US20150125446A1 - Combination therapy of an anti cd20 antibody with a btk inhibitor - Google Patents

Combination therapy of an anti cd20 antibody with a btk inhibitor Download PDF

Info

Publication number
US20150125446A1
US20150125446A1 US14/532,247 US201414532247A US2015125446A1 US 20150125446 A1 US20150125446 A1 US 20150125446A1 US 201414532247 A US201414532247 A US 201414532247A US 2015125446 A1 US2015125446 A1 US 2015125446A1
Authority
US
United States
Prior art keywords
antibody
ser
gly
val
cancer
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
US14/532,247
Other languages
English (en)
Inventor
Christian Klein
Toshio Yoshizawa
Tomoko Yasuhiro
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.)
Ono Pharmaceutical Co Ltd
Hoffmann La Roche Inc
Original Assignee
Ono Pharmaceutical Co Ltd
Hoffmann La Roche Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ono Pharmaceutical Co Ltd, Hoffmann La Roche Inc filed Critical Ono Pharmaceutical Co Ltd
Assigned to ROCHE GLYCART AG reassignment ROCHE GLYCART AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KLEIN, CHRISTIAN
Assigned to ONO PHARMACEUTICAL CO., LTD. reassignment ONO PHARMACEUTICAL CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: YASUHIRO, Tomoko, YOSHIZAWA, TOSHIO
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROCHE GLYCART AG
Assigned to HOFFMANN-LA ROCHE INC. reassignment HOFFMANN-LA ROCHE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: F. HOFFMANN-LA ROCHE AG
Publication of US20150125446A1 publication Critical patent/US20150125446A1/en
Priority to US16/679,483 priority Critical patent/US20200171153A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • 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
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • A61K31/52Purines, e.g. adenine
    • A61K31/522Purines, e.g. adenine having oxo groups directly attached to the heterocyclic ring, e.g. hypoxanthine, guanine, acyclovir
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • 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

