US20160067337A1 - Combination of dr5 agonist and anti-pd-1 antagonist and methods of use - Google Patents

Combination of dr5 agonist and anti-pd-1 antagonist and methods of use Download PDF

Info

Publication number
US20160067337A1
US20160067337A1 US14/774,382 US201414774382A US2016067337A1 US 20160067337 A1 US20160067337 A1 US 20160067337A1 US 201414774382 A US201414774382 A US 201414774382A US 2016067337 A1 US2016067337 A1 US 2016067337A1
Authority
US
United States
Prior art keywords
antibody
antagonist
seq
agonist
set forth
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/774,382
Other languages
English (en)
Inventor
Bryan BARNHART
Maria N. JURE-KUNKEL
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.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
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 Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Priority to US14/774,382 priority Critical patent/US20160067337A1/en
Assigned to BRISTOL-MYERS SQUIBB COMPANY reassignment BRISTOL-MYERS SQUIBB COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JURE-KUNKEL, MARIA N., BARNHART, Bryan
Assigned to BRISTOL-MYERS SQUIBB COMPANY reassignment BRISTOL-MYERS SQUIBB COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JURE-KUNKEL, MARIA N., BARNHART, Bryan
Publication of US20160067337A1 publication Critical patent/US20160067337A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • 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
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®

