US20230279117A1 - Anti-pd-l1 antibodies and their use as therapeutics and diagnostics - Google Patents

Anti-pd-l1 antibodies and their use as therapeutics and diagnostics Download PDF

Info

Publication number
US20230279117A1
US20230279117A1 US18/050,441 US202218050441A US2023279117A1 US 20230279117 A1 US20230279117 A1 US 20230279117A1 US 202218050441 A US202218050441 A US 202218050441A US 2023279117 A1 US2023279117 A1 US 2023279117A1
Authority
US
United States
Prior art keywords
seq
antibody
human
binding
igg1
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.)
Pending
Application number
US18/050,441
Inventor
Kang Li
Jing Song
Tong Zhang
Yucheng Li
Zhiying REN
Yanshen KANG
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.)
Beigene Ltd
Original Assignee
Beigene Ltd
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 Beigene Ltd filed Critical Beigene Ltd
Priority to US18/050,441 priority Critical patent/US20230279117A1/en
Assigned to BEIGENE, LTD. reassignment BEIGENE, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KANG, Yanshen, LI, KANG, LI, YUCHENG, REN, Zhiying, SONG, JING, ZHANG, TONG
Publication of US20230279117A1 publication Critical patent/US20230279117A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • PD-L1 was initially cloned as a member (known as B7-H1) of B7 protein family (Dong et al., 1999 Nature Med 5:1365). It binds to Programmed Death-1 (PD-1) receptor and activates negative regulatory signaling pathway, inhibiting T-cell proliferation and activities (Freeman et. al. 2000 J Exp Med 192:1027). Therefore, it was also termed as PD-1 ligand 1 (PD-L1 or CD274).
  • B7-H1 B7-H1 of B7 protein family
  • PD-L1 B7-H1
  • PD-L2 B7-DC
  • Human PD-L1 gene encodes a full-length protein of 290 amino acid residues (NCBI accession NP_054862.1) with a leader peptide, which is removed after PD-L1 is expressed on cell surface as a mature protein.
  • the calculated molecular weight of the full length PD-L1 is 33 kD. However, the observed molecular weight is around 50 kD due to glycosylation, based on Western blot data from ours and others.
  • PD-L1 was found constitutively expressed in human heart, lung, thymus and vascular endothelial cells, and expressed at a low level in many other human tissues and cell types including antigen presenting cells, peripheral blood monocytes and other immune cells (Freeman et. al. 2000 J Exp Med 192:1027; Eppihimer et. al. 2002 Microcirculation 9:133). When stimulated by IFN-y, IL-12 and type I interferons, many of those cell types were found expressing increased level of PD-L1 (Bald et. al. 2014 Cancer Discov 4:674-687; Planes et. al. 2014 J Virol 88:6672-6689).
  • Blockade of PD-L1 engaging PD-1 receptor by B7-H1Ig or anti-PD-L1 antibody stimulated T-cell proliferation and functional activities (Dong et. al. 1999 Nature Med 5:1365; Freeman et. al. 2000 J Exp Med 192:1027; Tamura et. al. 2001 Blood 97:1809; Iwai et. al. 2002 PNAS 99:12293), enhanced immune responses against tumor growth and viral infection (Iwai et. al. 2002 PNAS 99:12293).
  • the popular HIV infection frequently evades human immune system by similar mechanism.
  • Therapeutic modulation of PD-L1 induced signaling by antagonist molecules may revert immune cells from tolerance, and reactivated to eradicate cancer and chronic viral infection (Blank et. al. 2005 Cancer Immunol Immunother 54:307; Okazaki et. al. 2007 Int Immunol 19:813).
  • the invention provides methods and compositions for immune-activation by inhibiting PD-L1-mediated signaling and function.
  • the invention provides an antibody antigen binding domain which specifically binds human PD-L1, and comprises a complementarity determining region (CDR) sequence described herein.
  • CDRs are amenable to recombination into heavy chain variable region (Vh) and light chain variable regions (Vk) which comprise (CDR-H1, CDR-H2 and CDR-H3) and (CDR-L1, CDR-L2 and CDR-L3) sequences, respectively and retain PD-L1-specific binding and/or functionality.
  • the domain comprises CDR1, CDR2 and CDR3, in a combination selected from (a)-(r) as follows, wherein the antibody (Ab), heavy chain (HC) or light chain (LC) and CDR nomenclature system (Kabat, IMGT or composite) from which the CDR combinations derive are shown in the first column, and residues in bold text are Kabat system, and residues underlined are IMGT system:
  • SEQ ID NO:9 SEQ ID NO:10 SEQ ID NO:11 hu333-2B/3A2 vh GFSLT SYG VH V IWAGGST NYVDSVKG AK PYGNSAMDY d Kabat SEQ ID NO:9, res. 6-10 SEQ ID NO:24 SEQ ID NO:11, res. 3-11 e IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:24, res. 2-8 SEQ ID NO:11 f Comp.
  • SEQ ID NO:9 SEQ ID NO:24 SEQ ID NO:11 hu333-3C2/3H2 vh GFSLT SYG VH V IWAGGST NYADSVKG AK PYGNSAMDY g Kabat SEQ ID NO:9, res. 6-10 SEQ ID NO:25 SEQ ID NO:11, res. 3-11 h IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:25, res. 2-8 SEQ ID NO:11 i Comp.
  • SEQ ID NO:9 SEQ ID NO:25 SEQ ID NO:11 hu333-4A2 vh GFSLT SYG VH V IWAGGST NYVDSVKG AK PYGTSAMDY j Kabat SEQ ID NO:9, res. 6-10 SEQ ID NO:24 SEQ ID NO:26, res. 3-11 k IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:24, res. 2-8 SEQ ID NO:26 l
  • SEQ ID NO:9 SEQ ID NO:25 SEQ ID NO:11 hu333-4A2 vh GFSLT SYG VH V IWAGGST NYVDSVKG AK PYGTSAMDY j Kabat SEQ ID NO:9, res. 6-10
  • SEQ ID NO:24 SEQ ID NO:26, res. 3-11 k IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:24, res. 2-8
  • SEQ ID NO:9 SEQ ID NO:24 SEQ ID NO:26 hu333-4B2 vh GFSLT SYG VH V IWAGGST NYADSVKG AK PYGTSAMDY m Kabat SEQ ID NO:9, res. 6-10 SEQ ID NO:25 SEQ ID NO:26, res. 3-11 n IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:25, res. 2-8 SEQ ID NO:26 o Comp.
  • SEQ ID NO:9 SEQ ID NO:25 SEQ ID NO:26 mu333/ hu333′s vk KAS QDVGIV VA WAS IRHT QQYSNYPLYT p Kabat SEQ ID NO:12 SEQ ID NO:13 SEQ ID NO:14 q IMGT SEQ ID NO:12, res. 4-9 SEQ ID NO:13, res. 1-3 SEQ ID NO:14 r Comp. SEQ ID NO:12 SEQ ID NO:13 SEQ ID NO:14
  • the domain comprises a heavy chain variable region (Vh) comprising a CDR1, CDR2 and CDR3 combination selected from (a)-(o), and a light chain variable region (Vk) comprising a CDR1, CDR2 and CDR3 combination selected from (p)-(r).
  • Vh heavy chain variable region
  • Vk light chain variable region
  • the domain comprises CDR1, CDR2 and CDR3, in a combination selected from (c), (f), (i), (1), (o) and (r), as follows:
  • the domain comprises a heavy chain variable region (Vh) or a light chain variable region (Vk), comprising a sequence that is:
  • mu333 vh (SEQ ID NO:6); mu333 vk (SEQ ID NO:8); hu333-1A vh (SEQ ID NO:15), hu333-1A vk (SEQ ID NO:16); hu333-2B vh (SEQ ID NO:17); hu333-3A2 vh (SEQ ID NO:18); hu333-3C2 vh (SEQ ID NO:19); hu333-3H2 vh (SEQ ID NO:20); hu333-4A2 vh (SEQ ID NO:21); hu333-4B2 vh (SEQ ID NO:22); or hu333-4B2 vk (SEQ ID NO:23).
  • the domain comprises a heavy chain variable region (Vh) and a light chain variable region (Vk) comprising a sequence that is:
  • mu333 vh and vk (SEQ ID NOS:6 and 8); hu333-1A vh and vk (SEQ ID NOS:15 and 16); hu333-2B vh and vk (SEQ ID NOS:17 and 16); hu333-3A2 vh and vk (SEQ ID NOS:18 and 23); hu333-3C2 vh and vk (SEQ ID NOS:19 and 23); hu333-3H2 vh and vk (SEQ ID NOS:20 and 23); hu333-4A2 vh and vk (SEQ ID NOS:21 and 23); or hu333-4B2 vh and vk (SEQ ID NOS:22 and 23).
  • the domain comprises comprising a heavy chain variable region (Vh) or a light chain variable region (Vk) comprising:
  • hu333-4B2 vh (SEQ ID NO:22); or hu333-4B2 vk (SEQ ID NO:23).
  • the domain comprises comprising a heavy chain variable region (Vh) and a light chain variable region (Vk) comprising:
  • hu333-4A2 vh and vk (SEQ ID NOS:21 and 23); or hu333-4B2 vh and vk (SEQ ID NOS:22 and 23)
  • the domain specifically binds PD-L1 residues: D26 and R113.
  • the invention also provides antibodies, particularly monoclonal antibodies, and F(ab) or F(ab)2 comprising a subject PD-L1 binding domain.
  • the invention also provides novel polynucleotides such as cDNAs and expression vectors, encoding a subject PD-L1 antigen binding domain, and cells comprising such polynucleotides, and non-human animals comprising such cells.
  • the polynucleotides may be operably linked to a heterologous transcription regulating sequence for expression, and may be incorporated into such vectors, cells, etc.
  • the invention provides methods of using the subject domains by administering the domain to a person determined to have cancer or a viral infection or to otherwise be in need of PD-L1 antagonism.
  • compositions of the invention are useful for the treatment of cancer, neurodegenerative and infectious, particularly viral, diseases and other conditions in which inappropriate or detrimental expression of the human PD-1 and/or is a component of the etiology or pathology of the condition.
  • the invention provides methods for treating cancer or inhibiting tumor progression in a subject in need thereof with a subject anti-PD-L1 protein, and the humanized anti-PD-1 mAbs are used as therapeutic agents to treat human diseases that are involved in suppression of immune cells by PD-1 mediated intracellular signaling, leading to disease progression, particularly cancers and viral infections.
  • the invention further provides the use of subject polynucleotides for the manufacture of a medicament for treating cancer or inhibiting tumor progression in a subject.
  • FIG. 1 Schematic presentation of full length PD-L1 (top), PD-L1/Fc (middle) and PD-L1/His (bottom).
  • ECD extracellular domain.
  • L linker.
  • Fc y4Fc fragment from human IgG4.
  • H His tag.
  • N N-terminus.
  • C C-terminus.
  • FIG. 2 A and FIG. 2 B Dose-dependent binding to the purified human PD-L1/His in ELISA ( FIG. 2 A ) or to cell surface expressed PD-L1 in FACS ( FIG. 2 B ) by murine mAbs.
  • the murine mAbs and mouse IgG as negative control were indicated at top-left corner of each figure.
  • FIG. 2 A ELISA plate was coated by PD-L1/His at 100 ng per well. The binding signal strength in ELISA was indicated by direct OD 450 readings in y-axis. Concentrations of mAbs or mouse IgG were indicated by x-axis.
  • FIG. 2 B HEK293/PD-L1 cells were stained with a serial dilution of murine anti-PD-L1 mAbs or control mouse IgG.
  • the binding signal strength was indicated by MFI (mean fluorescence intensity) in FACS analyses. Concentrations of mAbs or mouse IgG were indicated by x-axis.
  • FIG. 3 Dose-dependent response curves of murine anti-PD-L1 mAb-induced IL-2 secretion in HuT78/PD-1 cells after co-culture with HEK293/OS8/PD-L1 cells.
  • Baseline Average IL-2 release induced by mouse IgG (mIgG) at all tested concentrations. Each represents average of duplicate data points.
  • Top line Highest IL-2 release based on regression calculation by Prizm.
  • FIG. 4 Dose-dependent response curves of murine anti-PD-L1 mAb-inhibited IL-2 secretion in HuT78/P3Z cells after co-culture with HEK293/PD-L1 cells. Engagement of PD-L1 and P3Z chimeric receptor leads to activation of P3Z chimeric receptor and IL-2 secretion.
  • FIG. 5 Dose-dependent competition curves of murine anti-PD-L1 mAbs against biotin-conjugated PD-1/Fc. Fixed amount of biotin-PD-1-ECD/Fc was mixed with increasing concentrations of anti-PD-L1 mAbs indicated in x-axis. Mean fluorescence intensity (MFI) analyzed by FACS was showed in y-axis. Mouse Gig (mug) was used as a negative control.
  • MFI mean fluorescence intensity
  • FIG. 6 Dose-dependent competition curves of murine anti-PD-L1 mAbs against biotin-conjugated CD80/Fc. Fixed amount of biotin-CD80-ECD/Fc was mixed with increasing amount of anti-PD-L1 mAbs indicated in x-aixs. Mean fluorescence intensity (MFI) analyzed by FACS was showed in y-axis. Mouse IgG (muIgG) was used as a negative control.
  • MFI mean fluorescence intensity
  • FIG. 7 A and FIG. 7 B IFN-y secretion induced by humanized anti-PD-L1 Fabs in primary human PBMCs from different healthy donors. (Donor 3 ( FIG. 7 A ) and Donor 17 ( FIG. 7 B )). PBMCs were co-cultured with HEK293/OS8/PD-L1 cells for overnight. IFN-y in conditioned medium was assayed by ELISA. BSA was used as a negative control.
  • FIG. 8 C1q bindings of humanized anti-PD-L1 mAbs in wild type (IgG1wt) or mutant human IgG1 formates (IgG1mc and IgG1mf). Fifty microliters of a serial dilution (x-axis) of humanized anti-PD-L1 mAb were coated on MaxiSorp ELISA plate. Human C1q bindings (y-axis) were assessed by ELISA OD 450 readings using a specific monoclonal antibody to human C1q.
  • FIG. 9 Antibody-dependent cell-cytotoxicity (ADCC) induced by humanized anti-PD-L1 mAbs in wild type (IgG1wt) or mutant human IgG1 formats (IgG1mc and IgG1mf).
  • ADCC antibody-dependent cell-cytotoxicity
  • Human NK92MI cells transfected with FcyRIIIA were used as effector cells and HEK293/PD-L1 cells were used as target cells.
  • Percentage of cytotoxicity (y-axis) was calculated based on lactate dehydrogenase (LDH) release assay as described in Example 5.
  • FIG. 10 Complement-dependent cytotoxicity (CDC) activities of humanized anti-PD-L1 mAbs in wild type (IgG1wt) or mutant human IgG1 formats (IgG1mc and IgG1mf). Daudi/PD-L1 cells were used as target cells and human sera from healthy donors were used as the source of complement components.
  • Rituximab (Roche) was used as positive control in classical CDC assay. Percentage of CDC (y-axis) was calculated based on cell-titer glow assay as described in Example 5.
  • FIG. 11 A and FIG. 11 B IFN-y secretion induced by humanized anti-PD-L1 mAbs in primary human PBMCs from different healthy donors.. (Donor 3 ( FIG. 11 A ) and Donor 17 ( FIG. 11 B )). PBMCs were co-cultured with HEK293/OS8/PD-L1 cells for overnight. IFN-y in conditioned medium was assayed by ELISA. Human IgG served as a negative control.
  • FIG. 12 A , FIG. 12 B , and FIG. 12 C Dose-dependent bindings of anti-PD-L1 mAbs to the purified human PD-L1/His ( FIG. 12 A ), cynomolgus monkey PD-L1/His ( FIG. 12 B ), and mouse PD-L1/His ( FIG. 12 C ) in ELISA. MaxiSorp ELISA plates were coated with 50 microliters of human, monkey and mouse PD-L1/His, respectively. Concentrations of anti-PD-L1 mAbs were indicated by x-axis. The binding signal strength was indicated by direct OD 450 readings (y-axis).
  • FIG. 13 A , FIG. 13 B , and FIG. 13 C Mapping the binding epitopes of anti-PD-L1 mAbs by ELISA (upper panel) and Western Blot (lower panel).
  • FIG. 13 A Binding activities to mutant PD-L1 by hu333-4B2-IgG1.
  • FIG. 13 B Binding activities to mutant PD-L1 by Y1-I g G1.
  • FIG. 13 C Binding activities to D 26 A mutant PD-L1 by anti-PD-L1 mAbs.
  • Conditioned media containing wild type or mutant PD-L1/His proteins were used to assess binding activity by ELISA and Western Blot. ** indicates the PD-L1 mutant to which the mAb binding activity reduced to 25-50% of that to wild type PD-L1. *** indicates the PD-L1 mutant to which the mAb binding activity reduced below 25% of that to wild type PD-L1.
  • FIG. 14 A and FIG. 14 B Binding assays of anti-PD-L1 mAbs to a mixture of human serum protein and PD-L1 antigen by ELISA.
  • FIG. 14 A Dose-dependent reaction curves of murine mAb or murine chimeric mAb binding to a mixture of human serum and PD-L1/His protein. Serial dilutions of PD-L1/His protein in PBS were coated onto 96-well MaxiSorp ELISA plate as indicated, and human serum pool (from three healthy donors) were added at a fixed final concentration of 5%. Three ug/mL indicated mAbs were added to each well and incubated for one hour at room temperature.
  • FIG. 14 B Histogram showed the average OD 450 readings of three data points in left side (coated mainly with human serum proteins and with very little PD-L1/His) of curves for each mAb and the negative control (without mAb).
  • FIG. 15 Mean tumor growth curves upon treatment with hu333-4B2-IgG1mf or vehicle.
  • NOD/SCID mice implanted with human cancer cells A431 and PBMCs from healthy donors were treated with hu333-4B2-IgG1mf at the dose regimen of 10 mg/kg, twice/week.
  • PD-L1 initiates inhibitory signaling in immune cells when engaged by its ligands, PD-L1 or PD-L2.
  • the activation of PD-1 signaling promotes immune tolerance, leading to the cancers or virus-infected cells escaping from immune surveillance and cancer metastasis or viral load increase.
  • Inhibition of PD-L1 mediated cellular signaling by therapeutic agents can activate immune cells including T-cells, B-cells and NK cells, and therefore enhance immune cell functions inhibiting cancer cell growth or viral infection, and restore immune surveillance and immune memory function to treat such human diseases.
  • the invention provides antibodies whose functions are antagonistic to PD-L1-induced cellular signaling in immune cells.
  • Murine anti-PD-L1 antibodies were humanized to a high degree of similarity to human antibodies in the framework regions.
  • the full antibodies made in the modified human IgG variant format have a unique set of features in the aspects of effector functions and physicochemical properties.
  • the disclosed anti-PD-L1 antibodies are suitable for therapeutic uses in cancer treatment, controlling viral infections and other human diseases that are mechanistically involved in exacerbated immune tolerance.
  • antibody is used in the broadest sense and specifically covers antibodies (including full length monoclonal antibodies) and antibody fragments so long as they recognize PD-L1.
  • An antibody molecule is usually monospecific, but may also be described as idiospecific, heterospecific, or polyspecific.
  • Antibody molecules bind by means of specific binding sites to specific antigenic determinants or epitopes on antigens.
  • Antibody fragments comprise a portion of a full length antibody, generally the antigen binding or variable region thereof. Examples of antibody fragments include Fab, Fab′, F(ab′).sub.2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Monoclonal antibodies may be obtained by methods known to those skilled in the art. See, for example Kohler et al (1975); U.S. Pat. No. 4,376,110; Ausubel et al (1987-1999); Harlow et al (1988); and Colligan et al (1993).
  • the mAbs of the invention may be of any immunoglobulin class including IgG, IgM, IgE, IgA, and any subclass thereof.
  • a hybridoma producing a mAb may be cultivated in vitro or in vivo.
  • High titers of mAbs can be obtained in in vivo production where cells from the individual hybridomas are injected intraperitoneally into mice, such as pristine-primed Balb/c mice to produce ascites fluid containing high concentrations of the desired mAbs.
  • MAbs of isotype IgM or IgG may be purified from such ascites fluids, or from culture supernatants, using column chromatography methods well known to those of skill in the art.
  • isolated polynucleotide refers to a polynucleotide segment or fragment which has been separated from sequences which flank it in a naturally occurring state, e.g., a DNA fragment which has been removed from the sequences which are normally adjacent to the fragment, e.g., the sequences adjacent to the fragment in a genome in which it naturally occurs.
  • the term therefore includes, for example, a recombinant DNA which is incorporated into a vector, into an autonomously replicating plasmid or virus, or into the genomic DNA of a prokaryote or eukaryote, or which exists as a separate molecule (e.g., as a cDNA or a genomic or cDNA fragment produced by PCR or restriction enzyme digestion) independent of other sequences. It also includes a recombinant DNA, which is part of a hybrid gene encoding additional polypeptide sequence.
  • a “construct” means any recombinant polynucleotide molecule such as a plasmid, cosmid, virus, autonomously replicating polynucleotide molecule, phage, or linear or circular single-stranded or double-stranded DNA or RNA polynucleotide molecule, derived from any source, capable of genomic integration or autonomous replication, comprising a polynucleotide molecule where one or more polynucleotide molecule has been linked in a functionally operative manner, i.e. operably linked.
  • a recombinant construct will typically comprise the polynucleotides of the invention operably linked to transcriptional initiation regulatory sequences that will direct the transcription of the polynucleotide in the intended host cell.
  • transcriptional initiation regulatory sequences that will direct the transcription of the polynucleotide in the intended host cell.
  • Both heterologous and non-heterologous (i.e., endogenous) promoters can be employed to direct expression of the nucleic acids of the invention.
  • a “vector” refers any recombinant polynucleotide construct that may be used for the purpose of transformation, i.e. the introduction of heterologous DNA into a host cell.
  • a “plasmid” refers to a circular double stranded DNA loop into which additional DNA segments can be ligated.
  • a viral vector Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • expression vectors are referred to herein as “expression vectors”.
  • an “expression vector” as used herein refers to a nucleic acid molecule capable of replication and expressing a gene of interest when transformed, transfected or transduced into a host cell.
  • the expression vectors comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desired, provide amplification within the host.
  • the expression vector further comprises a promoter to drive the expression of the polypeptide within the cells.
  • Suitable expression vectors may be plasmids derived, for example, from pBR322 or various pUC plasmids, which are commercially available. Other expression vectors may be derived from bacteriophage, phagemid, or cosmid expression vectors.
  • Murine anti-human PD-L1 monoclonal antibodies were generated using hybridoma fusion technology (Kohler and Milstein 1975 Nature 256:495-497; Mechetner 2007 Methods Mol Biol 378:1-13) with modifications. MAbs with high binding activities in enzyme-linked immunosorbent assay (ELISA) and fluorescence-activated cell sorting (FACS) assay were selected for further characterization in cell-based functional assays.
  • ELISA enzyme-linked immunosorbent assay
  • FACS fluorescence-activated cell sorting
  • the full length human PD-L1 cDNA was synthesized by GeneScript (Nanjing, China) based on published sequence (NCBI reference sequence NM_014143.3) (SEQ. NO. 1 and 2).
  • the extracellular domain consisting of amino acids (AA) 1-239 of human PD-L1 (SEQ. NO.
  • PD-L1/Fc was purified using a Protein G Sepharose Fast Flow column (Cat. No. 17061805, GE Life Sciences, Shanghai, China).
  • PD-L1/His was purified through Ni-Sepharose Fast Flow affinity chromatography (Cat. No. 17531801, GE Life Sciences), followed by size exclusion chromatography using a HiLoad 16/60 Superdex 200 column (Cat. No. 17106901, GE Life Sciences). Both PD-L1/Fc and PD-L1/His proteins were dialyzed against phosphate buffered saline (PBS) and stored in -80° C. freezer in small aliquots.
  • PBS phosphate buffered saline
  • Expression plasmid containing full-length human PD-1 cDNA was obtained from Origene (Cat. No. SC117011, NCBI Accession No: NM_005018.1, Beijing, China).
  • the extracellular domain consisting of amino acid (AA) 1-168 of PD-1 was PCR-amplified, and subcloned in pcDNA3.1-based expression vector (Invitrogen, Carlsbad, CA, USA) with C-terminus fused to the Fc domain of human IgG4 heavy chain, abbreviated as PD-1/Fc.
  • the human CD80 (B7-1) cDNA was synthesized by GeneScript according to the published sequence (NCBI access number NM_005191.3).
  • the extracellular domain (AA1-242) of CD80 was fused with human Fc at C-terminus and subcloned in a mammalian expression vector similar to the method described previously (Pat. US 8735553).
  • the fusion protein was named as CD80-ECD/Fc or CD80/Fc.
  • Stable cell line expressing human PD-L1 was established by transfection of pcDNA3.1 plasmid containing PD-L1 into HEK293 (ATCC, Manassas, VA, USA), and followed by selection with media containing 600 micrograms of hygromycin (Cat. No. 10687-010, Invitrogen) per milliliter. Single clones were isolated by picking up single colonies from culture-dish surface. All clones were screened by FACS analysis and Western blot using PD-L1 antibody (Cat. No. 17-5983, eBioscience, San Diego, USA), and the top expression clones were used for FACS binding analyses and functional assays.
  • Murine anti-human PD-L1 monoclonal antibodies were generated using the hybridoma fusion technology. All animal protocols were reviewed by and performed following BeiGene Animal Care and Use Procedure. Ten to twelve week-old Balb/c mice (HFK Bioscience, Beijing, China) were immunized three times (3 weeks apart between injections) subcutaneously and/or intra-peritoneally, each immunization was done with 100 uL of adjuvant (Cat. No. KX0210041, KangBiQuan, Beijing, China) containing 5-10 microgram of PD-L1/Fc. Two weeks after the 2 nd immunization, the mice sera were collected and evaluated for PD-L 1 binding by ELISA and FACS.
  • adjuvant Cat. No. KX0210041, KangBiQuan, Beijing, China
  • mice with high anti-PD-L1 binding titers in sera were selected and boosted intraperitoneally with 50 micrograms of PD-L1/Fc in PBS.
  • the splenocytes were isolated and fused with the murine myeloma cell line, SP2/0 (ATCC), using standard techniques (Mechetner et. al. 2007 Methods Mol Biol 378:1-13) with some modification.
  • the supernatants of hybridoma clones were initially screened for PD-L1 binding activities by a modified ELISA assay (Flanagan 2007 Methods Mol Biol 378:33-52). Briefly, 50-200 nanograms of PD-L1/His were diluted in 50 microliters of PBS and coated in each well of 96-well ELISA plates (JinCanHua, Shenzhen, China). After blocking with 3% bovine serum albumin in TBST (20 mM Tris, 150 mM NaCl, 0.05% Tween20, pH7.5) and incubating with culture supernatants of hybridoma clones, the HRP-conjugated horse anti-mouse IgG antibody (Cat. No.
  • tetramethylbenzidine (Cat. No. PA107-01, TianGen, Beijing, China) were used to detect binding signals by a plate reader (PHREAstar, BMG Labtech, Germany) as light absorbance at 450 nm.
  • the ELISA-positive clones were further verified by fluorescence-activated cell sorting (FACS).
  • PD-L1 expression cells, HEK293/PD-L1 (10 5 cells/well) were incubated with supernatants from hybridoma clones in V-bottom 96-well plates (Cat. No. 3897, Corning).
  • the cell surface bound PD-L1 antibodies were detected with Dylight 649-conjugated goat anti-mouse IgG antibody (Cat. No. 405312, Biolegend, San Diego, CA, USA) and cell fluorescence was monitored in a Guava EasyCyte 8HT flow cytometer (Millipore, USA).
  • hybridoma clones that were positive in both ELISA and FACS assays were then tested in human immune cell-based functional assays to identify antibodies with good functional activities.
  • the hybridoma clones with positive functional activities were further subcloned and characterized.
  • the positive hybridoma clones from primary screening through ELISA, FACS and functional assays were subcloned by limiting dilution. Three subclones from each original clone were selected and confirmed in FACS and functional assays. The subclones selected through functional assays were defined as monoclonal antibody. The top subclones were adapted to grow in the CDM4MAB medium (Cat. No. SH30801.02, Hyclone) with 0-3% fetal bovine serum for purification and further characterizations.
  • Hybridoma cells or 293-F cells transiently transfected with an antibody expression plasmid (Cat. No. R79007, Invitrogen) was cultured either in CDM4MAb medium (Cat. No. SH30801.02, Hyclone) or in FreestyleTM 293 Expression medium (Cat. No. 12338018, Invitrogen), and incubated in a CO 2 incubator for 5 to 7 days at 37° C.
  • the conditioned medium was collected through centrifugation at 10,000 g for 30 minutes to remove all cells and cell debris, and filtrated through a 0.22 ⁇ m membrane before purification.
  • Murine or recombinant antibodies containing supernatants were applied and bound to a Protein A column (Cat. No.
  • Murine mAb333 has a dose-dependent binding activities with EC 50 (effective concentration at 50% activity) of 0.036 ⁇ g/mL and 0.099 ⁇ g/mL, respectively, in ELISA and FACS assays.
  • control mouse IgG (muIgG) had no bindings to PD-L1 in both assays.
  • the stable cell lines for human T cell-based functional assays were essentially the same as described in Pat. US8735553. Briefly, a fusion protein expression plasmid, OS8, was generated containing a scFv of anti-human CD3 mAb OKT3 and a C-terminal domain of mouse CD8 ⁇ which included transmembrane and cytoplasmic domains. OS8 could function as a membrane anchored T cell engager that directly activates T-cell receptor (TCR). A stable cell line that co-expresses both OS8 and PD-L1 was generated by co-transfection of two expression constructs in HEK293 cells followed by hygromycin or G418 selection for 10-14 days.
  • TCR T-cell receptor
  • This cell line was named as HEK293/OS8/PD-L1.
  • a human T-cell line, HuT78/PD-1 was generated that expresses human PD-1.
  • a reverse signaling human T-cell line, HuT78/P3Z was generated by stable transfection with a chimeric PD-1 expression construct (named as P3Z) made by fusing the extracellular and transmembrane domains of human PD-1 to the cytoplasmic region of human CD3 ⁇ chain.
  • P3Z encoded a membrane bound receptor that would activate T cells upon ligation with PD-1 ligand (PD-L1 or PD-L2).
  • Cell lines were cloned by limiting dilution as described previously (Fuller 2001 Curr Protoc Mol Biol, Chap 11, unit 11.8).
  • HEK293/OS8/PD-L1 cells were pre-incubated with anti-PD-L1 mAbs for 15 minutes prior to co-culture with HuT78/PD-1 cells (1-3 ⁇ 10 4 per well) in a flat bottom plate fed with 200 ⁇ l of RPMI1640 growth medium per well at 37° C. After 16-18 hours of co-culture, supernatants were collected. IL-2 was assayed by ELISA using human IL-2 Ready-Set-Go! ELISA kits (Cat. No. 88-7025, eBiosciences, San Diego, CA). In this assay, blockade of PD-L1-PD-1 signaling with anti-PD-L1 antibodies resulted in enhanced TCR signaling and IL-2 production.
  • a reference anti-PD-L1 mAb (named Y1) was synthesized based on the variable regions of the published data (US 2010/0203056 A1), and both human and mouse format of Y1 antibodies were generated by fusing Y1 variable regions with mouse or human IgG1 ⁇ constant regions to generate Y1-muIgG1 or Y1-huIgG1, respectively (collectively termed asY1-hIgG1). Y1-mulgG1′s function was also confirmed in this assay.
  • FIG. 3 showed the dose response curves of the representative murine-anti-PD-L1 mAbs.
  • Table 5 summarized the EC 50 and the maximum IL-2 concentration these mAbs could induce.
  • Mu333 was a potent antagonist of the PD-L1-PD-1 signaling, and induced significant IL-2 production with a very low EC 50 .
  • one of the mAbs, mu277 had much weaker blocking activity than mu333, judged by the dose dependent response curve and the parameters of top line signal read-out and EC 50 .
  • muIgG could not block PD-L1/PD-1 signaling and stimulate IL-2 production.
  • HuT78/P3Z cells PD-1 mediated TCR signaling is reversed by design as described in the previous sections.
  • HEK293/PD-L1 cells were pre-incubated with purified PD-L1 antibodies for 15 minutes prior to co-culture with HuT78/P3Z cells in 96-well flat bottom plates at 37° C. After 16-18 hours of co-culture, supernatants were collected and IL-2 production was assayed by ELISA as described above.
  • Inhibitory activity of murine anti-PD-L1 mAbs was detected directly correlated to the decrease of IL-2 release in dose-dependent fashion. Consistent with the results shown above, mu333 had potent activities inhibiting IL-2 secretion by preventing PD-L1 engagement on P3Z chimeric receptor on HuT78 cells. As showed in Table 6 and FIG. 6 , mu333 was much more potent than mu277 in terms of IC 50 (the concentration of mAb at 50% inhibition of the maximum activity), consistent with the above results obtained with regular signaling in T-cell. The negative control, muIgG, could not inhibit PD-L1/P3Z induced IL-2 production.
  • HEK293/PD-L1 cells (1 ⁇ 10 5 cells per well) were incubated with the mixture of PD-L1 antibodies and biotin-conjugated PD-1/Fc fusion protein in V-bottom 96-well plate. Biotinylation of PD-1/Fc was done using the EZ-Link Sulfo-NHS-LC-Biotin reagent according to manufacturer’s instruction (Cat. No. 21327, Thermo Sci).
  • Mu333 showed better inhibitory efficacy with an IC 50 of 0.463 ⁇ g/mL, compared to an IC 50 of 2.172 ⁇ g/mL for mu277.
  • control antibody murine IgG
  • PD-L1 Besides interaction with PD-1, PD-L1 also binds to another B7 family protein, B7-1 or alternatively named as CD80 (Butte M.J. 2007 Immunity 27:111-122).
  • CD80 NCBI accession: NP _005182.1
  • HEK293/PD-L1 cells were incubated with the mixture of PD-L1 antibodies and biotin-conjugated CD80/Fc fusion protein.