Definitions

  • the present invention is directed to the combination therapy of an anti CD20 antibody with a BTK inhibitor for the treatment of cancer.
  • IgG1 type antibodies the most commonly used antibodies in cancer immunotherapy, are glycoproteins that have a conserved N-linked glycosylation site at Asn297 in each CH2 domain.
  • ADCC antibody dependent cellular cytotoxicity
  • the CD20 molecule (also called human B-lymphocyte-restricted differentiation antigen or Bp35) is a hydrophobic transmembrane protein located on pre-B and mature B lymphocytes that has been described extensively (Valentine, M. A., et al., J. Biol. Chem. 264 (1989) 11282-11287; and Einfeld, D. A., et al., EMBO J. 7 (1988) 711-717; Tedder, T. F., et al., Proc. Natl. Acad. Sci. U.S.A. 85 (1988) 208-212; Stamenkovic, I., et al., J. Exp. Med.
  • CD20 is expressed on greater than 90% of B cell non-Hodgkin's lymphomas (NHL) (Anderson, K. C., et al., Blood 63 (1984) 1424-1433) but is not found on hematopoietic stem cells, pro-B cells, normal plasma cells, or other normal tissues (Tedder, T. F., et al., J, Immunol. 135 (1985) 973-979).
  • NHL B cell non-Hodgkin's lymphomas
  • Type I antibodies as, e.g., rituximab (a non-afucosylated antibody with an amount of fucose of 85% or higher), ofatumumab, veltuzumab, ocrelizumab are potent in complement mediated cytotoxicity.
  • Type II antibodies as e.g. Tositumomab (B1), 11B8, AT80 or humanized B-Ly1 antibodies, effectively initiate target cell death via caspase-independent cell death induction with concomitant phosphatidylserine exposure.
  • Btk Bruton's tyrosine kinase or Bruton agammaglobulinemia tyrosine kinase
  • Btk Bruton agammaglobulinemia tyrosine kinase
  • BTK gene is located on the X chromosome. More than 400 mutations of the BTK gene have been identified.
  • BTK is activated upstream by Src-family kinases Blk, Lyn and Fyn and leads to downstream activation of essential cell survival pathways such as NF- ⁇ B and MAP kinase.
  • BTK contains a pleckstrin-homology (PH) domain that binds phosphatidylinositol (3,4,5)-trisphosphate (PI(3,4,5)P 3 ).
  • PI(3,4,5)P 3 binding induces Btk to phosphorylate phospholipase C ⁇ (PLC ⁇ ), which in turn hydrolyzes PI(4,5)P2, a phosphatidylinositol, into two second messengers, inositol triphosphate (IP3) and diacylglycerol (DAG), which then go on to modulate the activity of downstream proteins during B-cell signalling. Subsequently, Ca 2+ is mobilized and NF- ⁇ B and MAP kinase pathways are activated.
  • PLC ⁇ phosphorylate phospholipase C ⁇
  • IP3 inositol triphosphate
  • DAG diacylglycerol
  • An exemplary BTK inhibitor described in the art is ibrutinib (PCI-32765; Advani et al.; J Clin Oncol. 2013: Jan. 1; 31(1), page 88-94) which is a small molecule irreversible BTK inhibitor.
  • type I anti-CD20 antibody or an afucosylated, type II anti-CD20 antibody with a BTK inhibitor showed significantly enhanced antiproliferative effects. Surprisingly, this combination is more than additive, i.e. highly synergistic.
  • One aspect of the invention is an afucosylated anti-CD20 antibody with an amount of fucose of 60% or less of the total amount of oligosaccharides (sugars) at Asn297, for the treatment of cancer in combination with a BTK inhibitor.
  • Another aspect of the invention is the use of an afucosylated anti-CD20 antibody with an amount of fucose of 60% or less of the total amount of oligosaccharides (sugars) at Asn297, for the manufacture of a medicament for the treatment of cancer in combination with a BTK inhibitor.
  • Another aspect of the invention is a method of treatment of patient suffering from cancer by administering an afucosylated anti-CD20 antibody with an amount of fucose of 60% or less of the total amount of oligosaccharides (sugars) at Asn297, in combination with a BTK inhibitor, to a patient in the need of such treatment.
  • the amount of fucose is between 40% and 60% of the total amount of oligosaccharides (sugars) at Asn297. In another embodiment, the amount of fucose is 0% of the total amount of oligosaccharides (sugars) at Asn297.
  • the type I anti-CD20 antibody is rituximab.
  • the afucosylated anti-CD20 antibody is an IgG1 antibody.
  • said cancer is a CD20 expressing cancer, preferably a lymphoma or lymphocytic leukemia.
  • said afucosylated anti-CD20 antibody is humanized B-Ly1 antibody.
  • said afucosylated antibody is a type II anti-CD20 antibody.
  • said BTK inhibitor is a compound selected from the compounds described in WO2011/152351 and WO2013/081016.
  • Said BTK inhibitor preferably is a compound according to formula I or according to formula I-1 as disclosed herein.
  • the BTK inhibitor is 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof.
  • said type I anti-CD20 antibody is rituximab
  • said afucosylated anti-CD20 antibody is humanized B-Ly1 antibody
  • said BTK inhibitor is selected from the group consisting of: the compounds described in WO2011/152351 and WO2013/081016.
  • Said BTK inhibitor preferably is a compound according to formula I or according to formula I-1 as disclosed herein.
  • the BTK inhibitor is 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof, and said cancer is a CD20 expressing cancer, in one embodiment a lymphoma or lymphocytic leukemia.
  • the afucosylated anti-CD20 antibody binds CD20 with an KD of 10 ⁇ 8 M to 10 ⁇ 13 M.
  • One embodiment of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising a combination of a type I anti-CD20 antibody (in one embodiment rituximab) or an afucosylated anti-CD20 antibody with an amount of fucose of 60% or less of the total amount of oligosaccharides (sugars) at Asn297, (in one embodiment an afucosylated humanized B-Ly1 antibody), and a BTK inhibitor
  • the BTK inhibitor is selected from the group consisting of: the compounds described in WO2011/152351 and WO2013/081016.
  • Said BTK inhibitor preferably is a compound according to formula I or according to formula I-1 as disclosed herein.
  • the BTK inhibitor is 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof) for the treatment of cancer.
  • FIGS. 1A-1B Antitumour activity study: Mean tumour volume curves 1 A and Median tumour volume curves 1 B of mice bearing subcutaneous TMD8 cells.
  • mice were xenografted on D0. Randomization was performed on D14 (Day 0 as indicated in FIGS. 1A-1B ). Mice received one PO administration of vehicle and compound A at 10 mg/kg twice every day (Q0.5D ⁇ 24) alone or in combination with one IV injection of GA101 at 1 and 3 mg/kg once a week (Q7D ⁇ 4). Mice received one IV injection of Rituximab (at 3 mg/kg) or one IV injection of GA101 (at 1 and 3 mg/kg) once a week (D14 (Day 0), 21 (Day 7), 28 (Day 14) and D35 (Day 21): Q7D ⁇ 4). Tumor volumes were monitored and recorded twice a week.
  • FIGS. 2A-2B Antitumor activity study: Mean tumour volume curves 1 A and Median tumour volume curves 1 B of mice bearing subcutaneous TMD8 cells.
  • mice were xenografted on D0. Randomization was performed on D14 (400-450 mm 3 : Day 0 as indicated in FIGS. 2A-2B ). Mice received one PO administration of vehicle and compound A at 10 mg/kg twice every day (Q0.5D ⁇ 24) alone or in combination with one IV injection of GA101/RTX at 3 mg/kg once a week (Q7D ⁇ 4). Mice received one IV injection of Rituximab (at 3 mg/kg) or one IV injection of GA101 (at 3 mg/kg) once a week (D14 (Day 0), 21 (Day 7), 28 (Day 14) and 35 (Day 21): Q7D ⁇ 4). Tumor volumes were monitored and recorded twice a week.
  • FIG. 3 Antitumor activity study: Mean tumour volume curves of mice bearing subcutaneous TMD8 cells.
  • mice were xenografted on D0. Randomization was performed on D14 (around 450 mm 3 : Day 0 as indicated in FIG. 3 ). Mice fed a diet containing compound A at 0.0037% (converted to 6 mg/kg/day) or compound B at 0.012% (converted to 20 mg/kg/day) alone, or in combination with one IV injection of RTX at 3 mg/kg once a week (Q7D ⁇ 3). Mice received one IV injection of Rituximab (at 3 mg/kg) once a week (D14 (Day 0), 21 (Day 7) and 28 (Day 14): Q7D ⁇ 3). Tumor volumes were monitored and recorded at Day 3, Day 7, Day 11, Day 14, Day 18 and Day21.
  • the invention comprises a type I anti-CD20 antibody, or an afucosylated anti-CD20 antibody of IgG1 or IgG3 isotype with an amount of fucose of 60% or less of the total amount of oligosaccharides (sugars) at Asn297, for the treatment of cancer in combination with a BTK inhibitor.
  • the invention comprises the use of a type I anti-CD20 antibody, or an afucosylated anti-CD20 antibody of IgG1 or IgG3 isotype with an amount of fucose of 60% or less of the total amount of oligosaccharides (sugars) at Asn297, for the manufacture of a medicament for the treatment of cancer in combination with a BTK inhibitor.
  • the amount of fucose is between 40% and 60% of the total amount of oligosaccharides (sugars) at Asn297.
  • antibody encompasses the various forms of antibodies including but not being limited to whole antibodies, human antibodies, humanized antibodies and genetically engineered antibodies like monoclonal antibodies, chimeric antibodies or recombinant antibodies as well as fragments of such antibodies as long as the characteristic properties according to the invention are retained.
  • monoclonal antibody or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of a single amino acid composition.
  • human monoclonal antibody refers to antibodies displaying a single binding specificity which have variable and constant regions derived from human germline immunoglobulin sequences.
  • the human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic non-human animal, e.g. a transgenic mouse, having a genome comprising a human heavy chain transgene and a light human chain transgene fused to an immortalized cell.
  • a transgenic non-human animal e.g. a transgenic mouse
  • having a genome comprising a human heavy chain transgene and a light human chain transgene fused to an immortalized cell e.g. a transgenic mouse
  • chimeric antibody refers to a monoclonal antibody comprising a variable region, i.e., binding region, from one source or species and at least a portion of a constant region derived from a different source or species, usually prepared by recombinant DNA techniques.
  • Chimeric antibodies comprising a murine variable region and a human constant region are especially preferred.
  • Such murine/human chimeric antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding murine immunoglobulin variable regions and DNA segments encoding human immunoglobulin constant regions.
  • Other forms of “chimeric antibodies” encompassed by the present invention are those in which the class or subclass has been modified or changed from that of the original antibody.
  • Such “chimeric” antibodies are also referred to as “class-switched antibodies.”
  • Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques now well known in the art. See, e.g., Morrison, S. L., et al., Proc. Natl. Acad Sci. USA 81 (1984) 6851-6855; U.S. Pat. No. 5,202,238 and U.S. Pat. No. 5,204,244.
  • humanized antibody refers to antibodies in which the framework or “complementarity determining regions” (CDR) have been modified to comprise the CDR of an immunoglobulin of different specificity as compared to that of the parent immunoglobulin.
  • CDR complementarity determining regions
  • a murine CDR is grafted into the framework region of a human antibody to prepare the “humanized antibody.” See, e.g., Riechmann, L. et al., Nature 332 (1988) 323-327; and Neuberger, M. S. et al., Nature 314 (1985) 268-270.
  • Particularly preferred CDRs correspond to those representing sequences recognizing the antigens noted above for chimeric and bi- or multispecific antibodies.
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • Human antibodies are well-known in the state of the art (van Dijk, M. A., and van de Winkel, J. G., Curr. Opin. in Chem. Biol. 5 (2001) 368-374). Based on such technology, human antibodies against a great variety of targets can be produced. Examples of human antibodies are for example described in Kellermann, S. A., et al., Curr Opin Biotechnol. 13 (2002) 593-597.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell such as a NS0 or CHO cell or from an animal (e.g. a mouse) that is transgenic for human immunoglobulin genes or antibodies expressed using a recombinant expression vector transfected into a host cell.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences in a rearranged form.
  • the recombinant human antibodies according to the invention have been subjected to in vivo somatic hypermutation.
  • the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • bi- or multispecific antibody as used herein relates to monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, one of the binding specificities is for CD20 and the other is for any other antigen. In certain embodiments, bispecific antibodies may bind to two different epitopes of CD20. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express CD20. Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
  • binding refers to the binding of the antibody to an epitope of the tumor antigen in an in vitro assay, preferably in an plasmon resonance assay (BIAcore, GE-Healthcare Uppsala, Sweden) with purified wild-type antigen.
  • the affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), k D (dissociation constant), and K D (k D /ka).
  • Binding or specifically binding means a binding affinity (K D ) of 10 ⁇ 8 M or less, preferably 10 ⁇ 8 M to 10 ⁇ 13 M (in one embodiment 10 ⁇ 9 M to 10 ⁇ 13 M).
  • an afucosylated antibody according to the invention is specifically binding to the tumor antigen with a binding affinity (K D ) of 10 ⁇ 8 mol/l or less, preferably 10 ⁇ 8 M to 10 ⁇ 13 M (in one embodiment 10 ⁇ 9 M to 10 ⁇ 13 M).
  • nucleic acid molecule is intended to include DNA molecules and RNA molecules.
  • a nucleic acid molecule may be single-stranded or double-stranded, but preferably is double-stranded DNA.
  • the “constant domains” are not involved directly in binding the antibody to an antigen but are involved in the effector functions (ADCC, complement binding, and CDC).
  • variable region denotes each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen.
  • the domains of variable human light and heavy chains have the same general structure and each domain comprises four framework (FR) regions whose sequences are widely conserved, connected by three “hypervariable regions” (or complementarity determining regions, CDRs).
  • the framework regions adopt a ⁇ -sheet conformation and the CDRs may form loops connecting the ⁇ -sheet structure.
  • the CDRs in each chain are held in their three-dimensional structure by the framework regions and form together with the CDRs from the other chain the antigen binding site.
  • hypervariable region when used herein refer to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the hypervariable region comprises amino acid residues from the “complementarity determining regions” or “CDRs”.
