described herein may be a human fibrotic patient who has undergone or is subjecting to an anti- f1brotic therapy, for example medication, physical therapy, oxygen therapy, or surgery.
A subject suspected of having any of such target disease/disorder might show one or more symptoms of the disease/disorder. A subject at risk for the disease/disorder can be a t having one or more of the risk factors for that disease/disorder.
As used herein, “an effective amount” refers to the amount of each active agent required to confer therapeutic effect on the t, either alone or in combination with one or more other active agents. In some embodiments, the therapeutic effect is reduced in-9 activity and/or amount/expression, reduced Dectin-l signaling, reduced TIM-3 signaling, reduced CD206 signaling, or increased anti-tumor immune responses in the tumor microenvironment. Nonlimiting examples of increased anti-tumor responses e increased activation levels of effector T cells, or switching of the TAMs from the M2 to the M1 phenotype, and increased ADCC responses. Determination of whether an amount of the antibody ed the eutic effect would be evident to one of skill in the art. Effective amounts vary, as recognized by those d in the art, depending on the particular condition being d, the severity of the condition, the individual patient parameters including age, physical condition, size, gender and 2O weight, the duration of the treatment, the nature of rent therapy (if any), the specific route of stration and like s within the knowledge and expertise of the health practitioner.
These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose of the individual components or combinations f be used, that is, the highest safe dose according to sound medical judgment.
Empirical considerations, such as the half-life, generally will contribute to the ination of the dosage. For example, antibodies that are compatible with the human immune system, such as humanized antibodies or fully human antibodies, may be used to prolong half-life of the antibody and to prevent the antibody being attacked by the host's 3O immune system. Frequency of administration may be determined and ed over the course of therapy, and is generally, but not necessarily, based on treatment and/or suppression and/or amelioration and/or delay of a target disease/disorder. Alternatively, sustained continuous e ations of an antibody may be appropriate. Various formulations and devices for achieving sustained release are known in the art. -l77- In one example, dosages for an antibody as described herein may be determined empirically in individuals who have been given one or more administration(s) of the antibody.
Individuals are given incremental dosages of the antagonist. To assess efficacy of the antagonist, an tor of the disease/disorder can be followed.
Generally, for administration of any of the antibodies bed , such as those described in Table l or Table 2 herein, such as for example, antibody 9.2-17 and antibody 9.1- 8mutl, an initial candidate dosage can be about 2 mg/kg. For the purpose of the present disclosure, a typical daily dosage might range from about any of 0.1 ug/kg to 3 ug/kg to 30 ug/kg to 300 ug/kg to 3 mg/kg, to 30 mg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of symptoms occurs or until suff1cient eutic levels are achieved to alleviate a target disease or disorder, or a symptom thereof. An exemplary dosing regimen comprises administering an initial dose of about 2 mg/kg, followed by a weekly maintenance dose of about 1 mg/kg of the antibody, or followed by a maintenance dose of about 1 mg/kg every other week. However, other dosage regimens may be useful, depending on the pattern of pharmacokinetic decay that the practitioner wishes to achieve. For example, dosing from one-four times a week is contemplated. In some embodiments, dosing ranging from about 3 ug/mg to about 2 mg/kg (such as about 3 ug/mg, about 10 ug/mg, about 30 ug/mg, about 100 ug/mg, about 300 ug/mg, about 1 mg/kg, and about 2 mg/kg) may be used. In some embodiments, dosing frequency is once every week, every 2 weeks, every 4 weeks, every 5 weeks, every 6 weeks, every 7 weeks, every 8 weeks, every 9 weeks, or every 10 weeks, or once every month, every 2 months, or every 3 months, or longer.
The progress of this therapy is easily monitored by conventional techniques and assays. The dosing regimen (including the antibody used) can vary over time.
In some ments, for an adult patient of normal weight, doses ranging from about 0.3 to 5.00 mg/kg may be stered. In some examples, the dosage of the alectin-9 antibody described herein can be 10 mg/kg. The particular dosage regimen, i.e., dose, timing and repetition, will depend on the particular individual and that dual's medical history, as well as the properties of the individual agents (such as the half-life of the agent, and other considerations well known in the art).
For the purpose of the present disclosure, the appropriate dosage of an dy as described herein will depend on the specific antibody, antibodies, and/or non-antibody e (or compositions thereof) ed, the type and severity of the e/disorder, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's -l78- clinical history and response to the antagonist, and the discretion of the attending physician.
Typically, the clinician will administer an antibody, until a dosage is reached that es the desired result. In some ments, the desired result is an increase in anti-tumor immune response in the tumor microenvironment. Methods of determining whether a dosage resulted in the desired result would be evident to one of skill in the art. Administration of one or more antibodies can be continuous or intermittent, depending, for example, upon the recipient's physiological condition, r the purpose of the administration is therapeutic or lactic, and other factors known to skilled practitioners. The administration of an antibody may be essentially continuous over a preselected period of time or may be in a series of spaced dose, e. g., either before, during, or after developing a target e or disorder.
As used herein, the term “treating” refers to the application or administration of a composition including one or more active agents to a subject, who has a target disease or disorder, a symptom of the disease/disorder, or a predisposition toward the disease/disorder, with the purpose to cure, heal, alleviate, relieve, alter, remedy, rate, improve, or affect the disorder, the m of the disease, or the predisposition toward the disease or disorder.
Alleviating a target disease/disorder includes delaying the development or progression of the disease, or reducing e severity or prolonging survival. ating the disease or prolonging survival does not necessarily require curative results. As used therein, "delaying" the development of a target disease or disorder means to defer, hinder, slow, , stabilize, and/or postpone progression of the disease. This delay can be of varying lengths of time, ing on the history of the disease and/or individuals being d. A method that “delays” or alleviates the development of a disease, or delays the onset of the disease, is a method that reduces probability of developing one or more symptoms of the disease in a given time frame and/or reduces extent of the ms in a given time frame, when compared to not using the method.
Such comparisons are typically based on al studies, using a number of subjects sufficient to give a statistically significant result.
“Development” or “progression” of a disease means initial manifestations and/or ensuing progression of the disease. Development of the disease can be detectable and assessed using standard al techniques as well known in the art. However, development also refers to progression that may be undetectable. For purpose of this disclosure, development or progression refers to the biological course of the symptoms. “Development” es occurrence, recurrence, and onset. As used herein “onset” or “occurrence” of a target disease or disorder includes l onset and/or recurrence. -l79- In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are stered to a subject in need of the treatment at an amount sufficient to inhibit the ty of Galectin-9 (and/or Dectin-l or THVI-3 or CD206) in immune suppressive immune cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo. In other ments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered in an amount ive in reducing the activity level of Galectin-9 (and/or Dectin-l or THVI-3 or CD206) in immune suppressive immune cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) (as compared to levels prior to ent or in a control subject). In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, ing, but not limited to, 92-17 and 9. l3, are administered to a subject in need of the treatment at an amount sufficient to promote Ml-like programming in TAMs by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control subject).
In some embodiments, the antibodies described , e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are stered to a subject in need thereof at an amount sufficient to promote ADCC in target cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to promote ADCC in target cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or r) in vivo (as compared to levels prior to treatment or in a control t).
In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to promote CDC in target cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to promote CDC in target cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control subject).
In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not d to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the ent at an amount sufficient to promote ADCP in target cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to promote ADCP in target cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control subject).
In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to promote T cell activation in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to e T cell activation in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as ed to levels prior to treatment or in a l subject).
In some embodiments, the antibodies described , e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to promote CD4+ T cell activation in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, ing, but not limited to, 92-17 and 9. l3, are administered to a t in need of the treatment at an amount sufficient to promote CD4+ T cell activation in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control subject).
In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, ing, but not limited to, 92-17 and 9. l-8mutl3, are stered to a subject in need of the treatment at an amount sufficient to induce CD44 expression in CD4+ cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not d to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount ent to induce CD44 expression in CD4+ cells in a tumor by at least 20% (e.g., %, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control subject).
In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to induce TNFalpha expression in CD4+ cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to induce TNFalpha expression in CD4+ cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to ent or in a control subject).
In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not d to, 92-17 and 9. l-8mutl3, are stered to a subject in need of the treatment at an amount sufficient to promote CD8+ T cell activation in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to promote CD8+ T cell activation in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control subject).
In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to induce CD44 expression in CD8+ cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9. l-8mutl3, are stered to a subject in need of the treatment at an amount sufficient to induce CD44 expression in CD8+ cells in a tumor by at least 20% (e.g., %, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control t).
In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not d to, 92-17 and 9. l-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to induce TNFalpha expression in CD8+ cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, ing, but not d to, 92-17 and 9. l-8mutl3, are administered to a t in need of the treatment at an amount sufficient to induce TNFalpha expression in CD8+ cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or r) in vivo (as compared to levels prior to treatment or in a control subject).
In any of these embodiments, the antibodies described , e.g., in Table 1 and/or Table 2, including, but not d to, 92-17 and 9. l-8mutl3, are administered to a subject having cancer, wherein the cancer is selected from pancreatic cancer, e.g., pancreatic ductal adenocarcinoma, cholangiocarcinoma, hepatocellular oma, colorectal cancer, melanoma, renal cell carcinoma, and acute myeloid leukemia.
An analysis was conducted by the inventors using TCGA (The Cancer Genome Atlas) RNA Seq data from 29 different types of solid tumors utilizing approximately 40,000 individual samples, to analyze the correlation between T cell infiltration in a particular tumor type and levels of PDl, PD-Ll, IFNgamma and TNF alpha expression. Tumor types were ranked by median expression for a given gene. Gene expression for 4 TCR components (CD3 d, CD3 e, CD3g,CD3z) and the T cell specific effector protein kinase ZAP7O were used to establish the ve level of T cell ation with a given tumor type. TBX2l, a Th1 cell-specific transcription factor that controls the sion of the hallmark Th1 nes interferon-gamma (IFNg) and tumor necrosis factor (TNF) was also evaluated.
According to the analysis, T cell association levels (surrogate for T cell infiltration) with individual tumor types is generally proportional to the ranking of IFNg expression with the exception of pancreatic cancer (PDA). In PDA the level of IFNg transcription is significantly suppressed suggesting over that observed in other solid tumors implying that the immunosuppressive environment of FDA is particularly robust. In this dataset, the sion of TNF does not generally correlate with the degree of T cell infiltration. Of note, under most conditions TNF is produced by ted macrophages with less contribution by Thl T cells, NK cells, phils, mast cells, and eosinophils.
Without wishing to be bound by theory, increasing levels of IFNgamma may be particularly useful to combat the immunosuppressive environment and re-activate myeloid and lymphoid response particularly in PDA. Accordingly, in one embodiment, methods of increasing levels of ma in a cancer are provided herein, wherein the method comprises administering an anti-Galectin-9 antibody, e.g., as described herein in Table 1 and/or Table 2, 2O including, but not limited to, antibody 9.1-8ml3 and/or antibody 9.2-17, and wherein the levels of IFNgamma in the cancer are low prior prior the administration, e.g., as assessed ve to levels of expression of T cell markers. In some embodiments the cancer is PDA.
Accordingly, in some ments, methods of increasing levels of IFNgamma in a cancer are provided herein, wherein the method ses administering an anti-Galectin-9 antibody, e.g., as described herein in Table 1 and/or Table 2, ing, but not limited to, antibody 9.1-8ml3 and/or antibody 9.2-17, and wherein the cancer is PDA.In some embodiments, methods are provided herein, wherein the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9.1-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to induce IFNgamma expression in effector T cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, ing, but not limited to, 92-17 and 9.1-8mutl3, are administered to a subject in need of the ent at an amount sufficient to induce IFNgamma expression in effector T cells in a tumor. In In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not d to, 92-17 and 9.1-8mutl3, are administered to a t in need of the treatment at an amount sufficient to induce IFNgamma sion in CD4+ cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control subject).
In some embodiments, methods are provided herein, wherein the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and utl3, are administered to a subject in need of the treatment at an amount sufficient to induce IFNgamma expression in CD4+ cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not d to, 92-17 and 9.1-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to induce IFNgamma expression in CD4+ cells in a tumor. In some embodiments, the dies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9.1-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to induce IFNgamma expression in CD4+ cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control subject).
In some embodiments, methods are provided herein, wherein the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and utl3, are administered to a subject in need of the treatment at an amount sufficient to induce IFNgamma expression in CD8+ cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9.1-8mutl3, are administered to a subject in need of the treatment at an amount sufficient to induce IFNgamma expression in CD8+ cells in a tumor. In some embodiments, the dies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9.1-8mutl3, are administered to a subject in need of the ent at an amount sufficient to induce IFNgamma expression in CD8+ cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vivo (as compared to levels prior to treatment or in a control subject).
In some embodiments, methods are ed herein, n immune cell populations in tumor samples are analyzed in vitro or ex vivo. Accordingly s are provided herein, wherein the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and utl3, are provided in vitro or ex vivo at an amount sufficient to induce IFNgamma expression in effector T cells in a tumor. In some embodiments, . the antibodies described herein, e.g., in Table 1 and/or Table 2, including, but not limited to, 92-17 and 9.1-8mut13, are provided in vitro or ex vivo at an amount sufficient to induce IFNgamma expression in effector T cells in a tumor. In some embodiments, the antibodies described herein, e.g., in Table 1 and/or Table 2, ing, but not limited to, 92-17 and 9.1-8mutl3, induce IFNgamma expression in CD4+ cells in a tumor by at least 20% (e.g., 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) in vitro or ex vivo. In some embodiments, administration of one or more of the antibodies described herein results in a reduction in tumor size, reduction in tumor growth, elimination of the tumor, reduction in number of metastatic lesions over time, complete response, partial response, or stable disease. Conventional methods, known to those of ordinary skill in the art of medicine, can be used to administer the pharmaceutical composition to the subject, depending upon the type of disease to be treated or the site of the disease. This ition can also be administered via other conventional routes, e.g., administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term “parenteral” as used herein includes subcutaneous, intracutaneous, enous, intramuscular, rticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional, intratumoral, and intracranial injection or infusion techniques. In addition, it can be administered to the subject via injectable depot routes of stration such as using 1-, 3-, or 6-month depot injectable or biodegradable materials and methods. In some examples, the pharmaceutical composition is administered intraocularly or intravitreally.