  • blockade of biotin-CD80/Fc binding to PD-L1 by anti-PD-L1 antibodies resulted in decreased binding signals (MFI readings). As shown in FIG.
  • mu333 competed off the binding of CD80 to PD-L1 with100% of max inhibition and very low IC 50 (0.052 ⁇ g/mL), which compared to the IC 50 of 0.162 ⁇ g/mL for mu277.
  • murine IgG had no such competition effect ( FIG. 6 ).
  • the 1 st strand cDNA was synthesized using reverse transcriptase (Cat. No. AH301-02, TransGen, Beijing, China).
  • PCR amplification of heavy chain (Vh) and light chain variable region (V ⁇ ) of murine mAb was performed using PCR reagent kit (Cat. No. AP221-12, TransGen, Beijing, China) and a set of primers specific for cloning of murine Vh and V ⁇ as described (Brocks 2001 Mol Med 7:461-469).
  • the PCR products were subcloned into the pEASY-Blunt cloning vector (Cat. No. CB101-02, TransGen) and subsequently sequenced by Genewiz (Beijing, China).
  • the amino acid sequences of Vh and Vk were deduced from the DNA sequencing results.
  • CDRs Complementary determinant regions
  • the three dimensional structures were simulated for variable domain of mu333 in order to identify framework residues that might be important for supporting CDR loop structures. Potentially important framework residues were kept as the original murine residues in the first round antibody humanization.
  • the previously established structural modeling method for antibodies (Morea et al. Methods 2000 20:267-279) was adopted to simulate 3D structure of anti-PD-L1 mAb mu333 based on the known canonical structures of antibodies (Al-Lazikani et al. 1997 Journal of Molecular Biology 273:927-948).
  • Vk and Vh variable domains
  • PDB database Protein Data Bank, http:// blast.ncbi.nlm.nih.gov/
  • Selected structure templates for modeling mu333 (listed in Table 10) had the same classes of canonical loop structures in L-CDR1, L-CDR2, L-CDR3, H-CDR1, and H-CDR2 to the mu333 to be modeled.
  • V ⁇ and Vh came from different immunoglobulins, they were packed together by a least-squares fit of the main chain atoms to form a hybrid structure of Vk-Vh interface residues, which was used as the templates for structural homology modeling by Swiss-model program (Kiefer et al. 2009 Nucleic Acids Research 37, D387-D392). Certain side chain conformation was adjusted while the main chain conformations were retained. At the sites where the parental structure and the modeled structure had the same residue, the side chain conformation was retained. At sites where the residues were different, side chain conformations were modeled on the basis of template structure, rotamer libraries and packing considerations. After homology modeling, PLOP program (Jacobson et al.
  • human germline IgG genes were searched for sequences that share high degree of homology to the cDNA sequences of mu333 variable regions by blasting the human immunoglobulin gene database in IMGT (http://www.imgt.org/IMGT_vquest/share/textes/index.html) and NCBI (http://www.ncbi.nlm.nih.gov/igblast/)websites.
  • IMGT http://www.imgt.org/IMGT_vquest/share/textes/index.html
  • NCBI http://www.ncbi.nlm.nih.gov/igblast/
  • Humanization was carried out by CDR-grafting (Methods in Molecular Biology, Vol 248: Antibody Engineering, Methods and Protocols, Humana Press) and the humanization antibodies (hu333s) were engineered as the human Fab format using an in-house developed expression vector.
  • mutations from murine to human amino acid residues in framework regions were guided by the simulated 3D structure, and the murine framework residues of structural importance for supporting the canonical structures of CDRs were retained in the 1 st version of humanization antibody 333 (hu333-1A, SEQ. NO. 15-16).
  • CDRs of mu333 Vk were grafted into the framework of human germline variable gene IGVK1-5, and no murine framework residues were retained (SEQ NO 16).
  • CDRs of mu333 Vh were grafted into the framework of human germline variable gene IGVH3-7, with 4 murine framework residues retained, V 24 , L 67 , K 71 and V 78 (SEQ NO 15). All grafted CDRs were based on the Kabat’s CDR definition in hu333-1A (Table 9 and SEQ. NO. 15-16). In the following hu333 variants, only the N-terminal half of Kabat H-CDR2 was grafted, as only the N-terminal half was considered to be important for antigen binding according to the simulated 3D structure (Table 14).
  • Hu333-1A were constructed as human Fab format using in-house developed expression vectors that contain human IgG CH-1 and constant region of kappa chain, respectively, with easy adapting subcloning sites.
  • the hu333-1A joined IgG2a-CH1 was tagged at C-terminus with a 8xHis peptide to facilitate purification.
  • the C 232 S and C 233 S (Kabat residue numbering, Kabat et al. Sequence of proteins of immunologic interest, 5 th ed Bethesda, MD, NIH 1991) mutations were introduced in the IgG2 heavy chain to prevent disulfide bond exchange and stabilize human IgG2 in the IgG2a conformation (Lightle et al.
  • CM5 biosensor chip Cat. No. BR100530, GE Life Sciences
  • hu333 Fabs were expressed, purified and tested in binding and functional assays as described previously. Comparing to hu333-1A, hu333-2A, hu333-2C and hu333-2D had significantly reduced binding affinities and functionalities. Only hu333-2B (SEQ. NO. 16 and 17) had similar binding and functional activities to hu333-1A (Table 11). Taken together, hu333-2B (SEQ. NO. 16 and 17) reached a high level of humanization in the framework regions while maintained potent binding affinity and functional activities.
  • IL-2 release in HuT78/PD-1 IL-2 release induced by the Fabs in HuT78/PD-1 cells co-cultured with HEK293/OS8/PD-L1 cells
  • IL-2 release in HuT78/P3Z IL-2 release induced by the Fabs in HuT78/P3Z cells co-cultured with HEK293/PD-L1 cells
  • Vh and Vk sequence composition for hu333 that could be used as therapeutic antibody in human, we further engineered the hu333 by introducing mutations in CDRs and framework regions in considerations of the antibody’s molecular properties, such as physiochemical stabilities, amino acid compositions, projected isoelectronic points (pIs), expression level, antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) while maintaining functional activities.
  • pIs projected isoelectronic points
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • Deamination site NS 76-77 in Vh of hu333-1A was mutated to NT 76-77 to generate hu333-3A2 (SEQ. NO. 18 and 23).
  • V60 of hu333-3A2 -Vh was mutated to V60A, which is consistent to the consensus sequences of major human IGVH3 genes with reduced a surface-exposure of hydrophobicity.
  • This mutant construct was given the code name hu333-3C2 (SEQ. NO. 19 and 23).
  • Another deamidation site NS 73-74 was mutated to TS 73-74 on the template of hu333-3C2, which is also consistent to the consensus sequences of major human IGVH3 genes.
  • hu333-3H2 SEQ. NO. 20 and 23.
  • hu333-3A2 hu333-3C2 and hu333-3H2 all retained the potent functional activity, only with slight variations in binding affinity.
  • these engineered hu333 variants have better projected physiochemical properties.
  • IL-2 release in HuT78/PD-1 IL-2 release induced by the Fabs in HuT78/PD-1 cells co-cultured with HEK293/OS8/PD-L1 cells
  • IL-2 release in HuT78/P3Z IL-2 release induced by the Fabs in HuT78/P3Z cells co-cultured with HEK293/PD-L1 cells
  • hu333-4A2 SEQ. NO. 21 and 23
  • hu333-4B2 SEQ. NO. 22 and 23
  • IL-2 release in HuT78/PD-1 IL-2 release induced by the Fabs in HuT78/PD-1 cells co-cultured with HEK293/OS8/PD-L1 cells
  • IL-2 release in HuT78/P3Z IL-2 release induced by the Fabs in HuT78/P3Z cells co-cultured with HEK293/PD-L1 cells
  • PBMCs peripheral blood mononuclear cells
  • the activated PBMCs were co-cultured with HEK293/OS8/PD-L1 cells in each well of 96-well plates. Functional effect of anti-PD-L1 mAbs were tested by adding the mAb to the culture, co-cultured for 15-18 hours before harvesting culture supernatants to assess IFN-y level using Ready-Set-Go! ELISA kits (Cat. No. 88-7316, eBiosciences). As shown in FIG. 7 A and FIG.
  • PD-L1 is expressed on a wide range of normal human cells including hematopoietic cells such as T-cells, B-cells, dendritic cells, macrophages, mesenchymal stem cells and bone-marrow derived mast cells, and nonhematopoietic cells and tissues such as lung, hepatocytes, pancreatic islets, placental synctiotrophoblasts and vascular endothelium (Keir et. al. 2006 J Exp Med 203:883-895, Keir et. al. 2008 Ann Rev Immunol 26:677-704, Mu et. al. 2011 Medical Oncology 28:682-688).
  • hematopoietic cells such as T-cells, B-cells, dendritic cells, macrophages, mesenchymal stem cells and bone-marrow derived mast cells
  • nonhematopoietic cells and tissues such as lung, hepatocytes, pancreatic islets,
  • PD-L1 blocking antibodies linked to human wild type IgG-yFc moieties will induce yFc-mediated effector functions, for examples, antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), which might lead to unwanted toxicity to vital organs.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • hu333-4A2 and hu333-4B2 full antibody by linking the Vh sequences to mutated IgG1 constant regions, and screened for reduced or null Fc ⁇ receptors (FcyRs) binding or C1q binding activities, therefore, attenuating or eliminating ADCC and CDC effector functions.
  • FcyRs Fc ⁇ receptors
  • IgG2 has some sequence variations from IgG1 in the hinge region, which was attributed to weaker binding or no binding to most of the FcyRs except to Fc ⁇ RIIA H131 .
  • IgG1/IgG2 hybrid (IgGl ⁇ b) with most of IgG1 hinge sequence incorporating some IgG2 sequences was demonstrated having significantly reduced the binding activities to most FcyRs and attenuated ADCC and CDC effector functions (Armour et. al. 1999 Eur J Immunol 29:2613-2624).
  • V 234 A and G 237 A were designed to reduce the surface hydrophobic side chain at the yFc/FcyR binding interface to further reduce the binding to FcyRIIA and FcyRIIB, (Lund et. al. 1992 Mol Immunol 29:53-59, Lund et. al. 1996 J Immunol 157:4963-4969, Wines et. al. 2000 J Immunol 164:5313-5318).
  • the P 239 A mutation was designed to further reduce the C1q binding and CDC (Idusogie et. al. 2000 J of Immunol 164:4178-4184).
  • the IgG1mf SEQ. NO.
  • IgG1mc 29 was similar to IgG1mc except that the amino acid residue G 237 was not mutated.
  • the recombinant full length anti-PD-L1 antibodies, hu333-4A2-IgG1mc (SEQ. NO. 30 and 32), hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) were expressed in HEK293-F cells and purified as described in previous sections.
  • ADCC is initiated when an antibody binds to cell surface target protein followed by ligation to FcyRIIIA expressed on effector cells.
  • CDC is activated when an antibody cross-links a cell surface target by binding to C1q protein, which leads to a cascade reaction of complement complex formation and activation and target cell lysis.
  • biochemical assays for antibody binding to FcyRs and C1q may serve as the fundamental indicator of ADCC and CDC.
  • FcyRs The extracellular domains of FcyRs were fused to C-terminal His tags as described in previous sections. Recombinant proteins were expressed in 293-F cells by transient transfection and purified using Ni-Sepharose column followed by gel filtration column as described. 2-5 ⁇ g/mL of FcyRs were coated on Nunc MaxiSorp ELISA plates (Cat. No. 442404, Nunc, Thermo Fisher) except FcyRIIB and FcyRIIIB, Ni-chelate plates were used (Cat. No. 15242, Pierce, Thermo Fisher). After washing and blocking of the wells, a preformed immune-complex was added to each well and incubated at room temperature for 1-2 hours.
  • the preformed immune-complex contained 60 ng/mL streptavidin-HRP, 60 ng/mL of biotinylated-F(ab′) 2 goat anti-human IgG (Cat. No. 109-066-097, Jackson ImmunoRes, West Grove, PA, USA), and 1-5 ⁇ g/mL of indicated IgG1 Fc variants fused to the humanized anti-PD-L1 (hu333-4A2 or hu333-4B2) in the blocking buffer. After washing the plate four times, binding signals were detected by chemiluminescence using Immobilon Chemiluminescence Substrate A/B (Cat. No. WBKLS0500, Millipore).
  • Table 16 summarized the results of hu333-4A2-IgG1mc (SEQ. NO. 30 and 32), hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) binding to various FcyRs.
  • all three IgG1 mutant hu333 mAbs had very low binding activities to FcyRs, which indicated that all three hu333 mAbs above would have significantly reduced effector functions mediated by FcyRs.
  • Bindings of humanized anti-PD-L1 in various IgG1 formats (wt, IgG1mc, IgG1mf, SEQ. NO. 27-29) to FcyRs were also determined by flow cytometry.
  • a series of stable HEK293 transfectants expressing human FcyRs were established. These stable cell lines expressed FcyRI, Fc ⁇ RIIA H131 , Fc ⁇ RIIA R131 , Fc ⁇ RIIB, Fc ⁇ RIIIA F158 or Fc ⁇ RIIIA V158 , respectively.
  • Multi-subunit FcyRs i.e., Fc ⁇ RI and Fc ⁇ RIIIA were co-expressed with FcRy subunit.
  • a secondary antibody (goat anti-human IgG F(ab′) 2 -Alexa Fluor 488, Cat. No. 109-546-097, Jackson ImmunoResearch, West Grove, PA, USA) was used to detect the binding of monomeric anti-PD-L1 mAbs with the IgG1 variants (Table 17) to Fc ⁇ R expressing HEK293 cells.
  • hu333-4A2 in IgG1wt format had strong binding signals (MFI) to FcyRI, Fc ⁇ RIIA H131 and Fc ⁇ RIIIA V158 , and weak but significant binding signals to Fc ⁇ RIIA R131 , Fe ⁇ RIIB and Fc ⁇ RIIIA F158 (Table 17).
  • the modified IgG1 variants (hu333-4A2-1gGlmc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf, SEQ. NO. 30-33) had significantly reduced binding signals which were close to backgrounds.
  • Binding strength (MFI*) of monomeric IgG1 variants to FcyRs determined by FACS mAbs Fc ⁇ RI Fc ⁇ RIIA H131 Fc ⁇ RIIA R131 Fe ⁇ RIIB Fc ⁇ RIIIA F158 Fc ⁇ RIIIA V158 hu333-4A2-IgG1wt 1169.42 40.52 15.14 19.00 29.45 91.65 hu333-4A2-IgG1mc 4.29 5.78 3.80 3.71 5.20 3.87 hu333-4B2-IgG1mc 4.78 6.16 3.64 4.49 5.42 4.14 hu333-4B2-IgG1mf 4.56 6.12 3.99 3.73 5.09 3.91 MFI: Mean flourescence intensity from FACS analysis
  • the ELISA-based C1q binding assay was done by conventional ELISA method with minor modification. Briefly, indicated amounts of the humanized 333 antibodies fused to either wild type or modified IgG1 constant regions were coated onto the Maxisorp ELISA plate. After blocking and washing, the wells were incubated with 2 ⁇ g/mL of human C1q (Cat. No. A400, Quidel, San Diego, USA) at room temperature for 2 hours. After washing, the bound C1q was detected using a murine monoclonal antibody against human C1q (Cat. No. A201, Quidel) and HRP conjugated anti-murine IgG (Cat. No. A0168, Sigma, Shanghai, China). As shown in FIG.
  • ADCC Classical antibody-dependent cellular cytotoxicity
  • CD16 FcyRIIIA
  • NK92MI/CD16V cells which were generated from NK92MI cells (Cat. No. CRL-2408, ATCC) by co-transducing expression plasmids containing CD16 (V158 allele) and FcRy genes, were used as effector cells, and PD-L1-expressing HEK293 cell line, HEK293/PD-L1, was used as target cells.
  • Cytotoxicity of NK92MI/CD16 cells exerted against HEK293/PD-L1 cells was determined by lactate dehydrogenase (LDH) release assay using the CytoTox 96 Non-Radioactive Cytotoxicity Assay kit (Promega, Madison, WI). Specific lysis was determined by the following equation.
  • Human IgG1 antibodies in general, induce significant complement-dependent cytotoxicity (CDC) via classical pathway. Whether the humanized anti-PD-L1 antibodies in selected IgG1 mutant formats (IgG1mc and IgG1mf) trigger CDC was evaluated using a PD-L1-expressing B cell line, Daudi/PD-L1, and fresh human serum from healthy donors, which contains all necessary components for CDC. Cell lysis by CDC was determined by Celltiter glo assay kits (Promega). In brief, Daudi/PD-L1 cells (2 ⁇ 10 4 cells/well) were incubated in serum-free RPMI1640 (Invitrogen) with anti-PD-L1 Abs (0.001-10 ⁇ g/mL) at 37° C.
  • CDC complement-dependent cytotoxicity
  • % CDC activity [(RFU test - RFU background) / (RFU at total cell lysis - RFU background)] ⁇ 100.
  • % CDC activity [(RFU test - RFU background) / (RFU at total cell lysis - RFU background)] ⁇ 100.
  • Rituximab induced robust CDC in CD20 + Daudi/PD-L1 cells.
  • both hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) showed no CDC.
  • Ab 333-4A2-IgG1wt with wild type IgG1 Fc demonstrated low but above base level of CDC activities especially at concentrations of above 0.3 ⁇ g/mL.
  • FACS binding analysis was performed as described in previous sections. Serial dilutions of antibodies were incubated with HEK293/PD-L1 cells and the bindings were detected using the Goat anti-human IgG F(ab′) 2 -Alexa Fluor 488 (Cat. No. 109-546-097, Jackson ImmunoResearch). Dose-dependent binding activities were observed for the selected mAbs to native PD-L1 protein expressed on surface of HEK293 cells.
  • hu333-4A2-IgGlmc, hu333-4B2-IgGlmc and hu333-4B2-IgG1mf showed similar dose-dependent binding activities to the HEK293/PD-L1 cells with EC 50 (effective concentration at 50% activity) around 0.1 ⁇ g/mL.
  • the functionalities of the purified anti-PD-L1 mAbs were assessed in the HuT78/PD-1 and HEK293/OS8/PD-L1 co-culture system as described in previous section. As shown in Table 19, the humanized 333 mAbs were potent antagonists of PD-L1/PD-1 signaling in this co-culture system, and induced increased IL-2 secretions.
  • hu333-4A2-IgG1mc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf showed similar potencies in this assay with very close EC 50 (0.075-0.087 ⁇ g/mL) and maximum induction of IL-2 levels (287-300 pg/mL).
  • the functionalities of the purified anti-PD-L1 mAbs were also assessed in the reversed signaling system in which HuT78/P3Z and HEK293/PD-L1 were co-cultured as described. Consistently, the humanized 333 mAbs were potent inhibitor of PD-L1/P3Z signaling in this co-culture system, and inhibited IL-2 secretions induced by PD-L1/P3Z engagement.
  • hu333-4A2-IgG1mc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf showed similar potencies in the assay, as shown by similar IC 50 s (0.037-0.045 ⁇ g/mL) and maximum inhibition levels (99%) (Table 19).
  • IL-2 release in HuT78/PD-1 IL-2 release induced by the Fabs in HuT78/PD-1 cells co-cultured with HEK293/OS8/PD-L1 cells.
  • ⁇ IL-2 release in HuT78/P3Z IL-2 release induced by the Fabs in HuT78/P3Z cells co-cultured with HEK293/PD-L1 cells
  • PBMCs peripheral blood mononuclear cells
  • T-cells 50-70%)
  • B-cells and NK cells 15-30%)
  • monocytes 2-10%).
  • Human PBMCs were isolated from healthy donors by density gradient centrifugation using ficoll lymphocyte separation medium (Histopaque-1077; Cat. No. 10771, Sigma) according to manufacturer’s instruction. The human blood collections were done followed the Internal Procedure of BeiGene. PBMCs were then stimulated with 40 ng/mL of anti-CD3 mAb OKT3 (Cat. No.
  • PBMCs (1 ⁇ 10 4 cells) were co-cultured with HEK293/OS8/PD-L1 cells (3 ⁇ 10 4 cells) in each well of 96-well flat-bottom plates. Indicated concentrations of anti-PD-L1 antibodies were added to the culture. After 15-18 hours of co-culture, culture supernatants were assayed for IFN-y level by ELISA using Ready-Set-Go! ELISA kits (Cat. No.
  • the potencies of hu333-4A2-IgG1mc and hu333-4B2-IgG1mf were comparable to the parental murine antibody mu333-IgG.
  • the base level of IFN-y without antibody treatment varied among different donors, the increase of IFN-y secretion in PBMCs treated by hu333-4A2-IgG1mc, hu333-4B2-IgG1mf and mu333-IgG was statistically significant in the range of 0.01 to 10 ⁇ g/mL of antibody treatment (about 5-8 fold induction at 10 ⁇ g/mL depending on the donor).
  • hu333-4A2-IgG1mc, hu333-4B2-IgGlmc and hu333-4B2-IgG1mf were potent antagonists blocking PD-L1/PD-1 interactions and downstream signaling in all the cell line and primary immune cell-based assays. They were very similar in their functional activities and potencies, as they were very similar in sequences (only minor difference in framework regions), shared identical binding epitope and had very similar binding affinities and specificity (see below section).
  • the binding specificity was studied for the mAbs hu333 (hu333-4A2-IgG1mc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf) using human, cynomolgus monkey (Macaca fascicularis) and mouse (Mus musculus) PD-L1 as target proteins.
  • the monkey PD-L1/His and murine PD-L1/His were expressed and purified in a similar way to the human PD-L1/His as described earlier.
  • Y1 was a reference functional anti-PD-L1 mAb which was synthesized according to a published patent (US 2010/0203056 A1) and fused to human IgG1mc variant.
  • the synthesized full length mAb was named as Y1-IgG1mc.
  • the ELISA assay was performed essentially in the same way as described in the previous section. Briefly, 200 ng of PD-L1 protein was coated on each well of Nunc MaxiSorp ELISA plate (Cat. No. 442404, Nunc, Thermo Fisher). After washing and blocking, indicated concentrations of anti-PD-L1 mAbs were added and incubated at room temperature for 1 hour. After washing, the bound anti-PD-L1 mAbs were detected using a HRP-conjugated goat anti-human Fc antibody (Cat. No. A0170, Sigma). As shown in FIG. 12 A , FIG. 12 B , and FIG.
  • hu333-4A2-IgG1mc (SEQ. NO. 30 and 32), hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) bound to human and monkey PD-L1 in a dose-dependent manner, but not to murine PD-L1, which was consistent with the fact that the original mu333 was generated from mouse immunized with human PD-L1/Fc and that human and monkey PD-1 shares high degree of sequence homology (96%). In contrast, Y1-IgG1mc bound to all PD-L1 proteins from human, monkey and mouse.
  • Hu333-4A2 (SEQ. NO. 21 and 23), hu333-4B2 (SEQ. NO. 22 and 23) and the reference antibody Y1 were constructed as human IgG1 Fab format in which the Vh and Vk were fused to the N-terminus of human IgG1-CH1 and constant region of kappa chain, respectively.
  • the IgG1-CH1 was fused to a C-terminal His6 tag to facilitate purification. Expression and purification of recombinant Fabs were performed as described in the previous section.
  • the SPR-determined binding affinities of anti-PD-L1 Fabs were listed in Table 20.
  • Hu333-4A2 and hu333-4B2 Fabs bind to human PD-L1 with higher affinities than Y1 Fab does, which was indicated by the faster k on , slower k off and much smaller K d value.
  • the hu333-4A2 and hu333-4B2 Fabs bind to monkey PD-L1 almost equally well as their binding to human PD-L1.
  • Y1 Fab binds to the monkey PD-L1with about 100-fold lower affinity than its binding to human PD-L1 (K d of 0.18 nM to human PD-L1 and K d of 16.2 nM to monkey PD-L1).
  • the wild-type PD-L1/His ( FIG. 1 ) was used as template for rolling-circle mutagenesis using Fast Mutagenesis kit (Cat. No. FM111, Transgen Biotech, Beijing, China). All mutants were sub-cloned in our pcDNA-based expression vectors, and verified by sequencing. The mutant and wild-type PD-L1/His proteins were expressed by transient transfection as described in previous sections.
  • the conditioned media (CM) were collected after 4 to 6 days of culture, and analyzed by Western blot to verify the PD-L1/His protein expression in terms of quality and quantity.
  • the supernatants (CM) after clearing cell debris, were directly used in ELISA analysis or Western blot for epitope-mapping.
  • ELISA assays using the wild-type and mutant PD-L1/His were performed to study the binding epitopes of the anti-PD-L1 mAbs.
  • several murine mAbs from us and one reference antibody Y1-IgG1 (adapted from US 2010/0203056 A1 and fused to human IgG1kappa constant regions) were included in the study.
  • Equal volume of CM containing wild type or mutant PD-L1/His was coated in 96-well plate for all mAbs in the same ELISA assay.
  • the ELISA results were normalized using the mean ELISA readings of wild type PD-L1 binding signals as the standard.
  • ELISA binding signals to a specific mutant PD-L1 were further normalized against the highest antibody binding read-out (set as 100%) to the specific mutant PD-L1 to even out expression variations between PD-L1 mutants.
  • a mAb’s ELISA binding signal for a specific PD-L1 mutant dropped below 50% relative to wild type PD-L1, it was defined that the binding function is significantly impaired due to the corresponding amino acid mutation.
  • a mAb’s ELISA binding signal for a specific mutant dropped below 25%, it was defined to be very significant.
  • amino acid residues R 113 in PD-L1 is critical for hu333-4B2-IgG1 binding to PD-L1, whose mutation significantly impaired the hu333-4B2-IgG1 binding to PD-L1.
  • the reference antibody Y1-IgG1 had distinctive binding epitopes. F 19 , I 54 , D 122 and Y 123 were all significant epitopes for its binding besides R 113 ( FIG. 13 B ). Different signatures of binding epitopes were also observed in other anti-PD-L1 mAbs from our study. The data from Western blot for both hu333-4B2-IgG1 and Y1-IgG1 antibodies confirmed these results although the antigen proteins were denatured.
  • the mutation D 26 A Apart from the above key binding epitope mutations, we also made the mutation D 26 A. ELISA and Western blot results showed the mutation D 26 A in PD-L1 significantly inhibited the binding activities of all functional anti-PD-L1 mAbs including mAbs mu333, hu333-4B2-IgG1 and Y1-IgG1, but not inhibited the binding of non-functional antibodies, such as mu260 ( FIG. 13 C ).
  • hu333-4B2 binds to two key amino acid residues (epitopes) of human PD-L1, D 26 and R 113 ; in contrast, mAb Y1 binds to at least six amino acid residues.
  • anti-PD-L1 mAbs are capable of binding to different epitope signatures through molecular recognition, which might have profound impact on binding affinity, binding specificity and functional activity, e.g. hu333-4A2 and hu333-4B2 can only bind to human PD-L1 ( FIG. 12 A ), but not to mouse PD-L1 ( FIG. 12 C ); in contrast, Y1 binds to both human and mouse PD-L1 ( FIG. 12 A and FIG. 12 C ) although human and mouse PD-L1 have 26% sequence divergence.
  • ELISA study was performed using 96-well Nunc Maxisorp ELISA plates coated with 5% human serum (from healthy donors) and various concentrations of PD-L1/His antigen as indicated in FIG. 14 A , x-axis. Same amount of murine PD-L1 Abs or a chimeric Y1 (named as Y1-muIgG1), which was made of Y1 variable domains fused to murine IgG1kappa constant regions) were added in the ELISA reaction, and the bindings were detected with anti-mouse Fc-HRP (Cat. No. A9309, Sigma). The PBS buffer without any antibody was included as negative control ( FIG. 14 A and FIG. 14 B ).
  • the baseline O.D. reading (the reading at very low antigen concentration) for mu333 was almost identical to the negative control, while the baseline O.D. reading for Y1-muIgG1 is 4 folds higher than the negative control (PBS), indicating a differential property between mu333 and YI-mIgG1 in binding selectivity.
  • the humanized mAb hu333-4B2-IgG1mc and hu333-4B2-IgG1mf were also assessed for non-specific binding in similar assay method using fetal bovine serum (FBS) instead of human serum. Reminiscent to its parental mouse hybridoma mAb, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf had no binding to FBS.
  • FBS fetal bovine serum
  • mice Ten to twelve week-old female Balb/c nude mice (18-25 g) were used to study the pharmacokinetics of the humanized mAb hu333-4B2-IgG1mf (SEQ. NO. 32 and 33). Mice were dosed with 10 mg/kg of mAb hu333-4B2-IgG1mf either as a single intravenous (i.v.) or subcutaneous (s.c.) injection. Intravenous injections were administered via a tail vein, and subcutaneous injections were administered in the flank. In each injection group, mice were separated into different subgroups and in each subgroup blood sera were collected at certain time points. For i.v.
  • serum was harvested 2 days predose, and postdose at 15 min, 30 min, 60 min, 90 min, 6 h, 24 h and once on days 2, 3, 4, 5, 7, 10, 14, 21 and 28.
  • serum was harvested 2 days predose, and postdose at 1.5 h, 6 h, 24 h and once on days 2, 3, 4, 5, 7, 10, 14, 21, and 28.
  • Serum level of hu333-4B2-IgG1mf was determined by ELISA using human PD-L1/His protein. Briefly, Nunc MaxiSorp ELISA plates (Cat. No. 442404, Nunc, Thermo Fisher) were coated overnight at 4° C. with 100 ⁇ L per well of 3 ⁇ g/mL human PD-L1/His protein. Plates were blocked with 3% bovine serum albumin, 0.05% Tween 20 in PBS (blocking buffer) at room temperature for 1 hour. After washing, serially diluted serum samples and purified hu333-4B2-IgG1mf standards were added and incubated at room temperature for 1 hour.
  • hu333-4B2-IgG1mf was detected using a HRP-conjugated goat anti-human Fc antibody (Cat. No. A0170, Sigma) and color developed using TMB substrate (Cat. No. T0440, Sigma).
  • a standard curve was fit using nonlinear regression and the serum concentrations of hu333-4B2-IgG1mf were deduced from the standard curve and dilution factors.
  • the serum concentrations of hu333-4B2-IgG1mf versus time data were analyzed using the non-compartment model for the i.v. and s.c. doses (WinNonlin, Pharsight).
  • the clearance, volume of distribution, half-lives, mean residence time and bioavailability were deduced from WinNonlin data fitting.
  • mice The pharmacokinetics of hu333-4B2-IgG1mf in mice was summarized in Table 22.
  • hu333-4B2-IgG1mf SEQ. NO. 32 and 33 concentrations were cleared from the serum in a biphasic manner. The terminal half-life was about 10-14 days. After an i.v. dose of 10 mg/kg, the clearance was 7.9 mL/day/kg in mice.
  • peak concentrations of hu333-4B2-IgG1mf in the serum as approximately 30-50% of that noted after i.v. administration of the same dose. Comparison of the AUC after the 10 mg/kg s.c. and i.v.
  • the pharmacokinetics of hu333-4B2-IgGlmc was studied in cynomolgus monkeys.
  • humanized 333 bound to human and monkey PD-L1 with almost identical affinities the pharmacokinetic profile in cynomolgus monkeys should be very informative and scalable to predict the pharmacokinetic profile in humans.
  • the drug administrations and blood serum collections were done at 3D BioOptima Co. Ltd (Suzhou, China) following 3D BioOptima’s Animal Care and Use Procedure.
  • ELISA based bioanalyses and pharmacokinetic analyses were performed essentially as described above.
  • the averaged serum concentration from 2 monkeys was used for fitting except for the time points of 22 and 30 days post dose, where the data from only one monkey were used, as another monkey showed accelerated clearance and undetectable hu333-4B2-IgG1mc serum levels, presumably due to a monkey anti-drug immune response, at these two time points.
  • the serum concentration of hu333-4B2-IgG1mc versus time data were analyzed using the non-compartment model for the i.v. dose.
  • hu333-4B2-IgG1mc The pharmacokinetics of hu333-4B2-IgG1mc in cynomolgus monkeys was summarized in Table 23.
  • hu333-4B2-IgG1mc concentrations were cleared from the sera in a biphasic manner.
  • the terminal half-life was about 9 days.
  • i.v. dose of 10 mg/kg the clearance was 6.4 mL/day/kg in cynomolgus monkeys.
  • peak concentration of hu333-4B2-IgG1mc was 283 ⁇ g/mL at 5 min post dose.
  • PK parameters indicated that hu333-4B2-IgG1mc had normal pharmacokinetic profile in cynomolgus monkeys, which predicted normal pharmacokinetic behavior in humans (Deng et. al. 2011 mAbs 3:61-66).
  • mice Female NOD/SCID mice (6-7 weeks) were pre-treated with cyclophosphamide.
  • Human peripheral blood mononuclear cells PBMCs were isolated from blood of healthy human volunteer, mixed with A431 epidermoid carcinoma cells (Cat. No. CRL-1555 ATCC) and matrigel, and injected subcutaneously into the right front flank of the animals.
  • mice were randomly assigned into 3 groups with 8 mice per group. Mice were treated twice weekly (BIW i.p.) with vehicle (PBS) or 10 mg/kg hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) for 4 weeks. Individual animal body weight and tumor volume were recorded twice weekly, with mice being monitored daily for clinical signs of toxicity for the duration of the study. Tumor volumes were calculated using the following formula: [D ⁇ (d 2 )]/2, in which D represents the large diameter of the tumor, and d represents the small diameter.
  • the A431 tumor growth was slower compared to that in the PBS-treated group.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Provided are antibodies that specifically bind to Programmed Death-1 (PD1, Pdcd-1, or CD279) ligand (PD-L1) and inhibit PD-L1-mediated cellular signaling and activities in immune cells, antibodies binding to a set of amino acid residues required for its ligand binding, and uses of these antibodies to treat or diagnose cancer, infectious diseases or other pathological disorders modulated by PD-L1-mediated functions.