Definitions

  • PD-1 Programmed Cell Death 1
  • PD-1 is a cell surface signaling receptor that plays a critical role in the regulation of T cell activation and tolerance (Keir M E, et al., Annu Rev Immunol 2008; 26:677-704). It is a type I transmembrane protein and together with BTLA, CTLA-4, ICOS and CD28, comprise the CD28 family of T cell co-stimulatory receptors.
  • PD-1 is primarily expressed on activated T cells, B cells, and myeloid cells (Dong H, et al., Nat Med 1999; 5:1365-1369). It is also expressed on natural killer (NK) cells (Terme M, et al., Cancer Res 2011; 71:5393-5399).
  • PD-1 is highly expressed on tumor infiltrating lymphocytes, and its ligands are up-regulated on the cell surface of many different tumors (Dong H, et al., Nat Med 2002; 8:793-800).
  • Multiple murine cancer models have demonstrated that binding of ligand to PD-1 results in immune evasion. In addition, blockade of this interaction results in anti-tumor activity.
  • PD-L1 and PD-L2 Two cell surface glycoprotein ligands for PD-1 have been identified, PD-L1 and PD-L2, and have been shown to downregulate T cell activation and cytokine secretion upon binding to PD-1 (Freeman et al. (2000) J Exp Med 192:1027-34; Latchman et al. (2001) Nat Immunol 2:261-8; Carter et al. (2002) Eur J Immunol 32:634-43; Ohigashi et al. (2005) Clin Cancer Res 11:2947-53).
  • Both PD-L1 (B7-H1) and PD-L2 (B7-DC) are B7 homologs that bind to PD-1, but do not bind to other CD28 family members (Blank et al. (2004). Expression of PD-L1 on the cell surface has also been shown to be upregulated through IFN-gamma stimulation.
  • PD-L1 expression has been found in several murine and human cancers, including human lung, ovarian and colon carcinoma and various myelomas (Iwai et al. (2002) PNAS 99:12293-7; Ohigashi et al. (2005) Clin Cancer Res 11:2947-53).
  • PD-L1 has been suggested to play a role in tumor immunity by increasing apoptosis of antigen-specific T-cell clones (Dong et al. (2002) Nat Med 8:793-800). It has also been suggested that PD-L1 might be involved in intestinal mucosal inflammation and inhibition of PD-L1 suppresses wasting disease associated with colitis (Kanai et al. (2003) J Immunol 171:4156-63).
  • TRAIL tumor necrosis factor
  • TNF tumor necrosis factor
  • DR4 TRAIL-R1
  • DR5 TRAIL-R2
  • TRAIL-R1 apoptosis-inducing receptors, which each contain an intracellular death domain (see e.g., Pan G, et al., Science. 1997; 276:111-113, Pan G, et al., Science. 1997; 277:815-818, Sheridan J P, et al., Science.
  • DR4 and DR5 recruit FAS associated protein with death domain (FADD) and caspase-8 to form the death-inducing signaling complex (DISC), which activates caspase-8, subsequently leading to the activation of executioner caspases such as caspase-3 that induce apoptosis (see, e.g., Kischkel F C, et al., Immunity. 2000; 12:611-620, Thomas L R, et al., J Biol Chem.
  • FADD FAS associated protein with death domain
  • DISC death-inducing signaling complex
  • TRAIL and agonistic antibodies that recognize TRAIL receptors preferentially kill tumor cells and induce potent anti-tumor activity in a variety of experimental models (see, Griffith T S, et al., Curr Opin Immunol. 1998; 10:559-563, Ashkenazi A, et al., J Clin Invest. 1999; 104:155-162, Walczak H, et al, Nat Med. 1999; 5:157-163, Chuntharapai A, et al., J Immunol. 2001; 166:4891-4898, and Ichikawa K, et al., Nat Med. 2001; 7:954-960).
  • a DR5 agonist e.g., an antibody
  • an anti-PD-1 antagonist e.g., an antibody
  • the present invention provides a method for the treatment of cancer in a subject by co-administering an effective amount of a PD-1 antagonist and an agent that induces apoptosis in cancer cells, e.g., an agent that engages the DR4 or DR5 receptor, such as a DR4 or DR5 agonist.
  • Suitable anti-DR5 agonists for use in the methods of the invention include, without limitation, ligands, antibodies (e.g., monoclonal antibodies and bispecific antibodies) and multivalent agents.
  • the DR5 agonist is an antibody selected from the group consisting of Lexatumumab (also known as ETR2-ST01), Tigatuzumab (also known as CS-1008), Conatumumab (also known as AMG 655), Drozitumab, HGSTR2J/KMTRS, and LBY-135.
  • the DR5 agonist is a multivalent agent (e.g., TAS266).
  • the DR5 agonist is a ligand (e.g., a TNF-related apoptosis-inducing ligand (TRAIL), such as a recombinant human TRAIL, e.g., Dulanermin (also known as AMG951)).
  • a ligand e.g., a TNF-related apoptosis-inducing ligand (TRAIL), such as a recombinant human TRAIL, e.g., Dulanermin (also known as AMG951)).
  • TRAIL TNF-related apoptosis-inducing ligand
  • Dulanermin also known as AMG951
  • Suitable PD-1 antagonists for use in the methods of the invention include, without limitation, ligands, antibodies (e.g., monoclonal antibodies and bispecific antibodies), and multivalent agents.
  • the PD-1 antagonist is a fusion protein, e.g., an Fc fusion protein, such as AMP-244.
  • the PD-1 antagonist is an anti-PD-1 or anti-PD-L1 antibody.
  • the PD-1 antagonist is an antibody, such as MK-3475 or CT-011.
  • an exemplary anti-PD-1 antibody is 5C4 (referred to as 5C4 in WO 2006/121168; also known as MDX-1106, ONO-4538, and Nivolumab) comprising heavy and light chains having the sequences shown in SEQ ID NOs: 11 and 12, respectively, or antigen binding fragments and variants thereof.
  • the antibody comprises the heavy and light chain CDRs or VRs of 5C4. Accordingly, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH region of 5C4 having the sequence shown in SEQ ID NO: 13, and the CDR1, CDR2 and CDR3 domains of the VL region of 5C4 having the sequence shown in SEQ ID NO:15.
  • the antibody comprises the heavy chain CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 17, 18, and 19, respectively, and the light chain CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 20, 21, and 22, respectively.
  • the antibody comprises VH and/or VL regions having the amino acid sequences set forth in SEQ ID NO: 13 and/or SEQ ID NO: 15, respectively.
  • the antibody comprises the heavy chain variable (VH) and/or light chain variable (VL) regions encoded by the nucleic acid sequences set forth in SEQ ID NO: 14 and/or SEQ ID NO: 16, respectively.
  • the antibody competes for binding with, and/or binds to the same epitope on PD-1 as, the above-mentioned antibodies.
  • the antibody has at least about 90% variable region amino acid sequence identity with the above-mentioned antibodies (e.g., at least about 90%, 95% or 99% variable region identity with SEQ ID NO: 13 or SEQ ID NO: 15).
  • the PD-1 antagonist is an anti-PD-L1 antibody, such as MEDI4736 (also known as Anti-B7-H1) or MPDL3280A (also known as RG7446).
  • An exemplary anti-PD-L1 antibody is 12A4 (referred to as 12A4 in WO 2007/005874 and U.S. Pat. No. 7,943,743).
  • the antibody comprises the heavy and light chain CDRs or VRs of 12A4.
  • the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH region of 12A4 having the sequence shown in SEQ ID NO: 1, and the CDR1, CDR2 and CDR3 domains of the VL region of 5C4 having the sequence shown in SEQ ID NO: 3.
  • the antibody comprises the heavy chain CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 5, 6, and 7, respectively, and the light chain CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 8, 9, and 10, respectively.
  • the antibody comprises VH and/or VL regions having the amino acid sequences set forth in SEQ ID NO: 1 and/or SEQ ID NO: 3, respectively.
  • the antibody comprises the heavy chain variable (VH) and/or light chain variable (VL) regions encoded by the nucleic acid sequences set forth in SEQ ID NO: 2 and/or SEQ ID NO: 4, respectively.
  • the antibody competes for binding with, and/or binds to the same epitope on PD-L1 as, the above-mentioned antibodies.
  • the antibody has at least about 90% variable region amino acid sequence identity with the above-mentioned antibodies (e.g., at least about 90%, 95% or 99% variable region identity with SEQ ID NO: 1 or SEQ ID NO: 3).
  • the invention provides a method of treating cancer in a subject, the method comprising administering to the subject an effective amount of a PD-1 antagonist and a DR5 agonist, wherein
  • the PD-1 antagonist is an anti-PD-1 antibody comprising the CDR1, CDR2 and CDR3 domains in a heavy chain variable region having the sequence set forth in SEQ ID NO: 13, and the CDR1, CDR2 and CDR3 domains in a light chain variable region having the sequence set forth in SEQ ID NO: 15; and (b) the DR5 agonist is an antibody.
  • the invention provides a method cancer in a subject, the method comprising administering to the subject an effective amount of a PD-1 antagonist and a DR5 agonist, wherein
  • the PD-1 antagonist is an anti-PD-L1 antibody comprising the CDR1, CDR2 and CDR3 domains in a heavy chain variable region having the sequence set forth in SEQ ID NO: 1, and the CDR1, CDR2 and CDR3 domains in a light chain variable region having the sequence set forth in SEQ ID NO: 3; and (b) the DR5 agonist is an antibody.
  • the efficacy of the treatment methods provided herein can be assessed using any suitable means.
  • the treatment produces at least one therapeutic effect selected from the group consisting of reduction in size of a tumor, reduction in number of metastasic lesions over time, complete response, partial response, and stable disease.
  • administration of a PD-1 antagonist and a DR5 agonist results in at least a 1, 1.25, 1.50, 1.75, 2, 2.25, 2.50, 2.75, 3, 3.25, 3.5, 3.75, or 4-fold reduction in tumor volume, e.g., relative to treatment with the PD-1 antagonist or DR5 agonist alone, or relative to tumor volume before initiation of treatment.
  • administration of the PD-1 antagonist and DR5 agonist results in at least a 1-fold, 2-fold, or more preferably a 3-fold reduction in tumor volume, e.g., relative to treatment with the PD-1 antagonist or DR5 agonist alone, or relative to tumor volume before initiation of treatment.
  • administration of a PD-1 antagonist and a DR5 agonist results in tumor growth inhibition of at least 50%, 60%, 70% or 80%, e.g., relative to treatment with the PD-1 antagonist or DR5 agonist alone, or relative to tumor volume before initiation of treatment.
  • tumor volume is reduced by 50%, 60%, 70%, 80%, 90% or more, e.g., relative to tumor size before initiation of the treatment.
  • the PD-1 antagonist and DR5 agonist can be administered accordingly to a suitable dosage, route (e.g., intravenous, intraperitoneal, intramuscular, intrathecal or subcutaneous).
  • the antagonist and agonist can also be administered according to any suitable schedule.
  • the antagonist and agonist can be simultaneously administered in a single formulation.
  • the antagonist and agonist can be formulated for separate administration, wherein they are administered concurrently or sequentially.
  • the PD-1 antagonist is administered prior to administration of the DR5 agonist.
  • the DR5 agonist is administered prior to administration of the PD-1 antagonist.
  • the DR5 agonist and the PD-1 antagonist are administered simultaneously.
  • the cancer is a cancer selected from the group consisting of leukemia, lymphoma, blastoma, carcinoma and sarcoma.
  • the cancer is selected from the group consisting of chronic myeloid leukemia, acute lymphoblastic leukemia, Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ ALL), squamous cell carcinoma, small-cell lung cancer, non-small cell lung cancer, glioma, gastrointestinal cancer, renal cancer, ovarian cancer, liver cancer, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, stomach cancer, bladder cancer, hepatoma, breast cancer, colon carcinoma, and head and neck cancer, gastric cancer, germ cell tumor, pediatric sarcoma, sinonasal natural killer, multiple myeloma, acute myelogenous leukemia (AML), and chronic lymphocytic leuk
  • AML acute
  • Additional agents and therapies can be administered in combination with the agonists and antagonists described herein.
  • the methods comprise administration of an additional therapeutic agent (e.g., a cyotoxin or chemotherapeutic agent.
  • compositions comprising a PD-1 antagonist and a DR5 agonist.
  • the antagonist and/or agonist is a ligand, antibody (e.g., monoclonal antibody or bispecific antibody) or multivalent agent.
  • the PD-1 antagonist is an anti-PD-1 antibody comprising the heavy and light chain CDRs or VRs of 5C4.
  • the PD-1 antagonist is an anti-PD-L1 antibody comprising the heavy and light chain CDRs or VRs of 12A4.
  • kits for treating a cancer in a subject comprising:
  • the DR5 agonist is an antibody.
  • the PD-1 antagonist is an antibody.
  • the PD-1 antagonist is an anti-PD-1 antibody comprising the CDR1, CDR2 and CDR3 domains in a heavy chain variable region having the sequence set forth in SEQ ID NO: 13, and the CDR1, CDR2 and CDR3 domains in a light chain variable region having the sequence set forth in SEQ ID NO: 15.
  • the PD-1 antagonist is an anti-PD-L1 antibody comprising antibody comprises the CDR1, CDR2 and CDR3 domains in a heavy chain variable region having the sequence set forth in SEQ ID NO: 1, and the CDR1, CDR2 and CDR3 domains in a light chain variable region having the sequence set forth in SEQ ID NO: 3.
  • FIG. 1 is a graph depicting the median tumor volume in mice (mm 3 ) after administration of a control, an anti-DR5 antibody, an anti-PD-1 antibody, or combination of both an anti-DR5 antibody, an anti-PD-1 antibody, up to 23 days post implant.
  • FIG. 2 depicts the tumor volume in individual mice administered a control ( FIG. 2A ), an anti-PD-1 antibody on day 6 post-implant ( FIG. 2B ), an anti-PD-1 antibody on day 8 post-implant ( FIG. 2C ), an anti-PD-1 antibody on day 9 post-implant ( FIG. 2D ), an anti-DR5 antibody on day 8 post-implant ( FIG. 2E ), an anti-DR5 antibody on day 8 post-implant in combination with an anti-PD-1 antibody on day 6 post-implant ( FIG. 2F ), an anti-DR5 antibody on day 8 post-implant in combination with an anti-PD-1 antibody on day 8 post-implant ( FIG. 2G ), and an anti-DR5 antibody on day 8 post-implant in combination with an anti-PD-1 antibody on day 9 post-implant ( FIG. 211 ).
  • FIG. 3 is a graph depicting the percent body weight change following administration of a control, an anti-DR5 monoclonal antibody, an anti-PD-1 antibody, or combination of both an anti-DR5 monoclonal antibody and anti-PD-1 antibody, up to 24 days post implant.
  • the invention is based on the discovery that co-administration of a DR5 agonist (e.g., an antibody) and a PD-1 antagonist (e.g., an antibody) effectively inhibits tumor growth in vivo, even synergistically. Accordingly, the present invention provides a method for the treatment of cancer in a subject which comprises administering to a subject (e.g., human) an effective amount of a PD-1 antagonist and a DR5 agonist.