Injectable compositions may contain various carriers such as vegetable oils, dimethylactamide, dimethyformamide, ethyl lactate, ethyl carbonate, isopropyl myristate, ethanol, and polyols (glycerol, propylene glycol, liquid polyethylene glycol, and the like). For intravenous injection, water soluble antibodies can be stered by the drip method, whereby a pharmaceutical formulation containing the dy and a physiologically acceptable excipient is infused. Physiologically able excipients may include, for example, 5% dextrose, 0.9% saline, Ringer’s solution or other suitable excipients. Intramuscular preparations, e.g., a sterile ation of a suitable e salt form of the dy, can be dissolved and administered in a pharmaceutical excipient such as Water-for-Injection, 0.9% saline, or 5% glucose solution.
In one embodiment, an antibody is administered via site-speciflc or ed local delivery techniques. Examples of site-speciflc or targeted local delivery techniques include various implantable depot sources of the dy or local delivery catheters, such as infusion catheters, an indwelling catheter, or a needle catheter, synthetic grafts, itial wraps, shunts and stents or other implantable devices, site specific rs, direct injection, or direct application. See, e.g., PCT Publication No. WO 00/53211 and US. Pat. No. 5,981,568.
Targeted delivery of therapeutic compositions containing an antisense polynucleotide, expression , or subgenomic polynucleotides can also be used. or-mediated DNA delivery techniques are described in, for example, Findeis et al., Trends Biotechnol. (1993) 11:202, Chiou et al., Gene eutics: Methods And Applications Of Direct Gene Transfer (J.
A. Wolff, ed.) (1994), Wu et al., J. Biol. Chem. (1988) 263:621, Wu et al., J. Biol Chem. (1994) 269:542, Zenke et al., Proc. Natl. Acaa’. Sci. USA (1990) 87:3655, Wu et al., J. Biol. Chem. (1991) 266:338.
Therapeutic compositions containing a polynucleotide (e.g., those encoding the antibodies described herein) are stered in a range of about 100 ng to about 200 mg of DNA for local administration in a gene therapy protocol. In some embodiments, concentration ranges of about 500 ng to about 50 mg, about 1 ug to about 2 mg, about 5 ug to about 500 ug, and about 20 ug to about 100 ug ofDNA or more can also be used during a gene therapy protocol.
The therapeutic polynucleotides and polypeptides bed herein can be delivered using gene delivery vehicles. The gene delivery vehicle can be of viral or non-viral origin (see generally, Jolly, Cancer Gene Therapy (1994) 1:51, Kimura, Human Gene Therapy (1994) :845, Connelly, Human Gene Therapy (1995) 1:185, and Kaplitt, Nature cs (1994) 6: 148). Expression of such coding ces can be induced using endogenous mammalian or heterologous promoters and/or enhancers. Expression of the coding sequence can be either constitutive or regulated.
Viral-based vectors for delivery of a desired polynucleotide and expression in a desired cell are well known in the art. Exemplary viral-based es include, but are not limited to, recombinant retroviruses (see, e.g., PCT Publication Nos. WO 90/07936, WO 94/03622, WO 93/25698, WO 93/25234, WO 93/11230, WO 18, WO 91/02805, U.S. Pat. Nos. ,219,740 and 4,777,127, GB Patent No. 2,200,651, and EP Patent No. 0 345 242), alphavirus- based vectors (e.g., s virus vectors, Semliki forest virus (ATCC VR-67, ATCC VR-1247), Ross River virus (ATCC VR-373, ATCC VR—1246) and Venezuelan equine encephalitis virus (ATCC VR—923, ATCC VR-1250, ATCC VR 1249, ATCC VR-532)), and adeno-associated virus (AAV) vectors (see, e.g., PCT Publication Nos. W0 94/12649, WO 93/03769, WO 93/19191, WO 94/28938, WO 95/11984 and WO 95/00655). Administration of DNA linked to killed adenovirus as bed in Curiel, Hum. Gene Ther. (1992) 3:147 can also be employed.
Non-viral delivery vehicles and methods can also be employed, including, but not limited to, polycationic sed DNA linked or unlinked to killed adenovirus alone (see, e.g., Curiel, Hum. Gene Ther. (1992) 3:147), ligand-linked DNA (see, e.g., Wu, J Biol. Chem. (1989) 264: 16985), eukaryotic cell ry vehicles cells (see, e.g., U.S. Pat. No. 5,814,482, PCT Publication Nos. WO 95/07994, WO 96/17072, WO 95/30763, and WO 38) and nucleic charge neutralization or fusion with cell membranes. Naked DNA can also be ed. Exemplary naked DNA introduction methods are described in PCT Publication No.
WO 90/11092 and US. Pat. No. 5,580,859. Liposomes that can act as gene delivery vehicles are described in US. Pat. No. 5,422,120, PCT Publication Nos. WO 95/13796, WO 94/23697, W0 91/14445, and EP Patent No. 0524968. Additional approaches are described in , Mol.
Cell. Biol. (1994) 14:2411, and in Woffendin, Proc. Natl. Acad. Sci. (1994) 91 : 1581.
The particular dosage regimen, 1'. e., dose, timing and repetition, used in the method described herein will depend on the particular subject and that subject's medical y.
In some embodiments, more than one antibody, or a combination of an antibody and another suitable therapeutic agent, may be administered to a t in need of the ent.
The dy can also be used in ction with other agents that serve to enhance and/or complement the effectiveness of the agents. ent y for a target disease/disorder can be assessed by methods well-known in the art.
In some embodiments, the sure provides a method for suppressing Dectin-1 signaling e.g., in immune suppressive immune cells, e.g., tumor infiltrating immune cells, such as macrophages, the method comprising providing or administering an anti-Galectin-9 dy described , e.g., in Table 1 and/or Table 2, or antigen binding fragment thereof to a subject. In some embodiments, the alectin-9 antibody is a 9.1-8mut13 antibody and/or a 92-17 antibody. In some embodiments, the method suppresses Dectin-1 signaling, e.g., in immune suppressive immune cells, by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or r, including any increment therein).
In some embodiments, the disclosure provides a method for suppressing TIM-3 signaling e.g., in tumor infiltrating immune cells, the method comprising providing or administering an alectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen binding fragment thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9.1- 8mut13 antibody and/or a 92-17 antibody. In some embodiments, the method suppresses the THVI-3 signaling, e.g., in tumor infiltrating immune cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for suppressing CD206 signaling, e.g., in tumor infiltrating immune cells, e.g., in macrophages, the method comprising providing or administering an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen binding fragment thereof to a subject. In some embodiments, the anti- Galectin-9 dy is a 9.1-8mut13 antibody and/or a 92-17 antibody. In some embodiments, the method suppresses the CD206 signaling, e.g., in tumor infiltrating immune cells, e.g., macrophages, by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein). -l87- In some embodiments, the disclosure provides a method for inducing cell cytotoxicity, such as ADCC, in target cells expressing Galectin-9, e.g., wherein the target cells are cancer cells or immune suppressive immune cells, the method comprising providing or administering an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen g fragment thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9. l- 8mutl3 dy and/or a 92-17 dy. In some embodiments, the method induces apoptosis in immune cells such as T cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for inducing cell cytotoxicity such as complement-dependent cytotoxicity (CDC) against target cells expressing Galectin-9 in a subject, the method comprising providing or administering an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or n binding fragment thereof to a subject. In some embodiments, the anti-Galectin-9 dy is a 9.l-8mutl3 antibody and/or a 92-17 antibody. In some embodiments, the method induces cell cytotoxicity such as ment-dependent cytotoxicity (CDC) t target cells expressing Galectin-9 at least % (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, ing any increment therein).
In some embodiments, the disclosure provides a method for inducing cell cytotoxicity, such as ADCC, e.g., against target cells sing Galectin-9 in a subject, the method comprising ing or administering an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen binding fragment thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9. l-8mutl3 antibody and/or a 92-17 antibody. In some embodiments, the method induces cell xicity by at least 30% (e.g., 3 1%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for inducing phagocytosis of target cells expressing Galectin-9 (ADCP), the method comprising providing or administering an anti-Galectin-9 dy described herein, e.g., in Table 1 and/or Table 2, or antigen binding nt thereof to a t. In some embodiments, the anti-Galectin-9 antibody is a 9. l- 8mutl3 antibody and/or a 92-17 antibody. In some embodiments, the anti-Galectin-9 antibody increases phagocytosis of target cells by at least 30% (e.g., 3 1%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for inducing cell cytotoxicity such as complement-dependent cytotoxicity (CDC) against target cells expressing Galectin-9, the method comprising providing or administering an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen binding fragment thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9.l-8mutl3 antibody and/or a 92-17 dy.
In some embodiments, the method induces cell cytotoxicity against target cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, ing any increment therein).
In some embodiments, the disclosure provides a method for inducing T cell activation, e.g., in tumor infiltrating T cells, i.e., suppress Galectin-9 ed inhibition of T cell activation, either ly or indirectly., the method comprising providing or administering an alectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or n binding nt thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9. l- 8mutl3 antibody and/or a 92-17 antibody. In some embodiments, the method promotes T cell activation by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for promoting CD4+ cell activation, the method comprising providing or administering an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen binding fragment thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9. l-8mutl3 antibody and/or a 92-17 antibody. In some embodiments, the method promotes CD4+ cell activation by at least % (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for ng CD44 expression in CD4+ cells, the method comprising ing or stering an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen binding fragment thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9. l-8mutl3 antibody and/or a 92-17 antibody. In some ments, the method ses CD44 expression in CD4+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for inducing IFNgamma expression in CD4+ cells, the method comprising providing or administering an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen binding nt thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9.l-8mutl3 antibody and/or a 92-17 antibody. In some embodiments, the method increases IFNgamma sion in CD4+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for ng TNFalpha expression in CD4+ cells, the method comprising providing or administering an anti-Galectin-9 dy described herein, e.g., in Table 1 and/or Table 2, or n binding fragment thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9. l-8mutl3 antibody and/or a 92-17 antibody. In some embodiments, the method ses TNFalpha expression in CD4+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for inducing CD44 sion in CD8+ cells, the method comprising providing or administering an anti-Galectin-9 antibody bed , e.g., in Table 1 and/or Table 2, or antigen binding nt f to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9. l-8mutl3 dy and/or a 92-17 antibody. In some embodiments, the method ses CD44 expression in CD8+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure es a method for inducing IFNgamma expression in CD8+ cells, the method comprising providing or administering an effective amount of an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or n binding fragment thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9. l-8mutl3 antibody and/or a 92-17 antibody. In some embodiments, the method increases IFNgamma expression in CD8+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some embodiments, the disclosure provides a method for inducing TNFalpha expression in CD8+ cells, the method comprising providing or administering an effective amount of an anti-Galectin-9 dy described herein, e.g., in Table 1 and/or Table 2, or antigen binding fragment thereof to a subject. In some embodiments, the anti-Galectin-9 antibody is a 9. l-8mutl3 antibody and/or a 92-17 antibody. In some embodiments, the method increases TNFalpha expression in CD8+ cells by at least 30% (e.g., 31%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or greater, including any increment therein).
In some of these embodiments, the methods comprising providing or administering an effective amount of an anti-Galectin-9 antibody described herein, induce CD44, ma, and/or TNFalpha in CD4+ and CD8+ cells. The method embodiments described supra, for suppressing Dectin-l signaling, for suppressing TIM-3 signaling, for suppressing CD206 signaling, for inducing ADCC against target cells, for inducing CDC against target cell, for inducing ADCP against target cells, for inducing T cell activation, for promoting CD4+ cell -l90- tion, for inducing CD44 expression in CD4+ cells, for inducing IFNgamma expression in CD4-- cells, for inducing TNFalpha expression in CD4+ cells, for inducing CD44 expression in CD8-- cells, for inducing IFNgamma expression in CD8+ cells, method for inducing TNFalpha expression in CD8+ cells, wherein the method includes stering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen binding fragment thereof.
The method embodiments described supra (for suppressing Dectin-l signaling, for suppressing TIM-3 signaling, for suppressing CD206 signaling, for inducing ADCC t target cells, for inducing CDC against target cell, for inducing ADCP t target cells, for inducing T cell activation, for promoting CD4+ cell activation, for inducing CD44 expression in CD4+ cells, for inducing IFNgamma expression in CD4+ cells, for inducing TNFalpha expression in CD4-- cells, for inducing CD44 expression in CD8+ cells, for inducing IFNgamma sion in CD8-- cells, for inducing TNFalpha expression in CD8+ cells), wherein the method includes providing an effective amount of an alectin-9 antibody described herein, e.g., in Table 1 and/or Table 2, or antigen binding nt thereof, to a sample ed from a tumor, and measuring in vitro or ex vivo one or more parameters selected from Dectin-l suppression, for TIM-3 suppression, for CD206 suppression, ADCC induction, CDC induction, ADCP induction, induction of T cell activation, promotion of CD4+ cell activation, induction of CD44 expression in CD4+ cells, induction of IFNgamma expression in CD4+ cells, ion of 2O ha expression in CD4+ cells, induction of CD44 expression in CD8+ cells, induction of IFNgamma expression in CD8+ cells, induction of ha expression in CD8+ cells.
The in vivo methods embodiments described supra, wherein the subject in need of administration has , and wherein the cancer is selected from atic cancer, e.g., atic ductal adenocarcinoma, cholangiocarcinoma, hepatocellular carcinoma, colorectal cancer, melanoma, renal cell carcinoma, and acute myeloid leukemia. In some embodiments, cancer low levels of IFNgamma expression, relative to expression of T cell markers. In some embodiments, the cancer is PDA.
The in vitro or ex vivo method embodiments described supra, wherein the sample isolated from a a tumor is from a cancer selected from pancreatic cancer, e.g., pancreatic ductal 3O adenocarcinoma, cholangiocarcinoma, hepatocellular carcinoma, colorectal cancer, melanoma, renal cell oma, and acute myeloid leukemia.
Combination Therapy -l9l- Any of the anti-Galectin-9 dies described herein may be utilized in conjunction with other types of therapy for cancer or autoimmune diseases, such as chemotherapy, surgery, radiation, gene therapy, or in conjunction with other types of therapy for mune diseases, such as immunosuppressive mediation, hormone replacement therapy, blood transfusions, anti- inflammatory medication, and/or pain medication and so forth. Such therapies can be administered simultaneously or sequentially (in any order) with the immunotherapy according to the present disclosure.
In some embodiments, methods are provided herein, wherein the anti-Galectin-9 antibodies described herein are ed in conjunction with other types of therapy for cancer or autoimmune es, such as herapy, surgery, ion, gene therapy, or in conjunction with other types of therapy for autoimmune es, such as immunosuppressive mediation, hormone replacement therapy, blood transfusions, anti-inflammatory medication, and/or pain tion and so forth. In some embodiments, the s include the steps of administering the anti-Galectin-9 antibodies, such as any of the anti-Galectin-9 antibodies described herein, e.g., in Table 1 and/or Table 2 or sequentially (in any order) with the , simultaneously immunotherapy according to the present disclosure. When co-administered with an additional therapeutic agent, suitable therapeutically effective dosages for each agent may be lowered due to the additive action or synergy.
In some embodiments, the methods are provided herein, wherein the anti-Galectin-9 2O antibody, for example dy 92-17 or 9.1-8mutl3, is combined with other immunomodulatory treatments such as, e.g., inhibitors of a checkpoint molecule (e.g., PD-l, PD-Ll, PD-L2, , LAG3, TIM3, or A2aR), activators of a co-stimulatory receptor (e.g., DX40, GITR, CD137, CD40, CD27, and ICOS), and/or inhibitors of an innate immune cell target (e. g., KIR, NKG2A, CD96, TLR, and IDO). Without being bound by theory, it is thought that anti-Galectin-9 antibodies, through their inhibition of Dectin-l, can reprogram immune responses against tumor cells via, e.g., inhibiting the activity of y6 T cells inflltrated into tumor microenvironment, and/or ing immune surveillance against tumor cells by, e.g., activating CD4+ and/or CD8+ T cells. Thus, ed use of an anti-Galectin-9 antibody and an immunomodulatory agent such as those described herein would be expected to icantly enhance anti-tumor efficacy.
In some embodiments, the methods are provided, wherein the anti-Galectin-9 antibody is administered concurrently with a checkpoint inhibitor. In some embodiments, wherein the anti- Galectin-9 antibody is administered before or after a checkpoint inhibitor. In some -l92- embodiments, the checkpoint inhibitor is administered systemically. In some embodiments, the checkpoint inhibitor is administered locally.
In some embodiments, the s are provided, n the anti-Galectin-9 dy, such as any of the Galectin-9 antibodies described herein in Table 1 and/or Table 2, such as 9.2- 17 or 9.1-8mutl3, is capable of improving anti-tumor activity (e.g., reduced tumor proliferation, size, , weight, burden or load, reduction in number of metastatic lesions over time) of the co-administered checkpoint inhibitors (e.g., PD-l, PD-Ll and/or CTLA-4 or others listed herein or known in the art), e.g., by 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or more as compared to a checkpoint inhibitor therapy alone under the same conditions. In some embodiments, the anti-Galectin-9 dy, such as any of the Galectin-9 antibodies described herein in Table 1 and/or Table 2, for example antibody 92-17 or antibody 9. l-8mutl3, is e of ing antitumor activity (e.g., tumor proliferation, size, volume, weight, load or burden, or reduction in number of metastatic lesions over time) of the co- administered checkpoint inhibitors (e.g., PD-land/or CTLA-4 e.g., PD-l, PD-Ll and/or CTLA-4 or others listed herein or known in the art), e.g., l.0-l.2-fold, 1.2-1.4-fold, l.4-l.6-fold, 1.6-1.8- fold, l.8fold, or ld more or more as compared to a checkpoint inhibitor therapy alone under the same conditions. In some embodiments, the anti-Galectin-9 antibody, such as any of the Galectin-9 dies described herein in Table 1 and/or Table 2, ing but not limited to antibody 9.1-8ml3 and/or antibody 92-17, is capable of ing antitumor activity (e.g., 2O tumor proliferation, size, volume, weight, load or burden or reduction in number of metastatic lesions over time) of the co-administered checkpoint inhibitor (e.g., PD-l, PD-Ll and/or CTLA-4 or others listed herein or known in the art), e.g., about three-fold, four-fold, about threefold, four-fold, f1ve-fold, ld, seven-fold, eight-fold, nine-fold, ld, or more as compared to a checkpoint inhibitor therapy alone under the same conditions. In some embodiments, the co-administered checkpoint inhibitors (e.g., PD-l, PD-Ll and/or CTLA-4 or others listed herein or known in the art) are capable of improving anti-tumor activity of the anti- Galectin-9 antibody, such as any of the Galectin-9 antibodies described herein in Table 1 and/or Table 2, including but not limited to antibody 9.1-8ml3 and/or antibody 9.2-l7, (e.g., tumor proliferation, size, volume, weight, burden or load or reduction in number of metastatic lesions ), 3O e.g., by 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or more as compared to anti-Galectin-9 therapy alone under the same conditions. In some embodiments, the co-administered checkpoint inhibitors (e.g., PD-l, PD-Ll and/or CTLA-4 or others listed herein or known in the art) are capable of improving antitumor activity (e.g., tumor eration, size, volume, weight, load or burden or reduction in number of atic lesions over time) of -l93- the anti-Galectin-9 dy, such as any of the Galectin-9 antibodies described herein in Table 1 and/or Table 2, including but not limited to antibody 9. l-8ml3 and/or antibody 9.2-l7, e.g., l.0-l.2-fold, l.2-l.4-fold, l.4-l.6-fold, 1.6-1.8-fold, 1.8fold, or two-fold more or more as compared to an anti-Galectin-9 therapy alone under the same conditions. In some embodiments, the co-administered checkpoint inhibitors (e.g., PD-l, PD-Ll and/or CTLA-4 or others described herein or known in the art) are e of improving mor activity (e.g., tumor proliferation, size, volume, weight, load or burden or reduction in number of metastatic s over time) of the anti-Galectin-9 antibody, such as any of the Galectin-9 antibodies described herein in Table 1 and/or Table 2, including but not limited to, antibody 9.1-8ml3 and/or antibody 9.2-l7, e.g., about three-fold, four-fold, about threefold, old, ld, siX- fold, seven-fold, eight-fold, nine-fold, ld, or more as compared to a anti-Galectin-9 therapy alone under the same conditions.
In some embodiments, the methods are provided, wherein the anti-Galectin-9 antibody, such as any of the Galectin-9 antibodies described herein in Table 1 and/or Table 2, including but not limited to, antibody 9. l-8ml3 and/or antibody 92-17, is capable of improving the ability of the immunotherapy to activate T cells (e.g., as measured by cytokine markers described herein) (e.g., as described herein or known in the art), e.g., by 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or more as compared to a immunotherapy therapy alone under the same conditions. In some embodiments, the anti-Galectin-9 antibody is capable of improving the ability of the immunotherapy to activate T cells (e.g., as measured by cytokine markers described herein) (e.g., as described herein or known in the art), e.g., l.0-l.2-fold, 1.2- ld, l.4-l.6-fold, l.6-l.8-fold, 1.8fold, or two-fold more or more as compared to a immunotherapy therapy alone under the same conditions. In some ments, the anti- Galectin-9 antibody such as any of the Galectin-9 antibodies described herein in Table 1 and/or Table 2, including but not d to, antibody 9. l-8ml3 and/or antibody 92-17, is e of improving the ability of the immunotherapy to te T cells (e.g., as measured by cytokine s described herein) (e.g., as bed herein or known in the art), e.g., about three-fold, four-fold, about threefold, four-fold, f1ve-fold, siX-fold, seven-fold, eight-fold, nine-fold, ten- fold, or more as compared to a immunotherapy therapy alone under the same conditions.
In some embodiments, the methods are provided, wherein the co-administered immunotherapies (e.g., as bed herein or known in the art) are capable of improving the ability of the anti-Galectin-9 antibody, such as any of the Galectin-9 antibodies bed herein in Table 1 and/or Table 2, including but not limited to, antibody 9. l-8ml3 and/or antibody 9.2- 17, to activate T cells (e.g., as measured by cytokine markers described herein), e.g., by 10%, —194— %, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or more as compared to an anti-Galectin-9 therapy alone under the same conditions. In some embodiments, the co- administered immunotherapies (e.g., as described herein or known in the art) are capable of improving the y of the anti-Galectin-9 dy to te T cells (e.g., as measured by cytokine markers described herein ), e.g., l.0-l.2-fold, 1.2-1.4-fold, l.4-l.6-fold, l.6-l.8-fold, l.8fold, or two-fold more or more as ed to an anti-Galectin-9 therapy alone under the same conditions. In some ments, the co-administered therapies (e.g., as described herein or known in the art) are capable of improving the ability of the anti-Galectin-9 antibody to activate T cells (e.g., as measured by cytokine markers described herein), e.g., about three-fold, four-fold, about threefold, old, five-fold, six-fold, seven-fold, eight-fold, nine- fold, ten-fold, or more as compared to an anti-Galectin-9 therapy alone under the same conditions.
In other ments, the methods are provided herein, wherein the anti-Galectin-9 antibody, such as any of the Galectin-9 antibodies bed herein in Table 1 and/or Table 2, including but not limited to, antibody 9.l-8ml3 and/or antibody 92-17, is administered in combination with one or more of the ng modalities for treating autoimmune disorders ing, but not limited to: intravenous Ig therapy, roidal anti-inflammatory drugs (NSAID), and corticosteroids, and anti-inflammatory treatments such as cyclosporins, rapamycins or ascomycins, or their immunosuppressive analogs, e.g., cyclosporin A, cyclosporin G, FK-506, rapamycin, 40-O-(2-hydroxy)ethyl-rapamycin etc., cyclophosphamide, azathioprene, methotrexate, brequinar, FTY 720, leflunomide, mnizoribine, mycophenolic acid, mycophenolate mofetil, 15-deoxyspergualine, immunosuppressive onal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD25, CD28, B7, CD45, or CD58 or their s, or other immunomodulatory compounds, e.g., CTLA4Ig, or other adhesion molecule inhibitors, e.g. mAbs or low molecular weight inhibitors including selectin antagonists and VLA-4 antagonists. These combination therapies can be part of an immunomodulating regimens or a regimen for the treatment or prevention of inflammatory disorders or autoimmune disorders.
In some ments, the s are provided, wherein the anti-Galectin-9 antibody, such as any of the Galectin-9 antibodies described herein in Table 1 and/or Table 2, including but not limited to, antibody 9. l-8ml3 and/or antibody 9.2-l7, can also be co-used with a chemotherapeutic agent, including ting agents, anthracyclines, cytoskeletal disruptors (Taxanes), epothilones, histone deacetylase inhibitors, inhibitors of topoisomerase I, inhibitors of topoisomerase II, kinase inhibitors, nucleotide analogs and precursor analogs, peptide -l95- antibiotics, um-based agents, ids, vinca alkaloids and derivatives thereof.
Non-limiting examples include: (i) anti-angiogenic agents (e.g., INF-470, platelet factor 4, ospondin-l, tissue inhibitors of metalloproteases (TIMPl and THVIPZ), prolactin (l6- Kd fragment), angiostatin (3 8-Kd fragment of plasminogen), endostatin, bFGF soluble receptor, transforming growth factor beta, interferon alpha, e KDR and FLT-l receptors, tal proliferin-related protein, as well as those listed by Carmeliet and Jain (2000)), (ii) a VEGF antagonist or a VEGF receptor antagonist such as anti-VEGF antibodies, VEGF variants, soluble VEGF or fragments, aptamers capable of blocking VEGF or VEGFR, neutralizing anti- VEGFR antibodies, inhibitors of VEGFR tyrosine s and any combinations thereof, and (iii) herapeutic compounds such as, e.g., pyrimidine s (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine), purine analogs, folate antagonists and related inhibitors (mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)), oliferative/antimitotic agents ing natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disruptors such as taxane (paclitaxel, docetaxel), stine, vinblastine, nocodazole, epothilones, and navelbine, epidipodophyllotoxins (etoposide and teniposide), DNA damaging agents (actinomycin, amsacrine, anthracyclines, bleomycin, busulfan, camptothecin, carboplatin, chlorambucil, cisplatin, cyclophosphamide, cytoxan, dactinomycin, daunorubicin, doxorubicin, epirubicin, hexamethyhnelamineoxaliplatin, iphosphamide, lan, orehtamine, mitomycin, mitoxantrone, nitrosourea, plicamycin, procarbazine, taxol, taxotere, teniposide, triethylenethiophosphoramide and etoposide ), antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (adriamycin), icin, anthracyclines, mitoxantrone, bleomycin, plicamycin (mithramycin) and mitomycin, enzymes araginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine), antiplatelet agents, antiproliferative/antimitotic ting agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), nimines and methylmelamines (hexamethylmelamine and thiotepa), alkyl sulfonates-busulfan, nitrosoureas (carmustine (BCNU) and s, streptozocin), trazenes- dacarbazinine (DTIC), antiproliferative/antimitotic antimetabolites such as folic acid analogs 3O (methotrexate), platinum coordination complexes (cisplatin, carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide, hormones, hormone analogs (estrogen, tamoxifen, goserelin, bicalutamide, nilutamide) and aromatase inhibitors (letrozole, anastrozole), anticoagulants (heparin, synthetic heparin salts and other inhibitors of thrombin), f1brinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase), aspirin, -l96- dipyridamole, ticlopidine, clopidogrel, abciXimab; antimigratory agents; antisecretory agents (breveldin); immunosuppressives (cyclosporine, tacrolimus (PK-506), sirolimus ycin), azathioprine, mycophenolate mofetil); anti-angiogenic compounds (e.g., INF-470, genistein, bevacizumab) and growth factor inhibitors (e.g., fibroblast growth factor (FGF) inhibitors); angiotensin or blocker; nitric oxide donors; anti-sense oligonucleotides; antibodies (trastuzumab); cell cycle inhibitors and differentiation inducers (tretinoin); mTOR inhibitors, topoisomerase inhibitors (doxorubicin (adriamycin), amsacrine, camptothecin, daunorubicin, dactinomycin, eniposide, epirubicin, etoposide, idarubicin, mitoxantrone, topotecan, and ecan), corticosteroids (cortisone, dexamethasone, hydrocortisone, methylprednisolone, sone, and prednisolone); growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers and caspase activators; and chromatin disruptors.
In some embodiments, methods are provided herein wherein the alectin-9 antibody, such as any of the Galectin-9 antibodies described herein in Table 1 and/or Table 2, including but not limited to, antibody 9.1-8ml3 and/or dy 9.2-17, is administered concurrently with a chemotherapeutic agent. In some embodiments, methods are provided herein, wherein the anti-Galectin-9 antibody is administered before or after a chemotherapeutic agent. In some embodiments, methods are ed herein, wherein the chemotherapeutic agent is administered systemically. In some ments, methods are provided herein, wherine the chemotherapeutic agent is administered locally.
In some embodiments, the methods are provided, wherein the alectin-9 antibody, such as any of the antibodies described herein in Table 1 and/or Table 2, for example antibody 9.2-17 or antibody 9.1-8mutl3, is capable of improving anti-tumor activity (e.g., tumor proliferation, size, volume, weight, burden load or reduction in number of atic lesions over time) of the co-administered chemotherapeutic agents (e.g., as described herein or known in the art), e.g., by 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or more as compared to a chemotherapeutic agent therapy alone under the same conditions. In some embodiments, the alectin-9 antibody is capable of ing antitumor activity (e.g., tumor eration, size, volume, weight, load or burden or reduction in number of metastatic lesions over time) of the co-administered chemotherapeutic agents (e.g., 3O as described herein or known in the art), e.g., l.0-l.2-fold, l.2-l.4-fold, l.4-l.6-fold, 1.6-1.8- fold, 1.8fold, or two-fold more or more as ed to a chemotherapeutic agent therapy alone under the same conditions. In some embodiments, the anti-Galectin-9 antibody is capable of improving antitumor activity (e.g., tumor proliferation, size, volume, weight, load or burden or ion in number of metastatic lesions over time) of the co-administered -l97- chemotherapeutic agent (e.g., as described herein or known in the art), e.g., about three-fold, old, about threefold, four-fold, f1ve-fold, ld, seven-fold, eight-fold, nine-fold, tenfold , or more as compared to a chemotherapeutic agent therapy alone under the same conditions.
In some ments, the methods are provided, wherein the co-administered chemotherapeutic agents (e.g., as described herein or known in the art) are capable of improving anti-tumor activity of the anti-Galectin-9 antibody, such as any of the antibodies described herein in Table 1 and/or Table 2, for example antibody 92-17 or antibody utl3, (e.g., tumor proliferation, size, volume, weight, burden or load or reduction in number of metastatic s over time) of, e.g., by 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or more as compared to an anti-Galectin-9 therapy alone under the same conditions.
In some embodiments, the co-administered herapeutic agents (e.g., as described herein or known in the art) are capable of improving umor activity (e.g., tumor proliferation, size, volume, weight, burden or load or reduction in number of metastatic lesions over time) of the anti-Galectin-9 antibody, e.g., l.0-l.2-fold, l.2-l.4-fold, l.4-l.6-fold, l.6-l.8-fold, 1.8fold, or two-fold more or more as compared to an anti-Galectin-9 therapy alone under the same conditions. In some ments, the co-administered chemotherapeutic agents (e.g., as described herein or known in the art) are capable of improving antitumor activity (e.g., tumor eration, size, volume, weight, load or burden or reduction in number of metastatic lesions over time) of the anti-Galectin-9 antibody, e.g., about three-fold, four-fold, about threefold, four-fold, f1ve-fold, siX-fold, seven-fold, eight-fold, nine-fold, ten-fold, or more as compared to an anti-Galectin-9 therapy alone under the same conditions.
In some embodiments methods are provided herein, wherein the alectin-9 dy, such as any of the antibodies described herein in Table 1 and/or Table 2, for e antibody 92-17 or antibody 9. l-8mutl3, is capable of improving the ability of the chemotherapeutic agent to activate T cells (e.g., as measured by cytokine markers bed ) (e.g., as described herein or known in the art), e.g., by 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or more as compared to a chemotherapeutic agent therapy alone under the same conditions. In some embodiments, the anti-Galectin-9 antibody is capable of improving the ability of the chemotherapeutic agent to activate T cells (e.g., as measured by cytokine markers described herein) (e.g., as described herein or known in the art), e.g., l.0-l.2-fold, l.2-l.4-fold, l.4-l.6-fold, l.6-l.8-fold, 1.8fold, or two-fold more or more as compared to a chemotherapeutic agent therapy alone under the same conditions. In some embodiments, the anti-Galectin-9 antibody is capable of improving the ability of the -l98- chemotherapeutic agent to activate T cells (e.g., as measured by cytokine markers described herein) (e.g., as described herein or known in the art), e.g., about three-fold, four-fold, about threefold, four-fold, f1ve-fold, siX-fold, seven-fold, eight-fold, old, ten-fold, or more as compared to a chemotherapeutic agent therapy alone under the same conditions.
In some embodiments, methods are provided , n the co-administered chemotherapeutic agents (e.g., as described herein or known in the art) are capable of improving the ability of the anti-Galectin-9 antibody, such as any of the antibodies described herein in Table 1 and/or Table 2, for e antibody 92-17 or antibody 9.1-8mutl3, to activate T cells (e.g., as measured by cytokine markers described herein), e.g., by 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or more as compared to an anti- Galectin-9 therapy alone under the same conditions. In some embodiments, the co-administered chemotherapeutic agents (e.g., as described herein or known in the art) are capable of improving the ability of the anti-Galectin-9 antibody to activate T cells (e.g., as measured by cytokine s described herein), e.g., l.0-l.2-fold, l.2-l.4-fold, l.4-l.6-fold, l.6-l.8-fold, 1.8fold, or two-fold more or more as compared to an anti-Galectin-9 therapy alone under the same conditions. In some embodiments, the co-administered chemotherapeutic agents (e.g., as bed herein or known in the art) are capable of improving the ability of the anti-Galectin-9 antibody to activate T cells (e.g., as measured by cytokine s described herein), e.g., about three-fold, four-fold, about old, old, f1ve-fold, siX-fold, fold, eight-fold, nine- fold, ten-fold, or more as compared to an anti-Galectin-9 therapy alone under the same conditions.
In some these method embodiments, wherein the administration of an anti-Galectin-9 antibody is combined with the administration of a checkpoint inhibitor, the subject has a cancer selected from the group consisting of pancreatic cancer, e.g., atic ductal adenocarcinoma, giocarcinoma, hepatocellular carcinoma, ctal cancer, melanoma, renal cell carcinoma, and acute myeloid leukemia.In some embodiments, methods are provided herein, wherein the anti-Galectin-9 antibody is administered to a patient who is refractory to a previous treatment, e.g., checkpoint inhibitor therapy, such as PD-l.
In some embodiments, the disclosure provides a method for treating a cancer in a subject who is tory to checkpoint inhibitor therapy, the method sing stering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody described herein or antigen binding fragment thereof, wherein the checkpoint inhibitor molecule is selected from the group consisting of PD-l, PD-Ll, PD-L2, CTLA-4, LAG3, TIM3 and A2aR. In some embodiments, the disclosure provides a method for treating a cancer in a subject who is -l99- refractory to checkpoint inhibitor y, the method comprising administering to a subject in need thereof an effective amount of an anti-Galectin-9 antibody bed herein or antigen binding fragment thereof, wherein the checkpoint inhibitor molecule is PD-1. In some embodiments, the cancer is selected from the group consisting of pancreatic , e.g., pancreatic ductal adenocarcinoma, cholangiocarcinoma, hepatocellular carcinoma, colorectal cancer, melanoma, renal cell carcinoma, and acute myeloid leukemiaAdditional useful agents can be found in, e.g., ian's Desk Reference, 59.sup.th edition, (2005), Thomson P D R, Montvale N.J., Gennaro et al., Eds. ton's The Science and Practice of Pharmacy .sup.th edition, (2000), cott Williams and Wilkins, Baltimore Md, Braunwald et al., Eds. Harrison's Principles of Internal Medicine, 15.sup.th edition, (2001), McGraw Hill, NY, Berkow et al., Eds. The Merck Manual of Diagnosis and Therapy, (1992), Merck Research Laboratories, Rahway NJ.
It was reported that chemotherapy and/or immune therapy of solid tumors could enhance the level of immune modulators such as checkpoint molecules, resulting in suppressed immunity against tumor cells. Erisson et al., J. Translational Medicine (2016), 14:282, Grabosch et al., J.
Therapy ofCancer (2015), 3(suppl 2): P302, and Azad et al., EA/[BO J. (2016). Anti- Galectin-9 dies have been found to reprogram immune ses targeting tumor cells, ularly in PDA. As such, the co-use of an anti-Galectin-9 antibody and a chemotherapeutic agent (e.g., gemcitabine) or immunotherapeutic agent (e.g., anti-PD-Ll antibody) would be ed to result in significantly enhanced eutic activity against solid , such as PDA.
In any of the described combination therapies, the onal therapeutic agent or therapy can be stered prior to, simultaneously with, or following administration of the anti- Galectin-9 antibody.
Kits for Use in Treatment of Diseases Associated with Galectin-9 The present disclosure also provides kits for use in treating or alleviating a disease associated with Galectin-9, for example associated with Galectin-9 binding to a cell surface glycoprotein (e.g., Dectin-1, T11V13, etc), or pathologic cells (e.g., cancer cells) expressing Galectin-9. Examples include solid tumors such as PDA and others described herein, and autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus (SLE), autoimmune endocrine ers, autoimmune blood disorders, and others described herein.
Such kits can include one or more containers comprising an anti-Galectin-9 antibody, e.g., any of those described herein, and optionally a second therapeutic agent to be co-used with the anti- Galectin-9 antibody, which is also described herein.
In some ments, the kit can comprise instructions for use in accordance with any of the methods described herein. The included instructions can comprise a description of administration of the anti-Galectin-9 antibody, and optionally the second therapeutic agent, to treat, delay the onset, or alleviate a target disease as those described herein. The kit may r se a description of selecting an individual suitable for treatment based on identifying whether that individual has the target disease, e.g., applying the stic method as described herein. In still other embodiments, the instructions comprise a description of administering an antibody to an individual at risk of the target disease.
The instructions relating to the use of an anti-Galectin-9 antibody generally include information as to dosage, dosing le, and route of administration for the intended treatment. The containers may be unit doses, bulk packages (e.g., multi-dose es) or sub- unit doses. Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but e-readable instructions (e.g., instructions carried on a magnetic or l storage disk) are also acceptable.
The label or package insert indicates that the composition is used for treating, delaying the onset and/or alleviating the disease associated with in-9 (e.g., Dectin-l or TIM-3 signaling). Instructions may be ed for practicing any of the methods described herein.
The kits of this invention are in le packaging. Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. Also contemplated are packages for use in combination with a specific device, such as an inhaler, nasal stration device (e.g., an atomizer) or an infusion device such as a minipump. A kit may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
The container may also have a e access port (for example the ner may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
At least one active agent in the composition is an anti-Galectin-9 antibody as those bed herein.
Kits may optionally provide additional ents such as buffers and interpretive information. Normally, the kit comprises a container and a label or package insert(s) on or associated with the container. In some embodiments, the invention provides articles of manufacture comprising contents of the kits described above.
General Technigues The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art.
Such techniques are ned fully in the literature, such as, Molecular Cloning: A Laboratory , second edition (Sambrook, et al., 1989) Cold Spring Harbor Press, Oligonucleotide Synthesis (M. J. Gait, ed, 1984), Methods in lar Biology, Humana Press, Cell Biology: A Laboratory Notebook (J. E. , ed, 1998) Academic Press, Animal Cell Culture (R. I.
Freshney, ed, 1987), Introduction to Cell and Tissue Culture (J. P. Mather and P. E. Roberts, 1998) Plenum Press, Cell and Tissue Culture: Laboratory Procedures (A. Doyle, J. B. Griffiths, and D. G. Newell, eds, 1993-8) J. Wiley and Sons, Methods in logy (Academic Press, Inc), Handbook of Experimental Immunology (D. M. Weir and C. C. Blackwell, eds), Gene Transfer Vectors for Mammalian Cells (J. M. Miller and M. P. Calos, eds, 1987), Current Protocols in Molecular Biology (F. M. Ausubel, et al., eds, 1987), PCR: The rase Chain Reaction, (Mullis, et al., eds, 1994), Current Protocols in Immunology (J. E. Coligan et al., eds, 1991), Short Protocols in Molecular Biology (Wiley and Sons, 1999), Immunobiology (C. A.
Janeway and P. Travers, 1997), Antibodies (P. Finch, 1997), Antibodies: a practical approach (D. Catty., ed, IRL Press, 1988-1989), Monoclonal antibodies: a practical ch (P.
Shepherd and C. Dean, eds, Oxford University Press, 2000), Using antibodies: a laboratory manual (E. Harlow and D. Lane (Cold Spring Harbor Laboratory Press, 1999), The Antibodies (M. Zanetti and J. D. Capra, eds, d Academic Publishers, 1995).
Without r ation, it is believed that one skilled in the art can, based on the above description, utilize the present invention to its fullest extent. The following specific embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever. All publications cited herein are incorporated by reference for the es or t matter referenced herein.
EXAMPLES Example 1: Generation of alectin-9 Antibodies Codon-optimized genes encoding human Galectin-9 CRD1 (residues 1-148, SEQ ID NO: 3) and mouse Galectin-9 CRD1 (residues 1-147, SEQ ID NO: 5) were cloned as GST fusions using the pGEX vector including thrombin cleavage site and Avitag upstream of the cloned gene. Human Galectin-9 CRD2 (residues 218-355, SEQ ID NO: 4) and mouse Galectin- 9 CRD2 (residues 226-353, SEQ ID NO: 6) were cloned into the pHBT vector, an IPTG inducible expression vector that contains a hexahistadine tag, Avitag and TEV cleavage site upstream of the cloned gene (Sha et al., Proc Natl Acad Sci USA, 2013, 110: 14929).
Human and mouse Galectin-9 CRD2 samples were then purified via harose columns ed by gel filtration to apparent homogeneity and biotinylated in vitro using recombinant BirA. Human and mouse Galectin-9 CRDl samples were purified via GST affinity chromatography followed by thrombin cleavage. Samples were further purified using gel filtration chromatography and biotinylated in a similar manner to Galectin-9 CRD2.
Recombinant full-length mouse Galectin-9 (R&D Systems) was used as a control where necessary.
Antibody clones capable of binding to the human or mouse Galectin-9 fragments as noted above were isolated from a display Fab y. The library s the design of highly successful “Library E” (Miller et al., PloS One, 2012, 7, e43746) with improvements. A total of four rounds of phage library sorting were performed using CRDl and CRD2 samples as the targets, ially following published procedures (Miller et al., PloS One, 2012, 7, e43746, Fellouse et al., JM0] Biol, 2007, 373, 924-940). For CRD2, selection campaigns were performed using (a) only either mouse or human CRD2 as the target or (b) using human and mouse CRD2 s ately in successive rounds of library sorting. For CRDl, only human CRDl samples were used.
Binding to in-9 CRDs was determined by phage ELISA (Sidhu et al., Methods Enzymol, 2000, 328, 3). Biotinylated CRD samples were immobilized to neutravidin- coated wells and d with an excess of biotin. The wells were incubated with phage displaying single Fab clones and then bound phages were detected with HRP-conjugated anti- M13 phage antibody.
Then, phage-displayed Fab clones were pre-incubated with 50 nM non-biotinylated Galectin-9 CRD2 or CRDl prior to addition to ELISA plates. Reduction in the ELISA signal of clones with competitor compared to those without competitor indicated a high affinity and high specificity for Galectin-9 CRDl or CRD2.
From enriched pools of antibody clones, a total of 23 clones that bind to CRD2 (Figs. 1A-1B) and a total of 11 clones to CRDl (Figs. 2A-2B) were identified. Their amino acid sequences were deduced by determining the DNA sequences of the Fab genes in the phage clones (which are provided herein as SEQ ID NO: 7-75 and 77-85).
The genes for a subset of identified antibody clones were transferred into an E. coli expression vector that has previously been described (Zhang et al., Proc NatlAcad Sci USA, 2012, 109, 539). Fab proteins were sed in E. coli BL21 (EMD Millipore) and purified using HiTrap n G HP column (GE Healthcare) as described (Hattori et al., Nat Methods, 2013, 10, 992-995) followed by Superdex S200 or ResourceS column (GE Healthcare). When required, purified Fab was biotinylated via the Avitag attached to the C- terminus of the heavy chain using BirA.
Antibodies in the human IgG1, human IgG4, mouse IgG1 and mouse IgG2a formats were produced by g the genes for the VH and VL regions into mammalian expression vectors for IgG production (Invivogen). Accordingly, mIgGl and mIgG2a are human/mouse hybrids, e the Fc (1'. e. CH2 and CH2) is mouse IgGl, whereas CH1 and CL are human.
The proteins were produced by transient transfection of ExpiCHO cells (ThermoFisher) and purified using Protein G Sepharose chromatography ed by Superdex S200 or ResourceS tography (GE Healthcare).
Example 2: Characterization of alectin-9 Antibody Clones Epilope g Whether the antibody clones bind to distinct (non-overlapping) epitopes in Galectin-9 was examined using competition phage ELISA. The binding of all the CRD2-binding clones were significantly inhibited by pre-incubation of the purified G9.2-1, , 5 or G9.2- 17 Fab clone (Figs. 3A-3B), indicating that the isolated clones bind to an overlapping epitope within CRD2. Clones G9.2-15 and G9.2-17 were selected as representative clones for further characterization because of their strong binding activity and good cross-reactivity between human and mouse Galectin-9 (Figs. .
Epilope g The G9.2-17 clone was selected for further epitope analysis. To determine its epitope on Galectin-9 CRD2, a series of point mutants were constructed. Their ability to bind to G9.2-17 was assayed using phage ELISA, as shown in Fig. 10A. Reductions in ELISA signal indicates sites on Galectin-9 CRD2 that are critical to G9.2-17 binding. Notably, the W309K mutation (residue numbering is according to isoform 1, NCBI GenBank Accession No. BAB83625. 1) dramatically reduced the binding, while the other mutations had marginal effects, suggesting that G9.2-17 binds to a region including W309. Crystal structure analysis of the region showed that it is located opposite the sugar-binding site (Fig. 10B). The term “W309” or ue W309” refers to the tryptophan residue found at position 309 in SEQ ID NO: 1 (Galectin-9) or to the tryptophan residue located at position 277 in the sequence of Galectin-9 isoform 2, —204— t ID 000182-2 or to a residue in CRD2 of Galectin-9 that ponds to the residue found at position 309 in SEQ ID NO: 1 or at position 277 in the sequence of the isoform of t ID 000182-2. The terms “R253”, “R271”, “R334”, and “R341” refer to the arginine residue found at positions 253, 271, 334, and 341, respectively, in SEQ ID NO: 1 or the arginine residue found at positions 221, 239, 302, 309, respectively, in the sequence of Galectin-9 isoform 2, UniProt ID 000182-2. The terms “Y330” and “Y236” refer to the tyrosine residue found at positions 330 and 236, respectively, in SEQ ID NO: 1 or the ne residue found at positions 298 and 204, respectively, in the sequence of Galectin-9 isoform 2, UniProt ID 000182-2.
Mutation that sses the interaction of692-] 7 Fab with chromatography matrix Purified antibody (Fab or IgG) s were run on TOSOH TSngl Bioassist G2WXL columns in PBS and detected using absorbance at 280 nm. The Fab sample of G9.2-17 was found to exhibit a longer retention time than expected for its size, suggesting it interacts with the chromatography column material (Fig. 11). In comparison, the Fab sample of G9.2-Iso eluted with the expected time. A point mutant of G9.2-17, termed G9.2.17mut6, was found to have an improved chromatography profile while retaining the affinity to human and mouse Galectin-9 CRD2 (Fig. 12), suggesting that this mutant has a d level of off-target binding.
Antibodies that bind to a distinct epitope within CRD2 Potential additional epitopes were explored using additional clones that bind to Galectin- 9 CRD2. A phage display library selection using a modified scheme so as to enrich clones that bind to an epitope that is distinct from that of G9.2-17 was performed. Wild type human biotinylated Galectin-9 CRD2, the W309K Galectin-9 CRD2 mutant, or Galectin-9 CRD2 preincubated with 7 IgG was immobilized to neutravidin-coated wells and incubated with individual phage-displayed Fab clones. The results are shown in Fig. 13. Three clones (G9.2- 24, G9.2-25, and G9.2-26) ted similar levels of binding to the three targets tested, wild- type Galectin-9 CRD2, the W309K mutant, and wild-type CRD2 in complex with G9.2-17.
Their binding profiles suggest that they bind to an e that is distinct from that of G9.2-17.
Afinity Measurements The affinities of the antibodies were assessed using a bead-based assay as previously described kori et al., JMol Biol, 2012, 424, 391-399) and e plasmon resonance (SPR). In the bead-based assay, a biotinylated protein (either a Galectin-9 sample or a Fab sample) was immobilized on streptavidin-coated Dynabeads M280 via the biotin-streptavidin interaction. After ng the excess biotin-binding sites on the beads using unconjugated biotin, binding titration was performed by incubating the second component (1'.e., Fab for immobilized Galectin-9 or vice versa), ed by quantification using a dye-labeled neutravidin (ThermoFisher) and flow cytometry analysis. The results obtained from this ment are provided in Figs. 4 and 5. In experiments where the second component is an IgG, a dye-labeled anti-human IgG or anti-mouse IgG dy was used for detection.
In SPR experiments, a biotinylated Galectin-9 sample was immobilized on an Avicap chip (Pall ForteBio) that had been preloaded with neutravidin (ThermoFisher). dy samples were flowed using the OneStep method on a Pioneer SPR instrument (Pall ForteBio) and the results are ed in Fig. 6.
The noted two assays revealed that the analyzed Fab samples had dissociation constant (KD) values in the low or sub nanomolar range to their respective targets, as ed in Figs. 4-6.
Conversion of G9.2-l7 into the human IgG4 format substantially d the dissociation rate, as expected from the bivalent nature of IgG4 (Fig. 7). This was demonstrated using the OneStep method described above. ion ofEndogenous Galectin-Q on Cells To confirm that the antibodies bind to endogenous Galectin-9 produced in human cells, HEK293T and CRL-2134 cell lines were incubated with a biotinylated Fab, and bound Fab was detected using neutravidin conjugated with DyLight 650. s were then analyzed using flow try. Strong signals were observed for CRL-2l34 that expresses Galectin-9 but not for HEK293T that does not express Galectin-9 (Lahm et al., J Cancer Res Clin Oncol, 2001, 127, 375-386) (Fig. 8).
The data demonstrates that these antibodies recognize endogenous Galectin-9 and also show that they have minimal cross-reactivity for other cell surface proteins.To measure dy binding to endogenous Galectin-9 produced in human cells, CRL-2l34 cells were stained using g concentrations of anti-Galectin-9 antibody or a negative control. Samples were then washed and bound antibodies were detected using anti-Mouse IgG conjugated to Dylight 650 (Invitrogen, Carlsbad, CA). Prior to flow cytometry analysis, propidium iodide (l ug/mL) was added to each sample. Samples were then analyzed using flow cytometry. Percentage of galectin-9 positive cells was determined using unstained cells as a negative control. Cell-based iation constant (KD) was calculated from a saturation curve generated based on percentage of galectin-9 positive cells as a function of antibody concentration and the 1:1 binding model Inhibition ofGalectin-Q-mediatedActivation ofDectin-I Signaling Using a reporter cell line for human Dectin-l signaling (Invivogen), the effects of the anti-Galectin-9 antibodies on the signaling activation mediated by Galectin-9 were examined. In this assay, activation of the Dectin-l signaling pathway leads to the secretion of ne phosphatase into the cell media, which is detected as fiable metric changes. Cell lines were incubated with the ted molecules for 16 hours. In the absence of an antibody, Galectin-9 (R&D Systems) robustly activated the reporter on par with ed zymosan, a known ligand for Dectin-l, as shown in Fig. 9. As expected, Galectin-9 exhibited no activation on the matched cell line that did not express Dectin-l.
The antibodies inhibited the activation effect of in-9, suggesting that they block the interaction of Galectin-9 with Dectin-l on the cell surface.
Example 3: Evaluation of anti-Gal-9 antibodies in a mouse model of ductal adenocarcinoma (PDA) To test the effect of treatment with an anti-Gal9 antibody to atic ductal adenocarcinoma (PDA), two PDA mouse models can be used: the slowly progressive PDA model p48Cre,LSL-KrasG12D (KC) in which mice s oncogenic Kras in their pancreatic progenitor cells, and a more aggressive orthotopic PDA model utilizing tumor cells from delCre,LSL-KrasG12D,Tp53Rl72H (KPC) mice, which expresses mutant Kras and p53, as well as in human PDA42,43. A combination of immunohistochemical analysis, flow cytometry, or -fluorescent microscopy can be used to t immune profiling and assess the effect of treatment with anti-galectin antibodies as compared to isotype controls. Similar techniques are used to study human samples derived from FDA patients.
In one example of a mouse study, six week-old KC mice are treated with the anti- galectin antibody, e.g., G9.2-l7, to test the ability of anti-galectin antibody to reduce or prevent tumor growth. Tumor progression is assessed, one, two, three, four, five, six, seven, and eight weeks later compared to vehicle-treated animals. Animals are sacrificed and acinar architecture 3O in the pancreata is ed and scored. Immune profiling is performed according to FACS methods known in the art.
Example 4: Spheroid Preparation and luidic Culture of Patient Tumor Samples Fresh tumor specimens (human patients) are received in media (DMEM) on ice and minced in a 10-cm dish (on ice) using sterile forceps and scalpel. Minced tumor is resuspended in DMEM (4.5 mmol/L e, 100 mmol/L Na pyruvate, 1:100 penicillin—streptomycin, Corning CellGro) + 10% FBS (Gemini Bio- Products), 100 U/mL collagenase type IV (Life Technologies), and 15 mmol/L HEPES (Life Technologies). Samples are pelleted and resuspended in 10 to 20 mL media. Red blood cells (RBC) are removed from Visibly bloody samples using RBC lysis buffer (Boston Bio-Products). Samples are pelleted and then ended in fresh DMEM + 10% FBS and ed over 100-um filter and 40-um filters to generate S1 (>100 um), S2 (40—100 um), and S3 (<40 um) spheroid fractions, which are subsequently maintained in ultralow-attachment tissue culture . S2 fractions are used for ex vivo culture. An t of the S2 fraction is pelleted and resuspended in type I rat tail collagen (Corning) at a concentration of 2.5 mg/mL following the on of 10>< PBS with phenol red with pH adjusted using NaOH. pH 7.0—7.5 is confirmed using PANPEHA Whatman paper (Sigma-Aldrich). The spheroid—collagen e is then injected into the center gel region of a 3-D microfluidic culture deVice as described in Jenkins et al., Cancer Discov. 2018 Feb,8(2):196-215, EX Vivo Profiling of PD-l Blockade Using Organotypic Tumor Spheroids, the contents of which is herein incorporated by reference in its entirety. Collagen hydrogels containing patient-derived organotypic tumor spheroids (PDOTS) are hydrated with media with or without anti-Gal-9 monoclonal antibodies after 30 minutes at 37°C.
In some cases, to test synergy with checkpoint inhibitors or other immunotherapy agents, PDOTS are treated with anti—PD-l (pembrolizumab, 250 , anti-CTLA4 (ipilimumab, 50 ug/mL), or combination (250 ug/mL pembrolizumab + 50 ug/mL ipilimumab). For indicated PDOTS studies, uman PD-Ll (atezolizumab at 600 ug/mL + human IFNgamma. Immune profiling is performed by flow cytometry as bed in s et al.
Example 5: Generation and Analysis of ts of Clone G9.1-8 Mutants of clone G9.1-8 were designed by replacing CDR residues with Ser or by truncation of the CDR regions. The mutant genes were constructed using standard site-directed nesis methods and produced as described in Example 1. A total of 14 mutants were designated as G9.1-8m1- G9.1-8m1 (see Table 4-9). G9.1-8m1 and G9.1-8m2 have mutations to CDR-H2. G9.1-8m3, G9.1-8m4 and G9.1-8m5 mutations to CDR-H3. G9.1-8m6, G9.1-8m7, G9.1-8m8, G9.1-8m8, G9.1-8m10, and G9.1-8m11 have tions to CDR-H3. G9.1-8m12, G9.1-8m13, and G9.1-8m14 have mutations in CDR-H2 and CDR-H3. m12 comprises a combination of G9.1-8m2 and G9.1-8m8 mutations, G9.1-8m13 comprises a combination of G9.1-8m2 and G9.1-8m9 mutations; G9.1-ml4 comprises a combination of G9.1-8m2 and G9.1-8mll mutations.
Table 4. Light and Heavy Chain Sequences for G9.1-8 mutants Light chain SEQ ID Heavy Chain SEQ ID G9.1-8m3 G9.1-8m4 G9.1-8m5 G9.1-8m6 G9.1-8m7 G9.1-8m8 G9.1-8m9 G9.1-8mlO G9.1-8ml l G9.1-8m12 G9.1-8ml3 G9.1-8ml4 Figs. 18-22 depict graphs showing binding data for these clones as measured using the bead g assay as described in Example 2. Table 5-Table XXX list the KD for various clones. As described above, antibodies mouse IgG1 and mouse IgGZa formats were produced by cloning the genes for the VH and VL regions into ian expression vectors for IgG production (Invivogen). lO Table 5. Binding of purified G9.1-8 mutant mIgGl clones as characterized using a bead- based binding assay Clone Designation KD (nM) G9.1-8 WT mIgGl 2.4::1.l G9.1-8m2 mIgGl 6.51:0.65 G9.1-8m4 mIgGl 12.0::4.2 ml mIgGl no able binding G9.1-8m3 mIgGl no detectable binding G9.1-8m5 mIgGl no able binding Table 6. Binding of purified G9.1-8 mutant Fab as characterized using a bead-based binding assay Clone Designation KD (nM) G9.1-8 WT Fab 0.45::0.03 G9.1-8m6 Fab 0.52::0.05 G9.1-8m7 Fab 0.93::0.05 G9.1-8m8 Fab 0.04 G9.1-8m9 Fab 1.57::0.16 G9.1-8m10 Fab 58.4::3.4 G9.1-8m11 Fab 52.7::8.8 Table 7. Binding of purified G9.1-8 mutant mIgGZa as characterized using a bead-based binding assay Clone ation KD (nM) G9 l-8m8 mIgGZa 0.5 1:005 G9.l 8m9 mIgGZa 0.7l::0.04 G9.1-8ml l mIgGZa 1.46::0.05 Table 8. Binding of purified G9.1-8 mutant Fab as characterized using a bead-based binding assay G9.1-8ml4 Fab 58.5::7.7 Table 9. Binding of d G9.1-8 mutant mIgGZa as characterized using a bead-based binding assay Clone Designation KD(nM) G9.l-8 WT mIgGl 0.63:0.07 G9.1-8m12 mIgGZa 0.32::004 G9. l-8ml 3 mIgG2a G9. l-8ml4 mIgG2a These results show that certain es within CDR-H2 and CDR—H3 of the G9. 1-8 clone can be replaced or truncated with minimal effects on antigen binding. Conversion of G9. 1-8 mutant clones from the Fab into the IgG2A format can reduce the iation rate, as expected from the nt nature of IgG (see, e.g., Fig. 21 and Fig. 22, for example, G9. 1- 8ml3 and G9. l-8ml4). Of note, with the original G9. 1-8 clone and G9. l-8m8 reactivity with CRD2 was observed (Fig. 25 and data not shown). G9.l-8m12, G9.l-8ml3, and G9.l-8ml4 do not bind to CRD2 in a bead based assay.
Example 6: Cellular Preparation and Flow Cytometry for Analysis of Mouse and Human Tissues Fresh PDA tumors were placed in cold FACS buffer (PBS with 2 % FB S) with Collagenase IV (1 mg/mL, Worthington Biochemical, Lakewood, NJ), Trypsin inhibitor (1 mg/mL, EMD Millipore, Billerica, MA) and DNase I (2 U/mL, Promega, Madison, WI), and minced with scissors to sub-millimeter pieces. s were then ted at 37 0C for 20 min with gentle shaking every 5 min. Specimens were passed through a 70 um mesh and fuged at 350 g for 5 min. Cell pellets were re-suspended in the FACS buffer and 1x106 cells were first stained with zombie yellow (BioLegend) to exclude dead cells. After viability staining, cells were incubated with an anti-CDl6/CD32 mAb (eBiosciences, San Diego, CA) for blocking FcyRIII/II followed by antibody staining with 1 ug of fluorescently conjugated extracellular mAbs. Intracellular ng for cytokines and transcription factors was performed using the Fixation/ Permeabilization Solution Kit (eBiosciences). For mouse ens, we used mouse CD44 (1M7), PD-l (29F.1A12), CD3 (17A2), CD4 (RM4-5), CD8 (53-67), CD45 (30-F11), CD1 1b (Ml/70), Grl (RB6-8C5), MHC II (MS/114.152), lL-lO (JESS-16E3), IFNy()fl\/IG1.2), TNFoc (MP6-XT22), ICOS (15F9), CD69 (H1.2F3), 1L-17A (TC11-18H10.1), TGFoc (TW7- LFA-1 78), T-bet (eBio4BlO), RORyt (AFKJS-9), and FoxP3 (FJK-l6s, all eBiosciences). The antibodies were from BioLegend unless ise noted. Human flow cytometry antibodies included CD45 (H130), CD3 (UCHTl), CD4 (Al6lAl), CD8 (HIT8a), CD44 (BI18), TNFd (MAbl l), IFNy (4S.B3, all Biolegend). Flow cytometry was carried out on the LSR—II flow cytometer (BD Biosciences). Data were analyzed using FlowJo v. 10.1 (Treestar, Ashland, OR). e 7: Evaluation of Gal-9 Antibodies alone or in combination with Checkpoint Inhibition in a Mouse Model of Pancreatic Cancer and Tumor Mass and Immune Profile of Mice Treated with G9.2-17 mIgGl The effect of G9.2-17 mIgGl on tumor weight and on immune profile was assessed in a mouse model of pancreatic cancer. 8-week old C57BL/6 male (Jackson Laboratory, Bar Harbor, ME) mice were administered intra-pancreatic ions of FC1242 PDA cells derived from , KrasGlZD, Trp53Rl72H (KPC) mice (Zambirinis CP, et al., TLR9 on in pancreatic stellate cells es tumorigenesis. J Exp Med. 2015;212:2077-94). Tumor cells were suspended in PBS with 50% Matrigel (BD Biosciences, Franklin Lakes, NJ) and 1x105 tumor cells were injected into the body of the pancreas via tomy. Mice (n=10/group) received one pre-treatment dose i.p. followed by 3 doses (q.w.) of commercial aGalectin 9 mAb (RG9-1, 200ug, BioXcell, Lebanon, NH) or G9.2-17 mIgGl (200pg), or paired e, either so or rat IgG2a (LTF-2, BioXcell, Lebanon, NH) (200pg) (one dose per week for three . Mice were sacrificed 3 weeks later and tumors were harvested for analyses by flow cytometry. Tissue was processed and prepared and flow cytometric analysis was performed as described in Example 5.
Tumor Mass andImmune Profile ofA/[ice Treated with G9. 2-] 7 mIgG2a alone or in combination with aPD] mAb The effect of G9.2-17 mIgG2a on tumor weight and on immune profile was assessed in a mouse model of pancreatic cancer, alone or in combination with immunotherapy. 8-week old C57BL/6 male mice (Jackson Laboratory, Bar Harbor, ME) were administered intra-pancreatic ions of FC1242 PDA cells derived from delCre, KrasGlZD, Trp53Rl72H (KPC) mice.
Tumor cells were suspended in PBS with 50% Matrigel (BD Biosciences, Franklin Lakes, NJ) and 1x105 tumor cells were injected into the body of the pancreas via laparotomy. Mice received one pre-treatment dose i.p. ed by 3 doses (q.w.) of G9.2-17 mIgG2a (200pg) or a neutralizing aPD-l mAb (29F.1A12, 200 pg, BioXcell, Lebanon, NH), separately or in combination, or paired isotype (LTF-2 and C1.18.4, BioXcell, Lebanon, NH) as indicated. Mice were sacrificed on day 26 and tumors were harvested for analyses. Tissue was processed and prepared and flow cytometric analysis was performed as described in Example 5. Results are shown in Figs. l7A-17C. Each point represents one mouse, 05, >“"‘p<0.01, >“""“p<0.001, >“""“"‘p<0.0001, by unpaired Student’s t-test. These results show single-agent treatment with G9.2-17 mIgGZa s tumor growth at both of the dose levels, whereas anti-PD-l alone had no effect on tumor size.
Example 8: Spheroid Preparation and Analysis of Effect of al9 Antibody In Tumor Spheroids Derived from Patient Samples Patient-derived organotypic tumor spheroids (PDOTS) were prepared from fresh patient tumor ens (pancreatic adenocarcinoma, gall bladder cancer, and liver metastasis from a colorectal cancer). Briefly, specimens were received in media on ice and minced in 10cm dishes and resuspended in DMEM +10% FBS + 100 U/mL collagenase type IV. Partially digested samples were pelleted, re-suspended, and strained over both 100 um and 40 um filters to generate Sl (>100 um), 82 (40-100 um), and S3 (<40 um) spheroid fractions, which were subsequently maintained in low-attachment tissue culture plates. An aliquot of the $2 fraction was pelleted and resuspended in type I rat tail collagen at a concentration of 2.5 mg/mL following addition of 10x PBS with phenol red with pH adjusted using NaOH. The spheroid- collagen mixture was injected into the center gel region of the DAX-l 3D microfiuidic cell e chip. After 30 minutes at 37°C, collagen hydrogels containing PDOTS were ed with media and treated with Gal9 antibody (G9.2-17). Three days later, PDOTS were harvested and were flowed for immune changes. Preliminary results on single patient samples are shown in Figs. 26-29. If more than 100 cells were obtained, then cells were sorted for CD3+, CD4+ and CD8+, otherwise cells were only sorted for CD3+. e 9: Characterization of Effector function1 Cross-reactivity and Immunogenicifl of Gal-9 Antibodies Antibody-dependent cell-mediated cytotoxicily (ADCC) Assay In antibody-dependent cell-mediated cytotoxicity (ADCC), effector cells lyse target cells on which antibodies have bound to specific ns on the target cell ne. The ADCC which is developed is designed to characterize the Fc effector function of antibodies and e ADCC activity. Although the initial assay developed relied on both target and effector cells, the assay is further improved by ly coating the antigen of interest onto plates, bypassing the need for target cells. A recombinant Jurkat T-cell line expressing firefly luciferase gene under the control ofNFAT response elements with constitutive expression of human Ia is used as an effector cell line. Antigen coated onto sterile ELISA plates is used as a target and the test antibody is incubated with the antigen to allow biding through the Fab fragment. Effector cells displaying the correct type of chR are then incubated with the antigen/antibody complex on the plate. When the Fc n of the antibody binds to the a on the surface of effector cells, receptor linking leads to activation of the NFAT pathway, resulting in luciferase expression. Gene sion is allowed to proceed for 5-6 hours and rase activity is measured using a luminometer. A dose-response curve is generated for each antibody.
Antibody-dependent cell-mediatedphagocytosis (ADCP) Assay Antibody-dependent cell-mediated phagocytosis (ADCP) is an important mechanism of action for antibodies that mediate part or all of their action though phagocytosis. In that case, antibodies mediate uptake of specific antigens by antigen presenting cells. ADCP can be mediated by monocytes, macrophages, phils, and dendritic cells, through FcyRIIa, FcyRI, and FcyRIIIa, of which FcyRIIa (CD32a) on macrophages represent the predominant pathway.
In the ADCP assay being employed, THP-1 cells (human monocytic cell line derived from an acute monocytic leukemia patient) are used to measure ADCP. Fluorescently labeled beads are conjugated with the target antigen, then incubated with the test dy. THP-1 cells are then added to the plate to allow their binding to the Fc fraction of antibodies bound to antigen-coated fluorescent beads. Antibody g to the beads and engagement of the Fc-receptor results in an uptake of the beads by the THP-1 cells through a phagocytic mechanism. Phagocytosis events are analyzed using flow try. Total amount of fluorescence in each cell (representing the number of beads phagocytosed) and the percentage of fluorescence-positive cells (representing the frequency of ytosis) are measured. A dose-dependent curve is generated to assess the ADCP activity mediated by each test antibody.
Cross Reactivity Assay Off-target ty can present a significant problem during drug development of a therapeutic monoclonal antibody. As such, city is a critical factor to assess as part the characterization of any new monoclonal candidate and an important indicator of its predicted safety. In order to assess antibody specificity and cross-reactivity, test antibodies and test samples are tested for binding against a human proteomic array consisting of an extensive collection of human proteins — both native and denatured — and at two working concentrations.
Antibody specificity is evaluated using CDI’s HuProt Human Proteome Microarray (~75% of 3O the human proteome). The microarray is ted with the primary antibody, , incubated with a fluorescently-labelled secondary antibody and subsequently ed for the amount of fluorescence detected for each target protein. Results are compiled as microarray images. ion ofanti-drug antibodies (ADA) —214— Detection of anti-drug antibodies is performed on mouse sera from s treated with the anti-Gal9 mAbs in vivo. The ADA assay is run on the Mesoscale Discovery (MSD) platform due to its sed sensitivity and dynamic range over standard ELISA s. Biotin- conjugated Gal9 and sulfo-tagged anti-Gal9 antibodies (drug) will be incubated with test sera to form a bridge complex. ADA bridging complexes will be bound to streptavidin plates and the presence of ADA in the test serum samples will be detected by electrochemiluminescence detection.
Example 10: Evaluation of CRD2 clone 17 IgG1 and IgG4 human Galectin 9 monoclonal lO antibodies in a model of Acute Myeloid Leukemia (AML) in Humanized Mice.
A study is conducted to evaluate CRD2 clone 17 IgG1 and IgG4 human Galectin 9 monoclonal antibodies in a model of Acute Myeloid Leukemia in zed mice (CTG—2243, Champions).
The study protocol is depicted in Table 10.
Table 10. AML Efficacy Study : Mp fl Age—nt Dose tug/dose: ROA/ Schedule l 10 Vehicle Control p.o./qWX4 2 10 Control IgGl/4 100 p.o./qWX4 3 10 Control IgGl/4 200 p.o./qWX4 4 10 Control IgG 1/4 400 p.o./qWX4 10 Anti-Gal9 1/4 p.o./qwx4 6 10 Anti-Gal9 1/4 p.o./qwx4 7 10 Anti- Gal9 1/4 p.o./qWX4 8 10 Cytarabine QDXS 9 10 Cytarabine vehicle QDXS Study Animal Preparation Animals are sublethally ated and reconstituted with 1-5 million primary AML cells via tail vein injection. In-life blood collection is performed once monthly and flow try is conducted using the following flow panel: huCD45/muCD45/huCD3/huCD33 for determination of engraftment. Once human CD33+ levels reach 20-1000 counts/ul, 6 surrogate animals are ized for comprehensive immunophenotyping and spleen, bone marrow and peripheral blood is ed by the flow panel above. Animals are randomized into treatment groups based on peripheral blood counts. Disseminated Tumor growth/burden analysis is conducted up to 42 days dosing and ation. Terminal half whole blood is processed and analyzed for immune parameters and serum is used for Gal9 ELISA.
Terminal blood and bone marrow is collected for flow cytometry. 8-color cell surface flow cytometry is med from terminal bone marrow and peripheral blood from all animals: The flow panels are: LD/huCD45/huCD3/huCD33/huGalectin9/huTim9/huPD l/huCD34/huCD3 8/huCDl 17.
Fresh fecal samples are collected from all animals (1 /mouse) in a polypropylene tube at baseline (prior to ent initiation), at the end of Weekl of treatment, and at study endpoint. The collected samples will aree snap frozen and stored at -80°C. If possible, a terminal blood sample and tissues described is collected to assess drug toxicity.
Data Analysis To assess animal toxicity, beginning on Day 0, animals are observed daily and weighed 3x weekly using a digital scale, data including individual and mean gram weights (Mean We :: SEM), mean percent weight change versus Day 0 (%vDo) are recorded for each group and %vD0 is plotted at study completion. Any animal deaths are recorded daily and designated as drug- related (D), technical (T), tumor-related (B), or unknown (U) based on weight loss and gross observation, single agent or combination groups reporting a mean %vDo >20% and/or >10% mortality are considered above the maximum tolerated dose (MTD) for that treatment on the ted regimen. Maximum mean %vDo t nadir) for each treatment group is reported at study completion. To assess efficacy of the Gal-9 antibody, tumor growth inhibition is measured. Beginning on Day 0, tumor dimensions are measured 3x weekly by digital caliper and data, including individual and mean estimated tumor s (Mean TV :: SEM), are recorded for each group, tumor volume (TV) is calculated using the formula TV: width2 x length x 0.52.
At study completion, percent tumor growth tion (%TGI) values are calculated and reported for each treatment group (T) versus control (C) using initial (i) and final (f) tumor measurements by the formula %TGI = l - (Tf—Ti ) / (Cf-Ci). dual mice reporting a tumor volume £30% of the Day 0 measurement for two consecutive measurements are considered partial responders (PR). Individual mice lacking palpable tumors (0.00 mm3 for two consecutive measurements) are classified as complete responders (CR), a CR that persists until study completion is considered a tumor-free survivor (TF S). Tumor doubling time (DT) is determined for the vehicle treated groups using the formula DT = (Df — D1) * log2 / (logTVf — ) where D = Day and TV = Tumor Volume. All data collected in this study is d onically and stored on a ant server system.
Example 11: Evaluation of Gal-9 Antibody in a B16F10 Melanoma Syngeneic Tumor Model in Immunocompetent Mice Gal-9 antibody G9.2-17 was evaluated in the B16F10 syngeneic mouse model of melanoma immunocompetent mice. Pro-study animals (female C57BL/6, 6-8 weeks of age (Charles River Labs)) were unilaterally implanted subcutaneously on the left flank with 565 B16.F10 in 100 p1 PBS. Pro-study tumor volumes were recorded for each experiment beginning 2-3 days after implantation. When tumors reached an average tumor volume of 50- 100mm3 rably 50-75 mm3) animals were matched by tumor volume into ent or control groups (n=8) to be used for dosing and dosing was initiated on Day 0. Animals were dosed on day 0 and day 4 iv. The study design for testing of Anti-Ga19 7 IgG1 and Anti- Ga19 G9.2-17 IgG2 is summarized in Table 11 and Table 12.
Table 11. Anti-Gal9 IgGl Route of Dose Administr tug/mouse! Control mIgGl 200 ug 200 ul Anti-Ga19 mIgGl (G92- Table 12. Anti-Gal9 IgG2 Control mIgG2 200 ug 200 111 Control mIgG2 200 ug 200 111 Tumor volumes were taken and animals were weighed three times weekly. The study endpoint was set when the mean tumor volume of the control group (uncensored) d 1500mm3.
A final tumor volume was taken on the day the study reached endpoint. A final weight was taken on the day the study d end point (day 10). Tumor volume is shown in Fig. 30 and Fig. 31.
Example 12: Jurkat Apoptosis Assay The y of Galectin-9 G9.2-17 and G9. 1-8m9 antibodies to prevent Galectin induced apoptosis of Jurkat cells was assessed. Jurkat cells 52, ATCC, Manassas, VA) were grown in modified RPMI (2mM L-glutamine, lOmM HEPES, 1mM sodium pyruvate, 4.5 g/L e, 1.5 g/L sodium bicarbonate) with 10% FBS, lOOmU/mL penicillin and 100 ug/mL streptomycin at 37 °C with 5% C02. Cells (2X105 cells/well) were incubated in the wells of a 96-well culture plate with or without the addition of 280 nM Galectin-9 (2045-GA, RnD Systems, polis, MN), 1 uM G9.2-17 IgG, and/or 1 uM G9.l-8m9 IgG. Cells were incubated at 37 °C, 5% C02 for 5 hours and then resuspended in nV-binding buffer. Cells were then stained with AnneXinV-AlexaFluor488 (Invitrogen, Carlsbad, CA) at 4 °C for 30 minutes in the dark. Prior to flow cytometry, propidium iodide (PI)_ was added (1 ug/mL).
Cells were run on Guava easyCyte flow cytometer (MilliporeSigma, Burlington, MA) and analyzed using FlowJo (Treestar, Ashland, OR). Cell population was gated via forward and side scatter, then analyzed on AnneXinV for all apoptotic cells and PI for late apoptotic cell populations. Results are shown in Fig. 23.
Example 13. Anti-Galectin-9 antibodies disrupt the interaction between Galectin-9 and CD206 on high binding 96 well plates (Greiner Bio-One, unster, Austria) were coated with tin-9 (RnD Systems, Minneapolis, MN) (50uL, 4ug/mL in TBS) at room temperature for 1 hour. Plates were then blocked with TB S+O.5% BSA (150uL) at room temperature for 1 hour. Wells were washed three times with TB S+O. 1% BSA. G9. 1-8m13, and/or G9.2-17 antibodies were added to each well (50uL, lOOnM in TBS+O. 1% BSA) and incubated at room temperature for 30 minutes. CD206-His (RnD Systems, Minneapolis, MN) was then spiked 3O in to a final concentration of 13nM and incubated for an additional 30 s at room temperature. Wells were then washed 3 times with TBS+0.05% Tween20. aHis-HRP (ab1187, Abcam, Cambridge, MA) was added to each well (50uL, 1:2500 in TBS+0.05% Tween20, 0.1% BSA) and incubated an onal 30 minutes at room temperature. Wells were washed three times with TB S+0.05% Tween20 and once with TBS. lStep TMB-Ultra ELISA substrate (Thermof1sher, Waltham, MA) (SOuL) was added to each well.
The reaction was neutralized with 2M H2SO4 (SOuL) and absorbance signal at 450nm was read using an Epoch2 spectrophotometer (BioTek, Winooski, VT). Experiments were performed in triplicate; >“p<0.05; >“*p<0.01; >“""“p<0.001; ‘p<0.0001; by unpaired Student’s t-test).
Results are shown in Fig. 24A and Fig. 243 and indicate that both G9.l-8ml3 and G9.2-l7 antibodies inhibit the ction between in-9 and CD206 and their effects are additive.
Example 14. Evaluation of cytotoxic activity To evaluate complement-induced xic activity of 9.1-8mutl3 and 92-17, complement-dependent cytotoxicity (CDC) assays are performed and ed with 2 mouse monoclonal antibodies (mouse onal forms of g9.2-l7 and g9.l-8m9). Antibodies are incubated with the appropriate target cells expressing either gal-9, and species-specif1c serum is added as a source of complement proteins to bind to the cell-bound monoclonal antibodies and initiate complement-dependent cytolysis of the target cell. Cell death of target cells is determined by the differential ng obtained in cells with permeable vs. non-permeable membranes (i.e., lysed vs. unlysed cells) after incubation with a fluorescent cell Viability dye and assessed by flow cytometry.
Exam le 15: Evaluation of Gal-9 Antibod in Two S n eneic Models of Colorectal and Melanoma Cancer in Immunocompetent Mice Gal-9 antibodies G9.2-l7 and G9.l-8ml3 are ted in syngeneic models of colorectal and melanoma cancer in immunocompetent mice. Test articles are formulated and prepared on a weekly basis for the duration of the study according to Table 13.
Table 13. Test articles Master Workin Stock Stock For the For the duratlon of.
Control mIgGl duration of study study -2l9- For the For the duratlon of. l mIgG2 duration of study study For the Gal9-IgGl For the duration of duration of ' ' (G9 . 2- l 7) study study For the Gal9-IgG2 For the duration of duration of ' ' (G9 . 2- l 7) study study For the Gal9-IgGl For the duration of on of_ _ _ (G9. l-8ml3) study study For the mGal9-IgG2 For the duration of duration of ' ' (G9. ) study study For the For the duration of anti-mPD-l duration of study study Vehicle Control: mGal9-IgGl, and mGal9-IgG2; Control mIgGl, Control mIgG2, and anti- mPD-l: Sterile PBS Experimental Design Pre-study animals e C57BL/6, 6-8 weeks of age (Charles River Labs) are acclimatized for 3 days and then are unilaterally implanted aneously on the left flank with 5e5 Bl6.FlO (melanoma cell line) or MC38 cells (colorectal cancer cell line) ended in 100 pl PBS. Pre-study tumor volumes are recorded for each experiment beginning 2-3 days after implantation. When tumors reach an average tumor volume of 50-100mm3 (preferably 50-75 mm3) animals are matched by tumor volume into treatment or control groups to be used for dosing and dosing initiated on Day 0. The study design for testing of Anti-Gal9 IgG1 and Anti- Gal9 IgG2 is summarized in Table 14 and Table 15.
Table 14. Anti-Gal9 IgGl (B16F10 and MC38) Dose Route of m Volume Adminis Group Test Agent Schedule gumeousez tration Control Untreated - Control mlgGl 200 ug 200 111 1V Q4DX6 Control mlgGl 400 ug 200 111 IV Q4DX6 l mIgG2 200 ug 200 111 IP BIWX4 Anti-G319 mlgGl 200 ug 200 111 IV Q4DX6 Anti-G319 mlgGl 400 ug 200 111 IV Q4DX6 Anti-G319 mIgGl 200 111 200 ug 1V Q4DX6 (G9.l-8ml3) Anti-G319 mIgGl 200 111 400 ug 1V Q4DX6 (G9.l-8ml3) Anti-G319 mIgGl - 200 ug 200 200 111 Q4DX6 IV IP mAnti-PDl 11g 200 111 BIWX4 Anti-G319 mIgGl - 400 ug 200 200 111 Q4DX6 IV IP PDl ug 200 111 BIWX4 Anti-G319 mIgGl 200 111 200 ug 200 (G9.l-8ml3) + 200 111 mAnti-PDl Anti-G319 mIgGl 200 111 400 ug 200 (G9.l-8ml3) + 200 111 mAnti-PDl mAnti-PDl 200 ug IP BIWX4 Table 15. Anti-Gal9 IgGZ (B16F10 and MC38) H Dose Test Agent -10 l Untreated - - -10 Control n1IgG2 200 ug 200 111 -10 Control n1IgG2 400 ug 200 111 -10 Control 1111ng 200 ug 200 111 Anti-G319 mIgG2 200 ug 200 111 Anti-G319 mIgG2 400 ug 200 111 Anti-G319 mIgG2 200 111 200 ug (G9.l-8ml3) Anti-G319 mIgG2 200 111 400 ug (G9.l-8ml3) Anti-G319 mIgG2 + 200 ug 200 200 111 mAnti-PDl ug 200 111 Anti-G319 mIgG2 + 400 ug 200 200 111 Q4DX6 8 10 IV IP PDl ug 200 111 BIWX4 Anti-G319 mIgG2 200 111 200 ug 200 IV Q4DX6 7 10 (G9.1-8m13) + 200 111 ug IP BIWX4 mAnti-PDl Anti-G319 mIgG2 200 111 400 ug 200 Q4DX6 8 (G9.1-8m13) + 200 111 IV IP ug BIWX4 PD 1 9 10 mAnti-PDl 200 ug 200 111 IP BIWX4 8 Tumor volumes are taken three times weekly. A final tumor volume is taken on the day the study reaches endpoint. A final tumor volume is taken if an animal is found moribund.
Animals are weighed three times weekly. A final weight is taken on the day the study reaches end point or if animal is found moribund. Animals ting 210% weight loss when compared to Day 0 are provided DietGel® ad libitum. Any animal exhibiting >20% net weight loss for a period lasting 7 days or if mice display >30% net weight loss when ed to Day 0 is considered moribund and is euthanized. The study endpoint is set when the mean tumor volume of the control group (uncensored) reaches 3. If this occurs before Day 28, treatment groups and individual mice are dosed and ed up to Day 28. If the mean tumor volume of the control group (uncensored) does not reach 1500mm3 by Day 28, then the endpoint for all animals is the day when the mean tumor volume of the control group (uncensored) reaches 1500mm3 up to a maXimum of Day 60. Blood is collected from all animals from each group. For blood collection, as much blood as possible is collected via a cardiac puncture into KzEDTA tubes (400 ul) and serum separator tubes (remaining) under deep anesthesia d by isofiurane inhalation. The blood collected into KzEDTA tubes is placed on wet ice until used for performing immune panel flow as shown in Table 16.
Table 16. Flow Cytometry Panel 1 Antibody Conjugate Supplier Description BioLegend BioLegend BioLegend APC-R7OO BioLegend mCD44 1M7 BioLegend mCDl lb Ml/7O BioLegend mCD45 30-Fll BioLegend Live Dead ' BioLegend MEL-14 mCD62L BioLegend 29F-1A12 mPD-l BioLegend UC10'4B9 mCTLA4 BioLegend mCD27 LG~3A10 BioLegend Blood collected into serum separator tubes is allowed to clot at room temperature for at least 15 minutes. Samples are centrifuged at 3500 for 10 minutes at room temperature. The resultant serum is separated, transferred to uniquely labeled clear opylene tubes, and frozen immediately over dry ice or in a freezer set to maintain -80°C until shipment for the bridging ADA assay (shipped within one week).
Tumors from all animals are collected as follows. Tumors less than 400 mm3 in size are snap frozen, placed on dry ice, and stored at -80C until used for RT-qPCR analysis. For tumors of 400-500 mm3 in size, whole tumors are ted into MACS media for use in the Flow Panel (shown in Table 16A below). For tumors r than 500 mm3 in size, a small piece (about 50 lO mm3) is snap frozen placed on dry ice, and stored at -80C for RT-qPCR, and the remaining tumor is collected in MACS media for flow cytometry (as shown in Table 16A). For flow cytometry, tumors are placed in MACS media and stored on wet ice until processed. A summary of the flow cytometry is performed is shown in Table 16A.
Table 16A. Flow cytometry Panel 2 Antibody Con] ugate. Supplier.
Description BioLegend BioLegend APC-R7OO 53-67 BioLegend BV421 Hl .2F3 BioLegend mCDl 1b M1/70 BioLegend mCD45 30-F11 BioLegend Live Dead ' BioLegend MEL-14 mCD62L BioLegend 29F-1A12 mPD-1 end mCTLA4 UC10'4B9 BioLegend me1.1 PK136 BioLegend Spleen, liver, colon, lungs, heart, and kidneys from all animals are retained in 10% l buffered in (NBF) for 18-24 hours, transferred to 70% ethanol and stored at room temperature. Formalin fixed samples are paraffin embedded.
Exam le 16: Evaluation of Gal-9 Antibod in a Models of Cholan iocarcinoma The efficacy of Gal-9 antibody is assessed in a mouse model of cholangiocarcinoma as described in S. Rizvi, et al. (YAP-associated chromosomal instability and cholangiocarcinoma in mice, Oncotarget, 9 (2018) 5892-5905), the contents of which is herein incorporated by nce in its entirety. In this transduction model, in which oncogenes (AKT/YAP) are instilled directly into the biliary tree, tumors arise from the biliary tract in immunocompetent hosts with species-matched tumor microenvironment. Dosing is bed in Table 17.
Table 17. Dosing Route of Total Administ Schedule Number of u mouse ration Doses 1ROA1 Control mIgG2 200 ug 200 111 Q4DX6 6 Control mIgG2 400 ug 200 111 Q4DX6 6 l mIgG2 200 ug 200 111 BIWX4 8 Anti-G319 mIgG2 200 111 200 ug IV Q4DX6 6 (G9.2-17) Anti-G319 mIgG2 200 111 400 ug Q4DX6 6 (G9.2-17) Anti-G319 mIgG2 200 111 200 ug Q4DX6 6 (G9.1.8-m13) —224— In brief, murine CCA cells (described in S. Rizvi, et al) are harvested and washed in DMEM. Male C57BL/6 mice from Jackson Labs are anesthetized using l.5—3% isoflurane.
Under deep anesthesia, the abdominal cavity is opened by a 1 cm incision below the xiphoid process. A e cotton tipped applicator is used to expose the superolateral aspect of the medial lobe of the liver. Using a 27-gauge needle, 40 uL of standard media containing l X 10/‘6 cells is injected into the lateral aspect of the medial lobe. Cotton tipped applicator is held over the injection site to prevent cell leakage and blood loss. Subsequently, the abdominal wall and skin are closed in separate layers with absorbable chromic 3—0 gut suture material.
Two weeks post implantation, animals are matched by tumor volume into treatment or control groups to be used for dosing and dosing initiated on Day 0. Tumor volumes are measured and animals weighed three times weekly. A final tumor volume and weight is taken on the day the study reaches endpoint (4 weeks or when tumor burden of control becomes 1500mm3). Blood is collected from all animals from each group. Tumors from all animals are collected essentially as described in Example 15. Analysis is performed ially as bed in Example 15.
EQUIVALENTS From the above description, one skilled in the art can easily ascertain the essential teristics of the present invention, and without departing from the spirit and scope f, can make various changes and modifications of the invention to adapt it to various usages and conditions. Thus, other embodiments are also within the claims.
While several inventive embodiments have been described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the function and/or obtaining the results and/or one or more of the advantages described herein, and each of such ions and/or modifications is deemed to be within the scope of the inventive embodiments described herein. More lly, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be exemplary and that the actual parameters, ions, materials, and/or 3O configurations will depend upon the c ation or applications for which the inventive teachings is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routine mentation, many equivalents to the specific inventive embodiments described herein. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the ed claims and equivalents thereto, inventive embodiments may be practiced otherwise than as specifically described and claimed. Inventive ments of the present disclosure are directed to each individual feature, system, article, material, kit, and/or method described herein. In addition, any combination of two or more such features, systems, articles, materials, kits, and/or methods, if such features, systems, articles, materials, kits, and/or methods are not mutually inconsistent, is included within the inventive scope of the present disclosure.
All definitions, as defined and used herein, should be understood to control over nary definitions, ions in documents incorporated by reference, and/or ordinary meanings of the defined terms.
All references, patents and patent applications disclosed herein are incorporated by reference with t to the subject matter for which each is cited, which in some cases may encompass the entirety of the nt.
The indefinite articles “a” and “an,” as used herein in the specification and in the claims, unless clearly ted to the contrary, should be tood to mean “at least one.” The phrase “and/or,” as used herein in the cation and in the , should be understood to mean “either or bot ” of the elements so conjoined, 1'. e., elements that are conjunctively present in some cases and disjunctively present in other cases. le elements listed with “and/or” should be construed in the same fashion, i.e., “one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to “A and/or B”, when used in ction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B), in another embodiment, to B only nally including ts other than A), in yet another embodiment, to both A and B (optionally including other elements), etc.
As used herein in the specification and in the claims, “or” should be understood to have the same meaning as “and/or” as defined above. For example, when ting items in a list, “or” or “and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, ally, additional unlisted items. Only terms clearly indicated to the contrary, such as “only one of’ or “exactly one of,” or, when used in the claims, “consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e., “one or the other but not both”) when preceded by terms of exclusivity, such as “either,” “one of,” “only one of,” or ly one of.” “Consisting essentially of,” when used in the claims, shall have its ordinary meaning as used in the field of patent law.
As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of ts in the list of ts. This definition also allows that elements may optionally be t other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a miting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally ing more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, ally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
It should also be understood that, unless clearly indicated to the ry, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not arily limited to the order in which the steps or acts of the method are recited.
Definitions of specific embodiments of the invention as claimed herein follow.
According to a first embodiment of the invention, there is provided an isolated antibody, which binds human galectin-9 polypeptide, wherein the antibody comprises a heavy chain complementarity determining region 1 (CDR1) set forth as SEQ ID NO: 361, a heavy chain complementary determining region 2 (CDR2) set forth as SEQ ID NO: 388, and a heavy chain complementary determining region 3 (CDR3) set forth as SEQ ID NO: 406 and comprises a light chain complementarity determining region 1 (CDR1) set forth as SEQ ID NO: 328, a light chain complementary determining region 2 (CDR2) set forth as SEQ ID NO: 329, and a light chain complementary determining region 3 (CDR3) set forth as SEQ ID NO: 352.
According to a second embodiment of the invention, there is ed a ceutical composition comprising the antibody of the first embodiment and a pharmaceutically able carrier.
According to a third embodiment of the invention, there is provided the use of the isolated antibody of the first embodiment in the cture of a medicament for the inhibition of galectin-9 activity in a subject.
According to a fourth embodiment of the invention, there is provided the use of the isolated antibody of the first embodiment in the manufacture of a medicament for treating a solid tumor that expresses galectin-9 in a human subject.

Claims (24)

What We Claim Is
1. An isolated antibody, which binds human galectin-9, wherein the antibody comprises a heavy chain complementarity determining region 1 (CDR1) set forth as SEQ ID 5 NO: 361, a heavy chain complementarity determining region 2 (CDR2) set forth as SEQ ID NO: 388, and a heavy chain complementarity determining region 3 (CDR3) set forth as SEQ ID NO: 406 and comprises a light chain complementarity determining region 1 (CDR1) set forth as SEQ ID NO: 328, a light chain mentarity determining region 2 (CDR2) set forth as SEQ ID NO: 329, and a light chain complementarity determining region 3 (CDR3) set forth as SEQ ID 10 NO: 352.
2. The isolated antibody of claim 1, wherein the antibody binds to an e in a carbohydrate recognition domain (CRD) of galectin-9. 15
3. The ed dy of claim 2, wherein the CRD is CRD2.
4. The isolated antibody of claim 3, wherein the CRD2 consists of the amino acid sequence of SEQ ID NO: 4. 20
5. The isolated antibody of any one of claims 1 to 4, which is a full-length antibody or an antigen-binding fragment thereof.
6. The isolated antibody of any one of claims 1 to 5, which is a single chain dy. 25
7. The isolated antibody of any one of claims 1 to 6, which is a human antibody or a humanized antibody.
8. The isolated antibody of any one of claims 1 to 7, which is an IgG molecule. 30
9. The isolated antibody of any one of claims 1 to 8, wherein the antibody is an IgG1 or IgG4 molecule.
10. The isolated antibody of any one of claims 1 to 9, wherein the dy is an IgG4 molecule, wherein the IgG4 has a S228P mutation.
11. The isolated antibody of any one of claims 1 to 10, wherein the antibody ses a VH 5 set forth as SEQ ID NO: 55 and/or a VL set forth as SEQ ID NO: 54.
12. The isolated antibody of any one of claims 1 to 11, wherein the antibody comprises a heavy chain nt region set forth as SEQ ID NO: 422, and/or a light chain constant region set forth as SEQ ID NO: 418.
13. The isolated antibody of any one of claims 1 to 11, wherein the antibody comprises a VH set forth as SEQ ID NO: 55 and a VL set forth as SEQ ID NO: 54, and has a heavy chain constant region set forth as SEQ ID NO: 422 and a light chain constant region set forth as SEQ ID NO: 418.
14. The isolated antibody of claim 13, wherein the antibody comprises a heavy chain set forth as SEQ ID NO: 316 and a light chain set forth as SEQ ID NO:108.
15. The isolated antibody of any one of claims 1 to 10, wherein the antibody has a VL CDR1 20 set forth as SEQ ID NO: 328, a VL CDR2 set forth as SEQ ID NO: 329, a VL CDR3 set forth as SEQ ID NO: 352, a VH CDR1 set forth as SEQ ID NO: 361, a VH CDR2 set forth as SEQ ID NO: 388, a VH CDR3 set forth as SEQ ID NO: 406, and has a heavy chain constant region set forth as SEQ ID NO: 422 and a light chain nt region set forth as SEQ ID NO: 418. 25
16. The isolated antibody of any one of claims 1 to 10, wherein the dy comprises a heavy chain set forth as SEQ ID NO: 316 and/or a light chain set forth as SEQ ID NO:108.
17. A pharmaceutical composition comprising the antibody any one of claims 1 to 16 and a pharmaceutically acceptable carrier.
18. Use of the isolated antibody of any one of claims 1 to 16 in the manufacture of a medicament for the inhibition of galectin-9 activity in a subject.
19. Use of the isolated antibody of any one of claims 1 to 16 in the manufacture of a medicament for treating a solid tumor that expresses galectin-9 in a human subject.
20. The use of claim 18, n the subject is suspected of having, or is at risk for a solid 5 tumor.
21. The use of claim 19 or claim 20, wherein the solid tumor is selected from the group consisting of glioblastoma, glioma, melanoma, skin squamous cell carcinoma, sarcoma, upper and lower gastrointestinal cancers, carcinoid tumors, neuroendocrine , breast cancers, 10 lung cancers, head and neck cancers, and genitourinary cancers.
22. The use of claim 21, wherein the upper and lower intestinal disorders are selected from the group consisting of esophageal cancer, gastric cancer, pancreatic cancer, colorectal cancer, biliary tract s, cholangiocarcinoma, gall bladder cancer, liver cancer, and duodenal cancer.
23. The use of any one of claims 18 to 22, n the medicament is formulated to be administered to the human subject with an ive amount of a checkpoint tor.
24. The use of claim 23, wherein the checkpoint inhibitor is an anti-PD-1 antibody.
NZ764590A 2017-10-27 2018-10-29 Anti-galectin-9 antibodies and uses thereof NZ764590B2 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US201762578111P 2017-10-27 2017-10-27
US62/578,111 2017-10-27
US201862665175P 2018-05-01 2018-05-01
US62/665,175 2018-05-01
US201862736317P 2018-09-25 2018-09-25
US62/736,317 2018-09-25
PCT/US2018/058028 WO2019084553A1 (en) 2017-10-27 2018-10-29 Anti-galectin-9 antibodies and uses thereof

Publications (2)

Publication Number Publication Date
NZ764590A NZ764590A (en) 2020-12-18
NZ764590B2 true NZ764590B2 (en) 2021-03-19

Family

ID=

Similar Documents

Publication Publication Date Title
US11629191B2 (en) Anti-galectin-9 antibodies and uses thereof
US20220372155A1 (en) Anti-cd40 binding molecules and bi-specific antibodies comprising such
US11505615B2 (en) Anti-CD137 antibodies and uses thereof
US20220204629A1 (en) Anti-galectin-9 antibodies and uses thereof
US20210355220A1 (en) Antibodies specific to ctla-4 and uses thereof
NZ764590B2 (en) Anti-galectin-9 antibodies and uses thereof
US20240218069A1 (en) Bi-specific antibodies comprising anti-b7h3 binding molecules
US20230034659A1 (en) Anti-pdl1 antibodies and uses thereof
WO2024073522A2 (en) Antibodies binding to leukocyte immunoglobulin-like receptor subfamily b member 2 (lilrb2) and uses thereof