Description

    INTRODUCTION
  • PD-L1 was initially cloned as a member (known as B7-H1) of B7 protein family (Dong et al., 1999 Nature Med 5:1365). It binds to Programmed Death-1 (PD-1) receptor and activates negative regulatory signaling pathway, inhibiting T-cell proliferation and activities (Freeman et. al. 2000 J Exp Med 192:1027). Therefore, it was also termed as PD-1 ligand 1 (PD-L1 or CD274). To date, two sequence-related ligands, PD-L1 (B7-H1) and PD-L2 (B7-DC), were identified that interact with PD-1, induce negative signal transduction and inhibit TCR and CD28 mediated T-cell activation, cell proliferation and secretion of growth factors and cytokines such as IL-2 and IFN-y (Riley et. al. 2009 Immunol Rev 229:114).
  • Human PD-L1 gene encodes a full-length protein of 290 amino acid residues (NCBI accession NP_054862.1) with a leader peptide, which is removed after PD-L1 is expressed on cell surface as a mature protein. The calculated molecular weight of the full length PD-L1 is 33 kD. However, the observed molecular weight is around 50 kD due to glycosylation, based on Western blot data from ours and others.
  • PD-L1 was found constitutively expressed in human heart, lung, thymus and vascular endothelial cells, and expressed at a low level in many other human tissues and cell types including antigen presenting cells, peripheral blood monocytes and other immune cells (Freeman et. al. 2000 J Exp Med 192:1027; Eppihimer et. al. 2002 Microcirculation 9:133). When stimulated by IFN-y, IL-12 and type I interferons, many of those cell types were found expressing increased level of PD-L1 (Bald et. al. 2014 Cancer Discov 4:674-687; Planes et. al. 2014 J Virol 88:6672-6689).
  • Aberrant up-regulation of PD-L1 expression in tumor cells were reported in varieties of cancers involved in different types of tissues and organs such as lung (Konishi et. al. 2004 Clin Cancer Res 10:5094), liver (Shi et. al. 2008 Int J Cancer 128:887; Gao et. al., 2009 Clin Cancer Res 15:971), stomach (Wu et. al. 2006 Acta Histochem 108:19), kidney (Thompson et. al. 2004 Proc Natl Acad Sci 101:17174; Thompson et. al. 2007 Clin Cancer Res 13:1757), breast (Ghebeh et. al. 2006 Neoplasia 8:190), ovary (Hamanishi et. al. 2007 Proc Natl Acad Sci 104:3360), pancreas (Nomi et. al. 2007 Clin Cancer Res 13:2151), melanocytes (Hino et. al. 2010 Cancer 116:1757) and esophagus (Ohigashi et. al. 2005 Clin Cancer Res 11:2947). More frequently, the increased expression of PD-L1 in those cancers is associated to poor prognosis in patient survival outcome.
  • Blockade of PD-L1 engaging PD-1 receptor by B7-H1Ig or anti-PD-L1 antibody stimulated T-cell proliferation and functional activities (Dong et. al. 1999 Nature Med 5:1365; Freeman et. al. 2000 J Exp Med 192:1027; Tamura et. al. 2001 Blood 97:1809; Iwai et. al. 2002 PNAS 99:12293), enhanced immune responses against tumor growth and viral infection (Iwai et. al. 2002 PNAS 99:12293). Those observations suggested that inhibition of PD-L1/PD-1 signaling may activate immune responses not only against cancer cell growth, but also against viral infection and expansion in human. The prevalent hepatocyte infection viruses, HBV and HCV, induce overexpression of PD-1 ligands in hepatocytes and activate PD-1 signaling in T-effecter cells, resulting T-cell exhaustion and tolerance to the viral infection (Boni et. al. 2007 J Virol 81:4215; Golden-Mason et. al. 2008 J Immunol 180;3637). Likewise, the popular HIV infection frequently evades human immune system by similar mechanism. Therapeutic modulation of PD-L1 induced signaling by antagonist molecules may revert immune cells from tolerance, and reactivated to eradicate cancer and chronic viral infection (Blank et. al. 2005 Cancer Immunol Immunother 54:307; Okazaki et. al. 2007 Int Immunol 19:813).
  • Recently, it is discovered that PD-L1 also specifically interacts to B7-1 (another B7 family member, also known as CD80) besides binding to PD-1 (Butte et. al. 2007 Immunity 27:111). Initial evidences indicated that interaction of PD-L1 to CD80 exerts negative regulation to T-cell function and activity, and blockage of PD-L1 and CD80 interaction in mice elicited stronger immune responses to OVA antigen challenge (Park et. al. 2010 Blood 116:1291). Therefore, simultaneously blocking PD-L1 binding to PD-1 and CD80 may exert additive or synergistic effect against cancer and viral infection.
  • SUMMARY OF THE INVENTION
  • The invention provides methods and compositions for immune-activation by inhibiting PD-L1-mediated signaling and function. In one aspect, the invention provides an antibody antigen binding domain which specifically binds human PD-L1, and comprises a complementarity determining region (CDR) sequence described herein. The CDRs are amenable to recombination into heavy chain variable region (Vh) and light chain variable regions (Vk) which comprise (CDR-H1, CDR-H2 and CDR-H3) and (CDR-L1, CDR-L2 and CDR-L3) sequences, respectively and retain PD-L1-specific binding and/or functionality.
  • In particular embodiments, the domain comprises CDR1, CDR2 and CDR3, in a combination selected from (a)-(r) as follows, wherein the antibody (Ab), heavy chain (HC) or light chain (LC) and CDR nomenclature system (Kabat, IMGT or composite) from which the CDR combinations derive are shown in the first column, and residues in bold text are Kabat system, and residues underlined are IMGT system:
  • Ab, Chain System CDR1 CDR2 CDR3
    mu333/ hu333-1A vh GFSLTSYG VH VIWAGGSTNYNSALMS AKPYGNSAMDY
    a Kabat SEQ ID NO:9, res. 6-10 SEQ ID NO:10 SEQ ID NO:11, res. 3-11
    b IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:10, res. 2-8 SEQ ID NO:11
    c Comp. SEQ ID NO:9 SEQ ID NO:10 SEQ ID NO:11
    hu333-2B/3A2 vh GFSLTSYG VH VIWAGGSTNYVDSVKG AKPYGNSAMDY
    d Kabat SEQ ID NO:9, res. 6-10 SEQ ID NO:24 SEQ ID NO:11, res. 3-11
    e IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:24, res. 2-8 SEQ ID NO:11
    f Comp. SEQ ID NO:9 SEQ ID NO:24 SEQ ID NO:11
    hu333-3C2/3H2 vh GFSLTSYG VH VIWAGGSTNYADSVKG AKPYGNSAMDY
    g Kabat SEQ ID NO:9, res. 6-10 SEQ ID NO:25 SEQ ID NO:11, res. 3-11
    h IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:25, res. 2-8 SEQ ID NO:11
    i Comp. SEQ ID NO:9 SEQ ID NO:25 SEQ ID NO:11
    hu333-4A2 vh GFSLTSYG VH VIWAGGSTNYVDSVKG AKPYGTSAMDY
    j Kabat SEQ ID NO:9, res. 6-10 SEQ ID NO:24 SEQ ID NO:26, res. 3-11
    k IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:24, res. 2-8 SEQ ID NO:26
    l Comp. SEQ ID NO:9 SEQ ID NO:24 SEQ ID NO:26
    hu333-4B2 vh GFSLTSYG VH VIWAGGSTNYADSVKG AKPYGTSAMDY
    m Kabat SEQ ID NO:9, res. 6-10 SEQ ID NO:25 SEQ ID NO:26, res. 3-11
    n IMGT SEQ ID NO:9, res. 1-8 SEQ ID NO:25, res. 2-8 SEQ ID NO:26
    o Comp. SEQ ID NO:9 SEQ ID NO:25 SEQ ID NO:26
    mu333/ hu333′s vk KASQDVGIVVA WASIRHT QQYSNYPLYT
    p Kabat SEQ ID NO:12 SEQ ID NO:13 SEQ ID NO:14
    q IMGT SEQ ID NO:12, res. 4-9 SEQ ID NO:13, res. 1-3 SEQ ID NO:14
    r Comp. SEQ ID NO:12 SEQ ID NO:13 SEQ ID NO:14
  • In particular embodiments, the domain comprises a heavy chain variable region (Vh) comprising a CDR1, CDR2 and CDR3 combination selected from (a)-(o), and a light chain variable region (Vk) comprising a CDR1, CDR2 and CDR3 combination selected from (p)-(r).
  • In particular embodiments, the domain comprises CDR1, CDR2 and CDR3, in a combination selected from (c), (f), (i), (1), (o) and (r), as follows:
  • Ab, Chain CDR1 CDR2 CDR3
    mu333 vh (c) SEQ ID NO:9 SEQ ID NO:10 SEQ ID NO:11
    hu333 2B/3A2 vh (f) SEQ ID NO:9 SEQ ID NO:24 SEQ ID NO:11
    hu333 3C2/3H2 vh (i) SEQ ID NO:9 SEQ ID NO:25 SEQ ID NO:11
    hu333 4A2 vh (l) SEQ ID NO:9 SEQ ID NO:24 SEQ ID NO:26
    hu333 4B2 vh (o) SEQ ID NO:9 SEQ ID NO:25 SEQ ID NO:26
    mu333 vk (r) SEQ ID NO:12 SEQ ID NO:13 SEQ ID NO:14
  • In particular embodiments, the domain comprises a heavy chain variable region (Vh) or a light chain variable region (Vk), comprising a sequence that is:
  • mu333 vh (SEQ ID NO:6);
    mu333 vk (SEQ ID NO:8);
    hu333-1A vh (SEQ ID NO:15),
    hu333-1A vk (SEQ ID NO:16);
    hu333-2B vh (SEQ ID NO:17);
    hu333-3A2 vh (SEQ ID NO:18);
    hu333-3C2 vh (SEQ ID NO:19);
    hu333-3H2 vh (SEQ ID NO:20);
    hu333-4A2 vh (SEQ ID NO:21);
    hu333-4B2 vh (SEQ ID NO:22); or
    hu333-4B2 vk (SEQ ID NO:23).
  • In particular embodiments, the domain comprises a heavy chain variable region (Vh) and a light chain variable region (Vk) comprising a sequence that is:
  • mu333 vh and vk (SEQ ID NOS:6 and 8);
    hu333-1A vh and vk (SEQ ID NOS:15 and 16);
    hu333-2B vh and vk (SEQ ID NOS:17 and 16);
    hu333-3A2 vh and vk (SEQ ID NOS:18 and 23);
    hu333-3C2 vh and vk (SEQ ID NOS:19 and 23);
    hu333-3H2 vh and vk (SEQ ID NOS:20 and 23);
    hu333-4A2 vh and vk (SEQ ID NOS:21 and 23); or
    hu333-4B2 vh and vk (SEQ ID NOS:22 and 23).
  • In particular embodiments, the domain comprises comprising a heavy chain variable region (Vh) or a light chain variable region (Vk) comprising:
  • hu333-4B2 vh (SEQ ID NO:22); or
    hu333-4B2 vk (SEQ ID NO:23).
  • In particular embodiments, the domain comprises comprising a heavy chain variable region (Vh) and a light chain variable region (Vk) comprising:
  • hu333-4A2 vh and vk (SEQ ID NOS:21 and 23); or
    hu333-4B2 vh and vk (SEQ ID NOS:22 and 23)
  • In particular embodiments, the domain specifically binds PD-L1 residues: D26 and R113.
  • The invention also provides antibodies, particularly monoclonal antibodies, and F(ab) or F(ab)2 comprising a subject PD-L1 binding domain.
  • The invention also provides novel polynucleotides such as cDNAs and expression vectors, encoding a subject PD-L1 antigen binding domain, and cells comprising such polynucleotides, and non-human animals comprising such cells. The polynucleotides may be operably linked to a heterologous transcription regulating sequence for expression, and may be incorporated into such vectors, cells, etc.
  • The invention provides methods of using the subject domains by administering the domain to a person determined to have cancer or a viral infection or to otherwise be in need of PD-L1 antagonism.
  • The compositions of the invention are useful for the treatment of cancer, neurodegenerative and infectious, particularly viral, diseases and other conditions in which inappropriate or detrimental expression of the human PD-1 and/or is a component of the etiology or pathology of the condition. Hence, the invention provides methods for treating cancer or inhibiting tumor progression in a subject in need thereof with a subject anti-PD-L1 protein, and the humanized anti-PD-1 mAbs are used as therapeutic agents to treat human diseases that are involved in suppression of immune cells by PD-1 mediated intracellular signaling, leading to disease progression, particularly cancers and viral infections.
  • The invention further provides the use of subject polynucleotides for the manufacture of a medicament for treating cancer or inhibiting tumor progression in a subject.
  • The invention includes all combinations of the recited particular embodiments. Further embodiments and the full scope of applicability of the invention will become apparent from the detailed description given hereinafter. However, it should be understood that the detailed description and specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. All publications, patents, and patent applications cited herein, including citations therein, are hereby incorporated by reference in their entirety for all purposes.
  • BREIF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 . Schematic presentation of full length PD-L1 (top), PD-L1/Fc (middle) and PD-L1/His (bottom). ECD: extracellular domain. L: linker. Fc: y4Fc fragment from human IgG4. H: His tag. N: N-terminus. C: C-terminus.
  • FIG. 2A and FIG. 2B. Dose-dependent binding to the purified human PD-L1/His in ELISA (FIG. 2A) or to cell surface expressed PD-L1 in FACS (FIG. 2B) by murine mAbs. The murine mAbs and mouse IgG as negative control were indicated at top-left corner of each figure.
  • FIG. 2A. ELISA plate was coated by PD-L1/His at 100 ng per well. The binding signal strength in ELISA was indicated by direct OD450 readings in y-axis. Concentrations of mAbs or mouse IgG were indicated by x-axis.
  • FIG. 2B. HEK293/PD-L1 cells were stained with a serial dilution of murine anti-PD-L1 mAbs or control mouse IgG. The binding signal strength was indicated by MFI (mean fluorescence intensity) in FACS analyses. Concentrations of mAbs or mouse IgG were indicated by x-axis.
  • FIG. 3 . Dose-dependent response curves of murine anti-PD-L1 mAb-induced IL-2 secretion in HuT78/PD-1 cells after co-culture with HEK293/OS8/PD-L1 cells. Baseline: Average IL-2 release induced by mouse IgG (mIgG) at all tested concentrations. Each represents average of duplicate data points. Top line: Highest IL-2 release based on regression calculation by Prizm.
  • FIG. 4 . Dose-dependent response curves of murine anti-PD-L1 mAb-inhibited IL-2 secretion in HuT78/P3Z cells after co-culture with HEK293/PD-L1 cells. Engagement of PD-L1 and P3Z chimeric receptor leads to activation of P3Z chimeric receptor and IL-2 secretion.
  • FIG. 5 . Dose-dependent competition curves of murine anti-PD-L1 mAbs against biotin-conjugated PD-1/Fc. Fixed amount of biotin-PD-1-ECD/Fc was mixed with increasing concentrations of anti-PD-L1 mAbs indicated in x-axis. Mean fluorescence intensity (MFI) analyzed by FACS was showed in y-axis. Mouse Gig (mug) was used as a negative control.
  • FIG. 6 . Dose-dependent competition curves of murine anti-PD-L1 mAbs against biotin-conjugated CD80/Fc. Fixed amount of biotin-CD80-ECD/Fc was mixed with increasing amount of anti-PD-L1 mAbs indicated in x-aixs. Mean fluorescence intensity (MFI) analyzed by FACS was showed in y-axis. Mouse IgG (muIgG) was used as a negative control.
  • FIG. 7A and FIG. 7B. IFN-y secretion induced by humanized anti-PD-L1 Fabs in primary human PBMCs from different healthy donors. (Donor 3 (FIG. 7A) and Donor 17 (FIG. 7B)). PBMCs were co-cultured with HEK293/OS8/PD-L1 cells for overnight. IFN-y in conditioned medium was assayed by ELISA. BSA was used as a negative control.
  • FIG. 8 . C1q bindings of humanized anti-PD-L1 mAbs in wild type (IgG1wt) or mutant human IgG1 formates (IgG1mc and IgG1mf). Fifty microliters of a serial dilution (x-axis) of humanized anti-PD-L1 mAb were coated on MaxiSorp ELISA plate. Human C1q bindings (y-axis) were assessed by ELISA OD450 readings using a specific monoclonal antibody to human C1q.
  • FIG. 9 . Antibody-dependent cell-cytotoxicity (ADCC) induced by humanized anti-PD-L1 mAbs in wild type (IgG1wt) or mutant human IgG1 formats (IgG1mc and IgG1mf). Human NK92MI cells transfected with FcyRIIIA were used as effector cells and HEK293/PD-L1 cells were used as target cells. Percentage of cytotoxicity (y-axis) was calculated based on lactate dehydrogenase (LDH) release assay as described in Example 5.
  • FIG. 10 . Complement-dependent cytotoxicity (CDC) activities of humanized anti-PD-L1 mAbs in wild type (IgG1wt) or mutant human IgG1 formats (IgG1mc and IgG1mf). Daudi/PD-L1 cells were used as target cells and human sera from healthy donors were used as the source of complement components. Rituximab (Roche) was used as positive control in classical CDC assay. Percentage of CDC (y-axis) was calculated based on cell-titer glow assay as described in Example 5.
  • FIG. 11A and FIG. 11B. IFN-y secretion induced by humanized anti-PD-L1 mAbs in primary human PBMCs from different healthy donors.. (Donor 3 (FIG. 11A) and Donor 17 (FIG. 11B)). PBMCs were co-cultured with HEK293/OS8/PD-L1 cells for overnight. IFN-y in conditioned medium was assayed by ELISA. Human IgG served as a negative control.
  • FIG. 12A, FIG. 12B, and FIG. 12C. Dose-dependent bindings of anti-PD-L1 mAbs to the purified human PD-L1/His (FIG. 12A), cynomolgus monkey PD-L1/His (FIG. 12B), and mouse PD-L1/His (FIG. 12C) in ELISA. MaxiSorp ELISA plates were coated with 50 microliters of human, monkey and mouse PD-L1/His, respectively. Concentrations of anti-PD-L1 mAbs were indicated by x-axis. The binding signal strength was indicated by direct OD450 readings (y-axis).
  • FIG. 13A, FIG. 13B, and FIG. 13C. Mapping the binding epitopes of anti-PD-L1 mAbs by ELISA (upper panel) and Western Blot (lower panel). FIG. 13A: Binding activities to mutant PD-L1 by hu333-4B2-IgG1. FIG. 13B: Binding activities to mutant PD-L1 by Y1-IgG1. FIG. 13C: Binding activities to D26A mutant PD-L1 by anti-PD-L1 mAbs. Conditioned media containing wild type or mutant PD-L1/His proteins were used to assess binding activity by ELISA and Western Blot. ** indicates the PD-L1 mutant to which the mAb binding activity reduced to 25-50% of that to wild type PD-L1. *** indicates the PD-L1 mutant to which the mAb binding activity reduced below 25% of that to wild type PD-L1.
  • FIG. 14A and FIG. 14B. Binding assays of anti-PD-L1 mAbs to a mixture of human serum protein and PD-L1 antigen by ELISA. FIG. 14A: Dose-dependent reaction curves of murine mAb or murine chimeric mAb binding to a mixture of human serum and PD-L1/His protein. Serial dilutions of PD-L1/His protein in PBS were coated onto 96-well MaxiSorp ELISA plate as indicated, and human serum pool (from three healthy donors) were added at a fixed final concentration of 5%. Three ug/mL indicated mAbs were added to each well and incubated for one hour at room temperature. FIG. 14B: Histogram showed the average OD450 readings of three data points in left side (coated mainly with human serum proteins and with very little PD-L1/His) of curves for each mAb and the negative control (without mAb).
  • FIG. 15 . Mean tumor growth curves upon treatment with hu333-4B2-IgG1mf or vehicle. NOD/SCID mice implanted with human cancer cells A431 and PBMCs from healthy donors were treated with hu333-4B2-IgG1mf at the dose regimen of 10 mg/kg, twice/week.
  • DESCRIPTION OF PARTICULAR EMBODIMENTS OF THE INVENTION
  • PD-L1 initiates inhibitory signaling in immune cells when engaged by its ligands, PD-L1 or PD-L2. In the cases of cancer outgrowth and viral infection, the activation of PD-1 signaling promotes immune tolerance, leading to the cancers or virus-infected cells escaping from immune surveillance and cancer metastasis or viral load increase. Inhibition of PD-L1 mediated cellular signaling by therapeutic agents can activate immune cells including T-cells, B-cells and NK cells, and therefore enhance immune cell functions inhibiting cancer cell growth or viral infection, and restore immune surveillance and immune memory function to treat such human diseases.
  • The invention provides antibodies whose functions are antagonistic to PD-L1-induced cellular signaling in immune cells. Murine anti-PD-L1 antibodies were humanized to a high degree of similarity to human antibodies in the framework regions. The full antibodies made in the modified human IgG variant format have a unique set of features in the aspects of effector functions and physicochemical properties. The disclosed anti-PD-L1 antibodies are suitable for therapeutic uses in cancer treatment, controlling viral infections and other human diseases that are mechanistically involved in exacerbated immune tolerance.
  • Unless the context indicates otherwise, the term “antibody” is used in the broadest sense and specifically covers antibodies (including full length monoclonal antibodies) and antibody fragments so long as they recognize PD-L1. An antibody molecule is usually monospecific, but may also be described as idiospecific, heterospecific, or polyspecific. Antibody molecules bind by means of specific binding sites to specific antigenic determinants or epitopes on antigens. “Antibody fragments” comprise a portion of a full length antibody, generally the antigen binding or variable region thereof. Examples of antibody fragments include Fab, Fab′, F(ab′).sub.2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Natural and engineered antibody structures are well known in the art, e.g. Strohl et al., Therapeutic antibody engineering: Current and future advances driving the strongest growth area in the pharmaceutical industry, Woodhead Publishing Series in Biomedicine No. 11, October 2012; Holliger et al. Nature Biotechnol 23, 1126 - 1136 (2005); Chames et al. Br J Pharmacol. 2009 May; 157(2): 220-233.
  • Monoclonal antibodies (MAbs) may be obtained by methods known to those skilled in the art. See, for example Kohler et al (1975); U.S. Pat. No. 4,376,110; Ausubel et al (1987-1999); Harlow et al (1988); and Colligan et al (1993). The mAbs of the invention may be of any immunoglobulin class including IgG, IgM, IgE, IgA, and any subclass thereof. A hybridoma producing a mAb may be cultivated in vitro or in vivo. High titers of mAbs can be obtained in in vivo production where cells from the individual hybridomas are injected intraperitoneally into mice, such as pristine-primed Balb/c mice to produce ascites fluid containing high concentrations of the desired mAbs. MAbs of isotype IgM or IgG may be purified from such ascites fluids, or from culture supernatants, using column chromatography methods well known to those of skill in the art.
  • An “isolated polynucleotide” refers to a polynucleotide segment or fragment which has been separated from sequences which flank it in a naturally occurring state, e.g., a DNA fragment which has been removed from the sequences which are normally adjacent to the fragment, e.g., the sequences adjacent to the fragment in a genome in which it naturally occurs. The term therefore includes, for example, a recombinant DNA which is incorporated into a vector, into an autonomously replicating plasmid or virus, or into the genomic DNA of a prokaryote or eukaryote, or which exists as a separate molecule (e.g., as a cDNA or a genomic or cDNA fragment produced by PCR or restriction enzyme digestion) independent of other sequences. It also includes a recombinant DNA, which is part of a hybrid gene encoding additional polypeptide sequence.
  • A “construct” means any recombinant polynucleotide molecule such as a plasmid, cosmid, virus, autonomously replicating polynucleotide molecule, phage, or linear or circular single-stranded or double-stranded DNA or RNA polynucleotide molecule, derived from any source, capable of genomic integration or autonomous replication, comprising a polynucleotide molecule where one or more polynucleotide molecule has been linked in a functionally operative manner, i.e. operably linked. A recombinant construct will typically comprise the polynucleotides of the invention operably linked to transcriptional initiation regulatory sequences that will direct the transcription of the polynucleotide in the intended host cell. Both heterologous and non-heterologous (i.e., endogenous) promoters can be employed to direct expression of the nucleic acids of the invention.
  • A “vector” refers any recombinant polynucleotide construct that may be used for the purpose of transformation, i.e. the introduction of heterologous DNA into a host cell. One type of vector is a “plasmid”, which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “expression vectors”.
  • An “expression vector” as used herein refers to a nucleic acid molecule capable of replication and expressing a gene of interest when transformed, transfected or transduced into a host cell. The expression vectors comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desired, provide amplification within the host. The expression vector further comprises a promoter to drive the expression of the polypeptide within the cells. Suitable expression vectors may be plasmids derived, for example, from pBR322 or various pUC plasmids, which are commercially available. Other expression vectors may be derived from bacteriophage, phagemid, or cosmid expression vectors.
  • Description of Sequence Listing
  • SEQ ID No.1 PD-L1 full length cDNA SEQ ID No.18 hu333-3A2 pro-vh
    SEQ ID No.2 PD-L1 full length PRT SEQ ID No.19 hu333-3C2 pro-vh
    SEQ ID No.3 PD-L1 ECD cDNA SEQ ID No.20 hu333-3H2 pro-vh
    SEQ ID No.4 PD-L1 ECD PRT SEQ ID No.21 hu333-4A2 pro-vh
    SEQ ID No.5 mu333 cDNA-Vh SEQ ID No.22 hu333-4B2 pro-vh
    SEQ ID No.6 mu333 pro-Vh SEQ ID No.23 hu333-4B2 pro-vk
    SEQ ID No.7 mu333 cDNA-Vk SEQ ID No.24 hu333-4A2 H-CDR2
    SEQ ID No.8 mu333 pro-Vk SEQ ID No.25 hu333-4B2 H-CDR2
    SEQ ID No.9 mu333 H-CDR1 SEQ ID No.26 hu333-4B2 H-CDR3
    SEQ ID No.10 mu333 H-CDR2 SEQ ID No.27 huIgG1wt pro
    SEQ ID No.11 mu333 H-CDR3 SEQ ID No.28 huIgG1mc pro
    SEQ ID No.12 mu333 L-CDR1 SEQ ID No.29 huIgG1mf pro
    SEQ ID No.13 mu333 L-CDR2 SEQ ID No.30 hu333-4A2-IgG1mc HC pro
    SEQ ID No14 mu333 L-CDR3 SEQ ID No.31 hu333-4B2-IgG1mc HC pro
    SEQ ID No.15 hu333-1A pro-vh SEQ ID No.32 hu333-4B2-IgG1mf LC pro
    SEQ ID No.16 hu333-1A pro-vk SEQ ID No.33 hu333-4B2-IgG1mf HC pro
    SEQ ID No.17 hu333-2B pro-vh
  • EXAMPLES Example 1. Generation of Anti-PD-L1 Monoclonal Antibody
  • Murine anti-human PD-L1 monoclonal antibodies (mAbs) were generated using hybridoma fusion technology (Kohler and Milstein 1975 Nature 256:495-497; Mechetner 2007 Methods Mol Biol 378:1-13) with modifications. MAbs with high binding activities in enzyme-linked immunosorbent assay (ELISA) and fluorescence-activated cell sorting (FACS) assay were selected for further characterization in cell-based functional assays.
  • PD-L1 and CD80 Recombinant Proteins
  • The full length human PD-L1 cDNA was synthesized by GeneScript (Nanjing, China) based on published sequence (NCBI reference sequence NM_014143.3) (SEQ. NO. 1 and 2). The extracellular domain consisting of amino acids (AA) 1-239 of human PD-L1 (SEQ. NO. 3 and 4) was PCR-amplified and subcloned into pcDNA3.1-based expression vector (Invitrogen, Carlsbad, CA, USA) with C-terminus fused with either a Fc region of human IgG4 or a His tag, which resulted in two recombinant PD-L1 fusion constructs, PD-L1-ECD/Fc and PD-L1-ECD/His (abbreviated as PD-L1/Fc and PD-L1/His). The schematic diagram of the PD-L1 fusion proteins were shown in FIG. 1 . The recombinant PD-L1 fusion proteins were expressed in 293-F cells (Cat. No. R79007, Invitrogen) by transient transfection according to manufacturer’s instruction (Invitrogen). The conditioned media containing the secreted recombinant proteins was collected and cleared by centrifugation at 15000 g for 30 minutes. PD-L1/Fc was purified using a Protein G Sepharose Fast Flow column (Cat. No. 17061805, GE Life Sciences, Shanghai, China). PD-L1/His was purified through Ni-Sepharose Fast Flow affinity chromatography (Cat. No. 17531801, GE Life Sciences), followed by size exclusion chromatography using a HiLoad 16/60 Superdex 200 column (Cat. No. 17106901, GE Life Sciences). Both PD-L1/Fc and PD-L1/His proteins were dialyzed against phosphate buffered saline (PBS) and stored in -80° C. freezer in small aliquots.
  • Expression plasmid containing full-length human PD-1 cDNA was obtained from Origene (Cat. No. SC117011, NCBI Accession No: NM_005018.1, Beijing, China). The extracellular domain consisting of amino acid (AA) 1-168 of PD-1 was PCR-amplified, and subcloned in pcDNA3.1-based expression vector (Invitrogen, Carlsbad, CA, USA) with C-terminus fused to the Fc domain of human IgG4 heavy chain, abbreviated as PD-1/Fc.
  • The human CD80 (B7-1) cDNA was synthesized by GeneScript according to the published sequence (NCBI access number NM_005191.3). The extracellular domain (AA1-242) of CD80 was fused with human Fc at C-terminus and subcloned in a mammalian expression vector similar to the method described previously (Pat. US 8735553). The fusion protein was named as CD80-ECD/Fc or CD80/Fc.
  • Stable Cell Line Expressing PD-L1
  • Stable cell line expressing human PD-L1 was established by transfection of pcDNA3.1 plasmid containing PD-L1 into HEK293 (ATCC, Manassas, VA, USA), and followed by selection with media containing 600 micrograms of hygromycin (Cat. No. 10687-010, Invitrogen) per milliliter. Single clones were isolated by picking up single colonies from culture-dish surface. All clones were screened by FACS analysis and Western blot using PD-L1 antibody (Cat. No. 17-5983, eBioscience, San Diego, USA), and the top expression clones were used for FACS binding analyses and functional assays.
  • Immunization and Hybridoma Clone Generation
  • Murine anti-human PD-L1 monoclonal antibodies were generated using the hybridoma fusion technology. All animal protocols were reviewed by and performed following BeiGene Animal Care and Use Procedure. Ten to twelve week-old Balb/c mice (HFK Bioscience, Beijing, China) were immunized three times (3 weeks apart between injections) subcutaneously and/or intra-peritoneally, each immunization was done with 100 uL of adjuvant (Cat. No. KX0210041, KangBiQuan, Beijing, China) containing 5-10 microgram of PD-L1/Fc. Two weeks after the 2nd immunization, the mice sera were collected and evaluated for PD-L 1 binding by ELISA and FACS. An example of such assay results were shown in Table 1 and Table 2. The mice with high anti-PD-L1 binding titers in sera were selected and boosted intraperitoneally with 50 micrograms of PD-L1/Fc in PBS. Three days after boosting, the splenocytes were isolated and fused with the murine myeloma cell line, SP2/0 (ATCC), using standard techniques (Mechetner et. al. 2007 Methods Mol Biol 378:1-13) with some modification.
  • TABLE 1
    Binding activities of serial diluted mice sera to PD-L1/His protein in ELISA assay
    Mouse sera dilution factor ELISA binding (OD450)
    Immunized mouse #1 Immunized mouse #2 Immunized mouse #3
    1:300 5.749 5.546 5.586
    1:900 5.651 4.978 4.453
    1:8100 4.166 2.853 2.137
    1:24300 2.641 1.539 0.896
    1:72900 1.287 0.498 0.212
    1:218700 0.282 0.065 0.056
  • TABLE 2
    Binding activities of serial diluted mice sera to HEK293/PD-L1 cells in FACS assay
    Mouse sera dilution factor FACS binding (MFI)
    Immunized mouse #1 Immunized mouse #2 Immunized mouse #3
    1:300 2657.7 1675.8 1499
    1:900 1485.9 681.6 560.5
    1:8100 355.6 274.7 175.7
    1:24300 73.9 107.1 54.9
    1:72900 33.9 26.9 19.8
  • Assess PD-L1 Binding Activity of Murine mAbs by ELISA and FACS
  • The supernatants of hybridoma clones were initially screened for PD-L1 binding activities by a modified ELISA assay (Flanagan 2007 Methods Mol Biol 378:33-52). Briefly, 50-200 nanograms of PD-L1/His were diluted in 50 microliters of PBS and coated in each well of 96-well ELISA plates (JinCanHua, Shenzhen, China). After blocking with 3% bovine serum albumin in TBST (20 mM Tris, 150 mM NaCl, 0.05% Tween20, pH7.5) and incubating with culture supernatants of hybridoma clones, the HRP-conjugated horse anti-mouse IgG antibody (Cat. No. 7076S, Cell Signaling Technology) and tetramethylbenzidine (TMB) (Cat. No. PA107-01, TianGen, Beijing, China) were used to detect binding signals by a plate reader (PHREAstar, BMG Labtech, Germany) as light absorbance at 450 nm. The ELISA-positive clones were further verified by fluorescence-activated cell sorting (FACS). PD-L1 expression cells, HEK293/PD-L1 (105 cells/well), were incubated with supernatants from hybridoma clones in V-bottom 96-well plates (Cat. No. 3897, Corning). The cell surface bound PD-L1 antibodies were detected with Dylight 649-conjugated goat anti-mouse IgG antibody (Cat. No. 405312, Biolegend, San Diego, CA, USA) and cell fluorescence was monitored in a Guava EasyCyte 8HT flow cytometer (Millipore, USA).
  • The hybridoma clones that were positive in both ELISA and FACS assays were then tested in human immune cell-based functional assays to identify antibodies with good functional activities. The hybridoma clones with positive functional activities were further subcloned and characterized.
  • Subcloning and Hybridoma Cell Adaption to Serum-Free or Low Serum Medium
  • The positive hybridoma clones from primary screening through ELISA, FACS and functional assays were subcloned by limiting dilution. Three subclones from each original clone were selected and confirmed in FACS and functional assays. The subclones selected through functional assays were defined as monoclonal antibody. The top subclones were adapted to grow in the CDM4MAB medium (Cat. No. SH30801.02, Hyclone) with 0-3% fetal bovine serum for purification and further characterizations.
  • Assesses of Binding Activities of Purified Antibodies
  • Hybridoma cells or 293-F cells transiently transfected with an antibody expression plasmid (Cat. No. R79007, Invitrogen) was cultured either in CDM4MAb medium (Cat. No. SH30801.02, Hyclone) or in Freestyle™ 293 Expression medium (Cat. No. 12338018, Invitrogen), and incubated in a CO2 incubator for 5 to 7 days at 37° C. The conditioned medium was collected through centrifugation at 10,000 g for 30 minutes to remove all cells and cell debris, and filtrated through a 0.22 µm membrane before purification. Murine or recombinant antibodies containing supernatants were applied and bound to a Protein A column (Cat. No. 17127901, GE Life Sciences) following the manufacturer’s guide, washed with PBS, eluted in an acidic buffer (pH3.5) containing 20 mM citrate, 150 mM NaCl. The eluted materials were neutralized with 1 M Tris pH8.0. The procedure usually yielded antibodies with purity above 90%. The Protein A-affinity purified antibodies were either dialyzed against PBS or further purified using a HiLoad 16/60 Superdex200 column (Cat. No. 17531801, GE Life Sciences) to remove aggregates. Protein concentrations were determined by measuring absorbance at 280 nm or by Bradford assay (Cat. No. 1856210, Thermo Scientific, Rockford, IL, USA) using bovine IgG reference standard (Cat. No. 23212, Thermo Scientific). The final antibody preparations were stored in aliquots in -80° C. freezer.
  • The binding activities of the purified monoclonal antibodies were evaluated in ELISA and FACS assays as described in previous sections. The dose-dependent binding curves in ELISA and FACS were used to compare mAb potency. The results of two representative murine mAbs were illustrated in FIG. 2 and Table 3. Murine mAb333 (mu333) has a dose-dependent binding activities with EC50 (effective concentration at 50% activity) of 0.036 µg/mL and 0.099 µg/mL, respectively, in ELISA and FACS assays. Mu277 has a similar binding activity to Mu333 in ELISA (EC50=0.031 µg/mL), but a lower binding activity in FACS (EC50=0.371 µg/mL). In contrast, control mouse IgG (muIgG) had no bindings to PD-L1 in both assays.
