(19)
(11)EP 3 305 812 B1

(12)EUROPEAN PATENT SPECIFICATION

(45)Mention of the grant of the patent:
17.06.2020 Bulletin 2020/25

(21)Application number: 17186093.5

(22)Date of filing:  12.03.2014
(51)International Patent Classification (IPC): 
C07K 16/28(2006.01)

(54)

COMBINATION OF DR5 AGONIST AND ANTI-PD-1 ANTAGONIST AND METHODS OF USE

KOMBINATION VON DR5-AGONISTEN UND ANTI-PD1-ANTAGONISTEN UND VERFAHREN ZUR VERWENDUNG

COMBINAISON D'AGONISTE DR5 ET D'ANTAGONISTE ANTI-PD-1 ET PROCÉDÉS D'UTILISATION


(84)Designated Contracting States:
AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

(30)Priority: 14.03.2013 US 201361783184 P

(43)Date of publication of application:
11.04.2018 Bulletin 2018/15

(62)Application number of the earlier application in accordance with Art. 76 EPC:
14717294.4 / 2970473

(73)Proprietor: Bristol-Myers Squibb Company
Princeton, NJ 08543 (US)

(72)Inventors:
  • BARNHART, Bryan
    Princeton, NJ 08543 (US)
  • JURE-KUNKEL, Maria N.
    Princeton, NJ 08543 (US)

(74)Representative: Mewburn Ellis LLP 
Aurora Building Counterslip
Bristol BS1 6BX
Bristol BS1 6BX (GB)


(56)References cited: : 
  
  • K. TAKEDA ET AL: "Combination Therapy of Established Tumors by Antibodies Targeting Immune Activating and Suppressing Molecules", THE JOURNAL OF IMMUNOLOGY, vol. 184, no. 10, 16 April 2010 (2010-04-16), pages 5493-5501, XP055123617, ISSN: 0022-1767, DOI: 10.4049/jimmunol.0903033
  • TOMOYASU UNO ET AL: "Eradication of established tumors in mice by a combination antibody-based therapy", NATURE MEDICINE, vol. 12, no. 6, 7 May 2006 (2006-05-07), pages 693-698, XP055123624, ISSN: 1078-8956, DOI: 10.1038/nm1405
  • STEPHEN J DEMAREST ET AL: "Emerging antibody combinations in oncology", MABS, LANDES BIOSCIENCE, US, vol. 3, no. 4, 1 July 2011 (2011-07-01), pages 338-351, XP002690158, ISSN: 1942-0870, DOI: 10.4161/MABS.3.4.16615
  • MATTHEW VANNEMAN ET AL: "Combining immunotherapy and targeted therapies in cancer treatment", NATURE REVIEWS CANCER, vol. 12, no. 4, 1 January 2012 (2012-01-01), pages 237-251, XP055045378, ISSN: 1474-175X, DOI: 10.1038/nrc3237
  
Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


Description

CROSS-REFERENCE TO RELATED APPLICATIONS



[0001] This application claims priority to and the benefit of United States provisional patent application number 61/783, 184, filed March 14, 2013.

SEQUENCE LISTING



[0002] The instant application contains a Sequence Listing which has been submitted electronically in ASCII format. Said ASCII copy, created on March 3, 2014, is named MXI-529PC_SL.txt and is 24,778 bytes in size.

BACKGROUND



[0003] The National Cancer Institute has estimated that in the United States alone, 1 in 3 people will be struck with cancer during their lifetime. Moreover, approximately 50% to 60% of people contracting cancer will eventually succumb to the disease. The widespread occurrence of this disease underscores the need for improved anticancer regimens for the treatment of malignancy.

[0004] 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 ME, 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.

[0005] 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.

[0006] 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).

[0007] TRAIL (tumor necrosis factor (TNF)-related apoptosis-inducing ligand) is a member of the TNF superfamily with the ability to induce apoptosis of tumor cells. At least five receptors for TRAIL have been identified. DR4 (TRAIL-R1) and DR5 (TRAIL-R2) are 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 JP, et al., Science. 1997;277:818-821, and Walczak H, et al., EMBO J. 1997;16:5386-5397). Upon receptor activation, 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 FC, et al., Immunity. 2000;12:611-620, Thomas LR, et al., J Biol Chem. 2004;279:32780-32785, Thomas LR, et al., J Biol Chem. 2004;279:52479-52486, Varfolomeev E, et al., J Biol Chem. 2005;280:40599-40608, Ashkenazi A., Nat Rev Cancer. 2002;2:420-430, and Thorburn A. Cell Signal. 2004;16:139-144).

[0008] 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 TS, 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). Administration of TRAIL to mice bearing human tumors actively suppressed tumor progression and improved survival of the animal (Walczak H, et al, Nat Med. 1999;5:157-163). Accordingly, agonists against DR4 or DR5 by activating apoptosis are becoming dramatically meaningful as candidates for the treatment of cancer.

[0009] An anti-tumor combination therapy comprising a tumor cell death-inducing anti-DR5 monoclonal antibody (mAb) and immune activating anti-CD40 and anti-CD 137 mAbs is described in Uno et al. (Nature Medicine 12(6):693-698, 2006).

SUMMARY OF THE INVENTION



[0010] The present inventors have discovered for the first time that co-administration of a DR5 agonist (e.g., an antibody) and an anti-PD-1 antagonist (e.g., an antibody) effectively inhibits tumor growth in vivo, even synergistically. Accordingly, it is an object of the present invention to provide improved methods for treating subjects with cancer. Specifically, it is an object of the invention to provide efficacious combination treatment regimens wherein a DR5 agonist is combined with an anti-PD-1 antagonist for the treatment of cancer.

[0011] The present invention provides a PD-1 antagonist and/or a DR5 agonist for use in a method for the treatment of cancer in a subject, wherein the DR agonist is an anti-DR5 antibody and the PD-1 antagonist is an anti-PD-1 antibody comprising
  1. (i) 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; or
  2. (ii) heavy chain variable region CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 17, 18 and 19, respectively, and light chain variable region CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 20, 21 and 22, respectively; or
  3. (iii) heavy and light chain variable regions having the sequences set forth in SEQ ID NOs: 13 and 15, respectively; or
  4. (iv) heavy and light chains having the sequences set forth in SEQ ID NOs: 11 and 12, respectively,
wherein the anti-DR5 antibody and the anti-PD-1 antibody are for being administered in an amount that inhibits tumor growth in vivo. The DR5 agonist engages the DR5 receptor and is an agent that induces apoptosis in cancer cells.

[0012] The anti-DR5 agonists for use in the methods of treating cancer disclosed herein are anti-DR5 antibodies (e.g., monoclonal antibodies and bispecific antibodies). In one embodiment, 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. In another embodiment, the DR5 agonist is, e.g., TAS266. Further, DR5-agonistic ligands (e.g., a TNF-related apoptosis-inducing ligand (TRAIL), such as a recombinant human TRAIL, e.g., Dulanermin (also known as AMG951)) are described herein.

[0013] An exemplary anti-PD-1 antibody of the invention 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. In other embodiments, 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. In another embodiment, 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. In another embodiment, 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. In another embodiment, 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. In another embodiment, the antibody competes for binding with, and/or binds to the same epitope on PD-1 as, the above-mentioned antibodies. In another embodiment, 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).

[0014] Further described herein is a PD-1 antagonist that is an anti-PD-Ll 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 US Patent No. 7,943,743). The antibody may comprise the heavy and light chain CDRs or VRs of 12A4. The antibody may comprise 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 may comprise 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 may comprise 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 may comprise 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 may compete for binding with, and/or binds to the same epitope on PD-L1 as, the above-mentioned antibodies. The antibody may have 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).

[0015] In one embodiment, the invention provides a PD-1 antagonist and/or a DR5 agonist for use in a method of treating cancer in a subject, the method comprising administering to the subject an effective amount of the PD-1 antagonist and the DR5 agonist that inhibits tumor growth in vivo, wherein
  1. (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
  2. (b) the DR5 agonist is an antibody.


[0016] Also disclosed herein is a PD-1 antagonist and/or a DR5 agonist for use in a method of treating cancer in a subject, the method comprising administering to the subject an effective amount of the PD-1 antagonist and the DR5 agonist, wherein
  1. (a) the PD-1 antagonist is an anti-PD-Ll antibody; and
  2. (b) the DR5 agonist is an antibody.


[0017] The efficacy of the treatment methods provided herein can be assessed using any suitable means. In one embodiment, 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. In another embodiment, 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. In another embodiment, 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. In a further embodiment, 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. In certain embodiments, tumor volume is reduced by 50%, 60%, 70%, 80%, 90% or more, e.g., relative to tumor size before initiation of the treatment.