  • a subject e.g., human
  • the term “subject” or “patient” is a human patient (e.g., a patient having cancer).
  • a “solid tumor” includes, for example, sarcoma, melanoma, carcinoma, prostate carcinoma, lung carcinoma, colon carcinoma, or other solid tumor cancer.
  • cancer refers to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • examples of cancer include, for example, leukemia, lymphoma, blastoma, carcinoma and sarcoma.
  • cancers include chronic myeloid leukemia, acute lymphoblastic leukemia, Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ ALL), squamous cell carcinoma, small-cell lung cancer, non-small cell lung cancer, glioma, gastrointestinal cancer, renal cancer, ovarian cancer, liver cancer, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, stomach cancer, bladder cancer, hepatoma, breast cancer, colon carcinoma, and head and neck cancer, gastric cancer, germ cell tumor, pediatric sarcoma, sinonasal natural killer, multiple myeloma, acute myelogenous leukemia (AML), and chronic lymphocytic leukemia (CML).
  • CML chronic lymphocytic leukemia
  • effective treatment refers to treatment producing a beneficial effect, e.g., amelioration of at least one symptom of a disease or disorder.
  • a beneficial effect can take the form of an improvement over baseline, i.e., an improvement over a measurement or observation made prior to initiation of therapy according to the method.
  • a beneficial effect can also take the form of arresting, slowing, retarding, or stabilizing of a deleterious progression of a marker of cancer.
  • Effective treatment may refer to alleviation of at least one symptom of cancer. Such effective treatment may, e.g., reduce patient pain, reduce the size and/or number of lesions, may reduce or prevent metastasis of a tumor, and/or may slow tumor growth.
  • an effective amount refers to an amount of an agent that provides the desired biological, therapeutic, and/or prophylactic result. That result can be reduction, amelioration, palliation, lessening, delaying, and/or alleviation of one or more of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • an effective amount comprises an amount sufficient to cause a tumor to shrink and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation.
  • an effective amount is an amount sufficient to delay tumor development.
  • an effective amount is an amount sufficient to prevent or delay tumor recurrence.
  • An effective amount can be administered in one or more administrations.
  • the effective amount of the drug or composition may: (i) reduce the number of cancer cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and may stop cancer cell infiltration into peripheral organs; (iv) inhibit (i.e., slow to some extent and may stop tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer.
  • an “effective amount” is the amount of a PD-1 antagonist (e.g., an antibody) and DR5 agonist antibody (e.g., an antibody), in combination, to effect a significant decrease in cancer or slowing of progression of cancer, such as an advanced solid tumor.
  • a PD-1 antagonist e.g., an antibody
  • DR5 agonist antibody e.g., an antibody
  • the term “antagonist” refers to a molecule which blocks (e.g., reduces or prevents) a biological activity.
  • agonist refers to a molecule that triggers (e.g., initiates or promotes), partially or fully enhances, stimulates or activates one or more biological activities. Agonists often mimic the action of a naturally occurring substance. Whereas an agonist causes an action, an antagonist blocks the action of the agonist.
  • ligand refers to a molecule that forms a complex with a biomolecule (e.g., a receptor) to serve a biological purpose.
  • a biomolecule e.g., a receptor
  • a signal triggering molecule binding to a site on a target protein.
  • the binding occurs by intermolecular forces, such as ionic bonds, hydrogen bonds and van der Waals forces.
  • the docking (association) is usually reversible (dissociation).
  • Ligand binding to a receptor alters its chemical conformation (three dimensional shape). The conformational state of a receptor protein determines its functional state.
  • the terms “synergy”, “therapeutic synergy”, and “synergistic effect” refer to a phenomenon where treatment of patients with a combination of therapeutic agents (e.g., PD-1 antagonist in combination with DR5 agonist) manifests a therapeutically superior outcome to the outcome achieved by each individual constituent of the combination used at its optimum dose (see, e.g., T. H. Corbett et al., 1982 , Cancer Treatment Reports, 66, 1187).
  • a combination of therapeutic agents e.g., PD-1 antagonist in combination with DR5 agonist
  • a therapeutically superior outcome is one in which the patients either a) exhibit fewer incidences of adverse events while receiving a therapeutic benefit that is equal to or greater than that where individual constituents of the combination are each administered as monotherapy at the same dose as in the combination, or b) do not exhibit dose-limiting toxicities while receiving a therapeutic benefit that is greater than that of treatment with each individual constituent of the combination when each constituent is administered in at the same doses in the combination(s) as is administered as individual components.
  • a combination, used at its maximum tolerated dose, in which each of the constituents will be present at a dose generally not exceeding its individual maximum tolerated dose manifests therapeutic synergy when decrease in tumor growth achieved by administration of the combination is greater than the value of the decrease in tumor growth of the best constituent when the constituent is administered alone.
  • antibody includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding fragments” (also known as “antigen-binding portions”)) or single chains thereof.
  • Whole antibodies are glycoproteins comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as V H ) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, C H 1, C H 2 and C H 3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as V L ) and a light chain constant region.
  • the light chain constant region is comprised of one domain, C L .
  • V H and V L regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each V H and V L is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies can mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • antibody also encompasses chimeric antibodies, humanized antibodies, fully human antibodies, as well as multimeric forms of antibodies, such as minibodies, bis-scFv, diabodies, triabodies, tetrabodies and chemically conjugated Fab′ multimers.
  • antibody fragment also referred to as “antigen-binding fragment” or “antigen-binding portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term “antigen-binding fragment” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the V L , V H , C L and C H 1 domains; (ii) a F(ab′) 2 fragment, a bivalent fragment is essentially a Fab with part of the hinge region (see, FUNDAMENTAL IMMUNOLOGY (Paul ed., 3.sup.rd ed.
  • Single domain antibodies include V H H fragments (single-domain antibodies engineered from heavy-chain antibodies found in camelids, as well as VNAR fragments (single-domain antibodies obtained from heavy-chain antibodies (IgNAR, ‘immunoglobulin new antigen receptor’) of cartilaginous fishes).
  • Antigen binding scaffolds are proteins that bind specifically to a target (or antigen) or epitope, such as proteins comprising an Ig fold or an Ig-like fold, e.g., the DR5 binding proteins described in WO2009/058379 and WO2011/130328, Antibodies or antigen binding fragments thereof are also antigen binding scaffolds.
  • Antigen binding scaffolds can be monovalent, multivalent, e.g., bivalent, trivalent, tetravalent, or bind 5, 6 or more epitopes.
  • Multivalent antigen binding scaffolds can be monospecific or multispecific, i.e., binding to multiple (at least 2, 3, 4 or 5) epitopes that are different from one another.
  • a multivalent monospecific antigen binding scaffold is a protein that binds to at least 2, 3, 4 or 5 identical epitopes, and may be a protein comprising at least 2, 3, 4 or 5 identical antigen binding portions.
  • DR5 binding scaffolds may comprise 2-10, e.g., 2-6, 2-5, 2-4 or 2-3 DR5 binding portions, which may be the same or different from one another.
  • a multivalent antibody includes antibodies comprising at least 2, 3, 4, 5, 6, 7, 8, 9, 10 or more antigen binding portions of antibodies, which antigen binding portions may comprise a portion of a heavy chain and a portion of a light chain.
  • An antigen binding portion may be on a single polypeptide or comprise more than one polypeptide.
  • a multivalent antibody may comprise from 2-10 antigen binding portions, which may be the same or different from each other.
  • a multivalent antibody may be monospecific or multispecific.
  • a multispecific antibody may be bispecific, trispecific, tetraspecific or bind to 5 or more different epitopes.
  • the two domains of the Fv fragment, V L and V H are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the V L and V H regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term “antigen-binding fragment” of an antibody.
  • an antigen binding scaffold that “specifically binds” to an antigen or epitope thereof is an antigen binding scaffold that binds to the antigen or epitope thereof with a K D of 10 ⁇ 7 M, 5 ⁇ 10 ⁇ 8 M, 10 ⁇ 8 M, 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M, 5 ⁇ 10 ⁇ 11 M, 10 ⁇ 11 M, 5 ⁇ 10 ⁇ 12 M, 10 ⁇ 12 M or less.
  • an antigen binding scaffold that specifically binds to DR5 is an antigen binding scaffold that binds to DR5 with a K D of 10 ⁇ 7 M, 5 ⁇ 10 ⁇ 8 M, 10 ⁇ 8 M, 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M, 5 ⁇ 10 ⁇ 11 M, 10 ⁇ 11 M, 5 ⁇ 10 ⁇ 12 M, 10 ⁇ 12 M or less.
  • an antibody that “specifically binds to human PD-1” or “specifically binds to human PD-L1” is intended to refer to an antibody that binds to human PD-1 or PD-L1, respectively, with a K D of 10 ⁇ 7 M, 5 ⁇ 10 ⁇ 8 M, 10 ⁇ 8 M, 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M, 5 ⁇ 10 ⁇ 11 M, 10 ⁇ 11 M, 5 ⁇ 10 ⁇ 12 M, 10 ⁇ 12 M or less.
  • An antigen binding scaffold that comprises 2 or more regions binding to an antigen or epitope may bind specifically to the antigen or epitope even it has a lower affinity of binding to the antigen or epitope than the ranges provided above, as it will bind to the antigen or epitope with increased avidity.
  • bispecific or “bifunctional antibody” is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab′ fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol. 79:315-321 (1990); Kostelny et al., J. Immunol. 148, 1547-1553 (1992).
  • monoclonal antibody or “monoclonal antibody composition,” as used herein, refers to an antibody or a composition of antibodies that displays a single binding specificity and affinity for a particular epitope. Accordingly, the term “human monoclonal antibody” or “monoclonal antibody composition” refers to an antibody or a composition of antibodies which displays a single binding specificity and which has variable and optional constant regions derived from human germline immunoglobulin sequences.
  • human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic non-human animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.
  • epitopes refers to a site on an antigen to which an immunoglobulin or antibody specifically binds.
  • Epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acids in a unique spatial conformation.
  • Methods of determining spatial conformation of epitopes include techniques in the art and those described herein, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance (see, e.g., Epitope Mapping Protocols in Methods in Molecular Biology , Vol. 66, G. E. Morris, Ed. (1996)).
  • epitopope mapping refers to the process of identification of the molecular determinants for antibody-antigen recognition.
  • the term “binds to the same epitope,” with reference to two or more antibodies, means that the antibodies compete for binding to an antigen and bind to the same, overlapping, or encompassing continuous or discontinuous segments of amino acids.
  • the phrase “binds to the same epitope” does not necessarily mean that the antibodies bind to exactly the same amino acids.
  • the precise amino acids to which the antibodies bind can differ.
  • a first antibody can bind to a segment of amino acids that is completely encompassed by the segment of amino acids bound by a second antibody.
  • a first antibody binds one or more segments of amino acids that significantly overlap the one or more segments bound by the second antibody.
  • such antibodies are considered to “bind to the same epitope.”
  • antibodies that bind to an epitope which comprises all or a portion of an epitope recognized by the particular antibodies described herein (e.g., the same or an overlapping region or a region between or spanning the region).
  • antibodies that bind the same epitope and/or antibodies that compete for binding with the antibodies described herein are also encompassed by the present invention.
  • Antibodies that recognize the same epitope or compete for binding can be identified using routine techniques. Such techniques include, for example, an immunoassay, which shows the ability of one antibody to block the binding of another antibody to a target antigen, i.e., a competitive binding assay.
  • Competitive binding is determined in an assay in which the immunoglobulin under test inhibits specific binding of a reference antibody to a common antigen.
  • RIA solid phase direct or indirect radioimmunoassay
  • EIA solid phase direct or indirect enzyme immunoassay
  • sandwich competition assay see Stahli et al., Methods in Enzymology 9:242 (1983)
  • solid phase direct biotin-avidin EIA see Kirkland et al., J. Immunol. 137:3614 (1986)
  • solid phase direct labeled assay solid phase direct labeled sandwich assay
  • solid phase direct labeled sandwich assay see Harlow and Lane, Antibodies: A Laboratory Manual , Cold Spring Harbor Press (1988)
  • solid phase direct label RIA using I-125 label see Morel et al., Mol. Immunol.
  • epitope mapping methods such as, x-ray analyses of crystals of antigen:antibody complexes which provides atomic resolution of the epitope.
  • Other methods monitor the binding of the antibody to antigen fragments or mutated variations of the antigen where loss of binding due to a modification of an amino acid residue within the antigen sequence is often considered an indication of an epitope component.