  • TABLE 3
    Dose-dependent binding of anti-PD-L1 mAbs in ELISA and FACS assays
    Antibody ELISA EC50 (µg/mL) Top OD450 FACS EC50 (µg/mL) Top MFI
    mu333 0.036 4.026 0.099 1101
    mu277 0.031 3.730 0.371 793
    mulgG N/A N/A N/A N/A
    OD450: Absorbance signal at 450 nm in ELISA assay;
    MFI: Mean fluorescence intensity from FACS analysis
    N/A: Not applicable
  • Example 2. Functional Activities of Anti-PD-L1 Antibodies Generation of Stable Cell Lines
  • The stable cell lines for human T cell-based functional assays were essentially the same as described in Pat. US8735553. Briefly, a fusion protein expression plasmid, OS8, was generated containing a scFv of anti-human CD3 mAb OKT3 and a C-terminal domain of mouse CD8α which included transmembrane and cytoplasmic domains. OS8 could function as a membrane anchored T cell engager that directly activates T-cell receptor (TCR). A stable cell line that co-expresses both OS8 and PD-L1 was generated by co-transfection of two expression constructs in HEK293 cells followed by hygromycin or G418 selection for 10-14 days. This cell line was named as HEK293/OS8/PD-L1. Similarly, a human T-cell line, HuT78/PD-1, was generated that expresses human PD-1. And a reverse signaling human T-cell line, HuT78/P3Z, was generated by stable transfection with a chimeric PD-1 expression construct (named as P3Z) made by fusing the extracellular and transmembrane domains of human PD-1 to the cytoplasmic region of human CD3ζ chain. In this way, P3Z encoded a membrane bound receptor that would activate T cells upon ligation with PD-1 ligand (PD-L1 or PD-L2). Cell lines were cloned by limiting dilution as described previously (Fuller 2001 Curr Protoc Mol Biol, Chap 11, unit 11.8).
  • Determination of PD-L1 Antibody Functions by IL-2 Release in HuT78/PD-1 Cells
  • To determine whether anti-PD-L1 antibodies can block the PD-1 signaling induced by PD-L1, HEK293/OS8/PD-L1 cells were pre-incubated with anti-PD-L1 mAbs for 15 minutes prior to co-culture with HuT78/PD-1 cells (1-3 ×104 per well) in a flat bottom plate fed with 200 µl of RPMI1640 growth medium per well at 37° C. After 16-18 hours of co-culture, supernatants were collected. IL-2 was assayed by ELISA using human IL-2 Ready-Set-Go! ELISA kits (Cat. No. 88-7025, eBiosciences, San Diego, CA). In this assay, blockade of PD-L1-PD-1 signaling with anti-PD-L1 antibodies resulted in enhanced TCR signaling and IL-2 production.
  • As shown in Table 4, supernatants of ELISA and FACS-binding positive hybridoma clones were screened in this functional assay. Although all the tested clones bound to PD-L1 in ELISA and FACS assays, only a few of them could block PD-L1-PD-1 signaling and resulted in increase of IL-2 production. The remaining clones resulted in either no increase or very little increase of IL-2 production compared to the negative control with fresh medium only. In this experiment, an OD450 reading cut off was set at 2.5, i.e. clones that stimulated IL-2 production above this level were considered to have antagonist functions (Table 4). A reference anti-PD-L1 mAb (named Y1) was synthesized based on the variable regions of the published data (US 2010/0203056 A1), and both human and mouse format of Y1 antibodies were generated by fusing Y1 variable regions with mouse or human IgG1κ constant regions to generate Y1-muIgG1 or Y1-huIgG1, respectively (collectively termed asY1-hIgG1). Y1-mulgG1′s function was also confirmed in this assay.
  • TABLE 4
    Functional screening of anti-PD-L1 hybridoma clones in HEK293/OS8/PD-L1 coculture with HuT78/PD-1
    Sample/Clone ID OD450 in IL-2 ELISA assay
    negative control: medium only 1.30 ± 0.06
    mu31 1.28 ± 0.03
    mu32 1.33 ± 0.02
    mu33 1.24 ± 0.01
    mu34 1.19 ± 0.12
    mu35 1.27 ± 0.02
    mu36 2.95 ± 0.22
    mu37 3.10 ± 0.11
    mu38 1.33 ± 0.44
    mu39 2.94 ± 0.45
    mu310 1.90 ± 0.01
    mu311 1.38 ± 0.08
    mu312 1.40 ± 0.07
    mu313 1.49 ± 0.07
    mu314 1.26 ± 0.01
    mu315 1.36 ± 0.11
    mu316 1.23 ± 0.12
    mu317 1.72 ± 0.12
    mu318 2.21 ± 0.06
    mu319 1.38 ± 0.05
    mu320 1.32 ± 0.10
    mu321 1.33 ± 0.02
    mu322 1.34 ± 0.10
    mu323 1.52 ± 0.06
    mu324 3.09 ± 0.11
    mu325 1.44 ± 0.02
    mu326 1.35 ± 0.19
    mu327 2.55 ± 0.36
    mu328 3.10 ± 0.47
    mu329 1.43 ± 0.07
    mu330 1.46 ± 0.11
    mu331 1.37 ± 0.07
    mu332 1.44 ± 0.05
    mu333 3.01 ± 0.23
    mu334 3.22 ± 0.09
    mu335 3.03 ± 0.15
    mu336 3.12 ± 0.24
    mu337 1.28 ± 0.06
    mu338 1.34 ± 0.05
    *Functional clones were showed in bold
  • The purified murine anti-PD-L1 mAbs were compared in the same assay for quantitative assessments of the blocking activities. FIG. 3 showed the dose response curves of the representative murine-anti-PD-L1 mAbs. Table 5 summarized the EC50 and the maximum IL-2 concentration these mAbs could induce. Mu333 was a potent antagonist of the PD-L1-PD-1 signaling, and induced significant IL-2 production with a very low EC50. In contrast, one of the mAbs, mu277, had much weaker blocking activity than mu333, judged by the dose dependent response curve and the parameters of top line signal read-out and EC50. As the negative control, muIgG could not block PD-L1/PD-1 signaling and stimulate IL-2 production.
  • TABLE 5
    IL-2 release induced by anti-PD-L1 mAbs in HuT78/PD-1 cells co-cultured with HEK293/OS8/PD-L1 cells
    Antibody Baseline (pg/mL) Top line (pg/mL) EC50 (µg/mL)
    mu333 37 436 0.092
    mu277 37 225 0.510
    mulgG 37 N/A N/A
    Baseline: Average IL-2 release induced by mulgG at all tested concentrations, see FIG. 3
    Top line: Highest IL-2 release(pg/ml) based on regression calculation by Prizm Software, see FIG. 3
    N/A: Not applicable
  • Determination of PD-L1 Antibody Functions by Reverse Signaling of IL-2 Release in HuT78/P3Z Cells
  • In HuT78/P3Z cells, PD-1 mediated TCR signaling is reversed by design as described in the previous sections. In this assay, HEK293/PD-L1 cells were pre-incubated with purified PD-L1 antibodies for 15 minutes prior to co-culture with HuT78/P3Z cells in 96-well flat bottom plates at 37° C. After 16-18 hours of co-culture, supernatants were collected and IL-2 production was assayed by ELISA as described above.
  • Inhibitory activity of murine anti-PD-L1 mAbs was detected directly correlated to the decrease of IL-2 release in dose-dependent fashion. Consistent with the results shown above, mu333 had potent activities inhibiting IL-2 secretion by preventing PD-L1 engagement on P3Z chimeric receptor on HuT78 cells. As showed in Table 6 and FIG. 6 , mu333 was much more potent than mu277 in terms of IC50 (the concentration of mAb at 50% inhibition of the maximum activity), consistent with the above results obtained with regular signaling in T-cell. The negative control, muIgG, could not inhibit PD-L1/P3Z induced IL-2 production.
  • TABLE 6
    Inhibition of IL-2 secretion by anti-PD-L1 mAbs in HuT78/P3Z cells co-cultured with HEK293/PD-L1 cells
    Antibody IC50 (µg/mL) Maximum inhibition*
    mu333 0.021 100%
    mu277 0.331 100%
    mulgG N/A N/A
    *Maximum inhibition was calculated as percentage (%) of inhibition with anti-PD-L1 mAbs added to the highest concentration(10 µg/ml) in culture
  • Competitive Inhibition of PD-1 Binding to Cell Surface-Expressed PD-L1
  • To determine whether anti-PD-L1 antibodies can compete with PD-1 binding to PD-L1, HEK293/PD-L1 cells (1×105 cells per well) were incubated with the mixture of PD-L1 antibodies and biotin-conjugated PD-1/Fc fusion protein in V-bottom 96-well plate. Biotinylation of PD-1/Fc was done using the EZ-Link Sulfo-NHS-LC-Biotin reagent according to manufacturer’s instruction (Cat. No. 21327, Thermo Sci). Inhibition of PD-L1 and PD-1/Fc interaction by antibodies was assayed (Guava easyCyte 8HT Flow Cytometer, Millipore, USA) by mean fluorescence intensity (MFI) readout probed with Streptavidin-APC. Using this method, we evaluated functional strength of anti-PD-L1 mAbs. As shown in FIG. 5 and Table 7, the murine mAbs competitively bind to PD-L1, inhibiting MFI readout elicited by Biotin-PD-1/Fc binding to cell surface PD-L1 in the FACS assay. Mu333 showed better inhibitory efficacy with an IC50 of 0.463 µg/mL, compared to an IC50 of 2.172 µg/mL for mu277. In contrast, control antibody, murine IgG, had no such inhibitory effect (FIG. 5 ).
  • TABLE 7
    Inhibition of PD-1 binding to PD-L1 on HEK293 cells
    Antibody lC50 (µg/mL) Maximum inhibition
    mu333 0.463 100%
    mu277 2.172 98%
    mulgG N/A N/A
    Maximum inhibition was calculated as percentage (%) of inhibition with anti-PD-L1 mAbs added to the highest concentration(10 µg/ml); N/A: not applicable
  • Competitive Inhibition of CD80 Binding to Cell Surface PD-L1
  • Besides interaction with PD-1, PD-L1 also binds to another B7 family protein, B7-1 or alternatively named as CD80 (Butte M.J. 2007 Immunity 27:111-122). To determine whether the anti-PD-L1 antibodies compete against the binding of CD80 (NCBI accession: NP _005182.1) to PD-L1, HEK293/PD-L1 cells were incubated with the mixture of PD-L1 antibodies and biotin-conjugated CD80/Fc fusion protein. In this assay, blockade of biotin-CD80/Fc binding to PD-L1 by anti-PD-L1 antibodies resulted in decreased binding signals (MFI readings). As shown in FIG. 6 and Table 8, mu333 competed off the binding of CD80 to PD-L1 with100% of max inhibition and very low IC50 (0.052 µg/mL), which compared to the IC50 of 0.162 µg/mL for mu277. In contrast, murine IgG had no such competition effect (FIG. 6 ).
  • TABLE 8
    Inhibition of CD80 binding to cell surface expressed PD-L1 on HEK293 cells
    Antibody lC50 (µg/mL) Maximum inhibition
    mu333 0.052 100%
    mu277 0.162 99%
    mulgG N/A N/A
    Maximum inhibition was calculated as percentage (%) of inhibition with anti-PD-L1 mAbs added to the highest concentration(10 µg/ml) in culture; N/A: not applicable
  • Example 3. Sequence Analysis of Murine Anti-PD-L1 Antibodies
  • Cloning and sequencing of variable regions from the selected murine hybridoma clones were done based on commonly used methods with some modifications (Kontermann and Dubel, 2010 Antibody Engineering, Vol 1:3-14). Briefly, bybridoma cells were harvested, washed with PBS and collected by centrifugation at 1500 rpm in a swing bucket rotor. Total cellular RNA was isolated using Ultrapure RNA kit (Cat. No. CW0581, CW Biotech, Beijing, China) following the manufacturer’s protocol.
  • The 1st strand cDNA was synthesized using reverse transcriptase (Cat. No. AH301-02, TransGen, Beijing, China). PCR amplification of heavy chain (Vh) and light chain variable region (Vκ) of murine mAb was performed using PCR reagent kit (Cat. No. AP221-12, TransGen, Beijing, China) and a set of primers specific for cloning of murine Vh and Vκ as described (Brocks 2001 Mol Med 7:461-469). The PCR products were subcloned into the pEASY-Blunt cloning vector (Cat. No. CB101-02, TransGen) and subsequently sequenced by Genewiz (Beijing, China). The amino acid sequences of Vh and Vk were deduced from the DNA sequencing results.
  • The murine mAbs were analyzed by comparison of sequence homology, and grouped based on both sequence homology and epitope-mapping results (see Example 7). Complementary determinant regions (CDRs) were defined based on the Kabat (Wu and Kabat 1970 J. Exp. Med. 132:211-250) and IMGT (Lefranc 1999 Nucleic Acids Research 27:209-212) system by sequence annotation and by internet-based sequence analysis (http: //www.imgt.org/IMGT vquest/share/texts/index.html). Table 9 listed the CDRs of mu333 (SEQ. NO. 5-14), based on the definitions of Kabat and IMGT systems.
  • TABLE 9
    CDR sequences of mu333
    MAbs CDR1 SEQ ID NO CDR2 SEQ ID NO CDR3 SEQ ID NO
    mu333, Vh GFSLTSYG VH 9 VIWAGGSTNYNSALMS 10 AKPYGNSAMDY 11
    mu333, Vk KASQDVGIVVA 12 WASIRHT 13 QQYSNYPLYT 14
    Note: CDR sequences in bold face are defined based on Kabat system; CDR sequences underlined are defined based on IMGT system.
  • Example 4. Humanization of the Murine Anti-Human PD-L1 mAb Simulation of 3D Structure of Murine mAb
  • The three dimensional structures were simulated for variable domain of mu333 in order to identify framework residues that might be important for supporting CDR loop structures. Potentially important framework residues were kept as the original murine residues in the first round antibody humanization. The previously established structural modeling method for antibodies (Morea et al. Methods 2000 20:267-279) was adopted to simulate 3D structure of anti-PD-L1 mAb mu333 based on the known canonical structures of antibodies (Al-Lazikani et al. 1997 Journal of Molecular Biology 273:927-948). Briefly, the sequence of each variable domain (Vk and Vh) of mu333 was blasted in the PDB database (Protein Data Bank, http:// blast.ncbi.nlm.nih.gov/) to identify the most homologous antibody sequence with known high resolution structure (resolution less than 2.5 angstrom). Selected structure templates for modeling mu333 (listed in Table 10) had the same classes of canonical loop structures in L-CDR1, L-CDR2, L-CDR3, H-CDR1, and H-CDR2 to the mu333 to be modeled. As the templates for Vκ and Vh came from different immunoglobulins, they were packed together by a least-squares fit of the main chain atoms to form a hybrid structure of Vk-Vh interface residues, which was used as the templates for structural homology modeling by Swiss-model program (Kiefer et al. 2009 Nucleic Acids Research 37, D387-D392). Certain side chain conformation was adjusted while the main chain conformations were retained. At the sites where the parental structure and the modeled structure had the same residue, the side chain conformation was retained. At sites where the residues were different, side chain conformations were modeled on the basis of template structure, rotamer libraries and packing considerations. After homology modeling, PLOP program (Jacobson et al. 2002 Journal of Physical Chemistry 106:11673-11680) was used to refine the homology models to minimize all-atom energy and optimize Vk and Vh interface. This step was performed to improve the stereochemistry, especially in those regions where segments of structures coming from different antibodies had been joined together. The modeled 3D structure of mu333 variable domain was used to guide the structure-based humanization and engineering process.
  • TABLE 10
    Structure templates used in antibody structure simulation
    Antibody chain PDB code of template structure Sequence identity Sequence similarity
    mu333 Vk 1H8N 92% 94%
    mu333 Vh 3VFG 88% 91%
  • MAb Humanization and Engineering
  • For humanization of the anti-PD-L1 mAb, human germline IgG genes were searched for sequences that share high degree of homology to the cDNA sequences of mu333 variable regions by blasting the human immunoglobulin gene database in IMGT (http://www.imgt.org/IMGT_vquest/share/textes/index.html) and NCBI (http://www.ncbi.nlm.nih.gov/igblast/)websites. The human IGVH and IGVκ genes that are present in human antibody repertoires with high frequency (Glanville 2009 PNAS 106:20216-20221) and are highly homologous to mu333 were selected as the templates for humanization.
  • Humanization was carried out by CDR-grafting (Methods in Molecular Biology, Vol 248: Antibody Engineering, Methods and Protocols, Humana Press) and the humanization antibodies (hu333s) were engineered as the human Fab format using an in-house developed expression vector. In the initial round of humanization, mutations from murine to human amino acid residues in framework regions were guided by the simulated 3D structure, and the murine framework residues of structural importance for supporting the canonical structures of CDRs were retained in the 1st version of humanization antibody 333 (hu333-1A, SEQ. NO. 15-16). Specifically, CDRs of mu333 Vk were grafted into the framework of human germline variable gene IGVK1-5, and no murine framework residues were retained (SEQ NO 16). CDRs of mu333 Vh were grafted into the framework of human germline variable gene IGVH3-7, with 4 murine framework residues retained, V24, L67, K71 and V78 (SEQ NO 15). All grafted CDRs were based on the Kabat’s CDR definition in hu333-1A (Table 9 and SEQ. NO. 15-16). In the following hu333 variants, only the N-terminal half of Kabat H-CDR2 was grafted, as only the N-terminal half was considered to be important for antigen binding according to the simulated 3D structure (Table 14).
  • Hu333-1A were constructed as human Fab format using in-house developed expression vectors that contain human IgG CH-1 and constant region of kappa chain, respectively, with easy adapting subcloning sites. The hu333-1A joined IgG2a-CH1 was tagged at C-terminus with a 8xHis peptide to facilitate purification. The C232S and C233S (Kabat residue numbering, Kabat et al. Sequence of proteins of immunologic interest, 5th ed Bethesda, MD, NIH 1991) mutations were introduced in the IgG2 heavy chain to prevent disulfide bond exchange and stabilize human IgG2 in the IgG2a conformation (Lightle et al. 2010 Protein Sci 19(4): 753-762). Both constructs contained a signal peptide upstream of the Fab mature sequences. Secreted expression of hu333-1A Fab was achieved by co-transfection of the above two constructs into 293-F cells and cultured for 6-7 days before harvest. His8-tagged Fabs were purified from expression culture supernatants using a Ni-sepharose Fast Flow column (Cat. No. 17531801, GE Life Sciences) followed by size exclusion chromatography using a HiLoad 16/60 Superdex200 column (Cat. No. 17106901, GE Life Sciences). The purified Fabs were concentrated to 0.5-5 mg/mL in PBS and stored in aliquots in -80° C. freezer.
  • For affinity determinations of anti-PD-L1 Fabs, SPR assays were performed using BIAcore™ T-200 (GE Life Sciences). Briefly, human PD-L1/His was coupled to an activated CM5 biosensor chip (Cat. No. BR100530, GE Life Sciences) to achieve approximately 100-200 response units (RU), followed by blocking un-reacted groups with 1 M ethanolamine. A serial dilutions of 0.12 nM to 90 nM Fab samples were injected, mixed into the SPR running buffer (10 mM HEPES, 150 mM NaCl, 0.05% Tween20, pH7.4) at 30 µL/minute, and binding responses on human PD-L1/His were calculated by substracting of RU from a blank flow-cell. Association rates (kon) and dissociation rates (koff) were calculated using the one-to-one Langmuir binding model (BIA Evaluation Software, GE Life Sciences). The equilibrium dissociation constant (Kd) was calculated as the ratio koff/kon.
  • Functional activities of hu333 Fabs were confirmed in PD-1 competition assays described in previous sections. Data from SPR measurement and functional assays were summarized in Table 11. Hu333-1A Fab had very high affinity (Kd=9.88 pM) to PD-L1 indicated by a fast kon (1.61 × 106 M-1s-1) and very slow koff(1.59 × 10-5 s-1). It was observed that there was very slow or virtually no dissociations of hu333-1A Fab from the coated PD-L1during the 5-15 minutes of dissociation time in this experiment. It was apparent that the affinity of hu333-1A Fab to PD-L1 was close to the detection limit of the SPR technology. Such high affinity of hu333-1A Fab was consistent with the high potencies in all functional assays tested (Table 11).
  • Following on hu333-1A, we made individual mutations converting the four murine residues in framework region of Vh to corresponding human germline residues, respectively. At same time In order to further improve humanization level, we also changed the C-terminal part of H-CDR2 (Kabat’s definition) from murine sequence to corresponding human germline residues (Table 14, hu333-2B). Specifications of the four humanization Fabs were hu333-2A (V24A in Vh), hu333-2B (L67F in Vh), hu333-2C (K71R in Vh) and hu333-2D (V78L in Vh), which are illustrated in Table 13 with H-CDR2 changes. All humanization mutations were made using primers containing mutations at specific positions and a site directed mutagenesis kit (Cat. No. FM111-02, TransGen, Beijing, China). The desired mutations were verified by sequencing analyses. These hu333 Fabs were expressed, purified and tested in binding and functional assays as described previously. Comparing to hu333-1A, hu333-2A, hu333-2C and hu333-2D had significantly reduced binding affinities and functionalities. Only hu333-2B (SEQ. NO. 16 and 17) had similar binding and functional activities to hu333-1A (Table 11). Taken together, hu333-2B (SEQ. NO. 16 and 17) reached a high level of humanization in the framework regions while maintained potent binding affinity and functional activities.
  • TABLE 11
    Comparison of hu333-1A and hu333-2B Fabs by SPR and functional assays
    Assay/Parameter hu333-1A Fab hu333-2B Fab
    BiaCore SPR kon (M-1S-1) 1.61 × 106 1.36 × 106
    koff (S-1) * 1.59 × 10-5 2.09 × 10-6
    Kd (pM) * 9.88 1.54
    PD-1 binding competition (FACS) IC50 (µg/ml) 0.057 0.062
    Max inhibition 100% 100%
    CD80 binding competition (FACS) IC50 (µg/ml) 0.049 0.055
    Max inhibition 99% 99%
    IL-2 release in HuT78/PD-1# EC50 (µg/ml) 0.066 0.054
    Top line (pg/ml) 1369 1436
    IL-2 release in HuT78/P3Z§ IC50 (µg/ml) 0.012 0.011
    Max inhibition 100% 100%
    * koff might be too slow to be accurately measured during the 5-15 min dissociation time in the SPR experiment. Therefore, the affinity might be too strong to be accurately determined using current instrument/experiment setting #IL-2 release in HuT78/PD-1: IL-2 release induced by the Fabs in HuT78/PD-1 cells co-cultured with HEK293/OS8/PD-L1 cells §IL-2 release in HuT78/P3Z: IL-2 release induced by the Fabs in HuT78/P3Z cells co-cultured with HEK293/PD-L1 cells
  • To explore the best possible Vh and Vk sequence composition for hu333 that could be used as therapeutic antibody in human, we further engineered the hu333 by introducing mutations in CDRs and framework regions in considerations of the antibody’s molecular properties, such as physiochemical stabilities, amino acid compositions, projected isoelectronic points (pIs), expression level, antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) while maintaining functional activities.
  • Deamination site NS76-77 in Vh of hu333-1A was mutated to NT76-77 to generate hu333-3A2 (SEQ. NO. 18 and 23). V60 of hu333-3A2 -Vh was mutated to V60A, which is consistent to the consensus sequences of major human IGVH3 genes with reduced a surface-exposure of hydrophobicity. This mutant construct was given the code name hu333-3C2 (SEQ. NO. 19 and 23). Another deamidation site NS73-74 was mutated to TS73-74 on the template of hu333-3C2, which is also consistent to the consensus sequences of major human IGVH3 genes. The latter one was named as hu333-3H2 (SEQ. NO. 20 and 23). As summarized in Table 12, hu333-3A2, hu333-3C2 and hu333-3H2 all retained the potent functional activity, only with slight variations in binding affinity. On the other hand, these engineered hu333 variants have better projected physiochemical properties.
  • TABLE 12
    Comparison of hu333-3A2 and hu333-3C2 and hu333-3H2 Fabs by SPR and functional assays
    Assay/Parameter hu333-3A2 Fab hu333-3C2 Fab hu333-3H2 Fab
    BiaCore SPR kon (M-1S-1) 1.28 × 106 1.42 × 106 1.32 × 106
    koff (S-1) * 2.2 × 10-7 1.15 × 10-5 4.61 × 10-5
    Kd (pM) * 0.17 8.09 34.9
    PD-1 binding competition (FACS) IC50 (µg/ml) 0.068 0.065 0.071
    Max inhibition 100% 100% 100%
    CD80 binding competition (FACS) IC50 (µg/ml) 0.044 0.073 0.064
    Max inhibition 99% 99% 99%
    IL-2 release in HuT78/PD-1# EC50 (µg/ml) 0.057 0.046 0.057
    Top line (pg/ml) 2551 3124 3016
    IL-2 release in HuT78/P3Z§ IC50 (µg/ml) 0.014 0.013 0.014
    Max inhibition 95% 100% 100%
    * koff might be too slow to be accurately measured during the 5-15 min dissociation time in the SPR experiment. Therefore, the affinity might be too strong to be accurately determined using current instrument/experiment setting. #IL-2 release in HuT78/PD-1: IL-2 release induced by the Fabs in HuT78/PD-1 cells co-cultured with HEK293/OS8/PD-L1 cells §IL-2 release in HuT78/P3Z: IL-2 release induced by the Fabs in HuT78/P3Z cells co-cultured with HEK293/PD-L1 cells
  • To eliminate the last deamidation site in the CDR3 of Vh, we mutated NS101-102 to TS101- 102 on the templates of hu333-3A2 and hu333-3H2, respectively. The resulting humanization mAbs were constructed in human IgG1 Fab format, named as hu333-4A2 (SEQ. NO. 21 and 23) and hu333-4B2 (SEQ. NO. 22 and 23). The results from binding and functional assays indicated both hu333-4A2 and hu333-4B2 were very similar in affinity and functional activities such as blocking the PD-L1 binding to its targets (PD-1 and CD80) and inhibiting the PD-L1 and PD-1 mediated downstream signaling (Table 13 and Table 14). Several mutations made in the processes including hu333-3B2, -3D2, -3E2, -3G2 and -3I2 were dropped from further development for various considerations. The CDRs of the above mAbs were compared to those of mu333 were shown in Table 14.
  • TABLE 13
    Comparison of hu333-4A2 and hu333-4B2 Fabs by SPR and functional assays
    Assay/Parameter hu333-4A2 Fab hu333-4B2 Fab
    BiaCore SPR kon (M-1S-1) 3.88 × 106 3.78 × 106
    koff (S-1) * 1.03 × 10-5 1.32 × 10-5
    Kd (pM) * 2.65 3.50
    PD-1 binding competition (FACS) IC50 (µg/ml) 0.050 0.053
    Max inhibition 100% 100%
    CD80 binding competition (FACS) IC50 (µg/ml) 0.045 0.062
    Max inhibition 100% 100%
    IL-2 release in HuT78/PD-1# EC50 (µg/ml) 0.050 0.058
    Top line (pg/ml) 227.5 215.5
    IL-2 release in HuT78/P3Z§ IC50 (µg/ml) 0.005 0.016
    Max inhibition 100% 100%
    * koff might be too slow to be accurately measured during the 5-15 min dissociation time in the SPR experiment. Therefore, the affinity might be too strong to be accurately determined using current instrument/experiment setting. #IL-2 release in HuT78/PD-1: IL-2 release induced by the Fabs in HuT78/PD-1 cells co-cultured with HEK293/OS8/PD-L1 cells §IL-2 release in HuT78/P3Z: IL-2 release induced by the Fabs in HuT78/P3Z cells co-cultured with HEK293/PD-L1 cells
  • TABLE 14
    Comparison of CDRs among the selected 333 mAbs
    mAbs CDR1 SEQ ID NO CDR2 SEQ ID NO CDR3 SEQ ID NO
    mu333, vh GFSLTSYGVH 9 VIWAGGSTNYNSALMS 10 AKPYGNSAMDY 11
    hu333-1A, vh GFSLTSYGVH 9 VIWAGGSTNYNSALMS 10 AKPYGNSAMDY 11
    hu333-2B, vh GFSLTSYGVH 9 VIWAGGSTNYVDSVKG 24 AKPYGNSAMDY 11
    hu333-3A2, vh GFSLTSYGVH 9 VIWAGGSTNYVDSVKG 24 AKPYGNSAMDY 11
    hu333-3C2, vh GFSLTSYGVH 9 VIWAGGSTNYADSVKG 25 AKPYGNSAMDY 11
    hu333-3H2, vh GFSLTSYGVH 9 VIWAGGSTNYADSVKG 25 AKPYGNSAMDY 11
    hu333-4A2, vh GFSLTSYGVH 9 VIWAGGSTNYVDSVKG 24 AKPYGTSAMDY 26
    hu333-4B2, vh GFSLTSYGVH 9 VIWAGGSTNYADSVKG 25 AKPYGTSAMDY 26
    mu333, vk KASQDVGIVVA 12 WASIRHT 13 QQYSNYPLYT 14
    hu333-1A, vk KASQDVGIVVA 12 WASIRHT 13 QQYSNYPLYT 14
    hu333-2B, vk KASQDVGIVVA 12 WASIRHT 13 QQYSNYPLYT 14
    hu333-3A2, vk KASQDVGIVVA 12 WASIRHT 13 QQYSNYPLYT 14
    hu333-3C2, vk KASQDVGIVVA 12 WASIRHT 13 QQYSNYPLYT 14
    hu333-3H2, vk KASQDVGIVVA 12 WASIRHT 13 QQYSNYPLYT 14
    hu333-4A2, vk KASQDVGIVVA 12 WASIRHT 13 QQYSNYPLYT 14
    hu333-4B2, vk KASQDVGIVVA 12 WASIRHT 13 QQYSNYPLYT 14
    Note: AA residues underlined are changed from murine sequence to human sequence or mutated for improvement of physicochemical properties.
  • All the humanization mAbs shown above were also confirmed for functional effect on primary human immune cells, peripheral blood mononuclear cells (PBMCs), which were isolated from healthy donors by density gradient centrifugation using ficoll lymphocyte separation medium (Histopaque-1077; Cat. No. 10771, Sigma, St. Louis, USA) according to manufacturer’s instruction. PBMCs were then stimulated with 40 ng/mL of anti-CD3 mAb OKT3 (Cat. No. 16-0037, eBioscience, San Diego, CA, USA) for 3 days prior to the assay. The activated PBMC population mainly consisted of T-cells (50-70%), B-cells and NK cells (15-30%), and monocytes (2-10%). To better mimic the response of T cells to PD-L1 expressing tumor cells upon engagement of TCR/CD3 complex, the activated PBMCs were co-cultured with HEK293/OS8/PD-L1 cells in each well of 96-well plates. Functional effect of anti-PD-L1 mAbs were tested by adding the mAb to the culture, co-cultured for 15-18 hours before harvesting culture supernatants to assess IFN-y level using Ready-Set-Go! ELISA kits (Cat. No. 88-7316, eBiosciences). As shown in FIG. 7A and FIG. 7B, hu333-2B, hu333-3A2, hu333-3C2, hu333-4A2 and hu333-4B2 all resulted in increase of IFN-y secretion in a dose-dependent manner. In contrast, negative control, bovine serum albumin (BSA), had no such effect. The results were repeated using PBMCs from two different donors. Although the base level (without mAb) and magnitude changes of IFN-y secretion with mAb treatment varied among different donors, the fold of increase in IFN-y secretion remained similarly depending on dose concentration for all hu333.
  • Example 5. Generation of Recombinant Anti-PD-L1 mAbs Without Effector Functions by Fusion to Modified Human IgG1 Constant Region Design of Modified Human IgG1 Constant Region
  • PD-L1 is expressed on a wide range of normal human cells including hematopoietic cells such as T-cells, B-cells, dendritic cells, macrophages, mesenchymal stem cells and bone-marrow derived mast cells, and nonhematopoietic cells and tissues such as lung, hepatocytes, pancreatic islets, placental synctiotrophoblasts and vascular endothelium (Keir et. al. 2006 J Exp Med 203:883-895, Keir et. al. 2008 Ann Rev Immunol 26:677-704, Mu et. al. 2011 Medical Oncology 28:682-688). It is expected that PD-L1 blocking antibodies linked to human wild type IgG-yFc moieties will induce yFc-mediated effector functions, for examples, antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), which might lead to unwanted toxicity to vital organs.
  • To eliminate effector functions associated with anti-PD-L1mAbs while maintaining optimal physicochemical properties, we constructed hu333-4A2 and hu333-4B2 full antibody by linking the Vh sequences to mutated IgG1 constant regions, and screened for reduced or null Fcγ receptors (FcyRs) binding or C1q binding activities, therefore, attenuating or eliminating ADCC and CDC effector functions. The regions in IgG1 Fc that are involved in interactions with FcyRs and C1q have been studied extensively in the literature (Tao et al. 1993 J Exp Med 178:661-7; Cole et al. 1997 J Immunol 159:3613-21; Armour et. al. 1999 Eur J Immunol 29:2613-2624; Idusogie et. al. 2000 J of Immunol 164:4178-4184; Shields et. al. 2001 J of Biol Chem 276: 6591-6604; Lund et. al. 2002 Immunol Letters 82:57-65; reviewed by Strohl et. al. 2009 Current Opinion in Biotechnology 20:685-691). Taken together, these data have pointed to the essential role of lower hinge region (AA232-238 based on EU nomenclature) for binding to FcyRs and a structurally clustered region (D270, K322, P329 and P331based on EU nomenclature) of CH2 domain for binding to C1q. On the other hand, IgG2 has some sequence variations from IgG1 in the hinge region, which was attributed to weaker binding or no binding to most of the FcyRs except to FcγRIIAH131. Indeed, a IgG1/IgG2 hybrid (IgGlΔb) with most of IgG1 hinge sequence incorporating some IgG2 sequences was demonstrated having significantly reduced the binding activities to most FcyRs and attenuated ADCC and CDC effector functions (Armour et. al. 1999 Eur J Immunol 29:2613-2624).
  • By rational design of mutagenesis with considerations of good pharmaceutical and physicochemical properties, we generated a number of mutants IgG1 in the hinge and Fc regions described above, which were fused to the variable regions of hu333-4A2 and hu333-4B2, respectively, as full antibodies. Two of the IgG1 mutants, IgG1mc and IgG1mf, with favorable features in functional assays were shown in Table 15 in comparison to wild type IgG. The IgGlmc (SEQ. NO. 28) contains a combination of additional mutations, V234A, G237A and P239A, from the IgG1/IgG2 hybrid described above. The mutations of V234A and G237A were designed to reduce the surface hydrophobic side chain at the yFc/FcyR binding interface to further reduce the binding to FcyRIIA and FcyRIIB, (Lund et. al. 1992 Mol Immunol 29:53-59, Lund et. al. 1996 J Immunol 157:4963-4969, Wines et. al. 2000 J Immunol 164:5313-5318). The P239A mutation was designed to further reduce the C1q binding and CDC (Idusogie et. al. 2000 J of Immunol 164:4178-4184). The IgG1mf (SEQ. NO. 29) was similar to IgG1mc except that the amino acid residue G237 was not mutated. The recombinant full length anti-PD-L1 antibodies, hu333-4A2-IgG1mc (SEQ. NO. 30 and 32), hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) were expressed in HEK293-F cells and purified as described in previous sections.
  • TABLE 15
    Sequence modifications of IgG1 variants
    ... 231 232 233 234 235 236 237 238 ... 329 330 331 ...
    IgG1wt ... A P E L L G G P ... P A P ...
    IgG2wt ... A P P V A - G P ... P A P ...
    IgG4wt ... A P E F L G G P ... P S S ...
    IgG1mc ... A P P A A - A P ... A A P ...
    IgG1mf ... A P P A A - G P ... A A P ...
    Amino acid numbering is based on EU system. IgG1 sequence modifications are underlined.
  • ELISA Based FcγR and Clq Binding Assays
  • It was well documented that IgG mediated effector functions are triggered following on antibody-antigen complex binding to FcyRs or to complement component C1q (Nimmerjahn et. al. 2008 Nature Rev Immunol 8:34-47). For example, ADCC is initiated when an antibody binds to cell surface target protein followed by ligation to FcyRIIIA expressed on effector cells. CDC is activated when an antibody cross-links a cell surface target by binding to C1q protein, which leads to a cascade reaction of complement complex formation and activation and target cell lysis. As proxy of ADCC, CDC and other antibody mediated effector functions, biochemical assays for antibody binding to FcyRs and C1q may serve as the fundamental indicator of ADCC and CDC. We systematically assessed the bindings of the anti-PD-L1 antibodies with modified constant region to all major FcyRs and all known polymorphic variants, including FcyRI, FcγRIIAH131, FcγRIIAR131, FcγRIIIAF158, FcγRIIIAV158, FcγRIIB, and FcγRIIIB.