  • “Framework” or “FR” regions are those variable domain regions other than the hypervariable region residues as herein defined. Therefore, the light and heavy chains of an antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • CDR3 of the heavy chain is the region which contributes most to antigen binding.
  • CDR and FR regions are determined according to the standard definition of Kabat, et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991), and/or those residues from a “hypervariable loop”.
  • afucosylated antibody refers to an antibody of IgG1 or IgG3 isotype (preferably of IgG1 isotype) with an altered pattern of glycosylation in the Fc region at Asn297 having a reduced level of fucose residues.
  • Glycosylation of human IgG1 or IgG3 occurs at Asn297 as core fucosylated bianntennary complex oligosaccharide glycosylation terminated with up to 2 Gal residues.
  • G0, G1 ( ⁇ 1,6 or ⁇ 1,3) or G2 glycan residues depending from the amount of terminal Gal residues (Raju, T. S., BioProcess Int. 1 (2003) 44-53).
  • CHO type glycosylation of antibody Fc parts is e.g. described by Routier, F. H., Glycoconjugate J. 14 (1997) 201-207.
  • Antibodies which are recombinantely expressed in non glycomodified CHO host cells usually are fucosylated at Asn297 in an amount of at least 85%.
  • an afucosylated antibody as used herein includes an antibody having no fucose in its glycosylation pattern. It is commonly known that typical glycosylated residue position in an antibody is the asparagine at position 297 according to the EU numbering system (“Asn297”).
  • EU numbering system or “EU index” is generally used when referring to a residue in an immunoglobulin heavy chain constant region (e.g., the EU index reported in Kabat et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, Md. (1991) expressly incorporated herein by reference).
  • an afucosylated antibody means an antibody of IgG1 or IgG3 isotype (preferably of IgG1 isotype) wherein the amount of fucose is 60% or less of the total amount of oligosaccharides (sugars) at Asn297 (which means that at least 40% or more of the oligosaccharides of the Fc region at Asn297 are afucosylated). In one embodiment the amount of fucose is between 40% and 60% of the oligosaccharides of the Fc region at Asn297.
  • the amount of fucose is 50% or less, and in still another embodiment the amount of fucose is 30% or less of the oligosaccharides of the Fc region at Asn297.
  • “amount of fucose” means the amount of said oligosaccharide (fucose) within the oligosaccharide (sugar) chain at Asn297, related to the sum of all oligosaccharides (sugars) attached to Asn 297 (e. g. complex, hybrid and high mannose structures) measured by MALDI-TOF mass spectrometry and calculated as average value (for a detailed procedure to determine the amount of fucose, see e.g. WO 2008/077546).
  • the oligosaccharides of the Fc region are bisected.
  • the afucosylated antibody according to the invention can be expressed in a glycomodified host cell engineered to express at least one nucleic acid encoding a polypeptide having GnTIII activity in an amount sufficient to partially fucosylate the oligosaccharides in the Fc region.
  • the polypeptide having GnTIII activity is a fusion polypeptide.
  • ⁇ 1,6-fucosyltransferase activity of the host cell can be decreased or eliminated according to U.S. Pat. No. 6,946,292 to generate glycomodified host cells.
  • the amount of antibody fucosylation can be predetermined e.g. either by fermentation conditions (e.g.
  • afucosylated antibodies and respective glycoengineering methods are described in WO 2005/044859, WO 2004/065540, WO 2007/031875, Umana, P., et al., Nature Biotechnol. 17 (1999) 176-180, WO 99/154342, WO 2005/018572, WO 2006/116260, WO 2006/114700, WO 2005/011735, WO 2005/027966, WO 97/028267, US 2006/0134709, US 2005/0054048, US 2005/0152894, WO 2003/035835, WO 2000/061739.
  • glycoengineered antibodies have an increased ADCC.
  • Other glycoengineering methods yielding afucosylated antibodies according to the invention are described e.g. in Niwa, R. et al., J. Immunol. Methods 306 (2005) 151-160; Shinkawa, T., et al., J. Biol. Chem, 278 (2003) 3466-3473; WO 03/055993 or US 2005/0249722.
  • one aspect of the invention is a type I anti-CD20 antibody, or an afucosylated anti-CD20 antibody of IgG1 or IgG3 isotype (preferably of IgG1 isotype) specifically binding to CD20 with an amount of fucose of 60% or less of the total amount of oligosaccharides (sugars) at Asn297, for the treatment of cancer in combination with a BTK inhibitor.
  • an afucosylated anti-CD20 antibody of IgG1 or IgG3 isotype specifically binding to CD20 with an amount of fucose of 60% or less of the total amount of oligosaccharides (sugars) at Asn297, for the manufacture of a medicament for the treatment of cancer in combination with a BTK inhibitor.
  • the amount of fucose is between 60% and 20% of the total amount of oligosaccharides (sugars) at Asn297.
  • the amount of fucose is between 60% and 40% of the total amount of oligosaccharides (sugars) at Asn297.
  • the amount of fucose is between 0% of the total amount of oligosaccharides (sugars) at Asn297.
  • CD20 (also known as B-lymphocyte antigen CD20, B-lymphocyte surface antigen B1, Leu-16, Bp35, BM5, and LF5; the sequence is characterized by the SwissProt database entry P11836) is a hydrophobic transmembrane protein with a molecular weight of approximately 35 kD located on pre-B and mature B lymphocytes (Valentine, M. A. et al., J. Biol. Chem. 264 (1989) 11282-11287; Tedder, T. F., et al., Proc. Natl. Acad. Sci. U.S.A. 85 (1988) 208-212; Stamenkovic, I., et al., J. Exp. Med.
  • the corresponding human gene is Membrane-spanning 4-domains, subfamily A, member 1, also known as MS4A1. This gene encodes a member of the membrane-spanning 4A gene family. Members of this nascent protein family are characterized by common structural features and similar intron/exon splice boundaries and display unique expression patterns among hematopoietic cells and nonlymphoid tissues.
  • This gene encodes the B-lymphocyte surface molecule which plays a role in the development and differentiation of B-cells into plasma cells.
  • This family member is localized to 11q12, among a cluster of family members.
  • Alternative splicing of this gene results in two transcript variants which encode the same protein.
  • CD20 and CD20 antigen are used interchangeably herein, and include any variants, isoforms and species homologs of human CD20 which are naturally expressed by cells or are expressed on cells transfected with the CD20 gene. Binding of an antibody of the invention to the CD20 antigen mediate the killing of cells expressing CD20 (e.g., a tumor cell) by inactivating CD20. The killing of the cells expressing CD20 may occur by one or more of the following mechanisms: Cell death/apoptosis induction, ADCC and CDC.
  • CD20 Synonyms of CD20, as recognized in the art, include B-lymphocyte antigen CD20, B-lymphocyte surface antigen B1, Leu-16, Bp35, BM5, and LF5.
  • anti-CD20 antibody is an antibody that binds specifically to CD20 antigen.
  • two types of anti-CD20 antibodies can be distinguished according to Cragg, M. S., et al., Blood 103 (2004) 2738-2743; and Cragg, M. S., et al., Blood 101 (2003) 1045-1052, see Table 1.
  • type II anti-CD20 antibodies include e.g. humanized B-Ly1 antibody IgG1 (a chimeric humanized IgG1 antibody as disclosed in WO 2005/044859), 11B8 IgG1 (as disclosed in WO 2004/035607), and AT80 IgG1.
  • type II anti-CD20 antibodies of the IgG1 isotype show characteristic CDC properties.
  • Type II anti-CD20 antibodies have a decreased CDC (if IgG1 isotype) compared to type I antibodies of the IgG1 isotype.
  • type I anti-CD20 antibodies include e.g. rituximab, HI47 IgG3 (ECACC, hybridoma), 2C6 IgG1 (as disclosed in WO 2005/103081), 2F2 IgG1 (as disclosed and WO 2004/035607 and WO 2005/103081) and 2H7 IgG1 (as disclosed in WO 2004/056312).
  • rituximab HI47 IgG3 (ECACC, hybridoma)
  • 2C6 IgG1 as disclosed in WO 2005/103081
  • 2F2 IgG1 as disclosed and WO 2004/035607 and WO 2005/103081
  • 2H7 IgG1 as disclosed in WO 2004/056312
  • the afucosylated anti-CD20 antibodies according to the invention is in one embodiment a type II anti-CD20 antibody, in another embodiment an afucosylated humanized B-Ly1 antibody.
  • the afucosylated anti-CD20 antibodies according to the invention have an increased antibody dependent cellular cytotoxicity (ADCC) unlike anti-CD20 antibodies having no reduced fucose.
  • ADCC antibody dependent cellular cytotoxicity
  • ADCC antibody dependent cellular cytotoxicity
  • Said “increased ADCC” can be obtained by glycoengineering of said antibodies, that means enhance said natural, cell-mediated effector functions of monoclonal antibodies by engineering their oligosaccharide component as described in Umana, P., et al., Nature Biotechnol. 17 (1999) 176-180 and U.S. Pat. No. 6,602,684.
  • CDC complement-dependent cytotoxicity
  • CDC refers to lysis of human tumor target cells by the antibody according to the invention in the presence of complement.
  • CDC is measured preferably by the treatment of a preparation of CD20 expressing cells with an anti-CD20 antibody according to the invention in the presence of complement.
  • CDC is found if the antibody induces at a concentration of 100 nM the lysis (cell death) of 20% or more of the tumor cells after 4 hours.
  • the assay is performed preferably with 51 Cr or Eu labeled tumor cells and measurement of released 51 Cr or Eu. Controls include the incubation of the tumor target cells with complement but without the antibody.
  • the “rituximab” antibody (example of a type I anti-CD20 antibody) is a genetically engineered chimeric human gamma 1 murine constant domain containing monoclonal antibody directed against the human CD20 antigen.
  • the “rituximab” antibody (example of a type I anti-CD20 antibody) is a genetically engineered chimeric human gamma 1 murine constant domain containing monoclonal antibody directed against the human CD20 antigen.
  • This chimeric antibody contains human gamma 1 constant domains and is identified by the name “C2B8” in U.S. Pat. No. 5,736,137 (Anderson et. al.) issued on Apr. 17, 1998, assigned to IDEC Pharmaceuticals Corporation.
  • Rituximab is approved for the treatment of patients with relapsed or refracting low-grade or follicular, CD20 positive, B cell non-Hodgkin's lymphoma.
  • rituximab exhibits human complement-dependent cytotoxicity (CDC) (Reff, M. E., et. al., Blood 83 (1994) 435-445). Additionally, it exhibits significant activity in assays that measure antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular cytotoxicity
  • humanized B-Ly1 antibody refers to humanized B-Ly1 antibody as disclosed in WO 2005/044859 and WO 2007/031875, which were obtained from the murine monoclonal anti-CD20 antibody B-Ly1 (variable region of the murine heavy chain (VH): SEQ ID NO:1; variable region of the murine light chain (VL): SEQ ID NO:2 (see Poppema, S. and Visser, L., Biotest Bulletin 3 (1987) 131-139) by chimerization with a human constant domain from IgG1 and following humanization (see WO 2005/044859 and WO 2007/031875).
  • VH murine heavy chain
  • VL variable region of the murine light chain
  • SEQ ID NO:2 see Poppema, S. and Visser, L., Biotest Bulletin 3 (1987) 131-139
  • the “humanized B-Ly1 antibody” has variable region of the heavy chain (VH) selected from group of SEQ ID NO:3 to SEQ ID NO:19 (B-HH2 to B-HH9 and B-HL8 to B-HL17 of WO 2005/044859 and WO 2007/031875).
  • VH variable region of the heavy chain
  • such variable domain is selected from the group consisting of SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:11, SEQ ID NO:13 and SEQ ID NO:15 (B-HH2, BHH-3, B-HH6, B-HH8, B-HL8, B-HL11 and B-HL13 of WO 2005/044859 and WO 2007/031875).
  • the “humanized B-Ly1 antibody” has variable region of the light chain (VL) of SEQ ID NO:20 (B-KV1 of WO 2005/044859 and WO 2007/031875). In one specific embodiment, the “humanized B-Ly1 antibody” has a variable region of the heavy chain (VH) of SEQ ID NO:7 (B-HH6 of WO 2005/044859 and WO 2007/031875) and a variable region of the light chain (VL) of SEQ ID NO:20 (B-KV1 of WO 2005/044859 and WO 2007/031875). Furthermore in one embodiment, the humanized B-Ly1 antibody is an IgG1 antibody.
  • such afucosylated humanized B-Ly1 antibodies are glycoengineered (GE) in the Fc region according to the procedures described in WO 2005/044859, WO 2004/065540, WO 2007/031875, Umana, P. et al., Nature Biotechnol. 17 (1999) 176-180 and WO 99/154342.
  • the afucosylated glyco-engineered humanized B-Ly1 is B-HH6-B-KV1 GE.
  • the anti-CD20 antibody is obinutuzumab (recommended INN, WHO Drug Information, Vol. 26, No. 4, 2012, p. 453).
  • obinutuzumab is synonymous for GA101.
  • the tradename is GAZYVA.
  • the WHO Drug Information document replaces all previous versions (e.g. Vol. 25, No. 1, 2011, p. 75-76), and is formerly known as afutuzumab (recommended INN, WHO Drug Information, Vol. 23, No. 2, 2009, p. 176; Vol. 22, No. 2, 2008, p. 124).
  • BTK as used herein is “Bruton's tyrosine kinase” or “Bruton agammaglobulinemia tyrosine kinase” (abbreviated Btk or BTK), which is a member of the TEC family of kinases.
  • Btk Bruton agammaglobulinemia tyrosine kinase
  • the BTK gene encodes the BTK protein, which is crucial for the development and maturation of B cells as mast cell activation through the high-affinity IgE receptor.
  • the BTK gene is located on the X chromosome. More than 400 mutations of the BTK gene have been identified.
  • BTK inhibitor refers to agents that prevents kinase activity of BTK with an IC50 of 0.001 ⁇ M to about 2 ⁇ M, in one embodiment with 0.002 ⁇ M to about 2 ⁇ M.
  • the BTK inhibitors are antibodies, antisense oligonucleotides, peptides.
  • the BTK inhibitors are small molecular weight compounds with a molecular weight (MW) of less than 1500 Daltons (Da).
  • such small molecular weight BTK inhibitor compounds are the compounds described in WO2011/152351 and WO2013/081016.
  • a small molecular weight BTK inhibitor compound according to the invention is characterized by the general formula (I)
  • L represents (1) —O—, (2) —S—, (3) —SO—, (4) —SO 2 — (5) —NH—, (6) —C(O)—, (7) —CH 2 —O—, (8) —O—CH 2 —, (9) —CH 2 —, or (10) —CH(OH)—;
  • R 1 represents (1) a halogen atom, (2) a C 1-4 alkyl group, (3) a C 1-4 alkoxy group, (4) a C 1-4 haloalkyl group, or (5) a C 1-4 haloalkoxy group;
  • ring1 represents a 4- to 7-membered cyclic group, which may be substituted by from one to five substituents each independently selected from the group consisting of (1) halogen atoms, (2) C 1-4 alkyl groups, (3) C 1-4 alkoxy groups, (4) nitrile, (5) C 1-4 haloalkyl groups, and (6) C 1-4 haloalkoxy groups, wherein when two or more substituents are present on ring1, these substituents may form a 4- to 7-membered cyclic group together with the atoms in ring1 to which these substituents are bonded;