  • computational combinatorial methods for epitope mapping can also be used. These methods rely on the ability of the antibody of interest to affinity isolate specific short peptides from combinatorial phage display peptide libraries. The peptides are then regarded as leads for the definition of the epitope corresponding to the antibody used to screen the peptide library.
  • computational algorithms have also been developed which have been shown to map conformational discontinuous epitopes.
  • Chimeric molecules comprising an antigen binding domain, or equivalent, fused to another polypeptide or molecule are also encompassed by the present invention.
  • the polypeptides may be fused or conjugated to an antibody Fc region, or portion thereof (e.g., an Fc fusion protein).
  • the antibody portion fused to a polypeptide may comprise the constant region, hinge region, CH1 domain, CH2 domain, and CH3 domain or any combination of whole domains or portions thereof.
  • the polypeptides may also be fused or conjugated to the above antibody portions to form multimers.
  • Fc portions fused to the polypeptides of the present invention can form dimers through disulfide bonding between the Fc portions.
  • Higher multimeric forms can be made by fusing the polypeptides to portions of IgA and IgM.
  • Methods for fusing or conjugating the polypeptides of the present invention to antibody portions are known in the art. See, e.g., U.S. Pat. Nos. 5,336,603, 5,622,929, 5,359,046, 5,349,053, 5,447,851, and 5,112,946; EP 307,434; EP 367,166; PCT Publication Nos. WO 96/04388 and WO 91/06570; Ashkenazi et al., Proc. Natl. Acad. Sci. USA, 88:10535-10539 (1991); Zheng et al., J. Immunol., 154:5590-5600 (1995); and Vil et al., Proc. Natl. Acad. Sci. USA, 89:11337-11341 (1992).
  • immunoconjugate refers to an antibody linked to a therapeutic moiety, such as a cytotoxin, a drug or a radioisotope. When conjugated to a cytotoxin, these antibody conjugates are referred to as “immunotoxins.”
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to (e.g., kills) cells.
  • Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g.
  • Immunoconjugates can be used to modify a given biological response, and the drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • proteins may include, for example, an enzymatically active toxin, or active fragment thereof, such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor or interferon- ⁇ ; or, biological response modifiers such as, for example, lymphokines, interleukin-1 (“IL-1”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • IL-1 interleukin-1
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • GM-CSF
  • multivalent refers to a recombinant molecule that incorporates more than two biologically active segments.
  • the protein fragments forming the multivalent molecule optionally may be linked through a polypeptide linker which attaches the constituent parts and permits each to function independently.
  • “About” as used herein when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of .+ ⁇ .20% or .+ ⁇ .10%, more preferably .+ ⁇ .5%, even more preferably .+ ⁇ .1%, and still more preferably .+ ⁇ .0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.
  • Percent (%) amino acid sequence identity herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in a selected sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared.
  • the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows: 100 times the fraction X/Y where X is the number of amino acid residues scored as identical matches by a sequence alignment program, such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR), in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A.
  • the terms “Programmed Death 1,” “Programmed Cell Death 1,” “Protein PD-1,” “PD-1,” PD1,” “PDCD1,” “hPD-1” and “hPD-I” are used interchangeably, and include variants, isoforms, species homologs of human PD-1, and analogs having at least one common epitope with human PD-1.
  • the complete human PD-1 sequence can be found under GenBank Accession No. U64863 (SEQ ID NO:23).
  • the terms “Programmed Cell Death 1 Ligand 1”, “PD-L1”, “PDL1”, “PDCD1L1”, “PDCD1LG1”, “CD274”, “B7 homolog 1”, “B7-H1”, “B7-H”, and “B7H1” are used interchangeably, and include variants, isoforms, species homologs of human PDL-1, and analogs having at least one common epitope with human PDL-1.
  • the protein Programmed Death 1 is an inhibitory member of the CD28 family of receptors, that also includes CD28, CTLA-4, ICOS and BTLA. PD-1 is expressed on activated B cells, T cells, and myeloid cells (Agata et al., supra; Okazaki et al. (2002) Curr. Opin. Immunol. 14: 391779-82; Bennett et al. (2003) J Immunol 170:711-8).
  • the initial members of the family, CD28 and ICOS were discovered by functional effects on augmenting T cell proliferation following the addition of monoclonal antibodies (Hutloff et al. (1999) Nature 397:263-266; Hansen et al.
  • PD-1 was discovered through screening for differential expression in apototic cells (Ishida et al. (1992) EMBO J 11:3887-95).
  • the other members of the family, CTLA-4 and BTLA were discovered through screening for differential expression in cytotoxic T lymphocytes and TH1 cells, respectively.
  • CD28, ICOS and CTLA-4 all have an unpaired cysteine residue allowing for homodimerization.
  • PD-1 is suggested to exist as a monomer, lacking the unpaired cysteine residue characteristic in other CD28 family members.
  • the PD-1 gene is a 55 kDa type I transmembrane protein that is part of the Ig gene superfamily (Agata et al. (1996) Int Immunol 8:765-72).
  • PD-1 contains a membrane proximal immunoreceptor tyrosine inhibitory motif (ITIM) and a membrane distal tyrosine-based switch motif (ITSM) (Thomas, M. L. (1995) J Exp Med 181:1953-6; Vivier, E and Daeron, M (1997) Immunol Today 18:286-91).
  • ITIM immunoreceptor tyrosine inhibitory motif
  • ITSM membrane distal tyrosine-based switch motif
  • PD-1 lacks the MYPPPY motif (SEQ ID NO: 27) that is critical for B7-1 and B7-2 binding.
  • PD-1 deficient animals develop various autoimmune phenotypes, including autoimmune cardiomyopathy and a lupus-like syndrome with arthritis and nephritis (Nishimura et al. (1999) Immunity 11:141-51; Nishimura et al. (2001) Science 291:319-22). Additionally, PD-1 has been found to play a role in autoimmune encephalomyelitis, systemic lupus erythematosus, graft-versus-host disease (GVHD), type I diabetes, and rheumatoid arthritis (Salama et al.
  • GVHD graft-versus-host disease
  • PD-L1 and PD-L2 Two ligands for PD-1 have been identified, PD-L1 and PD-L2, that have been shown to downregulate T cell activation upon binding to PD-1 (Freeman et al. (2000) J Exp Med 192:1027-34; Latchman et al. (2001) Nat Immunol 2:261-8; Carter et al. (2002) Eur J Immunol 32:634-43). Both PD-L1 and PD-L2 are B7 homologs that bind to PD-1, but do not bind to other CD28 family members. PD-L1 is abundant in a variety of human cancers (Dong et al. (2002) Nat. Med. 8:787-9).
  • the methods of the present invention involve the use of a PD-1 antagonist (e.g., an antibody in combination with a DR5 agonist (e.g., an antibody), for treating cancer.
  • a PD-1 antagonist e.g., an antibody in combination with a DR5 agonist (e.g., an antibody)
  • PD-1 antagonists of the invention bind to ligands of PD-1 and interfere with, reduce, or inhibit the binding of one or more ligands to the PD-1 receptor, or bind directly to the PD-1 receptor, without engaging in signal transduction through the PD-1 receptor.
  • the PD-1 antagonist binds directly to PD-1 and blocks PD-1 inhibitory signal transduction.
  • the PD-1 antagonist binds to one or more ligands of PD-1 (e.g., PD-L1 and PD-L2) and reduces or inhibits the ligand(s) from triggering inhibitory signal transduction through the PD-1.
  • the PD-1 antagonist binds directly to PD-L1, inhibiting or preventing PD-L1 from binding to PD-1, thereby blocking PD-1 inhibitory signal transduction.
  • PD-1 antagonists used in the methods and compositions of the present invention include PD-1 binding scaffold proteins and include, but are not limited to, PD-1 ligands, antibodies and multivalent agents.
  • the antagonist is a fusion protein, such as AMP-224.
  • the antagonist is an anti-PD-1 antibody (“PD-1 antibody”).
  • Anti-human-PD-1 antibodies (or VH and/or VL domains derived therefrom) suitable for use in the invention can be generated using methods well known in the art. Alternatively, art recognized anti-PD-1 antibodies can be used. For example, antibodies MK-3475 or CT-011 can be used.
  • monoclonal antibodies 5C4, 17D8, 2D3, 4H1, 4A11, 7D3, and 5F4, described in WO 2006/121168, the teachings of which are hereby incorporated by reference, can be used.
  • Antibodies that compete with any of these art-recognized antibodies for binding to PD-1 also can be used.
  • an exemplary anti-PD-1 antibody is 5C4 comprising heavy and light chains having the sequences shown in SEQ ID NOs: 11 and 12, respectively, or antigen binding fragments and variants thereof.
  • the antibody comprises the heavy and light chain CDRs or variable regions of 5C4. Accordingly, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH of 5C4 having the sequence set forth in SEQ ID NO: 13, and the CDR1, CDR2 and CDR3 domains of the VL of 5C4 having the sequences set forth in SEQ ID NO: 15.
  • the antibody comprises CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 17, 18, and 19, respectively, and CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 20, 21, and 22, respectively.
  • the antibody comprises VH and/or VL regions having the amino acid sequences set forth in SEQ ID NO: 13 and/or SEQ ID NO: 15, respectively.
  • the antibody comprises the heavy chain variable (VH) and/or light chain variable (VL) regions encoded by the nucleic acid sequences set forth in SEQ ID NO: 14 and/or SEQ ID NO: 16, respectively.
  • the antibody competes for binding with and/or binds to the same epitope on PD-1 as the above-mentioned antibodies.
  • the antibody has at least about 90% variable region amino acid sequence identity with the above-mentioned antibodies (e.g., at least about 90%, 95% or 99% variable region identity with SEQ ID NO: 13 or SEQ ID NO: 15).
  • the PD1 antibodies exhibit one or more desirable functional properties, such as high affinity binding to PD-1, e.g., binding to human PD-1 with a K D of 10 ⁇ 7 M or less; lack of significant cross-reactivity to other CD28 family members, e.g., CD28, CTLA-4 and ICOS; the ability to stimulate T cell proliferation in a mixed lymphocyte reaction (MLR) assay; the ability to increase IFN- ⁇ and/or IL-2 secretion in an MLR; the ability to inhibit binding of one or more PD-1 ligands (e.g., PD-L1 and/or PD-L2) to PD-1; the ability to stimulate antigen-specific memory responses; the ability to stimulate antibody responses and/or the ability to inhibit growth of tumor cells in vivo.
  • MLR mixed lymphocyte reaction
  • the PD-1 antagonist is an anti-PD-L1 antibody.
  • Anti-human-PD-L1 antibodies (or VH and/or VL domains derived therefrom) suitable for use in the invention can be generated using methods well known in the art.
  • art recognized anti-PD-L1 antibodies can be used.
  • MEDI4736 also known as Anti-B7-H1
  • MPDL3280A also known as RG7446
  • Antibodies that compete with any of these art-recognized antibodies for binding to PD-L1 also can be used.
  • an exemplary anti-PD-L1 antibody is 12A4 (WO 2007/005874 and U.S. Pat. No. 7,943,743).
  • the antibody comprises the heavy and light chain CDRs or VRs of 12A4. Accordingly, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH region of 12A4 having the sequence shown in SEQ ID NO: 1 and the CDR1, CDR2 and CDR3 domains of the VL region of 12A4 having the sequence shown in SEQ ID NO: 3.
  • the antibody comprises the heavy chain CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 5, 6, and 7, respectively, and the light chain CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 8, 9, and 10, respectively.
  • the antibody comprises VH and/or VL regions having the amino acid sequences set forth in SEQ ID NO: 1 and/or SEQ ID NO: 3, respectively.
  • the antibody comprises the heavy chain variable (VH) and/or light chain variable (VL) regions encoded by the nucleic acid sequences set forth in SEQ ID NO: 2 and/or SEQ ID NO: 4, respectively.
  • the antibody competes for binding with, and/or binds to the same epitope on PD-L1 as, the above-mentioned antibodies.
  • the antibody has at least about 90% variable region amino acid sequence identity with the above-mentioned antibodies (e.g., at least about 90%, 95% or 99% variable region identity with SEQ ID NO: 1 or SEQ ID NO: 3).
  • Anti-PD-1 or anti-PD-L1 antibodies may bind to PD-1 or PD-L1, respectively, with a K D of 10 ⁇ 7 M, 5 ⁇ 10 ⁇ 8 M, 10 ⁇ 8 M, 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M or less.
  • apoptosis inducing agents include DR proteins, such as DR4 and DR5.
  • DR5 and “death receptor 5”, also known as “tumor necrosis factor receptor superfamily member 10b”, “TNFRSF10B”, “CD262”, “KILLER”, “TRICK2”, “TRICKB”, “ZTNFR9”, “TRAILR”, “TRAILR2”, “Apo-2” “TRICK2A”, “TRICK2B”, “TRAIL-R2”, “KILLER”, “KILLER/DR5”, “TR6”, “Tango-63”, “hAPO8”, and TRICK2 (see, e.g., Sheridan et al., Science, 277:818-821 (1997); Pan et al., Science, 277:815-818 (1997), WO98/51793; WO98/41629; Screaton et al., Curr.
  • TNF DR5 is a member of the tumor necrosis factor (TNF) receptor superfamily.
  • TNF ligands are known to be among the most pleiotropic cytokines, inducing a large number of cellular responses, including cytotoxicity, anti-viral activity, immunoregulatory activities, and the transcriptional regulation of several genes.
  • Cellular responses to TNF-family ligands include not only normal physiological responses, but also diseases associated with increased apoptosis or the inhibition of apoptosis.
  • Apoptosis i.e., programmed cell death
  • Diseases associated with increased cell survival, or the inhibition of apoptosis include cancers, autoimmune disorders, viral infections, inflammation, graft versus host disease, acute graft rejection, and chronic graft rejection.
  • Diseases associated with increased apoptosis include AIDS, neurodegenerative disorders, myelodysplastic syndromes, ischemic injury, toxin-induced liver disease, septic shock, cachexia and anorexia.
  • the death receptors are characterized by their cysteine rich domains in the extracellular region and death domains (DD) in the intracellular region. Death domain endows death receptor with function of inducing cell death by apoptosis, but sometime it also mediates other signals.
  • Tumor necrosis factor-related apoptosis-inducing ligand, TRAIL in combination with its death domains triggers two cell death signaling pathways, i.e., death receptor pathway and mitochondrion pathway, to kill various tumor cells, but is nontoxic to most normal human cells.
  • TRAIL receptors i.e., DR4 (death receptor 4 or named as TRAIL-R1), DR5, DcR1 (decoy receptor 1 or named as TRID/TRAIL-R3/LIT), DcR2 (TRAIL-R4 or named as TRUNDD), and osteoprotegerin (OPG), have been identified.
  • DR4 death receptor 4 or named as TRAIL-R1
  • DR5 contains three cysteine-rich domains in its extracellular portion and a single cytoplasmic death domain and be capable of signaling apoptosis upon ligand binding (or upon binding a molecule, such as an agonist (e.g., antibody), which mimics the activity of the ligand).
  • agonist refers to any molecule that partially or fully enhances, stimulates or activates one or more biological activities of DR5, in vitro, in situ, or in vivo. Examples of such biological activities binding of Apo2L/TRAIL to DR5, include apoptosis as well as those further reported in the literature.
  • DR5 agonists may function in a direct or indirect manner. For example, the DR5 agonist may function to partially or fully enhance, stimulate or activate one or more biological activities of DR5, in vitro, in situ, or in vivo as a result of its direct binding to DR5, which causes receptor activation or signal transduction.
  • the DR5 agonist may also function indirectly to partially or fully enhance, stimulate or activate one or more biological activities of DR5, in vitro, in situ, or in vivo as a result of, e.g., stimulating another effector molecule which then causes DR5 activation or signal transduction. It is contemplated that an agonist may act as an enhancer molecule which functions indirectly to enhance or increase DR5 activation or activity.
  • a DR5 agonist may be any molecule that directly or indirectly enhances the activity of DR5 and reduces tumor growth, whether on its own or in combination with another treatment, such as a PD-1 antagonist.
  • exemplary DR5 agonists include DR5 binding scaffolds, such as anti-DR5 antibodies (“DR5 antibodies”), e.g., chimeric, humanized or fully human antibodies, an antigen binding portion thereof, or molecules that are based on or derived from any of these.
  • DR5 agonists may also be non-antibody proteins.
  • DR5 agonist also include DR5 ligands, e.g., TRAIL and molecules that are derived from or based on TRAIL.
  • a DR5 agonist may be monovalent or multivalent.
  • a DR5 agonist is bivalent, trivalent, tetravalent, or binds to 5, 6, 7, 8, 9, 10 or more DR5 epitopes, which may be the same or different DR5 epitopes.
  • a DR5 agonist is a multivalent monospecific DR5 binding scaffold, e.g., a protein comprising a DR5 binding scaffold that comprises at least 2, 3, 4, 5, 6, 7, 8, 9, 10 or more regions that specifically bind to the same DR5 epitope, which binding regions may be composed of the same or a different amino acid sequence.
  • a DR5 agonist may be a DR5 binding scaffold comprising 2, 3, 4, 5, 6, 7, 8, 9, 10 or more repeats of the same DR5 binding region.
  • Multimeric DR5 binding scaffolds are described, e.g., in WO2009/058379, WO2011/130328, WO2010/042890 and WO2011/098520.
  • a DR5 agonist binds specifically to DR5, but does not bind significantly or specifically to other members of the TNF receptor superfamily, such as DR4. In other embodiments, a DR5 agonist binds specifically to DR5 and DR4.
  • the DR5 agonist is a recombinant human TRAIL (TNF-related apoptosis-inducing ligand), e.g., Dulanermin (also known as AMG-951; available from Amgen/Genentech).
  • TRAIL TNF-related apoptosis-inducing ligand
  • Dulanermin also known as AMG-951; available from Amgen/Genentech.
  • the DR5 agonist is an antibody, e.g., an antibody that binds to human DR5 with a K D of 10 ⁇ 7 M, 5 ⁇ 10 ⁇ 8 M, 10 ⁇ 8 M, 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M or less, wherein the antibody inhibits tumor growth and/or induces apoptosis of tumor cells.
  • an antibody e.g., an antibody that binds to human DR5 with a K D of 10 ⁇ 7 M, 5 ⁇ 10 ⁇ 8 M, 10 ⁇ 8 M, 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M or less, wherein the antibody inhibits tumor growth and/or induces apoptosis of tumor cells.
  • Numerous antibodies binding to human DR5 are known in the art and some of them have been used in clinical trials. Any of these antibodies may be used in combination with a PD-1 antagonist, provided that their combination
  • Exemplary antibodies that bind specifically to human DR5 include Conatumumab (a hTRAILR2-specific antibody also known as AMG655; available from Amgen), Drozitumab (a hTRAILR2-specific antibody also known as Apomab, DAB4, and PRO95780; available from Genentech), Lexatumumab (a hTRAILR2-specific antibody also known as HGS-ETR2; available from HGS/Kirin), Tigatuzumab (a humanized TRAILR2-specific antibody also known as CS-1008 and TRA-8; available from Daiichi Sankyo), HGSTR2J (a hTRAILR2-specific antibody also known as KMTRS), or LBY-135 (a TRAILR2-specific Ab; available from Novartis) (see, e.g., Ashkenazi et al., Journal of Clinical Investigation 2008; 118:1979-90).
  • the DR5 agonist is a bispecific death receptor agonist antibody, see, e.g., WO2011/039126; available from Roche Glycart).
  • the DR5 agonist is an antibody conjugated to targeting peptides or a cytotoxin, Fc-human TRAIL ligand fusion (see, e.g., WO2011/039126; available from Roche Glycart).
  • the DR5 agonist is a high affinity Fc-polypeptides (see, e.g., WO2011/143614; available from Amgen).
  • the DR5 agonist is a multivalent agent, such as TAS266 (a tetrameric nanobody agonist targeting DR5, see, e.g., WO2011/098520 and Cancer Research 2012; 72:Supplement 1; Abstract 3852; available from Novartis and Ablynx), multimeric Tn3 protein (see, e.g., WO2009/058379, WO2011/130328, and Cancer Research 2012; 72:Supplement 1; Abstract 239; available from Medimmune), a multimer (e.g., a polypeptide construct with trimerizing domain and a polypeptide that binds DR5; see WO2010/042890; available from Anaphore).
  • TAS266 a tetrameric nanobody agonist targeting DR5
  • multimeric Tn3 protein see, e.g., WO2009/058379, WO2011/130328, and Cancer Research 2012; 72:Supplement 1; Abstract 239; available from Medimmune
  • Agents which compete for binding to DR5 with any of the exemplary agents listed herein, and which inhibit tumor growth or reduces tumor size may also be used.
  • a DR5 agonist is replaced with a DR4 agonist.
  • a subject having cancer is treated with a combination of a DR4 agonist and a PD-1 antagonist.
  • any agent that induces apoptosis in tumor cells can be combined with a PD-1 antagonist for treating cancer.
  • an apoptosis inducing agent is an agent that binds specifically to DR5 and DR4, such as TRAIL or an agent that mimics TRAIL.
  • An exemplary DR4 agonist is Mapatumumab (HGS-ETR1), which has been used in phase 2 clinical trials.
  • the present invention provides composition comprising a PD-1 antagonist and a DR5 agonist (e.g., formulated together in a single composition or separately formulated).
  • the composition comprises a PD-1 antagonist and a DR5 agonist, wherein (a) the PD-1 antagonist is an anti-PD-1 antibody comprising the CDR1, CDR2 and CDR3 domains in a heavy chain variable region having the sequence set forth in SEQ ID NO: 13, and the CDR1, CDR2 and CDR3 domains in a light chain variable region having the sequence set forth in SEQ ID NO: 15; and (b) the DR5 agonist is an antibody.
  • the composition comprises a PD-1 antagonist and a DR5 agonist, wherein (a) the PD-1 antagonist is an anti-PD-L1 antibody comprising the CDR1, CDR2 and CDR3 domains in a heavy chain variable region having the sequence set forth in SEQ ID NO: 1, and the CDR1, CDR2 and CDR3 domains in a light chain variable region having the sequence set forth in SEQ ID NO: 3 and (b) the DR5 agonist is an antibody.
  • compositions suitable for administration to human patients are typically formulated for parenteral administration, e.g., in a liquid carrier, or suitable for reconstitution into liquid solution or suspension for intravenous administration.
  • compositions typically comprise a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a government regulatory agency or listed in the U.S. Pharmacopeia or another generally recognized pharmacopeia for use in animals, particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, glycerol polyethylene glycol ricinoleate, and the like.
  • Liquid compositions for parenteral administration can be formulated for administration by injection or continuous infusion. Routes of administration by injection or infusion include intravenous, intraperitoneal, intramuscular, intrathecal and subcutaneous.
  • the pharmaceutical compositions of the present invention may be administered, for example, in the form of tablets or capsules, powders, dispersible granules, or cachets, or as aqueous solutions or suspensions.
  • carriers which are commonly used include lactose, corn starch, magnesium carbonate, talc, and sugar, and lubricating agents such as magnesium stearate are commonly added.
  • useful carriers include lactose, corn starch, magnesium carbonate, talc, and sugar.
  • emulsifying and/or suspending agents are commonly added.
  • sweetening and/or flavoring agents may be added to the oral compositions.
  • sterile solutions of the active ingredient(s) are usually employed, and the pH of the solutions should be suitably adjusted and buffered.
  • the total concentration of the solute(s) should be controlled in order to render the preparation isotonic.
  • a low melting wax such as a mixture of fatty acid glycerides or cocoa butter is first melted, and the active ingredient is dispersed homogeneously in the wax, for example by stirring. The molten homogeneous mixture is then poured into conveniently sized molds and allowed to cool and thereby solidify.
  • Liquid preparations include solutions, suspensions and emulsions. Such preparations are exemplified by water or water/propylene glycol solutions for parenteral injection. Liquid preparations may also include solutions for intranasal administration.
  • Aerosol preparations suitable for inhalation may include solutions and solids in powder form, which may be in combination with a pharmaceutically acceptable carrier, such as an inert compressed gas.
  • a pharmaceutically acceptable carrier such as an inert compressed gas.
  • solid preparations which are intended for conversion, shortly before use, to liquid preparations for either oral or parenteral administration.
  • liquid forms include solutions, suspensions and emulsions.
  • a patient e.g., using a combination of a DR5 agonist and PD-1 antagonist.
  • the patient suffers from a cancer selected from the group consisting of leukemia, lymphoma, blastoma, carcinoma and sarcoma.
  • the patient suffers from a cancer selected from the group consisting of chronic myeloid leukemia, acute lymphoblastic leukemia, Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ ALL), squamous cell carcinoma, small-cell lung cancer, non-small cell lung cancer, glioma, gastrointestinal cancer, renal cancer, ovarian cancer, liver cancer, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, stomach cancer, bladder cancer, hepatoma, breast cancer, colon carcinoma, and head and neck cancer, gastric cancer, germ cell tumor, pediatric sarcoma, sinonasal natural killer, multiple myeloma, acute myelogenous leukemia (AML), and chronic lymphocytic leukemia (CML).
  • a cancer selected from the group consisting of chronic myeloid leukemia, acute lymphoblastic leukemia, Philadelphia chro
  • combinations of the present invention may also be used in conjunction with other well known therapies that are selected for their particular usefulness against the cancer that is being treated.
  • Combinations of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when inappropriate.
  • the PD-1 antagonists and DR5 agonists described herein can further be used in combination (e.g., simultaneously or separately) with an additional treatment, such as irradiation, chemotherapy (e.g., using camptothecin (CPT-11), 5-fluorouracil (5-FU), cisplatin, doxorubicin, irinotecan, paclitaxel, gemcitabine, cisplatin, paclitaxel, doxorubicin, 5-fu, or camptothecin+apo21/TRAIL (a 6 ⁇ combo)), one or more proteasome inhibitors (e.g., bortezomib or MG132), one or more Bcl-2 inhibitors (e.g., BH3I-2′ (bcl-xl inhibitor), AT-101 (R-( ⁇ )-gossypol derivative), ABT-263 (small molecule), GX-15-070 (obatoclax), or MCL-1 (myeloid leukemia cell
  • the PD-1 antagonists and DR5 agonists described herein can further be used in combination with one or more anti-proliferative cytotoxic agents.
  • Classes of compounds that may be used as anti-proliferative cytotoxic agents include, but are not limited to, the following:
  • Alkylating agents including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes: Uracil mustard, Chlormethine, Cyclophosphamide (CYTOXANTM) fosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylenemelamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, dacarbazine, and Temozolomide.
  • Uracil mustard Chlormethine
  • Melphalan Chlorambucil
  • Pipobroman Triethylenemelamine
  • Triethylenethiophosphoramine Triethylenethiophosphoramine
  • Busulfan Carmustine, Lomustine, Streptozocin, dacarbazine, and Temozolomide.
  • Antimetabolites including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors: Methotrexate, 5-Fluorouracil, Floxuridine, Cytarabine, 6-Mercaptopurine, 6-Thioguanine, Fludarabine phosphate, Pentostatine, and Gemcitabine.
  • Suitable anti-proliferative agents for use in the methods of the invention include, without limitation, taxanes, paclitaxel (paclitaxel is commercially available as TAXOL®), docetaxel, discodermolide (DDM), dictyostatin (DCT), Peloruside A, epothilones, epothilone A, epothilone B, epothilone C, epothilone D, epothilone E, epothilone F, furanoepothilone D, desoxyepothilone B1, [17]-dehydrodesoxyepothilone B, [18]dehydrodesoxyepothilones B, C12,13-cyclopropyl-epothilone A, C6-C8 bridged epothilone A, trans-9,10-dehydroepothilone D, cis-9,10-dehydroepothilone D, 16-desmethylepot
  • hormones and steroids can also be administered to the patient.
  • steroids including synthetic analogs
  • other agents used in the modulation of tumor growth or metastasis in a clinical setting such as antimimetics
  • chemotherapeutic agents are known to those skilled in the art. In addition, their administration is described in the standard literature. For example, the administration of many of the chemotherapeutic agents is described in the Physicians' Desk Reference (PDR), e.g., 1996 edition (Medical Economics Company, Montvale, N.J. 07645-1742, USA); the disclosure of which is incorporated herein by reference thereto.
  • PDR Physicians' Desk Reference
  • the chemotherapeutic agent(s) and/or radiation therapy can be administered according to therapeutic protocols well known in the art. It will be apparent to those skilled in the art that the administration of the chemotherapeutic agent(s) and/or radiation therapy can be varied depending on the disease being treated and the known effects of the chemotherapeutic agent(s) and/or radiation therapy on that disease. Also, in accordance with the knowledge of the skilled clinician, the therapeutic protocols (e.g., dosage amounts and times of administration) can be varied in view of the observed effects of the administered therapeutic agents on the patient, and in view of the observed responses of the disease to the administered therapeutic agents.
  • the therapeutic protocols e.g., dosage amounts and times of administration
  • Suitable treatment protocols for treating cancer in a patient include, for example, administering to the patient an effective amount of a PD-1 antagonist (e.g., antibody) and a DR5 agonist (e.g., antibody).
  • a PD-1 antagonist e.g., antibody
  • a DR5 agonist e.g., antibody
  • adjunctive or combined administration includes simultaneous administration of the antagonist and agonist in the same or different dosage form, or separate administration of the antagonist and agonist (e.g., sequential administration).
  • the PD-1 antagonist e.g., antibody
  • DR5 agonist e.g., antibody
  • the PD-1 antagonist and DR5 agonist can be simultaneously administered in a single formulation.
  • the PD-1 antagonist and DR5 agonist can be formulated for separate administration and are administered concurrently or sequentially.
  • the PD-1 antagonist can be administered first followed by (e.g., immediately followed by) the administration of the DR5 agonist, or vice versa.
  • the PD-1 antagonist is administered prior to administration of the DR5 agonist.
  • the DR5 agonist is administered prior to administration of the PD-1 antagonist.
  • concurrent or sequential administration preferably results in both the agonist and antagonist being simultaneously present in treated patients.
  • the DR5 agonist and the PD-1 antagonist are administered simultaneously.
  • a subject is dosed with a single dose of a DR5 agonist and at least 2 doses of a PD-1 antagonist, e.g., an anti-PD-1 or anti-PD-L1 antibody.
  • a subject receives a single dose of a DR5 agonist and at least 2, 3, 4, 5, or more doses of a PD-1 antagonist.
  • the multiple doses of PD-1 antagonist may be provided as one dose per day, one dose every 2 days, one dose every 3 days, one dose every 4 days, one dose every 5 days or less frequently.
  • the single dose of DR5 agonist may be provided on a day on which the PD-1 antagonist is provided or on a day on which it is not provided.
  • the total number of doses of PD-1 antagonist may be 2, 3, 4, 5, 6, 7, 8, 9, 10 or more.
  • multiple (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) doses of a DR5 agonist and multiple (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) doses of a PD-1 antagonist are administered to a subject in need of treatment.
  • Administration of the DR5 agonist and the PD-1 antagonist may be on the same day, or alternatively, the DR5 antagonist may be administered 1 or more days before or after the PD-1 antagonist.
  • Administrations of a DR5 agonist and a PD-1 antagonist may also be done weekly or monthly, in which regimen, they may be administered on the same day (e.g., simultaneously), or one after the other (e.g., one or more days before or after one another)
  • the dose of the PD-1 antagonist and/or DR5 agonist is varied over time.
  • the PD-1 antagonist and/or DR5 agonist may be initially administered at a high dose and may be lowered over time.
  • the the PD-1 antagonist and/or DR5 agonist is initially administered at a low dose and increased over time.
  • the amount of the PD-1 antagonist and/or DR5 agonist administered is constant for each dose. In another embodiment, the amount of the PD-1 antagonist and/or DR5 agonist varies with each dose. For example, the maintenance (or follow-on) dose of the antagonist and/or agonist can be higher or the same as the loading dose which is first administered. In another embodiment, the maintenance dose of the antagonist and/or agonist can be lower or the same as the loading dose.
  • a clinician may utilize preferred dosages as warranted by the condition of the patient being treated. The dose of may depend upon a number of factors, including stage of disease, etc. The specific dose that should be administered based upon the presence of one or more of such factors is within the skill of the artisan.
  • treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small amounts until the optimum effect under the circumstances is reached.
  • the total daily dosage may be divided and administered in portions during the day if desired. Intermittent therapy (e.g., one week out of three weeks or three out of four weeks) may also be used.
  • the DR5 agonist e.g., antibody
  • the PD-1 antagonist e.g., antibody
  • the PD-1 antagonist is administered at a dose of 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 mg/kg body weight.
  • 200 ⁇ g/mouse is approximately 10 mg/kg and 100 ⁇ g/mouse is approximately 5 mg/kg.
  • one or more doses of 1-20 mg/kg body weight, 1-10 mg/kg body weight, 5-20 mg/kg body weight or 5-10 mg/kg body weight of a DR5 agonist and PD-1 antagonist may be administered to a subject.
  • a dose of 0.3 mg/kg to 10 mg/kg body weight of a DR5 agonist is used and a dose of at least 1 mg/kg, e.g., 1-100 mg/kg body weight of a PD-1 antagonist is used.
  • Patients, e.g., humans, treated according to the methods disclosed herein preferably experience improvement in at least one sign of cancer.
  • improvement is measured by a reduction in the quantity and/or size of measurable tumor lesions.
  • lesions can be measured on chest x-rays or CT or MRI films.
  • cytology or histology can be used to evaluate responsiveness to a therapy.
  • the patient treated exhibits a reduction in size of a tumor, reduction in number of metastasic lesions over time, complete response, partial response, and stable disease.
  • the patient treated experiences tumor shrinkage and/or decrease in growth rate, i.e., suppression of tumor growth.
  • unwanted cell proliferation is reduced or inhibited.
  • CBR CR (complete response), PR (partial response) or SD (stable disease) ⁇ 6 months
  • the improvement of clinical benefit rate is about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% or more, e.g., compared to treatment with a PD-1 antagonist or DR5 agonist alone or relative to tumor growth on the first day of treatment or immediately before initiation of treatment.
  • administration of a PD-1 antagonist and a DR5 agonist results in at least a three-fold reduction (e.g., a 3.5-fold reduction) in tumor volume, e.g., relative to treatment with the PD-1 antagonist or the DR5 agonist alone or relative to tumor growth on the first day of treatment or immediately before initiation of treatment.
  • a three-fold reduction e.g., a 3.5-fold reduction
  • administration of a PD-1 antagonist and a DR5 agonist results in tumor growth inhibition of at least 80%, e.g., relative to treatment with the PD-1 antagonist or DR5 agonist alone or relative to tumor growth on the first day of treatment or immediately before initiation of treatment.
  • administration of a PD-1 antagonist and a DR5 agonist reduces tumor mass by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% relative to the tumor mass prior to initiation of the treatment or on the first day of treatment.
  • the tumor mass is no longer detectable following treatment as described herein.
  • a subject is in partial or full remission.
  • kits which include a pharmaceutical composition containing (a) a PD-1 antagonist and (b) a DR5 agonist and a pharmaceutically-acceptable carrier, in a therapeutically effective amount adapted for use in the preceding methods.
  • the PD-1 antagonist antagonist is an antibody (e.g., 5C4 or 12A4, respectively).
  • the DR5 agonist is an antibody.
  • the kits optionally also can include instructions, e.g., comprising administration schedules, to allow a practitioner (e.g., a physician, nurse, or patient) to administer the composition contained therein to a patient having cancer.
  • the kit also can include a syringe.
  • the present invention provides a kit for treating cancer in a patient, the kit comprising:
  • the DR5 agonist is an antibody.
  • the PD-1 antagonist is an antibody.
  • the PD-1 antagonist is an anti-PD-1 antibody comprising the CDR1, CDR2 and CDR3 domains in a heavy chain variable region having the sequence set forth in SEQ ID NO: 13, and the CDR1, CDR2 and CDR3 domains in a light chain variable region having the sequence set forth in SEQ ID NO: 15.
  • the PD-1 antagonist is an anti-PD-L1 antibody comprising antibody comprises the CDR1, CDR2 and CDR3 domains in a heavy chain variable region having the sequence set forth in SEQ ID NO: 1, and the CDR1, CDR2 and CDR3 domains in a light chain variable region having the sequence set forth in SEQ ID NO: 3.
  • mice Ten to eleven-week-old female C57/BL6 mice (Harlan) were used in the studies. Mice received food and water ad libitum and were maintained in a controlled environment according to Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC) International regulations. All animal studies have been approved by the appropriate ethics committee and have therefore been performed in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki and its later amendments.
  • AALAC Laboratory Animal Care
  • Anti-mouse PD-1 mAb (anti-mPD-1 mAb) clone 4H2, mouse IgG1 isotype was produced and purified by Bristol-Myers Squibb (Biologics Discovery, CA).
  • Stock solutions of anti-mPD-1 mAb and anti-mDR5 antibody were kept at 4° C. prior to use. Dosing solutions of anti-mPD-1 mAb and anti-mDR5 mAb were prepared in sterile phosphate buffered saline (pH 7.0) and maintained at 4° C.
  • Anti-mPD-1 mAb was administered intraperitoneally at its optimal dose of 10 mg/kg; anti-DR5 mAb at 5 mg/kg.
  • the MC38 colon carcinoma tumor line used in this study was maintained in vitro.
  • Cell suspensions were implanted in the subcutaneous space of the flank of mice of female C57/BL6 mice (2.0 ⁇ 10 6 MC-38 cells in 0.2 mL Hanks Balanced Salt Solution).
  • the tumor response endpoint was expressed as tumor growth delay (T ⁇ C value), calculated as the difference in time (days) between the treated (T) and control (C) groups for the tumor to reach a predetermined target size.
  • T ⁇ C value tumor growth delay
  • a delay in reaching target size by the treated groups of >1 times tumor volume doubling time was considered an active result.
  • Therapeutic synergy was defined as an antitumor effect in which the combination of agents demonstrated significant superiority (p ⁇ 0.05) relative to the activity shown by each agent alone.
  • mice Six days post tumor implant, mice were sorted into eight groups of 7 mice with a mean tumor volume of ⁇ 200 mm 3 . Antibodies were administered according to the dosing schedules descried in Table 1.
  • mice 1 Control 7 2: Control + anti-PD-1 mAb; 200 ug/mouse; 7 Q4D ⁇ 3 dosing initiated day 6 3: Control + anti-PD-1 mAb; 200 ug/mouse; 7 Q4D ⁇ 3 dosing initiated day 8 4: Control + anti-PD-1 mAb; 200 ug/mouse; 7 Q4D ⁇ 3 dosing initiated day 9 5: Anti-DR5 mAb; 100 ug/mouse; QD + 7 Control dosing initiated day 8 6: Anti-DR5 mAb; 100 ug/mouse dosed 7 day 8; QD + anti-PD-1 mAb; 200 ug/mouse; Q4D ⁇ 3 dosed day 6 7: Anti-DR5 mAb; 100 ug/mouse dosed 7 day 8; QD + anti-PD-1 mAb; 200 ug/mouse; Q4D ⁇ 3 dosed day 6 7: Anti-DR5 mAb;
  • the medium tumor volume (measured in mm 3 ) in mice treated with a combination of the DR5 mAb and the PD-1 mAb was significantly reduced, compared to mice treated with a control or either agent alone. Specifically, there was about a 3.5 fold reduction (e.g., at least a 3 fold reduction) in tumor volume in mice treated with both the DR5 mAb and the PD-1 mAb, compared to mice treated with a control or either agent alone.
  • the tumor volume in individual mice is shown in FIG. 2 .
  • the combination of the DR5 mAb and the PD-1 mAb resulted in enhanced activity compared to the activity elicited by single agents alone, independent of the schedule utilized. This synergy was statistically significant (p ⁇ 0.05, Wilcoxon). Out of the 3 administration schedules tested, a trend for better activity was observed in the groups which were treated with anti-mDR5 first or with anti-mPD-1 first. As shown in FIG. 3 , the combination therapy was well-tolerated (no significant body weight loss). In previous studies, significant body weight loss (>20%) was observed with multiple doses of the DR5 mAb alone or in combination.
  • results from this study demonstrate that a combination regimen that includes a single dose of anti-mDR5 mAb and multiple doses of PD-1 mAb is well-tolerated and result in marked antitumor activity.
  • VH Heavy Chain Variable Region
  • Amino Acid Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and U.S. Pat. No. 7,943,743)
  • QQRSNWPT 11 Heavy Chain Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (variable region underlined; constant region bold) QVQLVESGGGVVQPGRSLRLDCKASGITFSNSGMHWVRQAPGKGL EWVAVIWYDGSKRYYADSVKGRFTISRDNSKNTLFLQMNSLRAEDT AVYYCATNDDYWGQGTLVTVSS ASTKGPSVFPLAPCSRSTSESTAA LGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVT VPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFLG GPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGV EVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKG LPSSIEKTISKAKGQPREPQVYTLPPS
  • NP_054862.1 MRIFAVFIFM TYWHLLNAFT VTVPKDLYVV EYGSNMTIEC KFPVEKQLDL AALIVYWEME DKNIIQFVHG EEDLKVQHSS YRQRARLLKD QLSLGNAALQ ITDVKLQDAG VYRCMISYGG ADYKRITVKV NAPYNKINQR ILVVDPVTSE HELTCQAEGY PKAEVIWTSS DHQVLSGKTT TTNSKREEKL FNVTSTLRIN TTTNEIFYCT FRRLDPEENH TAELVIPELP LAHPPNERTH LVILGAILLC LGVALTFIFR LRKGRMMDVK KCGIQDTNSK KQSDTHLEET 25 Human PD-L1 amino acid sequence-isoform b precursor (GenBank Accession No.
  • NP_001254635.1 MRIFAVFIFM TYWHLLNAPY NKINQRILVV DPVTSEHELT CQAEGYPKAE VIWTSSDHQV LSGKTTTTNS KREEKLFNVT STLRINTTTN EIFYCTFRRL DPEENHTAEL VIPELPLAHP PNERTHLVIL GAILLCLGVA LTFIFRLRKG RMMDVKKCGI QDTNSKKQSD THLEET 26 Human DR5 amino acid sequence (GenBank Accession No.

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)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Biomedical Technology (AREA)
  • Endocrinology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US14/774,382 2013-03-14 2014-03-12 Combination of dr5 agonist and anti-pd-1 antagonist and methods of use Abandoned US20160067337A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/774,382 US20160067337A1 (en) 2013-03-14 2014-03-12 Combination of dr5 agonist and anti-pd-1 antagonist and methods of use

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361783184P 2013-03-14 2013-03-14
US14/774,382 US20160067337A1 (en) 2013-03-14 2014-03-12 Combination of dr5 agonist and anti-pd-1 antagonist and methods of use
PCT/US2014/024208 WO2014159562A1 (en) 2013-03-14 2014-03-12 Combination of dr5 agonist and anti-pd-1 antagonist and methods of use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/024208 A-371-Of-International WO2014159562A1 (en) 2013-03-14 2014-03-12 Combination of dr5 agonist and anti-pd-1 antagonist and methods of use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/993,687 Continuation US11103579B2 (en) 2013-03-14 2018-05-31 Combination of DR5 agonist and anti-PD-1 antagonist and methods of use

Publications (1)

Publication Number Publication Date
US20160067337A1 true US20160067337A1 (en) 2016-03-10

Family

ID=50483524

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/774,382 Abandoned US20160067337A1 (en) 2013-03-14 2014-03-12 Combination of dr5 agonist and anti-pd-1 antagonist and methods of use
US15/993,687 Active 2034-08-13 US11103579B2 (en) 2013-03-14 2018-05-31 Combination of DR5 agonist and anti-PD-1 antagonist and methods of use
US17/387,399 Abandoned US20220098302A1 (en) 2013-03-14 2021-07-28 Combination of dr5 agonist and anti-pd-1 antagonist and methods of use

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/993,687 Active 2034-08-13 US11103579B2 (en) 2013-03-14 2018-05-31 Combination of DR5 agonist and anti-PD-1 antagonist and methods of use
US17/387,399 Abandoned US20220098302A1 (en) 2013-03-14 2021-07-28 Combination of dr5 agonist and anti-pd-1 antagonist and methods of use

Country Status (15)

Country Link
US (3) US20160067337A1 (el)
EP (2) EP3305812B1 (el)
CY (1) CY1119510T1 (el)
DK (1) DK2970473T3 (el)
ES (2) ES2644022T3 (el)
HR (1) HRP20171758T1 (el)
HU (1) HUE035865T2 (el)
LT (1) LT2970473T (el)
ME (1) ME02917B (el)
NO (1) NO3080374T3 (el)
PL (1) PL2970473T3 (el)
PT (1) PT2970473T (el)
RS (1) RS56531B1 (el)
SI (1) SI2970473T1 (el)
WO (1) WO2014159562A1 (el)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017173360A3 (en) * 2016-03-31 2017-11-09 Stcube, Inc. Combination treatments directed toward programmed death ligand-1 (pd-l1) positive cancers
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10441654B2 (en) 2014-01-24 2019-10-15 Children's Hospital Of Eastern Ontario Research Institute Inc. SMC combination therapy for the treatment of cancer
WO2020150623A1 (en) * 2019-01-18 2020-07-23 University Of Georgia Research Foundation, Inc. Salt nanoparticles and compositions and methods of use thereof
US11103579B2 (en) 2013-03-14 2021-08-31 Bristol-Myers Squibb Company Combination of DR5 agonist and anti-PD-1 antagonist and methods of use
WO2022081869A1 (en) * 2020-10-14 2022-04-21 University Of Virginia Patent Foundation Compositions and methods for overcoming dr5-induced immune evasion by solid tumors

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2593027T3 (es) 2009-03-30 2016-12-05 Eisai R&D Management Co., Ltd. Composición liposomal
CN104470949A (zh) * 2012-05-15 2015-03-25 百时美施贵宝公司 通过破坏pd-1/pd-l1信号传输的免疫治疗
WO2015085847A1 (zh) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Pd-1抗体、其抗原结合片段及其医药用途
TWI680138B (zh) 2014-01-23 2019-12-21 美商再生元醫藥公司 抗pd-l1之人類抗體
TWI681969B (zh) 2014-01-23 2020-01-11 美商再生元醫藥公司 針對pd-1的人類抗體
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
GB201419084D0 (en) 2014-10-27 2014-12-10 Agency Science Tech & Res Anti-PD-1 antibodies
TWI595006B (zh) * 2014-12-09 2017-08-11 禮納特神經系統科學公司 抗pd-1抗體類和使用彼等之方法
AU2015380455A1 (en) 2015-01-26 2017-08-03 Macrogenics, Inc. Multivalent molecules comprising DR5-binding domains
EP3265122B1 (en) * 2015-03-04 2022-05-04 Merck Sharp & Dohme Corp. Combination of pembrolizumab and eribulin for treating triple-negative breast cancer
EP3744340A3 (en) 2015-07-16 2021-03-03 Biokine Therapeutics Ltd. Compositions and methods for treating cancer
CN108697791B (zh) 2015-11-03 2022-08-23 詹森生物科技公司 特异性结合pd-1的抗体及其用途
AU2016370376B2 (en) 2015-12-14 2023-12-14 Macrogenics, Inc. Bispecific molecules having immunoreactivity with PD-1 and CTLA-4, and methods of use thereof
SI3394103T1 (sl) 2015-12-22 2023-10-30 Regeneron Pharmaceuticals, Inc. Kombinacija protiteles proti-PD-1 in bispecifičnih protiteles proti-CD20/proti-CD3 za zdravljenje raka
ES2952680T3 (es) 2016-04-28 2023-11-03 Eisai R&D Man Co Ltd Eribulina para inhibir el crecimiento tumoral
TWI755395B (zh) 2016-05-13 2022-02-21 美商再生元醫藥公司 抗-pd-1抗體與輻射治療癌症之組合
WO2018029474A2 (en) 2016-08-09 2018-02-15 Kymab Limited Anti-icos antibodies
US9567399B1 (en) 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
CN116903741A (zh) 2016-06-20 2023-10-20 科马布有限公司 特异性结合于pd-l1的抗体或其抗原结合片段及其用途
TWI760352B (zh) 2016-08-09 2022-04-11 英商克馬伯有限公司 抗icos抗體
WO2018083248A1 (en) 2016-11-03 2018-05-11 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses & methods
WO2018167780A1 (en) 2017-03-12 2018-09-20 Yeda Research And Development Co. Ltd. Methods of prognosing and treating cancer
EP3596469A1 (en) 2017-03-12 2020-01-22 Yeda Research and Development Co., Ltd. Methods of diagnosing and prognosing cancer
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
GB201709808D0 (en) 2017-06-20 2017-08-02 Kymab Ltd Antibodies
GB201721338D0 (en) 2017-12-19 2018-01-31 Kymab Ltd Anti-icos Antibodies
US11629189B2 (en) 2017-12-19 2023-04-18 Kymab Limited Bispecific antibody for ICOS and PD-L1
US20210079102A1 (en) * 2018-02-08 2021-03-18 Dragonfly Therapeutics, Inc. Combination therapy of cancer involving multi-specific binding proteins that activate natural killer cells
CA3091140A1 (en) * 2018-02-26 2019-08-29 Igm Biosciences, Inc. Use of a multimeric anti-dr5 binding molecule in combination with a chemotherapeutic agent for treating cancer
CN108840932B (zh) * 2018-04-28 2022-03-29 中国科学院微生物研究所 一种pd-1特异性抗体及其抗肿瘤应用
EP3866812A1 (en) 2018-10-17 2021-08-25 BioLineRx Ltd. Treatment of metastatic pancreatic adenocarcinoma
US12036204B2 (en) 2019-07-26 2024-07-16 Eisai R&D Management Co., Ltd. Pharmaceutical composition for treating tumor
US11083705B2 (en) 2019-07-26 2021-08-10 Eisai R&D Management Co., Ltd. Pharmaceutical composition for treating tumor
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
CN110982824B (zh) * 2019-10-09 2022-04-15 天津大学 拮抗pd-1的抗体类似物bp基因及蛋白和应用
US20230293530A1 (en) 2020-06-24 2023-09-21 Yeda Research And Development Co. Ltd. Agents for sensitizing solid tumors to treatment
CA3182579A1 (en) 2020-07-07 2022-01-13 Ugur Sahin Therapeutic rna for hpv-positive cancer
WO2022135666A1 (en) 2020-12-21 2022-06-30 BioNTech SE Treatment schedule for cytokine proteins
TW202245808A (zh) 2020-12-21 2022-12-01 德商拜恩迪克公司 用於治療癌症之治療性rna
WO2022135667A1 (en) 2020-12-21 2022-06-30 BioNTech SE Therapeutic rna for treating cancer
TW202304506A (zh) 2021-03-25 2023-02-01 日商安斯泰來製藥公司 涉及抗claudin 18.2抗體的組合治療以治療癌症
TW202313682A (zh) 2021-05-18 2023-04-01 英商凱麥博有限公司 抗icos抗體之用途
GB202107994D0 (en) 2021-06-04 2021-07-21 Kymab Ltd Treatment of cancer
CA3225254A1 (en) 2021-07-13 2023-01-19 BioNTech SE Multispecific binding agents against cd40 and cd137 in combination therapy for cancer
WO2023051926A1 (en) 2021-09-30 2023-04-06 BioNTech SE Treatment involving non-immunogenic rna for antigen vaccination and pd-1 axis binding antagonists
TW202333802A (zh) 2021-10-11 2023-09-01 德商拜恩迪克公司 用於肺癌之治療性rna(二)
WO2023083439A1 (en) 2021-11-09 2023-05-19 BioNTech SE Tlr7 agonist and combinations for cancer treatment
WO2023222854A1 (en) 2022-05-18 2023-11-23 Kymab Limited Uses of anti-icos antibodies
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
WO2024126457A1 (en) 2022-12-14 2024-06-20 Astellas Pharma Europe Bv Combination therapy involving bispecific binding agents binding to cldn18.2 and cd3 and immune checkpoint inhibitors
WO2024160721A1 (en) 2023-01-30 2024-08-08 Kymab Limited Antibodies

Family Cites Families (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1763223A (en) 1927-04-02 1930-06-10 Owens Illinois Glass Co Glassware-annealing leer
EP0307434B2 (en) 1987-03-18 1998-07-29 Scotgen Biopharmaceuticals, Inc. Altered antibodies
US5336603A (en) 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
KR900005995A (ko) 1988-10-31 1990-05-07 우메모또 요시마사 변형 인터류킨-2 및 그의 제조방법
US5112946A (en) 1989-07-06 1992-05-12 Repligen Corporation Modified pf4 compositions and methods of use
WO1991006570A1 (en) 1989-10-25 1991-05-16 The University Of Melbourne HYBRID Fc RECEPTOR MOLECULES
US5349053A (en) 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
JP3242916B2 (ja) 1990-12-14 2001-12-25 セル ジェネシス,インコーポレイティド レセプター関連シグナル変換系のためのキメラ鎖
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
US5447851B1 (en) 1992-04-02 1999-07-06 Univ Texas System Board Of Dna encoding a chimeric polypeptide comprising the extracellular domain of tnf receptor fused to igg vectors and host cells
CN1117155C (zh) 1994-07-29 2003-08-06 史密丝克莱恩比彻姆有限公司 新型化合物
JP4435304B2 (ja) 1995-06-29 2010-03-17 イミュネックス・コーポレーション アポトーシスを誘導するサイトカイン
US6284236B1 (en) 1995-06-29 2001-09-04 Immunex Corporation Cytokine that induces apoptosis
US6046048A (en) 1996-01-09 2000-04-04 Genetech, Inc. Apo-2 ligand
US6713061B1 (en) 1996-03-12 2004-03-30 Human Genome Sciences, Inc. Death domain containing receptors
WO1997033904A1 (en) 1996-03-12 1997-09-18 Human Genome Sciences, Inc. Death domain containing receptors
US7357927B2 (en) 1996-03-12 2008-04-15 Human Genome Sciences, Inc. Death domain containing receptors
CA2249206A1 (en) 1996-04-01 1997-10-09 Genentech, Inc. Apo-2li and apo-3 apoptosis polypeptides
PT1012274E (pt) 1997-01-28 2007-08-14 Craig A Rosen Receptor 4 contendo um domínio de morte (dm4; receptor de morte 4), membro da super-família do ftn e ligação a liart (ap02)
US6433147B1 (en) 1997-01-28 2002-08-13 Human Genome Sciences, Inc. Death domain containing receptor-4
US7528239B1 (en) 1997-02-13 2009-05-05 Immunex Corporation Receptor that binds trail
US6072047A (en) 1997-02-13 2000-06-06 Immunex Corporation Receptor that binds trail
US20020160446A1 (en) 2000-11-14 2002-10-31 Holtzman Douglas A. Novel genes encoding proteins having prognostic diagnostic preventive therapeutic and other uses
US20010010924A1 (en) 1997-03-14 2001-08-02 Keith Charles Deen Tumor necrosis factor related receptor, tr6 polynecleotides
US6313269B1 (en) 1997-03-14 2001-11-06 Smithkline Beecham Corporation Tumor necrosis factor related receptor, TR6
US7803615B1 (en) 1997-03-17 2010-09-28 Human Genome Sciences, Inc. Death domain containing receptor 5
US6872568B1 (en) 1997-03-17 2005-03-29 Human Genome Sciences, Inc. Death domain containing receptor 5 antibodies
EP1007562A4 (en) 1997-04-16 2000-09-27 Millennium Pharm Inc TANGO-63d AND TANGO-63e PROTEINS RELATED TO THE TUMOR NECROSIS FACTOR RECEPTOR
US6342369B1 (en) 1997-05-15 2002-01-29 Genentech, Inc. Apo-2-receptor
EP1860187B1 (en) 1997-05-15 2011-07-13 Genentech, Inc. Apo-2 receptor
AU7705198A (en) 1997-05-30 1998-12-30 Human Genome Sciences, Inc. Human tumor necrosis factor receptor tr10
US6506569B1 (en) 1997-05-30 2003-01-14 Human Genome Sciences, Inc. Antibodies to human tumor necrosis factor receptor TR10
AU740227B2 (en) 1997-06-18 2001-11-01 Genentech Inc. Apo-2DcR
AU8400398A (en) 1997-07-11 1999-02-08 Trustees Of The University Of Pennsylvania, The Nucleic acid encoding a novel chemotherapy-induced protein, and methods of use
CA2301202A1 (en) 1997-08-15 1999-02-25 Idun Pharmaceuticals, Inc. Trail receptors, nucleic acids encoding the same, and methods of use thereof
WO1999011791A2 (en) 1997-09-05 1999-03-11 University Of Washington Tumor necrosis factor family receptors and ligands, encoding nucleic acids and related binding agents
ES2316182T3 (es) 1998-01-15 2009-04-01 Genentech, Inc. Ligando apo-2.
JP2002543151A (ja) 1999-05-04 2002-12-17 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド 死ドメイン含有レセプター5
EP1178828A4 (en) 1999-05-06 2004-10-13 Human Genome Sciences Inc RECEPTOR CONTAINING "DEATH DOMAIN"
CA2372246A1 (en) 1999-05-28 2000-12-07 Craig A. Rosen Human tumor necrosis factor receptor tr10
CN100594935C (zh) 2001-05-25 2010-03-24 人体基因组科学有限公司 免疫特异性结合trail受体的抗体
JP2005518782A (ja) 2001-09-17 2005-06-30 プロテイン デザイン ラブス, インコーポレイテッド ガンの診断方法、ガンのモジュレータのスクリーニング組成物及び方法
CA2471140A1 (en) 2001-12-20 2003-07-03 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to trail receptors
CA2494372C (en) 2002-08-15 2013-05-28 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to trail receptors
WO2005016326A2 (en) 2003-07-11 2005-02-24 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Analogs of thalidomide as potential angiogenesis inhibitors
CA3151350A1 (en) 2005-05-09 2006-11-16 E. R. Squibb & Sons, L.L.C. 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
RS54271B1 (en) 2005-07-01 2016-02-29 E. R. Squibb & Sons, L.L.C. HUMAN MONOCLONIC ANTIBODIES FOR LIGAND PROGRAMMED DEATH 1 (PD-L1)
CN101932720A (zh) 2007-10-31 2010-12-29 米迪缪尼有限公司 蛋白质支架
AU2009303304A1 (en) 2008-10-10 2010-04-15 Anaphore, Inc. Polypeptides that bind TRAIL-RI and TRAIL-R2
KR101508043B1 (ko) 2009-09-29 2015-04-06 로슈 글리카트 아게 이중특이적 사멸 수용체 아고니스트성 항체
US9120855B2 (en) 2010-02-10 2015-09-01 Novartis Ag Biologic compounds directed against death receptor 5
KR20130056870A (ko) 2010-04-13 2013-05-30 메디뮨 엘엘씨 피브로넥틴 ⅲ형 도메인에 기초한 다량체 스캐폴드
AU2011252841B2 (en) 2010-05-14 2014-04-03 Amgen Inc. Enhanced death receptor agonists
US9376437B2 (en) 2013-03-13 2016-06-28 Oncoceutics, Inc 7-benzyl-4-(2-methylbenzyl)-2,4,6,7,8,9-hexahydroimidazo[1,2-a]pyrido[3,4-e]pyrimidin-5(1H)-one, salts thereof and methods of using the same in combination therapy
RS59013B1 (sr) 2013-03-13 2019-08-30 Oncoceutics Inc 7-benzil-10-(2-metilbenzil)-2,6,7,8,9,10-heksahidroimidazo[1,2-a]pirido[4,3-d]pirimidin-5(3h)-on za upotrebu u lečenju kancera
EP3305812B1 (en) 2013-03-14 2020-06-17 Bristol-Myers Squibb Company Combination of dr5 agonist and anti-pd-1 antagonist and methods of use
US11229713B2 (en) * 2014-11-25 2022-01-25 Bristol-Myers Squibb Company Methods and compositions for 18F-radiolabeling of biologics
WO2018169993A1 (en) * 2017-03-14 2018-09-20 Five Prime Therapeutics, Inc. Antibodies binding to vista at acidic ph
CN113056483A (zh) * 2018-07-09 2021-06-29 戊瑞治疗有限公司 结合到ilt4的抗体

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Beckman et al. (Can. 109:170-179 (2007)) *
Cespdes et al. (Clin. Transl. Oncol. 8(5):318-329 (2006)) *
Dennis (Nature 442:739-741 (2006)) *
Fujimori et al. (J. Nuc. Med. 31:1191-1198 (1990)) *
Rudnick et al. (Can. Biotherp. & Radiopharm. 24: 155-162 (2009)) *
Talmadge et al. (Am. J. Pathol 170(3):793-804 (2007)) *
Thurber et al. (Adv. Drug Deliv. Rev. 60:1421-1434 (2008)) *
Voskoglou-Nomikos (Clin. Can. Res. 9:4227-4239 (2003)) *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11103579B2 (en) 2013-03-14 2021-08-31 Bristol-Myers Squibb Company Combination of DR5 agonist and anti-PD-1 antagonist and methods of use
US10441654B2 (en) 2014-01-24 2019-10-15 Children's Hospital Of Eastern Ontario Research Institute Inc. SMC combination therapy for the treatment of cancer
WO2017173360A3 (en) * 2016-03-31 2017-11-09 Stcube, Inc. Combination treatments directed toward programmed death ligand-1 (pd-l1) positive cancers
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10385130B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US11535670B2 (en) 2016-05-11 2022-12-27 Huyabio International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
WO2020150623A1 (en) * 2019-01-18 2020-07-23 University Of Georgia Research Foundation, Inc. Salt nanoparticles and compositions and methods of use thereof
WO2022081869A1 (en) * 2020-10-14 2022-04-21 University Of Virginia Patent Foundation Compositions and methods for overcoming dr5-induced immune evasion by solid tumors

Also Published As

Publication number Publication date
HRP20171758T1 (hr) 2017-12-29
EP3305812A1 (en) 2018-04-11
EP2970473A1 (en) 2016-01-20
US20180360962A1 (en) 2018-12-20
EP2970473B1 (en) 2017-08-16
CY1119510T1 (el) 2018-03-07
PT2970473T (pt) 2017-10-26
SI2970473T1 (sl) 2017-10-30
EP3305812B1 (en) 2020-06-17
ME02917B (me) 2018-10-20
RS56531B1 (sr) 2018-02-28
WO2014159562A1 (en) 2014-10-02
HUE035865T2 (en) 2018-05-28
US11103579B2 (en) 2021-08-31
ES2812208T3 (es) 2021-03-16
US20220098302A1 (en) 2022-03-31
PL2970473T3 (pl) 2018-01-31
ES2644022T3 (es) 2017-11-27
LT2970473T (lt) 2017-10-25
DK2970473T3 (da) 2017-11-27
NO3080374T3 (el) 2018-03-24

Similar Documents

Publication Publication Date Title
US20220098302A1 (en) Combination of dr5 agonist and anti-pd-1 antagonist and methods of use
US11466092B2 (en) Antibodies against OX-40 and uses thereof
ES2926969T3 (es) Anticuerpos contra el receptor de factor de necrosis de tumor inducido por glucocorticoides (gitr) y usos de los mismos
CA2951234C (en) Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
JP2020023511A (ja) 抗prlr抗体及びその使用
CN108473578A (zh) 治疗癌症的抗pd-1抗体和双特异性抗cd20/抗cd3抗体组合
US20220275091A1 (en) Combination of tim-4 antagonist and pd-1 antagonist and methods of use
JP7274426B2 (ja) 抗gitrアゴニスト抗体での癌の処置
BR112020018539A2 (pt) Anticorpos contra mica e/ou micb e usos dos mesmos
CN111542544A (zh) 用于治疗癌症的免疫刺激性激动性抗体
CA3123873A1 (en) Combination radioimmunotherapy and immune checkpoint therapy in the treatment of cancer
US11952427B2 (en) Anti-CD40 antibodies and uses thereof
US20240158510A1 (en) Antibodies against integrin heterodimers and uses thereof
US20230140384A1 (en) Antibodies to cd40 with enhanced agonist activity
CA3206413A1 (en) Antibodies against cd112r and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: BRISTOL-MYERS SQUIBB COMPANY, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BARNHART, BRYAN;JURE-KUNKEL, MARIA N.;SIGNING DATES FROM 20140417 TO 20140430;REEL/FRAME:032803/0267

AS Assignment

Owner name: BRISTOL-MYERS SQUIBB COMPANY, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BARNHART, BRYAN;JURE-KUNKEL, MARIA N.;SIGNING DATES FROM 20140417 TO 20140430;REEL/FRAME:036541/0947

STCB Information on status: application discontinuation

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