  • The extracellular domains of FcyRs were fused to C-terminal His tags as described in previous sections. Recombinant proteins were expressed in 293-F cells by transient transfection and purified using Ni-Sepharose column followed by gel filtration column as described. 2-5 µg/mL of FcyRs were coated on Nunc MaxiSorp ELISA plates (Cat. No. 442404, Nunc, Thermo Fisher) except FcyRIIB and FcyRIIIB, Ni-chelate plates were used (Cat. No. 15242, Pierce, Thermo Fisher). After washing and blocking of the wells, a preformed immune-complex was added to each well and incubated at room temperature for 1-2 hours. The preformed immune-complex contained 60 ng/mL streptavidin-HRP, 60 ng/mL of biotinylated-F(ab′)2 goat anti-human IgG (Cat. No. 109-066-097, Jackson ImmunoRes, West Grove, PA, USA), and 1-5 µg/mL of indicated IgG1 Fc variants fused to the humanized anti-PD-L1 (hu333-4A2 or hu333-4B2) in the blocking buffer. After washing the plate four times, binding signals were detected by chemiluminescence using Immobilon Chemiluminescence Substrate A/B (Cat. No. WBKLS0500, Millipore). Table 16 summarized the results of hu333-4A2-IgG1mc (SEQ. NO. 30 and 32), hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) binding to various FcyRs. In comparison to the hu333-4A2-IgG1wt, all three IgG1 mutant hu333 mAbs had very low binding activities to FcyRs, which indicated that all three hu333 mAbs above would have significantly reduced effector functions mediated by FcyRs.
  • TABLE 16
    ELISA-based bindings of IgG1 variants to FcyRs
    FcγRs hu333-4A2-IgG1wt hu333-4A2-IgG1mc hu333-4B2-IgG1mc hu333-4B2-IgG1mf
    Chemilumi nescence Relative binding Chemilumi nescence Relative binding Chemilumi nescence Relative binding Chemilumi nescence Relative binding
    FcγRI 42714 100% 136 0.3% 230 0.5% 175 0.4%
    FcγRIIAH131 54599 100% 61 0.1% 64 0.1% 82 0.1%
    FcγRIIAR131 50189 100% 138 0.3% 114 0.2% 158 0.3%
    FcγRIIIAF158 36402 100% 262 0.7% 252 0.7% 279 0.8%
    FcγRIIIAV158 57805 100% 323 0.6% 246 0.4% 225 0.4%
    FcγRIIB 136565 100% 2900 2.1% 2715 2.0% 2069 1.5%
    FcγRIIIB 40352 100% 2256 5.6% 2009 5.0% 1751 4.3%
    Chemiluminescence signal was read using a PheraStar FS microplate reader (BMG Labtech); For each FcyR, relative binding (percent) was normalized by the chemilumenescence signal of hu333-4A2-lgG1wt binding.
  • FACS Based FcγR Binding Assays
  • Bindings of humanized anti-PD-L1 in various IgG1 formats (wt, IgG1mc, IgG1mf, SEQ. NO. 27-29) to FcyRs were also determined by flow cytometry. In brief, a series of stable HEK293 transfectants expressing human FcyRs were established. These stable cell lines expressed FcyRI, FcγRIIAH131, FcγRIIAR131, FcγRIIB, FcγRIIIAF158 or FcγRIIIAV158, respectively. Multi-subunit FcyRs (i.e., FcγRI and FcγRIIIA) were co-expressed with FcRy subunit. A secondary antibody (goat anti-human IgG F(ab′)2-Alexa Fluor 488, Cat. No. 109-546-097, Jackson ImmunoResearch, West Grove, PA, USA) was used to detect the binding of monomeric anti-PD-L1 mAbs with the IgG1 variants (Table 17) to FcγR expressing HEK293 cells. As expected, hu333-4A2 in IgG1wt format (hu333-4A2-IgG1wt) had strong binding signals (MFI) to FcyRI, FcγRIIAH131 and FcγRIIIAV158, and weak but significant binding signals to FcγRIIAR131, FeγRIIB and FcγRIIIAF158 (Table 17). The modified IgG1 variants (hu333-4A2-1gGlmc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf, SEQ. NO. 30-33) had significantly reduced binding signals which were close to backgrounds.
  • TABLE 17
    Binding strength (MFI*) of monomeric IgG1 variants to FcyRs determined by FACS
    mAbs FcγRI FcγRIIAH131 FcγRIIAR131 FeγRIIB FcγRIIIAF158 FcγRIIIAV158
    hu333-4A2-IgG1wt 1169.42 40.52 15.14 19.00 29.45 91.65
    hu333-4A2-IgG1mc 4.29 5.78 3.80 3.71 5.20 3.87
    hu333-4B2-IgG1mc 4.78 6.16 3.64 4.49 5.42 4.14
    hu333-4B2-IgG1mf 4.56 6.12 3.99 3.73 5.09 3.91
    MFI: Mean flourescence intensity from FACS analysis
  • It has been shown that antibodies bind to FcγRs with much bigger strength in the forms of immune-complexes, which is due to the multivalent effect (Bruhns et. al. 2009 Blood 113:3716-3725). Such bindings are thought to be more relevant under physiological condition, as the binding strength between monomeric γFc and most of the FcγRs is very weak. Human immune system also takes advantage of this mechanism to avoid non-specific activation of FcγRs by monomeric IgG which are present at high levels in serum. In order to assess the bindings to FcyRs in the form of immune-complexes, 10 µg/mL of humanized 333 mAb as various IgG1 mutant forms were premixed with 3 µg/mL of biotin-PD-L1/His and 1.5 µg/mL of neutravidin (Cat. No. A-2666, Invitrogen) in FACS buffer to form the multivalent immune-complexes, before incubating with FcyR-expressing HEK293 cells. Goat anti-human IgG F(ab′)2-Alexa Fluor 488 (Cat. No. 109-546-097, Jackson ImmunoResearch) was used to detect the bindings. As shown in Table 18, hu333-4A2-IgG1wt in preformed immune-complex bound to the low affinity FcyRs (FcyRIIA, FeγRIIB, and FcyRIIIA) with much better strength than monomeric IgG1 does (Table 18 data vs. Table 17 data). And again, the anti-PD-L1 mAbs in selected IgG1 mutants (hu333-4A2-IgG1mc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf, SEQ. NO. 30-33) had significantly reduced binding signals which were close to backgrounds. Taken together, the humanized 333 in modified IgG1 formats had very little bindings to FcγRs, therefore they should have little FcyRs-mediated effector functions under physiological conditions.
  • TABLE 18
    Binding strength (MFI*) of IgG1 variants as immune-complexes to FcyRs determined by FACS
    mAbs FcγRI FcγRIIAH131 FcγRIIAR131 FeγRIIB FcγRIIIAF158 FcγRIIIAV158
    hu333-4A2-IgG1wt 3261.14 599.41 159.32 539.42 74.15 308.98
    hu333-4A2-IgG1mc 7.00 7.53 5.38 5.05 6.44 5.43
    hu333-4B2-IgG1mc 7.01 8.09 5.28 5.03 6.81 4.89
    hu333-4B2-IgG1mf 7.11 7.27 4.92 5.00 6.76 4.83
    MFI: Mean flourescence intensity from FACS analysis
  • The ELISA-based C1q binding assay was done by conventional ELISA method with minor modification. Briefly, indicated amounts of the humanized 333 antibodies fused to either wild type or modified IgG1 constant regions were coated onto the Maxisorp ELISA plate. After blocking and washing, the wells were incubated with 2 µg/mL of human C1q (Cat. No. A400, Quidel, San Diego, USA) at room temperature for 2 hours. After washing, the bound C1q was detected using a murine monoclonal antibody against human C1q (Cat. No. A201, Quidel) and HRP conjugated anti-murine IgG (Cat. No. A0168, Sigma, Shanghai, China). As shown in FIG. 8 , in contrast to hu333-4A2-IgG1wt, there were no detectable C1q bindings for the three mAbs with modified IgG1 Fc variants including hu333-4A2-IgG1mc (SEQ. NO. 30 and 32), hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33). The data indicated that humanized 333 antibodies either in IgG1mc (SEQ. NO. 28) or in IgG1mf format (SEQ. NO. 29) would have very low or null CDC effector function.
  • ADCC
  • Classical antibody-dependent cellular cytotoxicity (ADCC) involves activation of NK cells by antibodies engaging to FcyRIIIA (CD16). To test whether humanized anti-PD-L1 antibodies fused to selected human IgG1 variants induce ADCC, NK92MI/CD16V cells, which were generated from NK92MI cells (Cat. No. CRL-2408, ATCC) by co-transducing expression plasmids containing CD16 (V158 allele) and FcRy genes, were used as effector cells, and PD-L1-expressing HEK293 cell line, HEK293/PD-L1, was used as target cells. The effector cells (105 cells/well) were co-cultured with target cells (104 cells/well, E:T=10) in 96-well V-bottom plates in the presence of hu333-IgG1 variants (0.0001-1 µg/ml) for 5h. Cytotoxicity of NK92MI/CD16 cells exerted against HEK293/PD-L1 cells was determined by lactate dehydrogenase (LDH) release assay using the CytoTox 96 Non-Radioactive Cytotoxicity Assay kit (Promega, Madison, WI). Specific lysis was determined by the following equation.
  • % Specific lysis = Experimental Effector Spotaneous Target Spotaneous Target Maximum Target Spotaneous × 100
  • Consistent with the fact that hu333 in IgG1mc and IgG1mf formats had no or significantly reduced bindings to FcyRIIIA (see above section), the ADCC assays showed that both hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) had only base level of ADCC. In contrast, 333-4A2-IgG1wt with wild type IgG1 Fc induced 20% specific cell lysis at the concentration of 1 µg/mL (FIG. 9 ).
  • CDC
  • Human IgG1 antibodies, in general, induce significant complement-dependent cytotoxicity (CDC) via classical pathway. Whether the humanized anti-PD-L1 antibodies in selected IgG1 mutant formats (IgG1mc and IgG1mf) trigger CDC was evaluated using a PD-L1-expressing B cell line, Daudi/PD-L1, and fresh human serum from healthy donors, which contains all necessary components for CDC. Cell lysis by CDC was determined by Celltiter glo assay kits (Promega). In brief, Daudi/PD-L1 cells (2×104 cells/well) were incubated in serum-free RPMI1640 (Invitrogen) with anti-PD-L1 Abs (0.001-10 µg/mL) at 37° C. for 15 minutes before adding normal human serum to the final concentration of 16.6% in 96-well flat-bottom plates in a total volume of 120 µL. After overnight incubation at 37° C., cells were lysed and assayed for ATP concentration. Anti-CD20 mAb Rituximab (Roche) was used as a positive control as Daudi cells constitutively express CD20. The amount of ATP is directly proportional to the number of cells present in culture. Fluorescence was read using a 96-well fluorometer (PHERA Star FS, BMG LABTECH). The results are expressed in relative fluoresence units (RFU) that are proportional to the number of viable cells. The percent CDC activity was calculated as follows: % CDC activity = [(RFU test - RFU background) / (RFU at total cell lysis - RFU background)] × 100. As shown in FIG. 10 , Rituximab induced robust CDC in CD20+ Daudi/PD-L1 cells. In contrast, both hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) showed no CDC. Ab 333-4A2-IgG1wt with wild type IgG1 Fc demonstrated low but above base level of CDC activities especially at concentrations of above 0.3 µg/mL. These data were consistent with the fact that IgG1mc and IgG1mf Fc formats had very little or significantly reduced bindings to complement component C1q (see the previous section).
  • Example 6. Functional Activities of Humanized mAb 333 in Modified IgG1 Formats
  • The three humanized mAbs in modified IgG1 formats described above were characterized in cell-based binding assays and functional assessment. Table 19 summarized the study results about hu333-4A2-IgG1mc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf (SEQ. NO. 30-33).
  • FACS binding analysis was performed as described in previous sections. Serial dilutions of antibodies were incubated with HEK293/PD-L1 cells and the bindings were detected using the Goat anti-human IgG F(ab′)2-Alexa Fluor 488 (Cat. No. 109-546-097, Jackson ImmunoResearch). Dose-dependent binding activities were observed for the selected mAbs to native PD-L1 protein expressed on surface of HEK293 cells. As shown in Table 19, hu333-4A2-IgGlmc, hu333-4B2-IgGlmc and hu333-4B2-IgG1mf showed similar dose-dependent binding activities to the HEK293/PD-L1 cells with EC50 (effective concentration at 50% activity) around 0.1 µg/mL.
  • FACS based competition assays was performed as described earlier. The results shown in Table 19 demonstrated that hu333-4A2-IgGlmc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf compete off both PD-⅟Fc binding (IC50S of 0.167- 0.174 µg/mL) and CD80/Fc binding (IC50 s of 0.078-0.118 µg/mL) to HEK293/PD-L1 cells almost equally well.
  • The functionalities of the purified anti-PD-L1 mAbs were assessed in the HuT78/PD-1 and HEK293/OS8/PD-L1 co-culture system as described in previous section. As shown in Table 19, the humanized 333 mAbs were potent antagonists of PD-L1/PD-1 signaling in this co-culture system, and induced increased IL-2 secretions. Consistent with the result of FACS-based competition assay, hu333-4A2-IgG1mc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf showed similar potencies in this assay with very close EC50 (0.075-0.087 µg/mL) and maximum induction of IL-2 levels (287-300 pg/mL).
  • The functionalities of the purified anti-PD-L1 mAbs were also assessed in the reversed signaling system in which HuT78/P3Z and HEK293/PD-L1 were co-cultured as described. Consistently, the humanized 333 mAbs were potent inhibitor of PD-L1/P3Z signaling in this co-culture system, and inhibited IL-2 secretions induced by PD-L1/P3Z engagement. And again, hu333-4A2-IgG1mc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf showed similar potencies in the assay, as shown by similar IC50 s (0.037-0.045 µg/mL) and maximum inhibition levels (99%) (Table 19).
  • TABLE 19
    Activities of hu333-4A2-IgG1mc,hu333-4B2-IgG1mc and hu333-4B2-IgG1mf in cell based assays
    Assay/Parameter hu333-4A2-IgG1mc hu333-4B2-IgG1mc hu333-4B2-IgG1mf
    FACS binding EC50 (µg/ml) 0.098 0.092 0.102
    Top MFI* 1363 1391 1342
    PD-1 binding competition (FACS) IC50 (µg/ml) 0.172 0.167 0.174
    Max inhibition 100% 100% 100%
    CD80 binding competition (FACS) IC50 (µg/ml) 0.078 0.103 0.118
    Max inhibition 100% 100% 100%
    IL-2 release in HuT78/PD-1# EC50 (µg/ml) 0.087 0.084 0.075
    Top line (pg/ml) 299 300 287
    IL-2 release in HuT78/P3Z§ IC50 (µg/ml) 0.045 0.039 0.037
    Max inhibition 99% 99% 99%
    *MFI: Mean fluorescence intensity from FACS analysis. #IL-2 release in HuT78/PD-1: IL-2 release induced by the Fabs in HuT78/PD-1 cells co-cultured with HEK293/OS8/PD-L1 cells. §IL-2 release in HuT78/P3Z: IL-2 release induced by the Fabs in HuT78/P3Z cells co-cultured with HEK293/PD-L1 cells
  • To verify if the humanized 333 antibodies also exert functional effect on primary human immune cells, we assayed the antibody function using freshly isolated peripheral blood mononuclear cells (PBMCs), which are mainly consisted of T-cells (50-70%), B-cells and NK cells (15-30%), and monocytes (2-10%). Human PBMCs were isolated from healthy donors by density gradient centrifugation using ficoll lymphocyte separation medium (Histopaque-1077; Cat. No. 10771, Sigma) according to manufacturer’s instruction. The human blood collections were done followed the Internal Procedure of BeiGene. PBMCs were then stimulated with 40 ng/mL of anti-CD3 mAb OKT3 (Cat. No. 16-0037, eBioscience, CA) for 3 days prior to the assay. To mimic the response of pre-activated T cells to PD-L1 expressing tumor cells upon engagement of TCR/CD3 complex, PBMCs (1×104 cells) were co-cultured with HEK293/OS8/PD-L1 cells (3×104 cells) in each well of 96-well flat-bottom plates. Indicated concentrations of anti-PD-L1 antibodies were added to the culture. After 15-18 hours of co-culture, culture supernatants were assayed for IFN-y level by ELISA using Ready-Set-Go! ELISA kits (Cat. No. 88-7316, eBiosciences), which is the most prominent indicator of T-cell activation, as well as of other immune cell activation (Thakur et. al. 2012 Vaccine 30:4907-4920). As shown in FIG. 11A and FIG. 11B, the presence of mAb hu333-4A2-IgG1mc or hu333-4B2-IgG1mf in the co-culture of pre-activated PBMCs and HEK293/OS8/PD-L1 cells resulted in increasing IFN-y production in a dose-dependent manner. As a control, huIgG had no such effect of stimulating IFN-y secretion. The potencies of hu333-4A2-IgG1mc and hu333-4B2-IgG1mf were comparable to the parental murine antibody mu333-IgG. Although the base level of IFN-y without antibody treatment varied among different donors, the increase of IFN-y secretion in PBMCs treated by hu333-4A2-IgG1mc, hu333-4B2-IgG1mf and mu333-IgG was statistically significant in the range of 0.01 to 10 µg/mL of antibody treatment (about 5-8 fold induction at 10 µg/mL depending on the donor).
  • Taken together, these data demonstrated that hu333-4A2-IgG1mc, hu333-4B2-IgGlmc and hu333-4B2-IgG1mf were potent antagonists blocking PD-L1/PD-1 interactions and downstream signaling in all the cell line and primary immune cell-based assays. They were very similar in their functional activities and potencies, as they were very similar in sequences (only minor difference in framework regions), shared identical binding epitope and had very similar binding affinities and specificity (see below section).
  • Example 7. Binding Affinity and Specificity of Humanized Anti-PD-L1 mAbs Binding Specificity of Anti-PD-L1 mAbs to PD-L1 Proteins From Different Species
  • The binding specificity was studied for the mAbs hu333 (hu333-4A2-IgG1mc, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf) using human, cynomolgus monkey (Macaca fascicularis) and mouse (Mus musculus) PD-L1 as target proteins. The monkey PD-L1/His and murine PD-L1/His were expressed and purified in a similar way to the human PD-L1/His as described earlier. Y1 was a reference functional anti-PD-L1 mAb which was synthesized according to a published patent (US 2010/0203056 A1) and fused to human IgG1mc variant. The synthesized full length mAb was named as Y1-IgG1mc. The ELISA assay was performed essentially in the same way as described in the previous section. Briefly, 200 ng of PD-L1 protein was coated on each well of Nunc MaxiSorp ELISA plate (Cat. No. 442404, Nunc, Thermo Fisher). After washing and blocking, indicated concentrations of anti-PD-L1 mAbs were added and incubated at room temperature for 1 hour. After washing, the bound anti-PD-L1 mAbs were detected using a HRP-conjugated goat anti-human Fc antibody (Cat. No. A0170, Sigma). As shown in FIG. 12A, FIG. 12B, and FIG. 12C, hu333-4A2-IgG1mc (SEQ. NO. 30 and 32), hu333-4B2-IgG1mc (SEQ. NO. 31 and 32) and hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) bound to human and monkey PD-L1 in a dose-dependent manner, but not to murine PD-L1, which was consistent with the fact that the original mu333 was generated from mouse immunized with human PD-L1/Fc and that human and monkey PD-1 shares high degree of sequence homology (96%). In contrast, Y1-IgG1mc bound to all PD-L1 proteins from human, monkey and mouse.
  • Affinity Determination of Humanized Anti-PD-L1 Fabs by SPR
  • Hu333-4A2 (SEQ. NO. 21 and 23), hu333-4B2 (SEQ. NO. 22 and 23) and the reference antibody Y1 were constructed as human IgG1 Fab format in which the Vh and Vk were fused to the N-terminus of human IgG1-CH1 and constant region of kappa chain, respectively. The IgG1-CH1 was fused to a C-terminal His6 tag to facilitate purification. Expression and purification of recombinant Fabs were performed as described in the previous section.
  • For affinity determinations of anti-PD-L1 Fabs, SPR assays were conducted using BIAcore™ T-200 instrument (GE Life Sciences, Shanghai, China) as described earlier. Association rates (kon) and dissociation rates (koff) were calculated using the one-to-one Langmuir binding model (BIA Evaluation Software, GE Life Sciences). The equilibrium dissociation constant (Kd) was calculated as the ratio koff/kon.
  • The SPR-determined binding affinities of anti-PD-L1 Fabs were listed in Table 20. Hu333-4A2 and hu333-4B2 Fabs bind to human PD-L1 with higher affinities than Y1 Fab does, which was indicated by the faster kon, slower koff and much smaller Kdvalue. The hu333-4A2 and hu333-4B2 Fabs bind to monkey PD-L1 almost equally well as their binding to human PD-L1. In contrast, Y1 Fab binds to the monkey PD-L1with about 100-fold lower affinity than its binding to human PD-L1 (Kd of 0.18 nM to human PD-L1 and Kd of 16.2 nM to monkey PD-L1).
  • TABLE 20
    Affinities of anti-PD-L1 Fabs to human and monkey PD-L1 determined by SPR
    Binding kinetics/parameter hu333-4A2 Fab hu333-4B2 Fab Y1 Fab
    Affinity to human PD-L1 kon (M-1s-1) 3.88 × 106 3.78 × 106 4.97 × 105
    koff (s-1) * 1.03 × 10-5 1.32 × 10-5 9.00 × 10-5
    Kd* 2.65 pM 3.50 pM 0.18 nM
    Affinity to monkey PD-L1 kon (M-1s-1) 3.19 × 106 3.20 × 106 2.35 × 105
    koff (S-1) * 0.93 × 10-5 0.80 × 10-5 3.79 × 10-3
    Kd * 2.91 pM 2.5 pM 16.2 nM
    * koff might be too slow to be accurately measured during the 5-15 min dissociation time in the SPR experiment. Therefore, the affinity might be too strong to be accurately determined for hu333-4A2 Fab and hu333-4B2 Fab using current instrument/experiment setting.
  • Epitope Mapping of Anti-PD-L1 mAbs
  • The previous reports about the crystal structures of PD-⅟PD-L1 complexes shed light on the critical amino acid (AA) residues of PD-L1 that directly interact with receptor PD-1 (Zhang et. al. 2004 Immunity 20:337-347; Lin et. al. 2008 PNAS 105:3011-3016; Lazar-Molnar et. al. 2008 PNAS 105:10483-10488). Through point mutation analysis, eight AA residues in PD-L1 sequence were identified being required for its binding to PD-1. Based on the information from the structure-guided mutation analysis, we hypothesized that most effective way for the functional mAbs to block PD-L1 mediated signaling is to compete with PD-1 by binding to the eight critical AA residues, therefore, occupying the binding epitopes required for its binding to PD-1 receptor. To explore the hypothesis and to understand the mechanism of action by functional PD-L1 mAbs, we made eight mutants of PD-L1 by replacing each of the eight critical AAs to Ala, individually, i.e. F19A, I54A, R113A, M115A, D122A, Y123A, K124A and R125A (AA residue numbering based on Lin et. al. 2008 PNAS 105:3011-3016). The wild-type PD-L1/His (FIG. 1 ) was used as template for rolling-circle mutagenesis using Fast Mutagenesis kit (Cat. No. FM111, Transgen Biotech, Beijing, China). All mutants were sub-cloned in our pcDNA-based expression vectors, and verified by sequencing. The mutant and wild-type PD-L1/His proteins were expressed by transient transfection as described in previous sections. The conditioned media (CM) were collected after 4 to 6 days of culture, and analyzed by Western blot to verify the PD-L1/His protein expression in terms of quality and quantity. The supernatants (CM), after clearing cell debris, were directly used in ELISA analysis or Western blot for epitope-mapping.
  • ELISA assays using the wild-type and mutant PD-L1/His were performed to study the binding epitopes of the anti-PD-L1 mAbs. For comparison of antibody binding epitopes, several murine mAbs from us and one reference antibody Y1-IgG1 (adapted from US 2010/0203056 A1 and fused to human IgG1kappa constant regions) were included in the study. Equal volume of CM containing wild type or mutant PD-L1/His was coated in 96-well plate for all mAbs in the same ELISA assay. The ELISA results were normalized using the mean ELISA readings of wild type PD-L1 binding signals as the standard. ELISA binding signals to a specific mutant PD-L1 were further normalized against the highest antibody binding read-out (set as 100%) to the specific mutant PD-L1 to even out expression variations between PD-L1 mutants. For convenience of data analysis, when a mAb’s ELISA binding signal for a specific PD-L1 mutant dropped below 50% relative to wild type PD-L1, it was defined that the binding function is significantly impaired due to the corresponding amino acid mutation. Likewise, if a mAb’s ELISA binding signal for a specific mutant dropped below 25%, it was defined to be very significant.
  • As shown in FIG. 13A, amino acid residues R113 in PD-L1 is critical for hu333-4B2-IgG1 binding to PD-L1, whose mutation significantly impaired the hu333-4B2-IgG1 binding to PD-L1. On the other hand, the reference antibody Y1-IgG1 had distinctive binding epitopes. F19, I54, D122 and Y123 were all significant epitopes for its binding besides R113 (FIG. 13B). Different signatures of binding epitopes were also observed in other anti-PD-L1 mAbs from our study. The data from Western blot for both hu333-4B2-IgG1 and Y1-IgG1 antibodies confirmed these results although the antigen proteins were denatured.
  • Apart from the above key binding epitope mutations, we also made the mutation D26A. ELISA and Western blot results showed the mutation D26A in PD-L1 significantly inhibited the binding activities of all functional anti-PD-L1 mAbs including mAbs mu333, hu333-4B2-IgG1 and Y1-IgG1, but not inhibited the binding of non-functional antibodies, such as mu260 (FIG. 13C). As summarized in Table 21, similar to mu333 and its derivative humanization mAbs, hu333-4B2 binds to two key amino acid residues (epitopes) of human PD-L1, D26 and R113; in contrast, mAb Y1 binds to at least six amino acid residues.
  • TABLE 21
    Summary of epitope-mapping results
    mAb F19A D26A 154A R113A M115A D122A Y123A K124A R125A
    hu333-4B2-IgG1 ** ***
    Y1-IgG1 ** ** ** *** ** **
    ** Mutations inhibited > 50% binding comparing to wildtype PD-L1. *** Mutations inhibited > 75% binding comparing to wildtype PD-L1.
  • Through the epitope mapping study, we have demonstrated that anti-PD-L1 mAbs are capable of binding to different epitope signatures through molecular recognition, which might have profound impact on binding affinity, binding specificity and functional activity, e.g. hu333-4A2 and hu333-4B2 can only bind to human PD-L1 (FIG. 12A), but not to mouse PD-L1 (FIG. 12C); in contrast, Y1 binds to both human and mouse PD-L1 (FIG. 12A and FIG. 12C) although human and mouse PD-L1 have 26% sequence divergence.
  • Non-Specific Binding to Human Serum Proteins
  • In order to check whether mAb mu333 has non-specific binding to human serum proteins, ELISA study was performed using 96-well Nunc Maxisorp ELISA plates coated with 5% human serum (from healthy donors) and various concentrations of PD-L1/His antigen as indicated in FIG. 14A, x-axis. Same amount of murine PD-L1 Abs or a chimeric Y1 (named as Y1-muIgG1), which was made of Y1 variable domains fused to murine IgG1kappa constant regions) were added in the ELISA reaction, and the bindings were detected with anti-mouse Fc-HRP (Cat. No. A9309, Sigma). The PBS buffer without any antibody was included as negative control (FIG. 14A and FIG. 14B).
  • As showed in FIG. 14A and FIG. 14B, the baseline O.D. reading (the reading at very low antigen concentration) for mu333 was almost identical to the negative control, while the baseline O.D. reading for Y1-muIgG1 is 4 folds higher than the negative control (PBS), indicating a differential property between mu333 and YI-mIgG1 in binding selectivity. The humanized mAb hu333-4B2-IgG1mc and hu333-4B2-IgG1mf were also assessed for non-specific binding in similar assay method using fetal bovine serum (FBS) instead of human serum. Reminiscent to its parental mouse hybridoma mAb, hu333-4B2-IgG1mc and hu333-4B2-IgG1mf had no binding to FBS.
  • Example 8. Pharmacokinetics Pharmacokinetics of hu333-4B2-IgG1mf in Mice
  • All animal studies were performed following BeiGene Animal Care and Use Procedure. Ten to twelve week-old female Balb/c nude mice (18-25 g) were used to study the pharmacokinetics of the humanized mAb hu333-4B2-IgG1mf (SEQ. NO. 32 and 33). Mice were dosed with 10 mg/kg of mAb hu333-4B2-IgG1mf either as a single intravenous (i.v.) or subcutaneous (s.c.) injection. Intravenous injections were administered via a tail vein, and subcutaneous injections were administered in the flank. In each injection group, mice were separated into different subgroups and in each subgroup blood sera were collected at certain time points. For i.v. injection group, serum was harvested 2 days predose, and postdose at 15 min, 30 min, 60 min, 90 min, 6 h, 24 h and once on days 2, 3, 4, 5, 7, 10, 14, 21 and 28. For s.c. injection group, serum was harvested 2 days predose, and postdose at 1.5 h, 6 h, 24 h and once on days 2, 3, 4, 5, 7, 10, 14, 21, and 28.
  • Serum level of hu333-4B2-IgG1mf was determined by ELISA using human PD-L1/His protein. Briefly, Nunc MaxiSorp ELISA plates (Cat. No. 442404, Nunc, Thermo Fisher) were coated overnight at 4° C. with 100 µL per well of 3 µg/mL human PD-L1/His protein. Plates were blocked with 3% bovine serum albumin, 0.05% Tween 20 in PBS (blocking buffer) at room temperature for 1 hour. After washing, serially diluted serum samples and purified hu333-4B2-IgG1mf standards were added and incubated at room temperature for 1 hour. After washing, the bound hu333-4B2-IgG1mf was detected using a HRP-conjugated goat anti-human Fc antibody (Cat. No. A0170, Sigma) and color developed using TMB substrate (Cat. No. T0440, Sigma). A standard curve was fit using nonlinear regression and the serum concentrations of hu333-4B2-IgG1mf were deduced from the standard curve and dilution factors.
  • The serum concentrations of hu333-4B2-IgG1mf versus time data were analyzed using the non-compartment model for the i.v. and s.c. doses (WinNonlin, Pharsight). The clearance, volume of distribution, half-lives, mean residence time and bioavailability were deduced from WinNonlin data fitting.
  • The pharmacokinetics of hu333-4B2-IgG1mf in mice was summarized in Table 22. After i.v. administration, hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) concentrations were cleared from the serum in a biphasic manner. The terminal half-life was about 10-14 days. After an i.v. dose of 10 mg/kg, the clearance was 7.9 mL/day/kg in mice. After s.c. administration, peak concentrations of hu333-4B2-IgG1mf in the serum as approximately 30-50% of that noted after i.v. administration of the same dose. Comparison of the AUC after the 10 mg/kg s.c. and i.v. dose indicated a bioavailability of 90%. All these PK parameters were close to those of typical humanized monoclonal antibodies, indicating that hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) had good in vivo stability in mice.
  • Pharmacokinetics of hu333-4B2-IgGlmc in Cynomolgus Monkey
  • The pharmacokinetics of hu333-4B2-IgGlmc (SEQ. NO. 31 and 32) was studied in cynomolgus monkeys. As humanized 333 bound to human and monkey PD-L1 with almost identical affinities, the pharmacokinetic profile in cynomolgus monkeys should be very informative and scalable to predict the pharmacokinetic profile in humans. The drug administrations and blood serum collections were done at 3D BioOptima Co. Ltd (Suzhou, China) following 3D BioOptima’s Animal Care and Use Procedure. Briefly, two 3-5 year-old male monkeys were dosed with 10 mg/kg of mAb hu333-4B2-IgG1mc as a single intravenous (i.v.) dose. Blood samples (~1 mL) were collected at 2 days pre-dosing, 5 min, 30 min, 2 h, 6 h, 12 h, 24 h, 36 h, and 2, 3, 5, 7, 10, 15, 22, 30 days post-dosing via cephalic vein into tubes.
  • ELISA based bioanalyses and pharmacokinetic analyses were performed essentially as described above. At each time point, the averaged serum concentration from 2 monkeys was used for fitting except for the time points of 22 and 30 days post dose, where the data from only one monkey were used, as another monkey showed accelerated clearance and undetectable hu333-4B2-IgG1mc serum levels, presumably due to a monkey anti-drug immune response, at these two time points. The serum concentration of hu333-4B2-IgG1mc versus time data were analyzed using the non-compartment model for the i.v. dose.
  • TABLE 22
    Pharmacokinetics of hu333-4B2-IgG1mc after s.c./i.v. administration in nude mice
    Parameters
    10 mg/kg i.v. 10 mg/kg s.c.
    Cmax (mg/mL) 250 70.6
    Vss/W (mL/kg) 105 N/A
    CL/W (mL/day/kg) 7.9 N/A
    T½ terminal (day) 9.6 11.3
    AUC0-inf (mg/mL*day) 1266 1147
    MRT (day) 13.3 16.5
    Bioavailability (%) N/A 90%
    Cmax, maximum observed concentration; Vss/W, steady-state volume of distribution; CL/W, serum clearance; T½, half life; AUC, area under the curve; MRT, mean residence time in the body; N/A: not applicable
  • TABLE 23
    Pharmacokinetics of hu333-4B2-IgG1mc after i.v. administration in cynomolgus monkeys
    Parameters
    10 mg/kg i.v.
    Cmax (mg/mL) 283.4
    Vss/W (mL/kg) 80
    CL/W (mL/day/kg) 6.4
    T½ terminal (day) 9
    AUC0-inf (mg/mL*day) 1527
    MRT (day) 11.7
    Cmax, maximum observed concentration; Vss/W, steady-state volume of distribution; CL/W, serum clearance; T½, half life; AUC, area under the curve; MRT, mean residences time in the body
  • The pharmacokinetics of hu333-4B2-IgG1mc in cynomolgus monkeys was summarized in Table 23. After i.v. administration, hu333-4B2-IgG1mc concentrations were cleared from the sera in a biphasic manner. The terminal half-life was about 9 days. After an i.v. dose of 10 mg/kg, the clearance was 6.4 mL/day/kg in cynomolgus monkeys. After i.v. administration, peak concentration of hu333-4B2-IgG1mc was 283 µg/mL at 5 min post dose. These PK parameters indicated that hu333-4B2-IgG1mc had normal pharmacokinetic profile in cynomolgus monkeys, which predicted normal pharmacokinetic behavior in humans (Deng et. al. 2011 mAbs 3:61-66).
  • Example 9. Humanized Anti-PD-L1 mAb Inhibits Tumor Growth in a Mouse Xenograft Cancer Model In Vivo
  • The T-cell line and PBMC-based experiments indicated that the anti-PD-L1 mAb might work in mouse cancer models utilizing immune-compromised mice xenografted with human cancer cells, subsequently implanting human PBMCs and applying the mAb treatment to inhibit cancer cell growth in vivo. An allogeneic mouse cancer model was designed as follows. Female NOD/SCID mice (6-7 weeks) were pre-treated with cyclophosphamide. Human peripheral blood mononuclear cells (PBMCs) were isolated from blood of healthy human volunteer, mixed with A431 epidermoid carcinoma cells (Cat. No. CRL-1555 ATCC) and matrigel, and injected subcutaneously into the right front flank of the animals. Starting from day 0 after cell inoculation, animals were randomly assigned into 3 groups with 8 mice per group. Mice were treated twice weekly (BIW i.p.) with vehicle (PBS) or 10 mg/kg hu333-4B2-IgG1mf (SEQ. NO. 32 and 33) for 4 weeks. Individual animal body weight and tumor volume were recorded twice weekly, with mice being monitored daily for clinical signs of toxicity for the duration of the study. Tumor volumes were calculated using the following formula: [D × (d2)]/2, in which D represents the large diameter of the tumor, and d represents the small diameter.
  • As shown in FIG. 15 , in the anti-PD-L1 mAb (hu333-4B2-IgG1mf)-treated group, the A431 tumor growth was slower compared to that in the PBS-treated group. The results indicated that the anti-PD-L1 mAb described can activate human immune cells to inhibit tumor cells growth in the mouse in vivo cancer model, which is consistent with the in vitro experimental results described in previous sections.

