US20140363442A1 - Therapeutic cd47 antibodies - Google Patents

Therapeutic cd47 antibodies Download PDF

Info

Publication number
US20140363442A1
US20140363442A1 US14/302,348 US201414302348A US2014363442A1 US 20140363442 A1 US20140363442 A1 US 20140363442A1 US 201414302348 A US201414302348 A US 201414302348A US 2014363442 A1 US2014363442 A1 US 2014363442A1
Authority
US
United States
Prior art keywords
seq
binding fragment
antigen
cancer
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/302,348
Other languages
English (en)
Inventor
William A. Frazier
Pamela T. Manning
Gerhard Frey
Hwai Wen Chang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Arch Oncology Inc
Original Assignee
Individual
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 US14/302,348 priority Critical patent/US20140363442A1/en
Application filed by Individual filed Critical Individual
Assigned to VACULOX INC. reassignment VACULOX INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FRAZIER, WILLIAM A, MANNING, PAMELA T
Publication of US20140363442A1 publication Critical patent/US20140363442A1/en
Assigned to VASCULOX, INC. reassignment VASCULOX, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHANG, HWAI WEN, FREY, GERHARD
Priority to PCT/US2015/035345 priority patent/WO2015191861A1/fr
Priority to US14/737,053 priority patent/US20150274826A1/en
Assigned to TIOMA THERAPEUTICS, INC. reassignment TIOMA THERAPEUTICS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: VASCULOX, INC.
Assigned to TIOMA THERAPEUTICS, INC. reassignment TIOMA THERAPEUTICS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: VASCULOX, INC.
Priority to US15/723,523 priority patent/US10669336B2/en
Priority to US15/723,534 priority patent/US10676524B2/en
Assigned to VASCULOX, INC. reassignment VASCULOX, INC. CORRECTIVE ASSIGNMENT TO CORRECT THE THE ASSIGNEE IN THE ASSIGNMENT DOCUMENT PREVIOUSLY RECORDED AT REEL: 035323 FRAME: 0041. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: CHANG, HWAI WEN, FREY, GERHARD
Assigned to VASCULOX, INC. reassignment VASCULOX, INC. CORRECTIVE ASSIGNMENT TO CORRECT THE THE NAME OF THE ASSIGNEE IN THE ASSIGNMENT DOCUMENT. PREVIOUSLY RECORDED AT REEL: 033294 FRAME: 0616. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: FRAZIER, WILLIAM A., MANNING, PAMELA T.
Assigned to Arch Oncology, Inc. reassignment Arch Oncology, Inc. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: TIOMA THERAPEUITICS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal 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
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • 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
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/71Decreased effector function due to an Fc-modification
    • 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
    • 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
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present disclosure relates to antibodies that bind CD47, including that of humans and other mammalian species, and their use in treating conditions and disorders, such as ischemia-reperfusion injury (IRI) and cancers, mediated by this receptor.
  • IRI ischemia-reperfusion injury
  • CD47 is a cell surface receptor comprised of an extracellular IgV set domain, a 5 membrane spanning transmembrane domain, and a cytoplasmic tail that is alternatively spliced.
  • Two ligands bind CD47: thrombospondin-1 (TSP1), and signal inhibitory receptor protein alpha (SIRPalpha).
  • TSP1 binding to CD47 activates the heterotrimeric G protein Gi, which leads to suppression of intracellular cyclic AMP (cAMP) levels.
  • cAMP cyclic AMP
  • the TSP1-CD47 pathway opposes the beneficial effects of the nitric oxide pathway in all vascular cells.
  • the nitric oxide (NO) pathway consists of any of three nitric oxide synthase enzymes (NOS I, NOS II and NOS III) that generate bioactive gas NO using arginine as a substrate. NO can act within the cell in which it is produced, or in neighboring cells, to activate the enzyme soluble guanylyl cyclase that produces the messenger molecule cyclic GMP (cGMP).
  • cGMP messenger molecule cyclic GMP
  • the proper functioning of the NO-cGMP pathway is essential for protecting the cardiovascular system against stresses including, but not limited to, those resulting from wounding, inflammation, hypertension, metabolic syndrome, ischemia, and ischemia-reperfusion injury (IRI).
  • SIRPalpha is expressed on hematopoietic cells, including macrophages and dendritic cells. When it engages CD47 on a potential phagocytic target cell, phagocytosis is slowed or prevented.
  • the CD47-SIRPalpha interaction effectively sends a “don't eat me” signal to the phagocyte.
  • blocking the SIRPalpha-CD47 interaction with a monoclonal antibody in this therapeutic context can provide an effective anti-cancer therapy by promoting, i.e., increasing, the uptake and clearance of cancer cells by the host's immune system by increasing phagocytosis, This mechanism is effective in leukemias, lymphomas and many types of solid tumors.
  • U.S. Pat. No. 8,236,313 contemplates antibodies that could be useful in the field of ischemia and blood flow to reverse and/or prevent tissue ischemia and related and associated tissue and cell damage, including antibodies that block CD47.
  • U.S. Pat. No. 8,101,719 discloses humanized antibodies that bind to CD47 for use in treating hematological disorders.
  • Objects of the invention include humanized anti-CD47 antibodies and small antibody fragments exhibiting reduced antigenicity while retaining their CD47 binding activity and apoptosis-inducing activity.
  • Such antibodies and small fragments are contemplated for use in treating hematological disorders such as various types of leukemias, malignant lymphoma, aplastic anemia, myeodysplastic syndromes, and polycythemia vera.
  • PCT International Publication WO 2011/143624 discloses chimeric and humanized anti-CD47 monoclonal antibodies for use as reagents for the diagnosis and immunotherapy of diseases associated with CD47 in humans, particularly in cancer therapy, for example to increase phagocytosis of cancer cells expressing CD47.
  • Preferred antibodies are non-activating, i.e., block ligand binding, but do not signal.
  • Disclosed humanized B6H12 and 5F9 antibodies bound soluble human CD47; B6H12 also bound human CD47 on the surface of human CD47-transfected YB2/0 cells.
  • Humanized B6H12 and 5F9 antibodies enabled phagocytosis of CFSE-labeled HL-60 cells by mouse bone marrow- or peripheral blood-derived macrophages in vitro, respectively.
  • Humanized B6H12 utilized human VH-3-7 and VK3-11 frameworks.
  • U.S. 2013/0142786 discloses non-activating anti-CD47 antibodies that increase the phagocytosis of CD47 expressing cells.
  • PCT International Publication WO 2013/119714 discloses anti-CD47 antibodies that do not cause a significant level of hemagglutination of human red blood cells.
  • Antibody compounds disclosed herein meet these needs by exhibiting the following desirable therapeutic activities:
  • the present antibodies are useful in reducing, preventing, and/or treating CD47-mediated diseases or conditions (e.g., ischemia reperfusion injury and cancers).
  • CD47-mediated diseases or conditions e.g., ischemia reperfusion injury and cancers.
  • antibody compounds of the present disclosure should be therapeutically useful in treating many forms of IRI and cancers.
  • the present antibody compounds can possess a number of other desirable properties, including broad reactivity with CD47 of a wide variety of mammalian species, including that of human, mouse, rat, pig, cynomolgus monkey, and dog, making these antibodies useful in both human and veterinary medicine.
  • This feature is further advantageous in that it facilitates preclinical studies including, but not limited to, safety and efficacy studies, in a variety of mammalian species, and therefore the development of such antibodies as human and veterinary therapeutics.
  • the present disclosure provides:
  • FIGS. 1A , 1 B, 1 C, and 1 D show cross species binding curves to human, mouse, rat, and porcine RBCs, respectively, generated using various concentrations of purified antibodies from clones Cl 1, Cl 1.1, Cl 13, and Cl 13.1 as described in Example 3.
  • Clones Cl 1 and Cl 13 are as described in Table 3.
  • Clones Cl 1.1 and Cl 13.1 are Fc mutants of clones Cl 1 and Cl 13, respectively, modified to reduce effector function. Each has an Asn297 ⁇ Gln(N297Q) mutation in the Fc domain (Sazinsky et al. (2008) PNAS 105(51):20167-20172). All of these clones exhibit concentration-dependent binding to all of the species of RBCs tested.
  • RBCs are incubated for 60 minutes on ice with various concentrations of purified antibodies from clones Cl 1, Cl 1.1, Cl 13, and Cl 13.1. Cells are then washed with cold PBS containing EDTA, incubated for an additional hour on ice with FITC labeled donkey anti-human antibody, washed, and antibody binding is analyzed using a BD FACS Aria Cell Sorter (Becton Dickinson) or a C6 Accuri Flow Cytometer (Becton Dickinson).
  • FIG. 2 shows CD47 mAb-mediated phagocytosis of Jurkat cells.
  • Target Jurkat
  • effector human macrophages
  • target effector ratio 4:1
  • 10 ⁇ g/ml of the CD47 mAbs added and incubated for 2 hours at 37 C.
  • Percent phagocytosis is determined by flow cytometry as the percentage of CFSE + /CD14 + cells from the total CD14 + population. All of the clones, except the negative control mAb 2D3, increase phagocytosis of the Jurkat cells by the macrophages irrespective of their affinity to activate Fc receptors.
  • FIG. 3 shows reversal of TSP1 inhibition of NO-stimulated cGMP production by humanized antibodies of the present disclosure.
  • Jurkat JE6.1 cells are incubated overnight in serum-free medium and then incubated with humanized antibodies of the present disclosure or the control chimeric mAb, and with or without TSP1, followed by treatment with or without a NO donor. Cells are lysed 5 minutes later and cGMP is measured. None of the present humanized antibody clones tested, or the control chimeric mAb, has an effect on basal cGMP levels.
  • the control chimeric antibody reverses the TSP1 inhibition, as do humanized clones 1, 9, 11, and 24 disclosed herein (Cl 1; Cl 9; Cl 11; Cl 24, respectively).
  • PBS phosphate buffered saline
  • TSP or TSP1 thrombospondin-1
  • DEA diethylamine NONOate
  • chim chimeric antibody >VxP037-01LC (SEQ ID NO:7))/>VxP037-01HC (SEQ ID NO:57).
  • FIG. 4 shows reversal of TSP1 inhibition of NO-stimulated cGMP production by humanized antibodies of the present disclosure.
  • Jurkat JE6.1 cells are incubated overnight in serum-free medium and then incubated with purified humanized Clone 1 and 13 antibodies, or PBS as the control, and with or without TSP1, followed by treatment with or without a NO donor. Cells are lysed 5 minutes later and cGMP is measured.
  • the humanized antibody clones or PBS have no effect on basal cGMP levels.
  • the humanized clones 1 and 13 reverse the TSP1 inhibition, while PBS has no effect.
  • PBS phosphate buffered saline
  • TSP or TSP1 thrombospondin-1
  • DEA diethylamine NONOate.
  • FIG. 5 shows that treatment of a donor kidney with Clone 1 (Cl 1) at the time of organ harvest is effective in reducing IRI and kidney damage in vivo in a rat kidney transplantation model as assessed by measuring serum creatinine.
  • a syngeneic rat renal transplantation model of IRI with bilaterally nephrectomized recipients is used to evaluate the effect of the anti-CD47 monoclonal antibody Clone 1 on graft function following transplantation.
  • Male Lewis rats weighing 275-300 g are used as both donor and recipient animals.
  • Donor kidneys are flushed with 50 ⁇ g of purified Clone 1 or vehicle (phosphate buffered saline, pH 7.2), stored at 4° C.
  • kidney function is assessed by measuring circulating serum creatinine.
  • Treatment of donor kidneys with Clone 1 results in improved kidney function compared to controls as measured by a reduction in serum creatinine.
  • FIG. 6 shows the survival in a DCD (donation after cardiac death) rat transplant model.
  • Male Lewis rats weighing 275-300 g underwent 60 minutes of warm ischemia, prior to flushing the donor kidneys with 50 ⁇ g of purified Clone 1.1 or an IgG control mAb.
  • Kidneys are then stored at 4° C. in University of Wisconsin preservation solution (UW) for 6 hours prior to transplantation.
  • UW University of Wisconsin preservation solution
  • survival is monitored over a 7 day time period. All animals that received the IgG control mAb-treated kidney died within 4 days.
  • survival is significantly prolonged in the animals that received the Clone1.1 treated kidney, with 30% of the animals surviving for the 7 day duration of the experiment. This shows that with extended periods of warm ischemia, treatment of donor kidneys with Clone 1.1 reduces IRI and increases survival of the recipient.
  • FIG. 7 shows that purified, humanized antibody Clone 13 (Cl 13) reduces tumor burden in vivo in a syngeneic mouse Acute Promyelocytic Leukemia (APL) model.
  • Murine APL cells (B6APL1) are injected intravenously into C57BL/6 mice randomized into three groups (5-10 mice per group): Group 1: no APL; Group 2: APL with no treatment; Group 3: APL treated with anti-CD47mAb Cl 13.
  • Antibody treatment is initiated on the day of tumor inoculation (day 0), and given in single doses of 10 ⁇ g/dose (0.4 mg/kg) by intraperitoneal injection on days 0, 3, and 6.
  • Circulating APL cells (representing the tumor burden) are evaluated at day 25 following tumor inoculation by flow cytometry (CD34 + /CD117 + cells). Mice treated with Cl 13 have reduced tumor burden compared to untreated mice at 25 days after tumor inoculation, demonstrating anti-tumor activity of this humanized clone.
  • FIG. 8 shows that treatment with the CD47 mAb Clone 1.1 significantly reduced tumor growth of the HepG2 tumors.
  • Male NSG mice are obtained from The Jackson Laboratory (Bar Harbor, Me.) and housed in cages in temperature and light-controlled environments with access to water and food ad libitum.
  • HepG2-luc2 cells Perkin Elmer, Waltham, Mass. #134280
  • DMEM DMEM containing 25% (v/v)
  • 1,000,000 cells implanted subcutaneously into the dorsal subcutaneous space of 4- to 8-wk-old NSG mice.
  • Antibody compounds of the present disclosure can bind to epitopes in the extracellular IgV domain of CD47, inhibiting TSP1 and SIRPalpha binding to CD47 and receptor activation, while inducing little or no agonist activity, and promoting tumor cell phagocytic clearance. In view of these properties, antibody compounds of the present disclosure should be therapeutically useful in treating many forms of IRI and cancers.
  • the present antibody compounds can also possess a number of other desirable properties, including broad reactivity with CD47 of a wide variety of mammalian species, including that of human, mouse, rat, pig, cynomolgus monkey, and/or dog, i.e., any individual one of these mammalian species, or various combinations thereof, making these antibodies useful in both human and veterinary medicine.
  • This broad reactivity is further advantageous in that it facilitates preclinical studies including, but not limited to, safety and efficacy studies, in a variety of mammalian species, and therefore the development of such antibodies as human and veterinary therapeutics.
  • antibody compounds of the present disclosure exhibit the following desirable therapeutic activities:
  • a full-length antibody as it exists naturally is an immunoglobulin molecule comprising two heavy (H) chains and two light (L) chains interconnected by disulfide bonds.
  • the amino terminal portion of each chain includes a variable region of about 100-110 or more amino acids primarily responsible for antigen recognition via the complementarity determining regions (CDRs) contained therein.
  • the carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
