WO2010104949A2 - Anti-bcma antibodies - Google Patents

Anti-bcma antibodies Download PDF

Info

Publication number
WO2010104949A2
WO2010104949A2 PCT/US2010/026825 US2010026825W WO2010104949A2 WO 2010104949 A2 WO2010104949 A2 WO 2010104949A2 US 2010026825 W US2010026825 W US 2010026825W WO 2010104949 A2 WO2010104949 A2 WO 2010104949A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
cell
cells
cdr2
Prior art date
Application number
PCT/US2010/026825
Other languages
French (fr)
Other versions
WO2010104949A3 (en
Inventor
Susan L. Kalled
Yen-Ming Hsu
Original Assignee
Biogen Idec Ma 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
Priority to ES10708865.0T priority Critical patent/ES2593583T3/en
Priority to NZ59498510A priority patent/NZ594985A/en
Priority to EP10708865.0A priority patent/EP2406284B9/en
Priority to DK10708865.0T priority patent/DK2406284T5/en
Priority to CA2754938A priority patent/CA2754938C/en
Priority to SI201031281A priority patent/SI2406284T1/en
Priority to MX2011009430A priority patent/MX2011009430A/en
Priority to LTEP10708865.0T priority patent/LT2406284T/en
Priority to AU2010224160A priority patent/AU2010224160A1/en
Priority to MX2013013076A priority patent/MX341884B/en
Priority to JP2011554150A priority patent/JP6061469B2/en
Priority to US13/255,610 priority patent/US9034324B2/en
Application filed by Biogen Idec Ma Inc. filed Critical Biogen Idec Ma Inc.
Priority to CN201080020559.0A priority patent/CN102421801B/en
Publication of WO2010104949A2 publication Critical patent/WO2010104949A2/en
Publication of WO2010104949A3 publication Critical patent/WO2010104949A3/en
Priority to IL214996A priority patent/IL214996A/en
Priority to HK12107062.4A priority patent/HK1166333A1/en
Priority to US14/596,769 priority patent/US20150125460A1/en
Priority to US15/065,641 priority patent/US20170029518A1/en
Priority to CY20161100917T priority patent/CY1118069T1/en
Priority to HRP20161194TT priority patent/HRP20161194T1/en
Priority to US16/026,242 priority patent/US11111307B2/en
Priority to US17/444,107 priority patent/US20220213208A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/54F(ab')2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]

Definitions

  • This invention relates to antibodies that bind to the B cell surface antigen BCMA.
  • the invention also relates to the use of these antibodies to detect, deplete, and otherwise manipulate various B cell subtypes.
  • B cells are lymphocytes that play major roles in adaptive humoral immunity and production of antibodies that specifically recognize antigens.
  • Three subclasses of B cells are na ⁇ ve B cells, memory B cells, and plasma cells.
  • a B cell is said to be specific for an antigen that binds the antibodies made by that B cell.
  • B cells in general are stimulated by exposure to their specific antigen (Ag).
  • Na ⁇ ve B cells have not yet been exposed to their specific antigen. Such exposure (e.g., during an infection) results in proliferation of B cells and generation of sister clones.
  • Sister clones can develop into plasma cells, which produce high amounts of antibody. Plasma cells may either be short lived, or may migrate into bone marrow, where they can persist for an extended period of time.
  • a sister clone of an Ag-exposed B cell may also develop into a memory B cell that is quiescent until reexposed to the specific antigen.
  • Memory B cells respond rapidly to reexposure to antigen by dividing to produce both plasma cells and additional memory B cells.
  • Memory B cells include switched memory B cells (CD19 + CD27 high CD38 low lgD " ), unswitched memory B cells (CD19 + CD27 hi9h CD38 l0W IgD + ), and double negative memory B cells (CD19 + CD27 " CD38 l0W lgD-).
  • B cells Malignant transformation of B cells leads to cancers, including some lymphomas such as, for example, multiple myeloma and Hodgkins' Lymphoma. Some autoimmune diseases, including systemic lupus erythematosus (SLE), also involve B cells. Both cancer and autoimmune diseases that involve B cells may be considered gain of function conditions, in that the B cells overgrow and/or attack parts of the body inappropriately. A possible strategy to control such diseases is to use antibodies that target the pathological B cells.
  • SLE systemic lupus erythematosus
  • BCMA The B cell maturation antigen
  • TNFRSF17 and CD269 are proteins that has been shown to be expressed on the surface of plasmablasts (i.e., plasma cell precursors) and plasma cells, and is believed to stimulate survival. It therefore represents a potential target for B cell-related diseases.
  • BCMA is a member of the TNF receptor family and binds the TNF family ligands BAFF and APRIL (reviewed in Kalled et al. (2005), Curr Dir Autoimmun 8:206-242).
  • BCMA is a type III membrane protein, as it lacks the signal peptide associated with type I membrane proteins found in most TNF receptor family members.
  • BCMA RNA has been detected in the spleen, lymph nodes, thymus, adrenals and liver, and analysis of a number of B cell lines indicated that BCMA mRNA levels increased upon maturation.
  • Human BCMA protein has been detected on various subtypes of CD38 + B cells, particularly plasma cells (Zhang et al. (2005), lnt Immunol 17:779-788; Darce et al. (2007), J Immunol 179:7276- 7286; Sims et al. (2005), Blood 105:4390-4398; Avery et al. (2003), J Clin Invest 112:286-297).
  • BCMA The mechanism of action of BCMA is not fully understood. Mice that have been genetically altered to lack a functional gene for BCMA have normal lymphoid organs and cell populations, and a nearly normal functioning immune system (Xu and Lam (2001), MoI Cell Biol 21:4067-4074; Schiemann et al. (2001), Science 293:2111-2114). The only defect defined to date in these mice is a diminished survival of long-lived bone marrow (BM) plasma cells (O'Connor et al. (2004), J Exp Med 199:91-98). Therefore, it may be that BCMA, at least in the murine system, provides a survival signal to BM-resident plasma cells that is either BAFF or APRIL-mediated, or both.
  • BM bone marrow
  • the expression of BCMA on the surface of some B cells provides a marker by which those cells may be specifically targeted.
  • agents that specifically bind to BCMA and for a determination of which B cell subsets are bound by those BCMA-specific agents.
  • the invention provides antibodies that specifically bind to BCMA.
  • the antibodies of the invention may be used to target one or more of the following B cell subsets: plasma cells, memory B cells, and na ⁇ ve B cells.
  • Figure 1 depicts binding of anti-human BCMA mAbs on BCMA- transfected CHO cells. Binding of biotin-conjugated anti-human BCMA mAbs, visualized with Streptavidin-PE, was measured by flow cytometry on a BCMA- CHO stable cell line (A) and control, non-transfected CHO cells (B). The shaded area represents staining of cells with an isotype control mAb.
  • Figure 2 depicts anti-BCMA binding to B cell subsets.
  • B cell subsets in human peripheral blood were assessed by flow cytometry for reactivity to anti-BCMA mAbs. Visualization was as in Figure 1. The shaded area represents staining with an isotype control Ab.
  • B cell subsets were plasma cells (CD19 + CD27 h ⁇ gh CD38 h ⁇ gh lgD " ) (A), switched memory B cells (CD19 + CD27 high CD38 l0W lgD-) (B), unswitched memory B cells (CD19 + CD27 high CD38 l0W IgD + ) (C), double negative memory B cells (CD19 + CD27 ⁇ CD38 l0W lgD " ) (D), and naive B cells (CD19 + CD271gD + ) (E).
  • Figure 3 depicts anti-BCMA binding to B cell subsets isolated from healthy and SLE-afflicted individuals.
  • B cell subsets in human peripheral blood from a healthy volunteer and an SLE patient were assessed by flow cytometry for reactivity to the anti-BCMA mAbs C12A3.2 and A7D12.2.
  • B cell subsets were as in Figure 2.
  • Visualization was as in Figure 1.
  • FIG 4 depicts flow cytometric staining of human plasma cells within the human CD45 + splenocyte compartment isolated from HSC/NSG mice. Splenic plasma cells were stained with anti-BCMA antibodies A7D12.2 (left panel; bold line) and C12A3.2 (right panel; bold line) or an isoytpe control mouse lgG2b or IgGI Ab, respectively (thin line in both panels).
  • Figure 6 depicts flow cytometric staining for plasma cells (PCs) within the human CD45 + splenocyte compartment isolated from HSC/NSG mice treated with anti-BCMA antibody (chC11 D5.1 or chA7D12.2) or human IgGI control. Mice were injected i.p. with anti-BCMA Ab or HIgGI control twice weekly for 2 weeks. 45 huC13F12.1 H2 humanized mature heavy chain variable domain sequence
  • the invention provides antibodies that bind to BCMA and certain epitopes thereof.
  • the antibodies of the invention bind to one or more subsets of B cells, such as plasma cells, memory B cells, and na ⁇ ve B cells.
  • the invention also provides methods of depleting B cells or subclasses of B cells, including plasma cells, memory B cells, and na ⁇ ve B cells.
  • the invention provides an isolated antibody that binds to SEQ ID NO:9 and binds to plasma cells.
  • the invention provides an isolated antibody that binds to SEQ ID NO:9 and binds to memory B cells.
  • the invention provides an isolated antibody that binds to SEQ ID NO:9 and binds to na ⁇ ve B cells.
  • Certain anti-BCMA mAbs including clone C4E2.2 (hamster IgG) generated at Legacy Biogen (6); clone VICKY-1 (rat IgGI) (Alexis Biochemicals, Lausen, Switzerland, also sold as 6D10 by Santa Cruz Biotechnology, Santa Cruz, CA); and clone 335004 (rat lgG2a) (R&D Systems, Inc., Minneapolis, MN), are outside the scope of this invention.
  • the invention provides antibodies that bind specifically to SEQ ID NO:9.
  • the invention also provides to antibodies that bind to the surface of B cells or subclasses thereof, including plasma cells, memory B cells (including switched, unswitched, and double negative), and/or na ⁇ ve B cells.
  • the term "antibody” as used herein, includes both full-length immunoglobulins and antibody fragments that bind to the same antigens.
  • the antibodies can be, e.g., a monoclonal, polyclonal, chimeric, humanized, or single chain antibody.
  • the antibody fragments are Fab fragments or F(ab')2 fragments and retain the ability to specifically bind the protein of SEQ ID NO: 9.
  • the invention provides the antibodies A7D12.2, C11 D5.3, C12A3.2, and C13F12.1. Each of these is a murine monoclonal antibody.
  • A7D12.2 has a murine "miscellaneous" subgroup heavy chain, a heavy chain variable domain sequence that is SEQ ID NO:1 , a subgroup I kappa light chain, and a light chain variable domain sequence that is SEQ ID NO:2.
  • C11 D5.3 has a subgroup H(A) heavy chain, a heavy chain variable domain sequence that is SEQ ID NO:3, a murine subgroup III kappa light chain, and a light chain variable domain sequence that is selected from SEQ ID NO:4, SEQ ID NO:11 , and SEQ ID NO:12.
  • the light chain variable domain sequence of C11 D5.3 is SEQ ID NO: 12.
  • C12A3.2 has a subgroup H(A) heavy chain, a heavy chain variable domain sequence that is SEQ ID NO:5, a murine subgroup III kappa light chain, and a light chain variable domain sequence that is SEQ ID NO:6.
  • C13F12.1 has a subgroup II(A) heavy chain, a heavy chain variable domain sequence that is SEQ ID NO:7, a murine subgroup III kappa light chain, and a light chain variable domain sequence that is SEQ ID NO:8.
  • Techniques for producing single-chain antibodies specific to the protein of SEQ ID NO: 9 can be adapted from e.g., those described in U.S. Patent 4,946,778.
  • methods can be adapted for the construction of Fab expression libraries (see e.g., Huse et al. (1989) Science 246:1275-1281) to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for a BCMA protein or derivatives, fragments, analogs or homologs thereof.
  • Numerous techniques for humanizing non-human antibodies are well known in the art. See e.g., U.S. Patent 5,225,539, 6,632,927, or 5,648,237, all of which are incorporated by reference.
  • Antibody fragments that contain the idiotypes to a BCMA protein may be produced by any of a variety of techniques, including, but not limited to: (i) an F(ab')2 fragment produced by pepsin digestion of an antibody molecule; (ii) an Fab fragment generated by reducing the disulfide bridges of an F(ab')2 fragment; (iii) a Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent and (iv) Fv fragments.
  • recombinant anti-BCMA antibodies such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention.
  • Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques such as, for example, the methods described in U.S. Patent No. 7,112,421 ; Better et al. (1988) Science 240:1041-1043; or Liu et ai. (1987) Proc. Natl. Acad. Sci. USA 84:3439- 3443.
  • a chimeric form of A7D12.2 comprises a heavy chain comprising SEQ ID NO:13 and a light chain comprising SEQ ID NO:14.
  • a chimeric form of C11 D5.3 comprises a heavy chain comprising SEQ ID NO:15 and a light chain comprising a sequence selected from SEQ ID NO: 16 and SEQ ID NO: 17, preferably SEQ ID NO: 17.
  • a chimeric form of C12A3.2 comprises a heavy chain comprising SEQ ID NO:18 and a light chain comprising SEQ ID NO: 19.
  • a chimeric form of C13F12.1 comprises a heavy chain comprising SEQ ID NO:20 and a light chain comprising SEQ ID NO:21.
  • a humanized form of C11 D5.3 comprises a light chain variable domain comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 22-24 and a heavy chain variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 25-29.
  • a humanized form of C12A3.2 comprises a light chain variable domain comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 30-33 and a heavy chain variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 34-38.
  • a humanized form of C13F12.1 comprises a light chain variable domain comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 39-42 and a heavy chain variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 43-47.
  • the antibodies of the invention may comprise the heavy chain variable domain sequences of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7.
  • the heavy chain variable domain sequences may consist essentially of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7.
  • the antibodies of the invention may comprise the light chain variable domain sequences of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO:12.
  • the light chain variable domain sequences may consist essentially of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO:12.
  • the invention also provides a variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to a sequence selected from SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
  • the invention also provides a variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to a sequence selected from SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , and SEQ ID NO:12.
  • the invention also provides antibodies comprising a heavy chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:1 and a light chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:2.
  • the invention includes antibodies comprising a heavy chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:3 and a light chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO: 4, SEQ ID NO:11 , or SEQ ID NO: 12.
  • the invention includes antibodies comprising a heavy chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:5 and a light chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:6.
  • the invention includes antibodies comprising a heavy chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:7 and a light chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:8.
  • the invention also provides antibodies with particular complementarity determining regions (CDR). Table 2 defines the amino acid coordinates of CDR1 , CDR2, and CDR3 of SEQ ID NOs:1 through 8, 11 , and 12. Table 2. CDR Amino Acid Coordinates
  • CDRs are designated using the Kabat definitions (Johnson and Wu (2000), Nucleic Acids Res 28:214-218). As used herein, the "corresponding CDR” means the CDR in the most similar position within the variable domain amino acid sequence.
  • the heavy chain variable domain of antibodies of the invention may comprise CDRs such that one, two, or three of the CDRs are identical to the corresponding CDRs of SEQ ID NO:1 ; identical to the corresponding CDRs of SEQ ID NO:3; identical to the corresponding CDRs of SEQ ID NO:5; or identical to the corresponding CDRs of SEQ ID NO:7.
  • the light chain variable domain of antibodies of the invention may comprise CDRs such that one, two, or three of the CDRs are identical to the corresponding CDRs of SEQ ID NO:2; identical to the corresponding CDRs of SEQ ID NO:4; identical to the corresponding CDRs of SEQ ID NO:6; identical to the corresponding CDRs of SEQ ID NO:8; identical to the corresponding CDRs of SEQ ID NO:11 ; or identical to the corresponding CDRs of SEQ ID NO:12.
  • the heavy chain variable domain may comprise CDRs identical to each of the corresponding CDRs of one of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7 except that one or more amino acid substitutions have been made in said CDR regions.
  • CDRs of the heavy chain variable domain may have up to a total of 12 amino acid substitutions relative to SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7.
  • the heavy chain variable domain CDRs of the antibodies of the invention may have up to 10, up to 8, up to 5, or up to 3 substitutions relative to SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7.
  • the heavy chain variable domain CDRs of the antibodies of the invention are at least 80%, at least 85%, at least 90%, or at least 95% identical to the heavy chain variable domain CDRs of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7.
  • CDR2 of SEQ ID NO:7 is replaced by CDR2 (i.e., amino acids 50-66) of SEQ ID NO:46.
  • the heavy chain variable domain of an antibody of the invention may comprise CDR1 and CDR3 of SEQ ID NO:7 and CDR2 of SEQ ID NO:46.
  • the heavy chain variable domain of an antibody of the invention may also comprise CDR1 , CDR2, and CDR3 regions that are together at least 80%, at least 85%, at least 90%, or at least 95% identical to CDR1 and CDR3 of SEQ ID NO:7 and CDR2 of SEQ ID NO:46.
  • the light chain variable domain may comprise CDRs identical to the corresponding CDRs of one of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO:12, except for one or more amino acid substitutions in said CDR regions.
  • the antibodies of the invention comprise CDRs that are identical to the corresponding CDRs of one Of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO: 12, except for up to 12, up to 10, up to 8, up to 5, or up to 3 amino acid substitutions in said CDR regions.
  • the light chain variable domain CDRs of the antibodies of the invention are at least 80%, at least 85%, at least 90%, or at least 95% identical to the light chain variable domain CDRs of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO:12.
  • the substitutions in the CDR regions are conservative substitutions.
  • the invention also provides antibodies that bind particular epitopes. Whether a pair of antibodies binds the same epitope is determined based on cross-blocking experiments, as described in Example 4. Cross-blocking profiles are defined for seven antibodies in Table 3. For the purposes of this disclosure, two antibodies are considered to bind to the same epitope if each one reduces the other's binding to BCMA (i.e., they mutually cross-block) by at least 90% according to the procedure described in Example 4. Similarly, antibodies that do not reduce each other's binding by at least 90% as described in Example 4 are considered to bind to distinct epitopes.
  • the cross-blocking profiles of antibodies with certain variable domains are listed in Table 3. Pairs of antibodies that bind to distinct epitopes (as defined above) are noted with a "d.”
  • the invention further encompasses antibodies that bind to the same epitope as antibodies A7D12.2, C11 D5.3, C12A3.2, or C13F12.1.
  • the invention also provides antibodies that have cross-blocking profiles that match the profiles of A7D12.2, C11 D5.3, C12A3.2, or C13F12.1.
  • an antibody that has the same profile as C11 D5.3 binds to the same epitope as compared to C12A3.2 and C13F12.1 but binds to a distinct epitope as compared to A7D12.2, 335004, C4E2, and Vicky-1.
  • the antibodies of the invention mutually cross-block one or more of A7D12.2, C11 D5.3, C12A3.2, or C13F12.1 from binding the protein of SEQ ID NO: 9 by at least 80%, 85%, 90%, or 95%.
  • the extent of cross-blocking is measured according to the procedure described in Example 4.
  • the antibodies and antibody fragments of the invention bind to the extracellular domain of BCMA.
  • the antibodies bind to amino acids 1-52, 1-51 , 1-41 , or 8-41 of SEQ ID NO:9.
  • the invention also provides anti-BCMA antibodies that bind to one or more particular types of cell.
  • the antibodies or antibody fragments of the invention may bind one or more of the following: plasma cells, memory B cells, na ⁇ ve B cells, or cells that express BCMA (SEQ ID NO:9), a protein similar thereto, the extracellular domain thereof, or a polypeptide similar to the extracellular domain thereof.
  • the invention provides methods of depleting various types of cells.
  • the methods comprise administering the antibodies of the invention, as described above.
  • Types of cells that may be depleted by the methods of the invention include, without limitation, plasma cells, na ⁇ ve B cells, memory B cells (including switched, unswitched, and double negative), lymphoma cells derived from B cells, and cells that express BCMA, a protein similar thereto, the extracellular domain thereof, or a polypeptide similar to the extracellular domain thereof.
  • a cell may be in more than one of the foregoing categories.
  • antibody- mediated cell depletion methods see "Depletion of B Cells In Vivo by a Chimeric Mouse Human Monoclonal Antibody to CD20", Mitchell E. Reff, Blood, vol. 83, pp. 435-445, Jan. 15, 1994.
  • treatment with an antibody of the invention reduces the number of one or more of the above-listed cell types by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, and least 85%, at least 90%, or at least 95%. In some embodiments, treatment with an antibody of the invention reduces the number of plasma cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%.
  • treatment with an antibody of the invention reduces the number of switched memory B cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, or at least 60%. In some embodiments, treatment with an antibody of the invention reduces the number of unswitched memory B cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, or at least 60%. In some embodiments, treatment with an antibody of the invention reduces the number of double negative memory B cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, or at least 60%. In some embodiments, treatment with an antibody of the invention reduces the number of naive B cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, or at least 60%.
  • the invention also provides methods of reducing serum immunoglobulin levels comprising administering an antibody of the invention.
  • such methods reduce serum IgM levels.
  • treatment with an antibody of the invention reduces serum IgM levels by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, or at least 65%.
  • such methods reduce serum IgG levels.
  • such methods reduce the levels of one or both of lgG2 and lgG3.
  • such methods reduce the levels of lgG2 by at least 25%, at least 30%, at least 35%, at least 40%, at least 50%, at least 60%, at least 65%, or at least 70%. In some embodiments, such methods reduce the levels of lgG3 by at least 25%, at least 30%, at least 35%, at least 40%, at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, or at least 80%. In some embodiments, such methods reduce lgG2, lgG3, and IgM levels.
  • the invention also provides methods of reducing the level of at least one autoantibody comprising administering an antibody of the invention.
  • such methods reduce the level of one or more autoantibodies by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, and least 85%, at least 90%, or at least 95%.
  • the invention provides methods of treating or preventing or delaying a B-cell mediated condition disorder.
  • the method includes administering to a subject in which such treatment or prevention or delay is desired, an antibody of the invention in an amount sufficient to treat, prevent, or delay a tumorigenic or immunoregulatory condition in the subject.
  • the subject is a human.
  • the subject is a non-human mammal.
  • administration of the antibody of the invention blocks BCMA-mediated signalling in the subject, which may result in one or more of cell death, inhibition, reduction, or cessation of cell proliferation.
  • the antibodies or antibody fragments of the invention use BCMA to "target" B cell lymphomas.
  • targeting can be generalized as follows: antibodies or antibody fragments of the invention specific to the BCMA surface antigen of B cells are, e.g., injected into a subject and specifically bind to the BCMA cell surface antigen of (ostensibly) both normal and malignant B cells; this binding leads to the destruction and/or depletion of neoplastic B cells.
  • chemical agents or radioactive labels having the potential to destroy cancer cells and/or tumors can be conjugated to the antibodies or antibody fragments of the invention such that the agent is specifically "delivered" to the targeted B cells, such as, e.g., neoplastic B cells.
  • the methods of the invention comprise administering an antibody or antibody fragment that is not conjugated to a chemical agent or radioactive label. In some embodiments, the methods of the invention comprise administering an antibody or antibody fragment that is not conjugated to a cytotoxic agent.
  • B cell-related disorders include, without limitation, autoimmune diseases involving inappropriate B cell activity and B cell lymphomas.
  • B cell lymphomas include, without limitation, multiple myeloma, plasmacytoma, Hodgkins' lymphoma, follicular lymphomas, small non-cleaved cell lymphomas, endemic Burkitt's lymphoma, sporadic Burkitt's lymphoma, marginal zone lymphoma, extranodal mucosa-associated lymphoid tissue lymphoma, nodal monocytoid B cell lymphoma, splenic lymphoma, mantle cell lymphoma, large cell lymphoma, diffuse mixed cell lymphoma, immunoblastic lymphoma, primary mediastinal B cell lymphoma, pulmonary B cell angiocentric lymphoma, and small lymphocytic lymphoma.
  • the antibodies or antibody fragments of the invention may also be used to treat cancers in which the cancer cells express BCMA.
  • the B cell-related disorders additionally include B cell proliferations of uncertain malignant potential, such as, for example, lymphomatoid granulomatosis and post-transplant lymphoproliferative disorder.
  • the conditions diagnosed, treated, prevented or delayed using the antibodies or antibody fragments of the invention can additionally be an immunoregulatory disorder.
  • immunoregulatory disorders include those that are autoimmune in nature such as, for example, systemic lupus erythematosus, rheumatoid arthritis, myasthenia gravis, autoimmune hemolytic anemia, idiopathic thrombocytopenia purpura, anti-phospholipid syndrome, Chagas' disease, Grave's disease, Wegener's granulomatosis, poly-arteritis nodosa, Sjogren's syndrome, pemphigus vulgaris, scleroderma, multiple sclerosis, anti-phospholipid syndrome, ANCA associated vasculitis, Goodpasture's disease, Kawasaki disease, and rapidly progressive glomerulonephritis.
  • the antibodies or antibody fragments of the invention may also have application in plasma cell disorders such as heavy- chain disease, primary or immunocyte-associated amyloidosis, and monoclo
  • compositions and methods of treatment using the antibodies or antibody fragments of the invention can be used with any condition associated with undesired BCMA-expressing cell proliferation.
  • the antibodies of the invention may also be administered in conjunction with antibody C2B8 of US Patent 5,736,137, also known as RITUXANTM.
  • combined administration depletes or inhibits the proliferation of multiple B cell subtypes.
  • the invention further provides for the use of the antibodies of the invention to assay B cell phenotypes, such as determination of the presence, absence, or amount of a marker on the surface of a B cell or B cell subtype.
  • the antibodies of the invention may be used to measure the presence of a marker associated with SLE or another B cell-related condition on the surface of na ⁇ ve B cells, memory B cells, IgD+ memory B cells, IgD- memory B cells, or double negative memory B cells.
  • the antibodies of the invention may be used to measure the presence of a marker associated with SLE or another B cell-related condition on the surface of na ⁇ ve B cells, memory B cells, IgD+ memory B cells, IgD- memory B cells, or double negative memory B cells.
  • compositions suitable for administration typically comprise antibodies and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, diluents, and the like, compatible with pharmaceutical administration. Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incorporated herein by reference. Preferred examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin.
  • Liposomes and non-aqueous vehicles such as fixed oils may also be used.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the antibodies, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • a pharmaceutical composition comprising antibodies of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating antibodies of the invention in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the antibodies into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • methods of preparation are vacuum drying and freeze-drying that yields a powder of the antibodies plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the antibodies are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • the materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers.
  • the antibodies or antibody fragments of the invention may be used to target liposomal suspensions to B cells or subclasses thereof to which the particular antibody binds. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.
  • Parenteral compositions may be formulated in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of antibody calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved.
  • compositions comprising an antibody of the invention can be included in a container, pack, or dispenser together with instructions for administration.
  • a method of depleting na ⁇ ve B cells, memory B cells, and plasma cells comprising administering an antibody that binds to SEQ ID NO:9 and binds to na ⁇ ve B cells, memory B cells, and plasma cells.
  • a method of depleting memory B cells comprising administering an antibody that binds to SEQ ID NO:9 and binds to memory B cells.
  • a method of depleting na ⁇ ve B cells comprising administering an antibody that binds to SEQ ID NO:9 and binds to na ⁇ ve B cells.
  • an isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises at least one CDR chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
  • the antibody of embodiment 10 wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 3, and the antibody further comprises a variable domain comprising at least one of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 12, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 12, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 12.
  • the antibody of embodiment 10 wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 5, and the antibody further comprises a variable domain comprising at least one of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 6, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 6, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 6.
  • the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 7, and the antibody further comprises a variable domain comprising at least one of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 8, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 8, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 8.
  • an isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises: a. a variable domain comprising CDR1 , CDR2, and CDR3 regions identical to the CDR1 , CDR2, and CDR3 of a sequence which is chosen from SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7; or b. a variant of the variable domain of part (a) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
  • the antibody of embodiment 53 wherein the sequence is SEQ ID NO:1 and the antibody further comprises: c. a variable domain comprising a CDR1 region identical to amino acids 24-34 of SEQ ID NO:2, a CDR2 region identical to amino acids 50-56 of SEQ ID NO:2, and a CDR3 region identical to amino acids 89-97 of SEQ ID NO:2; or d. a variant of the variable domain of part (c) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
  • the antibody of embodiment 53 wherein the sequence is SEQ ID NO:3 and the antibody further comprises: c. a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 12, a CDR2 region identical to amino acids 54-60 of SEQ ID NO: 12, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO:12; or d. a variant of the variable domain of part (c) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
  • the antibody of embodiment 53 wherein the sequence is SEQ ID NO:5 and the antibody further comprises: c. a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO:6, a CDR2 region identical to amino acids 54-60 of SEQ ID NO:6, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO:6; or d. a variant of the variable domain of part (c) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
  • the antibody of embodiment 53 wherein the sequence is SEQ ID NO:7 and the antibody further comprises: c. a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO:8, a CDR2 region identical to amino acids 54-60 of SEQ ID NO:8, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO:8; or d. a variant of the variable domain of part (c) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
  • An isolated antibody that binds to the polypeptide of SEQ ID NO:9 wherein the antibody comprises at least one CDR chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8; SEQ ID NO:11 ; and SEQ ID NO:12.
  • a pharmaceutical composition comprising the antibody of any of embodiments 1-4, 7, 8, 10-21 , or 23-30 and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising the antigen binding portion, Fab fragment, or F(ab')2 fragment of embodiment 33 and a pharmaceutically acceptable carrier.
  • a method of depleting plasma cells comprising administering the antibody of any of embodiments 1-4, 8, or 10-31.
  • a method of treating a B cell-related disorder comprising administering the antibody of any one of embodiments 1-4, 8, or 10-31.
  • the B-cell related disorder is plasmacytoma, Hodgkins' lymphoma, follicular lymphomas, small non-cleaved cell lymphomas, endemic Burkitt's lymphoma, sporadic Burkitt's lymphoma, marginal zone lymphoma, extranodal mucosa-associated lymphoid tissue lymphoma , nodal monocytoid B cell lymphoma, splenic lymphoma, mantle cell lymphoma, large cell lymphoma, diffuse mixed cell lymphoma, immunoblastic lymphoma, primary mediastinal B cell lymphoma, pulmonary B cell angiocentric lymphoma, small lymphocytic lymphoma, B cell proliferations of uncertain malignant potential, lymphomatoid granulomatosis, post-transplant lymphoproliferative disorder, an immunoregulatory disorder, rheumatoid arthritis, myasthenia gravis
  • the antibody of embodiment 10 wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 1 , and the antibody further comprises a variable domain comprising at least two of a CDR1 region identical to amino acids 24-34 of SEQ ID NO:2, a CDR2 region identical to amino acids 50-56 of SEQ ID NO:2, or a CDR3 region identical to amino acids 89-97 of SEQ ID NO:2.
  • the antibody of embodiment 10 wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 3, and the antibody further comprises a variable domain comprising at least two of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 12, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 12, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 12.
  • the antibody of embodiment 10 wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 5, and the antibody further comprises a variable domain comprising at least two of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 6, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 6, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 6. 50.
  • the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:5 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:6.
  • the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:7 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:8.
  • the antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:5 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:6.
  • the antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:7 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:8.
  • the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:3 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:12.
  • the antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:5 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:6.
  • the antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:7 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:8.
  • a method of reducing serum IgM level comprising administering the antibody of any of embodiments 1-4, 8, and 10-31.
  • a method of reducing serum IgG level comprising administering the antibody of any of embodiments 1-4, 8, and 10-31.
  • a method of reducing serum lgG2 level comprising administering the antibody of any of embodiments 1-4, 8, and 10-31.
  • a method of reducing serum lgG3 level comprising administering the antibody of any of embodiments 1-4, 8, and 10-31.
  • the antibody of embodiment 87 wherein the sequence is SEQ ID NO:1 and the antibody further comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1 , CDR2, and CDR3 regions of SEQ ID NO:2.
  • the antibody of embodiment 87 wherein the sequence is SEQ ID NO:3 and the antibody further comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1 , CDR2, and CDR3 regions of SEQ ID NO:12.
  • the antibody of embodiment 87 wherein the sequence is SEQ ID NO:5 and the antibody further comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1 , CDR2, and CDR3 regions of SEQ ID NO:6.
  • the antibody of embodiment 87 wherein the sequence is SEQ ID NO:7 and the antibody further comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1, CDR2, and CDR3 regions of SEQ ID NO:8.
  • the B-cell related disorder is rheumatoid arthritis.
  • the B-cell related disorder is idiopathic thrombocytopenia purpura, or myasthenia gravis, or autoimmune hemolytic anemia.
  • Anti-BCMA monoclonal antibodies were generated by immunizing female RBF mice with BCMA-Fc/KLH conjugate protein i.p. in CFA, followed by additional immunizations at regular intervals with IFA, except that the last boost used RIBI instead of IFA, prior to splenocyte fusion to the FL653 myeloma cell line after the method of Harlow and Lane (1998), Using Antibodies: A Laboratory Manual: Portable Protocol No. I, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.
  • splenocytes isolated from a mouse 3 days after the final boost were washed twice and mixed in a 7:1 ratio with twice-washed log phase FL653 myeloma cells.
  • the cell mixture was split four ways, pelleted, and incubated in 37 0 C PEG for 1 min during which time cells were gently resuspended, followed by careful addition of 10 ml ice-cold DMEM.
  • Cells were mixed, pelleted, and resuspended in AAT hybridoma growth selection media. Cell supernatants were screened for BCMA-specific reactivity by ELISA and flow cytometry.
  • Total cellular RNA from murine hybridoma cells was prepared using a Qiagen RNeasy mini kit following the manufacturer's recommended protocol.
  • cDNAs encoding the variable regions of the heavy and light chains were cloned by RT-PCR from total cellular RNA, using random hexamers for priming of first strand cDNA.
  • a cocktail of degenerate forward primers hybridizing to multiple murine immunoglobulin gene family signal sequences and a single back primer specific for the 5' end of the murine constant domain were used.
  • PCR was performed using Clontech Advantage 2 Polymerase mix following the manufacturer's recommended protocol.
  • DHFR dihydrofolate reductase
  • CHO Chinese hamster ovary
  • the growth medium was changed to alpha minus MEM (Gibco, Rockville, MD), 10% dialyzed serum (Hyclone, Logan, UT), and 2 mM L- glutamine (Gibco, Rockville, MD), and cells were pooled and split into three T-75 tissue culture flasks and allowed to grow to confluence. Seven days post- transfection, the cells were pooled again and split into five T225 tissue culture flasks and allowed to grow to confluence.
  • the highest expressing clone was used for assessing new anti- human BCMA mAbs, with untransfected CHO cells as a control. Briefly, BCMA- CHO cells were pretreated with FACS buffer plus 5% normal mouse serum to block non-specific binding sites. Seven anti-BCMA mAbs — C11 D5.3, C12A3.2, C13F12.1 and A7D12.2 and the commercially available anti-BCMA mAbs, C4E2.2 (Legacy Biogen), VICKY-1 (Alexis) and 335004 (R&D Systems, Inc.)— and isotype control antibodies were incubated separately with cells for 30 minutes on ice at 1 ⁇ g/ml, and washed.
  • Biotin-conjugated isotype control Abs were as follows: mouse IgGI , eBioscience cat. no. 13-4714-85; mouse lgG2a, eBioscience, cat. no. 13- 4732-85; hamster IgGI , BD Pharmingen cat. no. 553970; rat lgG2a, eBioscience, cat. No. 13-4321-82.
  • Streptavidin-PE (Molecular Probes, Eugene, OR) was added to cells for 30 minutes on ice, after which cells were washed, fixed in 0.8% paraformaldehyde, and run on a FACScalibur flow cytometer (BDbiosciences, San Jose, CA) and analyzed using Flowjo software (Treestar, Ashland, OR). Results are shown in Figure 1.
  • Example 3 The seven anti-BCMA mAbs tested in Example 3 were then assessed in a cross-blocking assay to determine the presence or absence of epitope overlap between each antibody.
  • Corning 96-well flat-bottom plates were incubated overnight at 4°C with 10 ⁇ g/ml of mouse anti-human Fc in a 50 mM pH 9.6 sodium bicarbonate solution, washed, incubated with 2 ⁇ g/ml human BCMA- Fc (SEQ ID NO:10) at 37°C for 1 hr, washed again, and non-specific binding sites were blocked with blocking buffer (3% BSA in PBS) for 30 min at 37°C.
  • Antibodies were considered to bind the same epitope or very closely overlapping epitopes if they each reduced the other's binding (according to the fraction calculated as above) to below 20% of the positive control value. If they did not satisfy this condition, they were considered to have at least partially distinct epitopes. Table 3 shows which antibodies have at least partially distinct epitopes. Example 5. Flow cytometric analysis of antibody binding to human peripheral blood cells
  • PBMCs peripheral blood mononuclear cells
  • FACS buffer containing 5% normal mouse serum to block non-specific binding sites.
  • fluorphore-conjugated monoclonal antibodies directed against specific B cell and plasma cell markers were used: anti-CD19-PE-Cy5, anti-lgD-FITC, anti-CD27-APC, anti-CD38-PE-Cy7 (BD Biosciences, San Jose, CA).
  • Streptavidin-PE (Molecular Probes, Eugene, OR) was used to visualize biotin-conjugated anti-BCMA mAbs (10 ⁇ g/ml). Binding of an isotype control mAb as in Example 3 was also measured.
  • FIG. 3 shows a comparison between samples from a healthy volunteer and a representative SLE patient.
  • Antibody A7D12.2 bound to plasma cells from both healthy volunteers and SLE patients (Fig. 3A).
  • the A7D12.2 antibody bound na ⁇ ve B cells (Fig. 3E). Binding of the A7D12.2 antibody to memory B cells (Fig. 3B-D), particularly double negative memory B cells (Fig. 3D), was increased in SLE samples.
  • Example 7 Generation of cell lines producing chimeric anti-BCMA mAbs
  • CHO-DG44-I a dhfr-deficient, insulin-independent Chinese hamster ovary cell line, was used to construct anti-BCMA wild type cell lines.
  • the host cells were cultured in CHO-S-SFM Il medium with nucleosides prior to transfection.
  • Chimeric antibodies were produced by transfecting cells with expression plasmids encoding the mature heavy and light chain sequences listed in Table 4.
  • Chimeric anti-BCMA expression plasmids were transfected into the CHO host cell line DG44-I using a cationic lipid (Fugene HD) method. Briefly, 1x10 6 DG44-I cells were seeded in each of two wells of a 6-well plate containing 3 ml_ of CHO-S-SFMII medium w/nucleosides per well. Four ⁇ g of plasmid DNA (2 ⁇ g heavy chain, 2 ⁇ g light chain) was diluted into 200 ⁇ l_ CHO-S-SFM Il (Invitrogen) medium at room temperature. Sixteen ⁇ l_ of Fugene HD (Roche) reagent and allowed to complex with the DNA for approximately 15 minutes.
  • Fugene HD Fugene HD
  • CHO-DG44-I cells stably transfected with chimeric heavy and light chains were fermented in CHOM39 media, harvested, and the cells were removed by centrifugation. The pH of the cleared media was adjusted prior to passing through a protein A Fast Flow column.
  • Antibodies were eluted with 100 mM Na- citrate buffer, pH 3.0, neutralized to pH 7.0 using 10% (v/v) of 2M glycine, pH 8.9, and the recovered antibody solution was buffer-exchanged to PBS (pH 7.2) using a Superdex 200 size exclusion chromatography under endotoxin-free conditions. The purified protein was kept at -80 0 C.
  • JJN-3 human plasmacytoma cells (DSMZ ACC 541) were cultured in culture medium consisting of 40% Dulbecco's MEM, 40% Iscove's MDM, 20% FBS, 100 units/mL penicillin and 100 ⁇ g/mL streptomycin.
  • U266 human plasmacytoma cells (ATCC TIB 196) were cultured in culture medium consisting of RPMI 1640, 15% FBS, 20 mM HEPES, 100 units/mL penicillin and 100 ⁇ g/mL streptomycin. All cells were cultured at 37 0 C in a 5% CO2 atmosphere.
  • Peripheral blood mononuclear cells were isolated from a consented normal healthy donor by density centrifugation through Ficoll-Paque PLUS (GE Healthcare, Uppsala, Sweden).
  • ADCC antibody-dependent cellular cytotoxicity
  • Assay diluent was RPMI 1640, 1% BSA, 20 mM HEPES, 100 units/mL penicillin and 100 ⁇ g/mL streptomycin.
  • Human plasmacytoma cell lines JJN-3 and U266 were washed and resuspended in assay diluent to 0.4 x 10 6 cells per ml_. 50 ⁇ l_ of each cell suspension was plated into a 96-well U bottom microtiter plate in triplicates.
  • Controls included target cells alone (spontaneous LDH release), target cells alone with 2% Triton X-100 in assay diluent (maximum LDH release), effector cells with and without target cells, and human IgGI isotype control.
  • PBMCs peripheral blood mononuclear cells
  • chimeric mAbs C12A3.2 and C13F12.1 demonstrated marked ADCC activity relative to HIgGI controls.
  • Chimeric clones A7D12.2 and C11 D5.3 also mediated ADCC, although to a lesser degree than C12A3.2 and C13F12.1.
  • Table 5 ADCC of U266 cells by anti-human BCMA mAbs
  • ADCC assays were also performed using a second BCMA+ plasmacytoma cell line, JJN-3, as the target cells. As shown in Table 6, chimeric C12A3.2, C13F12.1 , and A7D12.2 also mediated ADCC of JJN-3 cells. Table 6. ADCC of JJN-3 cells by anti-human BCMA mAbs
  • NK cells were not the effector cells for chC 12A3.2, chC13F12.1 , or chA7D12.2 in the ADCC assays with U266 and JJN3 as target cells (data not shown).
  • Generation of afucosyl variants of chC12A3.2, chC13F12.1 , or chA7D12.2 did not improve activity of these mAbs (data not shown).
  • HSC/NSG mice provide a useful tool for testing in vivo biologic reagents specific for human protein targets since their immune system consists of functioning human cell types (Brehm et al. (2010), Clin Immunol [Epub ahead of print]). Humanized mice were generated as previously described (Pearson et al. (2008), Curr Protoc Immunol Chapter 15:Unit 15.21. PMID: 18491294). NOD/SCID/common gamma chain-deficient mice (NSG mice) were purchased from Jackson Laboratories (Bar Harbor, ME). Mice were maintained and bred under specific-pathogen-free conditions in isolator cages. Breeding pairs were established and dams were monitored for pregnancy and delivery.
  • Pups ranging in age from 2-6 days old were used to create humanized mice. Pups were lightly irradiated, receiving a dose of 1 Gy (100 rad) from a 137Cs irradiator. Pups were immediately injected intra-orbitally with approximately 50,000 CD34+CD3- human stem cells (HSC) derived from umbilical cord blood (All Cell LLC, purchased from StemCell Technologies Inc., Vancouver, BC, Canada), then returned to the dam. Pups were weaned at 21 days and caged with littermates according to gender. Starting at 3 months of age mice were bled via the facial vein into heparinized tubes and the whole blood was analyzed by flow cytometry for the presence of human cells.
  • HSC CD34+CD3- human stem cells
  • mice were considered successfully humanized if they had at least 20% human CD45+ cells in whole blood of which 10% or more were human CD3+, the remainder being human B cells and other human hematopoietic cells. Mice were occasionally bled and analyzed to ensure that the humanization was stable, and were routinely analyzed 2 weeks prior to study enrollment. Flow cytometry
  • PC plasma cells
  • HSC/NSG human stem cell humanized NSG mice
  • mAbs directed to human cell lineage and to human B cell markers.
  • the panel consisted of anti-human CD45, anti-human CD19, anti-human CD27, anti-human IgD, and anti-human CD38.
  • Two markers used to exclude specific cell populations were anti-mouse CD45 and anti-human CD3.
  • Cells identified as PC were human CD45+, human CD19+, human CD3-, human CD27+, human IgD- and human CD38bright .
  • BCMA+ cells the biotin-conjugated anti-human BCMA mAbs, C12A3.2 and A7D12.2, were used.
  • mice received chimeric anti-BCMA mAb i.p. Human IgGI with no known reactivity (Protos Immunoresearch) was used as a negative control. Blood was collected to prepare serum for analysis of human Ig isoform levels. At the study terminus the spleen was harvested, a single cell suspension was prepared, RBCs were lysed and cells were washed 3x with PBS/5% FCS and the cell number was determined. Cells were assessed by flow cytometry for T lineage cells, B lineage cells, and plasma cells.
  • Serum levels of human IgM and IgG were determined using an ELISA format (Bethyl Laboratories Inc., Montgomery, TX) and a human immunoglobulin isotyping kit (Millipore, Billerica, MA), respectively, according to the manufacturer's protocol. Results
  • HSC/NSG mice received various amounts of chimeric anti-human BCMA clones chAC11 D5.3, chC12A3.2, chC13F12.1 , and chA7D12.2 (Example 7) twice weekly i.p. for 2 weeks, after which the presence of splenic PCs was determined.
  • Splenic PCs from the HIgGI -treated control group were analyzed for expression of BCMA using the A7D12.2 and C12A3.2 clones and were confirmed to express cell surface BCMA (data not shown).
  • PCs were identified using the flow cytometric parameters described above, and total cell numbers were determined from the flow cytometric dot plots (calculated as a percentage of the total human cell number). The numbers of naive human B cells, unswitched memory human B cells, and switched memory human B cells were also determined.
  • Chimeric C12A3.2 resulted in a 63%, 62% and 30% reduction in IgM for the 200, 20 and 2 ⁇ g dose levels, respectively.
  • Chimeric C13F12.1 resulted in a 52%, 42% and 32% reduction in IgM for the 200, 20 and 2 ⁇ g dose levels, respectively.
  • Table 7 Serum human IgM levels ( ⁇ g/mL)
  • Table 8 Serum human immunoglobulin levels

Abstract

This invention provides antibodies that recognize the B Cell Maturation Antigen (BCMA) and that bind naϊve B cells, plasma cells, and/or memory B cells. The invention further provides methods for depleting naϊve B cells, plasma cells, and memory B cells, and for treating B cell-related disorders, including lymphomas and autoimmune diseases.

Description

ANTI-BCMA ANTIBODIES
[001] The instant application contains a Sequence Listing which has been submitted via EFS-Web and is hereby incorporated by reference in its entirety. The ASCII copy, created on March 10, 2010, is named 08201010.txt, and is 70,657 bytes in size.
[002] This invention relates to antibodies that bind to the B cell surface antigen BCMA. The invention also relates to the use of these antibodies to detect, deplete, and otherwise manipulate various B cell subtypes.
[003] B cells are lymphocytes that play major roles in adaptive humoral immunity and production of antibodies that specifically recognize antigens. Three subclasses of B cells are naϊve B cells, memory B cells, and plasma cells. The processes of VDJ recombination, in which two or three segments of DNA are chosen from a genomic library and recombined to generate a combinatorial array of antibody variable domains, and hypermutation, by which the variable domains encoded by different lineages of B cells are further varied, result in up to 109 distinct B cell lineages that produce antibodies with specificity for distinct targets. A B cell is said to be specific for an antigen that binds the antibodies made by that B cell. B cells in general are stimulated by exposure to their specific antigen (Ag). Naϊve B cells have not yet been exposed to their specific antigen. Such exposure (e.g., during an infection) results in proliferation of B cells and generation of sister clones. Sister clones can develop into plasma cells, which produce high amounts of antibody. Plasma cells may either be short lived, or may migrate into bone marrow, where they can persist for an extended period of time. A sister clone of an Ag-exposed B cell may also develop into a memory B cell that is quiescent until reexposed to the specific antigen. Memory B cells respond rapidly to reexposure to antigen by dividing to produce both plasma cells and additional memory B cells. Memory B cells include switched memory B cells (CD19+CD27highCD38low lgD"), unswitched memory B cells (CD19+CD27hi9hCD38l0W IgD+), and double negative memory B cells (CD19+CD27" CD38l0W lgD-).
[004] Several significant diseases involve B cells. Malignant transformation of B cells leads to cancers, including some lymphomas such as, for example, multiple myeloma and Hodgkins' Lymphoma. Some autoimmune diseases, including systemic lupus erythematosus (SLE), also involve B cells. Both cancer and autoimmune diseases that involve B cells may be considered gain of function conditions, in that the B cells overgrow and/or attack parts of the body inappropriately. A possible strategy to control such diseases is to use antibodies that target the pathological B cells.
[005] The B cell maturation antigen (BCMA, also known as TNFRSF17 and CD269) is a protein that has been shown to be expressed on the surface of plasmablasts (i.e., plasma cell precursors) and plasma cells, and is believed to stimulate survival. It therefore represents a potential target for B cell-related diseases. BCMA is a member of the TNF receptor family and binds the TNF family ligands BAFF and APRIL (reviewed in Kalled et al. (2005), Curr Dir Autoimmun 8:206-242). BCMA is a type III membrane protein, as it lacks the signal peptide associated with type I membrane proteins found in most TNF receptor family members.
[006] BCMA RNA has been detected in the spleen, lymph nodes, thymus, adrenals and liver, and analysis of a number of B cell lines indicated that BCMA mRNA levels increased upon maturation. Human BCMA protein has been detected on various subtypes of CD38+ B cells, particularly plasma cells (Zhang et al. (2005), lnt Immunol 17:779-788; Darce et al. (2007), J Immunol 179:7276- 7286; Sims et al. (2005), Blood 105:4390-4398; Avery et al. (2003), J Clin Invest 112:286-297). Independent laboratories have examined blood and/or tonsil B cell subsets and found that BCMA expression could not be detected on naϊve or memory B cells (Zhang et al. (2005), lnt Immunol 17:779-788; Darce et al. (2007), J Immunol 179:7276-7286; Chiu et al. (2007), Blood 109:729-739). Attempts to detect BCMA protein on the surface of germinal center B cells have had inconsistent results (Zhang et al. (2005), lnt Immunol 17:779-788; Chiu et al. (2007), Blood 109:729-739).
[007] The mechanism of action of BCMA is not fully understood. Mice that have been genetically altered to lack a functional gene for BCMA have normal lymphoid organs and cell populations, and a nearly normal functioning immune system (Xu and Lam (2001), MoI Cell Biol 21:4067-4074; Schiemann et al. (2001), Science 293:2111-2114). The only defect defined to date in these mice is a diminished survival of long-lived bone marrow (BM) plasma cells (O'Connor et al. (2004), J Exp Med 199:91-98). Therefore, it may be that BCMA, at least in the murine system, provides a survival signal to BM-resident plasma cells that is either BAFF or APRIL-mediated, or both. Indeed, signalling through BCMA activates the NF-κB pathway (Hatzoglou et al. (2000), J Immunol 165:1322-1330) which is involved in B cell survival, proliferation and maturation (Litinskiy et al. (2002) Nat Immunol 3:822-829; Pomerantz and Baltimore (2002) MoI Cell 10:693- 695; Huang et al. (2004) Proc Natl Acad Sci U S A 101 :17789-17794; He et al. (2004) J Immunol 172:3268-3279). Results with malignant human cells are generally consistent with a link between BCMA and cell survival. Primary multiple myeloma (MM) cells, MM cell lines (Novak et al. (2004) Blood 103:689-694), and Hodgkin and Reed-Stemberg (HRS) cells from Hodgkin lymphomas (Chiu et al. (2007), Blood 109:729-739; Novak et al. (2004), Blood 104:2247-2253) have been shown to express BCMA. Addition of BAFF and/or APRIL has further been shown to provide a survival signal for these malignant cells, although it is not clear that BCMA is predominantly responsible for this effect.
[008] Because different B cell subsets are implicated in different B cell related conditions, there exists a need for agents that specifically target one or more B cell subsets. The expression of BCMA on the surface of some B cells provides a marker by which those cells may be specifically targeted. To take advantage of BCMA as a marker of one or more B cell subsets, there is a need for agents that specifically bind to BCMA and for a determination of which B cell subsets are bound by those BCMA-specific agents. The invention provides antibodies that specifically bind to BCMA. The antibodies of the invention may be used to target one or more of the following B cell subsets: plasma cells, memory B cells, and naϊve B cells. BRIEF DESCRIPTION OF THE DRAWINGS
[009] Figure 1 depicts binding of anti-human BCMA mAbs on BCMA- transfected CHO cells. Binding of biotin-conjugated anti-human BCMA mAbs, visualized with Streptavidin-PE, was measured by flow cytometry on a BCMA- CHO stable cell line (A) and control, non-transfected CHO cells (B). The shaded area represents staining of cells with an isotype control mAb.
[010] Figure 2 depicts anti-BCMA binding to B cell subsets. B cell subsets in human peripheral blood were assessed by flow cytometry for reactivity to anti-BCMA mAbs. Visualization was as in Figure 1. The shaded area represents staining with an isotype control Ab. B cell subsets were plasma cells (CD19+CD27hιghCD38hιghlgD") (A), switched memory B cells (CD19+CD27highCD38l0W lgD-) (B), unswitched memory B cells (CD19+CD27highCD38l0W IgD+) (C), double negative memory B cells (CD19+CD27~ CD38l0W lgD") (D), and naive B cells (CD19+CD271gD+) (E).
[011] Figure 3 depicts anti-BCMA binding to B cell subsets isolated from healthy and SLE-afflicted individuals. B cell subsets in human peripheral blood from a healthy volunteer and an SLE patient were assessed by flow cytometry for reactivity to the anti-BCMA mAbs C12A3.2 and A7D12.2. B cell subsets were as in Figure 2. Visualization was as in Figure 1.
[012] Figure 4 depicts flow cytometric staining of human plasma cells within the human CD45+ splenocyte compartment isolated from HSC/NSG mice. Splenic plasma cells were stained with anti-BCMA antibodies A7D12.2 (left panel; bold line) and C12A3.2 (right panel; bold line) or an isoytpe control mouse lgG2b or IgGI Ab, respectively (thin line in both panels).
[013] Figure 5 depicts flow cytometric staining for plasma cells (PCs) within the human CD45+ splenocyte compartment isolated from HSC/NSG mice treated with anti-BCMA antibody (chC12A3.2 or chC13F12.1) or human IgGI control. Mice were injected i.p. with anti-BCMA Ab or HIgGI control twice weekly for 2 weeks. ** p<0.0001 ; * p=0.0066.
[014] Figure 6 depicts flow cytometric staining for plasma cells (PCs) within the human CD45+ splenocyte compartment isolated from HSC/NSG mice treated with anti-BCMA antibody (chC11 D5.1 or chA7D12.2) or human IgGI control. Mice were injected i.p. with anti-BCMA Ab or HIgGI control twice weekly for 2 weeks.
Figure imgf000007_0001
45 huC13F12.1 H2 humanized mature heavy chain variable domain sequence
46 huC13F12.1 H3 humanized mature heavy chain variable domain sequence
47 huC13F12.1 H4 humanized mature heavy chain variable domain sequence
DETAILED DESCRIPTION OF THE EMBODIMENTS
[015] The invention provides antibodies that bind to BCMA and certain epitopes thereof. In some embodiments, the antibodies of the invention bind to one or more subsets of B cells, such as plasma cells, memory B cells, and naϊve B cells. The invention also provides methods of depleting B cells or subclasses of B cells, including plasma cells, memory B cells, and naϊve B cells. In one embodiment, the invention provides an isolated antibody that binds to SEQ ID NO:9 and binds to plasma cells. In one embodiment, the invention provides an isolated antibody that binds to SEQ ID NO:9 and binds to memory B cells. In another embodiment, the invention provides an isolated antibody that binds to SEQ ID NO:9 and binds to naϊve B cells.
[016] Certain anti-BCMA mAbs, including clone C4E2.2 (hamster IgG) generated at Legacy Biogen (6); clone VICKY-1 (rat IgGI) (Alexis Biochemicals, Lausen, Switzerland, also sold as 6D10 by Santa Cruz Biotechnology, Santa Cruz, CA); and clone 335004 (rat lgG2a) (R&D Systems, Inc., Minneapolis, MN), are outside the scope of this invention.
A. Antibodies
[017] The invention provides antibodies that bind specifically to SEQ ID NO:9. The invention also provides to antibodies that bind to the surface of B cells or subclasses thereof, including plasma cells, memory B cells (including switched, unswitched, and double negative), and/or naϊve B cells. The term "antibody" as used herein, includes both full-length immunoglobulins and antibody fragments that bind to the same antigens. The antibodies can be, e.g., a monoclonal, polyclonal, chimeric, humanized, or single chain antibody. In some embodiments, the antibody fragments are Fab fragments or F(ab')2 fragments and retain the ability to specifically bind the protein of SEQ ID NO: 9.
[018] In part, the invention provides the antibodies A7D12.2, C11 D5.3, C12A3.2, and C13F12.1. Each of these is a murine monoclonal antibody. A7D12.2 has a murine "miscellaneous" subgroup heavy chain, a heavy chain variable domain sequence that is SEQ ID NO:1 , a subgroup I kappa light chain, and a light chain variable domain sequence that is SEQ ID NO:2.
[019] C11 D5.3 has a subgroup H(A) heavy chain, a heavy chain variable domain sequence that is SEQ ID NO:3, a murine subgroup III kappa light chain, and a light chain variable domain sequence that is selected from SEQ ID NO:4, SEQ ID NO:11 , and SEQ ID NO:12. In some embodiments, the light chain variable domain sequence of C11 D5.3 is SEQ ID NO: 12.
[020] C12A3.2 has a subgroup H(A) heavy chain, a heavy chain variable domain sequence that is SEQ ID NO:5, a murine subgroup III kappa light chain, and a light chain variable domain sequence that is SEQ ID NO:6.
[021] C13F12.1 has a subgroup II(A) heavy chain, a heavy chain variable domain sequence that is SEQ ID NO:7, a murine subgroup III kappa light chain, and a light chain variable domain sequence that is SEQ ID NO:8.
[022] Techniques for producing single-chain antibodies specific to the protein of SEQ ID NO: 9 can be adapted from e.g., those described in U.S. Patent 4,946,778. In addition, methods can be adapted for the construction of Fab expression libraries (see e.g., Huse et al. (1989) Science 246:1275-1281) to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for a BCMA protein or derivatives, fragments, analogs or homologs thereof. Numerous techniques for humanizing non-human antibodies are well known in the art. See e.g., U.S. Patent 5,225,539, 6,632,927, or 5,648,237, all of which are incorporated by reference. Antibody fragments that contain the idiotypes to a BCMA protein may be produced by any of a variety of techniques, including, but not limited to: (i) an F(ab')2 fragment produced by pepsin digestion of an antibody molecule; (ii) an Fab fragment generated by reducing the disulfide bridges of an F(ab')2 fragment; (iii) a Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent and (iv) Fv fragments.
[023] Additionally, recombinant anti-BCMA antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention. Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques such as, for example, the methods described in U.S. Patent No. 7,112,421 ; Better et al. (1988) Science 240:1041-1043; or Liu et ai. (1987) Proc. Natl. Acad. Sci. USA 84:3439- 3443.
[024] Some of the antibodies of the invention are chimeric forms of murine monoclonal antibodies A7D12.2, C11 D5.3, C12A3.2, and C13F12.1. In some embodiments, a chimeric form of A7D12.2 comprises a heavy chain comprising SEQ ID NO:13 and a light chain comprising SEQ ID NO:14. In some embodiments, a chimeric form of C11 D5.3 comprises a heavy chain comprising SEQ ID NO:15 and a light chain comprising a sequence selected from SEQ ID NO: 16 and SEQ ID NO: 17, preferably SEQ ID NO: 17. In some embodiments, a chimeric form of C12A3.2 comprises a heavy chain comprising SEQ ID NO:18 and a light chain comprising SEQ ID NO: 19. In some embodiments, a chimeric form of C13F12.1 comprises a heavy chain comprising SEQ ID NO:20 and a light chain comprising SEQ ID NO:21.
[025] Some of the antibodies of the invention are humanized forms of murine monoclonal antibodies A7D12.2, C11D5.3, C12A3.2, and C13F12.1. In some embodiments, a humanized form of C11 D5.3 comprises a light chain variable domain comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 22-24 and a heavy chain variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 25-29. In some embodiments, a humanized form of C12A3.2 comprises a light chain variable domain comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 30-33 and a heavy chain variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 34-38. In some embodiments, a humanized form of C13F12.1 comprises a light chain variable domain comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 39-42 and a heavy chain variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to a sequence selected from SEQ ID NOs 43-47.
B. Antibody variable domain sequence
[026] The antibodies of the invention may comprise the heavy chain variable domain sequences of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7. The heavy chain variable domain sequences may consist essentially of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7.
[027] The antibodies of the invention may comprise the light chain variable domain sequences of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO:12. The light chain variable domain sequences may consist essentially of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO:12.
[028] The invention also provides a variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to a sequence selected from SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7. The invention also provides a variable domain sequence comprising a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to a sequence selected from SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , and SEQ ID NO:12. The invention also provides antibodies comprising a heavy chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:1 and a light chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:2. The invention includes antibodies comprising a heavy chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:3 and a light chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO: 4, SEQ ID NO:11 , or SEQ ID NO: 12. The invention includes antibodies comprising a heavy chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:5 and a light chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:6. The invention includes antibodies comprising a heavy chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:7 and a light chain variable domain sequence that is at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO:8. [029] The invention also provides antibodies with particular complementarity determining regions (CDR). Table 2 defines the amino acid coordinates of CDR1 , CDR2, and CDR3 of SEQ ID NOs:1 through 8, 11 , and 12. Table 2. CDR Amino Acid Coordinates
Figure imgf000013_0001
[030] CDRs are designated using the Kabat definitions (Johnson and Wu (2000), Nucleic Acids Res 28:214-218). As used herein, the "corresponding CDR" means the CDR in the most similar position within the variable domain amino acid sequence.
[031] The heavy chain variable domain of antibodies of the invention may comprise CDRs such that one, two, or three of the CDRs are identical to the corresponding CDRs of SEQ ID NO:1 ; identical to the corresponding CDRs of SEQ ID NO:3; identical to the corresponding CDRs of SEQ ID NO:5; or identical to the corresponding CDRs of SEQ ID NO:7. The light chain variable domain of antibodies of the invention may comprise CDRs such that one, two, or three of the CDRs are identical to the corresponding CDRs of SEQ ID NO:2; identical to the corresponding CDRs of SEQ ID NO:4; identical to the corresponding CDRs of SEQ ID NO:6; identical to the corresponding CDRs of SEQ ID NO:8; identical to the corresponding CDRs of SEQ ID NO:11 ; or identical to the corresponding CDRs of SEQ ID NO:12.
[032] The heavy chain variable domain may comprise CDRs identical to each of the corresponding CDRs of one of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7 except that one or more amino acid substitutions have been made in said CDR regions. In certain embodiments, CDRs of the heavy chain variable domain may have up to a total of 12 amino acid substitutions relative to SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7. In other embodiments, the heavy chain variable domain CDRs of the antibodies of the invention may have up to 10, up to 8, up to 5, or up to 3 substitutions relative to SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7. In some embodiments, the heavy chain variable domain CDRs of the antibodies of the invention are at least 80%, at least 85%, at least 90%, or at least 95% identical to the heavy chain variable domain CDRs of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7.
[033] In some embodiments, CDR2 of SEQ ID NO:7 is replaced by CDR2 (i.e., amino acids 50-66) of SEQ ID NO:46. For example, the heavy chain variable domain of an antibody of the invention may comprise CDR1 and CDR3 of SEQ ID NO:7 and CDR2 of SEQ ID NO:46. The heavy chain variable domain of an antibody of the invention may also comprise CDR1 , CDR2, and CDR3 regions that are together at least 80%, at least 85%, at least 90%, or at least 95% identical to CDR1 and CDR3 of SEQ ID NO:7 and CDR2 of SEQ ID NO:46. [034] The light chain variable domain may comprise CDRs identical to the corresponding CDRs of one of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO:12, except for one or more amino acid substitutions in said CDR regions. In certain embodiments, the antibodies of the invention comprise CDRs that are identical to the corresponding CDRs of one Of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO: 12, except for up to 12, up to 10, up to 8, up to 5, or up to 3 amino acid substitutions in said CDR regions. In some embodiments, the light chain variable domain CDRs of the antibodies of the invention are at least 80%, at least 85%, at least 90%, or at least 95% identical to the light chain variable domain CDRs of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:11 , or SEQ ID NO:12.
[035] In some embodiments, the substitutions in the CDR regions are conservative substitutions.
C. Epitopes; antibody binding specificity
[036] The invention also provides antibodies that bind particular epitopes. Whether a pair of antibodies binds the same epitope is determined based on cross-blocking experiments, as described in Example 4. Cross-blocking profiles are defined for seven antibodies in Table 3. For the purposes of this disclosure, two antibodies are considered to bind to the same epitope if each one reduces the other's binding to BCMA (i.e., they mutually cross-block) by at least 90% according to the procedure described in Example 4. Similarly, antibodies that do not reduce each other's binding by at least 90% as described in Example 4 are considered to bind to distinct epitopes. The cross-blocking profiles of antibodies with certain variable domains are listed in Table 3. Pairs of antibodies that bind to distinct epitopes (as defined above) are noted with a "d."
Figure imgf000016_0001
[037] The invention further encompasses antibodies that bind to the same epitope as antibodies A7D12.2, C11 D5.3, C12A3.2, or C13F12.1. The invention also provides antibodies that have cross-blocking profiles that match the profiles of A7D12.2, C11 D5.3, C12A3.2, or C13F12.1. For example, following the definitions provided above, an antibody that has the same profile as C11 D5.3 binds to the same epitope as compared to C12A3.2 and C13F12.1 but binds to a distinct epitope as compared to A7D12.2, 335004, C4E2, and Vicky-1. In some embodiments, the antibodies of the invention mutually cross-block one or more of A7D12.2, C11 D5.3, C12A3.2, or C13F12.1 from binding the protein of SEQ ID NO: 9 by at least 80%, 85%, 90%, or 95%. The extent of cross-blocking is measured according to the procedure described in Example 4.
[038] In some embodiments, the antibodies and antibody fragments of the invention bind to the extracellular domain of BCMA. In particular embodiments, the antibodies bind to amino acids 1-52, 1-51 , 1-41 , or 8-41 of SEQ ID NO:9. [039] The invention also provides anti-BCMA antibodies that bind to one or more particular types of cell. The antibodies or antibody fragments of the invention may bind one or more of the following: plasma cells, memory B cells, naϊve B cells, or cells that express BCMA (SEQ ID NO:9), a protein similar thereto, the extracellular domain thereof, or a polypeptide similar to the extracellular domain thereof.
D. Methods
[040] The invention provides methods of depleting various types of cells. The methods comprise administering the antibodies of the invention, as described above. Types of cells that may be depleted by the methods of the invention include, without limitation, plasma cells, naϊve B cells, memory B cells (including switched, unswitched, and double negative), lymphoma cells derived from B cells, and cells that express BCMA, a protein similar thereto, the extracellular domain thereof, or a polypeptide similar to the extracellular domain thereof. A cell may be in more than one of the foregoing categories. For an example of antibody- mediated cell depletion methods, see "Depletion of B Cells In Vivo by a Chimeric Mouse Human Monoclonal Antibody to CD20", Mitchell E. Reff, Blood, vol. 83, pp. 435-445, Jan. 15, 1994.
[041] In some embodiments, treatment with an antibody of the invention reduces the number of one or more of the above-listed cell types by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, and least 85%, at least 90%, or at least 95%. In some embodiments, treatment with an antibody of the invention reduces the number of plasma cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%. In some embodiments, treatment with an antibody of the invention reduces the number of switched memory B cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, or at least 60%. In some embodiments, treatment with an antibody of the invention reduces the number of unswitched memory B cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, or at least 60%. In some embodiments, treatment with an antibody of the invention reduces the number of double negative memory B cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, or at least 60%. In some embodiments, treatment with an antibody of the invention reduces the number of naive B cells by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, or at least 60%.
[042] The invention also provides methods of reducing serum immunoglobulin levels comprising administering an antibody of the invention. In some embodiments, such methods reduce serum IgM levels. In particular embodiments, treatment with an antibody of the invention reduces serum IgM levels by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, or at least 65%. In some embodiments, such methods reduce serum IgG levels. In some embodiments, such methods reduce the levels of one or both of lgG2 and lgG3. In some embodiments, such methods reduce the levels of lgG2 by at least 25%, at least 30%, at least 35%, at least 40%, at least 50%, at least 60%, at least 65%, or at least 70%. In some embodiments, such methods reduce the levels of lgG3 by at least 25%, at least 30%, at least 35%, at least 40%, at least 50%, at least 60%, at least 65%, at least 70%, at least 75%, or at least 80%. In some embodiments, such methods reduce lgG2, lgG3, and IgM levels.
[043] The invention also provides methods of reducing the level of at least one autoantibody comprising administering an antibody of the invention. In some embodiments, such methods reduce the level of one or more autoantibodies by at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, and least 85%, at least 90%, or at least 95%.
[044] In a still further aspect, the invention provides methods of treating or preventing or delaying a B-cell mediated condition disorder. The method includes administering to a subject in which such treatment or prevention or delay is desired, an antibody of the invention in an amount sufficient to treat, prevent, or delay a tumorigenic or immunoregulatory condition in the subject. In some embodiments, the subject is a human. In other embodiments, the subject is a non-human mammal. In some embodiments, administration of the antibody of the invention blocks BCMA-mediated signalling in the subject, which may result in one or more of cell death, inhibition, reduction, or cessation of cell proliferation.
[045] In some embodiments, the antibodies or antibody fragments of the invention use BCMA to "target" B cell lymphomas. In essence, such targeting can be generalized as follows: antibodies or antibody fragments of the invention specific to the BCMA surface antigen of B cells are, e.g., injected into a subject and specifically bind to the BCMA cell surface antigen of (ostensibly) both normal and malignant B cells; this binding leads to the destruction and/or depletion of neoplastic B cells. Additionally, chemical agents or radioactive labels having the potential to destroy cancer cells and/or tumors can be conjugated to the antibodies or antibody fragments of the invention such that the agent is specifically "delivered" to the targeted B cells, such as, e.g., neoplastic B cells. In some embodiments, the methods of the invention comprise administering an antibody or antibody fragment that is not conjugated to a chemical agent or radioactive label. In some embodiments, the methods of the invention comprise administering an antibody or antibody fragment that is not conjugated to a cytotoxic agent.
[046] B cell-related disorders include, without limitation, autoimmune diseases involving inappropriate B cell activity and B cell lymphomas. B cell lymphomas include, without limitation, multiple myeloma, plasmacytoma, Hodgkins' lymphoma, follicular lymphomas, small non-cleaved cell lymphomas, endemic Burkitt's lymphoma, sporadic Burkitt's lymphoma, marginal zone lymphoma, extranodal mucosa-associated lymphoid tissue lymphoma, nodal monocytoid B cell lymphoma, splenic lymphoma, mantle cell lymphoma, large cell lymphoma, diffuse mixed cell lymphoma, immunoblastic lymphoma, primary mediastinal B cell lymphoma, pulmonary B cell angiocentric lymphoma, and small lymphocytic lymphoma. The antibodies or antibody fragments of the invention may also be used to treat cancers in which the cancer cells express BCMA. The B cell-related disorders additionally include B cell proliferations of uncertain malignant potential, such as, for example, lymphomatoid granulomatosis and post-transplant lymphoproliferative disorder.
[047] The conditions diagnosed, treated, prevented or delayed using the antibodies or antibody fragments of the invention can additionally be an immunoregulatory disorder. These disorders include those that are autoimmune in nature such as, for example, systemic lupus erythematosus, rheumatoid arthritis, myasthenia gravis, autoimmune hemolytic anemia, idiopathic thrombocytopenia purpura, anti-phospholipid syndrome, Chagas' disease, Grave's disease, Wegener's granulomatosis, poly-arteritis nodosa, Sjogren's syndrome, pemphigus vulgaris, scleroderma, multiple sclerosis, anti-phospholipid syndrome, ANCA associated vasculitis, Goodpasture's disease, Kawasaki disease, and rapidly progressive glomerulonephritis. The antibodies or antibody fragments of the invention may also have application in plasma cell disorders such as heavy- chain disease, primary or immunocyte-associated amyloidosis, and monoclonal gammopathy of undetermined significance (MGUS).
[048] Compositions and methods of treatment using the antibodies or antibody fragments of the invention can be used with any condition associated with undesired BCMA-expressing cell proliferation.
[049] The antibodies of the invention may also be administered in conjunction with antibody C2B8 of US Patent 5,736,137, also known as RITUXAN™. In some embodiments, such combined administration depletes or inhibits the proliferation of multiple B cell subtypes.
[050] The invention further provides for the use of the antibodies of the invention to assay B cell phenotypes, such as determination of the presence, absence, or amount of a marker on the surface of a B cell or B cell subtype. For example, the antibodies of the invention may be used to measure the presence of a marker associated with SLE or another B cell-related condition on the surface of naϊve B cells, memory B cells, IgD+ memory B cells, IgD- memory B cells, or double negative memory B cells. E. Pharmaceutical Compositions
[051] The antibodies of the invention can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically comprise antibodies and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, diluents, and the like, compatible with pharmaceutical administration. Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incorporated herein by reference. Preferred examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the antibodies, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
[052] A pharmaceutical composition comprising antibodies of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
[053] Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
[054] Sterile injectable solutions can be prepared by incorporating antibodies of the invention in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the antibodies into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the antibodies plus any additional desired ingredient from a previously sterile-filtered solution thereof.
[055] In one embodiment, the antibodies are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. Additionally, the antibodies or antibody fragments of the invention may be used to target liposomal suspensions to B cells or subclasses thereof to which the particular antibody binds. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811. [056] Parenteral compositions may be formulated in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of antibody calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved.
[057] The pharmaceutical compositions comprising an antibody of the invention can be included in a container, pack, or dispenser together with instructions for administration.
F. Listing of Exemplary Embodiments:
1. An isolated antibody that binds to SEQ ID NO:9 and binds to memory B cells.
2. The antibody of embodiment 1 , wherein the antibody also binds to plasma cells.
3. The antibody of embodiment 1 , wherein the antibody also binds to naive B cells.
4. The antibody of embodiment 1 , wherein the antibody also binds to naive B cells and plasma cells.
5. A method of depleting naϊve B cells, memory B cells, and plasma cells, comprising administering an antibody that binds to SEQ ID NO:9 and binds to naϊve B cells, memory B cells, and plasma cells.
6. A method of depleting memory B cells, comprising administering an antibody that binds to SEQ ID NO:9 and binds to memory B cells. 7. An isolated antibody that binds to SEQ ID NO:9 and binds to naϊve B cells.
8. The antibody of embodiment 7, wherein the antibody binds to plasma cells.
9. A method of depleting naϊve B cells, comprising administering an antibody that binds to SEQ ID NO:9 and binds to naϊve B cells.
10. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises at least one CDR chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
11. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
12. The antibody of embodiment 10, wherein the antibody comprises the CDRs at CDR1 , CDR2, and CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
13. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 1 , and the antibody further comprises a variable domain comprising at least one of a CDR1 region identical to amino acids 24-34 of SEQ ID NO:2, a CDR2 region identical to amino acids 50-56 of SEQ ID NO:2, or a CDR3 region identical to amino acids 89-97 of SEQ ID NO:2.
14. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 3, and the antibody further comprises a variable domain comprising at least one of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 12, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 12, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 12.
15. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 5, and the antibody further comprises a variable domain comprising at least one of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 6, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 6, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 6.
16. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 7, and the antibody further comprises a variable domain comprising at least one of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 8, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 8, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 8.
17. The antibody of embodiment 10, wherein the heavy chain variable domain comprises SEQ ID NO:1 and the light chain variable domain comprises SEQ ID NO:2.
18. The antibody of embodiment 10, wherein the heavy chain variable domain comprises SEQ ID NO:3 and the light chain variable domain comprises SEQ ID NO:12.
19. The antibody of embodiment 10, wherein the heavy chain variable domain comprises SEQ ID NO:5 and the light chain variable domain comprises SEQ ID NO:6. 20. The antibody of embodiment 10, wherein the heavy chain variable domain comprises SEQ ID NO:7 and the light chain variable domain comprises SEQ ID NO:8.
21. An isolated antibody that binds the same epitope as the antibody of embodiment 17.
22. An isolated antibody that binds the same epitope as the antibody of any of embodiments 18-20.
23. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises: a. a variable domain comprising CDR1 , CDR2, and CDR3 regions identical to the CDR1 , CDR2, and CDR3 of a sequence which is chosen from SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7; or b. a variant of the variable domain of part (a) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
24. The antibody of embodiment 53, wherein the sequence is SEQ ID NO:1 and the antibody further comprises: c. a variable domain comprising a CDR1 region identical to amino acids 24-34 of SEQ ID NO:2, a CDR2 region identical to amino acids 50-56 of SEQ ID NO:2, and a CDR3 region identical to amino acids 89-97 of SEQ ID NO:2; or d. a variant of the variable domain of part (c) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
25. The antibody of embodiment 53, wherein the sequence is SEQ ID NO:3 and the antibody further comprises: c. a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 12, a CDR2 region identical to amino acids 54-60 of SEQ ID NO: 12, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO:12; or d. a variant of the variable domain of part (c) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
26. The antibody of embodiment 53, wherein the sequence is SEQ ID NO:5 and the antibody further comprises: c. a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO:6, a CDR2 region identical to amino acids 54-60 of SEQ ID NO:6, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO:6; or d. a variant of the variable domain of part (c) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
27. The antibody of embodiment 53, wherein the sequence is SEQ ID NO:7 and the antibody further comprises: c. a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO:8, a CDR2 region identical to amino acids 54-60 of SEQ ID NO:8, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO:8; or d. a variant of the variable domain of part (c) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
28. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises at least one CDR chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8; SEQ ID NO:11 ; and SEQ ID NO:12.
29. The antibody of embodiment 28, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:12, SEQ ID NO:6, and SEQ ID NO:8.
30. The antibody of embodiment 28, wherein the antibody comprises the CDRs at CDR1 , CDR2, and CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:12, SEQ ID NO:6, or SEQ ID NO:8. 31. The antibody of any of embodiments 1-4, 7, 8, 10-21 , or 23-30, wherein the antibody is a chimeric, humanized, or single chain antibody.
32. A hybridoma that produces the antibody of any of embodiments 1-4, 7, 8, 10-21 , or 23-30.
33. A pharmaceutical composition comprising the antibody of any of embodiments 1-4, 7, 8, 10-21 , or 23-30 and a pharmaceutically acceptable carrier.
34. A polypeptide that binds to SEQ ID NO:9 and comprises the antigen binding portion, Fab fragment, or F(ab')2 fragment of the antibody of any of embodiments 1-4, 7, 8, 10-21 , or 23-30.
35. A hybridoma that produces the antigen binding portion, Fab fragment, or F(ab')2 fragment of embodiment 33.
36. A pharmaceutical composition comprising the antigen binding portion, Fab fragment, or F(ab')2 fragment of embodiment 33 and a pharmaceutically acceptable carrier.
37. A method of depleting plasma cells, comprising administering the antibody of any of embodiments 1-4, 8, or 10-31.
38. A method of treating a B cell-related disorder, comprising administering the antibody of any one of embodiments 1-4, 8, or 10-31.
39. The method of embodiment 38, wherein the B-cell related disorder is plasmacytoma, Hodgkins' lymphoma, follicular lymphomas, small non-cleaved cell lymphomas, endemic Burkitt's lymphoma, sporadic Burkitt's lymphoma, marginal zone lymphoma, extranodal mucosa-associated lymphoid tissue lymphoma , nodal monocytoid B cell lymphoma, splenic lymphoma, mantle cell lymphoma, large cell lymphoma, diffuse mixed cell lymphoma, immunoblastic lymphoma, primary mediastinal B cell lymphoma, pulmonary B cell angiocentric lymphoma, small lymphocytic lymphoma, B cell proliferations of uncertain malignant potential, lymphomatoid granulomatosis, post-transplant lymphoproliferative disorder, an immunoregulatory disorder, rheumatoid arthritis, myasthenia gravis, idiopathic thrombocytopenia purpura, anti-phospholipid syndrome, Chagas' disease, Grave's disease, Wegener's granulomatosis, poly-arteritis nodosa, Sjogren's syndrome, pemphigus vulgaris, scleroderma, multiple sclerosis, anti-phospholipid syndrome, ANCA associated vasculitis, Goodpasture's disease, Kawasaki disease, autoimmune hemolytic anemia, and rapidly progressive glomerulonephritis, heavy-chain disease, primary or immunocyte-associated amyloidosis, or monoclonal gammopathy of undetermined significance.
40. The method of embodiment 38, wherein the B cell-related disorder is a B cell malignancy.
41. The method of embodiment 38, wherein the B cell-related disorder is a plasma cell malignancy.
42. The method of embodiment 41 , wherein the plasma cell malignancy is multiple myeloma.
43. The method of embodiment 38, wherein the B cell-related disorder is an autoimmune disease.
44. The method of embodiment 43, wherein the autoimmune disease is systemic lupus erythematosus.
45. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 1 , and the antibody further comprises a variable domain comprising at least two of a CDR1 region identical to amino acids 24-34 of SEQ ID NO:2, a CDR2 region identical to amino acids 50-56 of SEQ ID NO:2, or a CDR3 region identical to amino acids 89-97 of SEQ ID NO:2.
46. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 1 , and the antibody further comprises a variable domain comprising a CDR1 region identical to amino acids 24-34 of SEQ ID NO:2, a CDR2 region identical to amino acids 50-56 of SEQ ID NO:2, and a CDR3 region identical to amino acids 89-97 of SEQ ID NO:2.
47. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 3, and the antibody further comprises a variable domain comprising at least two of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 12, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 12, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 12.
48. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 3, and the antibody further comprises a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 12, a CDR2 region identical to amino acids 54-60 of SEQ ID NO: 12, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 12.
49. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 5, and the antibody further comprises a variable domain comprising at least two of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 6, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 6, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 6. 50. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 5, and the antibody further comprises a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 6, a CDR2 region identical to amino acids 54-60 of SEQ ID NO: 6, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 6.
51. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 7, and the antibody further comprises a variable domain comprising at least two of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 8, a CDR2 region identical to amino acids 54- 60 of SEQ ID NO: 8, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 8.
52. The antibody of embodiment 10, wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 7, and the antibody further comprises a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 8, a CDR2 region identical to amino acids 54-60 of SEQ ID NO: 8, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 8.
53. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:1 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:2.
54. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:3 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:12.
55. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:5 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:6. 56. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:7 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:8.
57. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, and CDR3 of SEQ ID NO:1 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:2.
58. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID NO:3 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:12.
59. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID NO:5 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:6.
60. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID NO:7 and at least one CDR chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:8.
61. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:1 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:2.
62. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:3 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:12.
63. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:5 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:6. 64. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:7 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:8.
65. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, and CDR3 of SEQ ID NO:1 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:2.
66. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID NO:3 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:12.
67. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID NO:5 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:6.
68. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID NO:7 and at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID No:8.
69. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:1 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:2.
70. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:3 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:12.
71. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:5 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:6. 72. The antibody of embodiment 10, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of SEQ ID NO:7 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:8.
73. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, and CDR3 of SEQ ID NO:1 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:2.
74. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID NO:3 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:12.
75. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID NO:5 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:6.
76. The antibody of embodiment 10, wherein the antibody comprises CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID NO:7 and CDRs that are CDR1 , CDR2, or CDR3 of SEQ ID No:8.
77. An isolated antibody that mutually cross-blocks the antibody of embodiment 17 by at least 80%.
78. An isolated antibody that mutually cross-blocks at least one of the antibodies of embodiments 18-20 by at least 80%.
79. An isolated antibody that binds to the same epitope as the antibody of embodiment 18.
80. An isolated antibody that binds to the same epitope as the antibody of embodiment 19.
81. An isolated antibody that binds to the same epitope as the antibody of embodiment 20. 82. A method of reducing the level of at least one autoantibody, comprising administering the antibody of any of embodiments 1-4, 8, and 10-31.
83. A method of reducing serum IgM level, comprising administering the antibody of any of embodiments 1-4, 8, and 10-31.
84. A method of reducing serum IgG level, comprising administering the antibody of any of embodiments 1-4, 8, and 10-31.
85. A method of reducing serum lgG2 level, comprising administering the antibody of any of embodiments 1-4, 8, and 10-31.
86. A method of reducing serum lgG3 level, comprising administering the antibody of any of embodiments 1-4, 8, and 10-31.
87. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1 , CDR2, and CDR3 regions of a sequence which is chosen from SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7.
88. The antibody of embodiment 87, wherein the sequence is SEQ ID NO:1 and the antibody further comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1 , CDR2, and CDR3 regions of SEQ ID NO:2.
89. The antibody of embodiment 87, wherein the sequence is SEQ ID NO:3 and the antibody further comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1 , CDR2, and CDR3 regions of SEQ ID NO:12.
90. The antibody of embodiment 87, wherein the sequence is SEQ ID NO:5 and the antibody further comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1 , CDR2, and CDR3 regions of SEQ ID NO:6.
91. The antibody of embodiment 87, wherein the sequence is SEQ ID NO:7 and the antibody further comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1, CDR2, and CDR3 regions of SEQ ID NO:8.
92. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises a light chain variable domain comprising a sequence that is at least 95% identical to a sequence selected from SEQ ID NOs 22-24 and a heavy chain variable domain sequence comprising a sequence that is at least 95% identical to a sequence selected from SEQ ID NOs 25-29.
93. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises a light chain variable domain comprising a sequence that is at least 95% identical to a sequence selected from SEQ ID NOs 30-33 and a heavy chain variable domain sequence comprising a sequence that is at least 95% identical to a sequence selected from SEQ ID NOs 34-38.
94. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises a light chain variable domain comprising a sequence that is at least 95% identical to a sequence selected from SEQ ID NOs 39-42 and a heavy chain variable domain sequence comprising a sequence that is at least 95% identical to a sequence selected from SEQ ID NOs 43-47.
95. The method of embodiment 39, wherein the B-cell related disorder is rheumatoid arthritis. 96. The method of embodiment 39, wherein the B-cell related disorder is idiopathic thrombocytopenia purpura, or myasthenia gravis, or autoimmune hemolytic anemia.
97. The method of embodiment 37, further comprising administering RITUXAN ™ .
98. The method of embodiment 38, further comprising administering RITUXAN ™ .
Example 1. Generation and biotin conjugation of anti-human BCMA monoclonal antibodies
[058] Anti-BCMA monoclonal antibodies (mAbs) were generated by immunizing female RBF mice with BCMA-Fc/KLH conjugate protein i.p. in CFA, followed by additional immunizations at regular intervals with IFA, except that the last boost used RIBI instead of IFA, prior to splenocyte fusion to the FL653 myeloma cell line after the method of Harlow and Lane (1998), Using Antibodies: A Laboratory Manual: Portable Protocol No. I, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY. Briefly, splenocytes isolated from a mouse 3 days after the final boost were washed twice and mixed in a 7:1 ratio with twice-washed log phase FL653 myeloma cells. The cell mixture was split four ways, pelleted, and incubated in 370C PEG for 1 min during which time cells were gently resuspended, followed by careful addition of 10 ml ice-cold DMEM. Cells were mixed, pelleted, and resuspended in AAT hybridoma growth selection media. Cell supernatants were screened for BCMA-specific reactivity by ELISA and flow cytometry. Clones that scored positive for BCMA and negative for Fc-specificity in an ELISA format, positive on BCMA-transfected cells, and negative on mock- transfected cells were expanded and subcloned. Four BCMA-specific clones were selected for further evaluation: C11 D5.3 (IgGI), C12A3.2 (IgGI), C13F12.1 (IgGI) and A7D12.2 (lgG2b). Anti-BCMA mAbs were biotin-conjugated for use in ELISA and FACS experiments described below using a kit according to the manufacturer's recommendations (Molecular Probes, Eugene, OR).
Example 2. Cloning of murine anti-human BCMA mAb variable regions
[059] Total cellular RNA from murine hybridoma cells was prepared using a Qiagen RNeasy mini kit following the manufacturer's recommended protocol. cDNAs encoding the variable regions of the heavy and light chains were cloned by RT-PCR from total cellular RNA, using random hexamers for priming of first strand cDNA. For PCR amplification of the murine immunoglobulin variable domains with intact signal sequences, a cocktail of degenerate forward primers hybridizing to multiple murine immunoglobulin gene family signal sequences and a single back primer specific for the 5' end of the murine constant domain were used. PCR was performed using Clontech Advantage 2 Polymerase mix following the manufacturer's recommended protocol. The PCR products were gel-purified and subcloned into Invitrogen's pCR2.1TOPO vector using their TOPO cloning kit following the manufacturer's recommended protocol. Inserts from multiple independent subclones were sequenced to establish a consensus sequence. Deduced mature immunoglobulin N-termini were consistent with those determined by Edman degradation from the hybridoma. Assignment to specific subgroups was based upon BLAST analysis using consensus immunoglobulin variable domain sequences from the Kabat database (Johnson and Wu (2000), Nucleic Acids Res 28:214-218). CDRs were designated using the Kabat definitions (Johnson and Wu (2000), Nucleic Acids Res 28:214-218). Example 3. Stable BCMA-expressing CHO cell line development and assessment of anti-human BCMA mAbs
[060] To validate specific binding to BCMA, anti-BCMA mAbs were screened on a stable BCMA-expressing CHO cell line. The stable BCMA- expressing CHO cell line was generated using a previously described method (Brezinsky et al. (2003), J Immunol Methods 277:141-155). Briefly, approximately 1.5 million dihydrofolate reductase (DHFR) deficient DG44 Chinese hamster ovary (CHO) cells were transfected with 4 μg PV90 plasm id DNA containing the human BCMA gene using Fugene 6 Transfection Reagent (Roche, Indianapolis, IN). Following transfection, the cells were cultured in 6-well culture dishes. Twenty-four hours post-transfection, the growth medium was changed to alpha minus MEM (Gibco, Rockville, MD), 10% dialyzed serum (Hyclone, Logan, UT), and 2 mM L- glutamine (Gibco, Rockville, MD), and cells were pooled and split into three T-75 tissue culture flasks and allowed to grow to confluence. Seven days post- transfection, the cells were pooled again and split into five T225 tissue culture flasks and allowed to grow to confluence.
[061] Fourteen days post-transfection, the cells were incubated with the C4E2.2 Ab (6) and sorted for BCMA+ cells. These cells were grown in culture and sorted a second time, with positive cells sorted into 96-well plates. Clones were expanded and assessed for BCMA expression using clone C4E2.2.
[062] The highest expressing clone was used for assessing new anti- human BCMA mAbs, with untransfected CHO cells as a control. Briefly, BCMA- CHO cells were pretreated with FACS buffer plus 5% normal mouse serum to block non-specific binding sites. Seven anti-BCMA mAbs — C11 D5.3, C12A3.2, C13F12.1 and A7D12.2 and the commercially available anti-BCMA mAbs, C4E2.2 (Legacy Biogen), VICKY-1 (Alexis) and 335004 (R&D Systems, Inc.)— and isotype control antibodies were incubated separately with cells for 30 minutes on ice at 1 μg/ml, and washed. Biotin-conjugated isotype control Abs were as follows: mouse IgGI , eBioscience cat. no. 13-4714-85; mouse lgG2a, eBioscience, cat. no. 13- 4732-85; hamster IgGI , BD Pharmingen cat. no. 553970; rat lgG2a, eBioscience, cat. No. 13-4321-82. To visualize positive staining, Streptavidin-PE (Molecular Probes, Eugene, OR) was added to cells for 30 minutes on ice, after which cells were washed, fixed in 0.8% paraformaldehyde, and run on a FACScalibur flow cytometer (BDbiosciences, San Jose, CA) and analyzed using Flowjo software (Treestar, Ashland, OR). Results are shown in Figure 1.
[063] All seven antibodies showed specific recognition of the BCMA- expressing cells. C11 D5.3 showed a slightly higher basal binding to the negative control cells than the control mAb; basal binding of the other six antibodies to the negative control cells was similar to that of the control Ab.
Example 4. Analysis of anti-BCMA mAb epitope overlap by cross- blocking ELISA
[064] The seven anti-BCMA mAbs tested in Example 3 were then assessed in a cross-blocking assay to determine the presence or absence of epitope overlap between each antibody. Corning 96-well flat-bottom plates were incubated overnight at 4°C with 10 μg/ml of mouse anti-human Fc in a 50 mM pH 9.6 sodium bicarbonate solution, washed, incubated with 2 μg/ml human BCMA- Fc (SEQ ID NO:10) at 37°C for 1 hr, washed again, and non-specific binding sites were blocked with blocking buffer (3% BSA in PBS) for 30 min at 37°C. Triplicate wells were then incubated with each unconjugated anti-BCMA mAb clone in blocking buffer at a concentration that was ten times the concentration of the single biotin-conjugated anti-BCMA mAb used in each individual experiment. A single biotin-conjugated anti-BCMA mAb clone in blocking buffer was added to all wells at a concentration pre-determined to give 80% of maximal signal (EC80) and incubated for 1 hr at 37°C, after which wells were washed and incubated with a streptavidin-HRP solution for 30 min, 37°C, washed, and incubated with substrate, TMB, to visualize positive reactivity. Enzymatic reactivity was stopped by adding 2N sulfuric acid, and the absorbance at 450 nm was measured using a plate reader.
[065] For each species of biotin-conjugated antibody, control readings in which the unconjugated and conjugated antibodies were the same (except for the presence/absence of conjugated biotin) were considered background levels, i.e., the absorbance from this control was subtracted from each experimental reading for that antibody. In some cases, this background subtraction resulted in slightly negative values. Although it is possible that the labeled antibody was blocked slightly more effectively by a different unconjugated antibody than by itself, these slightly negative values might also result from experimental variation. Results were then expressed as a percentage of the positive control value, i.e., the background-adjusted absorbance reading for the biotin-conjugated antibody in the absence of a competing unconjugated antibody.
[066] Antibodies were considered to bind the same epitope or very closely overlapping epitopes if they each reduced the other's binding (according to the fraction calculated as above) to below 20% of the positive control value. If they did not satisfy this condition, they were considered to have at least partially distinct epitopes. Table 3 shows which antibodies have at least partially distinct epitopes. Example 5. Flow cytometric analysis of antibody binding to human peripheral blood cells
[067] Blood was obtained from consenting healthy volunteers and peripheral blood mononuclear cells (PBMCs) were enriched by centrifugation through Ficoll-Paque (GE Healthcare, UK) according to the manufacturer's recommendations. PBMCs were washed extensively in PBS prior to use. Cells were pretreated with FACS buffer containing 5% normal mouse serum to block non-specific binding sites. The following fluorphore-conjugated monoclonal antibodies directed against specific B cell and plasma cell markers were used: anti-CD19-PE-Cy5, anti-lgD-FITC, anti-CD27-APC, anti-CD38-PE-Cy7 (BD Biosciences, San Jose, CA). Streptavidin-PE (Molecular Probes, Eugene, OR) was used to visualize biotin-conjugated anti-BCMA mAbs (10 μg/ml). Binding of an isotype control mAb as in Example 3 was also measured.
[068] None of the anti-BCMA mAbs stained naive B cells from healthy volunteers (Fig. 2E), while all stained plasma cells, although with varying intensities (Fig. 2A). Only clone A7D12.2 stained a proportion of all three memory B cell subsets (Fig. 2B-D).
Example 6, Comparison of antibody binding to B cells of healthy and SLE-afflicted individuals
[069] Blood was obtained from consenting healthy volunteers and SLE patients and processed as in Example 5. Figure 3 shows a comparison between samples from a healthy volunteer and a representative SLE patient. Antibody A7D12.2 bound to plasma cells from both healthy volunteers and SLE patients (Fig. 3A). In the SLE samples but not the healthy samples, the A7D12.2 antibody bound naϊve B cells (Fig. 3E). Binding of the A7D12.2 antibody to memory B cells (Fig. 3B-D), particularly double negative memory B cells (Fig. 3D), was increased in SLE samples.
Example 7. Generation of cell lines producing chimeric anti-BCMA mAbs
[070] CHO-DG44-I , a dhfr-deficient, insulin-independent Chinese hamster ovary cell line, was used to construct anti-BCMA wild type cell lines. The host cells were cultured in CHO-S-SFM Il medium with nucleosides prior to transfection.
[071] Chimeric antibodies were produced by transfecting cells with expression plasmids encoding the mature heavy and light chain sequences listed in Table 4.
Table 4. Mature heavy and light chain sequences of chimeric anti-BCMA antibodies
Figure imgf000045_0001
[072] Chimeric anti-BCMA expression plasmids were transfected into the CHO host cell line DG44-I using a cationic lipid (Fugene HD) method. Briefly, 1x106 DG44-I cells were seeded in each of two wells of a 6-well plate containing 3 ml_ of CHO-S-SFMII medium w/nucleosides per well. Four μg of plasmid DNA (2 μg heavy chain, 2 μg light chain) was diluted into 200 μl_ CHO-S-SFM Il (Invitrogen) medium at room temperature. Sixteen μl_ of Fugene HD (Roche) reagent and allowed to complex with the DNA for approximately 15 minutes. 100 μl_ of the complexed DNA mixture was added to each well containing the cells. After three days, the transfected cells were combined and transferred to a T-75 flask containing 20 ml_ CHO-S-SFM Il medium w/o nucleosides containing 400 μg/mL geneticin (Invitrogen). Cells were monitored for viability and scaled-up accordingly. As the cells were scaled-up they were adapted to production medium. Clonal cell lines were obtained by FACS sorting individual cells from the stable population.
[073] CHO-DG44-I cells stably transfected with chimeric heavy and light chains were fermented in CHOM39 media, harvested, and the cells were removed by centrifugation. The pH of the cleared media was adjusted prior to passing through a protein A Fast Flow column. Antibodies were eluted with 100 mM Na- citrate buffer, pH 3.0, neutralized to pH 7.0 using 10% (v/v) of 2M glycine, pH 8.9, and the recovered antibody solution was buffer-exchanged to PBS (pH 7.2) using a Superdex 200 size exclusion chromatography under endotoxin-free conditions. The purified protein was kept at -800C.
Example 8. Anti-BCMA-mediated killing in vitro
Cells and cell culture
[074] JJN-3 human plasmacytoma cells (DSMZ ACC 541) were cultured in culture medium consisting of 40% Dulbecco's MEM, 40% Iscove's MDM, 20% FBS, 100 units/mL penicillin and 100 μg/mL streptomycin. U266 human plasmacytoma cells (ATCC TIB 196) were cultured in culture medium consisting of RPMI 1640, 15% FBS, 20 mM HEPES, 100 units/mL penicillin and 100 μg/mL streptomycin. All cells were cultured at 37 0C in a 5% CO2 atmosphere. Peripheral blood mononuclear cells were isolated from a consented normal healthy donor by density centrifugation through Ficoll-Paque PLUS (GE Healthcare, Uppsala, Sweden). In vitro antibody-dependent cellular cytotoxicity (ADCC) assay
[075] Assay diluent was RPMI 1640, 1% BSA, 20 mM HEPES, 100 units/mL penicillin and 100 μg/mL streptomycin. Human plasmacytoma cell lines JJN-3 and U266 were washed and resuspended in assay diluent to 0.4 x 106 cells per ml_. 50 μl_ of each cell suspension was plated into a 96-well U bottom microtiter plate in triplicates. 50 μl_ of serially diluted chimeric anti-BCMA antibodies (chC12A3.2, chC13F12.1 , chC11 D5.3, and chA7D12.2, as described in Example 7) and control human IgGI (HIgGI ; Protos Immunoresearch, Burlingame, CA) were added to wells containing the cell lines and incubated for 30 minutes at 37 0C. For an effectoπtarget ratio of 25:1 , 50 μl_ of PBMCs (500,000) were added and incubated for an additional four hours at 37 0C in a 5% CO2 atmosphere. Plates were centrifuged at 1200 rpm for 5 minutes, and 100 μL of supernatant was transferred to a 96-flat-bottom microwell plate. The level of cell lysis was determined by measuring the amount of lactate dehydrogenase (Cytotoxicity Detection Kit (LDH), #11 644 793 001 Roche) released from lysed cells. 100 μL of LDH kit reaction mixture was added to 100 μL of supernatant for up to 30 minutes as followed by manufacturer instructions. Absorbance was measured at 490 nm. Controls included target cells alone (spontaneous LDH release), target cells alone with 2% Triton X-100 in assay diluent (maximum LDH release), effector cells with and without target cells, and human IgGI isotype control.
Results
[076] The human BCMA+ plasmacytoma cell line, U266, was utilized to test the ability of chimeric anti-BCMA antibodies to kill via ADCC. Human peripheral blood mononuclear cells (PBMCs) were used as effector cells. As shown in Table 5, chimeric mAbs C12A3.2 and C13F12.1 demonstrated marked ADCC activity relative to HIgGI controls. Chimeric clones A7D12.2 and C11 D5.3 also mediated ADCC, although to a lesser degree than C12A3.2 and C13F12.1. Table 5. ADCC of U266 cells by anti-human BCMA mAbs
Figure imgf000048_0001
Maximum killing determination is incomplete
[077] ADCC assays were also performed using a second BCMA+ plasmacytoma cell line, JJN-3, as the target cells. As shown in Table 6, chimeric C12A3.2, C13F12.1 , and A7D12.2 also mediated ADCC of JJN-3 cells. Table 6. ADCC of JJN-3 cells by anti-human BCMA mAbs
Figure imgf000048_0002
Maximum killing determination is incomplete
[078] Using enriched human NK cells as effector cells did not result in improved killing for chC12A3.2, chC13F12.1 or chA7D12.2. Rather, percent killing by NKs was diminished relative to PBMCs, indicating that NK cells are not the effector cells for chC 12A3.2, chC13F12.1 , or chA7D12.2 in the ADCC assays with U266 and JJN3 as target cells (data not shown). Generation of afucosyl variants of chC12A3.2, chC13F12.1 , or chA7D12.2 did not improve activity of these mAbs (data not shown).
Example 9. Anti-BCMA-mediated cell depletion in vivo
Establishment of humanized (HSC/NSG) mice
[079] HSC/NSG mice provide a useful tool for testing in vivo biologic reagents specific for human protein targets since their immune system consists of functioning human cell types (Brehm et al. (2010), Clin Immunol [Epub ahead of print]). Humanized mice were generated as previously described (Pearson et al. (2008), Curr Protoc Immunol Chapter 15:Unit 15.21. PMID: 18491294). NOD/SCID/common gamma chain-deficient mice (NSG mice) were purchased from Jackson Laboratories (Bar Harbor, ME). Mice were maintained and bred under specific-pathogen-free conditions in isolator cages. Breeding pairs were established and dams were monitored for pregnancy and delivery. Pups ranging in age from 2-6 days old were used to create humanized mice. Pups were lightly irradiated, receiving a dose of 1 Gy (100 rad) from a 137Cs irradiator. Pups were immediately injected intra-orbitally with approximately 50,000 CD34+CD3- human stem cells (HSC) derived from umbilical cord blood (All Cell LLC, purchased from StemCell Technologies Inc., Vancouver, BC, Canada), then returned to the dam. Pups were weaned at 21 days and caged with littermates according to gender. Starting at 3 months of age mice were bled via the facial vein into heparinized tubes and the whole blood was analyzed by flow cytometry for the presence of human cells. In brief, 100 μl whole blood was stained with a cocktail of mAbs, anti- human CD45-FITC, anti-human CD3-PE, anti-human CD19-PerCp, and anti- mouse CD45-APC (BD Biosciences, San Jose, California). Mice were considered successfully humanized if they had at least 20% human CD45+ cells in whole blood of which 10% or more were human CD3+, the remainder being human B cells and other human hematopoietic cells. Mice were occasionally bled and analyzed to ensure that the humanization was stable, and were routinely analyzed 2 weeks prior to study enrollment. Flow cytometry
[080] To identify plasma cells (PC) in human stem cell humanized NSG (HSC/NSG) mice, collagenase-digested spleens were subjected to flow cytometric analysis. Cells were incubated with a cocktail of mAbs directed to human cell lineage and to human B cell markers. The panel consisted of anti-human CD45, anti-human CD19, anti-human CD27, anti-human IgD, and anti-human CD38. Two markers used to exclude specific cell populations were anti-mouse CD45 and anti-human CD3. Cells identified as PC were human CD45+, human CD19+, human CD3-, human CD27+, human IgD- and human CD38bright . To identify BCMA+ cells the biotin-conjugated anti-human BCMA mAbs, C12A3.2 and A7D12.2, were used.
In vivo cell depletion assay
[081] 5-6 month-old HSC/NSG mice received chimeric anti-BCMA mAb i.p. Human IgGI with no known reactivity (Protos Immunoresearch) was used as a negative control. Blood was collected to prepare serum for analysis of human Ig isoform levels. At the study terminus the spleen was harvested, a single cell suspension was prepared, RBCs were lysed and cells were washed 3x with PBS/5% FCS and the cell number was determined. Cells were assessed by flow cytometry for T lineage cells, B lineage cells, and plasma cells.
Assessment of serum human Ig isotypes
[082] Serum levels of human IgM and IgG were determined using an ELISA format (Bethyl Laboratories Inc., Montgomery, TX) and a human immunoglobulin isotyping kit (Millipore, Billerica, MA), respectively, according to the manufacturer's protocol. Results
[083] Analysis of HSC/NSG mice aged 5-6 months revealed that among the diverse cell subsets assessed BCMA+ cells were only found in the B cell lineage. Within the B cell lineage, BCMA was found only on splenic PCs (human CD19+, human CD27+, IgD-, CD38 bright) (Figure 4), and not on naive B cells (human CD19+, human CD27- , IgD+), unswitched memory B cells (human CD19+, human CD27+, human IgD+), or switched memory B cells (human CD19+, human CD27+, human IgD-) (data not shown).
[084] To assess the ability of chimeric anti-human BCMA mAbs to deplete human PC, HSC/NSG mice received various amounts of chimeric anti- human BCMA clones chAC11 D5.3, chC12A3.2, chC13F12.1 , and chA7D12.2 (Example 7) twice weekly i.p. for 2 weeks, after which the presence of splenic PCs was determined. Splenic PCs from the HIgGI -treated control group were analyzed for expression of BCMA using the A7D12.2 and C12A3.2 clones and were confirmed to express cell surface BCMA (data not shown). PCs were identified using the flow cytometric parameters described above, and total cell numbers were determined from the flow cytometric dot plots (calculated as a percentage of the total human cell number). The numbers of naive human B cells, unswitched memory human B cells, and switched memory human B cells were also determined.
[085] Treatment with chC12A3.2 (N=5) resulted in a statistically significant decline, 93% and 95%, in the number of splenic PCs at the 200 μg and 20 μg dose levels, respectively, when compared with control HlgG-treated mice (N=5) while the 2 μg dose also showed a marked decline (32%), although it was not statistically significant. Treatment with chC13F12.1 (N=5) resulted in a statistically significant decline, 88% and 51%, in the number of splenic PCs at the 200 μg and 20 μg dose levels, respectively (Figure 5). No impact on the number of other B cell subsets or T cells was observed with chC12A3.2 and chC13F12.1 (data not shown).
[086] Treatment with 200 μg of chC11 D5.1 (N=2) or chA7D12.2 (N=1) resulted in an 89% and 97% reduction, respectively, in human PCs within the spleen when compared to HlgG-treated control mice (N =5) (Figure 6). Treatment with chA7D12.1 also resulted in a 2.6-fold decline in the number of splenic human switched memory B cells when compared to HlgG-treated mice (575 vs. 217 PCs/105 HuCD45+ cells, for HIgG and chA7D12.1 , respectively). Although BCMA could not be detected on the surface of switched memory B cells in untreated humanized mice, it appears that while the level of BCMA was below the limit of detection by flow cytometry, it was sufficient to result in Ab-mediated killing.
[087] To determine the impact of human PC depletion on serum human Ig levels in the HSC/NSG mice described above, human Ig subsets were analyzed. The chimeric anti-BCMA mAbs used in these studies were of the human IgGI isoform, therefore human IgGI levels could not be accurately evaluated. In some experimental cohorts, control-treated mice had very little and variable amounts of isotypes IgGI , lgG2, IgA and IgE (data not shown). As shown in Table 7, both chC12A3.2 and chC13F12.1 resulted in marked reductions in serum human IgM, especially at the higher dose levels. Chimeric C12A3.2 resulted in a 63%, 62% and 30% reduction in IgM for the 200, 20 and 2 μg dose levels, respectively. Chimeric C13F12.1 resulted in a 52%, 42% and 32% reduction in IgM for the 200, 20 and 2 μg dose levels, respectively. Table 7. Serum human IgM levels (μg/mL)
Figure imgf000053_0001
SD=standard deviation; p=0.008; p=0.02; p=o.O5 , ND=not done
[088] In a separate experiment, mice received chC12A3.2 (N=14) or HIgGI control (N=9), and the control mice had readily detectable lgG2 and lgG3 isotypes as well as IgM. Chimeric C12A3.2-treated mice exhibited a significant depletion of splenic plasma cells, similar to that seen in Figure 5 (data not shown). As shown in Table 8, mice that received chC12A3.2 exhibited significantly reduced serum lgG2 and IgM levels when compared with HlgG-treated control mice. Chimeric C12A3.2-treated mice also had a marked reduction in serum lgG3, although the difference did not reach statistical significance when compared to control mice (Table 8). Table 8. Serum human immunoglobulin levels
Figure imgf000053_0002
SE=standard error; p=0.05; p=0.003
[089] The embodiments within the specification provide an illustration of embodiments of the invention and should not be construed to limit the scope of the invention. The skilled artisan readily recognizes that many other embodiments are encompassed by the invention. All publications and patents cited in this disclosure are incorporated by reference in their entirety. To the extent the material incorporated by reference contradicts or is inconsistent with this specification, the specification will supersede any such material. The citation of any references herein is not an admission that such references are prior art to the present invention.
[090] Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification, including claims, are to be understood as being modified in all instances by the term "about." Accordingly, unless otherwise indicated to the contrary, the numerical parameters are approximations and may vary depending upon the desired properties sought to be obtained by the present invention. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should be construed in light of the number of significant digits and ordinary rounding approaches.
[091] Unless otherwise indicated, the term "at least" preceding a series of elements is to be understood to refer to every element in the series. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

WHAT IS CLAIMED IS:
1. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises at least one CDR chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
2. The antibody of claim 1 , wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
3. The antibody of claim 1 , wherein the antibody comprises the CDRs at CDR1 , CDR2, and CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
4. The antibody of claim 1 , wherein the at least one CDR chosen from CDR1 , CDR2, or CDR3 is from SEQ ID NO: 5, and the antibody further comprises a variable domain comprising at least one of a CDR1 region identical to amino acids 24-38 of SEQ ID NO: 6, a CDR2 region identical to amino acids 54-60 of SEQ ID NO: 6, or a CDR3 region identical to amino acids 93-101 of SEQ ID NO: 6.
5. The antibody of claim 1 , wherein the heavy chain variable domain comprises SEQ ID NO:5 and the light chain variable domain comprises SEQ ID NO:6.
6. An isolated antibody that binds the same epitope as the antibody of claim 5.
7. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises: a. a variable domain comprising CDR1 , CDR2, and CDR3 regions identical to the CDR1 , CDR2, and CDR3 of a sequence which is chosen from SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7; or b. a variant of the variable domain of part (a) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
8. The antibody of claim 7, wherein the sequence is SEQ ID NO:5 and the antibody further comprises: c. a variable domain comprising a CDR1 region identical to amino acids 24-38 of SEQ ID NO:6, a CDR2 region identical to amino acids 54-60 of SEQ ID NO:6, and a CDR3 region identical to amino acids 93-101 of SEQ ID NO:6; or d. a variant of the variable domain of part (c) that is otherwise identical to said variable domain except for up to a total of 10 amino acid substitutions in said CDR regions.
9. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises at least one CDR chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:12, SEQ ID NO:6, and SEQ ID NO:8.
10. The antibody of claim 9, wherein the antibody comprises at least two CDRs chosen from CDR1 , CDR2, or CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:12, SEQ ID NO:6, and SEQ ID NO:8.
11. The antibody of claim 10, wherein the antibody comprises the CDRs at CDR1 , CDR2, and CDR3 of a protein sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:12, SEQ ID NO:6, or SEQ ID NO:8.
12. The antibody of any of claims 1-11 , wherein the antibody is a chimeric, humanized, or single chain antibody.
13. A hybridoma that produces the antibody of any of claims 1-11.
14. A pharmaceutical composition comprising the antibody of any of claims 1- 11 and a pharmaceutically acceptable carrier.
15. A polypeptide that binds to SEQ ID NO:9 and comprises the antigen binding portion, Fab fragment, or F(ab')2 fragment of the antibody of any of claims 1-11.
16. A hybridoma that produces the antigen binding portion, Fab fragment, or F(ab')2 fragment of claim 15.
17. A pharmaceutical composition comprising the antigen binding portion, Fab fragment, or F(ab')2 fragment of claim 15 and a pharmaceutically acceptable carrier.
18. A method of depleting plasma cells, comprising administering the antibody of any of claims 1-11.
19. A method of treating a B cell-related disorder, comprising administering the antibody of any one of claims 1-11.
20. The method of claim 19, wherein the B-cell related disorder is plasmacytoma, Hodgkins' lymphoma, follicular lymphomas, small non-cleaved cell lymphomas, endemic Burkitt's lymphoma, sporadic Burkitt's lymphoma, marginal zone lymphoma, extranodal mucosa-associated lymphoid tissue lymphoma , nodal monocytoid B cell lymphoma, splenic lymphoma, mantle cell lymphoma, large cell lymphoma, diffuse mixed cell lymphoma, immunoblastic lymphoma, primary mediastinal B cell lymphoma, pulmonary B cell angiocentric lymphoma, small lymphocytic lymphoma, B cell proliferations of uncertain malignant potential, lymphomatoid granulomatosis, post-transplant lymphoproliferative disorder, an immunoregulatory disorder, rheumatoid arthritis, myasthenia gravis, idiopathic thrombocytopenia purpura, anti-phospholipid syndrome, Chagas1 disease, Grave's disease, Wegener's granulomatosis, poly-arteritis nodosa, Sjogren's syndrome, pemphigus vulgaris, scleroderma, multiple sclerosis, anti-phospholipid syndrome, ANCA associated vasculitis, Goodpasture's disease, Kawasaki disease, autoimmune hemolytic anemia, and rapidly progressive glomerulonephritis, heavy-chain disease, primary or immunocyte-associated amyloidosis, or monoclonal gammopathy of undetermined significance.
21. The method of claim 20, wherein the B cell-related disorder is a B cell malignancy.
22. The method of claim 20, wherein the B cell-related disorder is a plasma cell malignancy.
23. The method of claim 22, wherein the plasma cell malignancy is multiple myeloma.
24. The method of claim 20, wherein the B cell-related disorder is an autoimmune disease.
25. The method of claim 24, wherein the autoimmune disease is systemic lupus erythematosus.
26. The method of claim 20, wherein the B-cell related disorder is rheumatoid arthritis.
27. The method of claim 20, wherein the the B-cell related disorder is idiopathic thrombocytopenia purpura, or myasthenia gravis, or autoimmune hemolytic anemia.
28. The method of claim 18, further comprising administering RITUXAN ™.
29. A method of reducing the level of at least one autoantibody, comprising administering the antibody of any of claims 1-11.
30. An isolated antibody that binds to the polypeptide of SEQ ID NO:9, wherein the antibody comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1 , CDR2, and CDR3 regions of a sequence which is chosen from SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7.
31. The antibody of claim 30, wherein the sequence is SEQ ID NO:5 and the antibody further comprises a variable domain comprising CDR1 , CDR2, and CDR3 regions that are together at least 95% identical to the CDR1 , CDR2, and CDR3 regions of SEQ ID NO:6.
PCT/US2010/026825 2009-03-10 2010-03-10 Anti-bcma antibodies WO2010104949A2 (en)

Priority Applications (21)

Application Number Priority Date Filing Date Title
JP2011554150A JP6061469B2 (en) 2009-03-10 2010-03-10 Anti-BCMA antibody
EP10708865.0A EP2406284B9 (en) 2009-03-10 2010-03-10 Anti-bcma antibodies
DK10708865.0T DK2406284T5 (en) 2009-03-10 2010-03-10 ANTI-BCMA ANTIBODIES
CA2754938A CA2754938C (en) 2009-03-10 2010-03-10 Anti-bcma antibodies
SI201031281A SI2406284T1 (en) 2009-03-10 2010-03-10 Anti-bcma antibodies
MX2011009430A MX2011009430A (en) 2009-03-10 2010-03-10 Anti-bcma antibodies.
LTEP10708865.0T LT2406284T (en) 2009-03-10 2010-03-10 Anti-bcma antibodies
AU2010224160A AU2010224160A1 (en) 2009-03-10 2010-03-10 Anti-BCMA antibodies
NZ59498510A NZ594985A (en) 2009-03-10 2010-03-10 Anti-bcma (b-cell maturation antigen, cd269, tnfrsf17) antibodies
US13/255,610 US9034324B2 (en) 2009-03-10 2010-03-10 Anti-BCMA antibodies
MX2013013076A MX341884B (en) 2009-03-10 2010-03-10 Anti-bcma antibodies.
ES10708865.0T ES2593583T3 (en) 2009-03-10 2010-03-10 Anti-BCMA antibodies
CN201080020559.0A CN102421801B (en) 2009-03-10 2010-03-10 Anti-BCMA antibody
IL214996A IL214996A (en) 2009-03-10 2011-09-06 Isolated antibody that binds a bmca polypeptide, a pharmaceutical composition comprising the same and uses thereof
HK12107062.4A HK1166333A1 (en) 2009-03-10 2012-07-18 Anti-bcma antibodies -bcma
US14/596,769 US20150125460A1 (en) 2009-03-10 2015-01-14 Anti-bcma antibodies
US15/065,641 US20170029518A1 (en) 2009-03-10 2016-03-09 Anti-bcma antibodies
CY20161100917T CY1118069T1 (en) 2009-03-10 2016-09-15 ANTI-BCMA ANTIBODIES
HRP20161194TT HRP20161194T1 (en) 2009-03-10 2016-09-19 Anti-bcma antibodies
US16/026,242 US11111307B2 (en) 2009-03-10 2018-07-03 Anti-BCMA antibodies
US17/444,107 US20220213208A1 (en) 2009-03-10 2021-07-30 Anti-bcma antibodies

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US15894209P 2009-03-10 2009-03-10
US61/158,942 2009-03-10
US16292409P 2009-03-24 2009-03-24
US61/162,924 2009-03-24

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US13/255,610 A-371-Of-International US9034324B2 (en) 2009-03-10 2010-03-10 Anti-BCMA antibodies
US14/596,769 Division US20150125460A1 (en) 2009-03-10 2015-01-14 Anti-bcma antibodies
US14/596,769 Continuation US20150125460A1 (en) 2009-03-10 2015-01-14 Anti-bcma antibodies

Publications (2)

Publication Number Publication Date
WO2010104949A2 true WO2010104949A2 (en) 2010-09-16
WO2010104949A3 WO2010104949A3 (en) 2010-11-25

Family

ID=42139515

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/026825 WO2010104949A2 (en) 2009-03-10 2010-03-10 Anti-bcma antibodies

Country Status (21)

Country Link
US (5) US9034324B2 (en)
EP (2) EP2406284B9 (en)
JP (2) JP6061469B2 (en)
KR (1) KR101589785B1 (en)
CN (2) CN102421801B (en)
AU (2) AU2010224160A1 (en)
CA (1) CA2754938C (en)
CY (1) CY1118069T1 (en)
DK (1) DK2406284T5 (en)
ES (1) ES2593583T3 (en)
HK (1) HK1166333A1 (en)
HR (1) HRP20161194T1 (en)
HU (1) HUE029619T4 (en)
IL (1) IL214996A (en)
LT (1) LT2406284T (en)
MX (2) MX341884B (en)
NZ (2) NZ594985A (en)
PL (1) PL2406284T3 (en)
PT (1) PT2406284T (en)
SI (1) SI2406284T1 (en)
WO (1) WO2010104949A2 (en)

Cited By (127)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012163805A1 (en) * 2011-05-27 2012-12-06 Glaxo Group Limited Bcma (cd269/tnfrsf17) -binding proteins
US20130101599A1 (en) * 2011-04-21 2013-04-25 Boehringer Ingelheim International Gmbh Bcma-based stratification and therapy for multiple myeloma patients
US20130156769A1 (en) * 2011-11-15 2013-06-20 Boehringer Ingelheim International Gmbh Binding molecules for bcma and cd3
WO2013154760A1 (en) 2012-04-11 2013-10-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting b-cell maturation antigen
US20130273055A1 (en) * 2010-11-16 2013-10-17 Eric Borges Agents and methods for treating diseases that correlate with bcma expression
WO2014068079A1 (en) 2012-11-01 2014-05-08 Max-Delbrück-Centrum für Molekulare Medizin An antibody that binds cd269 (bcma) suitable for use in the treatment of plasma cell diseases such as multiple myeloma and autoimmune diseases
EP2762496A1 (en) 2013-02-05 2014-08-06 EngMab AG Method for the selection of antibodies against BCMA
EP2762497A1 (en) 2013-02-05 2014-08-06 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
WO2014122144A1 (en) 2013-02-05 2014-08-14 Engmab Ag BISPECIFIC ANTIBODIES AGAINST CD3ε AND BCMA
WO2014140248A1 (en) * 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Binding molecules for bcma and cd3
WO2016014789A2 (en) 2014-07-24 2016-01-28 Bluebird Bio, Inc. Bcma chimeric antigen receptors
EP2982694A1 (en) 2014-08-04 2016-02-10 EngMab AG Bispecific antibodies against cd3epsilon and bcma
WO2016022630A1 (en) * 2014-08-05 2016-02-11 Jiping Zha Anti-pd-l1 antibodies
US9273141B2 (en) 2011-05-27 2016-03-01 Glaxo Group Limited B cell maturation antigen (BCMA) binding proteins
WO2016014565A3 (en) * 2014-07-21 2016-03-10 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
EP3023437A1 (en) 2014-11-20 2016-05-25 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
EP3029068A1 (en) 2014-12-03 2016-06-08 EngMab AG Bispecific antibodies against CD3epsilon and BCMA for use in the treatment of diseases
WO2016166629A1 (en) * 2015-04-13 2016-10-20 Pfizer Inc. Therapeutic antibodies and their uses
WO2017031104A1 (en) 2015-08-17 2017-02-23 Janssen Pharmaceutica Nv Anti-bcma antibodies, bispecific antigen binding molecules that bind bcma and cd3, and uses thereof
US20170166622A1 (en) * 2015-05-18 2017-06-15 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2017123741A1 (en) 2016-01-12 2017-07-20 Institute For Myeloma & Bone Cancer Research Improved methods for monitoring immune status of a subject
WO2017143069A1 (en) * 2016-02-17 2017-08-24 Seattle Genetics, Inc. Bcma antibodies and use of same to treat cancer and immunological disorders
WO2018083204A1 (en) 2016-11-02 2018-05-11 Engmab Sàrl Bispecific antibody against bcma and cd3 and an immunological drug for combined use in treating multiple myeloma
WO2018151836A1 (en) 2017-02-17 2018-08-23 Fred Hutchinson Cancer Research Center Combination therapies for treatment of bcma-related cancers and autoimmune disorders
WO2018158349A1 (en) 2017-02-28 2018-09-07 Affimed Gmbh Tandem diabody for cd16a-directed nk-cell engagement
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
WO2018204427A1 (en) 2017-05-01 2018-11-08 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound
US10144782B2 (en) 2014-04-30 2018-12-04 Max-Delbrück-Centrum Für Molekulare Medizin In Der Helmholtz-Gemeinschaft Humanized antibodies against CD269 (BCMA)
WO2018231949A1 (en) 2017-06-14 2018-12-20 Dana-Farber Cancer Institute, Inc. B-cell maturation antigen (bcma)-directed nanoparticles
WO2019025983A1 (en) 2017-08-01 2019-02-07 Medimmune, Llc Bcma monoclonal antibody-drug conjugate
US10208285B2 (en) 2016-10-07 2019-02-19 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2019090364A1 (en) 2017-11-06 2019-05-09 Juno Therapeutics, Inc. Combination of a cell therapy and a gamma secretase inhibitor
WO2019090003A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Chimeric antigen receptors specific for b-cell maturation antigen (bcma)
WO2019089969A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for b-cell maturation antigen
US10294304B2 (en) 2015-04-13 2019-05-21 Pfizer Inc. Chimeric antigen receptors targeting B-cell maturation antigen
WO2019113556A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Serum-free media formulation for culturing cells and methods of use thereof
WO2019113557A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Process for producing a composition of engineered t cells
WO2019149269A1 (en) * 2018-02-01 2019-08-08 信达生物制药(苏州)有限公司 Fully human anti-b cell maturation antigen (bcma) single chain variable fragment, and application thereof
US10383929B2 (en) 2014-12-12 2019-08-20 Bluebird Bio, Inc. BCMA chimeric antigen receptors
WO2019166650A1 (en) 2018-03-02 2019-09-06 Cdr-Life Ag Trispecific antigen binding proteins
US10428146B2 (en) 2014-07-22 2019-10-01 Cb Therapeutics, Inc. Anti PD-1 antibodies
WO2019198051A2 (en) 2018-04-13 2019-10-17 Affimed Gmbh Nk cell engaging antibody fusion constructs
US10479975B2 (en) 2014-06-06 2019-11-19 Bluebird Bio, Inc. Methods of making T cell compositions
WO2020033927A2 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
WO2020033916A1 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Methods for assessing integrated nucleic acids
US10562972B2 (en) 2014-12-05 2020-02-18 Memorial Sloan Kettering Cancer Center Antibodies targeting B-cell maturation antigen and methods of use
WO2020092854A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Chimeric antigen receptors specific for g protein-coupled receptor class c group 5 member d (gprc5d)
WO2020089343A1 (en) 2018-10-31 2020-05-07 Juno Therapeutics Gmbh Methods for selection and stimulation of cells and apparatus for same
WO2020092848A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Methods for treatment using chimeric antigen receptors specific for b-cell maturation antigen
WO2020097403A1 (en) 2018-11-08 2020-05-14 Juno Therapeutics, Inc. Methods and combinations for treatment and t cell modulation
EP3670535A1 (en) 2015-08-03 2020-06-24 EngMab Sàrl Monoclonal antibodies against bcma
US10774343B2 (en) 2014-04-25 2020-09-15 Bluebird Bio, Inc. MND promoter chimeric antigen receptors
US10821135B2 (en) 2014-12-05 2020-11-03 Memorial Sloan Kettering Cancer Center Nucleic acid molecules encoding chimeric antigen receptors targeting B-cell maturation antigen and uses thereof
WO2020224605A1 (en) 2019-05-07 2020-11-12 亘喜生物科技(上海)有限公司 Bcma-targeting engineered immune cell and use thereof
WO2020224606A1 (en) 2019-05-07 2020-11-12 亘喜生物科技(上海)有限公司 Engineered immune cell targeting bcma and use thereof
US10870701B2 (en) 2016-03-15 2020-12-22 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
WO2021018168A1 (en) 2019-07-30 2021-02-04 上海翰森生物医药科技有限公司 Anti-bcma antibody, antigen-binding fragment thereof and medical use thereof
US10927182B2 (en) 2019-01-16 2021-02-23 Caribou Biosciences, Inc. Humanized BCMA antibody and BCMA-CAR-T cells
US10967005B2 (en) 2013-03-15 2021-04-06 Celgene Corporation Modified T lymphocytes comprising a BAFF antibody-inducible caspase and methods of apoptosis
US11013800B2 (en) 2011-05-16 2021-05-25 Evive Biotech Ltd. Multi-specific Fab fusion proteins comprising a CD3-binding Fab fragment with N-terminal fusion to binding domains and methods of use
WO2021136323A1 (en) * 2020-01-03 2021-07-08 Salubris (Chengdu) Biotech Co., Ltd. Antibodies binding bcma and uses thereof
WO2021154887A1 (en) 2020-01-28 2021-08-05 Juno Therapeutics, Inc. Methods for t cell transduction
WO2021163389A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Bcma-directed chimeric antigen receptor t cell compositions and methods and uses thereof
US11130820B2 (en) 2012-12-20 2021-09-28 Celgene Corporation Chimeric antigen receptors
EP3689908A4 (en) * 2017-09-29 2021-09-29 Mogam Institute for Biomedical Research Anti-bcma antibody having high affinity for bcma and pharmaceutical composition for treatment of cancer, comprising same
WO2021207689A2 (en) 2020-04-10 2021-10-14 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
WO2021222330A2 (en) 2020-04-28 2021-11-04 Juno Therapeutics, Inc. Combination of bcma-directed t cell therapy and an immunomodulatory compound
WO2021231657A1 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Methods of identifying features associated with clinical response and uses thereof
WO2021228783A1 (en) 2020-05-11 2021-11-18 Janssen Pharmaceutica Nv Methods for treating multiple myeloma
US11242376B2 (en) 2016-08-02 2022-02-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2022035793A1 (en) 2020-08-10 2022-02-17 Precision Biosciences, Inc. Antibodies and fragments specific for b-cell maturation antigen and uses thereof
WO2022060806A1 (en) 2020-09-16 2022-03-24 Obsidian Therapeutics, Inc. Compositions and methods for expression of anti-bcma chimeric antigen receptors with small molecule-regulated il15 in t cells
WO2022094245A1 (en) 2020-10-30 2022-05-05 Vor Biopharma, Inc. Compositions and methods for bcma modification
WO2022098787A1 (en) 2020-11-04 2022-05-12 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified invariant cd3 immunoglobulin superfamily chain locus and related polynucleotides and methods
WO2022146891A2 (en) 2020-12-31 2022-07-07 Sana Biotechnology, Inc. Methods and compositions for modulating car-t activity
WO2022150731A1 (en) 2021-01-11 2022-07-14 Sana Biotechnology, Inc. Use of cd8-targeted viral vectors
US11390681B2 (en) 2020-04-29 2022-07-19 TeneoTwo, Inc. Multispecific heavy chain antibodies with modified heavy chain constant regions
WO2022175255A2 (en) 2021-02-16 2022-08-25 Janssen Pharmaceutica Nv Trispecific antibody targeting bcma, gprc5d, and cd3
US11434299B2 (en) 2016-12-21 2022-09-06 Teneobio, Inc. Anti-BCMA heavy chain-only antibodies
WO2022187406A1 (en) 2021-03-03 2022-09-09 Juno Therapeutics, Inc. Combination of a t cell therapy and a dgk inhibitor
WO2022204071A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Method to assess potency of viral vector particles
WO2022204070A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Methods of determining potency of a therapeutic cell composition
WO2022221726A2 (en) 2021-04-16 2022-10-20 Juno Therapeutics, Inc. Combination therapies with bcma-directed t cell therapy
WO2022221737A1 (en) 2021-04-16 2022-10-20 Juno Therapeutics, Inc. T cell therapy in patients who have had prior stem cell transplant
US11479755B2 (en) 2015-12-07 2022-10-25 2Seventy Bio, Inc. T cell compositions
US11492409B2 (en) 2018-06-01 2022-11-08 Novartis Ag Binding molecules against BCMA and uses thereof
WO2022234009A2 (en) 2021-05-06 2022-11-10 Juno Therapeutics Gmbh Methods for stimulating and transducing t cells
WO2022241430A1 (en) 2021-05-11 2022-11-17 Janssen Biotech, Inc. Methods and compositions for monitoring the treatment of relapsed and/or refractory multiple myeloma
US11505606B2 (en) 2016-09-14 2022-11-22 Teneobio, Inc. CD3 binding antibodies
WO2022246293A1 (en) 2021-05-19 2022-11-24 Sana Biotechnology, Inc. Hypoimmunogenic rhd negative primary t cells
WO2022251367A1 (en) 2021-05-27 2022-12-01 Sana Biotechnology, Inc. Hypoimmunogenic cells comprising engineered hla-e or hla-g
WO2023278811A1 (en) 2021-07-01 2023-01-05 Indapta Therapeutics, Inc. Engineered natural killer (nk) cells and related methods
WO2023287827A2 (en) 2021-07-14 2023-01-19 Sana Biotechnology, Inc. Altered expression of y chromosome-linked antigens in hypoimmunogenic cells
WO2023015217A1 (en) 2021-08-04 2023-02-09 Sana Biotechnology, Inc. Use of cd4-targeted viral vectors
WO2023019203A1 (en) 2021-08-11 2023-02-16 Sana Biotechnology, Inc. Inducible systems for altering gene expression in hypoimmunogenic cells
US11613572B2 (en) 2016-06-21 2023-03-28 Teneobio, Inc. CD3 binding antibodies
US11635435B2 (en) 2017-06-13 2023-04-25 Oncotracker, Inc. Diagnostic, prognostic, and monitoring methods for solid tumor cancers
WO2023069790A1 (en) 2021-10-22 2023-04-27 Sana Biotechnology, Inc. Methods of engineering allogeneic t cells with a transgene in a tcr locus and associated compositions and methods
WO2023081735A1 (en) 2021-11-03 2023-05-11 Celgene Corporation Chimeric antigen receptors specific for b-cell maturation antigen for use in treating myeloma
WO2023115039A2 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae fusion glycoproteins
WO2023115041A1 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae attachment glycoproteins
WO2023122337A1 (en) 2021-12-23 2023-06-29 Sana Biotechnology, Inc. Chimeric antigen receptor (car) t cells for treating autoimmune disease and associated methods
WO2023129929A1 (en) 2021-12-30 2023-07-06 Tr1X, Inc. Cd4+ t cells expressing il-10 and chimeric antigen receptors and uses thereof
WO2023133595A2 (en) 2022-01-10 2023-07-13 Sana Biotechnology, Inc. Methods of ex vivo dosing and administration of lipid particles or viral vectors and related systems and uses
WO2023147515A1 (en) 2022-01-28 2023-08-03 Juno Therapeutics, Inc. Methods of manufacturing cellular compositions
WO2023150518A1 (en) 2022-02-01 2023-08-10 Sana Biotechnology, Inc. Cd3-targeted lentiviral vectors and uses thereof
WO2023150647A1 (en) 2022-02-02 2023-08-10 Sana Biotechnology, Inc. Methods of repeat dosing and administration of lipid particles or viral vectors and related systems and uses
US11733236B2 (en) 2014-03-26 2023-08-22 Helmholtz Zentrum München—Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Flattop (fltp) is a novel biomarker for beta cell maturation
WO2023158836A1 (en) 2022-02-17 2023-08-24 Sana Biotechnology, Inc. Engineered cd47 proteins and uses thereof
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
WO2023215725A1 (en) 2022-05-02 2023-11-09 Fred Hutchinson Cancer Center Compositions and methods for cellular immunotherapy
WO2023213969A1 (en) 2022-05-05 2023-11-09 Juno Therapeutics Gmbh Viral-binding protein and related reagents, articles, and methods of use
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
WO2023220641A2 (en) 2022-05-11 2023-11-16 Juno Therapeutics, Inc. Methods and uses related to t cell therapy and production of same
US11827713B2 (en) 2017-06-27 2023-11-28 Nanjing Legend Biotech Co., Ltd. Chimeric antibody immune effector cell engagers and methods of use thereof
WO2023230581A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Methods of manufacturing t cell therapies
WO2023230548A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Method for predicting response to a t cell therapy
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2024007020A1 (en) 2022-06-30 2024-01-04 Indapta Therapeutics, Inc. Combination of engineered natural killer (nk) cells and antibody therapy and related methods
EP4311579A1 (en) * 2022-07-29 2024-01-31 Association Française contre les Myopathies B cell-specific mab-sirna conjugates improve myasthenia
WO2024026377A1 (en) 2022-07-27 2024-02-01 Sana Biotechnology, Inc. Methods of transduction using a viral vector and inhibitors of antiviral restriction factors
WO2024031091A2 (en) 2022-08-05 2024-02-08 Juno Therapeutics, Inc. Chimeric antigen receptors specific for gprc5d and bcma
US11905326B2 (en) 2019-06-14 2024-02-20 Teneobio, Inc. Multispecific heavy chain antibodies binding to CD22 and CD3
WO2024054944A1 (en) 2022-09-08 2024-03-14 Juno Therapeutics, Inc. Combination of a t cell therapy and continuous or intermittent dgk inhibitor dosing
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
WO2024064838A1 (en) 2022-09-21 2024-03-28 Sana Biotechnology, Inc. Lipid particles comprising variant paramyxovirus attachment glycoproteins and uses thereof
US11965022B2 (en) 2021-12-23 2024-04-23 Sana Biotechnology, Inc. Methods and compositions for modulating CAR-T activity

Families Citing this family (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2720682A1 (en) * 2008-04-25 2009-10-29 Zymogenetics, Inc. Levels of bcma protein expression on b cells and use in diagnostic methods
ES2959480T3 (en) 2014-02-07 2024-02-26 Univ Mcmaster T cell coupler - trifunctional antigen and methods and uses thereof
CN116808219A (en) * 2015-07-24 2023-09-29 安可初克公司 Gamma secretase modulators for the treatment of immune system dysfunction
TWI829617B (en) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Antibody constructs for flt3 and cd3
TWI796283B (en) 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Antibody constructs for msln and cd3
EP3858388A1 (en) 2015-09-28 2021-08-04 Regents of the University of Minnesota Chimeric antigen receptor (car) t cells as therapeutic interventions for auto- and allo-immunity
KR20190008171A (en) * 2015-11-13 2019-01-23 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 Anti-BCMA polypeptides and proteins
EP3411402B1 (en) 2016-02-03 2021-12-22 Amgen Research (Munich) GmbH Bcma and cd3 bispecific t cell engaging antibody constructs
DK3411404T3 (en) 2016-02-03 2023-01-30 Amgen Res Munich Gmbh PSMA-AND CD3-BISPECIFIC T CELL-ENGINEERING ANTIBODY CONSTRUCTS
EA039859B1 (en) 2016-02-03 2022-03-21 Эмджен Рисерч (Мюник) Гмбх Bispecific antibody constructs binding egfrviii and cd3
RU2018134771A (en) 2016-03-04 2020-04-06 Новартис Аг CELLS EXPRESSING MANY MOLECULES OF CHIMERIC ANTIGENIC RECEPTORS (CAR), AND THEIR APPLICATION
BR112018070073A2 (en) 2016-04-01 2019-02-12 Kite Pharma, Inc. chimeric antigen and t-cell receptors and methods of use
CR20210084A (en) 2016-04-01 2021-03-26 Amgen Inc Chimeric receptors and methods of use thereof
AU2017240150C1 (en) 2016-04-01 2022-10-27 Kite Pharma, Inc. BCMA binding molecules and methods of use thereof
CN110582509A (en) 2017-01-31 2019-12-17 诺华股份有限公司 Treatment of cancer using chimeric T cell receptor proteins with multispecific properties
AU2018240938A1 (en) 2017-03-24 2019-10-10 Zenyaku Kogyo Co., Ltd. Anti-IgM/B cell surface antigen bispecific antibody
US20200055948A1 (en) 2017-04-28 2020-02-20 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
CA3067584A1 (en) 2017-06-20 2018-12-27 Teneobio, Inc. Anti-bcma heavy chain-only antibodies
CN117567624A (en) 2017-06-20 2024-02-20 特纳奥尼股份有限公司 Heavy chain-only anti-BCMA antibodies
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. Multispecific molecules that bind to bcma and uses thereof
CN111133007A (en) 2017-09-13 2020-05-08 特尼奥生物股份有限公司 Heavy chain antibodies that bind extracellular enzymes
WO2019071358A1 (en) 2017-10-12 2019-04-18 Mcmaster University T cell-antigen coupler with y182t mutation and methods and uses thereof
US20200339704A1 (en) 2017-10-18 2020-10-29 James E. Bradner Compositions and methods for selective protein degradation
WO2019099639A1 (en) 2017-11-15 2019-05-23 Navartis Ag Bcma-targeting chimeric antigen receptor, cd19-targeting chimeric antigen receptor, and combination therapies
TW201925782A (en) 2017-11-30 2019-07-01 瑞士商諾華公司 BCMA-targeting chimeric antigen receptor, and uses thereof
KR20200104342A (en) 2017-12-22 2020-09-03 테네오바이오, 인코포레이티드 Heavy chain antibody that binds to CD22
EP3737408A1 (en) 2018-01-08 2020-11-18 Novartis AG Immune-enhancing rnas for combination with chimeric antigen receptor therapy
EP3746116A1 (en) 2018-01-31 2020-12-09 Novartis AG Combination therapy using a chimeric antigen receptor
TWI728308B (en) 2018-02-01 2021-05-21 大陸商南京馴鹿醫療技術有限公司 A chimeric antigen receptor (car) binding to bcma and use thereof
CN110662771B (en) 2018-02-01 2023-07-28 南京驯鹿生物技术股份有限公司 Chimeric Antigen Receptor (CAR) combined with BCMA and application thereof
EP3752203A1 (en) 2018-02-13 2020-12-23 Novartis AG Chimeric antigen receptor therapy in combination with il-15r and il15
WO2019226658A1 (en) 2018-05-21 2019-11-28 Compass Therapeutics Llc Multispecific antigen-binding compositions and methods of use
CN112105391A (en) * 2018-06-26 2020-12-18 爱必乐生物公司 anti-BCMA antibodies and uses thereof
CA3105448A1 (en) 2018-07-03 2020-01-09 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
US11110123B2 (en) 2018-07-17 2021-09-07 Triumvira Immunologics Usa, Inc. T cell-antigen coupler with various construct optimizations
KR20210034032A (en) 2018-07-19 2021-03-29 리제너론 파아마슈티컬스, 인크. Bispecific anti-BCMA x anti-CD3 antibodies and uses thereof
CN116769032A (en) 2018-07-20 2023-09-19 坦尼奥第二公司 Heavy chain antibodies that bind CD19
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
SG11202101825QA (en) 2018-08-31 2021-03-30 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
EP3870611A1 (en) 2018-10-26 2021-09-01 TeneoBio, Inc. Heavy chain antibodies binding to cd38
CA3126087A1 (en) 2019-02-25 2020-09-03 Novartis Ag Mesoporous silica particles compositions for viral delivery
EP3942025A1 (en) 2019-03-21 2022-01-26 Novartis AG Car-t cell therapies with enhanced efficacy
CA3133654A1 (en) 2019-04-05 2020-10-08 Teneobio, Inc. Heavy chain antibodies binding to psma
US20220168389A1 (en) 2019-04-12 2022-06-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
US20230374130A1 (en) 2019-07-30 2023-11-23 Qlsf Biotherapeutics, Inc. Bispecific anti lrrc15 and cd3epsilon antibodies
CN112409482B (en) * 2019-08-20 2022-08-26 杭州尚健生物技术有限公司 BCMA antibodies
WO2021051390A1 (en) * 2019-09-20 2021-03-25 上海吉倍生物技术有限公司 Bcma-targeted antibody and chimeric antigen receptor
EP4065158A2 (en) 2019-11-26 2022-10-05 Novartis AG Chimeric antigen receptors binding bcma and cd19 and uses thereof
WO2021132746A1 (en) * 2019-12-24 2021-07-01 Abl Bio, Inc. Anti-bcma/anti-4-1bb bispecific antibodies and uses thereof
CA3158579A1 (en) 2019-12-18 2021-06-24 Pranjali DALVI Heavy chain antibodies binding to cd38
WO2021146487A2 (en) 2020-01-14 2021-07-22 Synthekine, Inc. Il2 orthologs and methods of use
CN111234020B (en) * 2020-01-23 2020-10-23 和铂医药(苏州)有限公司 BCMA binding protein and preparation method and application thereof
CN113248611A (en) * 2020-02-13 2021-08-13 湖南华康恒健生物技术有限公司 anti-BCMA antibody, pharmaceutical composition and application thereof
US20230256017A1 (en) 2020-02-27 2023-08-17 Jennifer Brogdon Methods of making chimeric antigen receptor-expressing cells
CN115397460A (en) 2020-02-27 2022-11-25 诺华股份有限公司 Methods of making cells expressing chimeric antigen receptors
WO2021222578A1 (en) 2020-04-29 2021-11-04 Teneobio, Inc. Multispecific heavy chain antibodies with modified heavy chain constant regions
AU2021288224A1 (en) 2020-06-11 2023-01-05 Novartis Ag ZBTB32 inhibitors and uses thereof
US20230257473A1 (en) 2020-06-30 2023-08-17 Teneobio, Inc. Multi-specific antibodies binding to bcma
WO2022037528A1 (en) * 2020-08-20 2022-02-24 正大天晴药业集团股份有限公司 Single variable domain and antigen binding molecule binding bcma
TW202227124A (en) 2020-08-21 2022-07-16 瑞士商諾華公司 Compositions and methods for in vivo generation of car expressing cells
CA3191224A1 (en) 2020-10-16 2022-04-21 Shenda GU Multispecific binding compounds that bind to pd-l1
TW202233684A (en) 2020-11-18 2022-09-01 美商泰尼歐生物公司 Heavy chain antibodies binding to folate receptor alpha
CN112321713B (en) * 2020-12-31 2021-05-25 北京艺妙神州医药科技有限公司 anti-BCMA antibody and application thereof
CA3211138A1 (en) 2021-02-25 2022-09-01 Teneobio, Inc. Anti-psma antibodies and car-t structures
CN116917327A (en) 2021-02-26 2023-10-20 特尼奥生物股份有限公司 anti-MUC 1-C antibodies and CAR-T structures
CN116997654A (en) 2021-03-17 2023-11-03 第一三共株式会社 Genes encoding chimeric receptors for anti-acetylcholine receptor autoantibodies
TW202304994A (en) 2021-04-02 2023-02-01 美商泰尼歐生物公司 Agonistic anti-il-2r antibodies and methods of use
JP2024514107A (en) 2021-04-06 2024-03-28 テネオバイオ, インコーポレイテッド Anti-CD19 antibody and CAR-T construct
WO2022216993A2 (en) 2021-04-08 2022-10-13 Marengo Therapeutics, Inc. Multifuntional molecules binding to tcr and uses thereof
AU2022259688A1 (en) 2021-04-16 2023-10-05 Teneobio, Inc. Anti-cd20 antibodies and car-t structures
CA3218362A1 (en) 2021-04-27 2022-11-03 Novartis Ag Viral vector production system
WO2022271987A1 (en) 2021-06-23 2022-12-29 TeneoFour, Inc. Anti-cd38 antibodies and epitopes of same
US11453723B1 (en) 2021-06-25 2022-09-27 Mcmaster University BCMA T cell-antigen couplers and uses thereof
AU2022310862A1 (en) 2021-07-14 2024-02-01 2Seventy Bio, Inc. Engineered t cell receptors fused to binding domains from antibodies
WO2023004197A1 (en) 2021-07-23 2023-01-26 Teneoten, Inc. Heavy chain antibodies binding to hepatitis b surface antigen
CN117858901A (en) 2021-08-20 2024-04-09 诺华股份有限公司 Method for preparing cells expressing chimeric antigen receptor
WO2023064876A1 (en) 2021-10-14 2023-04-20 Teneobio, Inc. Mesothelin binding proteins and uses thereof
CN114276452A (en) * 2021-12-29 2022-04-05 源道隆(苏州)医学科技有限公司 Nano antibody capable of being combined with BCMA (brain cell activating antigen) and application thereof
WO2023152747A1 (en) 2022-02-09 2023-08-17 Hadasit Medical Research Services And Development Ltd. Anti-bcma car to target immune-related disorders, compositions and method thereof
WO2023196997A2 (en) 2022-04-08 2023-10-12 2Seventy Bio, Inc. Multipartite receptor and signaling complexes
WO2024026358A1 (en) 2022-07-27 2024-02-01 Teneobio, Inc. Mesothelin binding proteins and uses thereof
WO2024077044A1 (en) 2022-10-05 2024-04-11 Amgen Inc. Combination therapies comprising t-cell redirecting therapies and agonistic anti-il-2r antibodies or fragments thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US6632927B2 (en) 1989-12-21 2003-10-14 Celltech Therapeutics Limited Humanized antibodies
US7112421B2 (en) 2000-09-18 2006-09-26 Biogen Idec Ma Inc. Nucleic acids encoding BAFF receptor, chimeric proteins and methods and compositions related thereto

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060067933A1 (en) 1999-01-07 2006-03-30 Gross Jane A Soluble receptor BR43x2 and methods of using
DK1642973T3 (en) 1999-01-07 2009-11-02 Zymogenetics Inc Therapeutic applications of BR43X2-soluble receptors
US7833529B1 (en) 1999-01-07 2010-11-16 Zymogenetics, Inc. Methods for inhibiting B lymphocyte proliferation with soluble ztnf4 receptor
WO2000068378A1 (en) 1999-05-06 2000-11-16 National Jewish Medical And Research Center Tall-1 nucleic acid molecules, proteins, receptors and methods of use thereof
EE05673B1 (en) 1999-08-17 2013-08-15 Biogen, Inc. BAFF receptor (BCMA), an immunoregulatory agent
EP1666052B1 (en) 2000-02-16 2011-06-08 Genentech, Inc. Anti-APRIL monoclonal antibody and its use for the treatment of an immune related disease or cancer
US20040002068A1 (en) 2000-03-01 2004-01-01 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US7371388B1 (en) 2000-05-04 2008-05-13 Human Genome Sciences, Inc. Treatment of Sjogren's syndrome by administration of TR18 polypeptides
US20030012783A1 (en) 2001-02-20 2003-01-16 Wayne Kindsvogel Antibodies that bind both BCMA and TACI
CU23007A1 (en) * 2001-04-06 2004-12-17 Ct De Inmunologia Molecular Ct De Inmunologia Mole IMMUNOTHERAPEUTIC COMBINATIONS FOR THE TREATMENT IMMUNOTHERAPEUTIC COMBINATIONS FOR THE TREATMENT OF TUMORS THAT OVER-EXPRESS GANGLIOSIDES TO OF TUMORS THAT OVER-EXPRESS GANGLOSIDES
WO2003013582A1 (en) 2001-08-06 2003-02-20 Genset S.A. Genoxit agonists and antagonists for use in the treatment of metabolic disorders
AU2003221256A1 (en) * 2002-02-21 2003-09-09 Biogen Idec Ma Inc. Use of bcma as an immunoregulatory agent
ES2871905T3 (en) * 2002-03-01 2021-11-02 Immunomedics Inc Immunoconjugate comprising humanized RS7 antibodies
EP2322202A3 (en) 2002-10-29 2011-07-27 Genentech, Inc. Compositions and methods for the treatment of immune diseases
US20050163775A1 (en) 2003-06-05 2005-07-28 Genentech, Inc. Combination therapy for B cell disorders
WO2005005462A2 (en) 2003-06-05 2005-01-20 Genentech, Inc. Blys antagonists and uses thereof
US20090226440A1 (en) 2005-08-12 2009-09-10 Shane Grey Prophylactic and/or Therapeutic Method for Treatment of Autoimmune Disease
AU2006344395B2 (en) 2005-10-13 2013-05-02 Human Genome Sciences, Inc. Methods and compositions for use in treatment of patients with autoantibody positive diseases
DOP2006000277A (en) * 2005-12-12 2007-08-31 Bayer Pharmaceuticals Corp ANTI MN ANTIBODIES AND METHODS FOR USE
JP2007309746A (en) * 2006-05-17 2007-11-29 Osaka Univ Method of analyzing binding site of cell membrane surface substance
EP2069393A2 (en) 2006-10-10 2009-06-17 The Government of the United States of America as represented by the Secretary of the Department of Health and Human Services Avian influenza vaccine

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US6632927B2 (en) 1989-12-21 2003-10-14 Celltech Therapeutics Limited Humanized antibodies
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US7112421B2 (en) 2000-09-18 2006-09-26 Biogen Idec Ma Inc. Nucleic acids encoding BAFF receptor, chimeric proteins and methods and compositions related thereto

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
AVERY ET AL., J CLIN INVEST, vol. 112, 2003, pages 286 - 297
BETTER ET AL., SCIENCE, vol. 240, 1988, pages 1041 - 1043
BREZINSKY ET AL., J IMMUNOL METHODS, vol. 277, 2003, pages 141 - 155
CHIU ET AL., BLOOD, vol. 109, 2007, pages 729 - 739
DARCE ET AL., J IMMUNOL, vol. 179, 2007, pages 7276 - 7286
HARLOW; LANE: "Using Antibodies: A Laboratory Manual: Portable Protocol", 1998, COLD SPRING HARBOR LABORATORY
HATZOGLOU ET AL., J IMMUNOL, vol. 165, 2000, pages 1322 - 1330
HE ET AL., J IMMUNOL, vol. 172, 2004, pages 3268 - 3279
HUANG ET AL., PROC NATL ACAD SCI U S A, vol. 101, 2004, pages 17789 - 17794
HUSE ET AL., SCIENCE, vol. 246, 1989, pages 1275 - 1281
JOHNSON; WU, NUCLEIC ACIDS RES, vol. 28, 2000, pages 214 - 218
KALLED ET AL., CURR DIR AUTOIMMUN, vol. 8, 2005, pages 206 - 242
LITINSKIY ET AL., NAT IMMUNOL, vol. 3, 2002, pages 822 - 829
LIU, PROC. NATL. ACAD. SCI. USA, vol. 84, 1987, pages 3439 - 3443
MITCHELL E. REFF: "Depletion of B Cells In Vivo by a Chimeric Mouse Human Monoclonal Antibody to CD20", BLOOD, vol. 83, 15 January 1994 (1994-01-15), pages 435 - 445
NOVAK ET AL., BLOOD, vol. 103, 2004, pages 689 - 694
NOVAK ET AL., BLOOD, vol. 104, 2004, pages 2247 - 2253
O'CONNOR ET AL., J EXP MED, vol. 199, 2004, pages 91 - 98
POMERANTZ; BALTIMORE, MOL CELL, vol. 10, 2002, pages 693 - 695
SCHIEMANN ET AL., SCIENCE, vol. 293, 2001, pages 2111 - 2114
SIMS ET AL., BLOOD, vol. 105, 2005, pages 4390 - 4398
XU; LAM, MOL CELL BIOL, vol. 21, 2001, pages 4067 - 4074
ZHANG ET AL., INT IMMUNOL, vol. 17, 2005, pages 779 - 788

Cited By (281)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130273055A1 (en) * 2010-11-16 2013-10-17 Eric Borges Agents and methods for treating diseases that correlate with bcma expression
US20130101599A1 (en) * 2011-04-21 2013-04-25 Boehringer Ingelheim International Gmbh Bcma-based stratification and therapy for multiple myeloma patients
EP2699259B1 (en) 2011-04-21 2016-07-27 Boehringer Ingelheim International GmbH Bcma-based stratification and therapy for multiple myeloma patients
JP2014520248A (en) * 2011-04-21 2014-08-21 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング BCMA-based stratification and treatment of patients with multiple myeloma
US11013800B2 (en) 2011-05-16 2021-05-25 Evive Biotech Ltd. Multi-specific Fab fusion proteins comprising a CD3-binding Fab fragment with N-terminal fusion to binding domains and methods of use
JP2022033797A (en) * 2011-05-27 2022-03-02 グラクソ グループ リミテッド Bcma (cd269/tnfrsf17)-binding proteins
AU2012264890B2 (en) * 2011-05-27 2015-10-15 Glaxo Group Limited BCMA (CD269/TNFRSF17) -binding proteins
WO2012163805A1 (en) * 2011-05-27 2012-12-06 Glaxo Group Limited Bcma (cd269/tnfrsf17) -binding proteins
EP3693394A1 (en) * 2011-05-27 2020-08-12 Glaxo Group Limited Antigen binding proteins
KR20140036272A (en) * 2011-05-27 2014-03-25 글락소 그룹 리미티드 Bcma(cd269/tnfrsf17)-binding proteins
JP7260621B2 (en) 2011-05-27 2023-04-18 グラクソ グループ リミテッド BCMA (CD269/TNFRSF17) binding protein
JP2014520088A (en) * 2011-05-27 2014-08-21 グラクソ グループ リミテッド BCMA (CD269 / TNFRSF17) binding protein
JP7475515B2 (en) 2011-05-27 2024-04-26 グラクソ グループ リミテッド BCMA (CD269/TNFRSF17) binding protein
JP2019147816A (en) * 2011-05-27 2019-09-05 グラクソ グループ リミテッドGlaxo Group Limited Bcma (cd269/tnfrsf17)-binding proteins
EA028220B1 (en) * 2011-05-27 2017-10-31 Глаксо Груп Лимитед Immunoconjugate comprising bcma (cd269/tnfrsf17) binding protein, medical use thereof and pharmaceutical composition
JP7018910B2 (en) 2011-05-27 2022-02-14 グラクソ グループ リミテッド BCMA (CD269 / TNFRSF17) binding protein
TWI609883B (en) * 2011-05-27 2018-01-01 葛蘭素集團有限公司 Bcma binding proteins, immunoconjugate thereof and pharmaceutical composition comprising the same
AU2012264890C1 (en) * 2011-05-27 2016-03-10 Glaxo Group Limited BCMA (CD269/TNFRSF17) -binding proteins
JP2018087190A (en) * 2011-05-27 2018-06-07 グラクソ グループ リミテッドGlaxo Group Limited Bcma (cd269/tnfrsf17)-binding proteins
KR101972446B1 (en) 2011-05-27 2019-04-25 글락소 그룹 리미티드 Bcma(cd269/tnfrsf17)-binding proteins
US9273141B2 (en) 2011-05-27 2016-03-01 Glaxo Group Limited B cell maturation antigen (BCMA) binding proteins
TWI644924B (en) * 2011-05-27 2018-12-21 英商葛蘭素集團有限公司 Bcma binding protein, immunoconjugate thereof and pharmaceutical composition comprising the same
EP3415531A1 (en) * 2011-05-27 2018-12-19 Glaxo Group Limited Bcma (cd269/tnfrsf17) - binding proteins
US11419945B2 (en) 2011-05-27 2022-08-23 Glaxo Group Limited Antigen binding proteins
JP2020202838A (en) * 2011-11-15 2020-12-24 アムゲン リサーチ (ミュンヘン) ゲーエムベーハーAMGEN Research(Munich)GmbH Binding molecules for bcma and cd3
US10766969B2 (en) 2011-11-15 2020-09-08 Amgen Inc. Binding molecules for BCMA and CD3
US9150664B2 (en) 2011-11-15 2015-10-06 Amgen Research (Munich) Gmbh Binding molecules for BCMA and CD3
US9340621B2 (en) * 2011-11-15 2016-05-17 Boehringer Ingelheim International Gmbh Binding molecules for BCMA and CD3
US9598500B2 (en) 2011-11-15 2017-03-21 Amgen Inc. Binding molecules for BCMA and CD3
EP2780374B1 (en) 2011-11-15 2019-08-21 Amgen Research (Munich) GmbH Binding molecules for bcma and cd3
US20130156769A1 (en) * 2011-11-15 2013-06-20 Boehringer Ingelheim International Gmbh Binding molecules for bcma and cd3
JP7250736B2 (en) 2011-11-15 2023-04-03 アムゲン リサーチ (ミュンヘン) ゲーエムベーハー Binding molecules for BCMA and CD3
EP2780375B1 (en) 2011-11-15 2019-09-11 Amgen Research (Munich) GmbH Binding molecules for bcma and cd3
AU2017208279B2 (en) * 2012-04-11 2019-03-07 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting b-cell maturation antigen
CN110331154A (en) * 2012-04-11 2019-10-15 美国卫生和人力服务部 Target the Chimeric antigen receptor of B- cell maturation antigen
US10844387B2 (en) 2012-04-11 2020-11-24 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US10837019B2 (en) 2012-04-11 2020-11-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US10876123B2 (en) 2012-04-11 2020-12-29 The United States Of America,As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US9765342B2 (en) 2012-04-11 2017-09-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
WO2013154760A1 (en) 2012-04-11 2013-10-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting b-cell maturation antigen
US10829769B2 (en) 2012-04-11 2020-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting b-cell maturation antigen
US10829767B2 (en) 2012-04-11 2020-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US10829768B2 (en) 2012-04-11 2020-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US10815488B2 (en) 2012-04-11 2020-10-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
CN104379179A (en) * 2012-04-11 2015-02-25 美国卫生和人力服务部 Chimeric antigen receptors targeting b-cell maturation antigen
US10815487B2 (en) 2012-04-11 2020-10-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US10900042B2 (en) 2012-04-11 2021-01-26 The United States of Americans, as represented by the Secretary, Department of Health and Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US10767184B2 (en) 2012-04-11 2020-09-08 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US11773396B2 (en) 2012-04-11 2023-10-03 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
KR20150003306A (en) * 2012-04-11 2015-01-08 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 Chimeric antigen receptors targeting b-cell maturation antigen
JP2015513920A (en) * 2012-04-11 2015-05-18 アメリカ合衆国 Chimeric antigen receptor targeting B cell maturation antigen
US10738312B2 (en) 2012-04-11 2020-08-11 The United States Of America,As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
JP2018161140A (en) * 2012-04-11 2018-10-18 アメリカ合衆国 Chimeric antigen receptors targeting b-cell maturation antigen
US10738313B2 (en) 2012-04-11 2020-08-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US11377660B2 (en) 2012-04-11 2022-07-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
EP3689383A1 (en) 2012-04-11 2020-08-05 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting b-cell maturation antigen
AU2019203042B2 (en) * 2012-04-11 2021-03-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
US11359204B2 (en) 2012-04-11 2022-06-14 The United States of Americans represented by the Secretary, Department of Health and Human Services Chimeric antigen receptors targeting B-cell maturation antigen
JP2020078322A (en) * 2012-04-11 2020-05-28 アメリカ合衆国 Chimeric antigen receptors targeting b-cell maturation antigen
KR102229945B1 (en) 2012-04-11 2021-03-18 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 Chimeric antigen receptors targeting b-cell maturation antigen
KR20200027047A (en) * 2012-04-11 2020-03-11 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 Chimeric antigen receptors targeting b-cell maturation antigen
RU2766608C2 (en) * 2012-04-11 2022-03-15 Дзе Юнайтед Стейтс Оф Америка, Эз Репрезентед Бай Дзе Секретари, Департмент Оф Хелс Энд Хьюман Сёрвисез Chimeric antigen receptors targeted b-cell maturation antigen
KR102086874B1 (en) 2012-04-11 2020-03-10 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 Chimeric antigen receptors targeting b-cell maturation antigen
US11066674B2 (en) 2012-04-11 2021-07-20 The United States Of America,As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting b-cell maturation antigen
US11827889B2 (en) 2012-04-11 2023-11-28 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
CN110295186A (en) * 2012-04-11 2019-10-01 美国卫生和人力服务部 Target the Chimeric antigen receptor of B- cell maturation antigen
AU2013246443B2 (en) * 2012-04-11 2017-04-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting B-cell maturation antigen
EA033110B1 (en) * 2012-04-11 2019-08-30 Дзе Юнайтед Стейтс Оф Америка, Эз Репрезентед Бай Дзе Секретари, Департмент Оф Хелс Энд Хьюман Сёрвисез Chimeric antigen receptor targeting b-cell maturation antigen, nucleic acid encoding same, corresponding expression vector, host cell, uses and methods
JP2022058345A (en) * 2012-04-11 2022-04-12 アメリカ合衆国 Chimeric antigen receptors targeting b-cell maturation antigen
EP3508503A1 (en) 2012-11-01 2019-07-10 Max-Delbrück-Centrum für Molekulare Medizin Antibody against cd269 (bcma)
US10189906B2 (en) 2012-11-01 2019-01-29 Max-Delrück-Centrum Für Molekulare Medizin Antibody that binds CD269 (BCMA) suitable for use in the treatment of plasma cell diseases such as multiple myeloma and autoimmune diseases
US10745486B2 (en) 2012-11-01 2020-08-18 Max-Delbrück-Centrum für Molekulare Medizin Antibody that binds CD269 (BCMA) suitable for use in the treatment of plasma cell diseases such as multiple myeloma and autoimmune diseases
WO2014068079A1 (en) 2012-11-01 2014-05-08 Max-Delbrück-Centrum für Molekulare Medizin An antibody that binds cd269 (bcma) suitable for use in the treatment of plasma cell diseases such as multiple myeloma and autoimmune diseases
US11667722B2 (en) 2012-11-01 2023-06-06 Max-Delbrück-Centrum Für Molekulare Medizin In Der Helmholtz-Gemeinschaft Antibody that binds CD269 (BCMA) suitable for use in the treatment of plasma cell diseases such as multiple myeloma and autoimmune diseases
US11130820B2 (en) 2012-12-20 2021-09-28 Celgene Corporation Chimeric antigen receptors
WO2014122143A1 (en) 2013-02-05 2014-08-14 Engmab Ag Method for the selection of antibodies against bcma
EP3620468A1 (en) 2013-02-05 2020-03-11 EngMab Sàrl Method for the selection of antibodies against bcma
WO2014122144A1 (en) 2013-02-05 2014-08-14 Engmab Ag BISPECIFIC ANTIBODIES AGAINST CD3ε AND BCMA
US10851171B2 (en) 2013-02-05 2020-12-01 Engmab Sarl Method for the selection of antibodies against BCMA
US10077315B2 (en) 2013-02-05 2018-09-18 Engmab Sàrl Bispecific antibodies against CD3 and BCMA
EP2762497A1 (en) 2013-02-05 2014-08-06 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
US9963513B2 (en) 2013-02-05 2018-05-08 Engmab Sàrl Method for the selection of antibodies against BCMA
EP2762496A1 (en) 2013-02-05 2014-08-06 EngMab AG Method for the selection of antibodies against BCMA
US11806365B2 (en) 2013-03-15 2023-11-07 Celgene Corporation Modified T lymphocytes comprising a CD52 antibody-inducible caspase and methods of apoptosis
US10967005B2 (en) 2013-03-15 2021-04-06 Celgene Corporation Modified T lymphocytes comprising a BAFF antibody-inducible caspase and methods of apoptosis
US10752694B2 (en) 2013-03-15 2020-08-25 Amgen Inc. Binding molecules for BCMA and CD3
WO2014140248A1 (en) * 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Binding molecules for bcma and cd3
EP3912997A3 (en) * 2013-03-15 2021-12-15 Amgen Research (Munich) GmbH Binding molecules for bcma and cd3
TWI646107B (en) * 2013-03-15 2019-01-01 安進研究(慕尼黑)有限責任公司 BCMA and CD3 binding molecules
AU2014230217B2 (en) * 2013-03-15 2019-02-07 Amgen Inc. Binding molecules for BCMA and CD3
US11733236B2 (en) 2014-03-26 2023-08-22 Helmholtz Zentrum München—Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Flattop (fltp) is a novel biomarker for beta cell maturation
US10774343B2 (en) 2014-04-25 2020-09-15 Bluebird Bio, Inc. MND promoter chimeric antigen receptors
US11820829B2 (en) 2014-04-30 2023-11-21 Max-Delbrück-Centrum Für Molekulare Medizin In Der Helmholtz-Gemeinschaft Humanized antibodies against CD269 (BCMA)
US10144782B2 (en) 2014-04-30 2018-12-04 Max-Delbrück-Centrum Für Molekulare Medizin In Der Helmholtz-Gemeinschaft Humanized antibodies against CD269 (BCMA)
US10851172B2 (en) 2014-04-30 2020-12-01 Max-Delbrück-Centrum Für Molekulare Medizin In Der Helmholtz-Gemeinschaft Humanized antibodies against CD269 (BCMA)
US10479975B2 (en) 2014-06-06 2019-11-19 Bluebird Bio, Inc. Methods of making T cell compositions
US11560547B2 (en) 2014-06-06 2023-01-24 2Seventy Bio, Inc. Methods of making T cell compositions
JP2017527271A (en) * 2014-07-21 2017-09-21 ノバルティス アーゲー Treatment of cancer using a humanized anti-BCMA chimeric antigen receptor
US11084880B2 (en) 2014-07-21 2021-08-10 Novartis Ag Anti-BCMA chimeric antigen receptor
US10174095B2 (en) 2014-07-21 2019-01-08 Novartis Ag Nucleic acid encoding a humanized anti-BCMA chimeric antigen receptor
WO2016014565A3 (en) * 2014-07-21 2016-03-10 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
US10428146B2 (en) 2014-07-22 2019-10-01 Cb Therapeutics, Inc. Anti PD-1 antibodies
US11560429B2 (en) 2014-07-22 2023-01-24 Apollomics Inc. Anti PD-1 antibodies
US10981994B2 (en) 2014-07-22 2021-04-20 Apollomics Inc. Anti PD-1 antibodies
EP3172231A4 (en) * 2014-07-24 2018-01-10 Bluebird Bio, Inc. Bcma chimeric antigen receptors
WO2016014789A2 (en) 2014-07-24 2016-01-28 Bluebird Bio, Inc. Bcma chimeric antigen receptors
RU2747457C2 (en) * 2014-07-24 2021-05-05 Блубёрд Био, Инк. Chimeric antigenic receptors to bcma
US10253104B2 (en) 2014-08-04 2019-04-09 Engmab Ag Bispecific antibodies against CD3ϵ and BCMA
EP2982692A1 (en) 2014-08-04 2016-02-10 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
WO2016020332A1 (en) 2014-08-04 2016-02-11 Engmab Ag Bispecific antibodies against cd3epsilon and bcma
EP2982694A1 (en) 2014-08-04 2016-02-10 EngMab AG Bispecific antibodies against cd3epsilon and bcma
WO2016022630A1 (en) * 2014-08-05 2016-02-11 Jiping Zha Anti-pd-l1 antibodies
US10435470B2 (en) 2014-08-05 2019-10-08 Cb Therapeutics, Inc. Anti-PD-L1 antibodies
US11827707B2 (en) 2014-08-05 2023-11-28 Apollomics Inc. Anti PD-L1 antibodies
US11111300B2 (en) 2014-08-05 2021-09-07 Apollomics Inc. Anti PD-L1 antibodies
EP3023437A1 (en) 2014-11-20 2016-05-25 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
EP4282484A2 (en) 2014-12-03 2023-11-29 Bristol-Myers Squibb Company Bispecific antibodies against cd3epsilon and bcma for use in treatment of diseases
EP3029068A1 (en) 2014-12-03 2016-06-08 EngMab AG Bispecific antibodies against CD3epsilon and BCMA for use in the treatment of diseases
US10918665B2 (en) 2014-12-05 2021-02-16 Memorial Sloan Kettering Cancer Center Chimeric antigen receptors targeting B-cell maturation antigen and uses thereof
US10562972B2 (en) 2014-12-05 2020-02-18 Memorial Sloan Kettering Cancer Center Antibodies targeting B-cell maturation antigen and methods of use
US11000549B2 (en) 2014-12-05 2021-05-11 Memorial Sloan Kettering Cancer Center Methods of using chimeric antigen receptors targeting B-cell maturation antigen and uses thereof
US10947314B2 (en) 2014-12-05 2021-03-16 Memorial Sloan Kettering Cancer Center Antibodies targeting b-cell maturation antigen and methods of use
US11725059B2 (en) 2014-12-05 2023-08-15 Memorial Sloan Kettering Cancer Center Antibodies targeting B-cell maturation antigen and methods of use
RU2766094C2 (en) * 2014-12-05 2022-02-07 Мемориал Слоан-Кеттеринг Кэнсер Сентер Antibodies targeting b-cell maturation antigen and methods of the use thereof
US10821135B2 (en) 2014-12-05 2020-11-03 Memorial Sloan Kettering Cancer Center Nucleic acid molecules encoding chimeric antigen receptors targeting B-cell maturation antigen and uses thereof
US10624960B2 (en) 2014-12-12 2020-04-21 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US11382965B2 (en) 2014-12-12 2022-07-12 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US10639359B2 (en) 2014-12-12 2020-05-05 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US10639358B2 (en) 2014-12-12 2020-05-05 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US11633463B2 (en) 2014-12-12 2023-04-25 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US10646558B2 (en) 2014-12-12 2020-05-12 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US10383929B2 (en) 2014-12-12 2019-08-20 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US11351236B2 (en) 2014-12-12 2022-06-07 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US11020466B2 (en) 2014-12-12 2021-06-01 Bluebird Bio, Inc. BCMA chimeric antigen receptors
US11912776B2 (en) 2015-04-13 2024-02-27 Pfizer Inc. Chimeric antigen receptors targeting B-cell maturation antigen
IL290488B1 (en) * 2015-04-13 2024-03-01 Pfizer Therapeutic antibodies and their uses
WO2016166629A1 (en) * 2015-04-13 2016-10-20 Pfizer Inc. Therapeutic antibodies and their uses
US11155630B2 (en) 2015-04-13 2021-10-26 Pfizer Inc. Therapeutic antibodies and their uses
US10793635B2 (en) 2015-04-13 2020-10-06 Pfizer Inc. Therapeutic antibodies and their uses
US9969809B2 (en) 2015-04-13 2018-05-15 Pfizer Inc. Therapeutic antibodies and their uses
TWI703159B (en) * 2015-04-13 2020-09-01 美商輝瑞股份有限公司 Bcma-specific therapeutic antibodies and their uses
TWI703157B (en) * 2015-04-13 2020-09-01 美商輝瑞股份有限公司 Cd3-specific antibodies, therapeutic bispecific antibodies and their uses
US10294304B2 (en) 2015-04-13 2019-05-21 Pfizer Inc. Chimeric antigen receptors targeting B-cell maturation antigen
RU2722381C2 (en) * 2015-04-13 2020-05-29 Пфайзер Инк. Therapeutic antibodies and their application
US10040860B2 (en) 2015-04-13 2018-08-07 Pfizer Inc. Therapeutic antibodies and their uses
US11312782B2 (en) 2015-04-13 2022-04-26 Pfizer Inc. Chimeric antigen receptors targeting B-cell maturation antigen
TWI787645B (en) * 2015-04-13 2022-12-21 美商輝瑞股份有限公司 Cd3-specific antibodies, therapeutic bispecific antibodies and their uses
EP4234581A3 (en) * 2015-04-13 2023-10-04 Pfizer Inc. Therapeutic antibodies and their uses
EP3988117A1 (en) * 2015-04-13 2022-04-27 Pfizer Inc. Therapeutic antibodies and their uses
CN114874331A (en) * 2015-04-13 2022-08-09 辉瑞公司 Therapeutic antibodies and their use
US11814435B2 (en) 2015-04-13 2023-11-14 Pfizer Inc. Therapeutic antibodies and their uses
US11028142B2 (en) 2015-05-18 2021-06-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US20170166622A1 (en) * 2015-05-18 2017-06-15 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10358474B2 (en) * 2015-05-18 2019-07-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10358473B2 (en) * 2015-05-18 2019-07-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10442849B2 (en) * 2015-05-18 2019-10-15 Tcr2 Therabeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
EP3670535A1 (en) 2015-08-03 2020-06-24 EngMab Sàrl Monoclonal antibodies against bcma
US10072088B2 (en) 2015-08-17 2018-09-11 Janssen Pharmaceutica, Nv Anti-BCMA antibodies and uses thereof
EP3757131A1 (en) 2015-08-17 2020-12-30 Janssen Pharmaceutica NV Anti-bcma antibodies, bispecific antigen binding molecules that bind bcma and cd3, and uses thereof
WO2017031104A1 (en) 2015-08-17 2017-02-23 Janssen Pharmaceutica Nv Anti-bcma antibodies, bispecific antigen binding molecules that bind bcma and cd3, and uses thereof
US11479755B2 (en) 2015-12-07 2022-10-25 2Seventy Bio, Inc. T cell compositions
US11698369B2 (en) 2016-01-12 2023-07-11 Oncotracker, Inc. Methods for monitoring immune status of a subject
EP4079320A1 (en) * 2016-01-12 2022-10-26 Inc. Oncotracker Improved methods for monitoring immune status of a subject
WO2017123741A1 (en) 2016-01-12 2017-07-20 Institute For Myeloma & Bone Cancer Research Improved methods for monitoring immune status of a subject
EP3402515A4 (en) * 2016-01-12 2019-07-10 BERENSON, James, R. Improved methods for monitoring immune status of a subject
EP3416687A4 (en) * 2016-02-17 2019-10-23 Seattle Genetics, Inc. Bcma antibodies and use of same to treat cancer and immunological disorders
US11767365B2 (en) 2016-02-17 2023-09-26 Seagen Inc. BCMA antibodies and use of same to treat cancer and immunological disorders
AU2017219747B2 (en) * 2016-02-17 2023-09-28 Seagen Inc. BCMA antibodies and use of same to treat cancer and immunological disorders
US11078291B2 (en) 2016-02-17 2021-08-03 Seagen Inc. BCMA antibodies and use of same to treat cancer and immunological disorders
WO2017143069A1 (en) * 2016-02-17 2017-08-24 Seattle Genetics, Inc. Bcma antibodies and use of same to treat cancer and immunological disorders
US10870701B2 (en) 2016-03-15 2020-12-22 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
US11613572B2 (en) 2016-06-21 2023-03-28 Teneobio, Inc. CD3 binding antibodies
US11242376B2 (en) 2016-08-02 2022-02-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11505606B2 (en) 2016-09-14 2022-11-22 Teneobio, Inc. CD3 binding antibodies
US11377638B2 (en) 2016-10-07 2022-07-05 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11085021B2 (en) 2016-10-07 2021-08-10 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10208285B2 (en) 2016-10-07 2019-02-19 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018083204A1 (en) 2016-11-02 2018-05-11 Engmab Sàrl Bispecific antibody against bcma and cd3 and an immunological drug for combined use in treating multiple myeloma
EP4295918A2 (en) 2016-11-02 2023-12-27 Bristol-Myers Squibb Company Bispecific antibody against bcma and cd3 and an immunological drug for combined use in treating multiple myeloma
US11124577B2 (en) 2016-11-02 2021-09-21 Engmab Sàrl Bispecific antibody against BCMA and CD3 and an immunological drug for combined use in treating multiple myeloma
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11434299B2 (en) 2016-12-21 2022-09-06 Teneobio, Inc. Anti-BCMA heavy chain-only antibodies
WO2018151836A1 (en) 2017-02-17 2018-08-23 Fred Hutchinson Cancer Research Center Combination therapies for treatment of bcma-related cancers and autoimmune disorders
EP4269594A2 (en) 2017-02-17 2023-11-01 Fred Hutchinson Cancer Center Combination therapies for treatment of bcma-related cancers and autoimmune disorders
US11845803B2 (en) 2017-02-17 2023-12-19 Fred Hutchinson Cancer Center Combination therapies for treatment of BCMA-related cancers and autoimmune disorders
WO2018158349A1 (en) 2017-02-28 2018-09-07 Affimed Gmbh Tandem diabody for cd16a-directed nk-cell engagement
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
EP4327878A2 (en) 2017-05-01 2024-02-28 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound
WO2018204427A1 (en) 2017-05-01 2018-11-08 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound
US11635435B2 (en) 2017-06-13 2023-04-25 Oncotracker, Inc. Diagnostic, prognostic, and monitoring methods for solid tumor cancers
WO2018231949A1 (en) 2017-06-14 2018-12-20 Dana-Farber Cancer Institute, Inc. B-cell maturation antigen (bcma)-directed nanoparticles
US11827713B2 (en) 2017-06-27 2023-11-28 Nanjing Legend Biotech Co., Ltd. Chimeric antibody immune effector cell engagers and methods of use thereof
US11912782B2 (en) 2017-08-01 2024-02-27 Medimmune, Llc BCMA monoclonal antibody-drug conjugate
WO2019025983A1 (en) 2017-08-01 2019-02-07 Medimmune, Llc Bcma monoclonal antibody-drug conjugate
US10988546B2 (en) 2017-08-01 2021-04-27 Medimmune, Llc BCMA monoclonal antibody-drug conjugate
EP3689908A4 (en) * 2017-09-29 2021-09-29 Mogam Institute for Biomedical Research Anti-bcma antibody having high affinity for bcma and pharmaceutical composition for treatment of cancer, comprising same
US11066475B2 (en) 2017-11-01 2021-07-20 Juno Therapeutics, Inc. Chimeric antigen receptors specific for B-cell maturation antigen and encoding polynucleotides
WO2019090003A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Chimeric antigen receptors specific for b-cell maturation antigen (bcma)
WO2019089969A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for b-cell maturation antigen
US11623961B2 (en) 2017-11-01 2023-04-11 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for B-cell maturation antigen
WO2019090364A1 (en) 2017-11-06 2019-05-09 Juno Therapeutics, Inc. Combination of a cell therapy and a gamma secretase inhibitor
WO2019113556A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Serum-free media formulation for culturing cells and methods of use thereof
WO2019113557A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Process for producing a composition of engineered t cells
US11807663B2 (en) 2018-02-01 2023-11-07 Innovent Biologics (Suzhou) Co., Ltd. Fully humanized anti-B cell maturation antigen (BCMA) single-chain antibody and use thereof
WO2019149269A1 (en) * 2018-02-01 2019-08-08 信达生物制药(苏州)有限公司 Fully human anti-b cell maturation antigen (bcma) single chain variable fragment, and application thereof
WO2019166650A1 (en) 2018-03-02 2019-09-06 Cdr-Life Ag Trispecific antigen binding proteins
WO2019198051A2 (en) 2018-04-13 2019-10-17 Affimed Gmbh Nk cell engaging antibody fusion constructs
US11492409B2 (en) 2018-06-01 2022-11-08 Novartis Ag Binding molecules against BCMA and uses thereof
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11952428B2 (en) 2018-06-13 2024-04-09 Novartis Ag BCMA chimeric antigen receptors and uses thereof
WO2020033927A2 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
WO2020033916A1 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Methods for assessing integrated nucleic acids
WO2020089343A1 (en) 2018-10-31 2020-05-07 Juno Therapeutics Gmbh Methods for selection and stimulation of cells and apparatus for same
WO2020092854A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Chimeric antigen receptors specific for g protein-coupled receptor class c group 5 member d (gprc5d)
WO2020092848A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Methods for treatment using chimeric antigen receptors specific for b-cell maturation antigen
WO2020097403A1 (en) 2018-11-08 2020-05-14 Juno Therapeutics, Inc. Methods and combinations for treatment and t cell modulation
US10927182B2 (en) 2019-01-16 2021-02-23 Caribou Biosciences, Inc. Humanized BCMA antibody and BCMA-CAR-T cells
US11021542B1 (en) 2019-01-16 2021-06-01 Caribou Biosciences, Inc. Humanized BCMA antibody and BCMA-CAR-T cells
US11142583B2 (en) 2019-01-16 2021-10-12 Caribou Biosciences, Inc. BCMA-CAR-natural killer (NK) cells and methods related thereto
US11840575B2 (en) 2019-05-07 2023-12-12 Gracell Biotechnologies (Shanghai) Co., Ltd. Engineered immune cells targeting BCMA and their uses thereof
WO2020224605A1 (en) 2019-05-07 2020-11-12 亘喜生物科技(上海)有限公司 Bcma-targeting engineered immune cell and use thereof
WO2020224606A1 (en) 2019-05-07 2020-11-12 亘喜生物科技(上海)有限公司 Engineered immune cell targeting bcma and use thereof
US11905326B2 (en) 2019-06-14 2024-02-20 Teneobio, Inc. Multispecific heavy chain antibodies binding to CD22 and CD3
WO2021018168A1 (en) 2019-07-30 2021-02-04 上海翰森生物医药科技有限公司 Anti-bcma antibody, antigen-binding fragment thereof and medical use thereof
WO2021136323A1 (en) * 2020-01-03 2021-07-08 Salubris (Chengdu) Biotech Co., Ltd. Antibodies binding bcma and uses thereof
WO2021154887A1 (en) 2020-01-28 2021-08-05 Juno Therapeutics, Inc. Methods for t cell transduction
WO2021163389A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Bcma-directed chimeric antigen receptor t cell compositions and methods and uses thereof
WO2021207689A2 (en) 2020-04-10 2021-10-14 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
WO2021222330A2 (en) 2020-04-28 2021-11-04 Juno Therapeutics, Inc. Combination of bcma-directed t cell therapy and an immunomodulatory compound
US11390681B2 (en) 2020-04-29 2022-07-19 TeneoTwo, Inc. Multispecific heavy chain antibodies with modified heavy chain constant regions
US11952426B2 (en) 2020-05-11 2024-04-09 Janssen Biotech, Inc. Methods for treating multiple myeloma
WO2021228783A1 (en) 2020-05-11 2021-11-18 Janssen Pharmaceutica Nv Methods for treating multiple myeloma
WO2021231657A1 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Methods of identifying features associated with clinical response and uses thereof
WO2022035793A1 (en) 2020-08-10 2022-02-17 Precision Biosciences, Inc. Antibodies and fragments specific for b-cell maturation antigen and uses thereof
WO2022060806A1 (en) 2020-09-16 2022-03-24 Obsidian Therapeutics, Inc. Compositions and methods for expression of anti-bcma chimeric antigen receptors with small molecule-regulated il15 in t cells
WO2022094245A1 (en) 2020-10-30 2022-05-05 Vor Biopharma, Inc. Compositions and methods for bcma modification
WO2022098787A1 (en) 2020-11-04 2022-05-12 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified invariant cd3 immunoglobulin superfamily chain locus and related polynucleotides and methods
WO2022146891A2 (en) 2020-12-31 2022-07-07 Sana Biotechnology, Inc. Methods and compositions for modulating car-t activity
US11802157B2 (en) 2020-12-31 2023-10-31 Sana Biotechnology, Inc. Methods and compositions for modulating CAR-T activity
WO2022150731A1 (en) 2021-01-11 2022-07-14 Sana Biotechnology, Inc. Use of cd8-targeted viral vectors
WO2022175255A2 (en) 2021-02-16 2022-08-25 Janssen Pharmaceutica Nv Trispecific antibody targeting bcma, gprc5d, and cd3
WO2022187406A1 (en) 2021-03-03 2022-09-09 Juno Therapeutics, Inc. Combination of a t cell therapy and a dgk inhibitor
WO2022204071A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Method to assess potency of viral vector particles
WO2022204070A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Methods of determining potency of a therapeutic cell composition
WO2022221726A2 (en) 2021-04-16 2022-10-20 Juno Therapeutics, Inc. Combination therapies with bcma-directed t cell therapy
WO2022221737A1 (en) 2021-04-16 2022-10-20 Juno Therapeutics, Inc. T cell therapy in patients who have had prior stem cell transplant
WO2022234009A2 (en) 2021-05-06 2022-11-10 Juno Therapeutics Gmbh Methods for stimulating and transducing t cells
WO2022241430A1 (en) 2021-05-11 2022-11-17 Janssen Biotech, Inc. Methods and compositions for monitoring the treatment of relapsed and/or refractory multiple myeloma
WO2022246293A1 (en) 2021-05-19 2022-11-24 Sana Biotechnology, Inc. Hypoimmunogenic rhd negative primary t cells
WO2022251367A1 (en) 2021-05-27 2022-12-01 Sana Biotechnology, Inc. Hypoimmunogenic cells comprising engineered hla-e or hla-g
WO2023278811A1 (en) 2021-07-01 2023-01-05 Indapta Therapeutics, Inc. Engineered natural killer (nk) cells and related methods
WO2023287827A2 (en) 2021-07-14 2023-01-19 Sana Biotechnology, Inc. Altered expression of y chromosome-linked antigens in hypoimmunogenic cells
WO2023015217A1 (en) 2021-08-04 2023-02-09 Sana Biotechnology, Inc. Use of cd4-targeted viral vectors
WO2023019203A1 (en) 2021-08-11 2023-02-16 Sana Biotechnology, Inc. Inducible systems for altering gene expression in hypoimmunogenic cells
WO2023069790A1 (en) 2021-10-22 2023-04-27 Sana Biotechnology, Inc. Methods of engineering allogeneic t cells with a transgene in a tcr locus and associated compositions and methods
WO2023081735A1 (en) 2021-11-03 2023-05-11 Celgene Corporation Chimeric antigen receptors specific for b-cell maturation antigen for use in treating myeloma
WO2023115041A1 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae attachment glycoproteins
WO2023115039A2 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae fusion glycoproteins
US11965022B2 (en) 2021-12-23 2024-04-23 Sana Biotechnology, Inc. Methods and compositions for modulating CAR-T activity
WO2023122337A1 (en) 2021-12-23 2023-06-29 Sana Biotechnology, Inc. Chimeric antigen receptor (car) t cells for treating autoimmune disease and associated methods
WO2023129929A1 (en) 2021-12-30 2023-07-06 Tr1X, Inc. Cd4+ t cells expressing il-10 and chimeric antigen receptors and uses thereof
WO2023133595A2 (en) 2022-01-10 2023-07-13 Sana Biotechnology, Inc. Methods of ex vivo dosing and administration of lipid particles or viral vectors and related systems and uses
WO2023147515A1 (en) 2022-01-28 2023-08-03 Juno Therapeutics, Inc. Methods of manufacturing cellular compositions
WO2023150518A1 (en) 2022-02-01 2023-08-10 Sana Biotechnology, Inc. Cd3-targeted lentiviral vectors and uses thereof
WO2023150647A1 (en) 2022-02-02 2023-08-10 Sana Biotechnology, Inc. Methods of repeat dosing and administration of lipid particles or viral vectors and related systems and uses
WO2023158836A1 (en) 2022-02-17 2023-08-24 Sana Biotechnology, Inc. Engineered cd47 proteins and uses thereof
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
WO2023215725A1 (en) 2022-05-02 2023-11-09 Fred Hutchinson Cancer Center Compositions and methods for cellular immunotherapy
WO2023213969A1 (en) 2022-05-05 2023-11-09 Juno Therapeutics Gmbh Viral-binding protein and related reagents, articles, and methods of use
WO2023220641A2 (en) 2022-05-11 2023-11-16 Juno Therapeutics, Inc. Methods and uses related to t cell therapy and production of same
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
WO2023230548A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Method for predicting response to a t cell therapy
WO2023230581A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Methods of manufacturing t cell therapies
WO2024007020A1 (en) 2022-06-30 2024-01-04 Indapta Therapeutics, Inc. Combination of engineered natural killer (nk) cells and antibody therapy and related methods
WO2024026377A1 (en) 2022-07-27 2024-02-01 Sana Biotechnology, Inc. Methods of transduction using a viral vector and inhibitors of antiviral restriction factors
WO2024023289A1 (en) * 2022-07-29 2024-02-01 Association Francaise Contre Les Myopathies B cell-specific mab-sirna conjugates improve myasthenia
EP4311579A1 (en) * 2022-07-29 2024-01-31 Association Française contre les Myopathies B cell-specific mab-sirna conjugates improve myasthenia
WO2024031091A2 (en) 2022-08-05 2024-02-08 Juno Therapeutics, Inc. Chimeric antigen receptors specific for gprc5d and bcma
WO2024054944A1 (en) 2022-09-08 2024-03-14 Juno Therapeutics, Inc. Combination of a t cell therapy and continuous or intermittent dgk inhibitor dosing
WO2024064838A1 (en) 2022-09-21 2024-03-28 Sana Biotechnology, Inc. Lipid particles comprising variant paramyxovirus attachment glycoproteins and uses thereof
US11965012B2 (en) 2022-12-21 2024-04-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins

Also Published As

Publication number Publication date
US20150125460A1 (en) 2015-05-07
IL214996A (en) 2016-10-31
NZ612647A (en) 2015-03-27
US20220213208A1 (en) 2022-07-07
AU2014204447A1 (en) 2014-07-31
DK2406284T3 (en) 2016-09-26
HK1166333A1 (en) 2012-10-26
US20120082661A1 (en) 2012-04-05
EP2406284B9 (en) 2017-03-01
CN102421801B (en) 2016-03-16
PT2406284T (en) 2016-09-29
CN104877026A (en) 2015-09-02
MX341884B (en) 2016-09-07
CA2754938A1 (en) 2010-09-16
EP3141562A1 (en) 2017-03-15
US9034324B2 (en) 2015-05-19
KR101589785B1 (en) 2016-01-28
KR20110126740A (en) 2011-11-23
US11111307B2 (en) 2021-09-07
IL214996A0 (en) 2011-11-30
CN104877026B (en) 2019-10-25
HUE029619T2 (en) 2017-03-28
HUE029619T4 (en) 2017-07-28
AU2010224160A1 (en) 2011-09-22
CY1118069T1 (en) 2017-06-28
AU2014204447B2 (en) 2015-08-20
HRP20161194T1 (en) 2016-11-04
JP6061469B2 (en) 2017-01-25
EP2406284A2 (en) 2012-01-18
DK2406284T5 (en) 2017-06-12
US20170029518A1 (en) 2017-02-02
ES2593583T9 (en) 2017-06-05
WO2010104949A3 (en) 2010-11-25
MX2011009430A (en) 2011-11-18
PL2406284T3 (en) 2017-09-29
LT2406284T (en) 2016-10-10
US20190161552A1 (en) 2019-05-30
NZ594985A (en) 2013-07-26
SI2406284T1 (en) 2017-01-31
JP2012520308A (en) 2012-09-06
EP2406284B1 (en) 2016-07-27
CN102421801A (en) 2012-04-18
JP2015187164A (en) 2015-10-29
ES2593583T3 (en) 2016-12-09
CA2754938C (en) 2016-10-11

Similar Documents

Publication Publication Date Title
US20220213208A1 (en) Anti-bcma antibodies
US20220064319A1 (en) Human monoclonal antibody human cd134 (ox40) and methods of making and using same
EP2254911B1 (en) Humanized anti-c5ar antibodies
KR20200128018A (en) Chimeric antigen receptor targeting CD70
AU2018360800A1 (en) Chimeric antigen receptors specific for B-cell maturation antigen (BCMA)
KR20190004718A (en) Anti-human VISTA antibodies and uses thereof
JP2021040637A (en) Antibody cytokine engrafted compositions and methods of use for immunoregulation
KR20200106498A (en) Anti-MCT1 antibody and use thereof
KR20200044899A (en) TIM-3 antagonist for the treatment and diagnosis of cancer
WO2021207681A1 (en) Targeted reduction of activated immune cells
US11795230B2 (en) Anti-CD27 antibodies and use thereof
WO2023030539A1 (en) Anti-gpc3 chimeric antigen receptor and methods of use thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201080020559.0

Country of ref document: CN

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

Ref document number: 10708865

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 594985

Country of ref document: NZ

Ref document number: 2010224160

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2011554150

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2754938

Country of ref document: CA

Ref document number: MX/A/2011/009430

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2010224160

Country of ref document: AU

Date of ref document: 20100310

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20117023294

Country of ref document: KR

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2010708865

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2010708865

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13255610

Country of ref document: US