  • ring2 represents a 4- to 7-membered saturated heterocycle, which may be substituted by from one to three —K—
  • ring1-1 represents a benzene, cyclohexane, or pyridine ring, each of which may be substituted by from one to five substituents each independently selected from the group consisting of (1) halogen atoms, (2) C 1-4 alkyl groups, (3) C 1-4 alkoxy groups, (4) nitrile, and (5) CF 3
  • ring2-1 represents a 4- to 7-membered nitrogenous saturated heterocycle, which may be substituted by from one to three —K—R 2 , wherein the other symbols have the same definitions as above);
  • R 2 is a C 2-4 alkenyl group or a C 2-4 alkynyl group, each of which may be substituted by from one to five substituents each independently selected from the group consisting of (1) NR 3 R 4 , (2) halogen atoms, (3) CONR 5 R 6 , (4) CO 2 R 7 , and (5) OR 8 ;
  • the BTK inhibitor according to the invention is 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof.
  • Salt refers to salts of the compounds as a pharmaceutically acceptable salt.
  • Such salts can be exemplified by the salts with alkali metals (potassium, sodium, and the like), salts with alkaline-earth metals (calcium, magnesium, and the like), the ammonium salt, salts with pharmaceutically acceptable organic amines (tetramethylammonium, triethylamine, methylamine, dimethylamine, cyclopentylamine, benzylamine, phenethylamine, piperidine, monoethanolamine, diethanolamine, tris(hydroxymethyl)aminomethane, lysine, arginine, N-methyl-D-glucamine, and the like), and acid addition salts (inorganic acid salts (the hydrochloride, hydrobromide, hydroiodide, sulfate, phosphate, nitrate, and the like) and organic acid salts (the acetate, trifluoroacetate, lactate, tartrate,
  • IC50 refers to the concentration of a particular compound required to inhibit 50% of a specific measured activity. IC50 of the BTK inhibitors that inhibit the BTK kinase activity can be measured, inter alia, as is described subsequently.
  • the Btk enzyme inhibitory activity was measured, based on the protocol provided by the manufacturer, using Btk (Invitrogen Corporation) and the Z′-LYTETM Kinase Assay Kit-Tyr1 peptide (Invitrogen Corporation), which contained the following reagents: Tyr-1 peptide, Thy-1 phosphopeptide, 5 ⁇ kinase buffer, ATP, development reagent B, development buffer, and stop reagent.
  • a solution of a BTK inhibitor diluted with dimethyl sulfoxide (DMSO), or DMSO 5 ⁇ L/well of a solution of a BTK inhibitor diluted with dimethyl sulfoxide (DMSO), or DMSO, and 10 ⁇ L/well of the substrate/enzyme mixture solution were dispensed to a 96-well assay plate and a reaction was carried out for 20 minutes at 30° C.
  • the substrate/enzyme mixture solution was prepared by dilution with the kinase buffer (DL-dithiothreitol (DTT, 2.7 mM), 1.33 ⁇ kinase buffer) to provide a final concentration for the Tyr-1 peptide of 4 ⁇ M and a final Btk concentration of 5 nM.
  • DTT DL-dithiothreitol
  • 10 ⁇ L of a development solution provided by diluting the development reagent B to 128 ⁇ using the development buffer, was added and a reaction was carried out for an additional 1 hour at 30° C.
  • the enzymatic reaction was then stopped by adding 10 ⁇ L of the stop solution.
  • the fluorescence intensity at 445 nm and 520 nm in each well was measured using a Fusion Universal Microplate Analyzer (PerkinElmer Inc.) fluorescence plate reader. The percent phosphorylation was determined using the ratio of the emission at 445 nm (coumarin emission) to the emission at 520 nm (fluorescein emission) in accordance with the protocol provided with the kit.
  • the percent inhibition (%) by a BTK inhibitor was calculated using the following equation.
  • AX % phosphorylation when a BTK inhibitor has been added
  • AB % phosphorylation in the absence of ATP addition (blank)
  • AC % phosphorylation when only DMSO has been added (control)
  • the 50% inhibition value (IC50 value) for a BTK inhibitor was determined from the inhibition curve based on the % inhibition at each concentration of a BTK inhibitor.
  • the oligosaccharide component can significantly affect properties relevant to the efficacy of a therapeutic glycoprotein, including physical stability, resistance to protease attack, interactions with the immune system, pharmacokinetics, and specific biological activity. Such properties may depend not only on the presence or absence, but also on the specific structures, of oligosaccharides. Some generalizations between oligosaccharide structure and glycoprotein function can be made. For example, certain oligosaccharide structures mediate rapid clearance of the glycoprotein from the bloodstream through interactions with specific carbohydrate binding proteins, while others can be bound by antibodies and trigger undesired immune reactions (Jenkins, N., et al., Nature Biotechnol. 14 (1996) 975-981).
  • Mammalian cells are the excellent hosts for production of therapeutic glycoproteins, due to their capability to glycosylate proteins in the most compatible form for human application (Cumming, D. A., et al., Glycobiology 1 (1991) 115-130; Jenkins, N., et al., Nature Biotechnol. 14 (1996) 975-981).
  • Bacteria very rarely glycosylate proteins, and like other types of common hosts, such as yeasts, filamentous fungi, insect and plant cells, yield glycosylation patterns associated with rapid clearance from the blood stream, undesirable immune interactions, and in some specific cases, reduced biological activity.
  • Chinese hamster ovary (CHO) cells have been most commonly used during the last two decades.
  • these cells allow consistent generation of genetically stable, highly productive clonal cell lines. They can be cultured to high densities in simple bioreactors using serum free media, and permit the development of safe and reproducible bioprocesses.
  • Other commonly used animal cells include baby hamster kidney (BHK) cells, NSO- and SP2/0-mouse myeloma cells. More recently, production from transgenic animals has also been tested (Jenkins, N., et al., Nature Biotechnol. 14 (1996) 975-981).
  • All antibodies contain carbohydrate structures at conserved positions in the heavy chain constant regions, with each isotype possessing a distinct array of N-linked carbohydrate structures, which variably affect protein assembly, secretion or functional activity (Wright, A., and Morrison, S. L., Trends Biotech. 15 (1997) 26-32).
  • the structure of the attached N-linked carbohydrate varies considerably, depending on the degree of processing, and can include high-mannose, multiply-branched as well as biantennary complex oligosaccharides (Wright, A., and Morrison, S. L., Trends Biotech. 15 (1997) 26-32).
  • IgG1 type antibodies the most commonly used antibodies in cancer immunotherapy, are glycoproteins that have a conserved N-linked glycosylation site at Asn297 in each CH2 domain.
  • ADCC antibody dependent cellular cytotoxicity
  • GnTIII7y ⁇ (1,4)-N-acetylglucosaminyltransferase III
  • the antibody chCE7 belongs to a large class of unconjugated monoclonal antibodies which have high tumor affinity and specificity, but have too little potency to be clinically useful when produced in standard industrial cell lines lacking the GnTIII enzyme (Umana, P., et al., Nature Biotechnol. 17 (1999) 176-180). That study was the first to show that large increases of ADCC activity could be obtained by engineering the antibody producing cells to express GnTIII, which also led to an increase in the proportion of constant region (Fc)-associated, bisected oligosaccharides, including bisected, non-fucosylated oligosaccharides, above the levels found in naturally-occurring antibodies.
  • Fc constant region
  • cancer as used herein includes lymphomas, lymphocytic leukemias, lung cancer, non small cell lung (NSCL) cancer, bronchioloalveolar cell lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, gastric cancer, colon cancer, breast cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma
  • the term “expression of the CD20” antigen is intended to indicate an significant level of expression of the CD20 antigen in a cell, preferably on the cell surface of a T- or B-cell, more preferably a B-cell, from a tumor or cancer, respectively, preferably a non-solid tumor.
  • Patients having a “CD20 expressing cancer” can be determined by standard assays known in the art. For example CD20 antigen expression can be measured using immunohistochemical (IHC) detection, FACS or via PCR-based detection of the corresponding mRNA.
  • IHC immunohistochemical
  • CD20 expressing cancer refers to all cancers in which the cancer cells show an expression of the CD20 antigen.
  • CD20 expressing cancer refers to lymphomas (preferably B-Cell Non-Hodgkin's lymphomas (NHL)) and lymphocytic leukemias.
  • lymphomas and lymphocytic leukemias include e.g.
  • follicular lymphomas b) Small Non-Cleaved Cell Lymphomas/Burkitt's lymphoma (including endemic Burkitt's lymphoma, sporadic Burkitt's lymphoma and Non-Burkitt's lymphoma), c) marginal zone lymphomas (including extranodal marginal zone B cell lymphoma (Mucosa-associated lymphatic tissue lymphomas, MALT), nodal marginal zone B cell lymphoma and splenic marginal zone lymphoma), d) Mantle cell lymphoma (MCL), e) Large Cell Lymphoma (including B-cell diffuse large cell lymphoma (DLCL), Diffuse Mixed Cell Lymphoma, Immunoblastic Lymphoma, Primary Mediastinal B-Cell Lymphoma, Angiocentric Lymphoma-Pulmonary B-Cell Lymphoma), f) hairy cell leukemia, g) lymphocy
  • the CD20 expressing cancer is a B-Cell Non-Hodgkin's lymphomas (NHL).
  • the CD20 expressing cancer is a Mantle cell lymphoma (MCL), acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CLL), B-cell diffuse large cell lymphoma (DLCL), Burkitt's lymphoma, hairy cell leukemia, follicular lymphoma, multiple myeloma, marginal zone lymphoma, post transplant lymphoproliferative disorder (PTLD), HIV associated lymphoma, waldenstrom's macroglobulinemia, or primary CNS lymphoma.
  • MCL Mantle cell lymphoma
  • ALL acute lymphocytic leukemia
  • CLL chronic lymphocytic leukemia
  • DLCL B-cell diffuse large cell lymphoma
  • Burkitt's lymphoma hairy cell leukemia
  • follicular lymphoma multiple mye
  • a method of treating when applied to, for example, cancer refers to a procedure or course of action that is designed to reduce or eliminate the number of cancer cells in a patient, or to alleviate the symptoms of a cancer.
  • a method of treating does not necessarily mean that the cancer cells or other disorder will, in fact, be eliminated, that the number of cells or disorder will, in fact, be reduced, or that the symptoms of a cancer or other disorder will, in fact, be alleviated.
  • a method of treating cancer will be performed even with a low likelihood of success, but which, given the medical history and estimated survival expectancy of a patient, is nevertheless deemed to induce an overall beneficial course of action.
  • co-administration refers to the administration of said type I anti-CD20 antibody or said afucosylated anti-CD20, and said BTK inhibitor as two separate formulations (or as one single formulation).
  • the co-administration can be simultaneous or sequential in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • Said type I anti-CD20 antibody or said anti-CD20 afucosylated antibody and said BTK inhibitor are co-administered either simultaneously or sequentially (e.g. intravenous (i.v.) through a continuous infusion (one for the anti-CD20 antibody and eventually one for said BTK inhibitor; or e.g.
  • the anti-CD20 antibody is administered intravenous (i.v.) through a continuous infusion and said BTK inhibitor is administered orally).
  • the dose is administered either on the same day in two separate administrations, or one of the agents is administered on day 1 and the second is co-administered on day 2 to day 7, preferably on day 2 to 4.
  • the term “sequentially” means within 7 days after the dose of the first component (anti-CD20 antibody or BTK inhibitor), preferably within 4 days after the dose of the first component; and the term “simultaneously” means at the same time.
  • co-administration with respect to the maintenance doses of said type I anti-CD20 antibody or said afucosylated anti-CD20 antibody and said BTK inhibitor mean that the maintenance doses can be either co-administered simultaneously, if the treatment cycle is appropriate for both drugs, e.g. every week. Or BTK inhibitor is administered e.g. every first to third day and said afucosylated antibody is administered every week. Or the maintenance doses are co-administered sequentially, either within one or within several days.
  • the antibodies are administered to the patient in a “therapeutically effective amount” (or simply “effective amount”) which is the amount of the respective compound or combination that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • a “therapeutically effective amount” or simply “effective amount” which is the amount of the respective compound or combination that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • the amount of co-administration of said type I anti-CD20 antibody or said anti-CD20 afucosylated antibody and said BTK inhibitor and the timing of co-administration will depend on the type (species, gender, age, weight, etc.) and condition of the patient being treated and the severity of the disease or condition being treated.
  • Said type I anti-CD20 antibody or said afucosylated anti-CD20 antibody and said BTK inhibitor are suitably co-administered to the patient at one time or over a series of treatments e.g. on the same day or on the day after.
  • said afucosylated anti-CD20 antibody or said BTK inhibitor may be longer than subsequent infusion times, for instance approximately 90 minutes for the initial infusion, and approximately 30 minutes for subsequent infusions (if the initial infusion is well tolerated).
  • 0.1 mg/kg to 50 mg/kg (e.g. 0.1-20 mg/kg) of said type I anti-CD20 antibody or afucosylated anti-CD20 antibody; and 1 ⁇ g/kg to 50 mg/kg (e.g. 0.1-20 mg/kg) of said BTK inhibitor is an initial candidate dosage for co-administration of both drugs to the patient.
  • the preferred dosage of said afucosylated anti-CD20 antibody preferably the afucosylated humanized B-Ly1 antibody, more preferably obinutuzumab
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg, 10 mg/kg or 30 mg/kg (or any combination thereof) may be co-administered to the patient.
  • the preferred dosage of said BTK inhibitor preferably 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof
  • said BTK inhibitor preferably 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg, 10 mg/kg or 30 mg/kg (or any combination thereof) may be co-administered to the patient.
  • the dosage and the administration schedule of said type I anti-CD20 antibody or said afucosylated anti-CD20 antibody can differ from said BTK inhibitor.
  • the type I anti-CD20 antibody or said afucosylated anti-CD20 antibody may be administered e.g. every one to three weeks and said BTK inhibitor may be administered daily or every 2 to 10 days. An initial higher loading dose, followed by one or more lower doses may also be administered.
  • the preferred dosage of type I anti-CD20 antibody (preferably rituximab) will be in the range from about 100 mg/m 2 to about 1000 mg/m 2 on day 1, 8, 15, 22, 29, 36, 43, 50, 57 of a 8-weeks-dosage-cycle.
  • the preferred dosage of said afucosylated anti-CD20 antibody (preferably the afucosylated humanized B-Ly1 antibody, more preferably obinutuzumab) will be 800 to 1600 mg (in on embodiment 800 to 1200 mg) on day 1, 8, 15 of a 3- to 6-weeks-dosage-cycle and then in a dosage of 400 to 1200 (in one embodiment 800 to 1200 mg on day 1 of up to nine 3- to 4-weeks-dosage-cycles.
  • the dose for said BTK inhibitor selected from the group consisting of: (1) 9-(1-acryloyl-3-azetidinyl)-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (2) 6-amino-9- ⁇ (3R)-1-[(2E)-4-(dimethylamino)-2-butenoyl]-3-pyrrolidinyl ⁇ -7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (3) 9-[(1-acryloyl-4-piperidinyl)methyl]-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (4) 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8
  • the dose for said BTK inhibitor selected from the group of an optical isomer of said compounds (1) to (8) and the mixture thereof is as follows.
  • the dose for said BTK inhibitor 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof is as follows.
  • Said dose according to the invention is 10 mg/kg to 70 mg/kg, preferably 20 mg/kg to 55 mg/kg, once daily or every other day as oral administration.
  • the recommended dose may vary whether there is a further co-administration of chemotherapeutic agent and based on the type of chemotherapeutic agent.
  • this invention is useful for preventing or reducing metastasis or further dissemination in such a patient suffering from cancer, preferably CD20 expressing cancer.
  • This invention is useful for increasing the duration of survival of such a patient, increasing the progression free survival of such a patient, increasing the duration of response, resulting in a statistically significant and clinically meaningful improvement of the treated patient as measured by the duration of survival, progression free survival, response rate or duration of response.
  • this invention is useful for increasing the response rate in a group of patients.
  • cytotoxic, chemotherapeutic or anti-cancer agents or compounds or ionizing radiation that enhance the effects of such agents (e.g. cytokines) may be used in the type I anti-CD20 antibody or the afucosylated anti-CD20 antibody and said BTK inhibitor combination treatment of cancer.
  • cytokines e.g. cytokines
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • said type I anti-CD20 antibody preferably rituximab or said afucosylated anti-CD20 antibody, preferably the afucosylated humanized B-Ly1 antibody, more preferably obinutuzumab, and said BTK inhibitor combination treatment is used without such additional cytotoxic, chemotherapeutic or anti-cancer agents, or compounds that enhance the effects of such additional agents.
  • Such additional agents include, for example: alkylating agents or agents with an alkylating action, such as cyclophosphamide (CTX; e.g. Cytoxan®), chlorambucil (CHL; e.g. Leukeran®), cisplatin (CisP; e.g. Platinol®) busulfan (e.g. Myleran®), melphalan, carmustine (BCNU), streptozotocin, triethylenemelamine (TEM), mitomycin C, and the like; anti-metabolites, such as methotrexate (MTX), etoposide (VP16; e.g.
  • Vepesid® 6-mercaptopurine (6MP), 6-thiocguanine (6TG), cytarabine (Ara-C), 5-fluorouracil (5-FU), capecitabine (e.g. Xeloda®), dacarbazine (DTIC), and the like; antibiotics, such as actinomycin D, doxorubicin (DXR; e.g. Adriamycin®), daunorubicin (daunomycin), bleomycin, mithramycin and the like; alkaloids, such as vinca alkaloids such as vincristine (VCR), vinblastine, and the like; and other antitumor agents, such as paclitaxel (e.g.
  • Taxol® and paclitaxel derivatives
  • the cytostatic agents glucocorticoids such as dexamethasone (DEX; e.g. Decadron®) and corticosteroids such as prednisone
  • nucleoside enzyme inhibitors such as hydroxyurea
  • amino acid depleting enzymes such as asparaginase, leucovorin and other folic acid derivatives
  • arnifostine e.g.
  • Ethyol® dactinomycin, mechlorethamine (nitrogen mustard), streptozocin, cyclophosphamide, lomustine (CCNU), doxorubicin lipo (e.g. Doxil®), gemcitabine (e.g. Gemzar®), daunorubicin lipo (e.g. Daunoxome®), procarbazine, mitomycin, docetaxel (e.g.
  • Taxotere® aldesleukin, carboplatin, oxaliplatin, cladribine, camptothecin, CPT 11 (irinotecan), 10-hydroxy 7-ethyl-camptothecin (SN38), floxuridine, fludarabine, ifosfamide, idarubicin, mesna, interferon beta, interferon alpha, mitoxantrone, topotecan, leuprolide, megestrol, melphalan, mercaptopurine, plicamycin, mitotane, pegaspargase, pentostatin, pipobroman, plicamycin, tamoxifen, teniposide, testolactone, thioguanine, thiotepa, uracil mustard, vinorelbine, chlorambucil.
  • the afucosylated anti-CD20 antibody and said BTK inhibitor combination treatment is used without such additional agents.
  • cytotoxic and anticancer agents described above as well as antiproliferative target-specific anticancer drugs like protein kinase inhibitors in chemotherapeutic regimens is generally well characterized in the cancer therapy arts, and their use herein falls under the same considerations for monitoring tolerance and effectiveness and for controlling administration routes and dosages, with some adjustments.
  • the actual dosages of the cytotoxic agents may vary depending upon the patient's cultured cell response determined by using histoculture methods. Generally, the dosage will be reduced compared to the amount used in the absence of additional other agents.
  • Typical dosages of an effective cytotoxic agent can be in the ranges recommended by the manufacturer, and where indicated by in vitro responses or responses in animal models, can be reduced by up to about one order of magnitude concentration or amount.
  • the actual dosage will depend upon the judgment of the physician, the condition of the patient, and the effectiveness of the therapeutic method based on the in vitro responsiveness of the primary cultured malignant cells or histocultured tissue sample, or the responses observed in the appropriate animal models.
  • an effective amount of ionizing radiation may be carried out and/or a radiopharmaceutical may be used in addition to the type I anti-CD20 antibody or the afucosylated anti-CD20 antibody and said BTK inhibitor combination treatment of CD20 expressing cancer.
  • the source of radiation can be either external or internal to the patient being treated. When the source is external to the patient, the therapy is known as external beam radiation therapy (EBRT). When the source of radiation is internal to the patient, the treatment is called brachytherapy (BT).
  • Radioactive atoms for use in the context of this invention can be selected from the group including, but not limited to, radium, yttrium-90, cesium-137, iridium-192, americium-241, gold-198, cobalt-57, copper-67, technetium-99, iodine-123, iodine-131, and indium-111. Is also possible to label the antibody with such radioactive isotopes. In one embodiment, the type I anti-CD20 antibody or the afucosylated anti-CD20 antibody and said BTK inhibitor combination treatment is used without such ionizing radiation.
  • Radiation therapy is a standard treatment for controlling unresectable or inoperable tumors and/or tumor metastases. Improved results have been seen when radiation therapy has been combined with chemotherapy. Radiation therapy is based on the principle that high-dose radiation delivered to a target area will result in the death of reproductive cells in both tumor and normal tissues.
  • the radiation dosage regimen is generally defined in terms of radiation absorbed dose (Gy), time and fractionation, and must be carefully defined by the oncologist.
  • the amount of radiation a patient receives will depend on various considerations, but the two most important are the location of the tumor in relation to other critical structures or organs of the body, and the extent to which the tumor has spread.
  • a typical course of treatment for a patient undergoing radiation therapy will be a treatment schedule over a 1 to 6 week period, with a total dose of between 10 and 80 Gy administered to the patient in a single daily fraction of about 1.8 to 2.0 Gy, 5 days a week.
  • the inhibition of tumor growth by means of the agents comprising the combination of the invention is enhanced when combined with radiation, optionally with additional chemotherapeutic or anticancer agents.
  • Parameters of adjuvant radiation therapies are, for example, contained in WO 99/60023.
  • the type I anti-CD20 antibody or the afucosylated anti-CD20 antibodies are administered to a patient according to known methods, by intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intra-articular, intrasynovial, or intrathecal routes.
  • the administration of the antibody is intravenous or subcutaneous.
  • the BTK inhibitor is administered to a patient according to known methods, by intravenous administration as a bolus or by continuous infusion over a period of time, orally, by intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intra-articular, intrasynovial, or intrathecal routes.
  • the administration of the BTK inhibitor is intravenous or orally.
  • a “pharmaceutically acceptable carrier” is intended to include any and all material compatible with pharmaceutical administration including solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and other materials and compounds compatible with pharmaceutical administration. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • compositions can be obtained by processing the anti-CD20 antibody and/or the BTK inhibitor according to this invention with pharmaceutically acceptable, inorganic or organic carriers.
  • Lactose, corn starch or derivatives thereof, talc, stearic acids or it's salts and the like can be used, for example, as such carriers for tablets, coated tablets, dragées and hard gelatine capsules.
  • Suitable carriers for soft gelatine capsules are, for example, vegetable oils, waxes, fats, semi-solid and liquid polyols and the like. Depending on the nature of the active substance no carriers are, however, usually required in the case of soft gelatine capsules.
  • Suitable carriers for the production of solutions and syrups are, for example, water, polyols, glycerol, vegetable oil and the like.
  • Suitable carriers for suppositories are, for example, natural or hardened oils, waxes, fats, semi-liquid or liquid polyols and the like.
  • compositions can, moreover, contain preservatives, solubilizers, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. They can also contain still other therapeutically valuable substances.
  • the composition comprises both said type I anti-CD20 antibody (preferably rituximab) or said afucosylated anti-CD20 antibody with an amount of fucose is 60% or less (preferably the afucosylated humanized B-Ly1 antibody, more preferably obinutuzumab) and said BTK inhibitor for use in the treatment of cancer, in particular of CD20 expressing cancer (preferably a lymphoma or lymphocytic leukemia, more preferably a B-Cell Non-Hodgkin's lymphoma (NHL), Mantle cell lymphoma (MCL), acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CLL), B-cell diffuse large cell lymphoma (DLCL), Burkitt's lymphoma, hairy cell leukemia, follicular lymphoma, multiple myeloma, marginal zone lymphoma, post transplant lymphoproliferative disorder (PTLD), HIV associated
  • Said pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers.
  • the present invention further provides a pharmaceutical composition, e.g. for use in cancer, comprising (i) an effective first amount of a type I anti-CD20 antibody (preferably rituximab) or an afucosylated anti-CD20 antibody with an amount of fucose is 60% or less (preferably an afucosylated humanized B-Ly1 antibody), and (ii) an effective second amount of a BTK inhibitor.
  • a pharmaceutical composition e.g. for use in cancer, comprising (i) an effective first amount of a type I anti-CD20 antibody (preferably rituximab) or an afucosylated anti-CD20 antibody with an amount of fucose is 60% or less (preferably an afucosylated humanized B-Ly1 antibody), and (ii) an effective second amount of a BTK inhibitor.
  • Such composition optionally comprises pharmaceutically acceptable carriers and/or excipients.
  • compositions of the type I anti-CD20 antibody or the afucosylated anti-CD20 antibody alone used in accordance with the present invention are prepared for storage by mixing an antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. (ed.) (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • compositions of BTK inhibitors can be similar to those described above for the type I anti-CD20 antibody or the afucosylated anti-CD20 antibody.
  • compositions of small molecule BTK inhibitor include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, as well as the particular mode of administration.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of a BTK inhibitor which produces a therapeutic effect.
  • compositions of the BTK inhibitor are prepared by uniformly and intimately bringing into association a BTK inhibitor with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • compositions suitable for oral administration may be in the form of capsules, cachets, sachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a BTK inhibitor as an active ingredient.
  • a BTK inhibitor may also be administered as a bolus, electuary or paste.
  • the type I anti-CD20 antibody or the afucosylated anti-CD20 antibody and the BTK inhibitor are formulated into two separate pharmaceutical compositions.
  • the active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interracial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and gamma-ethyl-L-glutamate non-degradable ethylene-vinyl acetate
  • degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-( ⁇ )-3-hydroxybutyric acid.
  • the formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • One embodiment is a pharmaceutical composition
  • a pharmaceutical composition comprising a combination of a type I anti-CD20 antibody or a humanized B-Ly1 antibody which is afucosylated with an amount of fucose of 60% or less of the total amount of oligosaccharides (sugars) at Asn297, and 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof, for the treatment of cancer.
  • the present invention further provides a method for the treatment of cancer, comprising administering to a patient in need of such treatment (i) an effective first amount of a type I anti-CD20 antibody or an afucosylated anti-CD20 antibody with an amount of fucose is 60% or less, (preferably an afucosylated humanized B-Ly1 antibody); and (ii) an effective second amount of a BTK inhibitor.
  • the amount of fucose of is between 40% and 60%.
  • said cancer is a CD20 expressing cancer.
  • said CD20 expressing cancer is a lymphoma or lymphocytic leukemia.
  • type I anti-CD20 antibody is rituximab.
  • said afucosylated anti-CD20 antibody is a type II anti-CD20 antibody.
  • said antibody is a humanized B-Ly1 antibody as disclosed herein.
  • said antibody is obinutuzumab.
  • said BTK inhibitor is selected from the group consisting of: (1) 9-(1-acryloyl-3-azetidinyl)-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (2) 6-amino-9- ⁇ (3R)-1-[(2E)-4-(dimethylamino)-2-butenoyl]-3-pyrrolidinyl ⁇ -7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (3) 9-[(1-acryloyl-4-piperidinyl)methyl]-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (4) 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one
  • said BTK inhibitor is 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof.
  • said salt is a hydrochloride.
  • said type I anti-CD20 antibody is rituximab or said afucosylated anti-CD20 antibody is a humanized B-Ly1 antibody and said BTK inhibitor is selected from the group consisting of: (1) 9-(1-acryloyl-3-azetidinyl)-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (2) 6-amino-9- ⁇ (3R)-1-[(2E)-4-(dimethylamino)-2-butenoyl]-3-pyrrolidinyl ⁇ -7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (3) 9-[(1-acryloyl-4-piperidinyl)methyl]-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (4) 6-amino-9-[(3R)-1-
  • said BTK inhibitor is selected from the group of an optical isomer of said compounds (1) to (8) and the mixture thereof.
  • said BTK inhibitor is 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof and said cancer is a CD20 expressing cancer, preferably a lymphoma or lymphocytic leukemia.
  • the term “patient” preferably refers to a human in need of treatment with a type I anti-CD20 antibody or an afucosylated anti-CD20 antibody (e.g. a patient suffering from CD20 expressing cancer) for any purpose, and more preferably a human in need of such a treatment to treat cancer, or a precancerous condition or lesion.
  • the term “patient” can also refer to non-human animals, preferably mammals such as dogs, cats, horses, cows, pigs, sheep and non-human primates, among others.
  • the present invention further comprises a type I anti-CD20 antibody or an afucosylated anti-CD20 antibody with an amount of fucose is 60% or less, and a BTK inhibitor for use in the treatment of cancer.
  • said type I anti-CD20 antibody is rituximab.
  • said afucosylated anti-CD20 antibody is a humanized B-Ly1 antibody.
  • said afucosylated humanized B-Ly1 antibody is obinutuzumab.
  • said BTK inhibitor is selected from the group consisting of: (1) 9-(1-acryloyl-3-azetidinyl)-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (2) 6-amino-9- ⁇ (3R)-1-[(2E)-4-(dimethylamino)-2-butenoyl]-3-pyrrolidinyl ⁇ -7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (3) 9-[(1-acryloyl-4-piperidinyl)methyl]-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (4) 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one
  • said BTK inhibitor is 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof.
  • Said compound is also shown as the following structure:
  • said salt is a hydrochloride.
  • said type I anti-CD20 antibody is rituximab or said afucosylated anti-CD20 antibody is a humanized B-Ly1 antibody, more preferably obinutuzumab
  • said BTK inhibitor is selected from the group consisting of: (1) 9-(1-acryloyl-3-azetidinyl)-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (2) 6-amino-9- ⁇ (3R)-1-[(2E)-4-(dimethylamino)-2-butenoyl]-3-pyrrolidinyl ⁇ -7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (3) 9-[(1-acryloyl-4-piperidinyl)methyl]-6-amino-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one, (4) 6-
  • said BTK inhibitor is selected from the group of an optical isomer of said compounds (1) to (8) and the mixture thereof.
  • said BTK inhibitor is 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one or a salt thereof
  • said cancer is a CD20 expressing cancer, preferably a lymphoma or lymphocytic leukemia.
  • test substance 6-amino-9-[(3R)-1-(2-butynoyl)-3-pyrrolidinyl]-7-(4-phenoxyphenyl)-7,9-dihydro-8H-purin-8-one hydrochloride (hereinafter referred to as “compound A”), was stored in sealed containers at ambient temperature, protected from light.
  • GA101 bisnutuzumab, sold under the tradename Gazyva® or Gazyvaro®
  • Rituximab sold under the tradenames Rituxan® and MabThera®
  • Compound A was weighed out and suspended in a 0.5 w/v % methyl cellulose 400 cP solution (Wako Pure Chemical Industries, Ltd., hereinafter referred to as “0.5% MC”), using a mortar and pestle, to obtain a 1 mg/mL suspension.
  • the suspension was used within 7 days of preparation. The suspension was confirmed to be stable and uniform for 7 days under refrigeration, protected from light, and for a further 24 hours at ambient temperature exposed to indoor lighting.
  • the stock solutions of both GA101 (25 mg/mL) and Rituximab (10 mg/mL) were kept at 4° C. Before administration to mice, the stock solutions were diluted in NaCl 0.9% at 0.3, 1 and 3 mg/ml for dose finding study and at 0.1 mg/ml and 0.3 mg/ml for the combination study.
  • Compound A was per os (PO) administered to mice twice daily by oral gavage via a disposable oral feeding needle (Fuchigami Kikai Co.) at 10 ml/kg.
  • GA101/Rituximab were intravenously (IV) injected at 10 ml/kg in the caudal vein of mice.
  • IV intravenously
  • the compound A b.i.d. group and the vehicle group received compound A and 0.5% MC, respectively, in the morning and afternoon at an interval of 8 h or more.
  • mice Female C.B-17/lcr-SCID/SCIDJc1 mice (Clea Japan Inc., age at initiation of experiment: 6 weeks; hereinafter referred to as “Scid mice”) were used. The mice were kept in metal mouse cages, 3 to 5 to a cage, and underwent about 1 week acclimation period prior to being subjected to the experiment. While at our facility, the animals were allowed solid feed, CRF-1 (Oriental Yeast Co., Ltd.) and tap water (from an automatic dispenser) ad libitum. Excrement was processed with an automated cleaning system. The general condition of the animals was observed upon receipt and at the completion of acclimation. Animals having no problems with general condition were used in the experiment. The animals were maintained in rooms under controlled conditions of temperature: 24 ⁇ 2° C., humidity: 55 ⁇ 15%, ventilation: 15 ⁇ 5 cycles/h with 100% fresh outside air, Lighting: 12-h dark/light cycles with illumination from fluorescent lights (light: 08:00 to 20:00).
  • TMD8 was established from cells of a patient with diffuse large B-cell lymphoma (Tohda S et al., Leuk Res., 2006, 30: 1385-90).
  • RPMI Medium 1640 Invitrogen Corporation, hereinafter referred to as “RPMI”) containing 10 vol % inactivated FBS and 1 vol % Penicillin-Streptomycin liquid (Invitrogen Corporation) was prepared and used as medium. The medium was stored at 5° C. (allowable range: 1° C. to 9° C.). The cell suspension of 4 ⁇ 106 to 2.0 ⁇ 107 cells per dish was inoculated in a 150 mm dish containing the medium. The plates were incubated at 37° C., 5% CO 2 , 95% air. Cells were counted in a hemocytometer and their viability was at least 90% the day of cells injection.
  • the pellet was suspended in RPMI previously cooled on ice to obtain a 2 ⁇ 108 cells/mL cell suspension.
  • the RPMI cell suspension and equal volume of BD Matrigel GFR (BD Biosciences) were combined to prepare a 1 ⁇ 108 cells/mL cell suspension. This was used as the cell suspension for implantation. Then, 0.1 mL of cell suspension was subcutaneously injected in the right flank of the mice under pentobarbital anesthesia, using a 25-gauge needle (Terumo Corporation).
  • mice When the mean tumor volume reached 100 to 200 mm 3 and 300 to 400 mm 3 mice were randomized in groups of 8-10 according to their individual tumor volumes and treatment was initiated. The mean tumor volume of each group was not different from the others (analysis of variance).
  • TMD8 implantation After TMD8 implantation, the general condition of all animals was recorded on each day to measure tumor volume.
  • the long axis and short axis of the tumors were measured with electronic calipers (Mitutoyo Corporation) every 2 or 4 days from TMD8 implantation to the final assessment day.
  • the primary endpoint was tumor volume on the final assessment day.
  • the tumor growth inhibition rate (TGI, %) in the treatment groups versus the vehicle group was calculated.
  • Temporal change in tumor volume and body weight from Day 0 were used as secondary endpoints.
  • Tumor volume was calculated with the following formula.
  • Tumor volume Long axis of tumor ⁇ (short axis of tumor)2 ⁇ 0.5
  • mice were eventually be sacrificed and autopsied when tumors reach a maximum of 3,000 mm 3 .
  • TGI (%) in the treated group was displayed to the first decimal place.
  • FIGS. 1A-1B show that antitumor activity study of compound A in combination with GA101 in TMD8 xenograft model.
  • D14 when mean tumor volume was 400-450 mm 3 , randomization of mice into 7 groups of 10 mice according to tumor volume occurred.
  • GA101 monotherapy showed an antitumoral effect.
  • the combination therapy groups appeared more active than the corresponding monotherapy.
  • Statistical significance was observed at Days 14, 17, 21 and 24.
  • D35 Day 21
  • 4 vehicle treated mice were sacrificed when their tumor volume was higher than 3,000 mm 3 . All mice were sacrificed at D38 (Day 24).
  • Example 2 The experiment in Example 2 was performed according to the method described in Example 1 except for the following “substance vehicle” and “treatment schedule”.
  • Compound A was weighed out and suspended in a 0.5 w/v % methyl cellulose 400 cP solution (Wako Pure Chemical Industries, Ltd., hereinafter referred to as “0.5% MC”), using a mortar and pestle, to obtain a 1 mg/mL (10 mg/kg) suspension.
  • the suspension was used within 7 days of preparation. The suspension was confirmed to be stable and uniform for 7 days under refrigeration, protected from light, and for a further 24 hours at ambient temperature exposed to indoor lighting.
  • mice When the mean tumor volume reached 400 to 450 mm 3 mice were randomized in groups of 6 according to their individual tumor volumes and treatment was initiated. The mean tumor volume of each group was not different from the others (analysis of variance).
  • FIGS. 2A-2B show that antitumor activity study of compound A in combination with GA101/RTX in TMD8 xenograft model.
  • D14 Day 0
  • mean tumor volume was 400-450 mm 3
  • randomization of mice into 6 groups of 10 mice according to tumor volume occurred.
  • each monotherapy showed an antitumoural effect.
  • Statistical significance was observed after Day 4 for GA101 and RTX and after Day 11 for compound A.
  • compound A combined with GA101 or RTX at sub-optimal dose of 3 mg/kg once weekly resulted in a significant inhibition of tumor growth in the TMD8 xenograft model.
  • TGI Tumor growth inhibition
  • TGI Tumor Growth Inhibition
  • Tumor diameter was measured and tumor volume was calculated every 3 or 4 days after group assignment.
  • Example 3 The experiment in Example 3 was performed according to the method described in Example 1 except for the following “substance vehicle”, “route of drug administration” and “treatment schedule”
  • the comparative substance ibrutinib (hereinafter referred to as “compound B”), was stored in sealed containers at ambient temperature, protected from light.
  • mice were fed a commercial diet (CRF1, Oriental Yeast Co., Ltd.) supplemented with either 0.0037% (wt/wt incorporated in the pellets) of compound A or 0.012% (wt/wt incorporated in the pellets) of compound B, respectively.
  • the dose of compound A or compound B corresponds to a daily intake of 6 or 20 mg/kg of body weight. This calculation was based on the mean daily food intakes ranged from 160 to 180 mg/g of body weight.
  • mice and RTX Mice fed a normal diet (CRF1).
  • Treatment groups Mice fed the same diet containing compound A or compound B after randomization.
  • Rituximab was administered intravenously at a 3 mg/kg once weekly.
  • FIG. 3 shows that antitumor activity study of compound A or compound B in combination with RTX in TMD8 xenograft model.
  • D14 Day 0
  • each monotherapy showed an antitumoral effect.
  • Statistical significance was observed after Day 7 for RTX and compound A and after Day 11 for compound B. No statistical significance between the each.
  • the antitumor activity of compound A (0.0037%) is comparable to compound B (0.012%).
  • compound A in combination with RTX is more effective than the respective monotherapies in the TMD8 xenograft model.
  • Statistical significance was also observed between monotherapy and this combination therapy. Meanwhile, statistical significance was not observed between compound B in combination with RTX and respective monotherapy, indicating that compound A combined with RTX demonstrated synergistic effect.
  • Tumor growth inhibition at day 0, 3, 7, 11, 14, 18 and 21 of each group shown in below.
  • Compound A combination therapy showed superior anti-tumor activity compared with RTX monotherapy in terms of tumor growth inhibition (TGI, 91.1% vs 64.2% at day 21, p ⁇ 0.001).
  • Tumor diameter was measured and tumor volume was calculated every 3 or 4 days after group assignment.
  • Example 1 Example 2 and Example 3 the combination of compound A with GA101 or RTX may be an effective treatment for ABC-DLBCL. It has been reported that compound B antagonizes RTX-dependent NK cell-mediated cytotoxicity (See, Kohrt H E et al., Blood. Vol. 123, 1957-1960, 2014). Therefore it should be noted that compound A combined with RTX or GA101 is more effective than compound B combined with RTX in this DLBCL xenograft model, indicating rationale to use these combination (compound A+RTX or GA101) in the clinical setting.
US14/532,247 2013-11-07 2014-11-04 Combination therapy of an anti cd20 antibody with a btk inhibitor Abandoned US20150125446A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/679,483 US20200171153A1 (en) 2013-11-07 2019-11-11 Combination therapy of an anti cd20 antibody with a btk inhibitor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP13192006.8 2013-11-07
EP13192006 2013-11-07

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/679,483 Continuation US20200171153A1 (en) 2013-11-07 2019-11-11 Combination therapy of an anti cd20 antibody with a btk inhibitor

Publications (1)

Publication Number Publication Date
US20150125446A1 true US20150125446A1 (en) 2015-05-07

Family

ID=49552219

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/532,247 Abandoned US20150125446A1 (en) 2013-11-07 2014-11-04 Combination therapy of an anti cd20 antibody with a btk inhibitor
US16/679,483 Pending US20200171153A1 (en) 2013-11-07 2019-11-11 Combination therapy of an anti cd20 antibody with a btk inhibitor

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/679,483 Pending US20200171153A1 (en) 2013-11-07 2019-11-11 Combination therapy of an anti cd20 antibody with a btk inhibitor

Country Status (21)

Country Link
US (2) US20150125446A1 (fr)
EP (1) EP3066126B1 (fr)
JP (1) JP6478989B2 (fr)
KR (1) KR101874215B1 (fr)
CN (3) CN105792846A (fr)
AR (1) AR098328A1 (fr)
AU (1) AU2014345712B2 (fr)
BR (1) BR112016010271A2 (fr)
CA (1) CA2926893C (fr)
ES (1) ES2728350T3 (fr)
HK (1) HK1222803A1 (fr)
IL (1) IL244960B (fr)
MX (1) MX2016005801A (fr)
MY (1) MY178726A (fr)
NZ (1) NZ718792A (fr)
PH (1) PH12016500839A1 (fr)
PL (1) PL3066126T3 (fr)
RU (1) RU2727650C2 (fr)
TR (1) TR201907240T4 (fr)
TW (1) TWI524899B (fr)
WO (1) WO2015067586A2 (fr)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150274730A1 (en) * 2014-03-25 2015-10-01 Ono Pharmaceutical Co., Ltd. Prophylactic agent and/or therapeutic agent for diffuse large b-cell lymphoma
WO2017059252A1 (fr) * 2015-10-02 2017-04-06 Gilead Sciences, Inc. Combinaisons de l'inhibiteur de btk gs-4059 avec des inhibiteurs choisis parmi un inhibiteur de jak, d'ask1, de brd et/ou de mmp9 pour traiter le cancer, les troubles allergiques, les maladies auto-immunes ou les maladies inflammatoires
WO2017059224A3 (fr) * 2015-10-01 2017-05-11 Gilead Sciences, Inc. Combinaison d'un inhibiteur de btk et d'un inhibiteur de point de contrôle pour le traitement de cancers
US9879013B2 (en) 2010-05-31 2018-01-30 Ono Pharmaceutical Co., Ltd. Purinone derivative
US9896453B2 (en) 2011-11-29 2018-02-20 Ono Pharmaceutical Co., Ltd. Purinone derivative hydrochloride
WO2018183929A1 (fr) 2017-03-30 2018-10-04 Progenity Inc. Traitement d'une maladie du tractus gastro-intestinal avec un agent immunomodulateur libéré à l'aide d'un dispositif ingérable
US10239879B2 (en) 2015-04-09 2019-03-26 Ono Pharmaceutical Co., Ltd. Process for producing purinone derivative
WO2019246317A1 (fr) 2018-06-20 2019-12-26 Progenity, Inc. Traitement d'une maladie ou d'un état dans un tissu provenant de l'endoderme
WO2019246312A1 (fr) 2018-06-20 2019-12-26 Progenity, Inc. Traitement d'une maladie du tractus gastro-intestinal avec un immunomodulateur
WO2020106754A1 (fr) 2018-11-19 2020-05-28 Progenity, Inc. Méthodes et dispositifs pour traiter une maladie à l'aide d'agents biothérapeutiques
US10941205B2 (en) 2015-10-02 2021-03-09 Hoffmann-La Roche Inc. Bispecific anti-human A-beta/human transferrin receptor antibodies and methods of use
WO2021119482A1 (fr) 2019-12-13 2021-06-17 Progenity, Inc. Dispositif ingérable pour administrer un agent thérapeutique dans le tractus gastro-intestinal
US20220040153A1 (en) * 2014-09-23 2022-02-10 Genentech, Inc. METHODS OF USING ANTI-CD79b IMMUNOCONJUGATES
US11584793B2 (en) 2015-06-24 2023-02-21 Hoffmann-La Roche Inc. Anti-transferrin receptor antibodies with tailored affinity
US11603411B2 (en) 2015-10-02 2023-03-14 Hoffmann-La Roche Inc. Bispecific anti-human CD20/human transferrin receptor antibodies and methods of use
EP4252629A2 (fr) 2016-12-07 2023-10-04 Biora Therapeutics, Inc. Procédés, dispositifs et systèmes de détection du tractus gastro-intestinal

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112012023010A8 (pt) 2010-03-12 2017-12-26 Immunogen Inc Anticorpo ou fragmento de ligação a antígeno do mesmo que se liga especificamente a cd37, imunoconjugado compreendendo os mesmos, uso dos referidos anticorpo, fragmento e imunoconjugado, composição compreendendo os mesmos, kit, célula isolada, bem como método in vitro para inibir o crescimento de uma célula que expressa cd37
CA2986437A1 (fr) 2015-06-08 2016-12-15 Debiopharm International, S.A. Combinaisons d'immunoconjugues anti-cd37 et d'anticorps anti-cd20
EA201890347A1 (ru) 2015-08-28 2018-09-28 Дебиофарм Интернэшнл, С.А. Антитела и исследования для обнаружения cd37
JP2020510608A (ja) 2016-11-02 2020-04-09 デビオファーム インターナショナル, エス. アー. 抗cd37イムノコンジュゲート療法を改善するための方法
EP3658584A1 (fr) * 2017-07-26 2020-06-03 H. Hoffnabb-La Roche Ag Polythérapie avec un inhibiteur bet, un inhibiteur bcl-2 et un anticorps anti-cd20
MX2020001493A (es) 2017-08-08 2020-03-24 Hoffmann La Roche Tratamiento con obinutuzumab nuevo subgrupo de pacientes con nuevo linfoma de celulas b grandes difusas (dlbcl).

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011152351A1 (fr) * 2010-05-31 2011-12-08 小野薬品工業株式会社 Dérivé de purinone
WO2013081016A1 (fr) * 2011-11-29 2013-06-06 小野薬品工業株式会社 Chlorhydrate de dérivé de purinone
US20140303191A1 (en) * 2011-10-19 2014-10-09 Pharmacyclics, Inc. Use of inhibitors of bruton's tyrosine kinase (btk)

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT1034298E (pt) * 1997-12-05 2012-02-03 Scripps Research Inst Humanização de anticorpo murino
KR100944575B1 (ko) * 2002-10-17 2010-02-25 젠맵 에이/에스 Cd20에 대한 인간 모노클로날 항체
MY147651A (en) * 2007-07-31 2012-12-31 Regeneron Pharma Human antibodies to human cd20 and method of using thereof
US20130302274A1 (en) * 2011-11-25 2013-11-14 Roche Glycart Ag Combination therapy
TW201427667A (zh) * 2012-11-20 2014-07-16 Celgene Avilomics Res Inc 治療和布魯頓(bruton’s)酪胺酸激酶相關之疾病或失調的方法

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011152351A1 (fr) * 2010-05-31 2011-12-08 小野薬品工業株式会社 Dérivé de purinone
US20130079327A1 (en) * 2010-05-31 2013-03-28 Shingo Yamamoto Purinone derivative
US20140303191A1 (en) * 2011-10-19 2014-10-09 Pharmacyclics, Inc. Use of inhibitors of bruton's tyrosine kinase (btk)
WO2013081016A1 (fr) * 2011-11-29 2013-06-06 小野薬品工業株式会社 Chlorhydrate de dérivé de purinone
US9199997B2 (en) * 2011-11-29 2015-12-01 Ono Pharmaceutical Co., Ltd. Purinone derivative hydrochloride

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Cheah et al. Best Practice &Research Clinical Haematology 2018, 31:41-48. (Year: 2018) *

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9879013B2 (en) 2010-05-31 2018-01-30 Ono Pharmaceutical Co., Ltd. Purinone derivative
US10487085B2 (en) 2010-05-31 2019-11-26 Ono Pharmaceutical Co., Ltd. Purinone derivative
US10233185B2 (en) 2010-05-31 2019-03-19 Ono Pharmaceutical Co., Ltd. Purinone derivative
US9926322B2 (en) 2010-05-31 2018-03-27 Ono Pharmaceutical Co., Ltd. Purinone derivative
US9896453B2 (en) 2011-11-29 2018-02-20 Ono Pharmaceutical Co., Ltd. Purinone derivative hydrochloride
US10807981B2 (en) 2011-11-29 2020-10-20 Ono Pharmaceutical Co., Ltd. Purinone derivative hydrochloride
US11292795B2 (en) 2011-11-29 2022-04-05 Ono Pharmaceutical Co., Ltd. Purinone derivative hydrochloride
US9981966B2 (en) 2011-11-29 2018-05-29 Ono Pharmaceutical Co., Ltd. Purinone derivative hydrochloride
US11897885B2 (en) 2011-11-29 2024-02-13 Ono Pharmaceutical Co., Ltd. Purinone derivative hydrochloride
US10370377B2 (en) 2011-11-29 2019-08-06 Ono Pharmaceutical Co., Ltd. Purinone derivative hydrochloride
US20150274730A1 (en) * 2014-03-25 2015-10-01 Ono Pharmaceutical Co., Ltd. Prophylactic agent and/or therapeutic agent for diffuse large b-cell lymphoma
US10137129B2 (en) 2014-03-25 2018-11-27 Ono Pharmaceutical Co., Ltd. Prophylactic agent and/or therapeutic agent for diffuse large B-cell lymphoma
US10426778B2 (en) 2014-03-25 2019-10-01 Ono Pharmaceutical Co., Ltd. Prophylactic agent and/or therapeutic agent for diffuse large B-cell lymphoma
US9416131B2 (en) * 2014-03-25 2016-08-16 Ono Pharmaceutical Co., Ltd. Prophylactic agent and/or therapeutic agent for diffuse large B-cell lymphoma
US20220040153A1 (en) * 2014-09-23 2022-02-10 Genentech, Inc. METHODS OF USING ANTI-CD79b IMMUNOCONJUGATES
US10239879B2 (en) 2015-04-09 2019-03-26 Ono Pharmaceutical Co., Ltd. Process for producing purinone derivative
US11584793B2 (en) 2015-06-24 2023-02-21 Hoffmann-La Roche Inc. Anti-transferrin receptor antibodies with tailored affinity
WO2017059224A3 (fr) * 2015-10-01 2017-05-11 Gilead Sciences, Inc. Combinaison d'un inhibiteur de btk et d'un inhibiteur de point de contrôle pour le traitement de cancers
JP2018534263A (ja) * 2015-10-02 2018-11-22 ギリアード サイエンシーズ, インコーポレイテッド 癌、アレルギー性障害、自己免疫性疾患または炎症性疾患を治療するための、jak阻害物質、ask1阻害物質、brd阻害物質及び/またはmmp9阻害物質から選択される阻害物質とbtk阻害物質gs−4059の組み合わせ
US11603411B2 (en) 2015-10-02 2023-03-14 Hoffmann-La Roche Inc. Bispecific anti-human CD20/human transferrin receptor antibodies and methods of use
WO2017059252A1 (fr) * 2015-10-02 2017-04-06 Gilead Sciences, Inc. Combinaisons de l'inhibiteur de btk gs-4059 avec des inhibiteurs choisis parmi un inhibiteur de jak, d'ask1, de brd et/ou de mmp9 pour traiter le cancer, les troubles allergiques, les maladies auto-immunes ou les maladies inflammatoires
US10941205B2 (en) 2015-10-02 2021-03-09 Hoffmann-La Roche Inc. Bispecific anti-human A-beta/human transferrin receptor antibodies and methods of use
EP4252629A2 (fr) 2016-12-07 2023-10-04 Biora Therapeutics, Inc. Procédés, dispositifs et systèmes de détection du tractus gastro-intestinal
WO2018183929A1 (fr) 2017-03-30 2018-10-04 Progenity Inc. Traitement d'une maladie du tractus gastro-intestinal avec un agent immunomodulateur libéré à l'aide d'un dispositif ingérable
EP4108183A1 (fr) 2017-03-30 2022-12-28 Biora Therapeutics, Inc. Traitement d'une maladie du tractus gastro-intestinal avec un agent immunomodulateur libéré à l'aide d'un dispositif ingérable
WO2019246312A1 (fr) 2018-06-20 2019-12-26 Progenity, Inc. Traitement d'une maladie du tractus gastro-intestinal avec un immunomodulateur
WO2019246317A1 (fr) 2018-06-20 2019-12-26 Progenity, Inc. Traitement d'une maladie ou d'un état dans un tissu provenant de l'endoderme
WO2020106757A1 (fr) 2018-11-19 2020-05-28 Progenity, Inc. Dispositif ingérable pour administrer un agent thérapeutique au tube digestif
WO2020106704A2 (fr) 2018-11-19 2020-05-28 Progenity, Inc. Dispositif ingestible pour administrer un agent therapeutique dans le tractus digestif
WO2020106754A1 (fr) 2018-11-19 2020-05-28 Progenity, Inc. Méthodes et dispositifs pour traiter une maladie à l'aide d'agents biothérapeutiques
WO2021119482A1 (fr) 2019-12-13 2021-06-17 Progenity, Inc. Dispositif ingérable pour administrer un agent thérapeutique dans le tractus gastro-intestinal
EP4309722A2 (fr) 2019-12-13 2024-01-24 Biora Therapeutics, Inc. Dispositif ingérable pour l'administration d'un agent thérapeutique au tractus gastro-intestinal

Also Published As

Publication number Publication date
IL244960B (en) 2021-08-31
EP3066126A2 (fr) 2016-09-14
CN105792846A (zh) 2016-07-20
TWI524899B (zh) 2016-03-11
MY178726A (en) 2020-10-20
WO2015067586A3 (fr) 2015-07-30
BR112016010271A2 (pt) 2017-10-03
HK1222803A1 (zh) 2017-07-14
MX2016005801A (es) 2016-11-29
PL3066126T3 (pl) 2019-08-30
NZ718792A (en) 2020-05-29
RU2016122232A3 (fr) 2018-06-18
CN114632158A (zh) 2022-06-17
TR201907240T4 (tr) 2019-06-21
JP6478989B2 (ja) 2019-03-06
CN113995843A (zh) 2022-02-01
PH12016500839A1 (en) 2016-06-13
EP3066126B1 (fr) 2019-03-27
ES2728350T3 (es) 2019-10-23
US20200171153A1 (en) 2020-06-04
CA2926893C (fr) 2022-04-12
JP2016536298A (ja) 2016-11-24
TW201521767A (zh) 2015-06-16
AU2014345712A1 (en) 2016-05-05
AR098328A1 (es) 2016-05-26
RU2727650C2 (ru) 2020-07-22
AU2014345712B2 (en) 2019-10-17
RU2016122232A (ru) 2017-12-08
KR20160079869A (ko) 2016-07-06
IL244960A0 (en) 2016-05-31
WO2015067586A2 (fr) 2015-05-14
KR101874215B1 (ko) 2018-07-03
CA2926893A1 (fr) 2015-05-14

Similar Documents

Publication Publication Date Title
US20200171153A1 (en) Combination therapy of an anti cd20 antibody with a btk inhibitor
EP2464382B1 (fr) Thérapie combinée d'un anticorps cd20 afucosylaté et de bendamustine
US20150274834A1 (en) Combination therapy of an afucosylated cd20 antibody with fludarabine and/or mitoxantrone
US20140140988A1 (en) Combination therapy of an afucosylated cd20 antibody with a mdm2 inhibitor
US20200237905A1 (en) Combination therapy with a bet inhibitor, a bcl-2 inhibitor and an anti-cd20 antibody
RU2815679C2 (ru) Комбинированная терапия на основе антитела к cd20 и ингибитора btk

Legal Events

Date Code Title Description
AS Assignment

Owner name: ONO PHARMACEUTICAL CO., LTD., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:YOSHIZAWA, TOSHIO;YASUHIRO, TOMOKO;REEL/FRAME:035217/0546

Effective date: 20141016

Owner name: ROCHE GLYCART AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KLEIN, CHRISTIAN;REEL/FRAME:035217/0463

Effective date: 20140912

Owner name: HOFFMANN-LA ROCHE INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:F. HOFFMANN-LA ROCHE AG;REEL/FRAME:035217/0712

Effective date: 20141103

Owner name: F. HOFFMANN-LA ROCHE AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ROCHE GLYCART AG;REEL/FRAME:035217/0686

Effective date: 20140922

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION