WO2010027423A2 - Compositions of pd-1 antagonists and methods of use - Google Patents

Compositions of pd-1 antagonists and methods of use Download PDF

Info

Publication number
WO2010027423A2
WO2010027423A2 PCT/US2009/004825 US2009004825W WO2010027423A2 WO 2010027423 A2 WO2010027423 A2 WO 2010027423A2 US 2009004825 W US2009004825 W US 2009004825W WO 2010027423 A2 WO2010027423 A2 WO 2010027423A2
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
cancer
amino acids
compound
contiguous amino
Prior art date
Application number
PCT/US2009/004825
Other languages
French (fr)
Other versions
WO2010027423A3 (en
Inventor
Solomon Langerman
Original Assignee
Amplimmune, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=41725778&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2010027423(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to EP09811805.2A priority Critical patent/EP2350129B1/en
Priority to UAA201103619A priority patent/UA106050C2/en
Priority to NZ591130A priority patent/NZ591130A/en
Priority to EA201170373A priority patent/EA023148B1/en
Priority to MX2011002252A priority patent/MX2011002252A/en
Priority to CN2009801422563A priority patent/CN102203132A/en
Priority to SI200931266T priority patent/SI2350129T1/en
Priority to ES09811805.2T priority patent/ES2545609T3/en
Priority to CA2734908A priority patent/CA2734908A1/en
Priority to BRPI0917320A priority patent/BRPI0917320A2/en
Priority to PL09811805T priority patent/PL2350129T3/en
Priority to DK09811805.2T priority patent/DK2350129T3/en
Priority to AU2009288730A priority patent/AU2009288730B2/en
Priority to JP2011524986A priority patent/JP5798919B2/en
Priority to RS20150547A priority patent/RS54233B1/en
Application filed by Amplimmune, Inc. filed Critical Amplimmune, Inc.
Publication of WO2010027423A2 publication Critical patent/WO2010027423A2/en
Publication of WO2010027423A3 publication Critical patent/WO2010027423A3/en
Priority to ZA2011/01120A priority patent/ZA201101120B/en
Priority to IL211298A priority patent/IL211298A0/en
Priority to MA33714A priority patent/MA32646B1/en
Priority to HRP20150933TT priority patent/HRP20150933T1/en
Priority to SM201500212T priority patent/SMT201500212B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • compositions of PD-1 Antagonists Compositions of PD-1 Antagonists and Methods of Use
  • the present invention relates to therapeutic compositions containing a compound that prevents inhibitory signal transduction on T cells in combination with potentiating agents and the use of said components together or separately for the induction of T cell responses valuable in disease therapy.
  • T lymphocytes to disease states, such as infection and chronic diseases like cancer, is complicated and involves intercellular interactions and the production of soluble mediators (called cytokines or lymphokines).
  • cytokines soluble mediators
  • Activation of T cells normally depends on an antigen-specific signal following contact of the T cell receptor (TCR) with an antigenic peptide presented via the major histocompatibility complex (MHC) while the extent of this reaction is controlled by positive and negative antigen-independent signals eminating from a variety of co-stimulatory molecules.
  • TCR T cell receptor
  • MHC major histocompatibility complex
  • the latter are commonly members of the CD28/B7 family.
  • PD-1 is a member of the CD28 family of receptors that delivers a negative immune response when induced on T cells.
  • Contact between PD-1 and one of its ligands (B7-H1 or B7-DC) induces an inhibitory response that decreases T cell multiplication and/or the strength and/or duration of a T cell response.
  • the T lymphocyte response is regulated by various factors, including cell surface molecules that act as receptors, where the latter include both the TCR complex as well as other surface molecules.
  • an antigen specific T cell response is mediated by two signals: 1) engagement of the TCR with antigenic peptide presented in the context of HC (signal 1), and 2) a second antigen-independent signal delivered by contact between different receptor/ligand pairs (signal 2).
  • This "second signal” is critical in determining the type of T cell response (activation vs tolerance) as well as the strength and duration of that response, and is regulated by both positive and negative signals from costimulatory molecules, such as the B7 family of proteins.
  • T cell costimulatory pathway is B7- CD28, in which B7-1 (CD80) and B7-2 (CD86) each can engage the stimulatory CD28 receptor and the inhibitory CTLA-4 (CD152) receptor.
  • B7-1 CD80
  • B7-2 CD86
  • CD28 ligation increases antigen-specific proliferation of T cells, enhances production of cytokines, stimulates differentiation and effector function, and promotes survival of T cells (Lenshow, et al., Annu. Rev. Immunol., 14:233-258 (1996); Chambers and Allison, Curr. Opin. Immunol., 9:396-404 (1997); and Rathmell and Thompson, Annu. Rev. Immunol., 17:781-828 (1999)).
  • B7-H5 (described in WO 2006/012232) is a newly discovered member of the B7 family.
  • B7 family molecules have a membrane proximal IgC (constant) domain and a membrane distal IgV (variable) domain.
  • the CD28-like family of receptors for these ligands share a common extracellular IgV-like domain. Interactions of receptor-ligand pairs are mediated predominantly through residues in the IgV domains of the ligands and receptors (Schwartz, et al., Nature Immunol., 3:427-434 (2002)).
  • IgV domains are described as having two sheets that each contains a layer of ⁇ -strands (Williams and Barclay, Annu. Rev. Immunol., 6:381-405 (1988)).
  • the front and back sheets of CTLA-4 contain strands A'GFC'C and ABEDC, respectively (Ostrov, et al., Science, 290:816-819 (2000)), whereas the front and back sheets of the B7 IgV domains are composed of strands AGFCCC" and BED, respectively (Schwartz, et al., Nature, 410:604-608 (2001); Stamper, et al., Nature, 410:608-611 (2001); and Ikemizu, et al., Immunity, 12:51-60 (2000)).
  • B7-DC also called PD-L2 or CD273
  • B7 family and has an amino acid sequence that is about 34% identical to B7-
  • B7-H1 and B7-DC transcripts are found in various tissues (Dong, et al., Nature Med., 5:1365-1369 (1999); Latchman, et al., Nature Immunol., 2:261-268 (2001); and Tamura, Blood, 97:1809-1816
  • B7-H1 is broadly expressed on a wide variety of tissue and cell types, while B7-DC expression is predominantly restricted to activated dendritic cells (DC) and macrophages.
  • B7-H1 and B7-DC bind to PD-1 (Freeman, et al., J. Exp. Med., 192:1027-1034 (2000)), a distant member of the CD28 family with an immunoreceptor tyrosine-based inhibitory motif (ITIM) in its cytoplasmic domain (Ishida, et al., EMBO J., 11 :3887-3895 (1992)).
  • ITIM immunoreceptor tyrosine-based inhibitory motif
  • PD-1 a member of the CD28 family of receptors, is inducibly expressed on activated T cells, B cells, natural killer (NK) cells, monocytes, DC, and macrophages (Keir, et al Curr. Opin. Immunol. 19:309-314 (2007)).
  • the primary result of PD-1 ligation by its ligands is to inhibit signaling downstream of the T cell Receptor (TCR). Therefore, signal transduction via PD-1 usually provides a suppressive or inhibitory signal to the T cell that results in decreased T cell proliferation or other reduction in T cell activation.
  • B7-H1 is the predominant PD-1 ligand causing inhibitory signal transduction in T cells.
  • the present invention solves the problem of undesired T cell inhibition by providing agents that bind to PD-1 and thus prevent inhibitory signal transduction, or else bind to ligands of PD-1 such as B7-H1 , thereby preventing the ligand from binding to PD-1 to deliver an inhibitory signal. In either case, T cell responses, such as T cell proliferation or activation, are stimulated.
  • B7-H1 is the predominant PD-1 ligand, likely due to its broader distribution and higher expression levels. PD-1 inhibition occurs only when PD-1 and TCR are ligated in close proximity to each other, in the context of the immune synapse. PD-1 and its ligands have been the topic of several review articles.
  • B7-H1 is also over expressed in many cancers (including breast cancer, colon cancer, esophageal cancer, gastric cancer, glioma, leukemia, lung cancer, melanoma, multiple myeloma, ovarian cancer, pancreatic cancer, renal cell carcinoma, and urothelial cancer), and has been linked to poor prognosis.
  • B7-H1 is expressed by many tumor cell lines, especially following stimulation with interferon gamma (IFN- ⁇ ), and is also upregulated on tumor infiltrating myeloid derived suppressor cells (MDSC).
  • IFN- ⁇ interferon gamma
  • MDSC tumor infiltrating myeloid derived suppressor cells
  • PD-1 is up-regulated on tumor specific CD8 T cells and is associated with functional impairment, anergy, exhaustion, and apoptosis.
  • PD-1 upregulation has also been associated with dysfunctional and/or suppressive phenotypes on additional cell types, such as regulatory T cells (T
  • the present invention makes use of such molecular functions by providing treatment regimens for treating diseases through increased T cell activity, especially cancer and infectious diseases.
  • the present invention relates to a method of increasing T cell responses, for example, to an antigen, in a mammal in need of such increase, comprising administering to said mammal a compound that reduces inhibitory signal transduction in immune cells, especially T cells, and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal.
  • Compounds useful in the treatment regimen of the invention include those that bind to and block PD-1 receptors on T cells without triggering inhibitory signal transduction, compounds that bind to PD-1 ligands to prevent their binding to PD-1 , compounds that do both and compounds that prevent expression of genes that encode either PD-1 or natural ligands of PD-1. Such compounds are referred to herein as "PD-1 antagonists.”
  • PD-1 antagonists Compounds that bind to natural ligands of PD-1 include PD-1 itself, as well as active fragments of PD-1 , and in the case of the B7-H1 ligand, B7.1 proteins and fragments.
  • Such antagonists include proteins, antibodies, anti-sense molecules and small organics.
  • said T cell response is greater than that produced by either of said PD-1 antagonist or said potentiating agent when either is administered without the other.
  • compounds useful in the methods of the invention are those that bind to T cell surface molecules such as CTLA4 to prevent the inhibitory signals triggered by binding of natural ligands thereof or that bind to said natural ligands.
  • Such antagonists include proteins, antibodies, anti-sense molecules and small organics.
  • compounds useful in treatment regimens and compositions of the present invention include those that bind to PD-1 without triggering, inducing, increasing, facilitating and/or permitting co- ligation of PD-1 with TCR.
  • B7-DC comprises the amino acid sequence of SEQ ID NO: 1 , 2, 3 or 4.
  • Preferred such compounds are those incorporating the soluble domain of B7-DC (i.e., without transmembrane sequence).
  • Suitable fragments of B7-DC polypeptides include fragments containing the IgV and/or IgC domains or fragments containing only the IgV domain, with the latter being a preferred embodiment, with amino acids 20-121 of SEQ ID NO: 1 being a preferred example of an IgV domain.
  • Preferred PD-1 antagonists also include, but are not limited to, active fragments of natural ligands of PD-1 , such as B7-H1 polypeptides (disclosed in U.S. Patent No. 6,803,192, incorporated by reference herein in its entirety), especially soluble portions of these, including variants and homologs of these, as well as fusion proteins incorporating any of the foregoing, that bind to PD-1 without triggering inhibitory signal transduction.
  • active fragments of natural ligands of PD-1 such as B7-H1 polypeptides (disclosed in U.S. Patent No. 6,803,192, incorporated by reference herein in its entirety), especially soluble portions of these, including variants and homologs of these, as well as fusion proteins incorporating any of the foregoing, that bind to PD-1 without triggering inhibitory signal transduction.
  • Preferred compounds of the invention also include, but are not limited to, compounds, including active fragments, variants and homologs, that bind to natural ligands of PD-1 , such as fragments of B7-1 that bind to B7-H1 , as well as fusion proteins incorporating any of the foregoing, that bind to ligands of PD-1 to prevent the latter from binding to PD-1 to trigger inhibitory signal transduction.
  • compositions and methods of use thereof include a combination of a PD-1 receptor antagonist that binds to and blocks the PD-1 receptor, and a separate PD-1 receptor antagonist that binds to and blocks PD-1 receptor ligands.
  • a PD-1 receptor antagonist that binds to and blocks the PD-1 receptor
  • a separate PD-1 receptor antagonist that binds to and blocks PD-1 receptor ligands.
  • Another embodiment of the present invention provides PD-1 receptor antagonists that bind to the PD-1 receptor without triggering inhibitory signal transduction through the PD-1 receptor and also have the ability to bind and antagonize PD-1 receptor ligands, such as B7-H1 , that would otherwise trigger inhibitory signal transduction through the PD-1 receptor.
  • Other contemplated PD-1 receptor antagonists include bi-specific antibodies that can bind both the PD-1 receptor and PD-1 receptor ligands.
  • Preferred embodiments of compounds useful in the present invention also include antibodies that bind to PD-1 or CTLA4, thereby reducing, or abolishing, inhibitory signal transduction mediated by these sources.
  • Preferred compounds for use in the methods of the invention also include, but are not limited to, active fragments of ligands of CTLA4 (such as B7-1 and B7-2) that bind to CTLA4 to reduce subsequent inhibitory signals yet do not bind to CD28 or otherwise inhibit positive signal transduction by CD28.
  • Preferred compounds that prevent inhibitory signal transduction through PD-1 and thus act as PD-1 antagonists include, but are not limited to,
  • B7-DC antagonists especially soluble portions of these, including active fragments of these, variants and homologs of these, as well as fusion proteins incorporating any of the foregoing, that bind to B7-DC.
  • B7-DC polypeptides, fragments or variants thereof are coupled to other polypeptides to form fusion proteins that antagonize the PD-1 receptor by binding to the PD-1 receptor without causing inhibitory signal transduction through PD-1 , thereby reducing, or interfering with, ligand binding to PD-1 , particularly B7-H1 binding, and thereby interfering with inhibitory signal transduction through the PD-1 receptor.
  • fusion proteins are polypeptides comprising the amino acid sequence of SEQ ID NO: 9, 10, 12 or 13, as well as homologs thereof.
  • all or a portion of the extracellular domain (ECD) of B7-DC is part of a fusion protein wherein it is linked to a second polypeptide containing an Fc portion of an immunoglobulin.
  • ECD extracellular domain
  • B7-DC-lg especially where this structure is part of a homodimer wherein two B7-DC-lg molecules are linked to each other, such as by a disulfide linkage.
  • fragments useful in the compounds of the invention consist of at least 10, 15, 25, 50, 75, 100, 150, 200 or more contiguous amino acids of a polypeptide having the desired antagonist activity. Such fragments are also commonly part of fusion proteins for use in the invention.
  • the present invention relates to a method of increasing T cell responses in a mammal in need thereof, comprising administering to said mammal an effective treatment regimen comprising an anti-PD-1 antibody and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal.
  • the present invention relates to a method of increasing T cell responses in a mammal in need thereof, comprising administering to said mammal an effective treatment regimen comprising an immunomodulator, and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal.
  • immunomodulators include molecules that antagonize other CD28 family receptors (such as CTLA4) that inhibit T cell responses.
  • CTLA4 CD28 family receptors
  • a preferred embodiment uses an anti-CTLA4 antibody and a potentiating agent.
  • Additional immunomodulators include: molecules that agonize CD28 family receptors (such as CD28 and ICOS) that activate T cell responses; molecules that antagonize B7 family ligands (such as B7-H1 , B7-DC, B7-H4) that inhibit T cell responses; and molecules that agonize B7 family ligands (such as B7.1 and B7.2) that activate T cell responses.
  • CD28 family receptors such as CD28 and ICOS
  • B7 family ligands such as B7-H1 , B7-DC, B7-H4
  • agonize B7 family ligands such as B7.1 and B7.2
  • the treatment regimen of a PD-1 antagonist compound and a potentiating agent further comprises at least one additional therapeutic agent.
  • Additional therapeutic agents contemplated include immunomodulatory agents.
  • Exemplary immunomodulating agents for such methods include anti-PD-1 and anti-CTLA4 antibodies.
  • the potentiating agent is selected from cyclophosphamide and analogs of cyclophosphamide, Sunitinib (Sutent), anti- TGF ⁇ and lmatinib (Gleevac), a mitosis inhibitor, such as paclitaxel, an aromatase inhibitor, such as letrozole, an A2a adenosine receptor (A2AR) antagonist, an angiogenesis inhibitor, anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
  • Some of these agents reduce the number of Tregs (i.e., regulatory T lymphocytes or T-regs) within the tumor microenvironment.
  • the methods and/or compositions of the invention specifically contemplate use of any suitable adjuvant as part of said method and/or composition.
  • T cells can be contacted with PD-1 receptor antagonist and/or compositions thereof containing a potentiating agent in vitro, ex vivo or in vivo. Contacting T cells using PD-1 receptor antagonists and/or compositions thereof containing a potentiating agent can occur before, during or after activation of the T cell.
  • a molecule that prevents or reduces inhibitory signal transduction through PD-1 and the potentiating agent are administered at different times, such as where the potentiating agent is administered prior to administering the PD-1 antagonist. Such administration may be in conjunction with an additional therapeutic agent.
  • the treatment regimen includes administration of the potentiating agent at least 1 hour, or at least 2 hours, or at least 3 hours, or at least 5 hours, or at least 10 hours, or at least 15 hours, or at least 20 hours, or at least 24 hours, or at least 30 hours or even longer before administering any or all of the PD-1 antagonist, the anti-PD-1 antibody, the anti-CTLA4 antibody, and/or additional therapeutic agents.
  • Administration of the potentiating agent may also occur after administering any or all of the PD-1 antagonist, the anti-PD-1 antibody, the anti-CTLA4 antibody and/or additional therapeutic agents, such as no more than 1 hour, 2 hours, 3 hours, 5 hours, 10 hours, 15 hours, 20 hours, 24 hours, or even up to 30 hours after administering a PD-1 antagonist, or may occur in conjunction with administering the PD-1 antagonist.
  • the increased T cell response achieved as a result of the methods of the invention is sufficient to treat a disease, including one or more of cancer, viral infection, bacterial infection and parasitic infection.
  • a disease including one or more of cancer, viral infection, bacterial infection and parasitic infection.
  • the disease is cancer
  • such cancer is any one or more of bladder, brain, breast, cervical, colo-rectal, esophageal, kidney, liver, lung, nasopharangeal, pancreatic, prostate, skin, stomach, uterine, ovarian, testicular, or hematologic cancer.
  • the present invention includes compositions of the antagonists used in the methods of the invention, in a pharmaceutically acceptable carrier and wherein said PD-1 binding molecule and said potentiating agent are each present in an amount effective to produce increased T cell stimulation.
  • the invention includes medical kits comprising containers holding one or more of the agents for use in the invention together with pharmaceutical carriers for dilution thereof and instructions for administration.
  • both of said PD-1 receptor antagonist and potentiating agent may be present as components in a single container, in a pharmaceutically acceptable carrier, when said components are to be administered at the same time.
  • FIG. 1 shows that B7-DC-lg binds to PD-1.
  • Labeled B7-DC-lg was incubated at various concentrations with a CHO cell line constitutively expressing PD-1 or parent CHO cells that do not express PD-1. Binding was analyzed by flow cytometry. The median fluorescence intensity (MFI) of B7- DC-Ig (y-axis) is shown as a function of the concentration of probe (x-axis).
  • MFI median fluorescence intensity
  • B7-DC-lg binds to CHO.PD-1 cells (solid circle) but not untransfected CHO cells (gray triangle).
  • FIG. 2 shows that B7-DC-lg competes with B7-H1 for binding to PD- 1.
  • Unlabeled B7-DC-lg at various concentrations was first incubated with a CHO cell line constitutively expressing PD-1 before adding labeled B7-H1-lg to the cell mixture.
  • the median fluorescence intensity (MFI) of B7-H1-lg (y- axis) is shown as a function of the concentration of unlabeled B7-DC-lg competitor (x-axis) added.
  • B7-DC-lg As the concentration of unlabeled B7-DC-lg is increased the amount of labeled B7-H1-lg bound to CHO cells decreases, demonstrating that B7-DC-lg competes with B7-H1 for binding to PD-1.
  • Figure 3 shows the results of experiments wherein the combination of cyclophosphamide (CTX or Cytoxan®) and dimeric murine B7-DC-lg resulted in eradication of established CT26 tumors (colon carcinoma) in mice.
  • Graph A shows tumor volume (mm 3 ) versus days post tumor challenge in mice treated with 100 mg/kg of CTX on Day 10 while
  • Graph B shows tumor volume (mm 3 ) versus days post tumor challenge in mice treated with CTX on Day 10 followed a day later by the first B7-DC-lg administration. Each line in each graph represents one mouse.
  • Black arrow stands for B7-DC-lg administration.
  • Graph C shows average tumor volume.
  • Figure 4 shows the results of experiments wherein the combination of CTX and dimeric murine B7-DC-lg eradicated established CT26 tumors (colon carcinoma) in mice and protected against re-challenge with CT26.
  • Mice that were treated with CTX and B7-DC-lg and found to be free of tumor growth on day 44 following tumor inoculation were rechallenged with tumors. The mice were later rechallenged again on on Day 70. None of the mice displayed tumor growth by day 100.
  • FIG. 5 shows CTX and B7-DC-lg treatment resulted in generation of tumor specific memory CTL. Mice eradicated established CT26 subcutenous tumors post CTX and B7-DC-lg treatment were re-challenged with CT26 cells. Seven days later, splenocytes were isolated and pulsed with either ovalbumin, an irrelevant peptide, or AH1 , a CT26 specific peptide. Cells were stained with anti-CD8 antibody first followed by intracellular staining with anti- IFN ⁇ antibody prior to FACS analysis.
  • Figure 6 shows the effects of different doses of B7-DC-lg in combination with CTX on the eradication of established CT26 tumors in mice.
  • mice at age of 9 to 11 weeks were implanted subcutaneously with 1 E05 CT26 cells. On Day 9, mice were injected IP with 100 mg/kg of CTX. Twenty four hours later, on Day 10, mice were treated with 30, 100, or 300 ug of B7-DC-lg followed by 2 injections every week up to total 8 treatments. Tumor growth was measured two times per week.
  • Figure 7 shows the results of experiments wherein the combination of CTX and anti-PD-1 antibody resulted in eradication of established CT26 tumors (colon carcinoma) in mice.
  • Graph A shows tumor volume (mm 3 ) versus days post tumor challenge in untreated mice (i.e., mice treated with vehicle alone)
  • Graph B shows tumor volume (mm 3 ) versus days post tumor challenge in mice treated with anti-PD-1 alone starting on Day 11 at 300 ⁇ g per injection, 3 times per week, up to 12 injections
  • Graph C shows tumor volume (mm 3 ) versus days post tumor challenge in mice treated with CTX on Day 11 and the first anti-PD-1 administration on Day 12 at 300 ⁇ g per injection, 3 times per week, up to 12 injections.
  • Each line in each graph represents one mouse.
  • Black arrow stands for anti-PD-1 administration.
  • Figure 8 shows the results of experiments wherein the combination of CTX and anti-CTLA4 antibody resulted in eradication of established CT26 tumors (colon carcinoma) in mice.
  • Graph A shows tumor volume (mm 3 ) versus days post tumor challenge in mice treated with 100 mg/kg of CTX on Day 11
  • Graph B shows tumor volume (mm 3 ) versus days post tumor challenge in mice treated with CTX on Day 11 and anti-CTLA4 on Day 12 at 100 ⁇ g per injection, 2 times per week, up to 8 injections.
  • Each line in each graph represents one mouse.
  • Black arrow stands for anti-CTLA-4 administration.
  • Figure 9 shows the results of experiments wherein Balb/C mice at age of 9 to 11 weeks of age were implanted with 1 X 10 5 CT26 cells subcutaneously. On Day 9, mice were injected with 100 mg/kg of CTX, IP. Twenty four hours later, on Day 10, mice were treated with 100 ug of B7-DC- Ig. There were 5 groups: naive mice that did not receive any tumor cells, vehicle injected, CTX alone, CTX + B7-DC-lg or B7-DC-lg alone. Two naive mice and 4 mice from other groups were removed from the study on Day 11 (2 days post CTX) and Day 16 (7 days post CTX) for T cell analysis.
  • CTX and B7-DC-lg resulted in increased survival in mice with tail vein injection of a mouse prostate tumor cell line.
  • SP-1 cells were isolated from mouse lungs that were metastasized from TRAMP prostate tumor cell injection.
  • B10.D2 mice were first injected with 3x105 SP-1 cells via tail vein injection. On Day 5, 12 and 19, mice were injected with 50 mg/kg of CTX where was indicated. On Day 6, 13 and 20, mice were administered with 5 mg/kg of B7-DC-lg were it was indicated.
  • NT refers to "not treated”.
  • FIG. 11 Balb/C mice at age of 11-13 weeks were given isolated hepatic metastases using a hemispleen injection technique. The spleens of anesthetized mice were divided into two halves and the halves were clipped.
  • CT26 cells (1 E05) were injected into one hemispleen, and after 30 seconds, that hemispleen was resected and the splenic draining vein was clipped. On
  • mice received 1 injection of CTX at 50 mg/kg, IP. Twenty four hours later, on Day 11 , mice were treated with recombinant Listeria carrying AH 1 peptide, an immunodominant epitope of CT26, at 0.1 x LD 5O (1 x10 7 CFU), then on Day 14 and 17. Mice were also treated with B7-DC-lg on Day 11 and then on Day 18. Mouse overall survival was monitored. DEFINITIONS
  • inhibitory signal transduction is intended to mean any signal transduction having the effect of abolishing, or otherwise reducing, T cell responses against an antigen, whether by reducing T cell proliferation or by any other inhibitory mechanism, whereby the extent or duration of an immunogenic T cell response is decreased.
  • Such inhibitory signal transduction may be due to PD-1 binding to a natural ligand, such as binding of PD-1 by B7-H1 or some other member of this class of ligands, B7-DC, or may be due to binding of CTLA4 to ligands, such as B7-1 or B7-2.
  • compounds of the invention reduce such inhibitory signal transduction and include, but are not limited to, PD-1 antagonists and CTLA4 antagonists.
  • PD-1 antagonist means any molecule that attenuates inhibitory signal transduction mediated by PD-1 , found on the surface of T cells, B cells, natural killer (NK) cells, monocytes, DC, and macrophages.
  • Such an antagonist includes a molecule that disrupts any inhibitory signal generated by a PD-1 molecule on a T cell.
  • a PD-1 antagonist is a molecule that inhibits, reduces, abolishes or otherwise reduces inhibitory signal transduction through the PD-1 receptor signaling pathway. Such decrease may result where: (i) the PD-1 antagonist of the invention binds to a PD-1 receptor without triggering signal transduction, to reduce or block inhibitory signal transduction,; (ii) the PD-1 antagonist binds to a ligand (e.g.
  • a PD-1 antagonist of the invention is a molecule that effects a decrease in PD-1 inhibitory signal transduction, thereby increasing T cell response to one or more antigens.
  • CTLA4 antagonist means a compound that reduces CTLA4-mediated inhibition of T cell reactions.
  • CTLA4 delivers an inhibitory impulse upon binding of B7 ligands, such B7-1 and B7-2.
  • a CTLA4 antagonist is one that dirupts binding of said ligands to CTLA4 on activated T cells.
  • the antagonist is an anti- CTLA4 antibody that binds CTLA4 to prevent ligand binding.
  • active fragment refers to a portion of a natural polypeptide, or a polypeptide with high sequence homology (for example, at least 80%, 85%, 90%, 95%, 98%, or 99% amino acid sequence identity) to a natural polypeptide and that exhibits PD-1 antagonist activity, for example, by binding PD-1 or by binding to a ligand of PD-1.
  • a fragment would consist of the extracellular domain (ECD) of a B7-DC protein that binds to PD-1 , such as SEQ ID NO: 3, preferably amino acids 20 to 221 thereof.
  • an active fragment would be a portion of said polypeptide comprising a binding domain that binds to a natural ligand of PD-1 to prevent stimulation of PD-1 mediated inhibitory signal transduction by said ligand.
  • Active fragments may be identified by their ability to compete with the molecule they are derived from for binding to a natural binding site. For example, active fragments will compete with wild-type B7-DC for binding to PD-1.
  • active fragment means an antigen binding portion of an antibody that is less than an entire immunoglobulin.
  • fragments include Fab and F(ab 2 )' fragments, capable of reacting with and binding to any of the polypeptides disclosed herein as being receptors or ligands.
  • Fab and F(ab') 2 fragments lack the Fc portion of an intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody (Wahl et al., J. Nuc. Med. 24:316-325 (1983)).
  • Fv fragments Hochman, J. et al. (1973) Biochemistry 12:1130-1135; Sharon, J.
  • soluble portion of a PD-1 antagonist means that portion of the full length polypeptide that does not include any part of the transmembrane portion or segment.
  • a soluble portion would include the extracellular portion (with or without the N- terminal signal sequence) but would not include any part of the transmembrane portion (or, at least, not enough to reduce solubility).
  • ECD of human B7-DC is shown as SEQ ID NO: 3 and consists of both the
  • IgV-like and IgC-like domains of the full length molecule i.e., amino acids 20-
  • a "co-stimulatory polypeptide” is a polypeptide that, upon interaction with a cell-surface molecule on T cells, modulates the activity of the T cell.
  • the response of the T cell can be an effector (e.g., CTL or antibody-producing B cell) response, a helper response providing help for one or more effector (e.g., CTL or antibody-producing B cell) responses, or a suppressive response.
  • treatment regimen refers to a treatment of a disease or a method for achieving a desired physiological change, such as increased or decreased response of the immune system to an antigen or immunogen, such as an increase or decrease in the number or activity of one or more cells, or cell types, that are involved in such response, wherein said treatment or method comprises administering to an animal, such as a mammal, especially a human being, a sufficient amount of two or more chemical agents or components of said regimen to effectively treat a disease or to produce said physiological change, wherein said chemical agents or components are administered together, such as part of the same composition, or administered separately and independently at the same time or at different times (i.e., administration of each agent or component is separated by a finite period of time from one or more of the agents agents or components) and where administration of said one or more agents or components achieves a result greater than that of any of said agents or components when administered alone or in isolation.
  • a desired physiological change such as increased or decreased response of the immune system to an antigen or immunogen, such as an increase or
  • isolated is meant to describe a compound of interest (e.g., either a polynucleotide or a polypeptide) that is in an environment different from that in which the compound naturally occurs e.g. separated from its natural milieu such as by concentrating a peptide to a concentration at which it is not found in nature. "Isolated” is meant to include compounds that are within samples that are substantially enriched for the compound of interest and/or in which the compound of interest is partially or substantially purified.
  • polypeptide refers to a chain of amino acids of any length, regardless of modification (e.g., phosphorylation or glycosylation).
  • a polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide.
  • a "variant" polypeptide contains at least one amino acid sequence alteration as compared to the amino acid sequence of the corresponding wild-type polypeptide.
  • an “amino acid sequence alteration” can be, for example, a substitution, a deletion, or an insertion of one or more amino acids.
  • the terms "portion,” “segment,” and “fragment,” when used in relation to polypeptides refer to a continuous sequence of residues, such as amino acid residues, which sequence forms a subset of a larger sequence. For example, if a polypeptide were subjected to treatment with any of the common endopeptidases, such as trypsin or chymotrypsin, the oligopeptides resulting from such treatment would represent portions, segments or fragments of the starting polypeptide.
  • a “fragment” of a polypeptide thus refers to any subset of the polypeptide that is a shorter polypeptide of the full length protein. Generally, fragments will be five or more amino acids in length.
  • a derivative, analog or homolog, of a polypeptide (or fragment thereof) of the invention may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the mature polypeptide or a proprotein sequence.
  • Such derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
  • valency refers to the number of binding sites available per molecule.
  • the term "percent identity” or “percent identical,” when referring to a sequence, means that a sequence is compared to a claimed or described sequence after alignment of the sequence to be compared (the "Compared Sequence") with the described or claimed sequence (the “Reference Sequence”).
  • the Percent Identity is then determined according to the following formula:
  • C is the number of differences between the Reference Sequence and the Compared Sequence over the length of alignment between the Reference Sequence and the Compared Sequence wherein (i) each base or amino acid in the Reference Sequence that does not have a corresponding aligned base or amino acid in the Compared Sequence and (ii) each gap in the Reference Sequence and (iii) each aligned base or amino acid in the Reference Sequence that is different from an aligned base or amino acid in the Compared Sequence, constitutes a difference; and R is the number of bases or amino acids in the Reference Sequence over the length of the alignment with the Compared Sequence with any gap created in the Reference Sequence also being counted as a base or amino acid.
  • the Compared Sequence has the specified minimum percent identity to the Reference Sequence even though alignments may exist in which the hereinabove calculated Percent Identity is less than the specified Percent Identity.
  • the term "conservative amino acid substitution” means a substitution wherein the substituted amino acid has similar structural or chemical properties, and “non-conservative" amino acid substitutions are those in which the charge, hydrophobicity, or bulk of the substituted amino acid is significantly altered. Non-conservative substitutions will differ more significantly in their effect on maintaining (a) the structure of the peptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • conservative amino acid substitutions include those in which the substitution is within one of the five following groups: 1) small aliphatic, nonpolar or slightly polar residues (AIa 1 Ser, Thr, Pro, GIy); 2) polar, negatively charged residues and their amides (Asp, Asn, GIu, GIn); polar, positively charged residues (His, Arg, Lys); large aliphatic, nonpolar residues (Met, Leu, Me, VaI, Cys); and large aromatic resides (Phe, Tyr, Trp).
  • non-conservative amino acid substitutions are those where 1 ) a hydrophilic residue, e.g., seryl or threonyl, is substituted for (or by) a hydrophobic residue, e.g., leucyl, isoleucyl, phenylalanyl, valyl, or alanyl; 2) a cysteine or proline is substituted for (or by) any other residue; 3) a residue having an electropositive side chain, e.g., lysyl, arginyl, or histidyl, is substituted for (or by) an electronegative residue, e.g., glutamyl or aspartyl; or 4) a residue having a bulky side chain, e.g., phenylalanine, is substituted for (or by) a residue that does not have a side chain, e.g., glycine.
  • a hydrophilic residue e.g., seryl or th
  • the terms “individual”, “host”, “subject”, and “patient” are used interchangeably herein, and refer to a mammal, including, but not limited to, primates, for example, human beings, as well as rodents, such as mice and rats, and other laboratory animals.
  • the term "effective amount” or “therapeutically effective amount” means a dosage sufficient to treat, inhibit, or alleviate one or more symptoms of a disease state being treated or to otherwise provide a desired pharmacologic and/or physiologic effect, especially enhancing T cell response to a selected antigen.
  • the precise dosage will vary according to a variety of factors such as subject-dependent variables (e.g., age, immune system health, etc.), the disease, and the treatment being administered.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the therapeutic compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • antibody is meant to include both intact molecules as well as fragments thereof that include the antigen-binding site.
  • Whole antibody structure is often given as H 2 L 2 and refers to the fact that antibodies commonly comprise 2 light (L) amino acid chains and 2 heavy (H) amino acid chains. Both chains have regions capable of interacting with a structurally complementary antigenic target. The regions interacting with the target are referred to as “variable” or “V” regions and are characterized by differences in amino acid sequence from antibodies of different antigenic specificity.
  • the variable regions of either H or L chains contains the amino acid sequences capable of specifically binding to antigenic targets. Within these sequences are smaller sequences dubbed "hypervariable" because of their extreme variability between antibodies of differing specificity.
  • CDR regions Such hypervariable regions are also referred to as “complementarity determining regions” or “CDR” regions. These CDR regions account for the basic specificity of the antibody for a particular antigenic determinant structure.
  • the CDRs represent non-contiguous stretches of amino acids within the variable regions but, regardless of species, the positional locations of these critical amino acid sequences within the variable heavy and light chain regions have been found to have similar locations within the amino acid sequences of the variable chains.
  • the variable heavy and light chains of all antibodies each have 3 CDR regions, each non-contiguous with the others (termed L1 , L2, L3, H1 , H2, H3) for the respective light (L) and heavy (H) chains.
  • the accepted CDR regions have been described by Kabat et al, J. Biol.
  • the antibodies disclosed according to the invention may also be wholly synthetic, wherein the polypeptide chains of the antibodies are synthesized and, possibly, optimized for binding to the polypeptides disclosed herein as being receptors.
  • Such antibodies may be chimeric or humanized antibodies and may be fully tetrameric in structure, or may be dimeric and comprise only a single heavy and a single light chain.
  • the present invention provides a treatment regimen, or combination therapy, for treating disease in mammals comprising a compound that reduces or abolishes inhibitory signal transduction in T cells, preferably human T cells, administered in conjunction with a potentiating agent to increase an immune response.
  • the methods of the invention also relate to the use of broad spectrum immunomodulators and compositions of these.
  • the increased T cell response resulting from these methods is greater than any increased T cell response resulting from administering the same dose of either of said PD-
  • compositions and regimens are useful to stimulate or enhance immune responses involving T cells.
  • the methods of the invention are most useful in treating a disease condition that would benefit from an increase in T cell activity and where the increased T cell response is necessary or sufficient to treat said disease, even though the disease is not specifically caused or aggravated by a reduced T cell response.
  • the type of disease to be treated or prevented is a malignant tumor or a chronic infectious disease caused by a bacterium, virus, protozoan, helminth, or other intracellular microbial pathogen that is attacked, i.e., by cytotoxic T lymphocytes.
  • Activation of T cells using the disclosed compositions is also advantageous to treat or prevent conditions characterized by immunosuppression.
  • the T cell response can be regulated by molecules that bind to receptors on the T cell surface and molecules that bind to ligands of such receptors.
  • molecules that bind PD-1 to reduce its inhibitory effect and/or molecules that bind one or more PD-1 ligands to reduce their ability to bind PD-1 have the effect of reducing the ability of PD-1 to inhibit T cell response, thereby increasing this response and the immunological effects thereof.
  • compositions containing antagonists of PD-1 receptors include compounds or agents that either bind to and block a ligand of PD- 1 to interfere with or inhibit the binding of the ligand to the PD-1 receptor, or bind directly to and block the PD-1 receptor without inducing inhibitory signal transduction through the PD-1 receptor.
  • the PD-1 receptor antagonist binds directly to the PD-1 receptor without triggering inhibitory signal transduction and also binds to a ligand of the PD-1 receptor to reduce or inhibit the ligand from triggering signal transduction through the PD-1 receptor.
  • PD-1 signaling requires binding to a PD-1 ligand (such as B7-H1 or B7-DC) in close proximity to a peptide antigen presented by major histocompatibility complex (MHC) (see, for example, Freeman Proc. Natl. Acad. Sci. U. S. A 105:10275-10276 (2008)). Therefore, proteins, antibodies or small molecules that prevent co- ligation of PD-1 and TCR on the T cell membrane are useful PD-1 antagonists contemplated by this invention.
  • MHC major histocompatibility complex
  • Exemplary PD-1 receptor antagonists include, but are not limited to B7- DC polypeptides, including homologs and variants of these, as well as active fragments of any of the foregoing, and fusion proteins that incorporate any of these.
  • the fusion protein comprises the soluble portion of B7-DC coupled to the Fc portion of an antibody, such as human IgG, and does not incorporate all or part of the transmembrane portion of human B7-DC.
  • the PD-1 receptor antagonists can also be small molecule antagonists or antibodies that reduce or interfere with PD-1 receptor signal transduction by binding to ligands of PD-1 or to PD-1 itself, especially where co-ligation of PD-1 with TCR does not follow such binding, thereby not triggering inhibitory signal transduction through the PD-1 receptor.
  • the PD-1 receptor antagonists provided herein are generally useful in vivo and ex vivo as immune response-stimulating therapeutics.
  • the disclosed antagonist compositions are useful for treating a subject having or being predisposed to any disease or disorder to which the subject's immune system mounts an immune response.
  • B7-DC proteins can be used as PD-1 receptor antagonists.
  • B7-DC is a natural ligand of PD-1 and binds to PD-1 with higher affinity than B7-H1 , and can thus inhibit B7-H1 :PD-1 interactions.
  • Suitable B7- DC polypeptides, including variants, homologs and fragments thereof, can be obtained from the following full length human B7-DC polypeptides with (SEQ ID NO:1) or without (SEQ ID NO:2) the endogenous signal peptide.
  • the B7 family of molecules including B7-DC, are expressed at the cell surface with a membrane proximal constant IgC domain and a membrane distal IgV domain. Receptors for these ligands share a common extracellular IgV-like domain. Interactions of receptor-ligand pairs are mediated predominantly through residues in the IgV domains of the ligands and receptors.
  • IgV domains are described as having two sheets that each contains a layer of ⁇ -strands. These ⁇ -strands are referred to as A', B, C, C, C", D, E, F and G.
  • B7-DC a transmembrane protein, in its monomeric form, comprises IgV and IgC domains that make up the extracellular portion of the molecule (the extracellular domain, or ECD), with the IgV-like domain being responsible, in whole or in part, for PD-1 binding as well as other functions recited in the methods of the invention.
  • the IgV domain is characterized in that it possesses a disulfide bond linking the B and F strands (referred to above), which appears to be characteristic of many IgV domains and possesses a similar three-dimensional structure with the IgV domains of both B7-1 and B7-2 (see Molnar et al.(2008), supra).
  • the B7-DC variant polypeptides contain amino acid alterations (i.e., substitutions, deletions or insertions) within one or more of these ⁇ -strands in any possible combination.
  • B7-DC variants contain one or more amino acid alterations (i.e., substitutions, deletions or insertions) within the A', C, C, C", D, E, F or G ⁇ -strands.
  • B7-DC variants contain one or more amino acid alterations in the G ⁇ -strand.
  • variant B7-DC polypeptide fragments include the IgC and IgV domains of B7-DC.
  • variant B7-DC polypeptide fragments include the IgV domain of B7-DC.
  • variant B7-DC polypeptides include soluble fragments. Soluble B7-DC fragments are fragments of B7-DC that may be shed, secreted or otherwise extracted from the producing cells.
  • variant B7-DC polypeptide fragments include the entire extracellular domain of B7-DC.
  • the extracellular domain of B7-DC includes amino acids from about 20 to about amino acid 221 of murine or human B7-DC or active fragments thereof.
  • variant B7-DC polypeptide fragments include the IgC and IgV domains of B7-DC.
  • variant B7-DC polypeptide fragments include the IgV domain of B7-DC.
  • PD-1 signaling is thought to require binding to a PD-1 ligand (typically
  • MHC major histocompatibility complex
  • the PD-1 antagonist useful in the methods and compositions of the invention include fragments of the B7-DC protein incorporating the ECD.
  • the fragments of B7-DC include part of the extracellular domain that comprise the an IgV or IgV-like domain, preferably amino acids 20-221 , more preferably 20-121 , that are sufficient to bind to the PD-1 receptor to interfere with, or prevent, or otherwise reduce inhibitory signal transduction through the PD-1 receptor.
  • the B7-DC fragment competes with B7-H1 for binding to PD-1 receptors.
  • variant B7-DC polypeptide fragments may contain a region of the polypeptide that is important for binding to PD-1. These polypeptide fragments may be useful to compete for binding to PD-1 and to prevent native B7-DC from binding to PD-1. By competing for binding to PD- 1 , these fragments may be useful to enhance an immune response, as inhibiting interactions of B7-H1 and B7-DC with PD-1 inhibits the suppression of immune responses that would otherwise occur.
  • a polypeptide fragment of mouse or human B7-DC that could competitively bind to PD-1 can contain, for example, amino acids 101-108 or 110-114.
  • B7-DC fragments useful in the methods and/or compositions of the invention include, but are in no way limited to, the following B7-DC extracellular domains:
  • ECD Human B7-DC extracellular domain
  • a PD-1 antagonist useful in the compositions and methods of the invention also includes a fusion protein (as described below) that comprises first and second polypeptide portions, wherein said fusion protein, or at least the first polypeptide portion thereof, possesses PD-1 antagonist activity, especially where said fusion protein binds to and blocks PD-1 or binds to and blocks a ligand of PD-1.
  • the first polypeptide portion of such fusion protein can comprise, or consist of, any of the PD-1 antagonistic polypeptides, or PD- 1 binding fragments thereof, otherwise recited herein for use as PD-1 antagonists in the methods of the invention.
  • the recited first polypeptide portion is N-terminal to the recited second polypeptide portion.
  • the recited first polypeptide portion is linked to the recited second polypeptide portion by an oligopeptide in addition to the amino acids composing the recited first and second polypeptide portions, where said linking amino acids do not substantially decrease the PD-1 antagonist activity of said fusion protein.
  • the dimer results from the covalent bonding of Cys residues in the CH regions of two of the Ig heavy chains that are the same Cys residues that are disulfide linked in dimerized normal Ig heavy chains.
  • polypeptide sequences that are routinely used as fusion protein binding partners are well known in the art.
  • useful polypeptide binding partners include, but are not limited to, green fluorescent protein (GFP) 1 glutathione S-transferase (GST), polyhistidine, myc, hemaglutinin, FlagTM tag (Kodak, New Haven, CT), maltose E binding protein and protein A.
  • GFP green fluorescent protein
  • GST glutathione S-transferase
  • polyhistidine polyhistidine
  • myc myc
  • hemaglutinin FlagTM tag
  • FlagTM tag Kodak, New Haven, CT
  • maltose E binding protein and protein A protein A.
  • Still another embodiment provides a tetramer construct having a BirA substrate fused to the extracellular domain of a variant B7-DC polypeptide. Methods for making tetramer constructs are known in the art (see Pertovas, et al., J. Exp. Med.,
  • Exemplary murine B7-DC fusion proteins contain amino acids 20-221 of murine B7-DC fused to amino acids 237-469 of murine lgG2a (CAA49868).
  • human B7-DC fusion proteins contain amino acids 20-221 of human B7-DC fused to amino acids 245-476 of human IgGI (AAA02914).
  • the signal peptides for B7-DC fusion proteins include the endogenous signal peptides or any other signal peptide that facilitates secretion of the fusion protein from a host.
  • the first polypeptide would include only the IgV domain.
  • inventions may comprise the hinge and Fc domain of an IgG antibody, such IgGI , with none of the variable region present.
  • Other embodiments include use of the hinge and Fc region of lgG2 or lgG4, especially having an N297Q or other mutation that reduces effector function.
  • the polypeptide useful as a PD-1 antagonist, or the first polypeptide portion of a fusion protein useful as a PD-1 antagonist comprises an amino acid sequence that has at least 60%, or at least 65%, or at least 70%, or at least
  • amino acids 1-221 of SEQ ID NO: 1 preferably amino acids 20-221 of SEQ ID NO: 1 , or amino acids 26-221 of SEQ ID NO: 1 , or amino acids 1 -202 of SEQ ID NO: 3 or 4, more preferably amino acids 20-121 of SEQ ID NO: 1 or amino acids 1 -102 of SEQ ID NO: 3 or 4.
  • a polypeptide useful as a PD-1 antagonist or the first polypeptide portion of a fusion protein useful as a PD-1 antagonist, consists of amino acids 1-221 of SEQ ID NO: 1 , or consists of amino acids
  • SEQ ID NO: 1 20-221 of SEQ ID NO: 1 , or consists of amino acids 26-221 of SEQ ID NO: 1 , or consists of amino acids 1-202 of SEQ ID NO: 3 or 4. In one embodiment (SEQ ID NO: 2), it does not comprise amino acids 1-19 of SEQ ID NO: 1.
  • a PD-1 antagonist polypeptide, or first polypeptide portion of a PD-1 antagonist fusion protein comprises the amino acid sequence 20-121 of SEQ ID NO: 1 , preferably where it comprises the amino acid sequence WDYKY at residues 110-114 thereof, or where it comprises amino acids 1-102 of SEQ ID NO: 3, preferably where it comprises the amino acid sequence WDYKY at residues 91-95 thereof.
  • such percent identities are achieved by reliance on conservative amino acid substitutions as defined elsewhere herein.
  • the PD-1 antagonist polypeptide, or first polypeptide portion of a PD-1 antagonist fusion protein does not comprise amino acids 1-19 of SEQ ID NO: 1 , or does not comprise any portion of a transmembrane domain, especially not the entire such domain, or does not comprise any portion of the intracellular (or soluble) domain, especially not the entire such domain, of a PD-1 ligand or other PD-1 antagonist protein.
  • such antagonist, or first polypeptide portion comprises only the extracellular domain (ECD) of SEQ ID NO:1 and is thus comprised only of a soluble portion of the polypeptide of said sequence, or a fragment of said soluble portion.
  • the PD-1 antagonist polypeptide, or first polypeptide portion of a PD-1 antagonist fusion protein comprises the IgV domain, or IgV-like domain, or PD-1 binding fragment thereof, of a PD-1 ligand, or consists of the IgV domain, or IgV-like domain, or PD-1 binding fragment thereof, of a PD-1 ligand.
  • PD-1 ligand is a wild-type B7-DC or B7-H1 molecule, preferably mouse or primate, preferably human, wild-type B7-DC or B7-H1 molecule.
  • a PD-1 antagonist of the invention also includes a PD-1 binding fragment of amino acids 20-121 of SEQ ID NO: 1 (human full length), or amino acids 1-102 of SEQ ID NO: 3 (extracellular domain or ECD).
  • the polypeptide or PD-1 binding fragment also incorporates amino acids WDYKY at residues 110-114 of SEQ ID NO: 1 or WDYKY at residues 91-95 of SEQ ID NO: 3.
  • a PD-1 binding fragment comprises at least 10 , or at least 20, or at least 30, or at least 40, or at least 50, or at least 60, or at least 70, or at least 75, or at least 80, or at least 85, or at least 90, or at least 95, or at least 100 contiguous amino acids of the sequence of amino acids 20-121 of SEQ ID NO: 1 , wherein a preferred embodiment of each such PD-1 binding fragment would comprise as a sub-fragment the amino acids WDYKY found at residues 110-114 of SEQ ID NO: 1 or WDYKY at residues 91-95 of SEQ ID NO: 3.
  • polypeptides and PD-1 binding fragments specifically contemplated by the invention include the polypeptide sequence of amino acids 20-121 of SEQ ID NO: 1 (human full length) and PD-1 binding fragments thereof, wherein, in such polypeptide or PD-1 binding fragment, a cysteine is present at residues 42 and/or 102, with a cysteine at both positions being preferred, and/or wherein a phenylalanine is present at residue 21 , and/or wherein a glutamic acid is present at residue 28, and/or wherein a threonine, and/or wherein a glutamine is present at residue 60, and/or wherein a glutamic acid is present at residue 101 , and/or wherein isoleucine is present at residue 103, and/or wherein an isoleucine is present at residue 105, and/or wherein a glycine is present at residue 107, and/or wherein valine is present at residue 108, and/or wherein a tryp
  • Additional preferred polypeptides and PD-1 binding fragments specifically contemplated by the invention include the polypeptide sequence of amino acids 1-102 of SEQ ID NO: 3 (human ECD) or SEQ ID NO: 4 (murine ECD) and PD-1 binding fragments thereof, wherein, in such polypeptide or PD-1 binding fragment, a cysteine is present at residues 23 and/or 83, with a cysteine at both positions being preferred, and/or wherein a phenylalanine is present at residue 2, and/or wherein a glutamic acid is present at residue 9, and/or wherein a threonine or arginine is present at residue 37, with threonine preferred, and/or wherein a glutamine is present at residue 41 , and/or wherein arginine is present at residue 82, and/or wherein a leucine is present at residue 84, and/or wherein an isoleucine is present at residue 86, and/or wherein a glycine is present
  • any of the above polypeptides may also incorporate portions or fragments, for example, from 1 to 10 contiguous amino acids, drawn from the signal, transmembrane or C-terminal domains of the B7-DC or B7-H1 polypeptide, such as that of mouse or primate, preferably human.
  • Such polypeptides and/or PD-1 binding fragments can also be present in any of the fusion proteins of the invention, for example, where such polypeptide or PD-1 binding fragment represents the "first polypeptide" of such fusion protein.
  • the molecule combined with a potentiating agent for use in a treatment regimen of the invention, comprises a PD-1 binding fragment of amino acids 20-221 of SEQ ID NO: 1.
  • the fragment is from amino acids 20 - 121 of SEQ ID NO: 1 , preferably where the fragment contains amino acids 110-114 of SEQ ID NO: 1.
  • more than one such fragment is present (as described elsewhere herein) and the molecule comprises at least 2, 3, 4, 5 or more fragments of a B7-DC protein, especially where the fragment is part of, or contains part of, amino acids 20-221 of SEQ ID NO: 1.
  • At least one said fragment is from amino acids 20 - 121 of SEQ ID NO: 1 , more preferrably wherein at least one said fragment includes amino acids 110-114 of SEQ ID NO: 1 (i.e., the sequence WDYKY (SEQ ID NO: 14)).
  • the PD-1 binding fragment comprises at least 10, or at least 25, or at least 50, or at least 75, or at least 100 contiguous amino acids in length.
  • the endogenous human signal peptide has the following sequence MIFLLLMLSL ELQLHQIAA (SEQ ID NO:5) and represents the first 19 amino acids of SEQ ID NO: 1.
  • the polypeptide fragments of B7-DC can include 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 contiguous amino acids of the endogenous or heterologous signal peptide (which can be used to produce a recombinant B7-DC polypeptide by expression in and secretion from a transformed cell).
  • a useful B7-DC polypeptide can include 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 contiguous amino acids of the transmembrane domain of B7-DC, and/or 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 contiguous amino acids of the cytoplasmic domain, or combinations thereof provided the B7-DC fragment retains the ability to antagonize the PD-1 receptor.
  • the phenotypes of PD-1-/- mice provide direct evidence for PD-1 being a negative regulator of immune responses in vivo.
  • mice on the C57BL/6 background slowly develop a lupus-like glomerulonephritis and progressive arthritis (Nishimura, et al., Immunity, 11 :141-151 (1999)).
  • PD-1-/- mice on the BALB/c background rapidly develop a fatal autoimmune dilated cardiomyopathy (Nishimura, et al., Science. 291 :319-322 (2001)).
  • B7-DC can function to costimulate activate T cell responses.
  • B7-DC causes increased proliferation and production of cytokines in vitro (Tseng, et al., J. Exp. Med.
  • B7-DC proteins, variants, fragments and fusions thereof may have the advantage of directly enhancing T cell responses by binding to an unknown receptor that activates the T cell, in addition to enhancing T cell responses by preventing the PD-1 mediated inhibitory signal transduction.
  • the compound for use in combination with a potentiating agent in the treatment regimen of the invention is, or comprises, a fragment of a mammalian B7-H1 , preferably from mouse or primate, preferably human, wherein said fragment binds to and blocks PD-1 but does not result in inhibitory signal transduction through PD-1 and said fragment is at least 10, or at least 20, or at least 30, or at least 40, or at least 50, or at least 60, or at least 70, or at least 80, or at least 90, or at least 100 contiguous amino acids in length.
  • the fragment can be of variable length so long as it has the function of binding to PD-1 but does not produce inhibitory signal transduction that results in reduced T cell proliferation.
  • B7-H1 fragments also find use as part of the first polypeptide portion of fusion proteins of the invention.
  • B7-H1 sequences are as follows:
  • Murine B7-H1 (SEQ ID NO: 17)
  • Macaca mulatta PD-L1 (SEQ ID NO: 18) MRIFAVFIFT IYWHLLNAFT VTVPKDLYW EYGSNMTIEC RFPVEKQLGL 60
  • B7-H1-lg proteins are described in WO/2001/014557 (pub. 1 March 2001 ) and in WO/2002/079499 (pub. 10 October 2002).
  • polypeptides of the invention include those that bind to the ligands of the PD-1 receptor. These include the PD-1 receptor protein, or soluble fragments thereof, which can bind to the PD-1 ligands, such as B7-H1 or B7-DC, and prevent binding to the endogenous PD-1 receptor, thereby preventing inhibitory signal transduction. B7-H1 has also been shown to bind the protein B7.1 (Butte et al., Immunity, Vol. 27, pp. 111-122, (2007)).
  • Such fragments also include the soluble ECD portion of the PD-1 protein that includes mutations, such as the A99L mutation, that increases binding to the natural ligands (Molnar et al., Crystal structure of the complex between programmed death-1 (PD-1) and its ligand PD-L2, PNAS, Vol. 105, pp. 10483-10488 (29 July 2008)),.
  • B7-1 or soluble fragments thereof which can bind to the B7-H1 ligand and prevent binding to the endogenous PD-1 receptor, thereby preventing inhibitory signal transduction, are also useful.
  • PD-1 polypeptides useful in the methods of the invention are as follows:.
  • B7-1 and fragments thereof can also bind to B7-H1 and send inhibitory transducstion to T cells through B7-H1 , blocking of this interaction can also reduce inhibitory signal transduction that occurs through B7-H1.
  • Compounds for use in the invention include those molecules that block this type of interaction. Such molecules have been disclosed in Butte et al (2007), supra, and include anti-B7-H1 antibodies with dual-specificity that block either the B7-H1 :B7-1 and B7- H1 :PD-1 interaction as well as antibodies exhibiting mono-specificity that block the PD-L1 :B7-1 interaction. Compounds that block this interaction by blocking B7-1 are also useful, and include anti-B7-1 antibodies.
  • Polypeptides useful in the invention, as described, include those that are mutated to contain one or more amino acid substitutions, deletions, or insertions. Methods for mutagenesis are known in the art.
  • the mutated or variant polypeptides inhibit or reduce inhibitory signal transduction through PD-1 receptors by binding to ligands of PD-1.
  • the variants e.g. B7-DC polypeptides
  • the variant polypeptides may be of any species of origin. In one embodiment, the variant polypeptide is from a mammalian species. In a preferred embodiment, the variant polypeptide is of murine or primate, preferably human, origin.
  • the variant polypeptide is a B7-DC polypeptide that has the same binding affinity to PD-1 as wildtype or non-variant B7-DC but does not have or has less than 10% ability to trigger inhibitory signal transduction through the PD-1 receptor relative to a non-mutated B7-DC polypeptide.
  • the variant B7-DC polypeptide has 10%, 20%, 30%, 40%, 50%, or 60% more binding affinity to PD-1 than wildtype B7- DC without triggering PD-1 inhibitory signaling transduction.
  • a variant polypeptide (e.g. a variant B7-DC polypeptide) includes those having any combination of amino acid substitutions, deletions or insertions so long as the PD-1 antagonizing activity is not substantially reduced versus the wild type. However, where there is such a reduction, this should be by no more than half that of the wild type so that said variant has at least 50% of the PD-1 antagonist activity of the wild type protein, preferably at least 60%, more preferably at least 80%, most preferably at least 90% or 95%, with at least 100% being especially preferred. Increases in such activity resulting from said variant is even more desirable.
  • isolated B7-DC variant polypeptides have amino acid alterations such that their amino acid sequence shares at least 60, 70, 80, 85, 90, 95, 97, 98, 99, 99.5 or 100% identity with an amino acid sequence of a wild type B7-DC polypeptide, especially that from a mammal, preferably wild type murine or wild type primate, preferably human, B7-DC polypeptide.
  • Polypeptide sequence identity can be calculated using the definition of % identity provided hereinabove.
  • Amino acid substitutions in polypeptides may be "conservative" or
  • B7 family molecules including B7-DC, are expressed at the cell surface with a membrane proximal constant IgC domain and a membrane distal IgV domain. Receptors for these ligands share a common extracellular IgV-like domain. Interactions of receptor-ligand pairs are mediated predominantly through residues in the IgV domains of the ligands and receptors.
  • IgV domains are described as having two sheets that each contains a layer of ⁇ -strands. These ⁇ -strands are referred to as A', B, C, C, C", D, E, F and G.
  • the B7-DC variant polypeptides contain amino acid alterations (i.e., substitutions, deletions or insertions) within one or more of these ⁇ -strands in any possible combination.
  • B7-DC variants contain one or more amino acid alterations (i.e., substitutions, deletions or insertions) within the A', C, C 1 C", D, E, F or G ⁇ -strands.
  • B7-DC variants contain one or more amino acid alterations in the G ⁇ -strand.
  • a variant B7-DC polypeptide can contain, without limitation, substitutions, deletions or insertions at positions that do not substantially reduce binding to PD-1 relative to non-mutated B7-DC. It is understood, however, that substitutions at the recited amino acid positions can be made using any amino acid or amino acid analog.
  • substitutions at the recited positions can be made with any of the naturally-occurring amino acids (e.g., alanine, aspartic acid, asparagine, arginine, cysteine, glycine, glutamic acid, glutamine, histidine, leucine, valine, isoleucine, lysine, methionine, proline, threonine, serine, phenylalanine, tryptophan, or tyrosine).
  • amino acids e.g., alanine, aspartic acid, asparagine, arginine, cysteine, glycine, glutamic acid, glutamine, histidine, leucine, valine, isoleucine, lysine, methionine, proline, threonine, serine, phenylalanine, tryptophan, or tyrosine.
  • Preferred fragments include all or part of the extracellular domain of
  • B7-DC effective to bind to PD-1.
  • variant B7-DC polypeptide fragments are those that retain the ability to bind to PD-1 without triggering PD-1 inhibitory signal transduction.
  • One embodiment provides a variant B7-DC polypeptide that is a fragment of full-length B7-DC and typically has at least 20 percent, 30 percent, 40 percent, 50 percent, 60 percent, 70 percent, 80 percent, 90 percent, 95 percent, 98 percent, 99 percent, 100 percent, or even more than 100 percent of the PD-1 antagonist activity of the full-length variant B7-DC polypeptide.
  • variant B7-DC polypeptides include soluble fragments. Soluble B7-DC fragments are fragments of B7-DC that may be shed, secreted or otherwise extracted from the producing cells.
  • variant B7-DC polypeptide fragments include the entire extracellular domain of B7-DC.
  • the extracellular domain of B7-DC includes amino acids from about 20 to about amino acid 221 of murine or primate, preferably human, B7-DC.
  • variant B7-DC polypeptide fragments include the IgC and IgV domains of B7-DC.
  • variant B7-DC polypeptide fragments include the IgV domain of B7-DC.
  • variant B7-DC polypeptide fragments contain a region of the polypeptide that is important for binding affinity for PD-1. These polypeptide fragments are useful to bind to and block the PD-1 receptor to prevent native ligands from binding to PD-1 receptor, thereby enhancing an immune response. Inhibiting interactions of native B7-H1 or B7-DC with PD-1 inhibits the suppression of immune responses that would otherwise occur.
  • a polypeptide fragment of mouse or primate, preferably human, B7-DC that binds to PD-1 contains, by way of non-limiting example, amino acids 101-105, or 111-113.
  • the binding of B7-H1 to PD-1 receptor typically is inhibited by at least 50 percent, or by at least 60 percent, or by at least 70 percent, or by at least 75 percent, or by at least 80 percent, or by at least 90 percent, or by at least 95 percent, or more compared to the level of binding of B7-H1 to PD-1 in the absence of the fragment.
  • Human PD-1 mutant A99L binds B7-DC and B7-H1 with higher affinity than unmutated human PD-1 (Lazar Molnar et al PNAS 105 p. 10483-10488
  • the compound acting to reduce inhibitory signal transduction is a soluble protein, such as the ECD of PD-1 incorporating this mutation.
  • Polypeptides useful in the invention can be modified by chemical moieties found associated with polypeptides in the normal cellular environment, for example, by phosphorylation, methylation, amidation, sulfation, acylation, glycosylation, sumoylation and ubiquitylation of the polypeptide.
  • Such polypeptides may also be modified by chemical moieties that are not normally part of polypeptides in a cellular environment. Such modifications can be introduced into the molecule by reacting targeted amino acid residues of the polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or terminal residues. Another useful modification is cyclization of the protein.
  • Such modifications also include introduction of a label capable of providing a detectable signal, either directly or indirectly, including, but not limited to, radioisotopes and fluorescent compounds.
  • Examples of chemical derivatives of the polypeptides include lysinyl and amino terminal residues derivatized with succinic or other carboxylic acid anhydrides. Derivatization with a cyclic carboxylic anhydride has the effect of reversing the charge of the lysinyl residues.
  • Other suitable reagents for derivatizing amino-containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylisourea; 2,4 pentanedione; and transaminase-catalyzed reaction with glyoxylate.
  • aspartyl and glutamyl residues can be converted to asparaginyl and glutaminyl residues by reaction with ammonia.
  • Polypeptides of the invention can also include one or more D-amino acids that are substituted for one or more L-amino acids.
  • the potentiating agent such as CTX 1 may be itself part of the compound that reduces inhibitory signal transduction, such as where the potentiating agent is chemically linked to a PD-1 antagonist of the invention.
  • Fusion polypeptides having a first fusion partner, or polypeptide portion, comprising all or a part of a PD-1 antagonist protein, a B7-DC polypeptide for example, (including variants, homologs and fragments thereof) fused (i) directly to a second polypeptide or, (ii) optionally, fused to a linker peptide sequence that is fused to the second polypeptide are also provided.
  • the presence of the fusion partner can alter, for example, the solubility, affinity and/or valency of the PD-1 antagonist polypeptide.
  • the disclosed fusion proteins include any combination of amino acid alteration (i.e., substitution, deletion or insertion), fragment, and/or modification of a PD-1 antagonist polypeptide as described above.
  • B7-DC fusion proteins include the extracellular domain of a B7-DC protein as the first binding partner. In another embodiment, such B7-DC fusion proteins include the IgV and IgC domain of a B7-DC protein as the first binding partner. In another embodiment, variant B7-DC fusion proteins include the IgV domain of a B7-DC protein as the first binding partner.
  • Representative first fusion partners include primate, preferably human, or murine B7-DC polypeptide, fragments thereof, and variants thereof disclosed hereinabove.
  • Preferred fragments include the extracellular domain of B7-DC.
  • the extracellular domain can include 1-10 contiguous amino acids of a signal peptide, B7-DC transmembrane domain, or both.
  • compositions and/or products and/or methods of the invention utilize PD-1 receptor antagonist, especially polypeptides, including variants, homologs and fragments thereof, that are coupled to other polypeptides to form fusion proteins that antagonize the PD-1 receptor by binding a PD-1 ligand, such as B7-H1 , thereby inhibiting the ligand from interacting with PD-1.
  • PD-1 receptor antagonist polypeptides, or variants thereof are coupled to other polypeptides to form fusion proteins that antagonize the PD-1 receptor by binding to and blocking the PD-1 receptor and inhibit or reduce inhibitory signal transduction through PD-1.
  • the second polypeptide binding partner, or second polypeptide portion may be N-terminal or C-terminal relative to the PD-1 antagonist polypeptide.
  • the second polypeptide is C-terminal to the PD-1 antagonist polypeptide.
  • the fusion protein contemplated for use in the methods and compositions and/or products of the invention comprises at least a portion of an antibody.
  • an antibody With the advent of methods of molecular biology and recombinant technology, it is now possible to produce antibody molecules by recombinant means and thereby generate gene sequences that code for specific amino acid sequences found in the polypeptide structure of the antibodies.
  • Such antibodies can be produced by either cloning the gene sequences encoding the polypeptide chains of said antibodies or by direct synthesis of said polypeptide chains, with in vitro assembly of the synthesized chains to form active tetrameric (H 2 L 2 ) structures with affinity for specific epitopes and antigenic determinants. This has permitted the ready production of antibodies having sequences characteristic of neutralizing antibodies from different species and sources.
  • all antibodies have a similar overall 3 dimensional structure.
  • This structure is often given as H 2 L 2 and refers to the fact that antibodies commonly comprise 2 light (L) amino acid chains and 2 heavy (H) amino acid chains. Both chains have regions capable of interacting with a structurally complementary antigenic target. The regions interacting with the target are referred to as "variable” or "V” regions and are characterized by differences in amino acid sequence from antibodies of different antigenic specificity.
  • the PD-1 receptor antagonist polypeptides include fragments, mutants and other variants, have a first fusion partner having all or a part of a B7-DC protein or variant thereof fused (i) directly to a second polypeptide or, (ii) optionally, fused to a linker peptide sequence that is fused to the second polypeptide.
  • the presence of the fusion partner can alter the solubility, affinity and/or valency of the B7-DC polypeptide.
  • B7-DC polypeptides are fused to one or more domains of an Ig heavy chain constant region, more preferably an amino acid sequence corresponding to the hinge, C H 2 and C H 3 regions of a human immunoglobulin C ⁇ 1 chain or to the hinge, C H 2 and C H 3 regions of a murine immunoglobulin C ⁇ 2a chain.
  • the constant region preferably includes a mutation (for example N297Q) to eliminate or reduce Fc receptor binding.
  • the hinge, CH2 and C H 3 regions of a human immunoglobulin C ⁇ 1 chain has the following amino acid sequence:
  • the hinge, CH2 and C H 3 regions of a murine immunoglobulin C ⁇ 2a chain has the following amino acid sequence:
  • Exemplary murine B7-DC fusion proteins contain amino acids 20-221 of murine B7-DC fused to amino acids 237-469 of murine lgG2a (CAA49868).
  • Human B7-DC fusion proteins can contain amino acids 20-221 of human B7-DC fused to amino acids 245-476 of human IgGI (AAA02914).
  • the signal peptides for B7-DC fusion proteins can be the endogenous signal peptides or any other signal peptide that facilitates secretion of the fusion protein from a host.
  • a representative murine B7-DC-lg fusion protein is encoded by the nucleic acid sequence of SEQ ID NO:8.
  • nucleic acid sequences can be codon-optimized to increase levels of expression for synthesizing the fusion proteins useful in the methods and compositions of the present invention.
  • Methods for codon optimization are known in the art.
  • the murine B7-DC-lg fusion protein encoded by SEQ ID NO:8 has the following amino acid sequence:
  • SEQ ID NO: 10 provides the amino acid sequence for murine B7-DC-lg fusion protein without the signal sequence.
  • human B7-DC-lg is encoded by the nucleic acid sequence of SEQ ID NO:11 , encoding the amino acid sequence for human B7-DC-lg:
  • the present invention specifically contemplates embodiments where the mature fusion protein useful in the methods and compositions of the invention have the signal sequence removed.
  • the signal sequence is completely removed.
  • SEQ ID NO:13 provides the amino acid sequence for human B7-DC-lg without the signal sequence.
  • the present invention specifically contemplates embodiments where the disclosed B7-DC-lg fusion proteins used in the methods and compositions disclosed herein have at least about 80%, 85%, 90%, 99% or 100% sequence identity to SEQ ID NO: 9, 10, 12, or 13.
  • the fusion polypeptide may have bi-specific function whereby the first fusion partner binds to a ligand of
  • PD-1 such as B7-H1
  • the second fusion partner binds to the PD-1 receptor without triggering inhibitory signal transduction through the PD-1 receptor.
  • a polypeptide useful in the invention may be monomeric or dimeric
  • the fusion proteins themselves may be present in a monomeric or an oligomeric form, preferably as a dimer.
  • the fusion proteins useful as PD-1 antagonists in the methods and compositions of the invention may assemble spontaneously into oligomeric, especially dimeric, forms or may be chemically linked to form such oligomers by means well known in the art.
  • a fusion protein useful in practicing the invention may itself comprise a portion of a B7-DC polypeptide fused to a portion of an antibody and these may be further assembled into a dimer.
  • a polypeptide for use in the invention is fused as a single amino acid chain to the Fc region of an antibody (such as where this construct is expressed from a single recombinant polynucleotide), after which two such fusion products are linked to each other to form a homodimer, such as by a disulfide linkage between the respective Fc regions.
  • Such dimeric products may be homodimers (where both monomeric fusion proteins are identical) or may be heterodimers (where two different fusion proteins are linked to each other).
  • the individual monomers of such dimers may be linked by any means known in the art, such as by covalent linkage (e.g., a disulfide bond) or by non-covalent linkage (such as an ionic interaction).
  • covalent linkage e.g., a disulfide bond
  • non-covalent linkage such as an ionic interaction
  • the B7-DC-lg used in the examples of the invention were present in the form of a homodimer having 2 copies of SEQ ID NO: 10 linked together by a disulfide linkage.
  • heterodimers of the invention include bispecific proteins and fusion proteins wherein one monomeric portion binds to PD-1 and the other binds to a natural ligand of PD-1.
  • Such heterodimers are formed by coupling of polypeptides and fusion proteins fully described elsewhere herein.
  • the PD-1 antagonist is a heterodimer, such as where two fusion proteins are linked together but they are not of identical amino acid sequence.
  • each monomer may comprise an Fc portion of an antibody linked to an active fragment of a B7-DC polypeptide where these active fragments are from different portions of the B7-DC polypeptide or where a fusion protein comprising an Fc portion of an antibody fused to a full length native B7-DC polypeptide is linked (for example, cross-linked) to a fusion protein comprising an Fc portion of an antibody and an active fragment of a full length native BY- DC polypeptide.
  • the portion of the antibody used in forming each monomeric fusion protein may be different between the two monomeric units. Any such dimeric combination is specifically contemplated by the methods and compositions of the invention.
  • the dimer results from the covalent bonding of Cys residue in the CH regions of two of the Ig heavy chains that are the same Cys residues that are disulfide linked in dimerized normal Ig heavy chains.
  • Still another embodiment provides a tetramer construct having a BirA substrate fused to the extracellular domain of a variant B7-DC polypeptide.
  • Methods for making tetramer constructs are known in the art (see Pertovas, et al., J. Exp. Med., 203:2281 (2006)). 7. Anti-PD-1 and Other Antibodies
  • PD-1 antagonists contemplated by the methods of this invention include antibodies that bind to PD-1 or ligands of PD-1 , and other antibodies.
  • the present invention relates to a method of increasing a
  • T cell response in a mammal in need thereof comprising administering to said mammal an effective treatment regimen comprising an anti-PD-1 antibody and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal to said antigen.
  • Anti-PD-1 antibodies useful in the treatment regimens(s) of the invention include, but are not limited to, those described in the following publications:
  • PCT/JP03/08420 (Honjo et al., WO/2004/004771)
  • PCT/JP04/00549 (Honjo et al., WO/2004/072286)
  • an anti-PD-1 antibody useful in the methods of the invention is MDX-1106 (see Kosak, US 20070166281 (pub. 19 July 2007) at par. 42), a human anti-PD-1 antibody, preferably administered at a dose of 3 mg/kg.
  • the present invention relates to a method of increasing a T cell response in a mammal in need thereof, comprising administering to said mammal an effective treatment regimen comprising an anti-PD-1 ligand antibody, an anti-B7-H1 antibody for example, and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal to said antigen.
  • Anti-B7-H1 antibodies useful in the treatment regimens(s) of the invention include, but are not limited to, those described in the following publications:
  • PCT/US06/022423 (WO/2006/133396, pub. 14 December 2006)
  • PCT/US07/088851 (WO/2008/083174, pub. 10 July 2008)
  • US 2006/0110383 (pub. 25 May 2006)
  • an anti-B7-H1 antibody useful in the methods of the invention is MDX-1105 (WO/2007/005874, published 11 January 2007)), a human anti-B7-H1 antibody.
  • Another embodiment of the invention includes a bi-specific antibody that comprises an antibody that binds to the PD-1 receptor bridged to an antibody that binds to a ligand of PD-1 , such as B7-H1.
  • the PD-1 binding portion reduces or inhibits signal transduction through the PD-1 receptor.
  • the antibody for use in the invention need not be an anti-PD-1 or anti- PD-1 ligand antibody but may be another antibody useful in mediating the effects of T cells in an immune response.
  • the present invention relates to a method of increasing a T cell response to an antigen in a mammal in need thereof, comprising administering to said mammal an effective treatment regimen comprising an anti-CTLA4 antibody and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal to said antigen.
  • An example of an anti-CTLA4 antibody contemplated for use in the methods of the invention includes an antibody as described in PCT/US2006/043690 (Fischkoff et al., WO/2007/056539).
  • an anti-CTLA4 antibody useful in the methods of the invention are Ipilimumab, also known as MDX-010 or MDX-101 , a human anti- CTLA4 antibody, preferably administered at a dose of 10 mg/kg, and Tremelimumab a human anti-CTLA4 antibody, preferably administered at a dose of 15 mg/kg.
  • the PD-1 receptor antagonists can also be small molecule antagonists.