Claims (21)

What is claimed is:
1-12. (canceled)
13. A mutant IgG1 Fc-containing molecule having a decreased binding affinity to one or more Fcγ receptors and/or human C1q protein as compared to wild type human IgG1, comprising a mutated human IgG1 constant region comprising (i) E233P, L234A, L235A and P329A mutations in wild type human IgG1, as defined by the EU numbering system, or (ii) E233P, L234A, L235A, G237A and P329A mutations in wild type human IgG1, as defined by the EU numbering system.
14. The mutant IgG1 Fc-containing molecule of claim 13, wherein the mutated human IgG1 constant region comprises E233P, L234A, L235A and P329A mutations in wild type human IgG1, as defined by the EU numbering system.
15. The mutant IgG1 Fc-containing molecule of claim 13, wherein the mutated human IgG1 constant region comprises E233P, L234A, L235A, G237A and P329A mutations in wild type human IgG1, as defined by the EU numbering system.
16. A mutant IgG1 Fc-containing molecule comprising the amino acid sequence of SEQ ID NO:28 or SEQ ID NO:29.
17. The mutant IgG1 Fc-containing molecule of claim 16, which comprises the amino acid sequence of SEQ ID NO:28.
18. The mutant IgG1 Fc-containing molecule of claim 16, which comprises the amino acid sequence of SEQ ID NO:29.
19. An antibody comprising a mutated human IgG1 constant region comprising (i) E233P, L234A, L235A and P329A mutations in wild type human IgG1, as defined by the EU numbering system, or (ii) E233P, L234A, L235A, G237A and P329A mutations in wild type human IgG1, as defined by the EU numbering system; wherein the antibody has a decreased binding affinity to one or more Fcγ receptors and/or human C1q protein as compared to wild type human IgG1.
20. The antibody of claim 19, wherein the mutated human IgG1 constant region comprises E233P, L234A, L235A and P329A mutations in wild type human IgG1, as defined by the EU numbering system.
21. The antibody of claim 19, wherein the mutated human IgG1 constant region comprises E233P, L234A, L235A, G237A and P329A mutations in wild type human IgG1, as defined by the EU numbering system.
22. An antibody comprising a mutated human IgG1 constant region comprising the amino acid sequence of SEQ ID NO:28 or SEQ ID NO:29.
23. The antibody of claim 22, which comprises the amino acid sequence of SEQ ID NO:28.
24. The antibody of claim 22, which comprises the amino acid sequence of SEQ ID NO:29.
25. The antibody of claim 19, wherein the antibody is an antibody that specifically binds to human PD-L1.
26. The antibody of claim 22, wherein the antibody is an antibody that specifically binds to human PD-L1.
27. A method of treating cancer in a subject in need thereof, the method comprising administering to the subject a monoclonal antibody which binds human PD-L1, wherein the IgG Fc region of the antibody is an IgG1 Fc region containing E233P, L234A, L235A amino acid mutations and at least one additional amino acid mutation, wherein the at least one additional mutation causes the antibody to exhibit reduced binding to at least one Fcγ receptor relative to Fc binding of a reference IgG1 antibody which binds human PD-L1 having E233P, L234A, L235A amino acid mutations and no other Fc region mutation, and wherein the numbering of the residues in the IgG1 Fc region is that of the EU numbering system.
28. The method of claim 27, wherein the at least one additional amino acid mutation in the IgG1 Fc region is P329A mutation, as defined by the EU numbering system.
29. The method of claim 27, wherein the at least one additional amino acid mutation in the IgG1 Fc region are G237A and P329A mutations, as defined by the EU numbering system.
30. A method of treating cancer in a subject in need thereof, the method comprising administering to the subject a monoclonal antibody which binds human PD-L1, wherein the IgG Fc region of the antibody comprises the amino acid sequence of SEQ ID NO:28 or SEQ ID NO:29.
31. The method of claim 30, wherein the IgG Fc region of the antibody comprises the amino acid sequence of SEQ ID NO:28.
32. The method of claim 30, wherein the IgG Fc region of the antibody comprises the amino acid sequence of SEQ ID NO:29.
US18/050,441 2014-07-03 2022-10-27 Anti-pd-l1 antibodies and their use as therapeutics and diagnostics Pending US20230279117A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/050,441 US20230279117A1 (en) 2014-07-03 2022-10-27 Anti-pd-l1 antibodies and their use as therapeutics and diagnostics