[0018] 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. For example, the antagonist and agonist can be simultaneously administered in a single formulation. Alternatively, the antagonist and agonist can be formulated for separate administration, wherein they are administered concurrently or sequentially. In one embodiment, the PD-1 antagonist is administered prior to administration of the DR5 agonist. In another embodiment, the DR5 agonist is administered prior to administration of the PD-1 antagonist. In a further embodiment, the DR5 agonist and the PD-1 antagonist are administered simultaneously.

[0019] In one embodiment, the cancer is a cancer selected from the group consisting of leukemia, lymphoma, blastoma, carcinoma and sarcoma. In another embodiment, 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 leukemia (CML).

[0020] Additional agents and therapies can be administered in combination with the agonists and antagonists described herein. In one embodiment, the methods comprise administration of an additional therapeutic agent (e.g., a cytotoxin or chemotherapeutic agent.

[0021] Also provided herein are pharmaceutical compositions comprising a PD-1 antagonist and a DR5 agonist and a pharmaceutically acceptable carrier. The PD-1 antagonist is an anti-PD-1 antibody (e.g., monoclonal antibody or bispecific antibody). The DR5 agonist is an anti-DR5 antibody (e.g., monoclonal antibody or bispecific antibody). In one embodiment, the PD-1 antagonist is an anti-PD-1 antibody comprising the heavy and light chain CDRs or VRs of 5C4.

[0022] The invention relates to kits for use in a method of treating a cancer in a subject, the kit comprising:
  1. (a) a dose of a PD-1 antagonist;
  2. (b) a dose of a DR5 agonist;
wherein the method comprises administering the PD-1 antagonist and DR5 agonist in a therapeutically effective amount in the methods disclosed herein. The DR5 agonist is an anti-DR5 antibody. The PD-1 antagonist is an anti-PD-1 antibody (e.g., 5C4). In particular embodiment, 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.

BRIEF DESCRIPTION OF THE DRAWINGS



[0023] 

Figure 1 is a graph depicting the median tumor volume in mice (mm3) 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.

Figure 2 depicts the tumor volume in individual mice administered a control (Figure 2A), an anti-PD-1 antibody on day 6 post-implant (Figure 2B), an anti-PD-1 antibody on day 8 post-implant (Figure 2C), an anti-PD-1 antibody on day 9 post-implant (Figure 2D), an anti-DR5 antibody on day 8 post-implant (Figure 2E), an anti-DR5 antibody on day 8 post-implant in combination with an anti-PD-1 antibody on day 6 post-implant (Figure 2F), an anti-DR5 antibody on day 8 post-implant in combination with an anti-PD-1 antibody on day 8 post-implant (Figure 2G), and an anti-DR5 antibody on day 8 post-implant in combination with an anti-PD-1 antibody on day 9 post-implant (Figure 2H).

Figure 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.


DETAILED DESCRIPTION OF THE INVENTION



[0024] As disclosed herein, 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 PD-1 antagonist and/or a DR5 agonist for use in a method for the treatment of cancer in a subject, which method comprises administering to a subject (e.g., human) an effective amount of the PD-1 antagonist and the DR5 agonist that inhibits tumor growth in vivo, wherein the PD-1 antagonist is an anti-PD-1 antibody, and the DR5 agonist is an anti-DR5 antibody.

I. Definitions



[0025] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by the skilled artisan. Although any methods and compositions similar or equivalent to those described herein can be used in practice or testing of the present invention, the preferred methods and compositions are described herein.

[0026] As used herein, the term "subject" or "patient" is a human patient (e.g., a patient having cancer).

[0027] A "solid tumor" includes, for example, sarcoma, melanoma, carcinoma, prostate carcinoma, lung carcinoma, colon carcinoma, or other solid tumor cancer.

[0028] The terms "cancer", "cancerous", or "malignant" refer 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. More particular examples of such 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).

[0029] As used herein, "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.

[0030] The term "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. In reference to solid tumors, 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. In some embodiments, an effective amount is an amount sufficient to delay tumor development. In some embodiments, 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. In one example, 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.

[0031] As used herein, the term "antagonist" refers to a molecule which blocks (e.g., reduces or prevents) a biological activity.

[0032] As used herein, the term "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.

[0033] As used herein, the term "ligand" refers to a molecule that forms a complex with a biomolecule (e.g., a receptor) to serve a biological purpose. In a narrower sense, is 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). Actual irreversible covalent binding between a ligand and its target molecule is rare in biological systems. Ligand binding to a receptor (receptor protein) alters its chemical conformation (three dimensional shape). The conformational state of a receptor protein determines its functional state.

[0034] As used herein, 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). In this context 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. In xenograft models, 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.

[0035] As used herein, the term "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 VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL 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). Each VH and VL 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. The term "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.

[0036] The term "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. Examples of binding fragments encompassed within the term "antigen-binding fragment" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 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. 1993); (iv) a Fd fragment consisting of the VH and CH1 domains; (v) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (vi) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; (vii) an isolated complementarity determining region (CDR); and (viii) a nanobody (also known as a single-domain antibody (sdAb)), which is a heavy chain variable region containing a single variable domain and two constant domains. Single domain antibodies include VHH 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).

[0037] "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. For example, 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. For example, 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.

[0038] 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. For example, 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.

[0039] Furthermore, although the two domains of the Fv fragment, VL and VH, 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 VL and VH 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). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.

[0040] As used herein, 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 KD of 10-7 M, 5 x 10-8 M, 10-8 M, 5 x 10-9 M, 10-9 M, 5 x 10-10 M, 10-10 M, 5 x 10-11 M, 10-11 M, 5 x 10-12 M, 10-12 M or less. For example, an antigen binding scaffold that specifically binds to DR5 is an antigen binding scaffold that binds to DR5 with a KD of 10-7 M, 5 x 10-8 M, 10-8 M, 5 x 10-9 M, 10-9 M, 5 x 10-10 M, 10-10 M, 5 x 10-11 M, 10-11 M, 5 x 10-12 M, 10-12 M or less. For example, 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 KD of 10-7 M, 5 x 10-8 M, 10-8 M, 5 x 10-9 M, 10-9 M, 5 x 10-10 M, 10-10 M, 5 x 10-11 M, 10-11 M, 5 x 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.

[0041] A "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).

[0042] The term "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. In one embodiment, 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.

[0043] The term "epitope" or "antigenic determinant" 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)).

[0044] The term "epitope mapping" refers to the process of identification of the molecular determinants for antibody-antigen recognition.

[0045] 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. Those of skill in the art understand that 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. For example, 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. In another example, a first antibody binds one or more segments of amino acids that significantly overlap the one or more segments bound by the second antibody. For the purposes herein, such antibodies are considered to "bind to the same epitope."

[0046] Accordingly, the antibodies for use according to the present invention also encompass antibodies that bind to an epitope which comprises all or a portion of an epitope recognized by the particular antibodies disclosed herein (e.g., the same or an overlapping region or a region between or spanning the region).

[0047] The antibodies for use according to the invention also encompass antibodies that bind the same epitope and/or antibodies that compete for binding with the antibodies disclosed herein. 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. Numerous types of competitive binding assays are known, for example: solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), 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 (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. 25(1):7 (1988)); solid phase direct biotin-avidin EIA (Cheung et al., Virology 176:546 (1990)); and direct labeled RIA. (Moldenhauer et al., Scand. J. Immunol. 32:77 (1990)). Typically, such an assay involves the use of purified antigen bound to a solid surface or cells bearing either of these, an unlabeled test immunoglobulin and a labeled reference immunoglobulin. Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test immunoglobulin. Usually the test immunoglobulin is present in excess. Usually, when a competing antibody is present in excess, it will inhibit specific binding of a reference antibody to a common antigen by at least 50-55%, 55-60%, 60-65%, 65-70% 70-75% or more.

[0048] Other techniques include, for example, 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. In addition, 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. For epitope mapping, computational algorithms have also been developed which have been shown to map conformational discontinuous epitopes.

[0049] Further, chimeric molecules (or fusion molecules) comprising an antigen binding domain, or equivalent, fused to another polypeptide or molecule are described herein. For example, 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. For example, Fc portions fused to the polypeptides described herein 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 described herein 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).

[0050] As used herein, the term "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., vincristine and vinblastine). Antibodies for the use according to the present invention can be conjugated to a radioisotope, e.g., radioactive iodine, to generate cytotoxic radiopharmaceuticals for treating cancer.

[0051] 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. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such 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-y; 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.

[0052] Techniques for conjugating such therapeutic moiety to antibodies are well known, see, e.g., Arnon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Hellstrom et al., "Antibodies For Drug Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review", in Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., "The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates", Immunol. Rev., 62:119-58 (1982).

[0053] As used herein, the term "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.

[0054] "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.

[0055] "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.

[0056] For purposes herein, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity 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.

II. PD-1 Antagonists



[0057] As used herein, 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).

[0058] As used herein, 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 complete human PD-L1 amino acid sequence - isoform a precursor - can be found under GenBank Accession No. NP_054862.1 (SEQ ID NO:24). The complete human PD-L1 amino acid sequence -isoform b precursor - can be found under GenBank Accession No. NP_001254635.1 (SEQ ID NO:25).

[0059] The protein Programmed Death 1 (PD-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. (1980) Immunogenics 10:247-260). PD-1 was discovered through screening for differential expression in apoptotic 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. In contrast, PD-1 is suggested to exist as a monomer, lacking the unpaired cysteine residue characteristic in other CD28 family members.

[0060] 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). Although structurally similar to CTLA-4, PD-1 lacks the MYPPPY motif (SEQ ID NO: 27) that is critical for B7-1 and B7-2 binding.

[0061] Consistent with PD-1 being an inhibitory member of the CD28 family, 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. (2003) J Exp Med 198:71-78; Prokunina and Alarcon-Riquelme (2004) Hum Mol Genet 13:R143; Nielsen et al. (2004) Lupus 13:510). In a murine B cell tumor line, the ITSM of PD-1 was shown to be essential to block BCR-mediated Ca2+-flux and tyrosine phosphorylation of downstream effector molecules (Okazaki et al. (2001) PNAS 98:13866-71).

[0062] 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 interaction between PD-1 and PD-L1 results in a decrease in tumor infiltrating lymphocytes, a decrease in T-cell receptor mediated proliferation, and immune evasion by the cancerous cells (Dong et al. (2003) J. Mol. Med. 81:281-7; Blank et al. (2005) Cancer Immunol. Immunother. 54:307-314; Konishi et al. (2004) Clin. Cancer Res. 10:5094-100). Immune suppression can be reversed by inhibiting the local interaction of PD-1 with PD-L1, and the effect is additive when the interaction of PD-1 with PD-L2 is blocked as well (Iwai et al. (2002) Proc. Nat'l. Acad. Sci. USA 99:12293-7; Brown et al. (2003) J. Immunol. 170:1257-66).

[0063] The methods of treating cancer disclosed herein involve the use of a PD-1 antagonist (an anti-PD-1 antibody in combination with a DR5 agonist that is an anti-DR5 antibody), for treating cancer. Accordingly, PD-1 antagonists used according to the invention bind directly to the PD-1 receptor, without engaging in signal transduction through the PD-1 receptor. In one embodiment, the PD-1 antagonist binds directly to PD-1 and blocks PD-1 inhibitory signal transduction. Further, PD-1 antagonists are disclosed herein which bind to one or more ligands of PD-1 (e.g., PD-L1 and PD-L2) and reduce or inhibit the ligand(s) from triggering inhibitory signal transduction through the PD-1, e.g. PD-1 antagonists which binds directly to PD-L1, inhibiting or preventing PD-L1 from binding to PD-1, thereby blocking PD-1 inhibitory signal transduction.

[0064] The PD-1 antagonists used according to the present invention or in the pharmaceutical compositions of the present invention are anti-PD-1 antibodies ("PD-1 antibodies") (e.g., 5C4). Further, PD-1 antagonists are described herein which are PD-1 binding scaffold proteins, PD-1 ligands, or multivalent agents, for example fusion proteins, such as AMP-224. Anti-human-PD-1 antibodies (or VH and/or VL domains derived therefrom) disclosed herein can be generated using methods well known in the art. Alternatively, art recognized anti-PD-1 antibodies may be used. For example, antibodies MK-3475 or CT-011 may be used. Additionally, monoclonal antibodies 17D8, 2D3, 4H1, 4A11, 7D3, and 5F4, described in WO 2006/121168, may be used. Antibodies that compete with any of these art-recognized antibodies for binding to PD-1 may also be used.

[0065] An exemplary anti-PD-1 antibody of the invention 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. In other embodiments, 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. In another embodiment, 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. In another embodiment, 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. In another embodiment, 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. In another embodiment, the antibody competes for binding with and/or binds to the same epitope on PD-1 as the above-mentioned antibodies. In another embodiment, 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).

[0066] In certain embodiments, 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 KD 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-γ 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.

[0067] Further, PD-1 antagonists are disclosed herein which are anti-PD-Ll antibodies. Anti-human-PD-Ll antibodies (or VH and/or VL domains derived therefrom) can be generated using methods well known in the art. Alternatively, art recognized anti-PD-Ll antibodies can be used. For example, MEDI4736 (also known as Anti-B7-H1) or MPDL3280A (also known as RG7446) can be used. Additionally, monoclonal antibodies 12A4, 3G10, 10A5, 5F8, 10H10, 1B12, 7H1, 11E6, 12B7, and 13G4, described in WO 2007/005874 and US Patent No. 7,943,743, can be used. Antibodies that compete with any of these art-recognized antibodies for binding to PD-L1 also can be used.

[0068] An exemplary anti-PD-Ll antibody is 12A4 (WO 2007/005874 and US Patent No. 7,943,743). The antibody may comprise the heavy and light chain CDRs or VRs of 12A4. The antibody may comprise 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 may comprise 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 may comprise 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 may comprise 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 may compete for binding with, and/or binds to the same epitope on PD-L1 as, the above-mentioned antibodies. The antibody may have 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).

[0069] Anti-PD-1 or anti-PD-Ll antibodies may bind to PD-1 or PD-L1, respectively, with a KD of 10-7 M, 5 x 10-8 M, 10-8 M, 5 x 10-9 M, 10-9 M, 5 x 10-10 M, 10-10 M or less.

III. DR5 Agonists



[0070] Provided herein is a DR5 agonist (an anti-DR5 antibody) and/or a PD-1 antagonist (an anti-PD-1 antibody) for use in a method for treating cancer, the method comprising administering to a subject in need thereof (e.g., a subject having cancer), a therapeutically effective amount of the DR5 agonist (which is an agent that induces apoptosis in a cancer cell) and the PD-1 antagonist that inhibits tumor growth in vivo. Further apoptosis inducing agents include DR proteins, such as DR4 and DR5.

[0071] As used herein, the terms "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. Biol., 7:693-696 (1997); Walczak et al., EMBO J., 16:5386-5387 (1997); Wu et al., Nature Genetics, 17:141-143 (1997); WO98/35986; EP870,827; WO98/46643; WO99/02653; WO99/09165; WO99/11791; US 2002/0072091; US 2002/0098550; U.S. Pat. No. 6,313,269; US 2001/0010924; US 2003/01255540; US 2002/0160446, US 2002/0048785; U.S. Pat. No. 6,342,369; U.S. Pat. No. 6,569,642, U.S. Pat. No. 6,072,047, U.S. Pat. No. 6,642,358; US 6,743,625) are used interchangeably, and include variants, isoforms, species homologs of human DR5, and analogs having at least one common epitope with DR5. The complete human DR5 sequence can be found under GenBank Accession No. AAC01565.1 (SEQ ID NO: 26).

[0072] 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) is a physiological mechanism involved in the deletion of peripheral T lymphocytes of the immune system, and its dysregulation can lead to a number of different pathogenic processes. 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.

[0073] 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 (Wiley S R, Schooley K, Smolak P, et al., Immunity, 1995, 3:673-682) 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.

[0074] Five 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. Like DR4, 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).

[0075] The term "agonist" as used with reference to DR5 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.

[0076] 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 agonists also include DR5 ligands, e.g., TRAIL and molecules that are derived from or based on TRAIL.

[0077] A DR5 agonist may be monovalent or multivalent. In certain embodiments, 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. For example, a DR5 agonist may be 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. For example, 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.

[0078] In certain embodiments, 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.

[0079] One example of a DR5 agonist is a recombinant human TRAIL (TNF-related apoptosis-inducing ligand), e.g., Dulanermin (also known as AMG-951; available from Amgen/Genentech).

[0080] According to the invention, the DR5 agonist is an anti-DR5 antibody, e.g., an antibody that binds to human DR5 with a KD of 10-7 M, 5 x 10-8 M, 10-8 M, 5 x 10-9 M, 10-9 M, 5 x 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 results in inhibition of tumor growth or reduction in tumor size, e.g., in a subject having cancer. 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). In one embodiment, the DR5 agonist is a bispecific death receptor agonist antibody, see, e.g., WO2011/039126; available from Roche Glycart). In another embodiment, 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). Further, a DR5 agonist is described herein that is a high affinity Fc-polypeptide (see, e.g., WO2011/143614; available from Amgen).

[0081] DR5 agonists may bemultivalent agents, 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).

[0082] 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. Antibodies having VH and VL chains comprising an amino acid sequence that is at least 90%, 95%, 98% or 99% identical to those of any of the anti-DR5 antibodies listed herein may be used. In certain methods described herein, a DR5 agonist can be replaced with a DR4 agonist. Thus, a subject having cancer can be treated with a combination of a DR4 agonist and a PD-1 antagonist. Generally, any agent that induces apoptosis in tumor cells can be combined with a PD-1 antagonist for treating cancer. In certain cases, 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.

IV. Compositions



[0083] In one aspect, the present invention provides a pharmaceutical composition comprising a PD-1 antagonist that is an anti-PD-1 antibody and a DR5 agonist that is an anti-DR5 antibody (e.g., formulated together in a single pharmaceutical composition or separately formulated) and a pharmaceutically acceptable carrier. In one embodiment, the pharmaceutical composition comprises a PD-1 antagonist and a DR5 agonist and a pharmaceutically acceptable carrier, 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. Further, pharmaceutical compositions are disclosed herein comprising a PD-1 antagonist and a DR5 agonist and a pharmaceutically acceptable carrier, wherein (a) the PD-1 antagonist is an anti-PD-Ll antibody and (b) the DR5 agonist is an antibody.

[0084] Pharmaceutical 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.

[0085] In general, such compositions typically comprise a pharmaceutically acceptable carrier. As used herein, the term "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. The term "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. Water or aqueous solution saline and aqueous dextrose and glycerol solutions may be employed as carriers, particularly for injectable solutions. 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.

[0086] For oral use, 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. In the case of tablets for oral use, carriers which are commonly used include lactose, corn starch, magnesium carbonate, talc, and sugar, and lubricating agents such as magnesium stearate are commonly added. For oral administration in capsule form, useful carriers include lactose, corn starch, magnesium carbonate, talc, and sugar. When aqueous suspensions are used for oral administration, emulsifying and/or suspending agents are commonly added.

[0087] In addition, sweetening and/or flavoring agents may be added to the oral compositions. For intramuscular, intraperitoneal, subcutaneous and intravenous use, sterile solutions of the active ingredient(s) are usually employed, and the pH of the solutions should be suitably adjusted and buffered. For intravenous use, the total concentration of the solute(s) should be controlled in order to render the preparation isotonic.

[0088] For preparing suppositories according to the invention, 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.

[0089] 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.

[0090] 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.

[0091] Also included are solid preparations which are intended for conversion, shortly before use, to liquid preparations for either oral or parenteral administration. Such liquid forms include solutions, suspensions and emulsions.

V. Patient Populations



[0092] Provided herein are effective methods for treating cancer in a patient, e.g., using a combination of a DR5 agonist and PD-1 antagonist. In one embodiment, the patient suffers from a cancer selected from the group consisting of leukemia, lymphoma, blastoma, carcinoma and sarcoma. In another embodiment, 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).

VI. Additional Agents/Therapies



[0093] The combinations of the present invention (e.g., PD-1 antagonist in combination with DR5 agonist) 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.

[0094] For example, the PD-1 antagonists and DR5 agonists disclosed 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 6X 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 differentiation protein-1) antagonists), iAP (inhibitor of apoptosis protein) antagonists (e.g., smac7, smac4, small molecule smac mimetic, synthetic smac peptides (see Fulda et al., Nat Med 2002;8:808-15), ISIS23722 (LY2181308), or AEG-35156 (GEM-640)), HDAC (histone deacetylase) inhibitors, anti-CD20 antibodies (e.g., rituximab), angiogenesis inhibitors (e.g., bevacizumab), anti-angiogenic agents targeting VEGF and VEGFR, synthetic triterpenoids (see Hyer et al., Cancer Research 2005;65:4799-808), c-FLIP (cellular FLICE-inhibitory protein) modulators (e.g., natural and synthetic ligands of PPARγ (peroxisome proliferator-activated receptor γ), 5809354 or 5569100), kinase inhibitors (e.g., Sorafenib), and/or genotoxic drugs.

[0095] The PD-1 antagonists and DR5 agonists disclosed 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 (CYTOXAN™) fosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylenemelamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, Dacarbazine, and Temozolomide.

[0096] 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.

[0097] Suitable anti-proliferative agents for use in the methods described herein, include, without limitation, taxanes, paclitaxel (paclitaxel is commercially available as TAXOL.RTM.), docetaxel, discodermolide (DDM), dictyostatin (DCT), Peloruside A, epothilones, epothilone A, epothilone B, epothilone C, epothilone D, epothilone E, epothilone F, furanoepothilone D, desoxyepothilone Bl, [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-desmethylepothilone B, epothilone B10, discoderomolide, patupilone (EPO-906), KOS-862, KOS-1584, ZK-EPO, ABJ-789, XAA296A (Discodermolide), TZT-1027 (soblidotin), ILX-651 (tasidotin hydrochloride), Halichondrin B, Eribulin mesylate (E-7389), Hemiasterlin (HTI-286), E-7974, Cyrptophycins, LY-355703, Maytansinoid immunoconjugates (DM-1), MKC-1, ABT-751, T1-38067, T-900607, SB-715992 (ispinesib), SB-743921, MK-0731, STA-5312, eleutherobin, 17beta-acetoxy-2-ethoxy-6-oxo-B-homo-estra-1,3,5(10)-trien-3-ol, cyclostreptin, isolaulimalide, laulimalide, 4-epi-7-dehydroxy-14,16-didemethyl-(+)-discodermolides, and cryptothilone 1, in addition to other microtubuline stabilizing agents known in the art.

[0098] In cases where it is desirable to render aberrantly proliferative cells quiescent in conjunction with or prior to treatment with the chemotherapeutic methods described herein, hormones and steroids (including synthetic analogs), such as 17a-Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone, Dromostanolone propionate, Testolactone, Megestrolacetate, Methylprednisolone, Methyl-testosterone, Prednisolone, Triamcinolone, Chlorotrianisene, Hydroxyprogesterone, Aminoglutethimide, Estramustine, Medroxyprogesteroneacetate, Leuprolide, Flutamide, Toremifene, ZOLADEX™, can also be administered to the patient. When employing the medical uses or pharmaceutical compositions of the present invention, other agents used in the modulation of tumor growth or metastasis in a clinical setting, such as antimimetics, can also be administered as desired.

[0099] Methods for the safe and effective administration of 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).

[0100] 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.

VII. Treatment Protocols



[0101] 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).

[0102] As used herein, adjunctive or combined administration (co-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). Thus, the PD-1 antagonist (e.g., antibody) and DR5 agonist (e.g., antibody) can be simultaneously administered in a single formulation. Alternatively, the PD-1 antagonist and DR5 agonist can be formulated for separate administration and are administered concurrently or sequentially.

[0103] For example, the PD-1 antagonist can be administered first followed by (e.g., immediately followed by) the administration of the DR5 agonist, or vice versa. In one embodiment, the PD-1 antagonist is administered prior to administration of the DR5 agonist. In one embodiment, the DR5 agonist is administered prior to administration of the PD-1 antagonist. Such concurrent or sequential administration preferably results in both the agonist and antagonist being simultaneously present in treated patients. In another embodiment, the DR5 agonist and the PD-1 antagonist are administered simultaneously.

[0104] In an exemplary treatment, 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-Ll antibody. In certain embodiment, 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. In certain embodiments, in which a PD-1 antagonist is provided as 1 dose every 1, 2, 3, 4, 5 or more days, 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.

[0105] In certain embodiments, 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.

[0106] 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)

[0107] In one embodiment, the dose of the PD-1 antagonist and/or DR5 agonist is varied over time. For example, the PD-1 antagonist and/or DR5 agonist may be initially administered at a high dose and may be lowered over time. In another embodiment, the the PD-1 antagonist and/or DR5 agonist is initially administered at a low dose and increased over time.

[0108] In another embodiment, 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. Generally, 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. For convenience, 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.

[0109] In one embodiment, the DR5 agonist (e.g., antibody) is administered at a dose of 0.1, 0.3, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 mg/kg body weight. In another embodiment, the PD-1 antagonist (e.g., antibody) is administered at a dose of 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 mg/kg body weight. Generally, 200 µg/mouse is approximately 10 mg/kg and 100 µg/mouse is approximately 5 mg/kg. Therefore, based on the experiments disclosed herein, 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. In certain embodiments, 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.

VIII. Outcomes



[0110] Patients, e.g., humans, treated according to the methods disclosed herein preferably experience improvement in at least one sign of cancer. In one embodiment, improvement is measured by a reduction in the quantity and/or size of measurable tumor lesions. In another embodiment, lesions can be measured on chest x-rays or CT or MRI films. In another embodiment, cytology or histology can be used to evaluate responsiveness to a therapy.

[0111] In one embodiment, 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. In another embodiment, the patient treated experiences tumor shrinkage and/or decrease in growth rate, i.e., suppression of tumor growth. In another embodiment, unwanted cell proliferation is reduced or inhibited. In yet another embodiment, one or more of the following can occur: the number of cancer cells can be reduced; tumor size can be reduced; cancer cell infiltration into peripheral organs can be inhibited, retarded, slowed, or stopped; tumor metastasis can be slowed or inhibited; tumor growth can be inhibited; recurrence of tumor can be prevented or delayed; one or more of the symptoms associated with cancer can be relieved to some extent.

[0112] In another embodiment, the methods of treatment produce a comparable clinical benefit rate (CBR = CR (complete response), PR (partial response) or SD (stable disease) ≥ 6 months) better than that achieved by a PD-1 (e.g., antibody) or DR5 agonist (e.g., antibody) alone. In other embodiments, 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.

[0113] In another embodiment, 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.

[0114] In a further embodiment, 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.

[0115] In certain embodiments, 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. In some embodiment, the tumor mass is no longer detectable following treatment as disclosed herein. In some embodiments, a subject is in partial or full remission.

IX. Kits and Unit Dosage Forms



[0116] Also provided herein are 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 is an anti-PD-1 antibody (e.g., 5C4). The DR5 agonist is an anti-DR5 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.

[0117] Optionally, the kits include multiple packages of the single-dose pharmaceutical compositions each containing an effective amount of the PD-1 antagonist and the DR5 agonist for a single administration in accordance with the methods provided above. Instruments or devices necessary for administering the pharmaceutical composition(s) also may be included in the kits. For instance, a kit may provide one or more pre-filled syringes containing an amount of the PD-1 antagonist and the DR5 agonist.

[0118] In one embodiment, the present invention provides a kit for use in a method of treating cancer in a patient, the kit comprising:
  1. (a) a dose of a PD-1 antagonist;
  2. (b) a dose of a DR5 agonist;
wherein the method comprises administering the PD-1 antagonist and DR5 agonist in a therapeutically effective amount in the methods disclosed herein. The DR5 agonist is an anti-DR5 antibody. The PD-1 antagonist is an anti-PD-1 antibody (e.g., 5C4). In particular embodiment, 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.

[0119] The following examples are merely illustrative.

EXAMPLES


Example 1


1. Materials and Methods


Animals



[0120] 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.

Antibodies



[0121] 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). Agonist anti-mouse DR5 mAb, clone MD5-1, hamster IgG isotype, was purchased from BioxCell (West Lebanon, NH). Both antibodies were certified to have <0.5 EU/mg endotoxin levels, >95% purity and <5% high molecular weight species. 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.

[0122] Anti-mPD-1 mAb was administered intraperitoneally at its optimal dose of 10 mg/kg; anti-DR5 mAb at 5mg/kg.

Tumor Model



[0123] 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 (2.0×106 MC-38 cells in 0.2mL Hanks Balanced Salt Solution).

[0124] Tumor size and body weights were measured twice weekly. Tumor size (measured as mm3) was calculated by multiplying the tumor length by the square of the tumor width divided by 2. Treatments were initiated when subcutaneous tumors reached a median size of 200 mm3 (established model). Antitumor activity, defined as percentage tumor growth inhibition, was calculated with the formula % Tumor Growth Inhibition (%TGI)= 100 - [(Tt/To)/(Ct/Co)] /100 - (Ct/Co), where Tt = median tumor size of treated group at the end of treatment, To = median tumor size of treated group at treatment initiation, Ct = median tumor size of control group at the end of treatment, and Co = median tumor size at treatment initiation (see Table 3). Complete regressions were defined as absence of measurable tumor mass for at least 2 tumor volume doubling times.

[0125] 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. 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.

[0126] The antitumor effect of single dose anti-mouse DR5 mAb in combination an anti-PD-1 mAb given at various dose schedules was evaluated in MC-38 (murine colon) tumor bearing mice. Six days post tumor implant, mice were sorted into eight groups of 7 mice with a mean tumor volume of ∼ 200mm3. Antibodies were administered according to the dosing schedules descried in Table 1.
Table 1: Dosing Schedule
Treatment Group# mice
1: Control 7
2: Control + anti-PD-1 mAb; 200 ug/mouse; Q4D×3 dosing initiated day 6 7
3: Control + anti-PD-1 mAb; 200 ug/mouse; Q4D×3 dosing initiated day 8 7
4: Control + anti-PD-1 mAb; 200 ug/mouse; Q4D×3 dosing initiated day 9 7
5: Anti-DR5 mAb; 100 ug/mouse; QD + Control dosing initiated day 8 7
6: Anti-DR5 mAb; 100 ug/mouse dosed day 8; QD + anti-PD-1 mAb; 200 ug/mouse; Q4D×3 dosed day 6 7
7: Anti-DR5 mAb; 100 ug/mouse dosed day 8; QD + anti-PD-1 mAb; 200 ug/mouse; Q4D×3 dosed day 8 7
8: Anti-DR5 mAb; 100 ug/mouse dosed day 8; QD + anti-PD-1 mAb; 200 ug/mouse; Q4D×3 dosed day 9 7
QD: One dose administered on only one day.
Q4D×3: One dose administered every four days for a total of 3 doses.


[0127] The combination of DR5 mAb and PD-1 mAb were tested according to the three schedules set forth in Table 2.
Table 2: Administration Schedules
Group No.:Administration Schedule
Group 6 DR5 mAb was administered 2 days after PD-1 mAb therapy.
Group 7 DR5 mAb and PD-1 mAb were administered on the same day.
Group 8 DR5 mAb was administered 1 day before PD-1 mAb therapy.


[0128] It was hypothesized that since PD-1 mab induces IFN-gamma, which in tumors upregulates DR5 expression on tumor cells, it might be advantageous to administer the DR5 mAb after administration of PD-1 mAb therapy (Group 6). Alternatively, it was hypothesized that DR5 mAb induces tumor cell death, which in turn will prime antitumor immune responses, and PD-1 mAb will subsequently expand the induced antitumor immunity. To test this hypothesis, DR5 mAb was administered before PD-1 mAb therapy (Group 8).

2. Results



[0129] As shown in Table 3, at least 80% tumor growth inhibition was achieved in mice treated with a combination of both the DR5 mAb and PD1 mAb.
Table 3: Tumor Responses
TreatmentDose (mg/kg)Schedule (days post implant)% TGI% Complete Regressions (#/total mice)
Anti-PD-1 mAb 10 Day 6, 10, 14 31 0 (0/7)
Anti-PD-1 mAb 10 Day 8, 12, 16 14 0 (0/7)
Anti-PD-1 mAb 10 Day 9, 13, 17 -8 0 (0/7)
Anti-DR5 mAb 5 Day 8 -4 0 (0/7)
Anti-PD-1 mAb + Anti-DR5 mAb 10 Day 6, 10, 14 96 43 (3/7)
5 Day 8
Anti-PD-1 mAb + Anti-DR5 mAb 10 Day 8, 12, 16 82 14 (1/7)
5 Day 8
Anti-PD-1 mAb + Anti-DR5 mAb 10 Day 9, 13, 17 87 43 (3/7)
5 Day 8
Moreover, as shown in Figure 1, the medium tumor volume (measured in mm3) 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 Figure 2.

[0130] In sum, 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 Figure 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.

[0131] Therefore, 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.
SUMMARY OF THE SEQUENCE LISTING
SEQ ID NO:SEQUENCE
1 Heavy Chain Variable Region (VH) Amino Acid Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent No. 7,943,743)
 

 
2 Heavy Chain Variable Region (VH) Nucleotide Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent No. 7,943,743)
 

 
3 Light Chain Variable Region (VL) Amino Acid Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent No. 7,943,743)
 

 
4 Light Chain Variable Region (VL) Nucleotide Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent No. 7,943,743)
 

 
5 Heavy Chain CDR1 Amino Acid Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent No. 7,943,743) TYAIS
6 Heavy Chain CDR2 Amino Acid Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent No. 7,943,743)
  GIIPIFGKAHYAQKFQ
7 Heavy Chain CDR3 Amino Acid Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent No. 7,943,743)
  KFHFVSGSPFGMDV
8 Light Chain CDR1 Amino Acid Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent No. 7,943,743)
  RASQSVSSYLA
9 Light Chain CDR2 Amino Acid Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent No. 7,943,743)
  DASNRAT
10 Light Chain CDR3 Amino Acid Sequence Anti-PD-L1 mAb (12A4; 12A4 in WO 2007/005874 and US Patent 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)
 

 
12 Light Chain Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (variable region underlined; constant region bold)
 

 
13 Heavy Chain Variable Region (VH) Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:4 from WO 2006/121168)
 

 
14 Heavy Chain Variable Region (VH) Nucleotide Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:60 from WO 2006/121168)
 

 
15 Light Chain Variable Region (VL) Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:11 from WO 2006/121168)
 

 
16 Light Chain Variable Region (VL) Nucleotide Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:67 from WO 2006/121168)
 

 
17 Heavy Chain CDR1 Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:18 from WO 2006/121168)
  NSGMH
18 Heavy Chain CDR2 Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:25 from WO 2006/121168)
  VIWYDGSKRYYADSVKG
19 Heavy Chain CDR3 Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:32 from WO 2006/121168)
  NDDY
20 Light Chain CDR1 Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:39 from WO 2006/121168)
  RASQSVSSYLA
21 Light Chain CDR2 Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:46 from WO 2006/121168)
  DASNRAT
22 Light Chain CDR3 Amino Acid Sequence Anti-PD-1 mAb (5C4 in WO 2006/121168) (SEQ ID NO:53 from WO 2006/121168)
  QQSSNWPRT
23 Complete PD-1 sequence (GenBank Accession No.: U64863)
 

 
 

 
24 Human PD-L1 amino acid sequence - isoform a precursor (GenBank Accession No. NP_054862.1)
 

 
25 Human PD-L1 amino acid sequence - isoform b precursor (GenBank Accession No. NP_001254635.1)
 

 
26 Human DR5 amino acid sequence (GenBank Accession No. AAC01565.1)
 

 
 

 



Claims

1. A Programmed Cell Death 1 (PD-1) antagonist and a Death Receptor (DR) agonist for use in a method of treating cancer, wherein the DR agonist is an anti-DR5 antibody and the PD-1 antagonist is an anti-PD-1 antibody comprising

(i) 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; or

(ii) heavy chain variable region CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 17, 18 and 19, respectively, and light chain variable region CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 20, 21 and 22, respectively; or

(iii) heavy and light chain variable regions having the sequences set forth in SEQ ID NOs: 13 and 15, respectively; or

(iv) heavy and light chains having the sequences set forth in SEQ ID NOs: 11 and 12, respectively,

wherein the anti-DR5 antibody and the anti-PD-1 antibody are for being administered in an amount that inhibits tumor growth in vivo.
 
2. A PD-1 antagonist for use in a method of treating cancer, wherein the method further comprises administration of a DR agonist, wherein the DR agonist is an anti-DR5 antibody and the PD-1 antagonist is an anti-PD-1 antibody comprising

(i) 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; or

(ii) heavy chain variable region CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 17, 18 and 19, respectively, and light chain variable region CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 20, 21 and 22, respectively; or

(iii) heavy and light chain variable regions having the sequences set forth in SEQ ID NOs: 13 and 15, respectively; or

(iv) heavy and light chains having the sequences set forth in SEQ ID NOs: 11 and 12, respectively,

wherein the anti-DR5 antibody and the anti-PD-1 antibody are for being administered in an amount that inhibits tumor growth in vivo.
 
3. A DR agonist for use in a method of treating cancer, wherein the method further comprises administration of a PD-1 antagonist, wherein the DR agonist is an anti-DR5 antibody and the PD-1 antagonist is an anti-PD-1 antibody comprising

(i) 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; or

(ii) heavy chain variable region CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 17, 18 and 19, respectively, and light chain variable region CDR1, CDR2 and CDR3 domains having the sequences set forth in SEQ ID NOs: 20, 21 and 22, respectively; or

(iii) heavy and light chain variable regions having the sequences set forth in SEQ ID NOs: 13 and 15, respectively; or

(iv) heavy and light chains having the sequences set forth in SEQ ID NOs: 11 and 12, respectively,

wherein the anti-DR5 antibody and the anti-PD-1 antibody are for being administered in an amount that inhibits tumor growth in vivo.
 
4. The PD-1 antagonist and/or the DR agonist for the use according to any one of claims 1-3, wherein the anti-DR5 antibody is selected from Lexatumumab, Tigatuzumab, Conatumumab, Drozitumab, HGSTR2J/KMTRS and LBY-135.
 
5. The PD-1 antagonist and/or the DR agonist for the use according to any one of claims 1-4, wherein the anti-PDl antibody is nivolumab.
 
6. The PD-1 antagonist and/or the DR agonist for the use according to any one of claims 1-5, wherein

(i) administration of the PD-1 antagonist and the DR agonist reduces tumor size by at least 50%, particularly by at least 80%; or

(ii) administration of the PD-1 antagonist and the DR agonist results in tumor growth inhibition of at least 80%; or

(iii) the treatment produces at least one therapeutic effect chosen from a reduction in size of a tumor, reduction in number of metastatic lesions over time, complete response, partial response, and stable disease.


 
7. The PD-1 antagonist and/or the DR agonist for the use according to any one of claims 1-6, wherein the PD-1 antagonist and the DR agonist are formulated for intravenous administration.
 
8. The PD-1 antagonist and/or the DR agonist for the use according to any one of claims 1-7, wherein

(i) the PD-1 antagonist is for being administered prior to administration of the DR agonist; or

(ii) the DR agonist is for being administered prior to administration of the PD-1 antagonist; or

(iii) the DR agonist and the PD-1 antagonist are for being administered simultaneously.


 
9. The PD-1 antagonist and/or the DR agonist for the use according to any one of claims 1-8, wherein the cancer is a cancer selected from leukemia, lymphoma, blastoma, carcinoma and sarcoma.
 
10. The PD-1 antagonist and/or the DR agonist for the use according to any one of claims 1-8, wherein the cancer is selected from 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).
 
11. The PD-1 antagonist and/or the DR agonist for the use according to any one of claims 1-10, wherein the method for treating cancer further comprises administration of an additional therapeutic agent, particularly an additional therapeutic agent selected from a cyotoxin and a chemotherapeutic agent.
 
12. The PD-1 antagonist and/or the DR agonist for the use according to any one of claims 1-11, wherein the subject to be treated is a human.
 
13. A pharmaceutical composition comprising a PD-1 antagonist and a DR agonist as defined in any one of claims 1-5 and a pharmaceutically acceptable carrier.
 
14. The pharmaceutical composition of claim 13 for use in a method of treating cancer as defined in any one of claims 1-12.
 
15. A kit for use in a method of treating a cancer in a subject, the kit comprising:

(a) a dose of a PD-1 antagonist;

(b) a dose of a DR agonist;

wherein the PD-1 antagonist and the DR agonist are as defined in any one of claims 1-5 and wherein the method comprises administering a therapeutically effective amount adapted for use in a method of treating cancer as defined in any one of claims 1-12.
 


Ansprüche

1. Programmierter Zelltod 1 (PD-1)-Antagonist und ein Death-Rezeptor (DR)-Agonist für die Verwendung in einem Verfahren zum Behandeln von Krebs, wobei der DR-Agonist ein Anti-DR5-Antikörper ist und der PD-1-Antagonist ein Anti-PD-1-Antikörper ist, umfassend

(i) die CDR1-, CDR2-und CDR3-Domänen in einer variablen Schwerketten-Region, welche die in SEQ ID NO: 13 festgelegte Sequenz haben, und die CDR1-, CDR2- und CDR3-Domänen in einer variablen Leichtketten-Region, welche die in SEQ ID NO: 15 festgelegte Sequenz haben; oder

(ii) variable Schwerketten-Region CDR1-, CDR2- und CDR3-Domänen, welche die in SEQ ID NO: 17, 18 bzw. 19 festgelegten Sequenzen haben, und variable Leichtketten-Region CDR1-, CDR2- und CDR3-Domänen, welche die in SEQ ID NO: 20, 21 bzw. 22 festgelegten Sequenzen haben; oder

(iii) variable Schwer- und Leichtketten-Regionen, welche die in SEQ ID NO: 13 bzw. 15 festgelegten Sequenzen haben; oder

(iv) Schwer- und Leichtketten, aufweisend die Sequenzen, welche die in SEQ ID NO: 11 bzw. 12 festgelegten Sequenzen haben,

wobei der Anti-DR5-Antikörper und der Anti-PD-1-Antikörper in einer Menge verabreicht werden, die Tumorwachstum in vivo hemmt.
 
2. PD-1-Antagonist zur Verwendung in einem Verfahren zum Behandeln von Krebs, wobei das Verfahren ferner die Verabreichung eines DR-Agonisten umfasst, wobei der DR-Agonist ein Anti-DR5-Antikörper ist und der PD-1-Antagonist ein Anti-PD-1-Antikörper ist, umfassend

(i) die CDR1-, CDR2-und CDR3-Domänen in einer variablen Schwerketten-Region, welche die in SEQ ID NO: 13 festgelegte Sequenz haben, und die CDR1-, CDR2- und CDR3-Domänen in einer variablen Leichtketten-Region, welche die in SEQ ID NO: 15 festgelegte Sequenz haben; oder

(ii) variable Schwerketten-Region CDR1-, CDR2- und CDR3-Domänen, welche die in SEQ ID NO: 17, 18 bzw. 19 festgelegten Sequenzen haben, und variable Leichtketten-Region CDR1-, CDR2- und CDR3-Domänen, welche die in SEQ ID NO: 20, 21 bzw. 22 festgelegten Sequenzen haben; oder

(iii) variable Schwer- und Leichtketten-Regionen, welche die in SEQ ID NO: 13 bzw. 15 festgelegten Sequenzen haben; oder

(iv) Schwer- und Leichtketten, aufweisend die Sequenzen, welche die in SEQ ID NO: 11 bzw. 12 festgelegten Sequenzen haben,

wobei der Anti-DR5-Antikörper und der Anti-PD-1-Antikörper in einer Menge verabreicht werden, die Tumorwachstum in vivo hemmt.
 
3. DR-Agonist für die Verwendung in einem Verfahren zum Behandeln von Krebs, wobei das Verfahren ferner die Verabreichung eines PD-1-Antagonisten umfasst, wobei der DR-Agonist ein Anti-DR5-Antikörper ist und der PD-1-Antagonist ein Anti-PD-1-Antikörper ist, umfassend

(i) die CDR1-, CDR2-und CDR3-Domänen in einer variablen Schwerketten-Region, welche die in SEQ ID NO: 13 festgelegte Sequenz haben, und die CDR1-, CDR2- und CDR3-Domänen in einer variablen Leichtketten-Region, welche die in SEQ ID NO: 15 festgelegte Sequenz haben; oder

(ii) variable Schwerketten-Region CDR1-, CDR2- und CDR3-Domänen, welche die in SEQ ID NO: 17, 18 bzw. 19 festgelegten Sequenzen haben, und variable Leichtketten-Region CDR1-, CDR2- und CDR3-Domänen, welche die in SEQ ID NO: 20, 21 bzw. 22 festgelegten Sequenzen haben; oder

(iii) variable Schwer- und Leichtketten-Regionen, welche die in SEQ ID NO: 13 bzw. 15 festgelegten Sequenzen haben; oder

(iv) Schwer- und Leichtketten, aufweisend die Sequenzen, welche die in SEQ ID NO: 11 bzw. 12 festgelegten Sequenzen haben,

wobei der Anti-DR5-Antikörper und der Anti-PD-1-Antikörper in einer Menge verabreicht werden, die Tumorwachstum in vivo hemmt.
 
4. PD-1-Antagonist und/oder der DR-Agonist für die Verwendung nach einem der Ansprüche 1 bis 3, wobei der Anti-DR5-Antikörper ausgewählt ist aus Lexatumab, Tigatuzumab, Conatumab, Drozitumab, HGSTR2J/KMTRS und LBY-135.
 
5. PD-1-Antagonist und/oder der DR-Agonist für die Verwendung nach einem der Ansprüche 1 bis 4, wobei der Anti-PD1-Antikörper Nivolumab ist.
 
6. PD-1-Antagonist und/oder der DR-Agonist für die Verwendung nach einem der Ansprüche 1 bis 5, wobei

(i) Verabreichung des PD-1-Antagonisten und des DR-Agonisten die Tumorgröße um mindestens 50%, speziell um mindestens 80% reduziert; oder

(ii) Verabreichung des PD-1-Antagonisten und des DR-Agonisten zu einer Hemmung von Tumorwachstum von mindestens 80% führt; oder

(iii) die Behandlung mindestens eine therapeutische Wirkung erzielt, ausgewählt aus einer Verringerung der Größe eines Tumors, Verringerung der Anzahl von metastatischen Läsionen im Laufe der Zeit, vollständigem Ansprechen, partiellem Ansprechen und stabiler Erkrankung.


 
7. PD-1-Antagonist und/oder der DR-Agonist für die Verwendung nach einem der Ansprüche 1 bis 6, wobei der PD-1-Antagonist und der DR-Agonist zur intravenösen Verabreichung formuliert sind.
 
8. PD-1-Antagonist und/oder der DR-Agonist für die Verwendung nach einem der Ansprüche 1 bis 7, wobei

(i) der PD-1-Antagonist vor der Verabreichung des DR-Agonisten verabreicht wird; oder

(ii) der DR-Agonist vor der Verabreichung des PD-1-Antagonisten verabreicht wird; oder

(iii) der DR-Agonist und der PD-1-Antagonist gleichzeitig verabreicht werden.


 
9. PD-1-Antagonist und/oder der DR-Agonist für die Verwendung nach einem der Ansprüche 1 bis 8, wobei der Krebs ein Krebs ist, der ausgewählt ist aus Leukämie, Lymphom, Blastom, Karzinom und Sarkom.
 
10. PD-1-Antagonist und/oder der DR-Agonist für die Verwendung nach einem der Ansprüche 1 bis 8, wobei der Krebs ausgewählt ist aus chronischer myeloischer Leukämie, akuter lymphoblastischer Leukämie, Philadelphia- Chromosom- positiver akuter lymphoblastischer Leukämie (Ph + ALL), Plattenepithelkarzinom, kleinzelligem Lungenkrebs, nicht-kleinzelligem Lungenkrebs, Gliom, Magen-Darm-Krebs, nierenbedingtem Krebs, Krebs der Eierstöcke, Leberkrebs, kolorektalem Karzinom, Endometriumkarzinom, Nierenkrebs, Prostatakrebs, Schilddrüsenkrebs, Neuroblastom, Pankreaskrebs, Glioblastoma multiforme, Gebärmutterhalskrebs, Magenkrebs, Blasenkrebs, Hepatom, Brustkrebs, Kolonkarzinom und Kopf-und Halskrebs, Magenkarzinom, Keimzellkrebs, pädiatrischem Sarkom, sinusnasalem natürlichen Killer, multiplem Myelom, akuter myelogener Leukämie (AML) und chronischer lymphozytischer Leukämie (CML).
 
11. PD-1-Antagonist und/oder den DR-Agonist für die Verwendung nach einem der Ansprüche 1 bis 10, wobei das Verfahren zum Behandeln von Krebs ferner Verabreichung eines zusätzlichen therapeutischen Mittels umfasst, speziell eines zusätzlichen therapeutischen Mittels, das ausgewählt ist aus einem Cytotoxin und einem Chemotherapeutikum.
 
12. PD-1-Antagonist und/oder der DR-Agonist für die Verwendung nach einem der Ansprüche 1 bis 11, wobei das zu behandelnde Subjekt ein Mensch ist.
 
13. Pharmazeutische Zusammensetzung, umfassend einen PD-1-Antagonisten und einen DR-Agonisten, nach einem der Ansprüche 1 bis 5 und einen pharmazeutisch verträglichen Träger.
 
14. Pharmazeutische Zusammensetzung nach Anspruch 13 für die Verwendung in einem Verfahren zum Behandeln von Krebs, nach einem der Ansprüche 1 bis 12.
 
15. Kit für die Verwendung in einem Verfahren zum Behandeln von Krebs in einem Subjekt, wobei das Kit umfasst:

(a) eine Dosis eines PD-1-Antagonisten;

(b) eine Dosis eines DR-Agonisten;

wobei der PD-1-Antagonist und der DR-Agonist wie in einem der Ansprüche 1 bis 5 festgelegt sind, und wobei das Verfahren das Verabreichen einer therapeutisch wirksamen Menge umfasst, die für die Verwendung in einem Verfahren zum Behandeln von Krebs ausgelegt ist, wie in einem der Ansprüche 1 bis 12 festgelegt ist.
 


Revendications

1. Antagoniste de mort cellulaire programmée 1 (PD-1) et agoniste de récepteur de mort (DR) pour une utilisation dans une méthode de traitement d'un cancer, où l'agoniste de DR est un anticorps anti-DR5 et l'antagoniste de PD-1 est un anticorps anti-PD-1 comprenant:

(i) les domaines CDR1, CDR2 et CDR3 dans une région variable de chaîne lourde possédant la séquence présentée dans la SEQ ID NO: 13 et les domaines CDR1, CDR2 et CDR3 dans une région variable de chaîne légère possédant la séquence présentée dans la SEQ ID NO: 15; ou

(ii) les domaines CDR1, CDR2 et CDR3 de région variable de chaîne lourde possédant les séquences présentées dans les SEQ ID NOs: 17, 18 et 19, respectivement, et les domaines CDR1, CDR2 et CDR3 de région variable de chaîne légère possédant les séquences présentées dans les SEQ ID NOs: 20, 21 et 22, respectivement; ou

(iii) les régions variables de chaînes lourde et légère possédant les séquences présentées dans les SEQ ID NOs: 13 et 15, respectivement; ou

(iv) les chaînes lourde et légère possédant les séquences présentées dans les SEQ ID NOs: 11 et 12, respectivement,

où l'anticorps anti-DR5 et l'anticorps anti-PD-1 sont destinés à être administrés dans une quantité qui inhibe une croissance tumorale in vivo.
 
2. Antagoniste de PD-1 pour une utilisation dans une méthode de traitement d'un cancer, où la méthode comprend en outre l'administration d'un agoniste de DR, où l'agoniste de DR est un anticorps anti-DR5 et l'antagoniste de PD-1 est un anticorps anti-PD-1 comprenant:

(i) les domaines CDR1, CDR2 et CDR3 dans une région variable de chaîne lourde possédant la séquence présentée dans la SEQ ID NO: 13 et les domaines CDR1, CDR2 et CDR3 dans une région variable de chaîne légère possédant la séquence présentée dans la SEQ ID NO: 15; ou

(ii) les domaines CDR1, CDR2 et CDR3 de région variable de chaîne lourde possédant les séquences présentées dans les SEQ ID NOs: 17, 18 et 19, respectivement, et les domaines CDR1, CDR2 et CDR3 de région variable de chaîne légère possédant les séquences présentées dans les SEQ ID NOs: 20, 21 et 22, respectivement; ou

(iii) les régions variables de chaînes lourde et légère possédant les séquences présentées dans les SEQ ID NOs: 13 et 15, respectivement; ou

(iv) les chaînes lourde et légère possédant les séquences présentées dans les SEQ ID NOs: 11 et 12, respectivement,

où l'anticorps anti-DR5 et l'anticorps anti-PD-1 sont destinés à être administrés dans une quantité qui inhibe une croissance tumorale in vivo.
 
3. Agoniste de DR pour une utilisation dans une méthode de traitement d'un cancer, où la méthode comprend en outre l'administration d'un antagoniste de PD-1, où l'agoniste de DR est un anticorps anti-DR5 et l'antagoniste de PD-1 est un anticorps anti-PD-1 comprenant:

(i) les domaines CDR1, CDR2 et CDR3 dans une région variable de chaîne lourde possédant la séquence présentée dans la SEQ ID NO: 13 et les domaines CDR1, CDR2 et CDR3 dans une région variable de chaîne légère possédant la séquence présentée dans la SEQ ID NO: 15; ou

(ii) les domaines CDR1, CDR2 et CDR3 de région variable de chaîne lourde possédant les séquences présentées dans les SEQ ID NOs: 17, 18 et 19, respectivement, et les domaines CDR1, CDR2 et CDR3 de région variable de chaîne légère possédant les séquences présentées dans les SEQ ID NOs: 20, 21 et 22, respectivement; ou

(iii) les régions variables de chaînes lourde et légère possédant les séquences présentées dans les SEQ ID NOs: 13 et 15, respectivement; ou

(iv) les chaînes lourde et légère possédant les séquences présentées dans les SEQ ID NOs: 11 et 12, respectivement,

où l'anticorps anti-DR5 et l'anticorps anti-PD-1 sont destinés à être administrés dans une quantité qui inhibe une croissance tumorale in vivo.
 
4. Antagoniste de PD-1 et/ou agoniste de DR pour l'utilisation selon l'une quelconque des revendications 1-3, où l'anticorps anti-DR5 est choisi parmi Lexatumumab, Tigatuzumab, Conatumumab, Drozitumab, HGSTR2J/KMTRS et LBY-135.
 
5. Antagoniste de PD-1 et/ou agoniste de DR pour l'utilisation selon l'une quelconque des revendications 1-4, où l'anticorps anti-PD1 est nivolumab.
 
6. Antagoniste de PD-1 et/ou agoniste de DR pour l'utilisation selon l'une quelconque des revendications 1-5, où:

(i) l'administration de l'antagoniste de PD-1 et de l'agoniste de DR réduit la taille d'une tumeur d'au moins 50%, en particulier d'au moins 80%; ou

(ii) l'administration de l'antagoniste de PD-1 et de l'agoniste de DR conduit à une inhibition de croissance tumorale d'au moins 80%; ou

(iii) le traitement produit au moins un effet thérapeutique choisi parmi une réduction de la taille d'une tumeur, une réduction du nombre de lésions métastasiques au cours du temps, une réponse complète, une réponse partielle et une maladie stable.


 
7. Antagoniste de PD-1 et/ou agoniste de DR pour l'utilisation selon l'une quelconque des revendications 1-6, où l'antagoniste de PD-1 et l'agoniste de DR sont formulés pour une administration intraveineuse.
 
8. Antagoniste de PD-1 et/ou agoniste de DR pour l'utilisation selon l'une quelconque des revendications 1-7, où:

(i) l'antagoniste de PD-1 est destiné à être administré avant l'administration de l'agoniste de DR; ou

(ii) l'agoniste de DR est destiné à être administré avant l'administration de l'antagoniste de PD-1; ou

(iii) l'agoniste de DR et l'antagoniste de PD-1 sont destinés à être administrés simultanément.


 
9. Antagoniste de PD-1 et/ou agoniste de DR pour l'utilisation selon l'une quelconque des revendications 1-8, où le cancer est un cancer choisi parmi une leucémie, un lymphome, un blastome, un carcinome et un sarcome.
 
10. Antagoniste de PD-1 et/ou agoniste de DR pour l'utilisation selon l'une quelconque des revendications 1-8, où le cancer est choisi parmi une leucémie myéloïde chronique, une leucémie lymphoblastique aiguë, une leucémie lymphoblastique aiguë positive au chromosome Philadelphie (Ph+ ALL), un carcinome des cellules squameuses, un cancer pulmonaire à petites cellules, un cancer pulmonaire non à petites cellules, un gliome, un cancer gastro-intestinal, un cancer rénal, un cancer ovarien, un cancer du foie, un cancer colorectal, un cancer de l'endomètre, un cancer du rein, un cancer de la prostate, un cancer de la thyroïde, un neuroblastome, un cancer pancréatique, un glioblastome multiforme, un cancer du col de l'utérus, un cancer de l'estomac, un cancer de la vessie, un hépatome, un cancer du sein, un carcinome du côlon et un cancer de la tête et du cou, un cancer gastrique, une tumeur de cellules germinales, un sarcome pédiatrique, des cellules tueuses naturelles naso-sinusales, un myélome multiple, une leucémie myélogène aiguë (AML) et une leucémie lymphocytaire chronique (CML).
 
11. Antagoniste de PD-1 et/ou agoniste de DR pour l'utilisation selon l'une quelconque des revendications 1-10, où la méthode pour le traitement d'un cancer comprend en outre l'administration d'un agent thérapeutique supplémentaire, en particulier un agent thérapeutique supplémentaire choisi parmi une cytotoxine et un agent chimiothérapeutique.
 
12. Antagoniste de PD-1 et/ou agoniste de DR pour l'utilisation selon l'une quelconque des revendications 1-11, où le sujet à traiter est un humain.
 
13. Composition pharmaceutique comprenant un antagoniste de PD-1 et un agoniste de DR tels que définis dans l'une quelconque des revendications 1-5 et un véhicule pharmaceutiquement acceptable.
 
14. Composition pharmaceutique selon la revendication 13 pour une utilisation dans une méthode de traitement d'un cancer telle que définie dans l'une quelconque des revendications 1-12.
 
15. Kit pour une utilisation dans une méthode de traitement d'un cancer chez un sujet, le kit comprenant:

(a) une dose d'un antagoniste de PD-1;

(b) une dose d'un agoniste de DR;

où l'antagoniste de PD-1 et l'agoniste de DR sont tels que définis dans l'une quelconque des revendications 1-5 et où la méthode comprend l'administration d'une quantité thérapeutiquement efficace adaptée pour une utilisation dans une méthode de traitement d'un cancer telle que définie dans l'une quelconque des revendications 1-12.
 




Drawing














Cited references

REFERENCES CITED IN THE DESCRIPTION



This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description




Non-patent literature cited in the description