  • Each light chain variable region (LCVR) and heavy chain variable region (HCVR) is composed of 3 CDRs and 4 FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the 3 CDRs of the light chain are referred to as “LCDR1, LCDR2, and LCDR3” and the 3 CDRs of the heavy chain are referred to as “HCDR1, HCDR2, and HCDR3.”
  • the CDRs contain most of the residues which form specific interactions with the antigen.
  • CDR amino acid residues within the LCVR and HCVR regions are in accordance with the well-known Kabat numbering convention. While the light chain CDRs and heavy chain CDRs disclosed herein are numbered 1, 2, and 3, respectively, it is not necessary that they be employed in the corresponding antibody compound light and heavy chain variable regions in that numerical order, i.e., they can be present in any numerical order in a light or heavy chain variable region, respectively.
  • Light chains are classified as kappa or lambda, and are characterized by a particular constant region as known in the art.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the isotype of an antibody as IgG, IgM, IgA, IgD, or IgE, respectively.
  • IgG antibodies can be further divided into subclasses, e.g., IgG1, IgG2, IgG3, IgG4. Each heavy chain type is characterized by a particular constant region with a sequence well known in the art.
  • the monoclonal antibodies and other antibody compounds useful in the methods and compositions described herein can be any of these isotypes.
  • any of these isotypes can comprise amino acid modifications as follows.
  • the antibody constant region is of human IgG1 isotype.
  • the human IgG1 constant region is modified at amino acid Asn297 (Kabat Numbering) to prevent to glycosylation of the antibody.
  • this modification can be Asn297 ⁇ Ala (N297A) or Asn297 ⁇ Gln(N297Q) (Sazinsky et al. (2008) PNAS 105(51):20167-20172).
  • the constant region of the antibody is modified at amino acid Leu234 (Kabat Numbering) to alter Fc receptor interactions.
  • this modification can be Leu234 ⁇ Ala (L234A).
  • the constant region of the antibody is modified at amino acid Leu235 (Kabat Numbering) to alter Fc receptor interactions.
  • this modification can be Leu235 ⁇ Glu (L235E) or Leu235 ⁇ Ala (L235A).
  • the constant region of the antibody is altered at both amino acid 234 and 235.
  • these modifications can be Leu234 ⁇ Ala and Leu235 ⁇ Ala (L234A/L235A) (EU index of Kabat et al. (1991) Sequences of Proteins of Immunological Interest ).
  • the constant region of the antibody is of human IgG2 isotype.
  • the human IgG2 constant region is modified at amino acid Asn297 (Kabat Numbering) to prevent to glycosylation of the antibody.
  • this modification can be Asn297 ⁇ Ala (N297A) or Asn297 ⁇ Gln(N297Q).
  • the constant region of the antibody is of human IgG3 isotype.
  • the human IgG3 constant region is modified at amino acid Asn297 (Kabat Numbering) to prevent to glycosylation of the antibody.
  • this modification can be Asn297 ⁇ Ala (N297A) or Asn297 ⁇ Gln(N297Q).
  • the human IgG3 constant region is modified at amino acid 435 to extend the half-life.
  • this modification can be Arg435 ⁇ His (R435H) (EU index of Kabat et al. (1991) Sequences of Proteins of Immunological Interest ).
  • the constant region of the antibody is of human IgG4 isotype.
  • the human IgG4 constant region is modified within the hinge region to prevent or reduce strand exchange.
  • this modification can be Ser228 ⁇ Pro (S228P) (Angal et al. (1993) Molecular Immunology 30(1):105-108).
  • the human IgG4 constant region is modified at amino acid 235 to alter Fc receptor interactions.
  • this can be Leu235 ⁇ Glu (L235E).
  • the human IgG4 constant region is modified within the hinge and at amino acid 235.
  • this can be Ser228 ⁇ Pro and Leu235 ⁇ Glu (S228P/L235E).
  • the human IgG4 constant region is modified at amino acid Asn297 (Kabat Numbering) to prevent to glycosylation of the antibody.
  • Asn297 Kabat Numbering
  • this can be Asn297 ⁇ Ala (N297A).
  • EU index of Kabat et al. (1991) Sequences of Proteins of Immunological Interest ).
  • the human IgG constant region is modified to enhance FcRn binding.
  • Fc mutations that enhance binding to FcRn are Met252 ⁇ Tyr, Ser254 ⁇ Thr, Thr256 ⁇ Glu (M252Y, S254T, and T256E, respectively) (Kabat numbering, Dall'Acqua et al. (2006) J. Biol. Chem. 281(33) 23514-23524), or Met428 ⁇ Leu and Asn434 ⁇ Ser (M428L, N434S) (Zalevsky et al. (2010) Nature Biotech. 28(2):157-159). (EU index of Kabat et al. (1991) Sequences of Proteins of Immunological Interest ).
  • the human IgG constant region is modified to alter antibody-dependent cellular cytotoxicity (ADCC) and/or complement-dependent cytotoxicity (CDC), e.g., the amino acid modifications described in Natsume et al. (2008) Cancer Res. 68(10):3863-72; Idusogie et al. (2001) J. Immunol. 166(4):2571-5; Moore et al. (2010) mAbs 2(2):181-189; Lazar et al. (2006) PNAS 103(11):4005-4010; Shields et al. (2001) J. Biol. Chem. 276(9):6591-6604; Stavenhagen et al. (2007) Cancer Res.
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • the human IgG constant region is modified to induce heterodimerization.
  • having an amino acid modification within the CH3 domain at Thr366, which when replaced with a more bulky amino acid, such as Trp (T366W) is able to preferentially pair with a second CH3 domain having amino acid modifications to less bulky amino acids at positions Thr366, Leu368, and Tyr407, e.g., Ser, Ala, and Val, respectively (T366S/L368A/Y407V).
  • Heterodimerization via CH3 modifications can be further stabilized by the introduction of a disulfide bond, for example by changing Ser354 to Cys (S354C) and Tyr349 to Cys (Y349C) on opposite CH3 domains (reviewed in Carter (2001) Journal of Immunological Methods 248:7-15).
  • mAb monoclonal antibody
  • mAbs of the present disclosure preferably exist in a homogeneous or substantially homogeneous population, and can be chimeric or humanized. Complete mAbs contain two heavy chains and two light chains.
  • Antigen binding fragments of such monoclonal antibodies may be desirable for certain applications due to their small size and consequent superior tissue distribution, and include, for example, Fab fragments, Fab′ fragments, F(ab′) 2 fragments, Fd fragments, single chain Fv fragments (ScFv), and one-armed antibodies comprising a light chain and a heavy chain.
  • Preferred antigen binding fragments are those that bind to the antigen recognized by the intact antibody. Fc fragments can also be obtained.
  • Monoclonal antibodies and antigen-binding fragments thereof of the present disclosure can be produced, for example, by recombinant technologies, phage display technologies, synthetic technologies, e.g., CDR-grafting, or combinations of such technologies, or other technologies known in the art, including proteolytic digestion of intact antibodies.
  • Antibody compounds refers to mAbs, antigen-binding fragments thereof such as Fabs, etc., and competing antibodies, disclosed herein that specifically bind CD47 of various species, including human, rat, mouse, pig, cynomolgus monkey, and dog CD47, and that exhibit the properties disclosed herein.
  • mAb as used herein with respect to antibodies encompassed by the present disclosure includes Fabs and competing antibodies. Additional antibody compounds exhibiting similar functional properties according to the present disclosure can be generated by conventional methods.
  • mice can be immunized with human CD47 or fragments thereof, the resulting antibodies can be recovered and purified, and determination of whether they possess binding and functional properties similar to or the same as the antibody compounds disclosed herein can be assessed by the methods disclosed in Examples 3, 4, and 5, below.
  • Antigen-binding fragments can also be prepared by conventional methods. Methods for producing and purifying antibodies and antigen-binding fragments are well known in the art and can be found, for example, in Harlow and Lane (1988) Antibodies, A Laboratory Manual , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., chapters 5-8 and 15, ISBN 0-87969-314-2.
  • humanized antibodies refers to monoclonal antibodies and antigen binding fragments thereof, including the antibody compounds disclosed herein, that have binding and functional properties according to the disclosure similar to those disclosed herein, and that have framework and constant regions that are substantially human or fully human surrounding CDRs derived from a non-human antibody.
  • Framework region or “framework sequence” refers to any one of framework regions 1 to 4.
  • Humanized antibodies and antigen binding fragments encompassed by the present disclosure include molecules wherein any one or more of framework regions 1 to 4 is substantially or fully human, i.e., wherein any of the possible combinations of individual substantially or fully human framework regions 1 to 4, is present.
  • this includes molecules in which framework region 1 and framework region 2, framework region 1 and framework region 3, framework region 1, 2, and 3, etc., are substantially or fully human.
  • Substantially human frameworks are those that have at least 80% sequence identity to a known human germline framework sequence.
  • the substantially human frameworks have at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity, to a framework sequence disclosed herein, or to a known human germline framework sequence.
  • CDRs encompassed by the present disclosure include not only those specifically disclosed herein, but also CDR sequences having sequence identities of at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to a CDR sequence disclosed herein.
  • CDRs encompassed by the present disclosure include not only those specifically disclosed herein, but also CDR sequences having 1, 2, 3, 4, 5, or 6 amino acid changes at corresponding positions compared to CDR sequences disclosed herein. Such sequence identical, or amino acid modified, CDRs preferably bind to the antigen recognized by the intact antibody.
  • sequence identity means the percentage of identical nucleotide or amino acid residues at corresponding positions in two or more sequences when the sequences are aligned to maximize sequence matching, i.e., taking into account gaps and insertions. Identity can be readily calculated by known methods, including but not limited to those described in: Computational Molecular Biology , Lesk, A. M., ed., Oxford University Press, New York, 1988 ; Biocomputing: Informatics and Genome Projects , Smith, D. W., ed., Academic Press, New York, 1993 ; Computer Analysis of Sequence Data, Part I , Griffin, A. M., and Griffin, H.
  • Optimal alignment of sequences for comparison can be conducted, for example, by the local homology algorithm of Smith & Waterman, by the homology alignment algorithms, by the search for similarity method or, by computerized implementations of these algorithms (GAP, BESTFIT, PASTA, and TFASTA in the GCG Wisconsin Package, available from Accelrys, Inc., San Diego, Calif., United States of America), or by visual inspection. See generally, (Altschul, S. F. et al., J. Mol. Biol. 215: 403-410 (1990) and Altschul et al. Nucl. Acids Res. 25: 3389-3402 (1997)).
  • BLAST algorithm One example of an algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, which is described in (Altschul, S., et al., NCBI NLM NIH Bethesda, Md. 20894; & Altschul, S., et al., J. Mol. Biol. 215: 403-410 (1990).
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information.
  • This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold.
  • HSPs high scoring sequence pairs
  • initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them.
  • the word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always; 0) and N (penalty score for mismatching residues; always; 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value, the cumulative score goes to zero or below due to the accumulation of one or more negative-scoring residue alignments, or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • the BLASTP program uses as defaults a wordlength (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix.
  • the BLAST algorithm In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences.
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a test nucleic acid sequence is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid sequence to the reference nucleic acid sequence is in one embodiment less than about 0.1, in another embodiment less than about 0.01, and in still another embodiment less than about 0.001.
  • Fully human frameworks are those that are identical to a known human germline framework sequence.
  • Human framework germline sequences can be obtained from ImMunoGeneTics (IMGT) via their website or from The Immunoglobulin FactsBook by Marie-Paule Lefranc and Gerard Lefranc, Academic Press, 2001, ISBN 012441351.
  • germline light chain frameworks can be selected from the group consisting of: A11, A17, A18, A19, A20, A27, A30, LI, L1I, L12, L2, L5, L15, L6, L8, O12, O2, and O8, and germline heavy chain framework regions can be selected from the group consisting of: VH2-5, VH2-26, VH2-70, VH3-20, VH3-72, VHI-46, VH3-9, VH3-66, VH3-74, VH4-31, VHI-18, VHI-69, VI-13-7, VH3-11, VH3-15, VH3-21, VH3-23, VH3-30, VH3-48, VH4-39, VH4-59, and VH5-5I.
  • Humanized antibodies in addition to those disclosed herein exhibiting similar functional properties according to the present disclosure can be generated using several different methods.
  • the parent antibody compound CDRs are grafted into a human framework that has a high sequence identity with the parent antibody compound framework.
  • the sequence identity of the new framework will generally be at least 80%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identical to the sequence of the corresponding framework in the parent antibody compound.
  • frameworks having fewer than 100 amino acid residues In the case of frameworks having fewer than 100 amino acid residues, one, two, three, four, five, six, seven, eight, nine, or ten amino acid residues can be changed. This grafting may result in a reduction in binding affinity compared to that of the parent antibody. If this is the case, the framework can be back-mutated to the parent framework at certain positions based on specific criteria disclosed by Queen et al. (1991) Proc. Natl. Acad. Sci. USA 88:2869. Additional references describing methods useful in humanizing mouse antibodies include U.S. Pat. Nos. 4,816,397; 5,225,539; and 5,693,761; computer programs ABMOD and ENCAD as described in Levitt (1983) J. Mol. Biol.
  • the framework amino acid of the human germ-line sequence that is being used is replaced by a framework amino acid from a framework of the parent antibody compound (the “donor framework”): (a) the amino acid in the human framework region of the acceptor framework is unusual for human frameworks at that position, whereas the corresponding amino acid in the donor immunoglobulin is typical for human frameworks at that position; (b) the position of the amino acid is immediately adjacent to one of the CDRs; or (c) any side chain atom of a framework amino acid is within about 5-6 angstroms (center-to-center) of any atom of a CDR amino acid in a three dimensional immunoglobulin model.
  • Another approach to generating human engineered antibodies exhibiting similar functional properties to the antibody compounds disclosed herein involves randomly mutating amino acids within the grafted CDRs without changing the framework, and screening the resultant molecules for binding affinity and other functional properties that are as good as or better than those of the parent antibody compounds.
  • Single mutations can also be introduced at each amino acid position within each CDR, followed by assessing the effects of such mutations on binding affinity and other functional properties.
  • Single mutations producing improved properties can be combined to assess their effects in combination with one another.
  • Example 1 The method described in Example 1 below can also be employed.
  • amino acid substitution within the frameworks can include one, two, three, four, five, six, seven, eight, nine, or ten positions within any one or more of the 4 light chain and/or heavy chain framework regions disclosed herein.
  • amino acid substitution within the CDRs is restricted to one, two, three, four, or five positions within any one or more of the 3 light chain and/or heavy chain CDRs. Combinations of the various changes within these framework regions and CDRs described above are also possible.
  • the terms “specifically binds”, “bind specifically”, “specific binding”, and the like as applied to the present antibody compounds refer to the ability of a specific binding agent (such as an antibody) to bind to a target molecular species in preference to binding to other molecular species with which the specific binding agent and target molecular species are admixed.
  • a specific binding agent is said specifically to recognize a target molecular species when it can bind specifically to that target.
  • Binding affinity is a term that refers to the strength of binding of one molecule to another at a site on the molecule. If a particular molecule will bind to or specifically associate with another particular molecule, these two molecules are said to exhibit binding affinity for each other. Binding affinity is related to the association constant and dissociation constant for a pair of molecules, but it is not critical to the methods herein that these constants be measured or determined.
  • affinities as used herein to describe interactions between molecules of the described methods are generally apparent affinities (unless otherwise specified) observed in empirical studies, which can be used to compare the relative strength with which one molecule (e.g., an antibody or other specific binding partner) will bind two other molecules (e.g., two versions or variants of a peptide).
  • one molecule e.g., an antibody or other specific binding partner
  • two other molecules e.g., two versions or variants of a peptide.
  • epitope refers to a specific arrangement of amino acids located on a peptide or protein to which an antibody or antibody fragment binds.
  • Epitopes often consist of a chemically active surface grouping of molecules such as amino acids or sugar side chains, and have specific three dimensional structural characteristics as well as specific charge characteristics.
  • Epitopes can be linear, i.e., involving binding to a single sequence of amino acids, or conformational, i.e., involving binding to two or more sequences of amino acids in various regions of the antigen that may not necessarily be contiguous in the linear sequence.
  • Monoclonal antibodies or antigen-binding fragments thereof encompassed by the present disclosure that “compete” with the molecules disclosed herein are those that bind human CD47 at site(s) that are identical to, or overlapping with, the site(s) at which the present molecules bind. Competing monoclonal antibodies or antigen-binding fragments thereof can be identified, for example, via an antibody competition assay. For example, a sample of purified or partially purified human CD47 extracellular domain can be bound to a solid support. Then, an antibody compound, or antigen binding fragment thereof, of the present disclosure and a monoclonal antibody or antigen-binding fragment thereof suspected of being able to compete with such disclosure antibody compound are added. One of the two molecules is labeled.
  • the labeled compound and the unlabeled compound bind to separate and discrete sites on CD47, the labeled compound will bind to the same level whether or not the suspected competing compound is present. However, if the sites of interaction are identical or overlapping, the unlabeled compound will compete, and the amount of labeled compound bound to the antigen will be lowered. If the unlabeled compound is present in excess, very little, if any, labeled compound will bind.
  • competing monoclonal antibodies or antigen-binding fragments thereof are those that decrease the binding of the present antibody compounds to CD47 by about 50%, about 60%, about 70%, about 80%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, or about 99%.
  • Details of procedures for carrying out such competition assays are well known in the art and can be found, for example, in Harlow and Lane (1988) Antibodies, A Laboratory Manual , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., pages 567-569, ISBN 0-87969-314-2.
  • Such assays can be made quantitative by using purified antibodies.
  • a standard curve is established by titrating one antibody against itself, i.e., the same antibody is used for both the label and the competitor.
  • the capacity of an unlabeled competing monoclonal antibody or antigen-binding fragment thereof to inhibit the binding of the labeled molecule to the plate is titrated. The results are plotted, and the concentrations necessary to achieve the desired degree of binding inhibition are compared.
  • monoclonal antibodies or antigen-binding fragments thereof that compete with antibody compounds of the present disclosure in such competition assays possess the same or similar functional properties of the present antibody compounds can be determined via these methods in conjunction with the methods described in Examples 3-5, below.
  • competing antibodies possess about 50%, about 60%, about 70%, about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or identical biological activity compared to that of the antibody compounds disclosed herein as determined by the methods disclosed in the Examples presented below.
  • treating means slowing, interrupting, arresting, controlling, stopping, reducing, or reversing the progression or severity of a sign, symptom, disorder, condition, or disease, but does not necessarily involve a total elimination of all disease-related signs, symptoms, conditions, or disorders.
  • treating and the like refer to a therapeutic intervention that ameliorates a sign, symptom, etc., of a disease or pathological condition after it has begun to develop.
  • Acute events and chronic conditions can be treated.
  • an antibody or antigen binding fragment thereof is administered at the onset of a symptom, disorder, condition, disease, or procedure, and is discontinued when the acute event ends, or in the case of organ transplantation to the organ, at the time of organ harvest and/or to the transplant recipient at the time of organ transplantation.
  • a chronic symptom, disorder, condition, or disease is treated over a more protracted time frame.
  • an antibody compound of the present disclosure which, upon single or multiple dose administration to a patient or organ, provides the desired treatment or prevention.
  • an effective dose will generally be from about 0.01 mg/kg to about 50 mg/kg, or about 0.05 mg/kg to about 10 mg/kg of the compositions of the present disclosure in the individual to which it is administered.
  • Therapeutically effective amounts of the present antibody compounds can also comprise an amount in the range of from about 0.1 mg/kg to about 150 mg/kg, from about 0.1 mg/kg to about 100 mg/kg, or from about 0.1 mg/kg to about 50 mg/kg per single dose administered to a harvested organ or to a patient.
  • HerceptinTM is administered by intravenous infusion of a 21 mg/ml solution, with an initial loading dose of 4 mg/kg body weight and a weekly maintenance dose of 2 mg/kg body weight; RituxanTM is administered weekly at 375 mg/m 2 ; etc.
  • a therapeutically effective amount for any individual patient can be determined by the health care provider by monitoring the effect of the antibody compounds on a biomarker, such as serum biomarkers of injury of the treated organ, including but not limited to liver, kidney, lung, intestine, pancreas and heart, changes in pulmonary artery pressures, cell surface CD47 expression in tumor or non-tumor tissues, tumor regression, circulating tumor cells or tumor stem cells, etc.
  • a biomarker such as serum biomarkers of injury of the treated organ, including but not limited to liver, kidney, lung, intestine, pancreas and heart, changes in pulmonary artery pressures, cell surface CD47 expression in tumor or non-tumor tissues, tumor regression, circulating tumor cells or tumor stem cells, etc.
  • the dosing/treatment regimen can be modified over the course of therapy so that the lowest amounts of antibody compounds used alone or in combination that exhibit satisfactory efficacy are administered, and so that administration of such compounds is continued only so long as is necessary to successfully treat the patient.
  • Known antibody-based pharmaceuticals provide guidance relating to frequency of administration e.g., whether a pharmaceutical should be delivered daily, weekly, monthly, etc. Frequency and dosage may also depend on the severity of symptoms.
  • the antibody compounds of the present disclosure can be used as medicaments in human and veterinary medicine, administered by a variety of routes.
  • Veterinary applications include the treatment of companion/pet animals, such as cats and dogs; working animals, such as guide or service dogs, and horses; sport animals, such as horses and dogs; zoo animals, such as primates, cats such as lions and tigers, bears, etc.; and other valuable animals kept in captivity.
  • Antibody compounds can be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intraperitoneal, intrathecal, intraventricular, transdermal, transcutaneous, topical, subcutaneous, intranasal, enteral, sublingual, intravaginal or rectal routes. Hyposprays may also be used to administer the pharmaceutical compositions.
  • the therapeutic compositions can be prepared as injectables, either as liquid solutions or suspensions. Solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared.
  • Direct delivery of the compositions can generally be accomplished by injection, subcutaneously, intraperitoneally, intravenously, or intramuscularly, or delivered to the interstitial space of a tissue.
  • the compositions can also be administered into a lesion such as a tumor. Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • compositions are formulated for parenteral administration by, for example, intravenous, intramuscular, subcutaneous, etc., administration by infusion, injection, implantation, etc., as is well known in the art. Examples include bolus injection or continuous infusion. Intratumoral administration, for example by injection, is also contemplated.
  • compositions can be prepared by methods well known in the art. See, e.g., Remington: The Science and Practice of Pharmacy, 21 st Edition (2005), Lippincott Williams & Wilkins, Philadelphia, Pa., and comprise one or more antibody compounds disclosed herein, and a pharmaceutically or veterinarily acceptable, e.g., physiologically acceptable, carrier, diluent, or excipient.
  • a pharmaceutically or veterinarily acceptable e.g., physiologically acceptable, carrier, diluent, or excipient.
  • the therapeutic methods encompassed herein include the use of the antibodies disclosed herein alone, and/or in combinations with one another, and/or with antigen-binding fragments thereof, and/or with competing antibodies exhibiting appropriate biological/therapeutic activity, as well, i.e., all possible combinations of these antibody compounds.
  • the present therapeutic methods also encompass the use of these antibodies, antigen-binding fragments thereof, competing antibodies, etc., and combinations thereof further in combination with: (1) any one or more of the nitric oxide donor, precursor, or nitric oxide generating topical agents, and/or agents that activate soluble guanylyl cyclase, and/or agents that inhibit cyclic nucleotide phosphodiesterases disclosed herein, or (2) any one or more anti-tumor therapeutic treatments selected from surgery, radiation, anti-tumor or anti-neoplastic agents, and combinations of any of these, or (3) equivalents of any of the foregoing of (1) or (2) as would be apparent to one of ordinary skill in the art, in appropriate combination(s) to achieve the desired therapeutic treatment effect for the particular indication.
  • the monoclonal antibodies or antigen binding fragments thereof, and monoclonal antibodies or antigen binding fragments thereof that compete with these monoclonal antibodies or antigen binding fragments thereof of the present disclosure that bind to CD47 can be used alone, or in any appropriate combinations with one another, to achieve the greatest treatment efficacy.
  • the methods of the present disclosure can further comprise administering to a patient in need thereof an effective amount of a nitric oxide donor, precursor, or both; a nitric oxide generating topical agent; an agent that activates soluble guanylyl cyclase; an agent that inhibits cyclic nucleotide phosphodiesterases; or any combination of any of the foregoing.
  • the nitric oxide donor or precursor can be selected from NO gas, isosorbide dinitrate, nitrite, nitroprusside, nitroglycerin, 3-Morpholinosydnonimine (SIN-1), S-nitroso-N-acetylpenicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols, Bidil®, and arginine.
  • NO gas isosorbide dinitrate, nitrite, nitroprusside, nitroglycerin, 3-Morpholinosydnonimine (SIN-1), S-nitroso-N-acetylpenicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols, Bidil®, and arginine.
  • the agent that activates soluble guanylyl cyclase can be a non-NO (nitric oxide)-based chemical activator of soluble guanylyl cyclase that increases cGMP levels in vascular cells.
  • Such agents bind soluble guanylyl cyclase in a region other than the NO binding motif, and activate the enzyme regardless of local NO or reactive oxygen stress (ROS).
  • ROS reactive oxygen stress
  • Non-limiting examples of chemical activators of soluble guanylyl cyclase include organic nitrates (Artz et al. (2002) J. Biol. Chem. 277:18253-18256); protoporphyrin IX (Ignarro et al. (1982) Proc. Natl. Acad. Sci.
  • the agent that inhibits cyclic nucleotide phosphodiesterases can be selected from sildenafil, tadalafil, vardenafil, udenafil, and avanafil.
  • the methods of the present disclosure can further comprise treating the patient via surgery, radiation, and/or administering to a patient in need thereof an effective amount of a chemical small molecule or biologic drug including, but not limited to, a peptide, polypeptide, protein, nucleic acid therapeutic, etc., conventionally used, or currently being developed, to treat tumorous conditions.
  • a chemical small molecule or biologic drug including, but not limited to, a peptide, polypeptide, protein, nucleic acid therapeutic, etc., conventionally used, or currently being developed, to treat tumorous conditions.
  • combination therapies are often employed in cancer treatment as single-agent therapies or procedures may not be sufficient to treat or cure the disease or condition.
  • Conventional cancer treatments often involve surgery, radiation treatment, the administration of a combination of cytotoxic drugs to achieve additive or synergistic effects, and combinations of any or all of these approaches.
  • chemotherapeutic and biologic therapy combinations employ drugs that work via different mechanisms of action, increasing cancer cell control or killing, increasing the ability of the immune system to control cancer cell growth, reducing the likelihood of drug resistance during therapy, and minimizing possible overlapping toxicities by permitting the use of reduced doses of individual drugs.
  • Classes of conventional anti-tumor/anti-neoplastic agents useful in the combination therapies encompassed by the present methods are disclosed, for example, in Goodman & Gilman's The Pharmacological Basis of Therapeutics , Twelfth Edition (2010) L. L. Brunton, B. A. Chabner, and B. C. Knollmann Eds., Section VIII, “Chemotherapy of Neoplastic Diseases”, Chapters 60-63, pp. 1665-1770, McGraw-Hill, NY, and include, for example, alkylating agents; antimetabolites; natural products; a variety of miscellaneous agents; hormones and antagonists; and monoclonal antibodies.
  • Antibody and small molecule drugs that increase the immune response to cancer by modulating co-stimulatory or inhibitory interactions that influence the T cell response to tumor antigens are also of particular interest in the context of the combination therapeutic methods encompassed herein and include, but are not limited to, other anti-CD47 antibodies.
  • YERVOY® (ipilimumab; Bristol-Meyers Squibb) is an example of an approved anti-CTLA-4 antibody.
  • Another useful class of compounds for the combination therapies contemplated herein includes modulators of SIRPalpha/CD47 binding such as antibodies to SIRPalpha, as well as soluble protein fragments of this ligand, or CD47 itself, acting as “decoy” molecules inhibiting binding of, or interfering with binding of, SIRPalpha to CD47.
  • the present disclosure encompasses therapeutic methods comprising not only the administration of any of the individual monoclonal antibodies, antigen binding fragments thereof, or competing antibodies disclosed herein with any one or more of the molecules discussed immediately above, but also combinations of the disclosed monoclonal antibodies, antigen-binding fragments thereof, and competing antibodies in combinations with any one or more of the molecules discussed immediately above, i.e., all possible permutations and combinations of the presently disclosed molecules.
  • tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • cancer cancer
  • cancer cancerous
  • cancer cancerous
  • tumor tumor necrosis factor
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by aberrant cell growth/proliferation.
  • cancers include, but are not limited to, carcinomas, lymphomas, blastomas, sarcomas, and leukemias.
  • susceptible cancer refers to a cancer, cells of which express CD47 and that are responsive to treatment with an antibody or antigen binding fragment thereof, or competing antibody or antigen binding fragment thereof, of the present disclosure.
  • exemplary susceptible cancers include, but are not limited to, leukemias, including acute lymphocytic (lymphoblastic) leukemia, acute myeloid leukemia, myelogenous leukemia, chronic lymphocytic leukemia, multiple myeloma, chronic myeloid leukemia, and plasma cell leukemia; lymphomas, including Hodgkin lymphoma and Non-Hodgkin lymphoma, including B cell lymphoma, diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone B cell lymphoma, T cell lymphoma, and Waldenstrom macroglobulinemia; ovarian cancer; breast cancer; endometrial cancer; colon cancer; rectal cancer; bladder cancer; lung cancer
  • “Phagocytosis” of cancer cells refers to the engulfment and digestion of such cells by macrophages, and the eventual digestion or degradation of these cancer cells and their release extracellularly, or intracellularly, to undergo further processing.
  • Ischemia refers to a vascular phenomenon in which a decrease in the blood supply to a bodily organ, tissue, or part is caused, for instance, by constriction or obstruction of one or more blood vessels. Ischemia sometimes results from vasoconstriction or thrombosis or embolism. Ischemia can lead to direct ischemic injury, tissue damage due to cell death caused by reduced oxygen supply. Ischemia can occur acutely, as during surgery, or from trauma to tissue incurred in accidents, injuries and war settings, or following harvest of organs intended for subsequent transplantation, for example. It can also occur sub-acutely, as found in atherosclerotic peripheral vascular disease, where progressive narrowing of blood vessels leads to inadequate blood flow to tissues and organs.
  • ischemia When a tissue is subjected to ischemia, a sequence of chemical events is initiated that may ultimately lead to cellular dysfunction and necrosis. If ischemia is ended by the restoration of blood flow, a second series of injurious events ensue, producing additional injury.
  • the resultant injury involves two components—the direct injury occurring during the ischemic interval, and the indirect or reperfusion injury that follows.
  • Ischemic stroke can be caused by several different kinds of diseases. The most common problem is narrowing of the arteries in the neck or head. This is most often caused by atherosclerosis, or gradual cholesterol deposition. If the arteries become too narrow, blood cells may collect in them and form blood clots (thrombi). These blood clots can block the artery where they are formed (thrombosis), or can dislodge and become trapped in arteries closer to the brain (embolism). Cerebral stroke can occur when atherosclerotic plaque separates away partially from the vessel wall and occludes the flow of blood through the blood vessel.
  • Reperfusion refers to restoration of blood flow to tissue that is ischemic, due to decrease in blood flow. Reperfusion is a procedure for treating infarction or other ischemia, by enabling viable ischemic tissue to recover, thus limiting further necrosis. However, reperfusion can itself further damage the ischemic tissue, causing reperfusion injury.
  • ischemic/reperfusion injury involves tissue injury that occurs after blood flow is restored. Current understanding is that much of this injury is caused by chemical products, free radicals, and active biological agents released by the ischemic tissues.
  • “Nitric oxide donor, precursor, or nitric oxide generating topical agent” refers to a compound or agent that either delivers NO, or that can be converted to NO through enzymatic or non-enzymatic processes. Examples include, but are not limited to, NO gas, isosorbide dinitrite, nitrite, nitroprusside, nitroglycerin, 3-Morpholinosydnonimine (SIN-1), S-nitroso-N-acetyl-penicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols, Bidil®, and arginine.
  • NO gas isosorbide dinitrite, nitrite, nitroprusside, nitroglycerin, 3-Morpholinosydnonimine (SIN-1), S-nitroso-N-acetyl-penicillamine (SNAP), Diethylenetriamine/NO (DETA/NO), S-nitrosothiols
  • Soluble guanylyl cyclase is the receptor for nitric oxide in vascular smooth muscle.
  • nitric oxide is endogenously generated by endothelial nitric oxide synthase from L-arginine, and activates soluble guanylyl cyclase in adjacent vascular smooth muscle cells to increase cGMP levels, inducing vascular relaxation.
  • Nitric oxide binds to the normally reduced heme moiety of soluble guanylyl cyclase, and increases the formation of cGMP from GTP, leading to a decrease in intracellular calcium, vasodilation, and anti-inflammatory effects.
  • Oxidation of the heme iron on sGC decreases responsiveness of the enzyme to nitric oxide, and promotes vasoconstriction.
  • the nitric oxide-sGC-cGMP pathway therefore plays an important role in cardiovascular diseases.
  • Nitrogen-containing compounds such as sodium azide, sodium nitrite, hydroxylamine, nitroglycerin, and sodium nitroprusside have been shown to stimulate sGC, causing an increase in cGMP, and vascular relaxation.
  • activators of sGC activate the oxidized or heme-deficient sGC enzyme that is not responsive to nitric oxide, i.e., they stimulate sGC independent of redox state.
  • sGC activators are non-nitric oxide based. Note the reviews of Nossaman et al. (2012) Critical Care Research and Practice , Volume 2012, article 290805, and Derbyshire and Marietta (2012) Ann. Rev. Biochem. 81:533-559.
  • “An agent that activates soluble guanylyl cyclase” refers, for example, to organic nitrates (Artz et al. (2002) J. Biol. Chem. 277:18253-18256); protoporphyrin IX (Ignarro et al. (1982) Proc. Natl. Acad. Sci. USA 79:2870-2873); YC-1 (Ko et al. (1994) Blood 84:4226-4233); BAY 41-2272 and BAY 41-8543 (Stasch et al. (2001 Nature 410 (6825): 212-5), CMF-1571, and A-350619 (reviewed in Evgenov et al. (2006) Nat. Rev.
  • an agent that inhibits cyclic nucleotide phosphodiesterases examples include sildenafil, tadalafil, vardenafil, udenafil, and avanafil.
  • the term “about” as used herein is a flexible word with a meaning similar to “approximately” or “nearly”. The term “about” indicates that exactitude is not claimed, but rather a contemplated variation. Thus, as used herein, the term “about” means within 1 or 2 standard deviations from the specifically recited value, or ⁇ a range of up to 20%, up to 15%, up to 10%, up to 9%, up to 8%, up to 7%, up to 6%, up to 5%, up to 4%, up to 3%, up to 2%, or up to 1% compared to the specifically recited value.
  • IRI ischemia-reperfusion injury
  • IRI contributes to poor outcomes in many surgical procedures where IRI occurs due to the necessity to stop blood flow for a period of time, in many forms/causes of trauma in which blood flow is interrupted and later restored by therapeutic intervention and in procedures required for organ transplantation, cardio/pulmonary bypass procedures, reattachment of severed body parts, reconstructive and cosmetic surgeries and other situations involving stopping and restarting blood flow.
  • Ischemia itself causes many physiological changes that, by themselves would eventually lead to cell and tissue necrosis and death.
  • Reperfusion poses its own set of damaging events including generation of reactive oxygen species, thrombosis, inflammation and cytokine mediated damage.
  • TSP1-CD47 The pathways that are limited by the TSP1-CD47 system are precisely those that would be of most benefit in combating the damage of IRI. Thus, blocking the TSP1-CD47 pathway, as with the antibody compounds disclosed herein, will provide more robust functioning of these endogenous protective pathways.
  • humanized anti-CD47 antibodies, antigen binding fragments thereof, and competing antibodies and antigen binding fragments thereof, of the present disclosure can be used in the methods disclosed in U.S. Pat. No. 8,236,313, the contents of which are herein incorporated by reference in their entirety.
  • CD47 has been identified as a novel therapeutic target in hematologic cancers (Majeti et al. (2009) Cell 138(2):286-99), as well as in solid tumors such as colon, prostate, breast, and brain cancers (Willingham et al. (2012) Proc Natl Acad Sci USA 109(17):6662-7). Many human cancers up-regulate cell surface expression of CD47, and those expressing the highest levels of CD47 are the most aggressive and the most lethal for patients.
  • Increased CD47 expression is thought to protect cancer cells from phagocytic clearance by sending a “don't eat me” signal to macrophages via SIRPalpha, an inhibitory receptor that prevents phagocytosis of CD47-bearing cells (Jaiswal et al. (2009) Cell 138(2):271-851; Chao et al. (2010) Science Translational Medicine 2(63):63ra94).
  • SIRPalpha an inhibitory receptor that prevents phagocytosis of CD47-bearing cells
  • blocking mAbs Antibodies that block CD47 and prevent its binding to SIRPalpha (“blocking mAbs”) have shown efficacy in human tumor in mouse (xenograft) tumor models. Such blocking CD47mAbs exhibiting this property promote (increase) the phagocytosis of cancer cells by macrophages, which can reduce tumor burden (Majeti et al. (2009) Cell 138(2):286-99) and may ultimately lead to generation of an adaptive immune response to the tumor (Tseng et al. (2013) Proc Natl Acad Sci USA. 110(27): 11103-8).
  • a CD47 mAb or antigen binding fragment thereof disclosed herein can be used to treat a number of diseases and conditions in which IRI is a contributing feature, and to treat various autoimmune and inflammatory diseases.
  • diseases and conditions include: organ transplantation in which a mAb or antigen binding fragment thereof of the present disclosure is administered to the donor prior to organ harvest, to the harvested donor organ, to the organ preservation solution, to the recipient patient, or to any combination thereof; skin grafting; surgical resections or tissue reconstruction in which such mAb or fragment is administered either locally by injection to the affected tissue or parenterally to the patient; reattachment of body parts; treatment of traumatic injury; pulmonary hypertension; sickle cell disease (crisis); myocardial infarction; stroke; surgically-induced ischemia; acute kidney disease/kidney failure; any other condition in which IRI occurs and contributes to the pathogenesis of disease; and autoimmune/inflammatory diseases, including arthritis, multiple sclerosis, psoriasis, Crohn's disease, inflammatory
  • CD47 mAbs and antigen binding fragments thereof of the present disclosure can also be used to increase tissue perfusion in a subject in need of such treatment.
  • Such subjects can be identified by diagnostic procedures indicating a need for increased tissue perfusion.
  • the need for increased tissue perfusion may arise because the subject has had, is having, or will have, a surgery selected from integument surgery, soft tissue surgery, composite tissue surgery, skin graft surgery, resection of a solid organ, organ transplant surgery, or reattachment or an appendage or other body part.
  • mAbs and antigen binding fragments thereof effective as cancer therapeutics can be administered to patients, preferably parenterally, with susceptible hematologic cancers and solid tumors including, but not limited to, leukemias, including acute lymphocytic (lymphoblastic) leukemia, acute myeloid leukemia, myelogenous leukemia, chronic lymphocytic leukemia, multiple myeloma, chronic myeloid leukemia, and plasma cell leukemia; lymphomas, including Hodgkin lymphoma and Non-Hodgkin lymphoma, including B cell lymphoma, diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone B cell lymphoma, T cell lymphoma, and Waldenstrom macroglobulinemia; ovarian cancer; breast cancer; endometrial cancer; colon cancer; rectal cancer; bladder cancer; lung cancer; bronchial cancer; bone cancer; prostate cancer; pancreatic cancer; gastric
  • the mAb directly to the cancer by injection into the tumor.
  • CD47 expression is up-regulated on many cancers, it may also be desirable to use one or more of the disclosed mAbs as imaging and diagnostic agents when labeled with radioactive or other tracers known to those skilled in the art of in vivo imaging of cancers/tumors.
  • the humanized antibodies disclosed herein comprise frameworks derived from the human genome.
  • the collection covers the diversity found in the human germ line sequences, yielding functionally expressed antibodies in vivo.
  • the complementarity determining regions (CDRs) in the light and heavy chain variable regions of the target chimeric, non-human antibody VxP037-01LC/VxP037-01HC (SEQ ID NO:7/SEQ ID NO:57) are determined following commonly accepted rules disclosed, for example, in “Protein Sequence and Structure Analysis of Antibody Variable Domains”, In: Antibody Engineering Lab Manual , Eds. S. Duebel and R. Kontermann, Springer-Verlag, Heidelberg (2001)).
  • the CDR fragments are synthesized and combined with pools of frameworks to generate full length variable domains.
  • the humanized variable domains are then combined with a secretion signal and human kappa and human IgG1 constant domains, and cloned into a mammalian expression system (e.g., OptiCHO System, Lifetechnologies, Carlsbad, Calif.) to generate a library of humanized IgG1, IgG2, and IgG4 variants. An aliquot of the library is sequenced to ensure high diversity and integrity of the reading frames of the individual clones.
  • a mammalian expression system e.g., OptiCHO System, Lifetechnologies, Carlsbad, Calif.
  • the cell culture supernatant containing the humanized IgG variants is then screened for binding to the target antigen, CD47.
  • the concentration of each variant is determined in order to calculate specific activity for each clone.
  • the specific activity of each clone is compared to the specific activity of chimeric clone VxP037-01LC-Pro/VxP037-01HC-Pro (SEQ ID NO:107/SEQ ID NO:109) expressed on the same plate, and normalized. Top hits from each plate are re-arrayed and re-screened for confirmation.
  • the final candidates are selected by specific activity, functional activity, expression level, and sequence diversity, as well as other criteria, as described below.
  • IgG1-N297Q A non-glycosylated version (IgG1-N297Q) was created by site directed mutagenesis of heavy chain position 297 to change the asparagine to glutamine (pVxP037-01-HC-IgG1 N297Q-Pro; SEQ ID NO:111).
  • IgG2 and IgG4-S228P isotypes were constructed by cloning the heavy chain variable domain in frame with the human IgG2 and IgG4-S228P constant domains (pVxK7b-037-hum01-HC-IgG2-Pro, SEQ ID NO:112 or pVxK7b-037-hum01-HC_IgG4 S228P-Pro, SEQ ID NO:113).
  • amino acid sequences of the light chain and heavy chain variable regions, the complete light and heavy chains, and the respective encoding nucleotide sequences of the foregoing, of the present human engineered antibodies are listed below in the section entitled “Amino Acid and Nucleic Acid Sequences.”
  • SEQ ID NO:117 shows the amino acid sequence of framework 4+the light chain constant domain amino acid sequence of chimeric complete light chain amino acid sequence SEQ ID NO:107.
  • SEQ ID NOs:118, 119, 120, and 121 show the amino acid sequences of framework 4+the heavy chain constant domain amino acid sequences of complete heavy chain amino acid sequences SEQ ID NOs:109, 111, 112, and 113, respectively.
  • All the light chain variable regions SEQ ID NOs:7-31 can further comprise SEQ ID NO:117, and all the heavy chain amino acid sequences SEQ ID NOs:57-81 can further comprise any of SEQ ID NOs:118, 119, 120, and 121, thereby describing complete antibody sequences encompassed by this disclosure.
  • the light chain and heavy chain CDR amino acid sequences are shown in Tables 1 and 2, respectively.
  • Cross species reactivity of humanized antibodies of the present disclosure is determined using freshly isolated red blood cells (RBCs), which display CD47 on their surface, from human, mouse, rat, pig, cynomolgus monkey, and dog according to the methods disclosed in Kamel et al. (2010) Blood. Transfus. 8(4):260-266.
  • RBCs red blood cells
  • Supernatants containing secreted antibodies are collected from CHO cells transiently transfected with plasmids encoding antibody clones and used as collected, or antibodies are further purified from the supernatants using standard methods.
  • Transfected CHO cells are grown in F-12 medium containing 10% heat inactivated fetal bovine serum (BioWest; S01520).
  • Antibody concentration in the supernatants is determined utilizing a quantitative ELISA.
  • ELISA plates are coated with a donkey anti-human FC antibody (Sigma; Catalog #12136) at 10 ⁇ g/ml overnight at 4° C. (Promega; Catalog # W4031).
  • Plates are washed with PBS, and then blocked with casein blocking solution (ThermoScientific; Catalog #37532) for 60 minutes at room temperature. Plates are again washed with PBS, tissue culture supernatants are added, and the plates are incubated for 60 minutes at room temperature. Plates are then washed three times with PBS and incubated with peroxidase-conjugated goat anti-human IgG (Jackson Immunoresearch Labs; Catalog #109-035-003) for 60 minutes at room temperature. Plates are washed three times with PBS, and the peroxidase substrate 3,3′,5,5′-tetramethylbenzidine is added (Sigma; Catalog #T4444). Reactions are terminated by the addition of HCl to 0.7N, and absorbance at 450 nM is determined using a Tecan model Infinite M200 plate reader.
  • casein blocking solution ThermoScientific; Catalog #37532
  • RBCs are incubated for 60 minutes on ice with tissue culture supernatants containing the secreted humanized antibodies at a concentration of 10 ng/ml in a solution of phosphate buffered saline, pH 7.2, 2.5 mM EDTA (PBS+E), or with various concentrations of purified antibodies. Cells are then washed with cold PBS+E, and incubated for an additional hour on ice with FITC labeled donkey anti-human antibody (Jackson Immuno Research Labs, West Grove, Pa.; Catalogue #709-096-149) in PBS+E.
  • FITC labeled donkey anti-human antibody Jackson Immuno Research Labs, West Grove, Pa.; Catalogue #709-096-149
  • FIG. 1 shows cross species binding curves to human, mouse, rat, and porcine RBCs (panels A, B, C, and D, respectively, generated using various concentrations of purified antibodies from clones Cl 1, Cl 1.1, Cl 13, and Cl 13.1.
  • Clones Cl 1 and Cl 13 are as described above in Table 3.
  • Clones Cl 1.1 and Cl 13.1 are Fc mutants of clones Cl 1 and Cl 13, respectively, modified to reduce effector function. Each has an Asn297 ⁇ Gln(N297Q) mutation in the Fc domain (Sazinsky et al. (2008) PNAS 105(51):20167-20172). All of these clones exhibit concentration-dependent binding to all of the species of RBCs tested. These clones also bind to cynomolgus monkey RBCs (data not shown).
  • Table 4 shows the apparent affinities of these clones to human RBCs determined by non-linear fits (Prism Graphpad software) of the median fluorescence intensities at various antibody concentrations. Clones 1, 1.1, 13, and 13.1 all have apparent Kd values in the low nanomolar range.
  • Human derived macrophages are derived from leukapheresis of healthy human peripheral blood incubated in human AB serum (Sigma Aldrich) for 24 hours in culture. After 24 hours, all non-adherent cells are removed and the remaining adherent macrophages are incubated in RPMI medium (10% fetal bovine serum (FBS; Hyclone) and antibiotics) for two weeks. For the in vitro phagocytosis assay, macrophages are re-plated at a concentration of 5 ⁇ 10 4 cells per well in 1 ml of RPMI media in a 24 well plate and allowed to adhere for 24 hours.
  • RPMI medium % fetal bovine serum (FBS; Hyclone) and antibiotics
  • the target cancer cells (Jurkat) are labeled with 1 ⁇ M 5(6)-Carboxyfluorescein diacetate N-succinimidyl ester (CFSE; Sigma Aldrich) and added to the macrophage cultures at a concentration of 2 ⁇ 10 5 cells in 1 ml of RPMI media (4:1 target to effector ratio).
  • CD47mAbs (10 ⁇ g/ml) are added immediately upon mixture of target and effector cells and allowed to incubate at 37° C. for 2 hours. After 2 hours, all non-phagocytosed cells are removed and the remaining cells are washed three times with phosphate buffered saline (PBS; Sigma Aldrich).
  • Cells are trypsinized, collected into microcentrifuge tubes and incubated in 100 ng of allophycocyanin (APC) labeled CD14 antibodies (BD biosciences) for 30 minutes. Cells are washed once and analyzed by flow cytometry (Accuri C6; BD biosciences) for the percentage of CD14 positive cells that are also CFSE positive indicating complete phagocytosis.
  • APC allophycocyanin
  • both Clone 1 and Clone 13 humanized mAbs increase phagocytosis of Jurkat cells.
  • Clones 1.1 and Clone 13.1 are identical to Clones 1 and 13 except for a mutation of residue N297 to Q which reduces the affinity of the IgG1 molecule to the Fc receptor.
  • the IgG4 versions of Clone 1 and Clone 13 also increase phagocytosis of Jurkat cells.
  • the IgG4 isotype also has reduced affinity for activating Fc receptors.
  • cGMP cGMP Fluorescent Assay Kit
  • Molecular Devices Sunnyvale, Calif.
  • Jurkat JE6.1 cells ATCC, Manassas, Va.; Catalog # TIB-152 are used as these cells retain the NO-cGMP signaling pathway when grown in culture and exhibit a robust and reproducible inhibitory response to TSP1 ligation of CD47.
  • Cells are grown in Iscove's modified Dulbeccco's medium containing 5% (v/v) heat inactivated fetal bovine serum (BioWest; Catalogue # S01520), 100 units/mL penicillin, 100 ⁇ g mL streptomycin (Sigma; Catalogue # P4222) at densities less than 1 ⁇ 10 6 cells/mL.
  • cells are plated in 96 well tissue culture plates at a density of 1 ⁇ 10 5 cells/ml in Iscoves modified Dulbecco's medium containing 5% (v/v) heat inactivated fetal bovine serum (BioWest; Catalog # S01520), 100 units/mL penicillin, 100 ⁇ g/mL streptomycin (Sigma; #P4222) for 24 hours and then transferred to serum free medium overnight.
  • humanized antibodies as disclosed herein purified from transient transfections in CHO cells as described above in Example 3, as well as the control chimeric antibody, are then added at a final concentration of 20 ng/ml, followed 15 minutes later by 0 or 1 ⁇ g/ml human TSP1 (Athens Research and Technology, Athens, Ga., Catalogue #16-20-201319). After an additional 15 minutes, the NO donor, diethylamine (DEA) NONOate (Cayman Chemical, Ann Arbor, Mich., Catalog #82100), is added to half the wells at a final concentration of 1 ⁇ M. Five minutes later, the cells are lysed with buffer supplied in the cGMP kit, and aliquots of each well are assayed for cGMP content.
  • DEA diethylamine
  • Humanized clones 1, 9, 11, 13, and 24 of the present disclosure also significantly reverse TSP1 inhibition, demonstrating that they have the ability to increase NO signaling ( FIGS. 3 and 4 ), suggesting their utility in protecting the cardiovascular system against stresses including, but not limited to, those resulting from wounding, inflammation, hypertension, metabolic syndrome, ischemia, and ischemia-reperfusion injury (IRI).
  • IRI ischemia-reperfusion injury
  • the purpose of this experiment is to demonstrate that a humanized antibody clone disclosed herein, i.e., Clone 1, that is shown to regulate nitric oxide signaling in vitro in Example 5, is effective in reducing IRI and kidney damage in vivo in a rat kidney transplant model under standard conditions, i.e., with no warm ischemic time but with cold ischemic time.
  • IRI significantly contributes to delayed graft function and inflammation leading to graft loss, and is exacerbated by the thrombospondin-1/CD47 system through inhibition of nitric oxide signaling.
  • a syngeneic rat renal transplantation model of IRI with bilaterally nephrectomized recipients is used to evaluate the effect of the anti-CD47 monoclonal antibody Clone 1 on graft function following transplantation as described in Schumacher et al. (2003) Microsurg. 23:389-394 and Karatzas et al. (2007) Microsug. 27:668-672.
  • CD47mAb Clone 1 perfusion of donor kidneys results in improved kidney function compared to controls as measured by a reduction in serum creatinine.
  • APL Acute Promyelocytic Leukemia
  • the purpose of this experiment is to demonstrate that a humanized antibody clone disclosed herein, i.e., Clone 13, reduces tumor burden in vivo in a mouse leukemia model.
  • the anti-tumor activity of the anti-CD47mAb Clone13 (Cl 13; clone number as described above in Examples 2 and 3) is determined in a syngeneic murine model of Acute Promyelocytic Leukemia (APL) as described in Ramirez et al. (2009) Blood 113:6206-6214.
  • APL Acute Promyelocytic Leukemia
  • Murine APL cells (B6APL1) are injected intravenously into C57BL/6 mice that are randomized into three groups (5-10 mice per group): Group 1: no APL; Group 2: APL with no treatment; Group 3: APL with anti-CD47mAb Cl 13 treatment.
  • Antibody treatment is initiated on the day of tumor inoculation (day 0), and given in single doses of 10 ⁇ g/dose (0.4 mg/kg) in phosphate buffered saline, pH 7.2, by intraperitoneal injection on days 0, 3, and 6.
  • Tumor burden is evaluated at day 25 following tumor cell inoculation. Blood samples from each mouse are analyzed for white blood cell count using an automated hemocytometer, and circulating APL cells (representing the tumor burden) are quantified by flow cytometry (CD34 + /CD117 + cells).
  • mice treated with Cl 13 have reduced tumor burden compared to untreated mice at 25 days after tumor inoculation, thus demonstrating anti-tumor activity of this humanized clone.
  • HCC human hepatocellular carcinoma
  • mice Male NSG mice are obtained from The Jackson Laboratory (Bar Harbor, Me.) and housed in cages in temperature and light-controlled environments with access to water and food ad libitum.
  • HepG2-luc2 cells Perkin Elmer, Waltham, Mass. #134280
  • DMEM fetal methylcholine
  • 1,000,000 cells implanted subcutaneously into the dorsal subcutaneous space of 4- to 8-wk-old NSG mice.
  • antibody treatment is begun with twice-weekly intraperitoneal injections of 15 mg/kg of either anti-CD47 antibody Clone 1.1 or an IgG control for 6 weeks. Tumor volumes are calculated twice weekly using (length ⁇ width)/0.6. After 6 weeks of treatment, animals are euthanized and tumors were resected, weighed, and fixed in 10% formalin.
  • treatment with the CD47 mAb Clone 1.1 significantly reduced tumor growth of the HepG2 tumors (p ⁇ 0.01), demonstrating anti-tumor efficacy on solid tumors.
  • VxP037-01-LC-Pro represents a full length chimeric light chain variable domain (SEQ ID NO:7)+a constant domain amino acid sequence.
  • the underlined amino acid sequence framework 4+the constant domain.
  • All full length humanized light chain sequences can contain a light chain variable region sequence selected from SEQ ID NOs:7-31 in combination with framework 4+the same constant domain as VxP037-01-LC-Pro. However, while present, this constant domain is not shown for all the complete humanized light chain amino acid sequences.
  • the underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-01-LC-Pro, above.
  • VxP037-01-HC-Pro represents a full length chimeric heavy chain variable domain (SEQ ID NO:57)+a constant domain amino acid sequence.
  • the underlined amino acid sequence framework 4+the constant IgG1 domain.
  • All full length humanized heavy chain sequences can contain a heavy chain variable region sequence selected from SEQ ID NOs:57-81 in combination with framework 4+the same constant domain as VxP037-01-HC-Pro. However, while present, this constant domain is not shown for all the complete humanized heavy chain amino acid sequences.
  • the underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-01-HC-Pro, above.
  • >VxP037-01-HC-IgG1 N297Q-Pro represents a full length heavy chain variable domain (SEQ ID NO:57)+a constant domain amino acid sequence.
  • the underlined amino acid sequence framework 4+the constant IgG1 constant domain containing a N Q mutation at amino acid position 297.
  • All full length humanized heavy chain sequences can contain a heavy chain variable region sequence selected from SEQ ID NOs:57-81 in combination with framework 4+the same constant domain as >VxP037-01-HC-IgG1 N297Q-Pro. However, while present, this constant domain is not shown for all the complete humanized heavy chain amino acid sequences.
  • >pVxK7b-037-hum01-HC-IgG2-Pro represents a full length heavy chain variable domain (SEQ ID NO:58)+a constant domain amino acid sequence.
  • the underlined amino acid sequence framework 4+the constant IgG2 domain.
  • All full length humanized heavy chain sequences can contain a heavy chain variable region sequence selected from SEQ ID NOs:57-81 in combination with framework 4+the same constant domain as >pVxK7b-037-hum01-HC-IgG2-Pro. However, while present, this constant domain is not shown for all the complete humanized heavy chain amino acid sequences.
  • >pVxK7b-037-hum01-HC-IgG4 S228P-Pro represents a full length heavy chain variable domain (SEQ ID NO:58)+a constant domain amino acid sequence.
  • the underlined amino acid sequence framework 4+the constant IgG4 S228P domain.
  • All full length humanized heavy chain sequences can contain a heavy chain variable region sequence selected from SEQ ID NOs:57-81 in combination with framework 4+the same constant domain as >pVxK7b-037-hum01-HC-IgG4 S228P-Pro. However, while present, this constant domain is not shown for all the complete humanized heavy chain amino acid sequences.
  • the underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-01-HC-IgG1 N297Q-Pro, above.
  • the underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-hum 01-HC-IgG2-Pro, above.
  • the underlined nucleic acid sequence encodes the underlined protein sequence in >VxP037-hum 01-HC-IgG4 S228P Pro, above.
US14/302,348 2012-12-12 2014-06-11 Therapeutic cd47 antibodies Abandoned US20140363442A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US14/302,348 US20140363442A1 (en) 2012-12-12 2014-06-11 Therapeutic cd47 antibodies
US14/737,053 US20150274826A1 (en) 2012-12-12 2015-06-11 Therapeutic cd47 antibodies
PCT/US2015/035345 WO2015191861A1 (fr) 2012-12-12 2015-06-11 Anticorps cd47 thérapeutique
US15/723,534 US10676524B2 (en) 2012-12-12 2017-10-03 Therapeutic CD47 antibodies
US15/723,523 US10669336B2 (en) 2012-12-12 2017-10-03 Therapeutic CD47 antibodies

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261736301P 2012-12-12 2012-12-12
US201361833691P 2013-06-11 2013-06-11
PCT/US2013/074766 WO2014093678A2 (fr) 2012-12-12 2013-12-12 Anticorps anti-cd47 thérapeutiques
US14/302,348 US20140363442A1 (en) 2012-12-12 2014-06-11 Therapeutic cd47 antibodies

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/074766 Continuation-In-Part WO2014093678A2 (fr) 2012-12-12 2013-12-12 Anticorps anti-cd47 thérapeutiques

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/737,053 Continuation-In-Part US20150274826A1 (en) 2012-12-12 2015-06-11 Therapeutic cd47 antibodies
US15/723,534 Continuation US10676524B2 (en) 2012-12-12 2017-10-03 Therapeutic CD47 antibodies

Publications (1)

Publication Number Publication Date
US20140363442A1 true US20140363442A1 (en) 2014-12-11

Family

ID=50935081

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/302,348 Abandoned US20140363442A1 (en) 2012-12-12 2014-06-11 Therapeutic cd47 antibodies
US14/737,053 Abandoned US20150274826A1 (en) 2012-12-12 2015-06-11 Therapeutic cd47 antibodies
US15/723,534 Active 2034-05-10 US10676524B2 (en) 2012-12-12 2017-10-03 Therapeutic CD47 antibodies
US15/723,523 Active 2034-04-21 US10669336B2 (en) 2012-12-12 2017-10-03 Therapeutic CD47 antibodies

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/737,053 Abandoned US20150274826A1 (en) 2012-12-12 2015-06-11 Therapeutic cd47 antibodies
US15/723,534 Active 2034-05-10 US10676524B2 (en) 2012-12-12 2017-10-03 Therapeutic CD47 antibodies
US15/723,523 Active 2034-04-21 US10669336B2 (en) 2012-12-12 2017-10-03 Therapeutic CD47 antibodies

Country Status (16)

Country Link
US (4) US20140363442A1 (fr)
EP (1) EP2931751B1 (fr)
JP (1) JP6572131B2 (fr)
KR (1) KR20150093770A (fr)
CN (1) CN105102479B (fr)
AU (1) AU2013359167B2 (fr)
BR (1) BR112015013431A2 (fr)
CA (1) CA2894439A1 (fr)
ES (1) ES2786083T3 (fr)
HK (1) HK1216647A1 (fr)
IL (1) IL239339A0 (fr)
MX (1) MX2015007446A (fr)
PH (1) PH12015501318A1 (fr)
RU (1) RU2015122228A (fr)
SG (1) SG11201504469XA (fr)
WO (2) WO2014093678A2 (fr)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104804093A (zh) * 2015-05-27 2015-07-29 江苏春申堂药业有限公司 一种针对cd47的单域抗体
US9518117B2 (en) 2012-12-12 2016-12-13 Tioma Therapeutics, Inc. Therapeutic CD47 antibodies
US9650441B2 (en) 2015-09-21 2017-05-16 Erasmus University Medical Center Anti-CD47 antibodies and methods of use
WO2018176132A1 (fr) * 2017-03-28 2018-10-04 Trillium Therapeutics Inc. Thérapie par blocage de cd47
US10239945B2 (en) 2015-09-18 2019-03-26 Arch Oncology, Inc. Therapeutic CD47 antibodies
WO2019090355A1 (fr) * 2017-11-06 2019-05-09 Children's National Medical Center Cellules exprimant des anticorps et procédés de traitement les utilisant
CN110538321A (zh) * 2018-05-29 2019-12-06 江苏恒瑞医药股份有限公司 一种cd47抗体药物组合物及其用途
WO2020036977A1 (fr) * 2018-08-13 2020-02-20 Arch Oncology, Inc. Anticorps cd47 thérapeutiques
US10669336B2 (en) 2012-12-12 2020-06-02 Arch Oncology, Inc. Therapeutic CD47 antibodies
CN111635459A (zh) * 2020-06-27 2020-09-08 北京广未生物科技有限公司 抗cd47抗体及其在治疗癌症中的应用
US10894831B2 (en) 2015-08-26 2021-01-19 The Board Of Trustees Of The Leland Stanford Junior University Enhanced depletion of targeted cells with CD47 blockade and an immune costimulatory agonist
US10906954B2 (en) 2012-12-17 2021-02-02 Trillium Therapeutics Inc. Treatment of CD47+ disease cells with SIRPα-Fc fusions
WO2021116699A1 (fr) 2019-12-11 2021-06-17 Precision Immunotherapeutics Limited Molécules de liaison inhibées de manière réversible
US20210230276A1 (en) * 2016-01-21 2021-07-29 The Board Of Trustees Of The Leland Stanford Junior University Treatment of cancer with combinations of immunoregulatory agents
US11186640B2 (en) * 2014-07-31 2021-11-30 The University Of Western Australia Method for the identification of immunotherapy-drug combinations using a network approach
CN113993899A (zh) * 2019-06-19 2022-01-28 乐普生物科技股份有限公司 抗cd47抗体及其应用
US11400145B2 (en) 2015-11-27 2022-08-02 Cartherics Pty. Ltd. Genetically modified cells and uses thereof
WO2022177392A1 (fr) * 2021-02-19 2022-08-25 (주)샤페론 Anticorps à domaine unique dirigé contre cd47 et son utilisation
US11446315B2 (en) 2016-11-03 2022-09-20 Pf Argentum Ip Holdings Llc Enhancement of CD47 blockade therapy by proteasome inhibitors
US11692035B2 (en) 2016-10-21 2023-07-04 Arch Oncology, Inc. Therapeutic CD47 antibodies

Families Citing this family (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3054220C (fr) 2008-01-15 2022-10-11 The Board Of Trustees Of The Leland Stanford Junior University Procedes pour manipuler une phagocytose a mediation par cd47
WO2016057980A1 (fr) * 2014-10-10 2016-04-14 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Procédés d'élimination de cellules souches cancéreuses par ciblage de cd47
CN107406503B (zh) 2014-11-18 2021-07-16 詹森药业有限公司 Cd47抗体、方法和用途
KR102313073B1 (ko) 2014-12-05 2021-10-18 리제너론 파마슈티칼스 인코포레이티드 인간화 분화 클러스터 47 유전자를 가진 비인간 동물
US20170151281A1 (en) 2015-02-19 2017-06-01 Batu Biologics, Inc. Chimeric antigen receptor dendritic cell (car-dc) for treatment of cancer
TW202248215A (zh) * 2015-03-04 2022-12-16 美商索倫多醫療公司 結合cd47之抗體治療劑
WO2017044859A1 (fr) * 2015-09-10 2017-03-16 Affigen, Inc. Sélection orientée par le séquençage d'une théranostique des tumeurs
EP3383908A1 (fr) 2015-12-02 2018-10-10 Stsciences, Inc. Anticorps spécifiques au btla glycosylé (atténuateur de lymphocytes b et t)
KR20220163516A (ko) 2016-01-11 2022-12-09 포티 세븐, 인코포레이티드 인간화된, 마우스 또는 키메라 항-cd47 단클론 항체
AU2017250029C1 (en) * 2016-04-15 2022-03-24 Pfizer Inc. Macrophage stimulation in CD47 blockade therapy
CN106084052B (zh) 2016-06-17 2019-12-27 长春金赛药业股份有限公司 抗cd47单克隆抗体及其应用
CN106117354B (zh) * 2016-06-24 2020-01-14 安徽未名细胞治疗有限公司 一种全人源抗CD47的全分子IgG抗体及其应用
JP7241677B2 (ja) 2016-07-19 2023-03-17 テバ・ファーマシューティカルズ・オーストラリア・ピーティワイ・リミテッド 抗cd47併用療法
WO2018095428A1 (fr) * 2016-11-28 2018-05-31 江苏恒瑞医药股份有限公司 Anticorps cd47, fragment de liaison à l'antigène de l'anticorps cd47 et leur utilisation médicale
EP3345924A1 (fr) * 2017-01-10 2018-07-11 Universität Duisburg-Essen Emploi d'anticorps cd47
GB201700621D0 (en) 2017-01-13 2017-03-01 Guest Ryan Dominic Method,device and kit for the aseptic isolation,enrichment and stabilsation of cells from mammalian solid tissue
CA3051512A1 (fr) * 2017-01-26 2018-08-02 Zlip Holding Limited Unite de liaison a l'antigene cd47 et ses utilisations
BR112019020185A2 (pt) * 2017-03-27 2020-06-02 Celgene Corporation Métodos e composições para redução da imunogenicidade
WO2019014398A1 (fr) 2017-07-11 2019-01-17 Actym Therapeutics, Inc. Souches bactériennes immunostimulatrices modifiées et utilisations
US11401329B2 (en) 2017-08-02 2022-08-02 Phanes Therapeutics, Inc. Anti-CD47 antibodies and uses thereof
JP7256580B2 (ja) * 2017-08-18 2023-04-12 ウルトラヒューマン フォー リミティド 結合剤
CN109422811A (zh) 2017-08-29 2019-03-05 信达生物制药(苏州)有限公司 抗cd47抗体及其用途
EA039662B1 (ru) 2017-10-03 2022-02-24 Закрытое Акционерное Общество "Биокад" Антитела, специфичные к cd47 и pd-l1
CN117771364A (zh) 2017-10-18 2024-03-29 四十七公司 基于抗cd47剂的卵巢癌疗法
US20220298254A1 (en) 2017-11-01 2022-09-22 Hummingbird Bioscience Holdings Pte. Ltd. Cd47 antigen-binding molecules
EP3706775A4 (fr) 2017-11-06 2021-09-01 Trillium Therapeutics Inc. Blocage du cd47 associé à une radiothérapie
TWI831759B (zh) 2017-12-01 2024-02-11 美商思進公司 Cd47抗體及其用於治療癌症之用途
CN110144009B (zh) * 2018-02-14 2020-01-21 上海洛启生物医药技术有限公司 Cd47单域抗体及其用途
CN110305212A (zh) 2018-03-27 2019-10-08 信达生物制药(苏州)有限公司 抗cd47抗体及其用途
WO2019200462A1 (fr) * 2018-04-16 2019-10-24 Adaerata, Limited Partnership Méthodes de prévention ou de traitement de cancers positifs pour slamf7 et négatifs pour slamf7
CN108484770B (zh) * 2018-05-16 2020-11-13 武汉云克隆科技股份有限公司 重组大鼠抗小鼠cd4单克隆抗体,制备方法和应用
WO2019241732A1 (fr) * 2018-06-15 2019-12-19 Accurus Biosciences, Inc. Anticorps de blocage contre cd47 et leurs méthodes d'utilisation
CN110615841B (zh) * 2018-06-20 2022-01-04 瑞阳(苏州)生物科技有限公司 抗人cd47单克隆抗体及其应用
BR112021000315A2 (pt) 2018-07-11 2021-08-03 Actym Therapeutics, Inc. cepas bacterianas imunoestimulantes modificadas geneticamente e seus usos
EP3844276A2 (fr) 2018-08-28 2021-07-07 Actym Therapeutics, Inc. Souches bactériennes immunostimulatrices modifiées et utilisations associées
TWI830774B (zh) * 2018-08-31 2024-02-01 大陸商南京聖和藥業股份有限公司 抗cd47抗體及其應用
EP3876977A1 (fr) 2018-11-06 2021-09-15 The Regents Of The University Of California Récepteurs antigéniques chimériques pour la phagocytose
EP3892637A1 (fr) * 2018-12-03 2021-10-13 Shanghai Pharmaexplorer Co., Ltd. Anticorps cd47, son procédé de préparation et ses applications
US20230012428A1 (en) 2019-03-06 2023-01-12 Jiangsu Hengrui Medicine Co., Ltd. Bifunctional fusion protein and pharmaceutical use thereof
US11026973B2 (en) 2019-04-30 2021-06-08 Myeloid Therapeutics, Inc. Engineered phagocytic receptor compositions and methods of use thereof
EP3980747A1 (fr) 2019-06-07 2022-04-13 ALX Oncology Inc. Procédés et réactifs pour réduire les interférences de médicaments se liant au cd47 dans des dosages sérologiques
WO2020253785A1 (fr) * 2019-06-19 2020-12-24 Lepu Biopharma Co., Ltd. Anticorps anti-cd47 et leurs utilisations
EP3999107A1 (fr) 2019-07-16 2022-05-25 Gilead Sciences, Inc. Vaccins contre le vih et leurs procédés de fabrication et d'utilisation
WO2021043220A1 (fr) * 2019-09-03 2021-03-11 中山康方生物医药有限公司 Anticorps monoclonal anti-cd47 et son utilisation
AU2020341479A1 (en) 2019-09-03 2022-03-31 Myeloid Therapeutics, Inc. Methods and compositions for genomic integration
EP4349413A2 (fr) 2019-10-18 2024-04-10 Forty Seven, Inc. Polythérapies pour le traitement de syndromes myélodysplasiques et de leucémie myéloïde aiguë
JP2022553390A (ja) * 2019-10-25 2022-12-22 ウーシー バイオロジクス アイルランド リミテッド 新規抗cd47抗体及びその使用
CA3153636A1 (fr) 2019-10-31 2021-05-06 Forty Seven, Inc. Traitement d'un cancer du sang base sur une therapie anti-cd47 et anti-cd20
US10980836B1 (en) 2019-12-11 2021-04-20 Myeloid Therapeutics, Inc. Therapeutic cell compositions and methods of manufacturing and use thereof
JP2023507432A (ja) 2019-12-20 2023-02-22 インスティル バイオ (ユーケイ) リミテッド 腫瘍浸潤リンパ球を単離するためのデバイスおよび方法ならびにそれらの使用
IL294032A (en) 2019-12-24 2022-08-01 Carna Biosciences Inc Compounds that regulate diacylglycerol kinase
AU2021219668A1 (en) 2020-02-14 2022-08-25 Gilead Sciences, Inc. Antibodies and fusion proteins that bind to CCR8 and uses thereof
WO2021190441A1 (fr) * 2020-03-23 2021-09-30 倍而达药业(苏州)有限公司 Anticorps dirigé contre cd47/cd47 humanisé ou fragment de liaison à l'antigène ou fragment immunologiquement actif de celui-ci et utilisation associée
JP2023519620A (ja) * 2020-04-02 2023-05-11 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッド Cd47に結合する抗原結合ポリペプチド及び用途
JP2023550544A (ja) 2020-11-03 2023-12-01 アールディスカバリー エルエルシー がんおよびファゴサイトーシス不全に関連する疾患の処置のための療法
GB2617474A (en) 2020-11-04 2023-10-11 Myeloid Therapeutics Inc Engineered chimeric fusion protein compositions and methods of use thereof
AU2021373781A1 (en) * 2020-11-04 2023-06-22 The Trustees Of Dartmouth College Vista agonist for treatment/prevention of ischemic and/or reperfusion injury
EP4256336A1 (fr) 2020-12-06 2023-10-11 ALX Oncology Inc. Multimères pour réduire l'interférence de médicaments qui se lient à cd47 dans des dosages sérologiques
EP4262828A1 (fr) 2020-12-18 2023-10-25 Instil Bio (Uk) Limited Lymphocytes infiltrant les tumeurs et agents thérapeutiques anti-cd47
CA3212351A1 (fr) 2021-03-12 2022-09-15 Mendus B.V. Methodes de vaccination et utilisation d'un blocage de cd47
CN112979764B (zh) * 2021-03-26 2022-08-02 复旦大学附属中山医院 特异结合人cd47分子的多肽及其用途
TW202302145A (zh) 2021-04-14 2023-01-16 美商基利科學股份有限公司 CD47/SIRPα結合及NEDD8活化酶E1調節次單元之共抑制以用於治療癌症
KR20240023628A (ko) 2021-06-23 2024-02-22 길리애드 사이언시즈, 인코포레이티드 디아실글리세롤 키나제 조절 화합물
KR20240025616A (ko) 2021-06-23 2024-02-27 길리애드 사이언시즈, 인코포레이티드 다이아실글리세롤 키나제 조절 화합물
WO2022271650A1 (fr) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Composés de modulation de la diacylglycérol kinase
CN117355531A (zh) 2021-06-23 2024-01-05 吉利德科学公司 二酰基甘油激酶调节化合物
TW202330504A (zh) 2021-10-28 2023-08-01 美商基利科學股份有限公司 嗒𠯤—3(2h)—酮衍生物
CA3235986A1 (fr) 2021-10-29 2023-05-04 Gilead Science, Inc. Composes cd73
US20230242508A1 (en) 2021-12-22 2023-08-03 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
US20240124412A1 (en) 2021-12-22 2024-04-18 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
TW202340168A (zh) 2022-01-28 2023-10-16 美商基利科學股份有限公司 Parp7抑制劑
WO2023159220A1 (fr) * 2022-02-18 2023-08-24 Kenjockety Biotechnology, Inc. Anticorps anti-cd47
EP4245756A1 (fr) 2022-03-17 2023-09-20 Gilead Sciences, Inc. Agents de dégradation de la famille des doigts de zinc de l'ikaros et leurs utilisations
WO2023183817A1 (fr) 2022-03-24 2023-09-28 Gilead Sciences, Inc. Polythérapie pour le traitement de cancers exprimant trop -2
TW202345901A (zh) 2022-04-05 2023-12-01 美商基利科學股份有限公司 用於治療結腸直腸癌之組合療法
TW202400138A (zh) 2022-04-21 2024-01-01 美商基利科學股份有限公司 Kras g12d調節化合物
US20240116928A1 (en) 2022-07-01 2024-04-11 Gilead Sciences, Inc. Cd73 compounds
WO2024015741A1 (fr) 2022-07-12 2024-01-18 Gilead Sciences, Inc. Polypeptides immunogènes du vih et vaccins et utilisations de ceux-ci
WO2024064668A1 (fr) 2022-09-21 2024-03-28 Gilead Sciences, Inc. POLYTHÉRAPIE ANTICANCÉREUSE PAR RAYONNEMENT IONISANT FOCAL ET PERTURBATION CD47/SIRPα

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130224188A1 (en) * 2012-02-06 2013-08-29 Inhibrx Llc CD47 Antibodies and Methods of Use Thereof
US9017675B2 (en) * 2010-05-14 2015-04-28 The Board Of Trustees Of The Leland Sanford Junior University Humanized and chimeric monoclonal antibodies to CD47
US9221908B2 (en) * 2012-12-12 2015-12-29 Vasculox, Inc. Therapeutic CD47 antibodies

Family Cites Families (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1340456C (fr) 1986-07-07 1999-03-23 Hubert J.P. Schoemaker Immunoglobulines chimeriques de rongeur/homme specifiques pour antigenes associes a des tumeurs
US7531643B2 (en) 1997-09-11 2009-05-12 Chugai Seiyaku Kabushiki Kaisha Monoclonal antibody inducing apoptosis
AU740225B2 (en) 1997-09-11 2001-11-01 Chugai Seiyaku Kabushiki Kaisha Monoclonal antibody inducing apoptosis
CA2226962A1 (fr) * 1998-02-16 1999-08-16 Marie Sarfati Utilisation d'agents liants a cd47 et ces ligands pour le traitement ou prophylaxie de maladies inflammatoire, autoimmunitaire et allergique et pour le traitement de rejet de greffons
DE19813759C1 (de) 1998-03-27 1999-07-15 Gsf Forschungszentrum Umwelt Verfahren zur Induktion einer durch NK-Zellen vermittelten Immunantwort
US7696325B2 (en) 1999-03-10 2010-04-13 Chugai Seiyaku Kabushiki Kaisha Polypeptide inducing apoptosis
WO2000053634A1 (fr) 1999-03-10 2000-09-14 Chugai Seiyaku Kabushiki Kaisha Fv monocatenaire induisant l'apoptose
JP2004513066A (ja) * 1999-06-21 2004-04-30 インカイン ファーマシューティカル カンパニー,インコーポレイティド アンジオシジン:cys−ser−val−thr−cys−gly特異的腫瘍細胞付着受容体
US20010041670A1 (en) * 1999-12-06 2001-11-15 Ronit Simantov Thrombospondin-binding region of histidine-rich glycoprotein and method of use
US20050074457A1 (en) 2001-12-12 2005-04-07 Adeela Kamal Assays and implements for determining and modulating hsp90 binding activity
US20040213792A1 (en) 2003-04-24 2004-10-28 Clemmons David R. Method for inhibiting cellular activation by insulin-like growth factor-1
US8101719B2 (en) 2003-11-11 2012-01-24 Chugai Seiyaku Kabushiki Kaisha Humanized anti-CD47 antibody
WO2006031653A2 (fr) 2004-09-10 2006-03-23 Wyeth Anticorps anti-5t4 humanises et conjugues anticorps anti-5t4/calicheamicine
JP2007008895A (ja) 2005-07-04 2007-01-18 Chugai Pharmaceut Co Ltd 抗cd47抗体とインテグリンリガンドとの併用
US7514229B2 (en) 2005-09-29 2009-04-07 The Board Of Trustees Of The Leland Stanford Junior University Methods for diagnosing and evaluating treatment of blood disorders
NZ571208A (en) 2006-03-10 2011-12-22 Wyeth Corp Anti-5T4 antibodies and uses thereof
WO2008043072A2 (fr) 2006-10-05 2008-04-10 Biogen Idec Inc. Antagonistes de cd80 pour le traitement des affections néoplasiques
CA3163418A1 (fr) * 2006-10-06 2008-05-22 Jeffrey S. Isenberg Prevention de l'ischemie tissulaire et methodes et compositions associees
AU2014201010B2 (en) 2008-01-15 2016-03-03 The Board Of Trustees Of The Leland Stanford Junior University Methods for manipulating phagocytosis mediated by CD47
CA2711370C (fr) 2008-01-15 2017-06-13 The Board Of Trustees Of The Leland Stanford Junior University Marqueurs des cellules souches de la leucemie myeloide aigue
CA3054220C (fr) 2008-01-15 2022-10-11 The Board Of Trustees Of The Leland Stanford Junior University Procedes pour manipuler une phagocytose a mediation par cd47
ES2639857T3 (es) 2008-02-11 2017-10-30 Cure Tech Ltd. Anticuerpos monoclonales para el tratamiento del tumor
KR101604515B1 (ko) 2008-03-14 2016-03-17 알러간, 인코포레이티드 면역-기반 보툴리눔 독소 세로타입 a 활성 검정
EP2111869A1 (fr) 2008-04-23 2009-10-28 Stichting Sanquin Bloedvoorziening Compositions et procédés pour renforcer le système immunitaire
AU2009279676C1 (en) 2008-08-07 2015-08-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Radioprotectants targeting thrombospondin-1 and CD47
EP4134095A1 (fr) 2009-09-15 2023-02-15 The Board of Trustees of the Leland Stanford Junior University Thérapie par anti-cd47 synergique pour les cancers hématologiques
WO2011083141A2 (fr) 2010-01-08 2011-07-14 Ablynx Nv Procédé de production de séquences d'immunoglobulines par utilisation de particules de lipoprotéines
CN102812041A (zh) 2010-01-21 2012-12-05 英穆尔诺基公司 治疗卵巢癌的组合物及方法
WO2012047427A2 (fr) 2010-08-31 2012-04-12 The Regents Of The University Of California Anticorps pour neurotoxines botuliques
US20140271683A1 (en) 2010-12-21 2014-09-18 The Board Of Trustees Of The Leland Stanford Junior University Therapeutic and Diagnostic Methods for Manipulating Phagocytosis Through Calreticulin and Low Density Lipoprotein-Related Receptor
US20140140989A1 (en) 2012-02-06 2014-05-22 Inhibrx Llc Non-Platelet Depleting and Non-Red Blood Cell Depleting CD47 Antibodies and Methods of Use Thereof
AU2013278843A1 (en) 2012-06-21 2014-03-27 Compugen Ltd. LSR antibodies, and uses thereof for treatment of cancer
US11059910B2 (en) 2012-12-03 2021-07-13 Novimmune Sa Anti-CD47 antibodies and methods of use thereof
BR112015013431A2 (pt) 2012-12-12 2017-11-14 Vasculox Inc anticorpos monoclonais ou respectivos fragmentos ligantes de antígenos, composição farmacêutica, e usos de anticorpo monoclonal ou respectivo fragmento ligante de antígenos
EP4137518A1 (fr) 2013-02-06 2023-02-22 Inhibrx, Inc. Anticorps cd47 n'induisant ni l'appauvrissement en globules rouges ni l'appauvrissement en plaquettes
ES2728066T3 (es) 2013-03-15 2019-10-22 Univ Leland Stanford Junior Métodos para la obtención de dosis terapéuticamente eficaces de agentes anti-CD47
CN103665165B (zh) 2013-08-28 2016-02-24 江苏匡亚生物医药科技有限公司 一种靶向人CD47-SIRPα信号通路的双特异性抗体及其制备方法和用途
US20160130336A1 (en) 2013-12-31 2016-05-12 Development Center For Biotechnology Anti-vegf antibodies and use thereof
RU2748401C2 (ru) 2015-09-18 2021-05-25 Арч Онколоджи, Инк. Терапевтические антитела к CD47
US10946042B2 (en) 2015-12-01 2021-03-16 The Trustees Of The University Of Pennsylvania Compositions and methods for selective phagocytosis of human cancer cells
WO2018075960A1 (fr) 2016-10-21 2018-04-26 Tioma Therapeutics, Inc. Anticorps cd47 thérapeutiques
US20190309066A1 (en) 2017-03-22 2019-10-10 Arch Oncology, Inc. Combination therapy for the treatment of solid and hematological cancers
CA3057139A1 (fr) 2017-03-22 2018-09-27 Arch Oncology, Inc. Polytherapie pour le traitement de cancers solides et hematologiques

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9017675B2 (en) * 2010-05-14 2015-04-28 The Board Of Trustees Of The Leland Sanford Junior University Humanized and chimeric monoclonal antibodies to CD47
US9382320B2 (en) * 2010-05-14 2016-07-05 The Board Of Trustees Of The Leland Stanford Junior University Humanized and chimeric monoclonal antibodies to CD47
US20130224188A1 (en) * 2012-02-06 2013-08-29 Inhibrx Llc CD47 Antibodies and Methods of Use Thereof
US9221908B2 (en) * 2012-12-12 2015-12-29 Vasculox, Inc. Therapeutic CD47 antibodies
US20160137733A1 (en) * 2012-12-12 2016-05-19 Vasculox Inc. Therapeutic cd47 antibodies
US20160137734A1 (en) * 2012-12-12 2016-05-19 Vasculox Inc. Therapeutic cd47 antibodies
US9518117B2 (en) * 2012-12-12 2016-12-13 Tioma Therapeutics, Inc. Therapeutic CD47 antibodies
US9518116B2 (en) * 2012-12-12 2016-12-13 Tioma Therapeutics, Inc. Therapeutic CD47 antibodies

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9518117B2 (en) 2012-12-12 2016-12-13 Tioma Therapeutics, Inc. Therapeutic CD47 antibodies
US9518116B2 (en) 2012-12-12 2016-12-13 Tioma Therapeutics, Inc. Therapeutic CD47 antibodies
US10676524B2 (en) 2012-12-12 2020-06-09 Arch Oncology, Inc. Therapeutic CD47 antibodies
US10669336B2 (en) 2012-12-12 2020-06-02 Arch Oncology, Inc. Therapeutic CD47 antibodies
US10259873B2 (en) 2012-12-12 2019-04-16 Arch Oncology, Inc. Therapeutic CD47 antibodies
US11292834B2 (en) 2012-12-12 2022-04-05 Arch Oncology, Inc. Therapeutic CD47 antibodies
US10906954B2 (en) 2012-12-17 2021-02-02 Trillium Therapeutics Inc. Treatment of CD47+ disease cells with SIRPα-Fc fusions
US11186640B2 (en) * 2014-07-31 2021-11-30 The University Of Western Australia Method for the identification of immunotherapy-drug combinations using a network approach
CN104804093A (zh) * 2015-05-27 2015-07-29 江苏春申堂药业有限公司 一种针对cd47的单域抗体
EP3341015B2 (fr) 2015-08-26 2023-12-27 The Board of Trustees of the Leland Stanford Junior University Déplétion accrue de cellules cibles avec un blocage de cd47 et un agoniste de co-stimulation immunitaire
EP3341015B1 (fr) 2015-08-26 2021-07-28 The Board of Trustees of the Leland Stanford Junior University Déplétion accrue de cellules cibles avec un blocage de cd47 et un agoniste de co-stimulation immunitaire
JP7198083B2 (ja) 2015-08-26 2022-12-28 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー Cd47遮断及び免疫同時刺激アゴニストを用いた標的細胞の枯渇亢進
US10894831B2 (en) 2015-08-26 2021-01-19 The Board Of Trustees Of The Leland Stanford Junior University Enhanced depletion of targeted cells with CD47 blockade and an immune costimulatory agonist
AU2016310348B2 (en) * 2015-08-26 2023-01-05 The Board Of Trustees Of The Leland Stanford Junior University Enhanced depletion of targeted cells with CD47 blockade and an immune costimulatory agonist
US11608377B2 (en) 2015-08-26 2023-03-21 The Board Of Trustees Of The Leland Stanford Junior University Enhanced depletion of targeted cells with CD47 blockade and an immune costimulatory agonist
US10844124B2 (en) 2015-09-18 2020-11-24 Arch Oncology, Inc. Therapeutic CD47 antibodies
US10239945B2 (en) 2015-09-18 2019-03-26 Arch Oncology, Inc. Therapeutic CD47 antibodies
US10570201B2 (en) 2015-09-21 2020-02-25 Erasmus University Medical Center Anti-CD47 antibodies and methods of use
US9650441B2 (en) 2015-09-21 2017-05-16 Erasmus University Medical Center Anti-CD47 antibodies and methods of use
US9803016B2 (en) 2015-09-21 2017-10-31 Erasmus University Medical Center Anti-CD47 antibodies and methods of use
US10442858B2 (en) 2015-09-21 2019-10-15 Erasmus University Medical Center Anti-CD47 antibodies and methods of use
US11400145B2 (en) 2015-11-27 2022-08-02 Cartherics Pty. Ltd. Genetically modified cells and uses thereof
US20210230276A1 (en) * 2016-01-21 2021-07-29 The Board Of Trustees Of The Leland Stanford Junior University Treatment of cancer with combinations of immunoregulatory agents
US11692035B2 (en) 2016-10-21 2023-07-04 Arch Oncology, Inc. Therapeutic CD47 antibodies
US11446315B2 (en) 2016-11-03 2022-09-20 Pf Argentum Ip Holdings Llc Enhancement of CD47 blockade therapy by proteasome inhibitors
WO2018176132A1 (fr) * 2017-03-28 2018-10-04 Trillium Therapeutics Inc. Thérapie par blocage de cd47
WO2019090355A1 (fr) * 2017-11-06 2019-05-09 Children's National Medical Center Cellules exprimant des anticorps et procédés de traitement les utilisant
CN110538321A (zh) * 2018-05-29 2019-12-06 江苏恒瑞医药股份有限公司 一种cd47抗体药物组合物及其用途
WO2020036977A1 (fr) * 2018-08-13 2020-02-20 Arch Oncology, Inc. Anticorps cd47 thérapeutiques
CN113993899A (zh) * 2019-06-19 2022-01-28 乐普生物科技股份有限公司 抗cd47抗体及其应用
WO2021116699A1 (fr) 2019-12-11 2021-06-17 Precision Immunotherapeutics Limited Molécules de liaison inhibées de manière réversible
CN111635459A (zh) * 2020-06-27 2020-09-08 北京广未生物科技有限公司 抗cd47抗体及其在治疗癌症中的应用
WO2022177392A1 (fr) * 2021-02-19 2022-08-25 (주)샤페론 Anticorps à domaine unique dirigé contre cd47 et son utilisation

Also Published As

Publication number Publication date
PH12015501318A1 (en) 2015-08-24
WO2015191861A1 (fr) 2015-12-17
AU2013359167A1 (en) 2015-06-11
JP2016501892A (ja) 2016-01-21
US20180057592A1 (en) 2018-03-01
HK1216647A1 (zh) 2016-11-25
US10669336B2 (en) 2020-06-02
NZ708445A (en) 2020-10-30
SG11201504469XA (en) 2015-07-30
MX2015007446A (es) 2015-12-07
CN105102479A (zh) 2015-11-25
RU2015122228A (ru) 2017-01-19
EP2931751A4 (fr) 2016-07-13
WO2014093678A2 (fr) 2014-06-19
BR112015013431A2 (pt) 2017-11-14
EP2931751A2 (fr) 2015-10-21
WO2014093678A3 (fr) 2014-11-27
CA2894439A1 (fr) 2014-06-19
US20150274826A1 (en) 2015-10-01
EP2931751B1 (fr) 2020-02-05
US20180051081A1 (en) 2018-02-22
JP6572131B2 (ja) 2019-09-04
IL239339A0 (en) 2015-07-30
CN105102479B (zh) 2019-06-28
US10676524B2 (en) 2020-06-09
ES2786083T3 (es) 2020-10-08
KR20150093770A (ko) 2015-08-18
AU2013359167B2 (en) 2018-08-23

Similar Documents

Publication Publication Date Title
US10676524B2 (en) Therapeutic CD47 antibodies
US11292834B2 (en) Therapeutic CD47 antibodies
US10844124B2 (en) Therapeutic CD47 antibodies
US20210324075A1 (en) Therapeutic cd47 antibodies
KR102166083B1 (ko) 브라디키닌 b1 수용체 리간드에 대한 항체
JP2020511481A (ja) 固形及び血液癌の治療のための併用療法
NZ708445B2 (en) Therapeutic cd47 antibodies

Legal Events

Date Code Title Description
AS Assignment

Owner name: VACULOX INC., MISSOURI

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FRAZIER, WILLIAM A;MANNING, PAMELA T;REEL/FRAME:033294/0616

Effective date: 20140625

AS Assignment

Owner name: VASCULOX, INC., MISSOURI

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FREY, GERHARD;CHANG, HWAI WEN;REEL/FRAME:035323/0041

Effective date: 20150401

AS Assignment

Owner name: TIOMA THERAPEUTICS, INC., MISSOURI

Free format text: CHANGE OF NAME;ASSIGNOR:VASCULOX, INC.;REEL/FRAME:040044/0357

Effective date: 20160803

AS Assignment

Owner name: TIOMA THERAPEUTICS, INC., MISSOURI

Free format text: CHANGE OF NAME;ASSIGNOR:VASCULOX, INC.;REEL/FRAME:040165/0166

Effective date: 20160803

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE

AS Assignment

Owner name: VASCULOX, INC., MISSOURI

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE THE ASSIGNEE IN THE ASSIGNMENT DOCUMENT PREVIOUSLY RECORDED AT REEL: 035323 FRAME: 0041. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNORS:FREY, GERHARD;CHANG, HWAI WEN;REEL/FRAME:060204/0656

Effective date: 20150401

AS Assignment

Owner name: VASCULOX, INC., MISSOURI

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE THE NAME OF THE ASSIGNEE IN THE ASSIGNMENT DOCUMENT. PREVIOUSLY RECORDED AT REEL: 033294 FRAME: 0616. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNORS:FRAZIER, WILLIAM A.;MANNING, PAMELA T.;REEL/FRAME:060218/0592

Effective date: 20140625

AS Assignment

Owner name: ARCH ONCOLOGY, INC., MISSOURI

Free format text: CHANGE OF NAME;ASSIGNOR:TIOMA THERAPEUITICS, INC.;REEL/FRAME:060247/0083

Effective date: 20171113