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
WOPCT/CN2014/081581 2014-07-03
CN2014081581 2014-07-03
PCT/CN2015/083066 WO2016000619A1 (en) 2014-07-03 2015-07-01 Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
US201615323153A 2016-12-30 2016-12-30
US16/704,738 US11512132B2 (en) 2014-07-03 2019-12-05 Anti-PD-L1 antibodies and their use as therapeutics and diagnostics
US18/050,441 US20230279117A1 (en) 2014-07-03 2022-10-27 Anti-pd-l1 antibodies and their use as therapeutics and diagnostics

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/704,738 Continuation US11512132B2 (en) 2014-07-03 2019-12-05 Anti-PD-L1 antibodies and their use as therapeutics and diagnostics

Publications (1)

Publication Number Publication Date
US20230279117A1 true US20230279117A1 (en) 2023-09-07

Family

ID=55018460

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/323,153 Active 2036-09-24 US10544225B2 (en) 2014-07-03 2015-07-01 Anti-PD-L1 antibodies and their use as therapeutics and diagnostics
US16/704,738 Active 2036-09-06 US11512132B2 (en) 2014-07-03 2019-12-05 Anti-PD-L1 antibodies and their use as therapeutics and diagnostics
US18/050,441 Pending US20230279117A1 (en) 2014-07-03 2022-10-27 Anti-pd-l1 antibodies and their use as therapeutics and diagnostics

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US15/323,153 Active 2036-09-24 US10544225B2 (en) 2014-07-03 2015-07-01 Anti-PD-L1 antibodies and their use as therapeutics and diagnostics
US16/704,738 Active 2036-09-06 US11512132B2 (en) 2014-07-03 2019-12-05 Anti-PD-L1 antibodies and their use as therapeutics and diagnostics

Country Status (7)

Country Link
US (3) US10544225B2 (en)
EP (1) EP3160505A4 (en)
JP (2) JP6526189B2 (en)
KR (2) KR102003754B1 (en)
CN (2) CN106604742B (en)
TW (2) TWI726608B (en)
WO (1) WO2016000619A1 (en)

Families Citing this family (195)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HRP20240240T1 (en) 2008-12-09 2024-04-26 F. Hoffmann - La Roche Ag Anti-pd-l1 antibodies and their use to enhance t-cell function
US9217040B2 (en) 2011-01-14 2015-12-22 The Regents Of The University Of California Therapeutic antibodies against ROR-1 protein and methods for use of same
WO2013013188A1 (en) 2011-07-21 2013-01-24 Tolero Pharmaceuticals, Inc. Heterocyclic protein kinase inhibitors
ME02855B (en) 2011-12-31 2018-04-20 Beigene Ltd Fused tetra or penta-cyclic dihydrodiazepinocarbazolones as parp inhibitors
AU2013359907B2 (en) 2012-12-11 2018-01-18 Albert Einstein College Of Medicine, Inc. Methods for high throughput receptor:ligand identification
CN107011441B (en) 2013-09-13 2020-12-01 百济神州(广州)生物科技有限公司 anti-PD 1 antibodies and their use as therapeutic and diagnostic agents
EP3049442A4 (en) 2013-09-26 2017-06-28 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
JOP20200096A1 (en) 2014-01-31 2017-06-16 Children’S Medical Center Corp Antibody molecules to tim-3 and uses thereof
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
EP3160505A4 (en) 2014-07-03 2018-01-24 BeiGene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
US10428146B2 (en) 2014-07-22 2019-10-01 Cb Therapeutics, Inc. Anti PD-1 antibodies
AU2015301126B2 (en) 2014-08-05 2021-03-11 Cb Therapeutics, Inc. Anti-PD-L1 antibodies
KR20170060042A (en) 2014-09-13 2017-05-31 노파르티스 아게 Combination therapies of alk inhibitors
MX2017004810A (en) 2014-10-14 2017-10-16 Novartis Ag Antibody molecules to pd-l1 and uses thereof.
IL292449B2 (en) 2015-03-13 2024-02-01 Cytomx Therapeutics Inc Nucleic acids encoding anti-pdl1 antibodie and methods of producing same
GB201506411D0 (en) 2015-04-15 2015-05-27 Bergenbio As Humanized anti-axl antibodies
AU2016271591A1 (en) 2015-05-29 2017-12-21 Agenus Inc. Anti-CTLA-4 antibodies and methods of use thereof
NZ739876A (en) 2015-08-25 2022-09-30 Beigene Ltd Process for preparing parp inhibitor, crystalline forms, and uses thereof
BR112018003186A2 (en) 2015-09-01 2018-09-25 Agenus Inc. anti-pd-1 antibodies and their methods of use
EP3195878A1 (en) 2016-01-22 2017-07-26 Werner Lubitz Bacterial ghosts for the treatment of cancer
JP6432121B2 (en) 2016-03-04 2018-12-05 シチュアン ケルン−バイオテック バイオファーマシューティカル カンパニー リミテッド PDL-1 antibody, pharmaceutical composition thereof and use thereof
WO2017196867A1 (en) 2016-05-09 2017-11-16 Igm Biosciences, Inc. Anti-pd-l1 antibodies
KR20190044029A (en) * 2016-05-18 2019-04-29 알버트 아인슈타인 컬리지 오브 메디슨, 인크. Variant PD-L1 polypeptides, T-cell modulated multimeric polypeptides and methods for their use
WO2017201210A1 (en) 2016-05-18 2017-11-23 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
MY195089A (en) 2016-05-27 2023-01-10 Agenus Inc Anti-Tim-3 Antibodies and Methods of use thereof
CN109414500B (en) * 2016-06-13 2022-02-25 奥美药业有限公司 PD-L1 specific monoclonal antibody for treatment and diagnosis
EP3468581A1 (en) 2016-06-13 2019-04-17 Torque Therapeutics, Inc. Methods and compositions for promoting immune cell function
CA3026474A1 (en) 2016-06-20 2017-12-28 Kymab Limited Anti-pd-l1 and il-2 cytokines
US9567399B1 (en) 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
WO2018029474A2 (en) 2016-08-09 2018-02-15 Kymab Limited Anti-icos antibodies
JP6993056B2 (en) 2016-07-05 2022-02-15 ベイジーン リミテッド Combination of PD-1 antagonist and RAF inhibitor for cancer treatment
KR102048477B1 (en) 2016-08-05 2019-11-26 주식회사 와이바이오로직스 Antibodies Against Programmed death-ligand 1 and Uses Thereof
CN116640214A (en) 2016-08-09 2023-08-25 科马布有限公司 Isolated antibodies and uses thereof
IL264784B (en) 2016-08-16 2022-07-01 Beigene Ltd Crystalline form of (s)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo(1,5-a)pyrimidine-3-carboxamide, preparation, and uses thereof
CA3034326A1 (en) 2016-08-19 2018-02-22 Beigene, Ltd. Use of a combination comprising a btk inhibitor for treating cancers
SG11201901548SA (en) * 2016-08-26 2019-03-28 Beigene Ltd Anti-tim-3 antibodies and use thereof
EP3519051B1 (en) 2016-09-27 2021-09-22 Beigene, Ltd. Treatment of cancers using combination comprising parp inhibitors
CN117586403A (en) 2016-10-11 2024-02-23 艾吉纳斯公司 anti-LAG-3 antibodies and methods of use thereof
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
US11279694B2 (en) 2016-11-18 2022-03-22 Sumitomo Dainippon Pharma Oncology, Inc. Alvocidib prodrugs and their use as protein kinase inhibitors
EA201991383A1 (en) 2016-12-07 2019-12-30 Эйдженус Инк. ANTIBODIES AGAINST CTLA-4 AND WAYS OF THEIR APPLICATION
CN110300599A (en) 2016-12-07 2019-10-01 艾吉纳斯公司 Antibody and its application method
EP3554535A4 (en) 2016-12-14 2020-10-21 Janssen Biotech, Inc. Pd-l1 binding fibronectin type iii domains
WO2018111978A1 (en) 2016-12-14 2018-06-21 Janssen Biotech, Inc. Cd137 binding fibronectin type iii domains
EP3554562A4 (en) 2016-12-14 2020-11-04 Janssen Biotech, Inc. Cd8a-binding fibronectin type iii domains
IL297617B2 (en) 2016-12-22 2023-11-01 Cue Biopharma Inc T-cell modulatory multimeric polypeptides and methods of use thereof
US11851471B2 (en) 2017-01-09 2023-12-26 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
EP3568412A2 (en) 2017-01-13 2019-11-20 Agenus Inc. T cell receptors that bind to ny-eso-1 and methods of use thereof
EP3573989A4 (en) 2017-01-25 2020-11-18 Beigene, Ltd. Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
IL268836B2 (en) 2017-02-24 2024-04-01 Macrogenics Inc Bispecific binding molecules that are capable of binding cd137 and tumor antigens, and uses thereof
CN115433187B (en) 2017-02-28 2023-10-27 百济神州(苏州)生物科技有限公司 Crystalline form composition of salt of fused tetracyclic or pentacyclic dihydro-diazepin carbazolone and use thereof
JP2020514375A (en) 2017-03-15 2020-05-21 キュー バイオファーマ, インコーポレイテッド Methods for modulating the immune response
EP3609921A2 (en) 2017-04-13 2020-02-19 Agenus Inc. Anti-cd137 antibodies and methods of use thereof
CN110536703A (en) 2017-04-20 2019-12-03 Adc治疗有限公司 Use Anti-AXL antibodies-drug conjugate combination treatment
WO2018193104A1 (en) 2017-04-20 2018-10-25 Adc Therapeutics Sa Combination therapy with an anti-cd25 antibody-drug conjugate
AR111651A1 (en) 2017-04-28 2019-08-07 Novartis Ag CONJUGATES OF ANTIBODIES THAT INCLUDE TOLL TYPE RECEIVER AGONISTS AND COMBINATION THERAPIES
WO2018204363A1 (en) 2017-05-01 2018-11-08 Agenus Inc. Anti-tigit antibodies and methods of use thereof
JP2020522486A (en) 2017-06-01 2020-07-30 サイトメックス セラピューティクス インコーポレイテッド Activatable anti-PDL1 antibody and method of using the same
MX2019015042A (en) 2017-06-14 2020-08-06 Adc Therapeutics Sa Dosage regimes for the administration of an anti-cd19 adc.
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
GB201709808D0 (en) 2017-06-20 2017-08-02 Kymab Ltd Antibodies
SG11201912473PA (en) 2017-06-22 2020-01-30 Novartis Ag Antibody molecules to cd73 and uses thereof
WO2018235056A1 (en) 2017-06-22 2018-12-27 Novartis Ag Il-1beta binding antibodies for use in treating cancer
EP3642240A1 (en) 2017-06-22 2020-04-29 Novartis AG Antibody molecules to cd73 and uses thereof
JP2020524694A (en) 2017-06-22 2020-08-20 ノバルティス アーゲー IL-1β binding antibodies for use in the treatment of cancer
JP2020525411A (en) * 2017-06-26 2020-08-27 ベイジーン リミテッド Immunotherapy for hepatocellular carcinoma
MX2019015738A (en) 2017-06-27 2020-02-20 Novartis Ag Dosage regimens for anti-tim-3 antibodies and uses thereof.
WO2019018730A1 (en) 2017-07-20 2019-01-24 Novartis Ag Dosage regimens of anti-lag-3 antibodies and uses thereof
CA3071599A1 (en) 2017-08-07 2019-02-14 Amgen Inc. Treatment of triple negative breast cancer or colorectal cancer with liver metastases with an anti pd-l1 antibody and an oncolytic virus
WO2019046856A1 (en) 2017-09-04 2019-03-07 Agenus Inc. T cell receptors that bind to mixed lineage leukemia (mll)-specific phosphopeptides and methods of use thereof
US11497756B2 (en) 2017-09-12 2022-11-15 Sumitomo Pharma Oncology, Inc. Treatment regimen for cancers that are insensitive to BCL-2 inhibitors using the MCL-1 inhibitor alvocidib
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
TW201923089A (en) 2017-11-06 2019-06-16 美商建南德克公司 Diagnostic and therapeutic methods for cancer
CN111344305B (en) * 2017-11-27 2022-09-16 山东博安生物技术股份有限公司 Antibodies against PD-L1 and uses thereof
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors
CA3083949A1 (en) 2017-11-30 2020-06-06 Novartis Ag Bcma-targeting chimeric antigen receptor, and uses thereof
GB201721338D0 (en) 2017-12-19 2018-01-31 Kymab Ltd Anti-icos Antibodies
WO2019122882A1 (en) 2017-12-19 2019-06-27 Kymab Limited Bispecific antibody for icos and pd-l1
KR102311838B1 (en) * 2017-12-27 2021-10-14 주식회사 파멥신 Anti-PD-L1 Antibody and Use Thereof
CA3078849A1 (en) * 2017-12-28 2019-07-04 Nanjing Legend Biotech Co., Ltd. Antibodies and variants thereof against pd-l1
TW201930591A (en) 2018-01-08 2019-08-01 瑞士商諾華公司 Immune-enhancing RNAs for combination with chimeric antigen receptor therapy
WO2019139896A1 (en) 2018-01-09 2019-07-18 Cue Biopharma, Inc. Multimeric t-cell modulatory polypeptides and methods of use thereof
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
EP3752203A1 (en) 2018-02-13 2020-12-23 Novartis AG Chimeric antigen receptor therapy in combination with il-15r and il15
AU2019241345A1 (en) * 2018-03-29 2020-10-15 Adagene Inc. Anti-PD-L1 antibodies and use thereof
CN110891975B (en) * 2018-03-29 2023-11-10 天境生物科技(上海)有限公司 anti-PD-L1 antibodies and uses thereof
US20210147547A1 (en) 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
EP3784688A2 (en) 2018-04-26 2021-03-03 Agenus Inc. Heat shock protein-binding peptide compositions and methods of use thereof
MX2020012567A (en) * 2018-05-23 2021-01-29 Beigene Ltd Anti-ox40 antibodies and methods of use.
MX2020012418A (en) 2018-05-23 2021-04-28 Adc Therapeutics Sa Molecular adjuvant.
UY38247A (en) 2018-05-30 2019-12-31 Novartis Ag ANTIBODIES AGAINST ENTPD2, COMBINATION THERAPIES AND METHODS OF USE OF ANTIBODIES AND COMBINATION THERAPIES
US20210214459A1 (en) 2018-05-31 2021-07-15 Novartis Ag Antibody molecules to cd73 and uses thereof
UY38251A (en) 2018-06-01 2019-12-31 Novartis Ag BINDING MOLECULES AGAINST BCMA AND USES OF THEM
CA3103017A1 (en) 2018-06-23 2019-12-26 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, a platinum agent, and a topoisomerase ii inhibitor
ES2963694T3 (en) 2018-07-10 2024-04-01 Novartis Ag 3-(5-hydroxy-1-oxoisoindolin-2-yl)piperidin-2,6-dione derivatives and their use in the treatment of diseases dependent on zinc finger protein 2 of the ikaros family (ikzf2)
AR116109A1 (en) 2018-07-10 2021-03-31 Novartis Ag DERIVATIVES OF 3- (5-AMINO-1-OXOISOINDOLIN-2-IL) PIPERIDINE-2,6-DIONA AND USES OF THE SAME
US20200171146A1 (en) 2018-07-18 2020-06-04 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, an antimetabolite, and a platinum agent
CN112805267B (en) 2018-09-03 2024-03-08 豪夫迈·罗氏有限公司 Carboxamide and sulfonamide derivatives as TEAD modulators
US20230053449A1 (en) 2018-10-31 2023-02-23 Novartis Ag Dc-sign antibody drug conjugates
WO2020117988A1 (en) 2018-12-04 2020-06-11 Tolero Pharmaceuticals, Inc. Cdk9 inhibitors and polymorphs thereof for use as agents for treatment of cancer
CA3123511A1 (en) 2018-12-20 2020-06-25 Novartis Ag Dosing regimen and pharmaceutical combination comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
AU2019409139A1 (en) 2018-12-21 2021-06-03 Novartis Ag Use of IL-1β binding antibodies
WO2020128637A1 (en) 2018-12-21 2020-06-25 Novartis Ag Use of il-1 binding antibodies in the treatment of a msi-h cancer
AU2019406840A1 (en) 2018-12-21 2021-06-03 Novartis Ag Use of IL-1 beta antibodies in the treatment or prevention of myelodysplastic syndrome
US20220025036A1 (en) 2018-12-21 2022-01-27 Novartis Ag Use of il-1beta binding antibodies
MX2021007271A (en) 2018-12-21 2021-07-15 Onxeo New conjugated nucleic acid molecules and their uses.
KR20210116525A (en) 2019-01-14 2021-09-27 제넨테크, 인크. Methods of treating cancer with PD-1 axis binding antagonists and RNA vaccines
WO2020156507A1 (en) * 2019-02-01 2020-08-06 信达生物制药(苏州)有限公司 Novel anti-pd-l1 antibodies and use thereof
EP3924351A4 (en) 2019-02-12 2022-12-21 Sumitomo Pharma Oncology, Inc. Formulations comprising heterocyclic protein kinase inhibitors
CA3123519A1 (en) 2019-02-15 2020-08-20 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2020165833A1 (en) 2019-02-15 2020-08-20 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
US11793802B2 (en) 2019-03-20 2023-10-24 Sumitomo Pharma Oncology, Inc. Treatment of acute myeloid leukemia (AML) with venetoclax failure
KR20210141621A (en) 2019-03-22 2021-11-23 스미토모 다이니폰 파마 온콜로지, 인크. Compositions comprising PKM2 modulators and methods of treatment using same
WO2020209645A1 (en) * 2019-04-11 2020-10-15 (재) 스크립스코리아항체연구원 Antibody to programmed cell death protein ligand-1 (pd-l1), and use thereof
JP2022529154A (en) 2019-04-19 2022-06-17 ジェネンテック, インコーポレイテッド Anti-MERTK antibody and how to use it
JP2022530674A (en) 2019-05-03 2022-06-30 ジェネンテック, インコーポレイテッド Cancer treatment method using anti-PD-L1 antibody
EP3976090A1 (en) 2019-05-24 2022-04-06 Pfizer Inc. Combination therapies using cdk inhibitors
CN114302878A (en) 2019-07-03 2022-04-08 大日本住友制药肿瘤公司 Tyrosine kinase non-receptor 1(TNK1) inhibitors and uses thereof
WO2021024020A1 (en) 2019-08-06 2021-02-11 Astellas Pharma Inc. Combination therapy involving antibodies against claudin 18.2 and immune checkpoint inhibitors for treatment of cancer
JP2022545741A (en) 2019-08-30 2022-10-28 アジェナス インコーポレイテッド ANTI-CD96 ANTIBODY AND METHODS OF USE THEREOF
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
CN114502590A (en) 2019-09-18 2022-05-13 诺华股份有限公司 ENTPD2 antibodies, combination therapies, and methods of using these antibodies and combination therapies
WO2021076574A2 (en) 2019-10-14 2021-04-22 Aro Biotherapeutics Company Fn3 domain-sirna conjugates and uses thereof
CN114786682A (en) 2019-10-14 2022-07-22 Aro生物疗法公司 Fibronectin type III domain of CD71
WO2021079195A1 (en) 2019-10-21 2021-04-29 Novartis Ag Tim-3 inhibitors and uses thereof
CA3158298A1 (en) 2019-10-21 2021-04-29 Novartis Ag Combination therapies with venetoclax and tim-3 inhibitors
CR20220207A (en) 2019-11-13 2022-06-06 Genentech Inc Therapeutic compounds and methods of use
WO2021098774A1 (en) * 2019-11-21 2021-05-27 Beigene, Ltd. Methods of cancer treatment using anti-ox40 antibodies in combination with anti-pd1 or anti-pdl1 antibodies
US20230000864A1 (en) 2019-11-22 2023-01-05 Sumitomo Pharma Oncology, Inc. Solid dose pharmaceutical composition
GB201917254D0 (en) 2019-11-27 2020-01-08 Adc Therapeutics Sa Combination therapy
CA3165274A1 (en) 2019-12-20 2021-06-24 Novartis Ag Combination of anti tim-3 antibody mbg453 and anti tgf-beta antibody nis793, with or without decitabine or the anti pd-1 antibody spartalizumab, for treating myelofibrosis and myelodysplastic syndrome
CN113121686A (en) * 2019-12-31 2021-07-16 迈威(上海)生物科技股份有限公司 anti-PD-L1 antibody and application thereof
TW202140037A (en) 2020-01-17 2021-11-01 瑞士商諾華公司 Combination therapies
CA3161513A1 (en) 2020-01-28 2021-08-05 Irwin DAVIDSON Antisense oligonucleotide targeting linc00518 for treating melanoma
CN116650628A (en) 2020-01-31 2023-08-29 基因泰克公司 Method for inducing neoepitope specific T cells with PD-1 axis binding antagonists and RNA vaccines
JP2023526723A (en) 2020-05-12 2023-06-23 キュー バイオファーマ, インコーポレイテッド Multimeric T cell regulatory polypeptides and methods of use thereof
AR122644A1 (en) 2020-06-19 2022-09-28 Onxeo NEW CONJUGATED NUCLEIC ACID MOLECULES AND THEIR USES
JP2023531676A (en) 2020-06-23 2023-07-25 ノバルティス アーゲー Dosing Regimens Containing 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione Derivatives
JP2023532768A (en) 2020-07-07 2023-07-31 バイオエヌテック エスエー RNA for treatment of HPV-positive cancer
US11787775B2 (en) 2020-07-24 2023-10-17 Genentech, Inc. Therapeutic compounds and methods of use
WO2022029573A1 (en) 2020-08-03 2022-02-10 Novartis Ag Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022093981A1 (en) 2020-10-28 2022-05-05 Genentech, Inc. Combination therapy comprising ptpn22 inhibitors and pd-l1 binding antagonists
EP4240493A2 (en) 2020-11-04 2023-09-13 Genentech, Inc. Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies and anti-cd79b antibody drug conjugates
JP7402381B2 (en) 2020-11-04 2023-12-20 ジェネンテック, インコーポレイテッド Administration for treatment with anti-CD20/anti-CD3 bispecific antibodies
TW202225191A (en) 2020-11-04 2022-07-01 美商建南德克公司 Subcutaneous dosing of anti-cd20/anti-cd3 bispecific antibodies
EP4240491A1 (en) 2020-11-06 2023-09-13 Novartis AG Cd19 binding molecules and uses thereof
KR20230088780A (en) 2020-11-13 2023-06-20 제넨테크, 인크. Methods and compositions comprising a KRASG12C inhibitor and a PD-L1 binding antagonist for the treatment of lung cancer
CA3202523A1 (en) 2020-12-02 2022-06-09 Genentech, Inc. Methods and compositions for neoadjuvant and adjuvant urothelial carcinoma therapy
TW202237119A (en) 2020-12-10 2022-10-01 美商住友製藥腫瘤公司 Alk-5 inhibitors and uses thereof
WO2022135666A1 (en) 2020-12-21 2022-06-30 BioNTech SE Treatment schedule for cytokine proteins
WO2022135667A1 (en) 2020-12-21 2022-06-30 BioNTech SE Therapeutic rna for treating cancer
TW202245808A (en) 2020-12-21 2022-12-01 德商拜恩迪克公司 Therapeutic rna for treating cancer
TWI789679B (en) * 2021-01-12 2023-01-11 大陸商江蘇先聲藥業有限公司 Anti-human programmed death ligand-1 (pd-l1) antibodies and uses thereof
JP2024505049A (en) 2021-01-29 2024-02-02 ノバルティス アーゲー Administration modes for anti-CD73 and anti-ENTPD2 antibodies and their uses
GB202102396D0 (en) 2021-02-19 2021-04-07 Adc Therapeutics Sa Molecular adjuvant
JP2024511373A (en) 2021-03-18 2024-03-13 ノバルティス アーゲー Biomarkers and their use for cancer
TW202304506A (en) 2021-03-25 2023-02-01 日商安斯泰來製藥公司 Combination therapy involving antibodies against claudin 18.2 for treatment of cancer
TW202304979A (en) 2021-04-07 2023-02-01 瑞士商諾華公司 USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
US20220363696A1 (en) 2021-04-13 2022-11-17 Nuvalent, Inc. Amino-substituted heterocycles for treating cancers with egfr mutations
EP4322938A1 (en) 2021-04-14 2024-02-21 Institut National de la Santé et de la Recherche Médicale (INSERM) New method to improve nk cells cytotoxicity
KR20240005691A (en) 2021-04-30 2024-01-12 에프. 호프만-라 로슈 아게 Dosage for combination therapy with anti-CD20/anti-CD3 bispecific antibody and anti-CD79B antibody drug conjugate
JP2024516230A (en) 2021-04-30 2024-04-12 ジェネンテック, インコーポレイテッド Therapeutic and diagnostic methods and compositions for cancer
CA3219336A1 (en) 2021-05-18 2022-11-24 Kymab Limited Uses of anti-icos antibodies
AR125874A1 (en) 2021-05-18 2023-08-23 Novartis Ag COMBINATION THERAPIES
GB202107994D0 (en) 2021-06-04 2021-07-21 Kymab Ltd Treatment of cancer
AU2022302170A1 (en) 2021-07-02 2023-12-21 F. Hoffmann-La Roche Ag Methods and compositions for treating cancer
IL309831A (en) 2021-07-13 2024-02-01 BioNTech SE Multispecific binding agents against cd40 and cd137 in combination therapy for cancer
IL309120A (en) 2021-07-28 2024-02-01 Hoffmann La Roche Methods and compositions for treating cancer
EP4377350A2 (en) 2021-07-28 2024-06-05 Genentech, Inc. Methods and compositions for treating cancer
TW202321308A (en) 2021-09-30 2023-06-01 美商建南德克公司 Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
WO2023051926A1 (en) 2021-09-30 2023-04-06 BioNTech SE Treatment involving non-immunogenic rna for antigen vaccination and pd-1 axis binding antagonists
WO2023057882A1 (en) 2021-10-05 2023-04-13 Pfizer Inc. Combinations of azalactam compounds with a pd-1 axis binding antagonist for the treatment of cancer
TW202333802A (en) 2021-10-11 2023-09-01 德商拜恩迪克公司 Therapeutic rna for lung cancer
WO2023079428A1 (en) 2021-11-03 2023-05-11 Pfizer Inc. Combination therapies using tlr7/8 agonist
WO2023080900A1 (en) 2021-11-05 2023-05-11 Genentech, Inc. Methods and compositions for classifying and treating kidney cancer
WO2023083439A1 (en) 2021-11-09 2023-05-19 BioNTech SE Tlr7 agonist and combinations for cancer treatment
US20230202984A1 (en) 2021-11-24 2023-06-29 Genentech, Inc. Therapeutic compounds and methods of use
US20230203062A1 (en) 2021-11-24 2023-06-29 Genentech, Inc. Therapeutic compounds and methods of use
WO2023111203A1 (en) 2021-12-16 2023-06-22 Onxeo New conjugated nucleic acid molecules and their uses
WO2023137161A1 (en) 2022-01-14 2023-07-20 Amgen Inc. Triple blockade of tigit, cd112r, and pd-l1
WO2023174210A1 (en) 2022-03-14 2023-09-21 Laekna Limited Combination treatment for cancer
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023220703A1 (en) 2022-05-12 2023-11-16 Genentech, Inc. Methods and compositions comprising a shp2 inhibitor and a pd-l1 binding antagonist
WO2023222854A1 (en) 2022-05-18 2023-11-23 Kymab Limited Uses of anti-icos antibodies
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024049949A1 (en) 2022-09-01 2024-03-07 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
WO2024077166A1 (en) 2022-10-05 2024-04-11 Genentech, Inc. Methods and compositions for classifying and treating lung cancer
WO2024077095A1 (en) 2022-10-05 2024-04-11 Genentech, Inc. Methods and compositions for classifying and treating bladder cancer
WO2024091991A1 (en) 2022-10-25 2024-05-02 Genentech, Inc. Therapeutic and diagnostic methods for multiple myeloma
WO2024115725A1 (en) 2022-12-01 2024-06-06 BioNTech SE Multispecific antibody against cd40 and cd137 in combination therapy with anti-pd1 ab and chemotherapy

Family Cites Families (205)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE792533A (en) 1971-12-09 1973-06-08 Int Chem & Nuclear Corp NEW PYRAZOLO (1.5A) PYRIMIDINES AND THEIR PREPARATION PROCESS
EP0307434B2 (en) 1987-03-18 1998-07-29 Scotgen Biopharmaceuticals, Inc. Altered antibodies
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US5994514A (en) 1991-08-14 1999-11-30 Genentech, Inc. Immunoglobulin variants
AU4116793A (en) 1992-04-24 1993-11-29 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
EP0586740B1 (en) 1992-09-11 1996-12-18 Siemens-Elema AB Device for preventing passage of air bubbles
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
JPH07291996A (en) 1994-03-01 1995-11-07 Yuu Honshiyo Polypeptide related to programmed cell death in human, dna coding the same, vector consisting of the same dna, host cell transformed with the same vector, antibody of the same polypeptide and pharmaceutical composition containing the same polypeptide or the same antibody
JP2778921B2 (en) 1994-11-18 1998-07-23 三共株式会社 Imidazopyrazole derivatives
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
KR100887482B1 (en) 1999-01-15 2009-03-10 제넨테크, 인크. Polypeptide Variants with Altered Effector Function
BR0013542A (en) 1999-08-23 2002-05-14 Dana Farber Cancer Inst Inc B7-4 molecules and uses for them
JP4896327B2 (en) 1999-08-23 2012-03-14 ダナ−ファーバー キャンサー インスティテュート,インコーポレイテッド PD-1, B7-4 receptors and uses thereof
EP1206474B1 (en) 1999-08-27 2004-05-26 Abbott Laboratories Sulfonylphenylpyrazole compounds useful as cox-2 inhibitors
MXPA02002898A (en) 1999-09-17 2003-10-14 Abbott Gmbh & Co Kg Pyrazolopyrimidines as therapeutic agents.
AU2001260153B2 (en) 2000-03-24 2006-08-17 Micromet Ag Multifunctional polypeptides comprising a binding site to an epitope of the NKG2D receptor complex
AU5345901A (en) 2000-04-13 2001-10-30 Univ Rockefeller Enhancement of antibody-mediated immune responses
US20020094989A1 (en) 2000-10-11 2002-07-18 Hale Jeffrey J. Pyrrolidine modulators of CCR5 chemokine receptor activity
KR20030066657A (en) 2000-11-15 2003-08-09 오노 야꾸힝 고교 가부시키가이샤 Pd-1-lacking mouse and use thereof
WO2002060919A2 (en) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules with extended half-lives, compositions and uses thereof
EP1347971B1 (en) 2000-12-21 2006-03-01 Bristol-Myers Squibb Company Thiazolyl inhibitors of tec family tyrosine kinases
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
DK1366067T3 (en) 2001-03-07 2012-10-22 Merck Patent Gmbh EXPRESSION TECHNOLOGY FOR PROTEINS CONTAINING A HYBRID ISOTYPE ANTIBODY UNIT
MXPA03008142A (en) 2001-03-09 2003-12-12 Pfizer Prod Inc Benzimidazole anti-inflammatory compounds.
AR036993A1 (en) 2001-04-02 2004-10-20 Wyeth Corp USE OF AGENTS THAT MODULATE THE INTERACTION BETWEEN PD-1 AND ITS LINKS IN THE SUBMODULATION OF IMMUNOLOGICAL ANSWERS
JPWO2003004497A1 (en) 2001-07-05 2004-10-28 住友製薬株式会社 New heterocyclic compounds
US7662925B2 (en) 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
ATE481985T1 (en) 2002-07-03 2010-10-15 Ono Pharmaceutical Co IMMUNOPOTENTATING COMPOSITIONS
ZA200500782B (en) 2002-08-26 2007-10-31 Takeda Pharmaceutical Calcium receptor modulating compound and use thereof
JP4494205B2 (en) 2002-08-26 2010-06-30 武田薬品工業株式会社 Calcium receptor modulating compound and use thereof
AU2003287345A1 (en) 2002-10-31 2004-06-07 Genentech, Inc. Methods and compositions for increasing antibody production
ES2367430T3 (en) 2002-12-23 2011-11-03 Wyeth Llc ANTIBODIES AGAINST PD-1 AND ITS USES.
CA2512729C (en) 2003-01-09 2014-09-16 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
ES2729974T3 (en) 2003-01-23 2019-11-07 Ono Pharmaceutical Co Specific antibody of human PD-1 and CD3
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
WO2005014599A1 (en) 2003-06-04 2005-02-17 Cellular Genomics, Inc. Imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of bruton’s tyrosine kinase by such compounds
US20060183746A1 (en) 2003-06-04 2006-08-17 Currie Kevin S Certain imidazo[1,2-a]pyrazin-8-ylamines and method of inhibition of Bruton's tyrosine kinase by such compounds
US7393848B2 (en) 2003-06-30 2008-07-01 Cgi Pharmaceuticals, Inc. Certain heterocyclic substituted imidazo[1,2-A]pyrazin-8-ylamines and methods of inhibition of Bruton's tyrosine kinase by such compounds
US7371750B2 (en) 2003-07-29 2008-05-13 Irm Llc Compounds and compositions as protein kinase inhibitors
CN1871259A (en) 2003-08-22 2006-11-29 比奥根艾迪克Ma公司 Improved antibodies having altered effector function and methods for making the same
US20060134105A1 (en) 2004-10-21 2006-06-22 Xencor, Inc. IgG immunoglobulin variants with optimized effector function
WO2005047290A2 (en) 2003-11-11 2005-05-26 Cellular Genomics Inc. Imidazo[1,2-a] pyrazin-8-ylamines as kinase inhibitors
EP1697520A2 (en) 2003-12-22 2006-09-06 Xencor, Inc. Fc polypeptides with novel fc ligand binding sites
GB0400440D0 (en) 2004-01-09 2004-02-11 Isis Innovation Receptor modulators
US8911726B2 (en) 2004-09-22 2014-12-16 Kyowa Hakko Kirin Co., Ltd Stabilized human Igg4 antibodies
AU2005304473A1 (en) 2004-11-10 2006-05-18 Cgi Pharmaceuticals, Inc. Imidazo[1 , 2-a] pyrazin-8-ylamines useful as modulators of kinase activity
CA2591413A1 (en) 2004-12-16 2006-06-22 Vertex Pharmaceuticals Incorporated Pyrid-2-ones useful as inhibitors of tec family protein kinases for the treatment of inflammatory, proliferative and immunologically-mediated diseases
TW200639163A (en) 2005-02-04 2006-11-16 Genentech Inc RAF inhibitor compounds and methods
TW200716551A (en) 2005-03-10 2007-05-01 Cgi Pharmaceuticals Inc Certain substituted amides, method of making, and method of use thereof
EP1896582A4 (en) 2005-05-09 2009-04-08 Ono Pharmaceutical Co Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
KR101600225B1 (en) 2005-06-08 2016-03-04 다나-파버 캔서 인스티튜트 인크. Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (pd-1) pathway
CA2612241C (en) * 2005-07-01 2018-11-06 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
ATE548363T1 (en) 2005-08-29 2012-03-15 Vertex Pharma 3,5-DISUBSTITUTED PYRID-2-ONES, WHICH ARE SUITABLE AS INHIBITORS OF THE TEC FAMILY OF NON-RECEPTOR TYROSINE KINASES
EP1919906B1 (en) 2005-08-29 2011-10-12 Vertex Pharmaceuticals Incorporated 3, 5-disubstituted pyrid-2-ones useful as inhibitors of tec family of non-receptor tyrosine kinases
WO2007026720A1 (en) 2005-08-31 2007-03-08 Taisho Pharmaceutical Co., Ltd. Ring-fused pyrazole derivative
WO2007026950A1 (en) 2005-09-01 2007-03-08 Astellas Pharma Inc. Pyridazinone derivatives used for the treatment of pain
KR20080074220A (en) 2005-12-08 2008-08-12 밀레니엄 파머슈티컬스 인코퍼레이티드 Bicyclic compounds with kinase inhibitory activity
CA2636696A1 (en) 2006-01-13 2007-08-02 Pharmacyclics, Inc. Inhibitors of tyrosine kinases and uses thereof
KR101413615B1 (en) 2006-03-23 2014-07-18 바이오악틱 뉴로사이언스 에이비 Improved antibody selective for human protofibrils and the use thereof
CA2651153C (en) 2006-05-15 2014-04-29 Merck & Co., Inc. Antidiabetic bicyclic compounds
WO2007136790A2 (en) 2006-05-18 2007-11-29 Mannkind Corporation Intracellular kinase inhibitors
JO3235B1 (en) 2006-05-26 2018-03-08 Astex Therapeutics Ltd Pyrrolopyrimidine compounds and their uses
CN101104640A (en) 2006-07-10 2008-01-16 苏州大学 Preparation for anti human PD-L1 monoclonal antibody and application thereof
PE20080839A1 (en) 2006-09-11 2008-08-23 Cgi Pharmaceuticals Inc CERTAIN AMIDAS SUBSTITUTED, METHOD OF PREPARATION AND METHOD OF USE OF THE SAME
AR063946A1 (en) 2006-09-11 2009-03-04 Cgi Pharmaceuticals Inc CERTAIN REPLACED PIRIMIDINS, THE USE OF THE SAME FOR THE TREATMENT OF DISEASES MEDIATED BY THE INHIBITION OF THE ACTIVITY OF BTK AND PHARMACEUTICAL COMPOSITIONS THAT UNDERSTAND THEM.
PE20081370A1 (en) 2006-09-11 2008-11-28 Cgi Pharmaceuticals Inc CERTAIN AMIDAS SUBSTITUTED, METHOD OF PREPARATION AND METHOD OF USE OF THE SAME
EP2270200A3 (en) 2006-09-11 2011-07-13 CGI Pharmaceuticals, Inc. Kinase inhibitors, and methods of using and identifying kinase inhibitors
NZ601278A (en) 2006-09-22 2013-09-27 Pharmacyclics Inc Inhibitors of Bruton's tyrosine kinase
MX2009003649A (en) 2006-10-06 2009-04-22 Irm Llc Protein kinase inhibitors and methods for using thereof.
ES2403546T3 (en) 2006-11-03 2013-05-20 Pharmacyclics, Inc. Bruton tyrosine kinase activity probe and method of use
JP2010522241A (en) 2007-03-21 2010-07-01 ブリストル−マイヤーズ スクイブ カンパニー Condensed heterocyclic compounds useful as proliferative diseases, allergic diseases, autoimmune diseases or inflammatory diseases
WO2008144253A1 (en) 2007-05-14 2008-11-27 Irm Llc Protein kinase inhibitors and methods for using thereof
AU2008255352B2 (en) 2007-05-31 2014-05-22 Genmab A/S Stable IgG4 antibodies
PL2170959T3 (en) 2007-06-18 2014-03-31 Merck Sharp & Dohme Antibodies to human programmed death receptor pd-1
US9243052B2 (en) 2007-08-17 2016-01-26 Daniel Olive Method for treating and diagnosing hematologic malignancies
CL2008002793A1 (en) 2007-09-20 2009-09-04 Cgi Pharmaceuticals Inc Compounds derived from substituted amides, inhibitors of btk activity; pharmaceutical composition comprising them; Useful in the treatment of cancer, bone disorders, autoimmune diseases, among others
JO3076B1 (en) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
US7989465B2 (en) 2007-10-19 2011-08-02 Avila Therapeutics, Inc. 4,6-disubstituted pyrimidines useful as kinase inhibitors
US7982036B2 (en) 2007-10-19 2011-07-19 Avila Therapeutics, Inc. 4,6-disubstitued pyrimidines useful as kinase inhibitors
JP5587193B2 (en) 2007-10-23 2014-09-10 エフ.ホフマン−ラ ロシュ アーゲー Novel kinase inhibitors
JP5643105B2 (en) 2007-12-14 2014-12-17 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Novel imidazo [1,2-a] pyridine and imidazo [1,2-b] pyridazine derivatives
MY158411A (en) 2008-02-05 2016-10-14 Hoffmann La Roche Novel pyridinones and pyridazinones
MX2010008578A (en) 2008-02-08 2010-11-10 Medimmune Llc Anti-ifnar1 antibodies with reduced fc ligand affinity.
EP2262837A4 (en) 2008-03-12 2011-04-06 Merck Sharp & Dohme Pd-1 binding proteins
JP5529852B2 (en) 2008-05-06 2014-06-25 ジリード コネティカット,インコーポレイティド Substituted amides, their preparation and use as Btk inhibitors
EP2361248B1 (en) 2008-06-27 2018-09-19 Celgene CAR LLC Heteroaryl compounds and uses thereof
US8338439B2 (en) 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
KR101523451B1 (en) 2008-07-02 2015-05-27 에프. 호프만-라 로슈 아게 Novel phenylpyrazinones as kinase inhibitors
WO2010006947A1 (en) 2008-07-15 2010-01-21 F. Hoffmann-La Roche Ag Novel phenyl-imidazopyridines and pyridazines
AU2009270856B2 (en) 2008-07-16 2013-07-25 Pharmacyclics Llc Inhibitors of Bruton's tyrosine kinase for the treatment of solid tumors
MX2010013190A (en) 2008-07-18 2010-12-20 Hoffmann La Roche Novel phenylimidazopyrazines.
WO2010011837A1 (en) 2008-07-24 2010-01-28 Bristol-Myers Squibb Company Fused heterocyclic compounds useful as kinase modulators
KR20110074850A (en) 2008-08-25 2011-07-04 앰플리뮨, 인크. Pd-1 antagonists and methods of use thereof
BRPI0918970A2 (en) 2008-09-05 2019-09-24 Avila Therapeutics Inc algorithm for irreversible inhibitor design
WO2010029435A1 (en) 2008-09-12 2010-03-18 Isis Innovation Limited Pd-1 specific antibodies and uses thereof
EP2342228B1 (en) 2008-09-12 2017-09-06 Oxford University Innovation Limited Pd-1 specific antibodies and uses thereof
CA2998281C (en) 2008-09-26 2022-08-16 Dana-Farber Cancer Institute, Inc. Human anti-pd-1 antobodies and uses therefor
KR101050829B1 (en) * 2008-10-02 2011-07-20 서울대학교산학협력단 Anticancer agents comprising an anti-PD-1 antibody or an anti-PD-L1 antibody
AU2009308675A1 (en) 2008-10-31 2010-05-06 Genentech, Inc. Pyrazolopyrimidine JAK inhibitor compounds and methods
US20120028981A1 (en) 2008-11-05 2012-02-02 Principia Biopharma Inc. Kinase Knockdown Via Electrophilically Enhanced Inhibitors
WO2010056875A1 (en) 2008-11-12 2010-05-20 Cgi Pharmaceuticals, Inc. Pyridazinones and their use as btk inhibitors
WO2010063011A2 (en) 2008-11-28 2010-06-03 Emory University Methods for the treatment of infections and tumors
US8426428B2 (en) 2008-12-05 2013-04-23 Principia Biopharma, Inc. EGFR kinase knockdown via electrophilically enhanced inhibitors
HRP20240240T1 (en) * 2008-12-09 2024-04-26 F. Hoffmann - La Roche Ag Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2010068810A2 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Certain substituted amides, method of making, and method of use thereof
WO2010068788A1 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Heterocyclic amides as btk inhibitors
WO2010068806A1 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Amide derivatives as btk inhibitors in the treatment of allergic, autoimmune and inflammatory disorders as well as cancer
AU2009335821A1 (en) 2008-12-19 2010-07-15 Bristol-Myers Squibb Company Carbazole carboxamide compounds useful as kinase inhibitors
US20100197924A1 (en) 2008-12-22 2010-08-05 Millennium Pharmaceuticals, Inc. Preparation of aminotetralin compounds
WO2010080712A2 (en) 2009-01-06 2010-07-15 Dana Farber Cancer Institute Pyrimido-diazepinone kinase scaffold compounds and methods of treating disorders
EP2393835B1 (en) * 2009-02-09 2017-04-05 Université d'Aix-Marseille Pd-1 antibodies and pd-l1 antibodies and uses thereof
US8299077B2 (en) 2009-03-02 2012-10-30 Roche Palo Alto Llc Inhibitors of Bruton's tyrosine kinase
CA2748414A1 (en) 2009-04-24 2010-10-28 F. Hoffmann-La Roche Ag Inhibitors of bruton's tyrosine kinase
WO2010126960A1 (en) 2009-04-29 2010-11-04 Locus Pharmaceuticals, Inc. Pyrrolotriazine compounds
US8586751B2 (en) 2009-06-12 2013-11-19 Bristol-Myers Squibb Company Nicotinamide compounds useful as kinase modulators
AR077468A1 (en) 2009-07-09 2011-08-31 Array Biopharma Inc PIRAZOLO COMPOUNDS (1,5-A) PYRIMIDINE SUBSTITUTED AS TRK-QUINASA INHIBITORS
US8846673B2 (en) 2009-08-11 2014-09-30 Bristol-Myers Squibb Company Azaindazoles as kinase inhibitors and use thereof
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
WO2011029043A1 (en) 2009-09-04 2011-03-10 Biogen Idec Ma Inc. Heteroaryl btk inhibitors
TWI711610B (en) 2009-09-04 2020-12-01 美商百健Ma公司 Bruton's tyrosine kinase inhibitors
US7718662B1 (en) 2009-10-12 2010-05-18 Pharmacyclics, Inc. Pyrazolo-pyrimidine inhibitors of bruton's tyrosine kinase
US8389689B2 (en) 2009-10-28 2013-03-05 Janssen Biotech, Inc. Anti-GLP-1R antibodies and their uses
US10053513B2 (en) 2009-11-30 2018-08-21 Janssen Biotech, Inc. Antibody Fc mutants with ablated effector functions
LT2536745T (en) 2010-02-19 2016-09-26 Xencor, Inc. Novel ctla4-ig immunoadhesins
JP2013523098A (en) 2010-03-29 2013-06-17 ザイムワークス,インコーポレイテッド Antibody with enhanced or suppressed effector function
CN103038233B (en) 2010-05-07 2017-06-20 吉利德康涅狄格有限公司 Pyridone and azepine pyridinone compounds and application method
TWI500617B (en) 2010-05-31 2015-09-21 Ono Pharmaceutical Co Purine ketone derivatives
AU2011261185A1 (en) 2010-06-03 2013-01-10 Pharmacyclics, Inc. The use of inhibitors of Bruton's tyrosine kinase (Btk)
US8685969B2 (en) 2010-06-16 2014-04-01 Bristol-Myers Squibb Company Carboline carboxamide compounds useful as kinase inhibitors
HUE042165T2 (en) 2010-06-23 2019-06-28 Hanmi Science Co Ltd Novel fused pyrimidine derivatives for inhibition of tyrosine kinase activity
US20120053189A1 (en) 2010-06-28 2012-03-01 Pharmacyclics, Inc. Btk inhibitors for the treatment of immune mediated conditions
WO2012009705A1 (en) 2010-07-15 2012-01-19 Zyngenia, Inc. Ang-2 binding complexes and uses thereof
CA2807051A1 (en) 2010-08-10 2012-02-16 Celgene Avilomics Research, Inc. Besylate salt of a btk inhibitor
AR082590A1 (en) 2010-08-12 2012-12-19 Hoffmann La Roche INHIBITORS OF THE TIROSINA-QUINASA DE BRUTON
US9249123B2 (en) 2010-09-01 2016-02-02 Genentech, Inc. Pyridinones/pyrazinones, method of making, and method of use thereof
BR112013007499A2 (en) 2010-09-01 2016-07-12 Genentech Inc pyridazinones - rearing methods and uses
CN103429261A (en) 2010-12-22 2013-12-04 塞法隆澳大利亚股份有限公司 Modified antibody with improved half-life
CN105949313B (en) 2011-03-29 2021-06-15 罗切格利卡特公司 Antibody Fc variants
PL2691112T3 (en) 2011-03-31 2018-09-28 Merck Sharp & Dohme Corp. Stable formulations of antibodies to human programmed death receptor pd-1 and related treatments
WO2012135801A1 (en) 2011-04-01 2012-10-04 University Of Utah Research Foundation Substituted n-(3-(pyrimidin-4-yl)phenyl)acrylamide analogs as tyrosine receptor kinase btk inhibitors
CA2760174A1 (en) 2011-12-01 2013-06-01 Pharmascience Inc. Protein kinase inhibitors and uses thereof
RU2625034C2 (en) 2011-04-20 2017-07-11 МЕДИММЬЮН, ЭлЭлСи Antibodies and other molecules binding b7-h1 and pd-1
EP2699577A1 (en) 2011-04-20 2014-02-26 Glaxo Group Limited Tetrahydropyrazolo [1,5 -a]pyrimidine as anti -tuberculosis compounds
HUE033019T2 (en) 2011-05-17 2017-11-28 Principia Biopharma Inc Tyrosine kinase inhibitors
WO2012158795A1 (en) 2011-05-17 2012-11-22 Principia Biopharma Inc. Pyrazolopyrimidine derivatives as tyrosine kinase inhibitors
US20140107151A1 (en) 2011-05-17 2014-04-17 Principia Biophama Inc. Tyrosine kinase inhibitors
SG194728A1 (en) 2011-05-17 2013-12-30 Hoffmann La Roche Inhibitors of bruton's tyrosine kinase
DK3424953T3 (en) 2011-06-06 2020-11-02 Novo Nordisk As Terapeutiske antistoffer
US9096642B2 (en) * 2011-06-08 2015-08-04 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
RS63418B1 (en) 2011-06-10 2022-08-31 Merck Patent Gmbh Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
AU2012273955A1 (en) * 2011-06-22 2014-01-09 Inserm (Institut National De La Sante Et De La Recherche Medicale) Anti-Axl antibodies and uses thereof
CA2841111A1 (en) 2011-07-08 2013-01-17 Novartis Ag Novel pyrrolo pyrimidine derivatives
MY170117A (en) 2011-08-01 2019-07-05 Genentech Inc Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
US9624298B2 (en) 2011-11-28 2017-04-18 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US20150210763A1 (en) 2012-02-09 2015-07-30 Chugai Seiyaku Kabushiki Kaisha MODIFIED Fc REGION OF ANTIBODY
CA2871934C (en) 2012-04-30 2023-06-13 Medimmune, Llc Molecules with reduced effector function and extended half-lives, compositions, and uses thereof
BR122022015975B1 (en) 2012-05-15 2024-01-02 Bristol-Myers Squibb Company MONOCLONAL ANTIBODIES, KIT FOR THE TREATMENT OF AN INDIVIDUAL AFFLICTED WITH A CANCER, PROCESS FOR MEASURING MEMBRANOUS PD-L1 ON ISOLATED TUMOR CELLS AND USE OF THE ANTIBODY OR A PORTION THAT BIDS TO THE ANTIGEN THEREOF
EP3553086A1 (en) 2012-05-31 2019-10-16 Sorrento Therapeutics Inc. Antigen binding proteins that bind pd-l1
CA2877516A1 (en) 2012-07-03 2014-01-03 Janssen Alzheimer Immunotherapy C-terminal and central epitope a-beta antibodies
KR101947702B1 (en) * 2012-10-04 2019-02-14 다나-파버 캔서 인스티튜트 인크. Human monoclonal anti-pd-l1 antibodies and methods of use
EP2934577A1 (en) 2012-12-19 2015-10-28 Adimab, LLC Multivalent antibody analogs, and methods of their preparation and use
AR093984A1 (en) 2012-12-21 2015-07-01 Merck Sharp & Dohme ANTIBODIES THAT JOIN LEGEND 1 OF SCHEDULED DEATH (PD-L1) HUMAN
TWI682941B (en) 2013-02-01 2020-01-21 美商再生元醫藥公司 Antibodies comprising chimeric constant domains
RU2017114166A (en) 2013-02-20 2019-01-29 Регенерон Фармасьютикалс, Инк. NON-HUMAN ANIMALS WITH MODIFIED SEQUENCES OF HEAVY IMMUNOGLOBULIN CHAINS
CA2904377C (en) 2013-03-15 2021-07-13 Regeneron Pharmaceuticals, Inc. Il-33 antagonists and uses thereof
SG11201506764WA (en) 2013-04-25 2015-09-29 Beigene Ltd Fused heterocyclic compounds as protein kinase inhibitors
EP3004169B1 (en) 2013-05-31 2023-03-22 Sorrento Therapeutics, Inc. Antigen binding proteins that bind pd-1
CN107011441B (en) 2013-09-13 2020-12-01 百济神州(广州)生物科技有限公司 anti-PD 1 antibodies and their use as therapeutic and diagnostic agents
MX2016005283A (en) 2013-10-25 2017-02-20 Pharmacyclics Llc Treatment using bruton's tyrosine kinase inhibitors and immunotherapy.
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
EP3160505A4 (en) 2014-07-03 2018-01-24 BeiGene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
WO2016008411A1 (en) 2014-07-18 2016-01-21 Beigene, Ltd. 5-amino-4-carbamoyl-pyrazole compounds as selective and irreversible t790m over wt-egfr kinase inhibitors and use thereof
WO2016024228A1 (en) 2014-08-11 2016-02-18 Acerta Pharma B.V. Therapeutic combinations of a btk inhibitor, a pi3k inhibitor, a jak-2 inhibitor, a pd-1 inhibitor and/or a pd-l1 inhibitor
US10035802B2 (en) 2014-08-14 2018-07-31 Teva Pharmaceuticals Usa, Inc. Solid state forms of ibrutinib
TW201625304A (en) 2014-10-24 2016-07-16 美國禮來大藥廠 Therapy for urothelial carcinoma
WO2016087994A1 (en) 2014-12-05 2016-06-09 Acerta Pharma B.V. Btk inhibitors to treat solid tumors through modulation of the tumor microenvironment
EP3233103B1 (en) 2014-12-18 2020-10-14 Principia Biopharma Inc. Treatment of pemphigus
DK3236943T3 (en) 2014-12-24 2023-04-11 Principia Biopharma Inc COMPOSITIONS FOR ILEO-JEJUNAL DRUG ADMINISTRATION
US9139653B1 (en) 2015-04-30 2015-09-22 Kymab Limited Anti-human OX40L antibodies and methods of treatment
AU2016270625B2 (en) 2015-05-29 2022-10-06 Genentech, Inc. Therapeutic and diagnostic methods for cancer
WO2017024465A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
US20190022092A1 (en) 2015-09-15 2019-01-24 Acerta Pharma B.V. Therapeutic Combinations of a BTK Inhibitor and a GITR Binding Molecule, a 4-1BB Agonist, or an OX40 Agonist
TW201725044A (en) 2015-10-01 2017-07-16 基利科學股份有限公司 Combination of a BTK inhibitor and a checkpoint inhibitor for treating cancers
JP6993056B2 (en) 2016-07-05 2022-02-15 ベイジーン リミテッド Combination of PD-1 antagonist and RAF inhibitor for cancer treatment
IL264784B (en) 2016-08-16 2022-07-01 Beigene Ltd Crystalline form of (s)-7-(1-acryloylpiperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetra-hydropyrazolo(1,5-a)pyrimidine-3-carboxamide, preparation, and uses thereof
CA3034326A1 (en) 2016-08-19 2018-02-22 Beigene, Ltd. Use of a combination comprising a btk inhibitor for treating cancers
SG11201901548SA (en) 2016-08-26 2019-03-28 Beigene Ltd Anti-tim-3 antibodies and use thereof
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
EP3519051B1 (en) 2016-09-27 2021-09-22 Beigene, Ltd. Treatment of cancers using combination comprising parp inhibitors
EP3526241A1 (en) 2016-10-14 2019-08-21 Xencor, Inc. Il15/il15r heterodimeric fc-fusion proteins
EP3573989A4 (en) 2017-01-25 2020-11-18 Beigene, Ltd. Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
AU2018253951A1 (en) 2017-04-20 2019-09-19 Adc Therapeutics Sa Combination therapy
JP2020525411A (en) 2017-06-26 2020-08-27 ベイジーン リミテッド Immunotherapy for hepatocellular carcinoma
CN110997677A (en) 2017-08-12 2020-04-10 百济神州有限公司 Btk inhibitors with improved dual selectivity
US11786529B2 (en) 2017-11-29 2023-10-17 Beigene Switzerland Gmbh Treatment of indolent or aggressive B-cell lymphomas using a combination comprising BTK inhibitors
CN112351794A (en) 2018-02-09 2021-02-09 百济神州有限公司 Immunotherapy method for urothelial cancer
GB201803746D0 (en) 2018-03-08 2018-04-25 Ultrahuman Eight Ltd PD1 binding agents
SG11202009137PA (en) 2018-03-21 2020-10-29 Mei Pharma Inc Combination therapy
JP2022538215A (en) 2019-06-10 2022-09-01 ベイジーン スウィッツァーランド ゲーエムベーハー Oral capsule and its preparation method
EP3981399A4 (en) 2019-06-10 2023-05-31 BeiGene Switzerland GmbH Oral solid tablet comprising bruton's tyrosine kinase inhibitor and preparation method therefor

Also Published As

Publication number Publication date
CN106604742B (en) 2019-01-11
KR102003754B1 (en) 2019-07-25
KR102130600B1 (en) 2020-07-08
US20180215825A1 (en) 2018-08-02
US20200283527A1 (en) 2020-09-10
JP6526189B2 (en) 2019-06-05
WO2016000619A1 (en) 2016-01-07
TW201625681A (en) 2016-07-16
CN106604742A (en) 2017-04-26
US10544225B2 (en) 2020-01-28
TWI726608B (en) 2021-05-01
KR20170023102A (en) 2017-03-02
TW202019970A (en) 2020-06-01
CN110156892B (en) 2023-05-16
KR20190089090A (en) 2019-07-29
JP2017522905A (en) 2017-08-17
JP2019172677A (en) 2019-10-10
EP3160505A4 (en) 2018-01-24
EP3160505A1 (en) 2017-05-03
CN110156892A (en) 2019-08-23
TWI687438B (en) 2020-03-11
US11512132B2 (en) 2022-11-29
JP6734436B2 (en) 2020-08-05

Similar Documents

Publication Publication Date Title
US20230279117A1 (en) Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
US11186637B2 (en) Anti-PD1 antibodies and their use as therapeutics and diagnostics
JP7430137B2 (en) Antibodies and methods of use

Legal Events

Date Code Title Description
AS Assignment

Owner name: BEIGENE, LTD., CAYMAN ISLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LI, KANG;SONG, JING;ZHANG, TONG;AND OTHERS;REEL/FRAME:062711/0879

Effective date: 20170103

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION