WO2023077172A2 - Nouveaux anticorps anti-lilrb2 et produits dérivés - Google Patents

Nouveaux anticorps anti-lilrb2 et produits dérivés Download PDF

Info

Publication number
WO2023077172A2
WO2023077172A2 PCT/US2022/079097 US2022079097W WO2023077172A2 WO 2023077172 A2 WO2023077172 A2 WO 2023077172A2 US 2022079097 W US2022079097 W US 2022079097W WO 2023077172 A2 WO2023077172 A2 WO 2023077172A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
lilrb2
antigen
binding
binding fragment
Prior art date
Application number
PCT/US2022/079097
Other languages
English (en)
Inventor
Hongyu Tian
Krista Mccutcheon
Ryan STAFFORD
Kyu Hee Hong
Maria Jose COSTA
Jing-Tyan Ma
Original Assignee
Immune-Onc Therapeutics, Inc.
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 Immune-Onc Therapeutics, Inc. filed Critical Immune-Onc Therapeutics, Inc.
Publication of WO2023077172A2 publication Critical patent/WO2023077172A2/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • 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
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • 8016W001_ST26 which is 554,405 bytes (as measured in Microsoft Windows) and was created on November 01, 2022, is filed herewith by electronic submission and is incorporated by reference herein.
  • the present disclosure relates generally to the fields of biochemistry, molecular biology, cell biology, immunology and oncology. More particular, the disclosure relates to antibodies that bind to LILRB2 and their use in detecting LILRB2.
  • LILR Leukocyte Immunoglobulin-Like Receptors
  • ILTs Immunoglobulin-like transcripts
  • CD85 are a family of receptors that can exert immunomodulatory effects on a wide range of immune cells.
  • the LILRs belong to the Ig superfamily and have either two or four Ig-like extracellular domains. There are inhibitory (LILRB1-5) and activating (LILRA1-6) receptors.
  • LILRB1-5 inhibitory
  • LILRA1-6 activating receptors.
  • the expression of LILRs varies widely on immune cells, but they are found primarily on antigen presenting cells (APC) of the myeloid lineage such as monocytes, macrophages and dendritic cells, as well as on B cells and on subsets of NK and T cells.
  • APC antigen presenting cells
  • LILRB2 LIR subfamily B member 2
  • ILT4 Immunoglobulin-like transcript 4
  • LIR2 or LIR-2 Leukocyte Immunoglobulin-like Receptor 2
  • CD85d or CD85D has emerged as a key immune checkpoint mediating the tolerogenic activity of myeloid cells associated with cancer and is thought to be expressed primarily by myeloid cells (monocytes, macrophages, dendritic cells and neutrophils).
  • myeloid cells monocytes, macrophages, dendritic cells and neutrophils.
  • the accurate identification of LILRB2 expression by immunohistochemistry and immunofluorescence staining has been a problem due to the lack of antibodies specifically binding to LILRB2 and not cross-reacting with other LILR family members. Therefore, there is a significant need for novel anti-LILRB2 antibodies to provide highly sensitive and specific detection of LILRB2 by immunohistochemistry.
  • the present disclosure provides anti-LILRB2 antibodies and antigen-binding fragment thereof, amino acid and nucleotide sequences thereof, anti-LILRB2 chimeric antigen receptors, and uses thereof.
  • the present disclosure provides an isolated anti-LILRB2 antibody or an antigen-binding fragment thereof.
  • the anti-LILRB2 antibody or an antigen-binding fragment comprises a heavy chain (HC) variable region (VH) and a light chain (LC) variable region (VL), wherein the VH and VL comprise clone-paired complementarity determining region (CDR) sequences as set forth in Table 1, and variants thereof wherein one or more of the HC-CDRs and/or LC-CDRs has one, two, or three amino acid substitutions, additions, deletions or combination thereof.
  • CDR complementarity determining region
  • the VH and VL have amino acid sequences at least 90% or 95% identical to clone-paired sequences of Table 2. In some embodiments, the VH and VL have amino acid sequences identical to clone-paired sequences of Table 2.
  • the isolated antibody is a mouse, a rat, a hamster, a guinea pig, sheep, goat, donkey, horse, llama, or a rabbit antibody.
  • the antigen-binding fragment is a recombinant ScFv (single chain fragment variable) antibody, a Fab fragment, a F(ab’)2 fragment, or a Fv fragment.
  • the isolated antibody or an antigen-binding fragment thereof disclosed herein further comprises an immunoglobulin constant region, optionally a constant region of Ig, or optionally a constant region of IgG.
  • the isolated antibody or an antigen-binding fragment thereof disclosed herein is linked to a detectable label.
  • the detectable label is a peptide tag, an enzyme, a magnetic particle, a chromophore, a fluorescent molecule, a chemo-luminescent molecule, or a dye.
  • the present disclosure provides a pharmaceutical composition comprising the isolated antibody or an antigen-binding fragment thereof disclosed herein, and a pharmaceutically acceptable carrier.
  • the present disclosure provides an isolated polynucleotide encoding the isolated antibody or antigen-binding fragment thereof disclosed herein.
  • the present disclosure provides a vector comprising the isolated polynucleotide encoding the isolated antibody or antigen-binding fragment thereof disclosed herein.
  • the present disclosure provides a host cell comprising the vector disclosed herein.
  • the present disclosure provides a method of producing an antibody or antigen-binding fragment thereof, comprising culturing the host cell disclosed herein under the condition at which the antibody or antigen-binding fragment thereof is expressed, and recovering the antibody or antigen-binding fragment thereof.
  • the present disclosure provides a method for detecting LILRB2 in a sample.
  • the method comprises: (a) contacting a sample containing LILRB2 with the antibody or an antigen-binding fragment thereof disclosed herein, and (b) detecting binding of the antibody or an antigen-binding fragment thereof to the LILRB2 in the sample.
  • the sample is a cell, tissue or organ. In some embodiments, the sample is obtained from a human subject.
  • the binding of the antibody or an antigen-binding fragment thereof to the LILRB2 is detected by immunohistochemistry, immunofluorescence, flow cytometry, Western blot, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA) or other immunoassays.
  • the present disclosure provides a method of generating an antibody specifically binding to LILRB2.
  • the method comprises immunizing a non-human animal with a peptide having an amino acid sequence as set forth in any one of SEQ ID NO: 25-27.
  • the method further comprises generating from the animal a hybridoma expressing an antibody specifically binding to LILRB2.
  • the method further comprises obtaining a polynucleotide sequence encoding the antibody specifically binding to LILRB2.
  • the non-human animal is a mouse, a rat, a hamster, a guinea pig, llama, goat, sheep, donkey, horse or a rabbit.
  • the present disclosure provides a kit comprising the antibody or antigen-binding fragment thereof disclosed herein, useful in detecting LILRB2.
  • Figure 1 shows the screening of anti-LILRB2 hybridoma clones in the immunohistochemistry (IHC) assay.
  • Formalin-fixed paraffin-embedded (FFPE) cell pellets of HEK293 stably expressing full length LILRB2 (hereafter designated as HEK293 LILRB2) were processed and stained with supernatant of hybridoma clones generated from mouse immunization with antigen GQIHGTPFI ( Figure 1A, 1 : 10 dilution) or MGSSPPPTGPISTP ( Figure IB, 1 :5 dilution). Clone 16B9 was used as positive control for results shown in Figure 1A.
  • Figure 2 shows the results for binding specificity testing of anti-LILRB2 hybridoma clones in the IHC assay.
  • HEK293 cells cultured on microscope slide coverslips were transiently transfected with LILRB2, LILRB1, LILRB3 or LILRA6. Non-transfected cells were used as negative control.
  • the specificity of 16B9, 29F12, 49G11 ( Figure 2A) or 32G8, 37B2 ( Figure 2B) were tested using IHC staining with supernatant diluted 1 :5 from each hybridoma clone.
  • Figure 3 shows the titration of anti-LILRB2 recombinant antibodies in the IHC assay.
  • FFPE sections of HEK293 or HEK293 LILRB2 cell pellets were processed and stained with a 2-fold dilution series (10-0.15625 pg/mL) of recombinant 16B9 or 37B2. Nuclei were counterstained with hematoxylin.
  • Figure 4 shows results of specificity testing for recombinant 16B9 and 37B2 antibodies.
  • Figure 4A Sequence alignment of peptides among different members of LILR family compared to peptide sequence of LILRB2 antigen (MGSSPPPTGPISTP).
  • Figure 4B- 4D Evaluation of cross-reactivity of recombinant 16B9 (Figure 4B and Figure 4C) and 37B2 ( Figure 4D) antibodies to the peptides from different LILR, as assessed by ELISA ( Figure 4B) and bio-layer interferometry (BLI) ( Figure 4C and Figure 4D).
  • Figure 4E HEK293 cells cultured on microscope slide coverslips were transiently transfected with LILRB2, LILRB1 or LILRB3, then the specificity of 16B9 was tested at different concentrations, using IHC staining.
  • Figure 5 shows the detection of endogenous LILRB2 in human normal tissues by IHC using recombinant 16B9 and 37B2.
  • FFPE sections of lung, spleen or bone marrow from healthy human donors were stained by IHC using 16B9 ( Figure 5 A and Figure 5B) or 37B2 ( Figure 5B) at different concentrations.
  • the nuclei were counterstained with hematoxylin.
  • Figure 6 shows the detection of endogenous LILRB2 in human normal tissues using recombinant 16B9 by IHC and immunofluorescent staining.
  • Figure 6A FFPE tissue sections of human normal lung were stained by IHC with recombinant 16B9 antibody or anti- CDl lb. The nuclei were counterstained with hematoxylin.
  • Figure 6B FFPE tissue sections of human normal lung and spleen were stained with recombinant 16B9 and anti-CDl lb antibodies by immunofluorescence staining; nuclei were counterstained with 4’,6-diamidino- 2-phenylindoJe (DAPI).
  • DAPI diamidino- 2-phenylindoJe
  • Figure 7 shows the detection of endogenous LILRB2 in human tumor tissues by IHC or immunofluorescent staining using recombinant 16B9 and 37B2 antibodies.
  • Figure 7 A FFPE sections of human tumors of breast, colon, ovary, liver or lung were stained by IHC with 2.5 pg/mL recombinant 16B9 or anti-CD163. The nuclei were counterstained with hematoxylin.
  • Figure 7B FFPE sections of human tumors of colon and ovary were stained with recombinant 16B9 and anti-CD163 antibodies by immunofluorescence; nuclei were counterstained with DAPI.
  • Figure 7C FFPE sections of human ovarian tumor were stained by IHC with different concentrations of recombinant 16B9 or 37B2 antibodies. The nuclei were counterstained with hematoxylin.
  • Figure 8 shows the identification of the critical LILRB2 amino acids for binding of 16B9 or 37B2 antibodies.
  • Figure 8A and Figure 8B Binding sensorgrams obtained by BLI analysis of captured biotinylated peptides interacting with recombinant 16B9 ( Figure 8A) or 37B2 ( Figure 8B).
  • Figure 8D Peptides synthesized to determine which amino acid residues are necessary for antibody specificity to LILRB2 versus LILRA6 or LILRB3.
  • FIG. 8E and Figure 8F Binding sensorgrams obtained by BLI analysis of captured biotinylated peptide interacting with recombinant 16B9 ( Figure 8E) or 37B2 ( Figure 8F).
  • Figure 9 shows that 16B9 recombinant antibody can detect LILRB2 by IHC staining in FFPE samples of HEK293 LILRB2 cells that have been pre-treated with a therapeutic anti-LILRB2, in this case an antagonist antibody (e.g., a B2-19 variant ⁇ Cultured HEK293 LILRB2 cells were detached from tissue culture dishes, resuspended in culture medium, and then treated, or left untreated, with 20 pg/mL anti-LILRB2 antagonist antibody in suspension for 1 hour at 37 °C. The cells were then washed in PBS, processed into FFPE sections and stained with 16B9 recombinant antibody at different concentrations. The nuclei were counterstained with hematoxylin.
  • Figure 10 shows the logo plot of enriched NGS sequences after ELISA binding selection of a 16B9 ribosome display scFv NNK walk library on peptide #1, #5 or #8. Sequences with >100 reads were included in the analysis and the total number of reads derived from sequencing of each amplicon sample is shown. Positions indicated by an astrix were changed in IgG variants.
  • Figure 11 shows the bilayer interferometry (BLI) sensorgrams of five different versions of the 16B9 antibody. Peptides #1, A6, B3, Bl, #5, #6 and #8 are shown in order from top to bottom. After peptides were captured at a concentration of 7ug/ml on streptavidin sensors, kinetic binding of lOOnM of mouse IgGi kappa antibody was evaluated (association and dissociation).
  • BLI bilayer interferometry
  • Figure 12 shows the IHC staining with 16B9 and its variants in human heart tissue and lung cancer tissues.
  • Human lung cancer and left ventricle FFPE slides were IHC stained using 5 ug/ml 16B9 or 16B9 V2, V3, V4 and V5 for 30 minutes at room temperature. Nuclear counterstain was carried out with haematoxylin. Images were acquired using 40x objective.
  • Figure 13 shows the IHC staining with 16B9 and its variants in human lung cancer tissues with different antigen retrieval methods.
  • Human lung cancer FFPE slides were subjected to different antigen retrieval and then IHC stained using 15 ug/ml 16B9 and its variants overnight at 4°C. Nuclear counterstain was carried out with haematoxylin. Images were acquired using 40x objective.
  • Figures 14A-14C show the staining and scoring of human cancer samples based on LILRB2, CD3 and CD163 IHC.
  • CD163, CD3 and LILRB2 were stained with respective IHC antibodies in 195 evaluable samples from 19 different human cancer indications.
  • Figure 14A shows the number of samples in each LILRB2 IHC reactivity score (0-5) in macrophages across solid tumor types.
  • Figure 14B shows the average score and correlation between LILRB2, CD 163 and CD3 IHC staining across tumor types.
  • Figure 14C shows the ranking of tumor types stained by IHC based on reactivity scores. Each tissue was evaluated by CRO’s board-certified pathologist separately for LILRB2, CD163, and CD3 reactivity.
  • TIS tumor and tumor-induced stroma
  • antibody includes any immunoglobulin, monoclonal antibody, polyclonal antibody, multivalent antibody, bivalent antibody, monovalent antibody, multi-specific antibody, or bispecific antibody that binds to a specific antigen.
  • a native intact antibody comprises two heavy (H) chains and two light (L) chains.
  • Mammalian heavy chains are classified as alpha, delta, epsilon, gamma, and mu, each heavy chain consists of a variable domain (VH) and a constant region including a first, second, and third constant domain (CHI, CH2, CH3, respectively); mammalian light chains are classified as X or K, while each light chain consists of a variable domain (V L ) and a constant domain (CL).
  • a typical IgG antibody has a “Y” shape, with the stem of the Y typically consisting of the second and third constant domains of two heavy chains bound together via disulfide bonding. Each arm of the Y includes the variable domain and first constant domain of a single heavy chain bound to the variable and constant domains of a single light chain.
  • variable domains of the light and heavy chains are responsible for antigen binding.
  • the variable domains in both chains generally contain three highly variable loops called the complementarity determining regions (CDRs) (light chain CDRs including LCDR1, LCDR2, and LCDR3, heavy chain CDRs including HCDR1, HCDR2, HCDR3).
  • CDR boundaries for the antibodies and antigenbinding fragments disclosed herein may be defined or identified by the conventions of Kabat, IMGT, Chothia, or Al-Lazikani (Al-Lazikani, B., Chothia, C., Lesk, A. M., J. Mol. Biol., 273(4), 927 (1997); Chothia, C. et al., J Mol Biol.
  • the three CDRs are interposed between flanking stretches known as framework regions (FRs), which are more highly conserved than the CDRs and form a scaffold to support the hypervariable loops.
  • FRs framework regions
  • the constant domains of the heavy and light chains are not involved in antigen-binding but exhibit various effector functions.
  • Antibodies are assigned to classes based on the amino acid sequence of the constant region of their heavy chain.
  • the five major classes or isotypes of antibodies are IgA, IgD, IgE, IgG, and IgM, which are characterized by the presence of alpha, delta, epsilon, gamma, and mu heavy chains, respectively.
  • IgGl gammal heavy chain
  • IgG2 gamma2 heavy chain
  • IgG3 gamma3 heavy chain
  • IgG4 gamma4 heavy chain
  • IgAl alpha 1 heavy chain
  • IgA2 alpha2 heavy chain
  • Suitable antigens include without limitation parts of bacteria (coats, capsules, cell walls, flagella, fimbrai, and toxins), viruses, and other microorganisms.
  • Antigens also include tumor antigens, e.g., antigens generated by mutations in tumors.
  • antigens also include immunogens and haptens.
  • antigen-binding fragment refers to an antibody fragment formed from a portion of an antibody comprising one or more CDRs, or any other antibody fragment that binds to an antigen but does not comprise an intact native antibody structure.
  • antigen-binding fragment examples include, without limitation, a diabody, a Fab, a Fab’, a F(ab’) 2 , an Fv fragment, a disulfide stabilized Fv fragment (dsFv), a (dsFv) 2 , a bispecific dsFv (dsFv-dsFv’), a disulfide stabilized diabody (ds diabody), a single-chain antibody molecule (scFv), an scFv dimer (bivalent diabody), a bispecific antibody, a multispecific antibody, a camelized single domain antibody, a nanobody, a domain antibody, and a bivalent domain antibody.
  • An antigen-binding fragment is capable of binding to the same antigen to which the parent antibody binds.
  • a “Fab fragment” comprises one light chain and the CHI and variable domains of one heavy chain.
  • the heavy chain of a Fab molecule cannot form a disulfide bond with another heavy chain molecule.
  • a “Fab' fragment” comprises one light chain and a portion of one heavy chain that contains the VH domain and the CHI domain and also the region between the CHI and C H 2 domains, such that an interchain disulfide bond can be formed between the two heavy chains of two Fab' fragments to form an F(ab') 2 molecule.
  • a “F(ab') 2 fragment” contains two light chains and two heavy chains containing a portion of the constant region between the CHI and CH2 domains, such that an interchain disulfide bond is formed between the two heavy chains.
  • a F(ab') 2 fragment thus is composed of two Fab' fragments that are held together by a disulfide bond between the two heavy chains.
  • Fv with regard to an antibody refers to the smallest fragment of the antibody to bear the complete antigen-binding site.
  • An Fv fragment consists of the variable domain of a single light chain bound to the variable domain of a single heavy chain.
  • Single-chain Fv antibody or “scFv” refers to an engineered antibody consisting of a light chain variable domain and a heavy chain variable domain connected to one another directly or via a peptide linker sequence (Huston JS et al., Proc Natl Acad Sci USA (1988) 85:5879).
  • An “Fc” region comprises two heavy chain fragments comprising the CH2 and CH3 domains of an antibody.
  • the two heavy chain fragments are held together by two or more disulfide bonds and by hydrophobic interactions of the C H 3 domains.
  • the Fc region of the antibody is responsible for various effector functions such as antibody-dependent cell- mediated cytotoxicity (ADCC), and complement dependent cytotoxicity (CDC), but does not function in antigen binding.
  • ADCC antibody-dependent cell- mediated cytotoxicity
  • CDC complement dependent cytotoxicity
  • Single-chain Fv-Fc antibody or “scFv-Fc” refers to an engineered antibody consisting of a scFv connected to the Fc region of an antibody.
  • a “dsFv” refers to a disulfide-stabilized Fv fragment that the linkage between the variable domain of a single light chain and the variable domain of a single heavy chain is a disulfide bond.
  • a “(dsFv) 2 ” or “(dsFv-dsFv’)” comprises three peptide chains: two V H domains linked by a peptide linker (e.g., a long flexible linker) and bound to two VL domains, respectively, via disulfide bridges.
  • dsFv- dsFv’ is bispecific in which each disulfide paired heavy and light chain has a different antigen specificity.
  • Camelized single domain antibody refers to an antibody that contains two VH domains and no light chains (Riechmann L. and Muyldermans S., J Immunol Methods. Dec 10;231 (1 -2):25-38 (1999); Muyldermans S., J Biotechnol. Jun;74(4):277-302 (2001); WO94/04678; WO94/25591; U.S. Patent No. 6,005,079).
  • Heavy chain antibodies were originally derived from Camelidae (camels, dromedaries, and llamas). Although devoid of light chains, camelized antibodies have an authentic antigen-binding repertoire (Hamers-Casterman C.
  • variable domain of a heavy chain antibody represents the smallest known antigen-binding unit generated by adaptive immune responses (Koch-Nolte F. et al., FASEB J. (2007) 21 :3490-8).
  • a “nanobody” refers to an antibody fragment that consists of a VHH domain from a heavy chain antibody and two constant domains, CH2 and CH3.
  • “Diabodies” or “dAbs” include small antibody fragments with two antigenbinding sites, wherein the fragments comprise a VH domain connected to a VL domain in the same polypeptide chain (V H -V L or V L -V H ) (see, e.g., Holliger P. et al., Proc Natl Acad Sci U S A. Jul 15;90(14):6444-8 (1993); EP404097; WO93/11161).
  • a “bispecific ds diabody” is a diabody target two different antigens (or epitopes).
  • an “scFv dimer” is divalent (or bivalent) single-chain variable fragments (di-scFvs, bi-scFvs) that can be engineered by linking two scFvs.
  • an “scFv dimer” is a bispecific diabody comprising V H i-V L 2 (linked by a peptide linker) associated with VLI-V H2 (also linked by a peptide linker) such that VHI and VLI coordinate and VH2 and VL2 coordinate and each coordinated pair has a different antigen specificity.
  • a “domain antibody” refers to an antibody fragment containing only the variable domain of a heavy chain or the variable domain of a light chain.
  • two or more VH domains are covalently joined with a peptide linker to create a bivalent or multivalent domain antibody.
  • the two VH domains of a bivalent domain antibody may target the same or different antigens.
  • a “bispecific” antibody refers to an artificial antibody which has fragments derived from two different monoclonal antibodies and is capable of binding to two different epitopes.
  • the two epitopes may present on the same antigen, or they may present on two different antigens.
  • Cancer refers to any medical condition characterized by malignant cell growth or neoplasm, abnormal proliferation, infiltration or metastasis, and includes both solid tumors and non-solid cancers (hematologic malignancies) such as leukemia.
  • solid tumor refers to a solid mass of neoplastic and/or malignant cells.
  • cancer or tumors include hematological malignancies, oral carcinomas (for example of the lip, tongue or pharynx), digestive organs (for example esophagus, stomach, small intestine, colon, large intestine, or rectum), peritoneum, liver and biliary passages, pancreas, respiratory system such as larynx or lung (small cell and non-small cell), bone, connective tissue, skin (e.g., melanoma), breast, reproductive organs (fallopian tube, uterus, cervix, testicles, ovary, or prostate), urinary tract (e.g., bladder or kidney), brain and endocrine glands such as the thyroid.
  • oral carcinomas for example of the lip, tongue or pharynx
  • digestive organs for example esophagus, stomach, small intestine, colon, large intestine, or rectum
  • peritoneum liver and biliary passages
  • pancreas respiratory system
  • respiratory system such
  • the cancer is selected from ovarian cancer, breast cancer, head and neck cancer, renal cancer, bladder cancer, hepatocellular cancer, and colorectal cancer. In certain embodiments, the cancer is selected from a lymphoma, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma and B-cell lymphoma.
  • chimeric means an antibody or antigen-binding fragment, having a portion of heavy and/or light chain derived from one species, and the rest of the heavy and/or light chain derived from a different species.
  • a chimeric antibody may comprise a constant region derived from human and a variable region from a non-human animal, such as from mouse or rabbit.
  • the nonhuman animal is a mammal, for example, a mouse, a rat, a rabbit, a goat, a sheep, a guinea pig, a llama, a horse, a donkey or a hamster.
  • the term “specific binding” or “specifically binds” as used herein refers to a non-random binding reaction between two molecules, such as for example between an antibody and an antigen.
  • the antibodies or antigen-binding fragments provided herein specifically bind to human LILRB2 with a binding affinity (K D ) of ⁇ 10' 6 M (e.g., ⁇ 5xl0' 7 M, ⁇ 2xl0' 7 M, ⁇ 10' 7 M, ⁇ 5xl0' 8 M, ⁇ 2xl0' 8 M, ⁇ 10' 8 M, ⁇ 5xl0' 9 M, ⁇ 4xlO' 9 M, ⁇ 3X10' 9 M, ⁇ 2X10' 9 M, or ⁇ 10' 9 M).
  • K D binding affinity
  • KD used herein refers to the ratio of the dissociation rate to the association rate (k o ff/k on ), which may be determined by using any conventional method known in the art, including, but not limited to, BLI, surface plasmon resonance method, microscale thermophoresis method, HPLC-MS method and flow cytometry (such as FACS) method.
  • the K D value can be appropriately determined by using flow cytometry.
  • LILRB2 Leukocyte immunoglobulin-like receptor subfamily B member 2
  • LIR leukocyte immunoglobulin-like receptor
  • the encoded protein belongs to the subfamily B class of LIR receptors which contain two or four extracellular immunoglobulin domains, a transmembrane domain, and two to four cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIM).
  • the receptor is expressed on myeloid cells, binds to MHC class I (MHC-I) and other ligands and transduces a negative signal that inhibits stimulation of an immune response.
  • MHC-I MHC class I
  • the receptor can also compete with CD8 to the binding of MHC-I. It is thought to negatively regulate immune inflammatory responses and cytotoxicity to help focus the immune response and limit autoimmunity. Multiple LILRB2 isoform and polymorphic variants have been identified.
  • LILRB2 has been shown to interact with classical and non-classical MHC-I (most notably, human leukocyte antigen G (HLA-G)), angiopoietin-like protein (ANGPTL) 2 and 5, Semaphorin-4A (SEMA4A), complement split products (CSPs), CDlc/d, myelin-associated glycoprotein (MAG), oligodendrocyte myelin glycoprotein (Omgp), P-amyloid protein and RIFIN.
  • HLA-G human leukocyte antigen G
  • ANGPTL angiopoietin-like protein
  • SEMA4A Semaphorin-4A
  • CSPs complement split products
  • CDlc/d CDlc/d
  • MAG myelin-associated glycoprotein
  • Omgp oligodendrocyte myelin glycoprotein
  • P-amyloid protein P-amyloid protein and RIFIN.
  • anti-LILRB2 antibody refers to an antibody that is capable of specifically binding to LILRB2.
  • LILRB2-related disease or condition refers to any disease or condition caused by, exacerbated by, or otherwise linked to increased or decreased expression or activities of LILRB2.
  • the LILRB2 related condition is immune-related disorder, such as, for example, cancer, autoimmune disease, inflammatory disease or infectious disease.
  • a “conservative substitution” with reference to amino acid sequence refers to replacing an amino acid residue with a different amino acid residue having a side chain with similar physiochemical properties.
  • conservative substitutions can be made among amino acid residues with hydrophobic side chains (e.g., Met, Ala, Vai, Leu, and He), among residues with neutral hydrophilic side chains (e.g., Cys, Ser, Thr, Asn and Gin), among residues with acidic side chains (e.g., Asp, Glu), among amino acids with basic side chains (e.g., His, Lys, and Arg), or among residues with aromatic side chains (e.g., Trp, Tyr, and Phe).
  • conservative substitution usually does not cause significant change in the protein conformational structure, and therefore could retain the biological activity of a protein.
  • epitope refers to the specific group of atoms or amino acids on an antigen to which an antibody binds.
  • Two antibodies may bind the same or a closely related epitope within an antigen if they exhibit competitive binding for the antigen. For example, if an antibody or antigen-binding fragment blocks binding of a reference antibody to the antigen by at least 85%, or at least 90%, or at least 95%, then the antibody or antigen-binding fragment may be considered to bind the same/closely related epitope as the reference antibody.
  • homologue and “homologous” as used herein are interchangeable and refer to nucleic acid sequences (or its complementary strand) or amino acid sequences that have sequence identity of at least 80% (e.g., at least 85%, 88%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%) to another sequence when optimally aligned.
  • host cell refers to a cell into which an exogenous polynucleotide and/or a vector has been introduced.
  • an “isolated” substance has been altered by the hand of man from the natural state. If an “isolated” composition or substance occurs in nature, it has been changed or removed from its original environment, or both.
  • a polynucleotide or a polypeptide naturally present in a living animal is not “isolated,” but the same polynucleotide or polypeptide is “isolated” if it has been sufficiently separated from the coexisting materials of its natural state so as to exist in a substantially pure state.
  • An “isolated nucleic acid sequence” refers to the sequence of an isolated nucleic acid molecule.
  • an “isolated antibody or antigen-binding fragment thereof’ refers to the antibody or antigenbinding fragments having a purity of at least 60%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% as determined by electrophoretic methods (such as SDS-PAGE, isoelectric focusing, capillary electrophoresis), or chromatographic methods (such as ion exchange chromatography or reverse phase HPLC).
  • electrophoretic methods such as SDS-PAGE, isoelectric focusing, capillary electrophoresis
  • chromatographic methods such as ion exchange chromatography or reverse phase HPLC.
  • leader peptide refers to a peptide having a length of about 5-30 amino acids that is present at the N-terminus of newly synthesized proteins that form part of the secretory pathway. Proteins of the secretory pathway include, but are not limited to proteins that reside either inside certain organelles (the endoplasmic reticulum, Golgi or endosomes), are secreted from the cell, or are inserted into a cellular membrane. In some embodiments, the leader peptide forms part of the transmembrane domain of a protein.
  • link refers to the association via intramolecular interaction, e.g., covalent bonds, metallic bonds, and/or ionic bonding, or inter-molecular interaction, e.g., hydrogen bond or noncovalent bonds.
  • operably linked refers to an arrangement of elements wherein the components so described are configured so as to perform their usual function.
  • a given signal peptide that is operably linked to a polypeptide directs the secretion of the polypeptide from a cell.
  • a promoter that is operably linked to a coding sequence will direct the expression of the coding sequence.
  • the promoter or other control elements need not be contiguous with the coding sequence, so long as they function to direct the expression thereof. For example, intervening untranslated yet transcribed sequences can be present between the promoter sequence and the coding sequence and the promoter sequence can still be considered “operably linked” to the coding sequence.
  • Percent (%) sequence identity with respect to amino acid sequence (or nucleic acid sequence) is defined as the percentage of amino acid (or nucleic acid) residues in a candidate sequence that are identical to the amino acid (or nucleic acid) residues in a reference sequence, after aligning the sequences and, if necessary, introducing gaps, to achieve the maximum number of identical amino acids (or nucleic acids). Conservative substitution of the amino acid residues may or may not be considered as identical residues. Alignment for purposes of determining percent amino acid (or nucleic acid) sequence identity can be achieved, for example, using publicly available tools such as BLASTN, BLASTp (available on the website of U.S. National Center for Biotechnology Information (NCBI), see also, Altschul S.F.
  • polynucleotide or “nucleic acid” includes both single-stranded and double-stranded nucleotide polymers.
  • the nucleotides comprising the polynucleotide can be ribonucleotides or deoxyribonucleotides or a modified form of either type of nucleotide.
  • Said modifications include base modifications such as bromouridine and inosine derivatives, ribose modifications such as 2', 3 '-dideoxyribose, and internucleotide linkage modifications such as phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phoshoraniladate and phosphoroamidate.
  • polypeptide or “protein” means a string of at least two amino acids linked to one another by peptide bonds. Polypeptides and proteins may include moieties in addition to amino acids (e.g., may be glycosylated) and/or may be otherwise processed or modified. Those of ordinary skill in the art will appreciate that a “polypeptide” or “protein” can be a complete polypeptide chain as produced by a cell (with or without a signal sequence), or can be a functional portion thereof. Those of ordinary skill will further appreciate that a polypeptide or protein can sometimes include more than one polypeptide chain, for example linked by one or more disulfide bonds or associated by other means. The term also includes amino acid polymers in which one or more amino acids are chemical analogs of a corresponding naturally-occurring amino acid and polymers.
  • sample refers to any sample that is taken from a subject (e.g., a human, such as a person with a disease of interest, or a person suspected of having such disease) and contains one or more molecule(s) of interest.
  • the biological sample includes but not limited to cells (e.g., bacteria, yeast, virus, plant cells, animal cells and the like), tissues (e.g., biopsy tissue, paraffin embedded tissue and the like), and body fluids (e.g., blood, plasma, serum, saliva, pleural effusion, ascites, amniocentesis fluid, seroperitoneum and the like).
  • the term “subject” refers to a human or any non-human animal (e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate).
  • a human includes pre- and post-natal forms.
  • a subject is a human being.
  • a subject can be a patient, which refers to a human presenting to a medical provider for diagnosis or treatment of a disease.
  • the term “subject” is used herein interchangeably with “individual” or “patient.”
  • a subject can be afflicted with or is susceptible to a disease or disorder but may or may not display symptoms of the disease or disorder.
  • vector refers to a vehicle into which a polynucleotide encoding a protein may be operably inserted so as to bring about the expression of that protein.
  • a vector may be used to transform, transduce, or transfect a host cell so as to bring about expression of the genetic element it carries within the host cell.
  • vectors include plasmids, phagemids, cosmids, artificial chromosomes such as yeast artificial chromosome (YAC), bacterial artificial chromosome (BAC), or Pl -derived artificial chromosome (PAC), bacteriophages such as lambda phage or Ml 3 phage, and animal viruses.
  • a vector may contain a variety of elements for controlling expression, including promoter sequences, transcription initiation sequences, enhancer sequences, selectable elements, and reporter genes. In addition, the vector may contain an origin of replication.
  • a vector may also include materials to aid in its entry into the cell, including but not limited to a viral particle, a liposome, or a protein coating.
  • a vector can be an expression vector or a cloning vector.
  • the present disclosure provides vectors (e.g., expression vectors) containing the nucleic acid sequence provided herein encoding the antibody or antigen-binding fragment thereof, at least one promoter (e.g., SV40, CMV, EF-la) operably linked to the nucleic acid sequence, and at least one selection marker.
  • promoter e.g., SV40, CMV, EF-la
  • vectors include, but are not limited to, retrovirus (including lentivirus), adenovirus, adeno-associated virus, herpesvirus (e.g., herpes simplex virus), poxvirus, baculovirus, papillomavirus, papovavirus (e.g., SV40), lambda phage, and M13 phage, plasmid pcDNA3.3, pMD18-T, pOptivec, pCMV, pEGFP, pIRES, pQD-Hyg-Gseu, pALTER, pBAD, pcDNA, pCal, pL, pET, pGEMEX, pGEX, pCI, pEGFT, pSV2, pFUSE, pVITRO, pVIVO, pMAL, pMONO, pSELECT, pUNO, pDUO, Psg5L, pBABE, pW
  • the present disclosure in one aspect provides an anti-LILRB2 antibody and antigen-binding fragment thereof that has a high sensitivity and specificity to LILRB2.
  • Binding affinity of the antibody and antigen-binding fragment provided herein can be represented by K D value, which represents the ratio of dissociation rate to association rate (k of f/k on ) when the binding between the antigen and antigen-binding molecule reaches equilibrium.
  • the antigen-binding affinity e.g., K D
  • K D can be appropriately determined using suitable methods known in the art, including, for example, bio-layer interferometry (BLI).
  • Binding of the antibodies to LILRB2 can also be represented by “half maximal effective concentration” (EC 50 ) value, which refers to the concentration of an antibody where 50% of its maximal effect (e.g., binding or inhibition etc.) is observed.
  • the EC50 value can be measured by methods known in the art, for example, sandwich assay such as ELISA, flow cytometry assay, and other quantitative binding assays.
  • the present disclosure in one aspect provides an anti-LILRB2 antibody and antigen-binding fragment thereof comprising one or more (e.g., 1, 2, 3, 4, 5, or 6) CDR sequences of an anti-LILRB2 antibody disclosed herein.
  • CDRs are known to be responsible for antigen binding, however, it has been found that not all of the 6 CDRs are indispensable or unchangeable. In other words, it is possible to replace or change or modify one or more CDRs in anti-LILRB2 antibody disclosed herein, yet substantially retain the specific binding affinity to LILRB2.
  • the anti-LILRB2 antibody has CDR sequences as listed in Table 1 below.
  • the anti-LILRB2 antibodies and antigen-binding fragments thereof provided herein comprise H-CDR sequences derived from clone 16B9, which have the formula as shown below:
  • HC-CDR1 GX 1 SIX 2 SX 3 YAX 4 X 5 (SEQ ID NO: 75), wherein Xi is Y, I or H, X 2 is T or A, X 3 is D or G, X 4 is W or R, X 5 is N, S or T;
  • HC-CDR2 X
  • HC-CDR3 X!WX 2 GX 3 (SEQ ID NO: 77), wherein X 4 is S or P, X 2 is F or L, X 3 is R, T, S, H, K, N, Q, or P.
  • the anti-LILRB2 antibodies and antigen-binding fragments thereof provided herein comprise suitable framework region (FR) sequences, as long as the antibodies and antigen-binding fragments thereof can specifically bind to LILRB2.
  • the CDR sequences provided in Table 1 can be grafted to any suitable FR sequences of any suitable species such as mouse, human, rat, rabbit, sheep, llama, horse, donkey, guinea pig, hamster, among others, using suitable methods known in the art such as recombinant techniques.
  • the anti-LILRB2 antibodies and antigen-binding fragments thereof provided herein comprise heavy chain and light chain variable region amino acid sequences as provided in Table 2 below.
  • the anti-LILRB2 antibodies and the antigen-binding fragments provided herein comprise all or a portion of the heavy chain variable domain and/or all or a portion of the light chain variable domain.
  • the anti- LILRB2 antibodies and the antigen-binding fragments provided herein is a single domain antibody which consists of all or a portion of the heavy chain variable domain provided herein. More information of such a single domain antibody is available in the art (see, e.g., U.S. Pat. No. 6,248,516).
  • the anti-LILRB2 antibodies and the fragments thereof provided herein further comprise an immunoglobulin constant region.
  • an immunoglobulin constant region comprises a heavy chain and/or a light chain constant region.
  • the heavy chain constant region comprises CHI, hinge, and/or CH2-CH3 regions.
  • the heavy chain constant region comprises an Fc region.
  • the light chain constant region comprises CK or Ck.
  • the antibodies or antigen-binding fragments thereof provided herein can be a monoclonal antibody, polyclonal antibody, humanized antibody, chimeric antibody, recombinant antibody, bispecific antibody, labeled antibody, bivalent antibody, or anti- idiotypic antibody.
  • a recombinant antibody is an antibody prepared in vitro using recombinant methods rather than in animals.
  • anti-LILRB2 antigen-binding fragments are also provided herein.
  • Various types of antigen-binding fragments are known in the art and can be developed based on the anti-LILRB2 antibodies provided herein, including for example, the exemplary antibodies whose CDR and variable sequences are provided herein, and their different variants (such as affinity variants, glycosylation variants, Fc variants, cysteine-engineered variants and so on).
  • an anti-LILRB2 antigen-binding fragment provided herein is a camelized single domain antibody, a diabody, a single chain Fv fragment (scFv), an scFv dimer, a BsFv, a dsFv, a (dsFv)2, a dsFv-dsFv’, an Fv fragment, a Fab, a Fab’, a F(ab’)2, a bispecific antibody, a ds diabody, a nanobody, a domain antibody, a single domain antibody, or a bivalent domain antibody.
  • scFv single chain Fv fragment
  • scFv dimer a BsFv, a dsFv, a (dsFv)2, a dsFv-dsFv’, an Fv fragment, a Fab, a Fab’, a F(ab’)2, a bispecific antibody, a ds diabody
  • a Single Chain Variable Fragment is a fusion of the variable regions of the heavy and light chains of immunoglobulins, linked together with a short (usually serine, glycine) linker.
  • This chimeric molecule retains the specificity of the original immunoglobulin, despite removal of the constant regions and the introduction of a linker peptide. This modification usually leaves the specificity unaltered.
  • These molecules were created historically to facilitate phage display where it is highly convenient to express the antigen binding domain as a single peptide.
  • scFv can be created directly from subcloned heavy and light chains derived from a hybridoma.
  • Single chain variable fragments lack the constant Fc region found in complete antibody molecules, and thus, the common binding sites (e.g., protein A/G) used to purify antibodies. These fragments can often be purified/immobilized using Protein L since Protein L interacts with the variable region of kappa light chains.
  • Flexible linkers generally are comprised of helix- and turn-promoting amino acid residues such as alanine, serine and glycine. However, other residues can function as well.
  • Tang et al. (1996) used phage display as a means of rapidly selecting tailored linkers for single-chain antibodies (scFvs) from protein linker libraries.
  • scFvs single-chain antibodies
  • a random linker library was constructed in which the genes for the heavy and light chain variable domains were linked by a segment encoding an 18-amino acid polypeptide of variable composition.
  • the scFv repertoire (21 itteri. 5 x io 6 different members) was displayed on filamentous phage and subjected to affinity selection with hapten.
  • the recombinant antibodies of the present disclosure may also involve sequences or moieties that permit dimerization or multimerization of the receptors.
  • sequences include those derived from IgA, which permit formation of multimers in conjunction with the J-chain.
  • Another multimerization domain is the Gal4 dimerization domain.
  • the chains may be modified with agents such as biotin/avidin, which permit the combination of two antibodies.
  • a single-chain antibody can be created by joining receptor light and heavy chains using a non-peptide linker or chemical unit.
  • the light and heavy chains will be produced in distinct cells, purified, and subsequently linked together in an appropriate fashion (i.e., the N-terminus of the heavy chain being attached to the C-terminus of the light chain via an appropriate chemical bridge).
  • Cross-linking reagents are used to form molecular bridges that tie functional groups of two different molecules, e.g., a stabilizing and coagulating agent.
  • a stabilizing and coagulating agent e.g., a stabilizing and coagulating agent.
  • dimers or multimers of the same analog or heteromeric complexes comprised of different analogs can be created.
  • hetero-bifunctional cross-linkers can be used that eliminate unwanted homopolymer formation.
  • An exemplary hetero-bifunctional cross-linker contains two reactive groups: one reacting with primary amine group (e.g., Nhydroxy succinimide) and the other reacting with a thiol group (e.g., pyridyl disulfide, maleimides, halogens, etc.).
  • primary amine group e.g., Nhydroxy succinimide
  • a thiol group e.g., pyridyl disulfide, maleimides, halogens, etc.
  • the cross-linker may react with the lysine residue(s) of one protein (e.g., the selected antibody or fragment) and through the thiol reactive group, the cross-linker, already tied up to the first protein, reacts with the cysteine residue (free sulfhydryl group) of the other protein (e.g., the selective agent).
  • SMPT is a bifunctional cross-linker containing a disulfide bond that is “sterically hindered” by an adjacent benzene ring and methyl groups.
  • steric hindrance of the disulfide bond serves a function of protecting the bond from attack by thiolate anions such as glutathione which can be present in tissues and blood, and thereby help in preventing decoupling of the conjugate prior to the delivery of the attached agent to the target site.
  • the SMPT cross-linking reagent lends the ability to cross-link functional groups such as the SH of cysteine or primary amines (e.g., the epsilon amino group of lysine).
  • Another possible type of crosslinker includes the hetero-bifunctional photoreactive phenylazides containing a cleavable disulfide bond such as sulfosuccinimidyl-2-(p-azido salicylamido) ethyl- 1,3’- dithiopropionate.
  • the N-hydroxy-succinimidyl group reacts with primary amino groups and the phenylazide (upon photolysis) reacts non-selectively with any amino acid residue.
  • non-hindered linkers also can be employed in accordance herewith.
  • Other useful cross-linkers include SATA, SPDP and 2-iminothiolane (Wawrzynczak & Thorpe, 1987). The use of such cross-linkers is well understood in the art. Another embodiment involves the use of flexible linkers.
  • U.S. Patent 4,680,338 describes bifunctional linkers useful for producing conjugates of ligands with amine-containing polymers and/or proteins, especially for forming antibody conjugates with chelators, drugs, enzymes, detectable labels and the like.
  • U.S. Patents 5,141,648 and 5,563,250 disclose cleavable conjugates containing a labile bond that is cleavable under a variety of mild conditions. This linker is particularly useful in that the agent of interest may be bonded directly to the linker, with cleavage resulting in release of the active agent. Particular uses include adding a free amino or free sulfhydryl group to a protein, such as an antibody, or a drug.
  • U.S. Patent 5,856,456 provides peptide linkers for use in connecting polypeptide constituents to make fusion proteins, e.g., single chain antibodies.
  • the linker is up to about 50 amino acids in length, contains at least one occurrence of a charged amino acid (preferably arginine or lysine) followed by a proline, and is characterized by greater stability and reduced aggregation.
  • U.S. Patent 5,880,270 discloses aminooxy-containing linkers useful in a variety of immunodiagnostic and separative techniques.
  • Various techniques can be used for the production of such antigen-binding fragments.
  • Illustrative methods include, enzymatic digestion of intact antibodies (see, e.g., Morimoto et al., Journal of Biochemical and Biophysical Methods (1992) 24: 107-117; and Brennan et al., Science (1985) 229:81), recombinant expression by host cells such as E. Coli (e.g. for Fab, Fv and ScFv antibody fragments), screening from a phase display library as discussed above (e.g. for ScFv), and chemical coupling of two Fab’-SH fragments to form F(ab’) 2 fragments (Carter et al., Bio/Technology (1992) 10: 163-167). Other techniques for the production of antibody fragments will be apparent to a skilled practitioner.
  • the antigen-binding fragment is a scFv.
  • Generation of scFv is described in, for example, WO 93/16185; U.S. Pat. Nos. 5,571,894; and 5,587,458.
  • scFv may be fused to an effector protein at either the amino or the carboxyl terminus to provide for a fusion protein (see, for example, Antibody Engineering, ed. Borrebaeck).
  • the anti-LILRB2 antibodies and antigen-binding fragments thereof further comprise a conjugate moiety.
  • the conjugate moiety can be linked to the antibodies and antigen-binding fragments thereof.
  • a conjugate moiety is a proteinaceous or non-proteinaceous moiety that can be attached to the antibody or antigenbinding fragment thereof. It is contemplated that a variety of conjugate moieties may be linked to the antibodies or antigen-binding fragments provided herein (see, for example, “Conjugate Vaccines”, Contributions to Microbiology and Immunology, J. M. Cruse and R. E. Lewis, Jr. (eds.), Carger Press, New York, (1989)). These conjugate moieties may be linked to the antibodies or antigen-binding fragments by covalent binding, affinity binding, intercalation, coordinate binding, complexation, association, blending, or addition, among other methods.
  • the antibodies and antigen-binding fragments disclosed herein may be engineered to contain specific sites outside the epitope binding portion that may be utilized for binding to one or more conjugate moieties.
  • a site may include one or more reactive amino acid residues, such as for example cysteine or histidine residues, to facilitate covalent linkage to a conjugate moiety.
  • the antibodies may be linked to a conjugate moiety indirectly, or through another conjugate moiety.
  • the antibody or antigen-binding fragments may be conjugated to biotin, then indirectly conjugated to a second conjugate that is conjugated to avidin.
  • the conjugate can be a detectable label (e.g., a radioactive isotope, a lanthanide, a luminescent label, a fluorescent label, or an enzyme-substrate label), a clearance-modifying agent, or purification moiety.
  • detectable label may include a fluorescent labels (e.g. fluorescein, rhodamine, dansyl, phycoerythrin, or Texas Red), enzyme-substrate labels (e.g. horseradish peroxidase, alkaline phosphatase, luciferases, glucoamylase, lysozyme, saccharide oxidases or p-D-galactosidase), radioisotopes (e.g.
  • the conjugate moiety can be a clearance-modifying agent which helps increase half-life of the antibody.
  • Illustrative examples include water- soluble polymers, such as PEG, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, copolymers of ethylene glycol/propylene glycol, and the like.
  • the polymer may be of any molecular weight and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules.
  • the conjugate moiety can be a purification moiety such as a magnetic bead.
  • the antibodies and antigen-binding fragments thereof provided herein is used for a base for a conjugate.
  • the present disclosure provides isolated polynucleotides that encode the anti- LILRB2 antibodies and antigen-binding fragments thereof disclosed herein.
  • the isolated polynucleotides comprise one or more nucleotide sequences listed in Table 1 that encodes the variable region of the exemplary antibodies provided herein.
  • DNA encoding the monoclonal antibody is 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 antibody).
  • the encoding DNA may also be obtained by synthetic methods.
  • the isolated polynucleotide that encodes the anti-LILRB2 antibodies and antigen-binding fragments can be inserted into a vector for further cloning (amplification of the DNA) or for expression, using recombinant techniques known in the art.
  • Many vectors are available.
  • the vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter (e.g., SV40, CMV, EF-la), and a transcription termination sequence.
  • the present disclosure provides vectors (e.g., expression vectors) containing the nucleic acid sequence provided herein encoding the antibodies or antigen-binding fragments, at least one promoter (e.g., SV40, CMV, EF-la) operably linked to the nucleic acid sequence, and at least one selection marker.
  • promoter e.g., SV40, CMV, EF-la
  • vectors include, but are not limited to, retrovirus (including lentivirus), adenovirus, adeno-associated virus, herpesvirus (e.g., herpes simplex virus), poxvirus, baculovirus, papillomavirus, papovavirus (e.g., SV40), lambda phage, and M13 phage, plasmid pcDNA3.3, pMD18-T, pOptivec, pCMV, pEGFP, pIRES, pQD-Hyg-Gseu, pALTER, pBAD, pcDNA, pCal, pL, pET, pGEMEX, pGEX, pCI, pEGFT, pSV2, pFUSE, pVITRO, pVIVO, pMAL, pMONO, pSELECT, pUNO, pDUO, Psg5L, pBABE, pW
  • Vectors comprising the polynucleotide sequence encoding the antibody or antigen-binding fragment can be introduced to a host cell for cloning or gene expression.
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above.
  • Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as Escherichia, e.g., E.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for anti-LILRB2 antibody-encoding vectors.
  • Saccharomyces cerevisiae, or common baker’s yeast is the most commonly used among lower eukaryotic host microorganisms.
  • Schizosaccharomyces pombe ' Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K.
  • waltii ATCC 56,500
  • K. drosophilarum ATCC 36,906
  • K. thermotolerans K. marxianus
  • Pichia pastoris EP 183,070
  • Candida Trichoderma reesia
  • Neurospora crassa Schwanniomyces such as Schwanniomyces occidentalism
  • filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated antibodies or antigenfragment provided here are derived from multicellular organisms.
  • invertebrate cells include plant and insect cells.
  • Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruit fly), and Bombyx mori have been identified.
  • a variety of viral strains for transfection are publicly available, e.g., the L-l variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures of cotton, com, potato, soybean, petunia, tomato, and tobacco can also be utilized as hosts.
  • vertebrate cells have been greatest in vertebrate cells, and propagation of vertebrate cells in culture (tissue culture) has become a routine procedure.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. (1977) 36:59); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese Hamster Ovary cells (CHO), CHO cells deficient in dihydrofolate reductase (DHFR) activity, CHO-DHFR (Urlaub et al., Proc. Natl. Acad. Sci.
  • DHFR dihydrofolate reductase
  • mice 27itteri cells (TM4, Mather, Biol. Reprod. (1980) 23:243-251); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. (1982) 383:44-68); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2).
  • the host cell is 293F cell.
  • Host cells are transfected with the above-described expression or cloning vectors for anti-LILRB2 antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the antibody may be produced by homologous recombination known in the art.
  • the host cells used to produce the antibodies or antigen-binding fragments provided herein may be cultured in a variety of media.
  • Commercially available media such as Ham’s F10 (Sigma), Minimal Essential Medium (MEM) (Sigma), RPMI-1640 (Sigma), and Dulbecco’s Modified Eagle’s Medium (DMEM), Sigma) are suitable for culturing the host cells.
  • any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as GENTAMYCINTM drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art.
  • the culture conditions such as temperature, pH, and the like, are those previously used with the host cell selected for expression and will be apparent to the ordinarily skilled artisan.
  • the antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, is removed, for example, by centrifugation or ultrafiltration. Carter et al., Bio/Technology (1992) 10: 163-167 describe a procedure for isolating antibodies which are secreted to the periplasmic space of E. coli. Briefly, cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 min.
  • sodium acetate pH 3.5
  • EDTA EDTA
  • PMSF phenylmethylsulfonylfluoride
  • Cell debris can be removed by centrifugation.
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • the anti-LILRB2 antibodies and antigen-binding fragments thereof prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, DEAE-cellulose ion exchange chromatography, ammonium sulfate precipitation, salting out, and affinity chromatography, with affinity chromatography being the preferred purification technique.
  • Protein A immobilized on a solid phase is used for immunoaffinity purification of the antibody and antigen-binding fragment thereof.
  • the suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc domain that is present in the antibody.
  • Protein A can be used to purify antibodies that are based on human gamma 1, gamma2, or gamma4 heavy chains (Lindmark et al., J. Immunol. Meth. (1983) 62: 1-13). Protein G is recommended for all mouse isotypes and for human gamma3 (Guss et al., EMBO J. (1986)5: 1567-75).
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the antibody comprises a CH3 domain, the Bakerbond ABXTM resin (J. T. Baker, Phillipsburg, N.J.) is useful for purification.
  • the mixture comprising the antibody of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5-4.5, preferably performed at low salt concentrations (e.g., from about 0-0.25M salt).
  • the antibodies of the present disclosure may be purified.
  • purified is intended to refer to a composition, isolatable from other components, wherein the protein is purified to any degree relative to its naturally obtainable state.
  • a purified protein therefore also refers to a protein, free from the environment in which it may naturally occur.
  • substantially purified this designation will refer to a composition in which the protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95% or more of the proteins in the composition.
  • Protein purification techniques are well known to those of skill in the art. These techniques involve, at one level, the crude fractionation of the cellular milieu to polypeptide and non-polypeptide fractions. Having separated the polypeptide from other proteins, the polypeptide of interest may be further purified using chromatographic and electrophoretic techniques to achieve partial or complete purification (or purification to homogeneity). Analytical methods particularly suited to the preparation of a pure peptide are ion-exchange chromatography, exclusion chromatography; polyacrylamide gel electrophoresis; isoelectric focusing.
  • protein purification include precipitation with ammonium sulfate, PEG, antibodies and the like or by heat denaturation, followed by centrifugation; gel filtration, reverse phase, hydroxylapatite and affinity chromatography; and combinations of such and other techniques.
  • polypeptide in a prokaryotic or eukaryotic expression system and extract the protein using denaturing conditions.
  • the polypeptide may be purified from other cellular components using an affinity column, which binds to a tagged portion of the polypeptide.
  • affinity column which binds to a tagged portion of the polypeptide.
  • antibodies are fractionated utilizing agents (i.e., protein A) that bind the Fc portion of the antibody.
  • agents i.e., protein A
  • antigens may be used to simultaneously purify and select appropriate antibodies.
  • Such methods often utilize the selection agent bound to a support, such as a column, filter or bead.
  • the antibodies are bound to a support, contaminants removed (e.g., washed away), and the antibodies released by applying conditions (salt, heat, etc.).
  • the present disclosure further provides methods of using the anti-LILRB2 antibodies or antigen-binding fragments thereof to detect presence or amount of LILRB2 in a sample.
  • the method comprises contacting the sample with the antibody or antigen-binding fragment thereof disclosed herein and determining the presence or the amount of LILRB2 in the sample.
  • the methods of detecting LILRB2 using an anti- LILRB2 antibody are generally immunoassays.
  • An immunoassay is a biochemical test that measures the presence or concentration of a macromolecule or a small molecule in a sample through the use of an antibody or an antigen.
  • Immunoassays come in many different format and variants, including, without limitation, enzyme linked immunosorbent assay (ELISA), Western-blot, immunohistochemistry, immunofluorescence, flow cytometry and FACS. Additional immunoassays include radioimmunoassay (RIA), immunoradiometric assay, fluoroimmunoassay, chemiluminescent assay, bioluminescent assay.
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • fluoroimmunoassay fluoroimmunoassay
  • chemiluminescent assay chemiluminescent assay
  • bioluminescent assay bioluminescent assay.
  • Immunoassays in their most simple and direct sense, are binding assays. Certain preferred immunoassays are the various types of enzyme linked immunosorbent assays (ELISAs) and radioimmunoassays (RIA) known in the art. Immunohistochemical detection using tissue sections is also particularly useful. However, it will be readily appreciated that detection is not limited to such techniques, and western blotting, dot blotting, FACS analyses, and the like may also be used.
  • the antibodies of the disclosure are immobilized onto a selected surface exhibiting protein affinity, such as a well in a polystyrene microtiter plate. Then, a test sample suspected of containing LILRB2 is added to the wells. After binding and washing to remove non-specifically bound immune complexes, the bound antigen may be detected. Detection may be achieved by the addition of another anti-LILRB2 antibody that is linked to a detectable label.
  • ELISA is a simple “sandwich ELISA.” Detection may also be achieved by the addition of a second anti-LILRB2 antibody, followed by the addition of a third antibody that has binding affinity for the second antibody, with the third antibody being linked to a detectable label.
  • the samples suspected of containing the LILRB2 are immobilized onto the well surface and then contacted with the anti- LILRB2 antibodies of the disclosure. After binding and washing to remove non-specifically bound immune complexes, the bound anti-LILRB2 antibodies are detected. Where the initial anti- LILRB2 antibodies are linked to a detectable label, the immune complexes may be detected directly. Again, the immune complexes may be detected using a second antibody that has binding affinity for the first anti-LILRB2 antibody, with the second antibody being linked to a detectable label.
  • ELIS Irrespective of the format employed, ELIS As have certain features in common, such as coating, incubating and binding, washing to remove non-specifically bound species, and detecting the bound immune complexes. These are described below.
  • a plate with either antigen or antibody In coating a plate with either antigen or antibody, one will generally incubate the wells of the plate with a solution of the antigen or antibody, either overnight or for a specified period of hours. The wells of the plate will then be washed to remove incompletely adsorbed material. Any remaining available surfaces of the wells are then “coated” with a nonspecific protein that is antigenically neutral with regard to the test antisera. These include bovine serum albumin (BSA), casein or solutions of milk powder.
  • BSA bovine serum albumin
  • the coating allows for blocking of nonspecific adsorption sites on the immobilizing surface and thus reduces the background caused by nonspecific binding of antisera onto the surface.
  • a secondary or tertiary detection means rather than a direct procedure.
  • the immobilizing surface is contacted with the biological sample to be tested under conditions effective to allow immune complex (antigen/antibody) formation. Detection of the immune complex then requires a labeled secondary binding ligand or antibody, and a secondary binding ligand or antibody in conjunction with a labeled tertiary antibody or a third binding ligand.
  • Under conditions effective to allow immune complex (antigen/antibody) formation means that the conditions preferably include diluting the antigens and/or antibodies with solutions such as BSA, bovine gamma globulin (BGG) or phosphate buffered saline (PBS)/Tween. These added agents also tend to assist in the reduction of nonspecific background.
  • the “suitable” conditions also mean that the incubation is at a temperature or for a period of time sufficient to allow effective binding. Incubation steps are typically from about 1 to 2 to 4 hours or so, at temperatures preferably on the order of 25°C to 27°C or may be overnight at about 4°C or so.
  • the contacted surface is washed so as to remove non-complexed material.
  • a preferred washing procedure includes washing with a solution such as PBS/Tween, or borate buffer. Following the formation of specific immune complexes between the test sample and the originally bound material, and subsequent washing, the occurrence of even minute amounts of immune complexes may be determined.
  • the second or third antibody will have an associated label to allow detection.
  • this will be an enzyme that will generate color development upon incubating with an appropriate chromogenic substrate.
  • a urease, glucose oxidase, alkaline phosphatase or hydrogen peroxidase-conjugated antibody for a period of time and under conditions that favor the development of further immune complex formation (e.g., incubation for 2 hours at room temperature in a PBS-containing solution such as PBS-Tween).
  • the amount of label is quantified, e.g., by incubation with a chromogenic substrate such as urea, or bromocresol purple, or 2,2 ’-azino-di-(3 -ethyl - benzthiazoline-6-sulfonic acid (ABTS), or H2O2, in the case of peroxidase as the enzyme label. Quantification is then achieved by measuring the degree of color generated, e.g., using a visible spectra spectrophotometer.
  • a chromogenic substrate such as urea, or bromocresol purple, or 2,2 ’-azino-di-(3 -ethyl - benzthiazoline-6-sulfonic acid (ABTS), or H2O2
  • Quantification is then achieved by measuring the degree of color generated, e.g., using a visible spectra spectrophotometer.
  • the Western blot is an analytical technique used to detect specific proteins in a given sample of tissue homogenate or extract. It uses gel electrophoresis to separate native or denatured proteins by the length of the polypeptide (denaturing conditions) or by the 3-D structure of the protein (native/ nondenaturing conditions). The proteins are then transferred to a membrane (typically nitrocellulose or PVDF), where they are probed (detected) using antibodies specific to the target protein.
  • a membrane typically nitrocellulose or PVDF
  • Samples may be taken from whole tissue or from cell culture. In most cases, solid tissues are first broken down mechanically using a blender (for larger sample volumes), using a homogenizer (smaller volumes), or by sonication. Cells may also be broken open by one of the above mechanical methods. However, it should be noted that bacteria, virus or environmental samples can be the source of protein and thus Western blotting is not restricted to cellular studies only. Assorted detergents, salts, and buffers may be employed to encourage lysis of cells and to solubilize proteins. Protease and phosphatase inhibitors are often added to prevent the digestion of the sample by its own enzymes. Tissue preparation is often done at cold temperatures to avoid protein denaturing.
  • the proteins of the sample are separated using gel electrophoresis. Separation of proteins may be by isoelectric point (pl), molecular weight, electric charge, or a combination of these factors. The nature of the separation depends on the treatment of the sample and the nature of the gel. This is a very useful way to determine a protein. It is also possible to use a two-dimensional (2-D) gel which spreads the proteins from a single sample out in two dimensions. Proteins are separated according to isoelectric point (pH at which they have neutral net charge) in the first dimension, and according to their molecular weight in the second dimension.
  • isoelectric point pH at which they have neutral net charge
  • the proteins are moved from within the gel onto a membrane made of nitrocellulose or polyvinylidene difluoride (PVDF).
  • PVDF polyvinylidene difluoride
  • the membrane is placed on top of the gel, and a stack of filter papers placed on top of that. The entire stack is placed in a buffer solution which moves up the paper by capillary action, bringing the proteins with it.
  • Another method for transferring the proteins is called electroblotting and uses an electric current to pull proteins from the gel into the PVDF or nitrocellulose membrane.
  • the proteins move from within the gel onto the membrane while maintaining the organization they had within the gel. As a result of this blotting process, the proteins are exposed on a thin surface layer for detection (see below).
  • Both varieties of membrane are chosen for their non-specific protein binding properties (i.e., binds all proteins equally well). Protein binding is based upon hydrophobic interactions, as well as charged interactions between the membrane and protein. Nitrocellulose membranes are cheaper than PVDF but are far more fragile and do not stand up well to repeated probing. The uniformity and overall effectiveness of transfer of protein from the gel to the membrane can be checked by staining the membrane with Coomassie Brilliant Blue or Ponceau S dyes. Once transferred, proteins are detected using labeled primary antibodies, or unlabeled primary antibodies followed by indirect detection using labeled protein A or secondary labeled antibodies binding to the Fc region of the primary antibodies.
  • the antibodies of the present disclosure may also be used in conjunction with both fresh-frozen and/or formalin-fixed, paraffin-embedded tissue blocks prepared for study by immunohistochemistry (IHC) or immunofluorescence staining.
  • IHC immunohistochemistry
  • the method of preparing tissue blocks from these particulate specimens has been successfully used in previous IHC and immunofluorescence studies of various prognostic factors and is well known to those of skill in the art (Brown et al., 1990; Abbondanzo et al., 1990; Allred et al., 1990).
  • frozen-sections may be prepared by rehydrating 50 ng of frozen “pulverized” tissue at room temperature in phosphate buffered saline (PBS) in small plastic capsules; pelleting the particles by centrifugation; resuspending them in a viscous embedding medium (OCT); inverting the capsule and/or pelleting again by centrifugation; snap-freezing in 70°C isopentane; cutting the plastic capsule and/or removing the frozen cylinder of tissue; securing the tissue cylinder on a cryostat microtome chuck; and/or cutting 25-50 serial sections from the capsule.
  • whole frozen tissue samples may be used for serial section cuttings.
  • Permanent sections may be prepared by a similar method involving rehydration of the 50 mg sample in a plastic microfuge tube; pelleting; resuspending in 10% formalin for 4 hours fixation; washing/pelleting; resuspending in warm 2.5% agar; pelleting; cooling in ice water to harden the agar; removing the tissue/agar block from the tube; infiltrating and/or embedding the block in paraffin; and/or cutting up to 50 serial permanent sections. Again, whole tissue samples may be substituted.
  • the antibodies of the present disclosure may also be used in flow cytometry or FACS.
  • Flow cytometry is a laser- or impedance-based technology employed in many detection assays, including cell counting, cell sorting, biomarker detection and protein engineering. The technology suspends cells in a stream of fluid and passing them through an electronic detection apparatus, which allows simultaneous multiparametric analysis of the physical and chemical characteristics of up to thousands of particles per second.
  • Flow cytometry is routinely used in the diagnosis disorders, especially blood cancers, but has many other applications in basic research, clinical practice and clinical trials.
  • Fluorescence-activated cell sorting is a specialized type of cytometry. It provides a method for sorting a heterogenous mixture of biological cells into two or more containers, one cell at a time, based on the specific light scattering and fluorescent characteristics of each cell.
  • the technology involves a cell suspension entrained in the center of a narrow, rapidly flowing stream of liquid. The flow is arranged so that there is a large separation between cells relative to their diameter. A vibrating mechanism causes the stream of cells to break into individual droplets. Just before the stream breaks into droplets, the flow passes through a fluorescence measuring station where the fluorescence of each cell is measured. An electrical charging ring is placed just at the point where the stream breaks into droplets.
  • a charge is placed on the ring based immediately prior to fluorescence intensity being measured, and the opposite charge is trapped on the droplet as it breaks from the stream.
  • the charged droplets then fall through an electrostatic deflection system that diverts droplets into containers based upon their charge.
  • the antibodies of the present disclosure are labeled with fluorophores and then allowed to bind to the cells of interest, which are analyzed in a flow cytometer or sorted by a FACS machine.
  • the present disclosure concerns immunodetection kits for use with the immunodetection methods described above.
  • the immunodetection kits will thus comprise, in suitable container means, a first antibody that binds to LILRB2, and optionally an immunodetection reagent.
  • the antibody may be pre-bound to a solid support, such as a column matrix and/or well of a microtiter plate.
  • the immunodetection reagents of the kit may take any one of a variety of forms, including those detectable labels that are associated with or linked to the given antibody. Detectable labels that are associated with or attached to a secondary binding ligand are also contemplated. Exemplary secondary ligands are those secondary antibodies that have binding affinity for the first antibody.
  • suitable immunodetection reagents for use in the present kits include the two-component reagent that comprises a secondary antibody that has binding affinity for the first antibody, the second antibody being linked to a detectable label.
  • a number of exemplary labels are known in the art and all such labels may be employed in connection with the present disclosure.
  • kits may further comprise a suitably aliquoted composition of LILRBs, whether labeled or unlabeled, as may be used to prepare a standard curve for a detection assay.
  • the kits may contain antibody-label conjugates either in fully conjugated form, in the form of intermediates, or as separate moieties to be conjugated by the user of the kit.
  • the components of the kits may be packaged either in aqueous media or in lyophilized form.
  • the container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which the antibody may be placed, or preferably, suitably aliquoted.
  • the kits of the present disclosure will also typically include a means for containing the antibody, antigen, and any other reagent containers in close confinement for commercial sale.
  • Such containers may include injection or blow-molded plastic containers into which the desired vials are retained.
  • the present disclosure provides methods of diagnosing a LILRB2 related disease or condition in a subject.
  • the method comprises: a) contacting a sample obtained from the subject with the antibody or antigenbinding fragment thereof provided herein; b) determining presence or amount of LILRB2 in the sample; and c) correlating the existence or the amount of the LILRB2 to the LILRB2- related disease or condition in the subject.
  • kits comprising the antibody or antigen-binding fragment thereof provided herein, optionally conjugated with a detectable moiety.
  • the kits may be useful in detection of LILRB2 or diagnosis of LILRB2 related disease.
  • the present disclosure also provides use of the antibody or antigen-binding fragment thereof provided herein in the manufacture of a medicament for treating a LILRB2 related disease or condition in a subject, in the manufacture of a diagnostic reagent for diagnosing a LILRB2 related disease or condition.
  • the present disclosure provides a method of generating an antibody specifically binding to LILRB2.
  • the method comprises immunizing a non-human animal with a peptide fragment of LILRB2 that has low homology to other LILR family members.
  • the peptide has an amino acid sequence as set forth in any one of SEQ ID NO: 25-27.
  • a given composition for immunization may vary in its immunogenicity. It is often necessary therefore to boost the host immune system, as may be achieved by coupling a peptide or polypeptide immunogen to a carrier.
  • Exemplary and preferred carriers are keyhole limpet hemocyanin (KLH) and bovine serum albumin (BSA).
  • albumins such as ovalbumin, mouse serum albumin or rabbit serum albumin can also be used as carriers.
  • Means for conjugating a polypeptide to a carrier protein are well known in the art and include glutaraldehyde, m- maleimidobencoyl-N-hydroxysuccinimide ester, 38ittering38de and bis-biazotized benzidine.
  • the immunogenicity of a particular immunogen composition can be enhanced by the use of non-specific stimulators of the immune response, known as adjuvants.
  • adjuvants include complete Freund’s adjuvant (a nonspecific stimulator of the immune response containing killed Mycobacterium tuberculosis), incomplete Freund’s adjuvants and aluminum hydroxide adjuvant.
  • the amount of immunogen composition used in the production of polyclonal antibodies varies upon the nature of the immunogen as well as the animal used for immunization.
  • a variety of routes can be used to administer the immunogen (subcutaneous, intramuscular, intradermal, intravenous and intraperitoneal).
  • the production of polyclonal antibodies may be monitored by sampling blood of the immunized animal at various points following immunization. A second, booster injection, also may be given. The process of boosting and 38ittering is repeated until a suitable titer is achieved.
  • the immunized animal can be bled and the serum isolated and stored, and polyclonal antibodies specific to LILRB2 can be purified.
  • the method further comprises the steps of generating monoclonal antibodies (mAbs) against LILRB2.
  • the methods for generating monoclonal antibodies (mAbs) generally begin along the same lines as those for preparing polyclonal antibodies.
  • the first step is immunization of an appropriate host. When a desired level of immunogenicity is obtained, the immunized animal can be used to generate mAbs.
  • somatic cells with the potential for producing antibodies, specifically B lymphocytes (B cells), are selected for use in the mAb generating protocol. These cells may be obtained from biopsied spleens or lymph nodes, or from circulating blood. The antibody-producing B lymphocytes from the immunized animal are then fused with cells of an immortal myeloma cell, generally one of the same species as the animal that was immunized or human or human/mouse chimeric cells.
  • B lymphocytes B lymphocytes
  • Myeloma cell lines suited for use in hybridoma-producing fusion procedures preferably are non-antibodyproducing, have high fusion efficiency, and enzyme deficiencies that render then incapable of growing in certain selective media which support the growth of only the desired fused cells (hybridomas). Any one of a number of myeloma cells may be used, as are known to those of skill in the art (Goding, pp. 65-66, 1986; Campbell, pp. 75-83, 1984).
  • Methods for generating hybrids of antibody-producing spleen or lymph node cells and myeloma cells usually comprise mixing somatic cells with myeloma cells in a 2: 1 proportion, though the proportion may vary from about 20: 1 to about 1 : 1, respectively, in the presence of an agent or agents (chemical or electrical) that promote the fusion of cell membranes.
  • Fusion methods using Sendai virus have been described by Kohler and Milstein (1975; 1976), and those using polyethylene glycol (PEG), such as 37% (v/v) PEG, by Gefter et al. (1977).
  • PEG polyethylene glycol
  • the use of electrically induced fusion methods also is appropriate (Goding, pp. 71-74, 1986).
  • Fusion procedures usually produce viable hybrids at low frequencies, about 1 x 10' 6 to 1 x 10' 8 . However, this does not pose a problem, as the viable, fused hybrids are differentiated from the parental, infused cells (particularly the infused myeloma cells that would normally continue to divide indefinitely) by culturing in a selective medium.
  • the selective medium is generally one that contains an agent that blocks the de novo synthesis of nucleotides in the tissue culture media.
  • Exemplary and preferred agents are aminopterin, methotrexate, and azaserine. Aminopterin and methotrexate block de novo synthesis of both purines and pyrimidines, whereas azaserine blocks only purine synthesis.
  • the media is supplemented with hypoxanthine and thymidine as a source of nucleotides (HAT medium).
  • HAT medium a source of nucleotides
  • azaserine is used, the media is supplemented with hypoxanthine.
  • Ouabain is added if the B cell source is an Epstein Barr virus (EBV) transformed human B cell line, in order to eliminate EBV transformed lines that have not fused to the myeloma.
  • EBV Epstein Barr virus
  • the preferred selection medium is HAT or HAT with ouabain. Only cells capable of operating nucleotide salvage pathways are able to survive in HAT medium.
  • the myeloma cells are defective in key enzymes of the salvage pathway, e.g., hypoxanthine phosphoribosyl transferase (HPRT), and they cannot survive.
  • HPRT hypoxanthine phosphoribosyl transferase
  • the B cells can operate this pathway, but they have a limited life span in culture and generally die within about two weeks. Therefore, the only cells that can survive in the selective media are those hybrids formed from myeloma and B cells.
  • ouabain is also used for drug selection of hybrids as EBV- transformed B cells are susceptible to drug killing, whereas the myeloma partner used is chosen to be ouabain resistant.
  • Culturing provides a population of hybridomas from which specific hybridomas are selected. Typically, selection of hybridomas is performed by culturing the cells by single-clone dilution in microtiter plates, followed by testing the individual clonal supernatants (after about two to three weeks) for the desired reactivity.
  • the assay should be sensitive, simple and rapid, such as radioimmunoassays, enzyme immunoassays, cytotoxicity assays, plaque assays dot immunobinding assays, and the like.
  • the selected hybridomas are then serially diluted or single-cell sorted by flow cytometric sorting and cloned into individual antibody-producing cell lines, which clones can then be propagated indefinitely to provide mAbs.
  • the cell lines may be exploited for Mab production in two basic ways.
  • a sample of the hybridoma can be injected (often into the peritoneal cavity) into an animal (e.g., a mouse).
  • the animals are primed with a hydrocarbon, especially oils such as pristane (tetramethylpentadecane) prior to injection.
  • pristane tetramethylpentadecane
  • the injected animal develops tumors secreting the specific monoclonal antibody produced by the fused cell hybrid.
  • the body fluids of the animal such as serum or ascites fluid, can then be tapped to provide Mabs in high concentration.
  • the individual cell lines could also be cultured in vitro, where the Mabs are naturally secreted into the culture medium from which they can be readily obtained in high concentrations.
  • human hybridoma cells lines can be used in vitro to produce immunoglobulins in cell supernatant.
  • the cell lines can be adapted for growth in serum-free medium to optimize the ability to recover human monoclonal immunoglobulins of high purity.
  • Mabs produced by either means may be further purified, if desired, using filtration, centrifugation and various chromatographic methods such as FPLC or affinity chromatography.
  • Fragments of the monoclonal antibodies of the disclosure can be obtained from the purified monoclonal antibodies by methods which include digestion with enzymes, such as pepsin or papain, and/or by cleavage of disulfide bonds by chemical reduction.
  • monoclonal antibody fragments encompassed by the present disclosure can be synthesized using an automated peptide synthesizer.
  • RNA can be isolated from the hybridoma line and the antibody genes obtained by RT-PCR and cloned into an immunoglobulin expression vector.
  • combinatorial immunoglobulin phagemid libraries are prepared from RNA isolated from the cell lines and phagemids expressing appropriate antibodies are selected by panning using viral antigens.
  • the advantages of this approach over conventional hybridoma techniques are that approximately 10 4 times as many antibodies can be produced and screened in a single round, and that new specificities are generated by H and L chain combination which further increases the chance of finding appropriate antibodies.
  • LILRB1 (Q8NHL6), LILRB2 (Q8N423), LILRB3 (075022), LILRB4 (Q8NHJ6), LILRB5 (075023), LILRA1 (075019), LILRA2 (Q8N149), LILRA3 (Q8N6C8), LILRA4 (P59901), LILRB5 (LILRA5), and LILRA6 (Q6PI73).
  • the chosen peptides were synthesized with a C-terminal Cys at 2 mg scale using standard solid-phase methods to >85% purity (GenScript Biotech, Piscataway, NJ).
  • the synthesized peptides were characterized by mass spectrometry (MS) and high-pressure liquid chromatography (HPLC) and then conjugated to keyhole limpet hemocyanin (KLH).
  • MS mass spectrometry
  • HPLC high-pressure liquid chromatography
  • KLH keyhole limpet hemocyanin
  • Each KLH-peptide conjugate was used for immunizing 3 separate mice (Balb/c).
  • Anti-peptide titers were monitored by ELISA dilution series (1 : 1000 to 1 :512,000).
  • hybridoma cells (5xl0 6 ) for the mouse IgGik isotype monocl onals cryopreserved in FBS with 10% DMSO were used to isolate total RNA.
  • Cells were rapidly thawed in a 37°C water bath, immediately diluted into 5ml of DMEM with 10% FBS, and centrifuged at 300xg for 5 min.
  • Cells were resuspended in ice-cold PBS and centrifuged at 300xg for 5min. The washed pellets were resuspended in TRI reagent with a 1ml syringe and 20-gauge needle, and snap frozen on dry ice.
  • RNA samples were diluted 1 : 1 with absolute ethanol and total RNA purified according to the kit instructions (Direct-zol RNA miniprep kit, Zymo Research, Irvine, CA, USA). Total RNA was quantified on the Nanodrop and 5ug of each used to prepare 5’ RACE-ready cDNA via the SMART (switching mechanism at 5’ end of RNA template) technology (SMART er RACE 573’ kit, Takara Bio, Mountain View, CA, USA).
  • SMART switching mechanism at 5’ end of RNA template
  • RACE was performed using the Universal primer A (provided in kit) and custom ordered gene-specific primers (IDT DNA, Coralville, Iowa) directed to the constant regions of the mouse IgGi heavy chain (1 : 1 mix of GATTACGCCAAGCTTTCTTGTCCACCTTGGTGCTGCTGGCCGG (SEQ ID NO: 28), GATTACGCCAAGCTTTTTGTCCACCGTGGTGCTGCTGGCTGGT (SEQ ID NO: 29)) or kappa light chain (GATTACGCCAAGCTTGATCAGTCCAACTGTTCAGGACGCC (SEQ ID NO: 30)).
  • Gene-specific primers included a 15bp pRACE vector cloning sequence (GATTACGCCAAGCTT (SEQ ID NO: 31)) at the 5’ ends for downstream fusion cloning.
  • PCR was carried out for 25 cycles of 94°C 30sec, 68°C 30sec and 72°C 3 min. PCR products were run on and extracted from 1.2% agarose gels, followed by elution in 15ul from a NucleoSpin Gel clean-up column (provided in kit). Purified PCR RACE products were cloned into the linearized pRACE vector by In-Fusion HD cloning and 5ul transformed into 50ul of Stellar competent cells (provided in kit). Twelve colonies from each VH and Vk product were sequenced using the standard pucl9 vector M13F primer (Sequetech, Mountain View, CA, USA).
  • This example illustrates the screening of anti-LILRB2 antibodies suitable for immunohistochemistry assay.
  • the inventors screened the anti-LILRB2 hybridoma clones in immunohistochemistry (IHC) assay using HEK293 cells stably expressing LILRB2.
  • IHC immunohistochemistry
  • FFPE Formalin-fixed paraffin-embedded
  • HEK293 cells stably expressing LILRB2 were made into FFPE cells pellets samples.
  • FFPE sections mounted on microscope slides were deparaffinized twice in xylene for 10 minutes and then sequentially rehydrated in 100%, 95%, 70% and 50% ethanol baths for 5 minutes each. After rinsing in tap water for 5 minutes, the sections were subjected to antigen-unmasking in a 2100-Retriever (EMS, Cat. No. 62706) using Target Retrieval Solution, Citrate pH 6 (Agilent, Cat. No. S236984-2).
  • Sections were rinsed in tap water for 5 minutes, then blocked and stained using ImmPRESS® HRP Horse Anti-Mouse IgG PLUS Polymer Kit (for mouse antibody; Vector Laboratories, Cat. No. MP-7802-15) or ImmPRESS® HRP Horse Anti-Rabbit IgG PLUS Polymer Kit (for rabbit antibody; Vector Laboratories, Cat. No. MP-7801) according to the manufacturer’s manual. Briefly, the sections were incubated in BLOXALL Blocking Solution for 10 minutes, washed in PBS, blocked with 2.5% horse serum for 20 minutes, then incubated with diluted primary antibody for 30 minutes at room temperature. The sections were then washed in PBS, incubated with ImmPRESS Horse Reagent, washed in PBS.
  • the signal was developed using ImmPACT DAB Eqv working solution.
  • the cell nuclei were counterstained with hematoxylin for 30 seconds.
  • the sections were then rinsed in tap water for 5 minutes and dehydrated in 50%, 70%, 95%, 100% ethanol baths and twice in xylene baths for 5 minutes each.
  • the sections were air-dried, and the slides mounted with VectaMountTM (Vector Laboratories; Cat. No. H- 5000).
  • a group of hybridomas (listed in Table 4) can recognize LILRB2 expressed in HEK293 cells.
  • the inventors then assessed the binding specificities of the anti-LILRB2 antibodies (recombinant and/or hybridoma supernatant) to synthetic peptides from LILRs by ELISA. Based on a multiple sequence alignment, peptides from each LILR as listed in Table 5 were assessed. Each peptide with a C-terminal lysine-biotin moiety was synthesized using standard methods at Elim Biopharmaceuticals and purified by HPLC to >95% and characterized by mass spectrometry prior to use.
  • Peptides were coated at 10 pg/mL in sodium bicarbonate, pH 9.6 overnight at 4°C on a 96-well MaxiSorp plate, washed 3x with PBS/Tween and blocked with 1% BSA in PBS. After washing 3x with PBS/Tween the mouse antibodies were incubated in PBS/Tween + 0.5% BSA for 2 hours. After washing 6x with PBS/Tween, the mouse antibodies were bound using a 1 :10,000 dilution of goat anti-mouse IgG-HRP conjugation in PBS/Tween + 0.5% BSA. Samples were washed 6x with PBS/Tween and detected with 3, 3’, 5,5’- Tetram ethylbenzidine (TMB).
  • TMB Tetram ethylbenzidine
  • the inventors then assessed the specificity of anti-LILRB2 hybridoma clones in the IHC assay using HEK293 cells expressing LILRs.
  • HEK293 cells cultured on microscope slide coverslips were transiently transfected with LILRB2, LILRB1, LILRB3 or LILRA6. Non-transfected cells were used as negative control.
  • the specificity of 16B9, 29F12, 49G11, 32G8, and 37B2 were tested using IHC staining with 1 :5 diluted supernatant from each hybridoma clone. A more detailed description of the method is provided below.
  • HEK293 cells were seeded on 20 x 20 millimeter (mm) microscope slide coverslips placed in 6-well plates and cultured overnight to reach 80% confluency.
  • DNA plasmids (5 pg) coding full length LILRB2 protein were transfected using LipofectamineTM 3000 following manufacturer’s instruction.
  • Transfected HEK293 cells were cultured for 4 days in a 5% CO 2 incubator.
  • the transiently transfected HEK293 cells were stained by IHC using VECTASTAIN Elite ABC HRP Kit according to the manual.
  • the coverslips were fixed in 4% paraformaldehyde in phosphate-buff ered saline (PBS) for 10 minutes, washed with PBS, incubated in BLOXALL Blocking Solution for 10 minutes, washed with PBS, blocked with 2.5% normal horse serum for 20 minutes, blocked with Avidin/Biotin Blocking Kit, incubated with diluted primary antibody overnight at 4°C, washed 3 times with PBS, incubated with diluted biotinylated anti -mouse secondary antibody for 1 hour at room temperature, washed 3 times with PBS, incubated with VECTASTAIN ABC buffer for 30 minutes, washed 3 times with PBS, and then developed with 3,3’ Di aminobenzidine (DAB) for 3 minutes. After wash with PBS 3 times, the coverslips were mounted on microscope slides with VectaMountTM.
  • PBS phosphate-buff ered saline
  • the inventors titrated the recombinant anti-LILRB2 antibodies derived from clones 16B9 and 37B2 using FFPE sections of HEK293 or HEK293 LILRB2 cell pellets.
  • the methods of IHC assay using FFPE sections of HEK293 or HEK293 LILRB2 cell pellets have been described in Example 2 supra.
  • recombinant 16B9 and 37B2 antibodies bind to LILRB2 in the IHC assay at the concentrations of 10 to 0.15625 pg/mL.
  • the inventors then evaluated the cross-reactivity of recombinant 16B9 and 37B2 antibodies to the peptides from different LILR members using ELISA and bio-layer interferometry (BLI).
  • the recombinant 16B9 and 37B2 antibodies were used to detect endogenous LILRB2 in human normal tissues by IHC staining of FFPE sections of lung, spleen or bone marrow from healthy human donors.
  • FFPE tissue sections mounted on microscope slides were deparaffinized twice in xylene baths for 10 minutes and then sequentially rehydrated in 100%, 95%, 70% and 50% ethanol baths for 5 minutes each. After rinsing in tap water for 5 minutes, the sections were subjected to antigen-unmasking in a 2100-Retriever using Target Retrieval Solution, Citrate pH 6. Sections were rinsed in tap water for 5 minutes, then blocked and stained using ImmPRESS® HRP Horse Anti -Mouse IgG PLUS Polymer Kit or ImmPRESS® HRP Horse Anti-Rabbit IgG PLUS Polymer according to the manufacturer’s manual.
  • the sections were incubated in BLOXALL Blocking Solution for 10 minutes, washed in PBS, blocked with 2.5% horse serum for 20 minutes, then incubated with diluted primary antibodies (anti-LILRB2, anti-CDl lb or anti-CD163) for 30 minutes at room temperature.
  • the sections were then washed in PBS, incubated with ImmPRESS Horse Reagent, washed in PBS.
  • the signal was developed using ImmPACT DAB Eqv working solution.
  • the cell nuclei were counterstained with hematoxylin for 30 seconds.
  • the sections were then rinsed in tap water for 5 minutes and dehydrated in 50%, 70%, 95%, 100% ethanol baths and twice in xylene baths for 5 minutes each.
  • the sections were air-dried, and the slides mounted with VectaMountTM.
  • the recombinant 16B9 and 37B2 antibodies detected endogenous LILRB2 in human normal or tumor tissues by IHC or immunofluorescence staining, at a concentration 0.3125-2.5 pg/mL (16B9 antibody) and 0.625 -2.5 pg/mL (37B2 antibody).
  • the staining pattern of the myeloid cell markers CDl lb or CD163 was also analyzed to compare with the staining pattern of 16B9 or 37B2,
  • Recombinant 16B9 antibody was used to detect endogenous LILRB2 in human normal tissues by immunofluorescence staining.
  • FFPE tissue sections mounted on microscope slides were deparaffinized twice in xylene baths for 10 minutes and then sequentially rehydrated in 100%, 95%, 70% and 50% ethanol baths for 5 minutes each. After rinsing in tap water for 5 minutes, the sections were subjected to antigen-unmasking in 2100- Retriever using Target Retrieval Solution, Citrate pH 6.
  • the slides were rinsed in tap water for 5 minutes, permeabilized with 0.4% Triton X-100 for 10 minutes, washed with PBS, and block with 5% goat and donkey serum in PBS for 1-2 hours, incubated with diluted primary antibodies (anti-LILRB2 or anti-CDl lb or anti-CD163) overnight at 4°C, washed with PBS 3 times, incubated with Goat anti-Rabbit IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor Plus 594 and Donkey anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 488 at 1 : 1000 dilution for 1 hour.
  • the sections were washed 3 times with PBS and stained with DAPI for 1 minute, rinsed with PBS and mount the slides with ProLongTM Gold Antifade Mountant.
  • the inventors then identified amino acid residues of LILRB2 that are critical for the binding of 16B9 and 37B2 antibodies using BLI analysis of captured biotinylated peptides.
  • BLI analysis has been described supra.
  • an isoleucine residue is critical for the binding of 16B9 and 37B2 antibodies to LILRB2. Binding to this isoleucine is important for the specificity of 16B9 and 37B2 to LILRB2 over LILRA6 and LILRB3 which have closely related peptides.
  • 16B9 antibody detects LILRB2 in FFPE samples pre-treated with therapeutic anti-LILRB2 antibody
  • the inventors also tested whether 16B9 recombinant antibody can detect LILRB2 by IHC staining in FFPE sections prepared from samples that have been exposed to, or treated with, a therapeutic antibody (e.g. antagonist or agonist antibody) targeting LILRB2.
  • a therapeutic antibody e.g. antagonist or agonist antibody
  • Cultured HEK293 LILRB2 cells were detached from tissue culture dishes, resuspended in culture medium, and then treated, or left untreated, with 20 pg/mL anti-LILRB2 therapeutic antibody in suspension for 1 hour at 37°C. The cells were pelleted by centrifugation, washed in PBS and processed into FFPE sections. The sections were stained by IHC using 16B9 recombinant antibody at different concentrations.
  • 16B9 recombinant antibody can detect LILRB2 by IHC staining in HEK293 LILRB2 FFPE samples that have been pre-treated with a therapeutic anti-LILRB2 antibody.
  • This example illustrates the epitope mapping and LILRB2 peptide specificity of 16B9 antibody.
  • the 16B9 antibody from lOug/ml with 1 :3 dilutions in 0.5%BSA/PBST was allowed to bind for 1 hour at ambient temperature, washed in PBST and the secondary antibody peroxidase-AffiniPure Goat Anti-Mouse IgG, Fey Fragment Specific (min X Hu, Bov, Hrs Sr Prot) was added for 1 hour at ambient temperature.
  • the plates were developed with a TMB (3,3',5,5'-Tetramethylbenzidine) substrate.
  • the 16B9 antibody is a specific IHC reagent in tissues important to LILRB2 therapeutic applications.
  • Off-target IHC staining to unknown cytoplasmic antigens were observed in some normal human tissues such as heart and testes.
  • the inventors explored whether the paratope of 16B9 could be edited to decrease this off-target staining while maintaining sensitivity and specificity to LILRB2.
  • the mouse 16B9 antibody was converted to a scFv format and tested to show it retained its binding properties.
  • a systematic “NNK walk” across each of the heavy chain CDRs was designed into an oligonucleotide pool (Table 8) used to generate a scFv library.
  • each CDR residue is tested one at a time as either wildtype or any of the other 20 amino acids in the context of the otherwise wildtype sequence.
  • the 16B9 binding is strongly dependent on the PTGPI motif. This motif can be identified by the NCBI Blastp tool in other proteins, such as titin.
  • the inventors compared binding of the 16B9 scFv NNK walk library to peptide #1 (wildtype), peptide #5 (TAA) and peptide #8 (IAA), with the hope to select for changes in the heavy chain CDRs that would de-emphasize the importance of the contacts with threonine and isoleucine in PTGPI and recruit or strengthen alternative contacts within the 16 amino acid long peptide. Binding was quantified statistically by the enrichment of sequences in NGS data in each selected library after three rounds of ELISA binding.
  • FIG. 10 A logo plot of a selection of the NGS sequences is shown in Figure 10, with the top 100 sequences obtained for peptide #1 selection listed in SEQ ID NOs: 78-183, the top 100 sequences obtained for peptide #5 selection listed in SEQ ID NOs: 184-346, and the top 100 sequences obtained for peptide #8 selection listed in SEQ ID NOs: 347-539.
  • Ten variants representing single amino acid CDR changes enriched in NGS data after binding both peptide #5 and #8 but not peptide #1 were made as recombinant IgG (Table 9).
  • This example illustrates the staining and scoring of human cancer samples based on LILRB2, CD3 and CD 163 IHC.
  • CD163, CD3 and LILRB2 were stained with respective IHC antibodies in 195 evaluable samples from 19 different human cancer indications.
  • Figure 14A shows the number of samples in each LILRB2 IHC reactivity score (0-5) in macrophages across solid tumor types.
  • Figure 14B shows the average score and correlation between LILRB2, CD 163 and CD3 IHC staining across tumor types.
  • Figure 14C shows the ranking of tumor types stained by IHC based on reactivity scores. Each tissue was evaluated by CRO’s board- certified pathologist separately for LILRB2, CD163, and CD3 reactivity. Positively staining immune cells were assessed and scored within tumor and tumor-induced stroma (TIS).
  • TIS tumor-induced stroma
  • the number of reactive cells for each biomarker in each sample was counted at 20X magnification over multiple fields and the cell counts were converted to IHC reactivity score of 0-5 as defined here: 5: >100 reactive cells; 4: 51-100 reactive cells; 3: 26-50 reactive cells; 2: 11-25 reactive cells; 1 : 1-10 reactive cells; 0: ⁇ 1 reactive cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
PCT/US2022/079097 2021-11-01 2022-11-01 Nouveaux anticorps anti-lilrb2 et produits dérivés WO2023077172A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163274511P 2021-11-01 2021-11-01
US63/274,511 2021-11-01

Publications (1)

Publication Number Publication Date
WO2023077172A2 true WO2023077172A2 (fr) 2023-05-04

Family

ID=86160676

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/079097 WO2023077172A2 (fr) 2021-11-01 2022-11-01 Nouveaux anticorps anti-lilrb2 et produits dérivés

Country Status (1)

Country Link
WO (1) WO2023077172A2 (fr)

Similar Documents

Publication Publication Date Title
JP7034950B2 (ja) 抗hla-g特異的抗体
CN110305210B (zh) 新型抗体分子、其制备方法及其用途
US11834501B2 (en) ROR2 antibody compositions and related methods
TWI655209B (zh) 結合人類計畫性死亡(programmed death)配位子1(pd-l1)之抗體
KR20210099052A (ko) 항-pd-l1/항-4-1bb 이중특이적 항체 및 이의 용도
KR20180101623A (ko) Psma 및 cd3 이중특이성 t 세포 맞물림 항체 작제물
JP6827485B2 (ja) ムスカリン性アセチルコリン受容体の結合剤およびそれらの使用
CN115768793A (zh) 新型lilrb2抗体和其用途
KR101504039B1 (ko) 인간 및 마우스 l1cam 단백질에 특이적으로 결합하는 항체 및 이의 용도
KR20150008095A (ko) 암 진단에 유용한 클로딘 18.2에 대한 항체
NL2011406C2 (en) Method for obtaining april-binding peptides, process for producing the peptides, april-binding peptides obtainable with said method/process and use of the april-binding peptides.
JP2019512207A (ja) Foxp3由来のペプチドに特異的なt細胞受容体様抗体
US20230331836A1 (en) Anti-cldn18.2 antibodies and diagnostic uses thereof
JP2023540526A (ja) ネクチン-4抗体およびそれの使用
CN113195516A (zh) 由特异性结合物识别的表位标签
US11505614B2 (en) Antibodies binding to soluble BCMA
WO2013162748A1 (fr) Variants d'anticorps anti-marqueur 1 endothélial de tumeur (tem1) et leurs utilisations
WO2023077172A2 (fr) Nouveaux anticorps anti-lilrb2 et produits dérivés
TW202233680A (zh) 具有增加的選擇性之多靶向性雙特異性抗原結合分子
CN115917317A (zh) 用于测定表达嵌合抗原的免疫细胞的体外肿瘤杀伤活性的基于细胞的测定法
JP7082112B2 (ja) 抗gpr20抗体
US11174310B2 (en) Disulfide-type HMGB1-specific antibody, method for measuring disulfide-type HMGB1 and kit for said measurement, and measurement method capable of quantitating all of HMGB1 molecules including reduced HMGB1, disulfide-type HMGB1 and thrombin-cleavable HMGB1 and kit for said measurement
JP2012500818A (ja) ヒトepo受容体に対する抗体
RU2815883C1 (ru) Антитела, пригодные в диагностике рака
CN115867352A (zh) 抗α4β7抗体

Legal Events

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

Ref document number: 22888598

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2022888598

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022888598

Country of ref document: EP

Effective date: 20240603