  • small molecule refers to small organic compounds having a molecular weight of more than 100 and less than about 2,500 daltons, preferably between 100 and 2000, more preferably between about 100 and about 1250, more preferably between about 100 and about 1000, more preferably between about 100 and about 750, more preferably between about 200 and about 500 daltons.
  • the small molecules often include cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more functional groups.
  • the small molecule antagonists reduce or interfere with PD-1 receptor signal transduction by binding to ligands of PD-1 such as B7-H1 and B7-DC and preventing the ligand from interacting with PD-1 or by binding directly to and blocking the PD- 1 receptor without triggering signal transduction through the PD-1 receptor.
  • such a small molecule may be administered in combination with another PD-1 antagonist or CTLA4 antagonist, such as an antibody specific for PD-1 or one of its ligands or an antibody specific for
  • CTLA4 or one of its ligands may be administered as compounds in one or more of the methods of the invention or may be administered in combination with other compounds useful in the methods of the invention.
  • small molecules may be administered as compounds in one or more of the methods of the invention or may be administered in combination with other compounds useful in the methods of the invention.
  • a series of small organic compounds have been shown to bind to the B7-1 ligand to prevent binding to CTLA4 (see Erbe et al., J. Biol. Chem., Vol. 277, pp. 7363-7368 (2002).
  • Such small organics could be administered alone or together with an anti-CTLA4 antibody, in combination with CTX administration, to reduce inhibitory signal transduction of T cells.
  • PD-1 antagonists or CTLA4 antagonists contemplated for use in the methods of the invention include anti-sense nucleic acids, both DNA and RNA, as well as siRNA molecules.
  • anti- sense molecules prevent expression of PD-1 on T cells as well as production of T cell ligands, such as B7-H1 , PD-L1 and PD-L2.
  • siRNA for example, of about 21 nucleotides in length, which is specific for the gene encoding PD-1 , or encoding a PD-1 ligand, and which oligonucleotides can be readily purchased commercially
  • carriers such as polyethyleneimine (see Cubillos-Ruiz et al., J. CHn.
  • Invest. 119(8): 2231-2244 (2009), are readily taken up by cells that express PD-1 as well as ligands of PD-1 and reduce expression of these receptors and ligands to achieve a decrease in inhibitory signal transduction in T cells, thereby activating T cells.
  • the activity of the PD-1 antagonist is increased, preferably synergistically, by the presence of a potentiating agent.
  • the potentiating agent acts to increase the efficacy of the PD-1 receptor antagonist, possibly by more than one mechanism, although the precise mechanism of action is not essential to the broad practice of the present invention.
  • the potentiating agent is cyclophosphamide.
  • Cyclophosphamide (CTX, Cytoxan ® , or Neosar ® ) is an oxazahosphorine drug and analogs include ifosfamide (IFO, Ifex), perfosfamide, trophosphamide (trofosfamide; Ixoten), and pharmaceutically acceptable salts, solvates, prodrugs and metabolites thereof (US patent application 20070202077 which is incorporated in its entirety), lfosfamide (MITOXANA ® ) is a structural analog of cyclophosphamide and its mechanism of action is considered to be identical or substantially similar to that of cyclophosphamide.
  • Perfosfamide (4-hydroperoxycyclophosphamide) and trophosphamide are also alkylating agents, which are structurally related to cyclophosphamide. For example, perfosfamide alkylates DNA, thereby inhibiting DNA replication and RNA and protein synthesis.
  • New oxazaphosphorines derivatives have been designed and evaluated with an attempt to improve the selectivity and response with reduced host toxicity (Liang J, Huang M, Duan W, Yu XQ, Zhou S. Design of new oxazaphosphorine anticancer drugs. Curr Pharm Des. 2007;13(9):963-78. Review).
  • Mafosfamide is an oxazaphosphorine analog that is a chemically stable 4-thioethane sulfonic acid salt of 4-hydroxy-CPA.
  • Glufosfamide is IFO derivative in which the isophosphoramide mustard, the alkylating metabolite of IFO, is glycosidically linked to a beta-D-glucose molecule. Additional cyclophosphamide analogs are described in US patent 5,190,929 entitled “Cyclophosphamide analogs useful as anti-tumor agents” which is incorporated herein by reference in its entirety.
  • the potentiating agent is an agent that reduces activity and/or number of regulatory T lymphocytes (T-regs), preferably Sunitinib (SUTENT ® ), anti-TGF ⁇ or lmatinib (GLEEVAC ® ).
  • T-regs regulatory T lymphocytes
  • SUTENT ® Sunitinib
  • anti-TGF ⁇ anti-TGF ⁇
  • GLEEVAC ® lmatinib
  • the recited treatment regimen may also include administering an adjuvant.
  • Useful potentiating agents also include mitosis inhibitors, such as paclitaxol, aromatase inhibitors (e.g. Letrozole) and angiogenesis inhibitors
  • VEGF inhibitors e.g. Avastin, VEGF-Trap
  • Vascular endothelial growth factor blockade reduces intratumoral regulatory T cells and enhances the efficacy of a GM-CSF-secreting cancer immunotherapy. Clin Cancer Res. 2006 Nov 15;12(22):6808-16.), anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
  • the invention relates to a therapeutic composition, comprising a molecule that prevents inhibitory signal transduction through PD- 1 , or a CTLA4 antagonist, and a potentiating agent in a pharmaceutically acceptable carrier.
  • the components of said composition are present in an amount effective to increase a T cell response in a mammal.
  • the potentiating agent is cyclophosphamide or an analog of cyclophosphamide, examples of such analogs having been recited above.
  • the potentiating agent is an agent that reduces activity of regulatory T lymphocytes (T-regs), preferably where the activity is reduced due to a decrease in the number of said T-regs.
  • the agent is Sunitinib (SUTENT ® ), anti- TGF ⁇ or lmatinib (GLEEVAC ® ).
  • the potentiating agent useful in formulating compositions of the invention also include mitosis inhibitors, such as paclitaxol, aromatase inhibitors (e.g. Letrozole), agniogenesis inhibitors (VEGF inhibitors e.g. Avastin, VEGF-Trap), anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
  • mitosis inhibitors such as paclitaxol, aromatase inhibitors (e.g. Letrozole), agniogenesis inhibitors (VEGF inhibitors e.g. Avastin, VEGF-Trap), anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
  • mitosis inhibitors such as paclitaxol, aromatase inhibitors (e.g. Letrozole), agniogenesis inhibitors
  • a therapeutic composition of the invention also optionally comprises at least one additional agent that may be one or more of an anti-PD-1 antibody, an anti-CTLA4 antibody, a mitosis inhibitor, an aromatase inhibitor, an A2a adenosine receptor (A2AR) antagonist, or an angiogenesis inhibitor. Any of the therapeutic compositions of the invention may also contain one or more adjuvants as described herein.
  • a PD-1 antagonist useful as a component of a therapeutic composition of the invention includes any of the PD-1 antagonists recited herein for use in any of the methods of the invention.
  • such PD-1 antagonist includes any of the fusion proteins recited herein.
  • Such antagonist can also be any of the polypeptides or PD-1 binding fragments recited herein for use as the first polypeptide portion of any of the fusion proteins described for use in any of the methods of the invention.
  • Such antagonist can further be an antibody, such as any of the known anti-PD-1 , -B7-DC or -B7-H1 antibodies mentioned herein.
  • a therapeutic composition of the invention also includes, in addition to or in place of the aforementioned PD-1 antagonist, an anti-CTLA4 antibody.
  • an anti-CTLA4 antibody Such a composition would therefore contain such an anti-CTLA4 antibody and a potentiating agent of the kind already described herein.
  • a therapeutic composition of the invention finds use in any of the methods of the invention disclosed herein. Such composition, while intended for use as an active treatment of a disease condition, may also find use as prophylactic compositions to prevent any of the diseases recited herein.
  • the present invention contemplates a therapeuitc composition
  • a therapeuitc composition comprising a PD-1 antagonist and a potentiating agent in a pharmaceutically acceptable carrier, wherein the PD-1 antagonist and the potentiating agent are together present in an amount effective to increase a T cell response in a mammal.
  • compositions within the scope of the invention include compositions comprising any and all combinations of the PD-1 antagonists and/or antibodies disclosed herein with any of the recited potentiating agents.
  • a therapeutic composition of the invention includes a composition comprising an effective amount of one or more PD-1 antagonists, such as a combination of any or all of the full length polypeptides enumerated herein as specific SEQ ID NOs.
  • the composition comprises at least one PD-1 anatgonist and/or antibody mediating T cell activity and at least one potentiating agent.
  • compositions of the invention may also include additional active agents.
  • the pharmaceutical or therapeutic composition further comprises at least one additional agent selected from the group consisting of an anti-PD-1 antibody, an anti-CTLA4 antibody, a mitosis inhibitor, such as paclitaxel, an aromatase inhibitor, such as letrozole, an A2AR antagonist, an angiogenesis inhibitor, anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
  • the PD-1 antagonist and/or potentiating agent may be administered by any suitable means.
  • the PD-1 antagonist and/or potentiating agent is administered in an aqueous solution, by parenteral injection.
  • the formulation may also be in the form of a suspension or emulsion.
  • pharmaceutical compositions are provided including effective amounts of a peptide or polypeptide, and optionally include pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers.
  • compositions include diluents sterile water, buffered saline of various buffer content (e.g., Tris-HCI, acetate, phosphate), pH and ionic strength; and optionally, additives such as detergents and solubilizing agents (e.g., TWEEN 20, TWEEN 80, Polysorbate 80), anti- oxidants (e.g., ascorbic acid, sodium metabisulfite), and preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol).
  • buffered saline of various buffer content e.g., Tris-HCI, acetate, phosphate
  • pH and ionic strength e.g., Tris-HCI, acetate, phosphate
  • additives e.g., Tris-HCI, acetate, phosphate
  • additives e.g., TWEEN 20, TWEEN 80,
  • non-aqueous solvents or vehicles examples include propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate.
  • the formulations may be lyophilized and redissolved/resuspended immediately before use.
  • the formulation may be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions.
  • compositions of the invention may be administered by parenteral (intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection), transdermal (either passively or using iontophoresis or electroporation), or transmucosal (nasal, vaginal, rectal, or sublingual) routes of administration.
  • parenteral intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection
  • transdermal either passively or using iontophoresis or electroporation
  • transmucosal nasal, vaginal, rectal, or sublingual routes of administration.
  • the methods of the invention do not preclude administering the PD-1 antagonist and the potentiating agent by separate and different routes (e.g. topically).
  • the PD-1 antagonist and the potentiating agent may be administered at the same time, or at different times, with the potentiating agent being administered before or after the PD-1 antagonist.
  • a potentiating agent is administered both before and after the PD-1 antagonist.
  • the same potentiating agent is administered before and after the PD-1 antagonist.
  • the potentiating agent administered before the PD-1 antagonist is administered before the PD-1 antagonist.
  • the term "effective amount” or “therapeutically effective amount” means a dosage sufficient to treat, inhibit, or alleviate one or more symptoms of the disorder being treated or to otherwise provide a desired pharmacologic and/or physiologic effect.
  • the precise dosage will vary according to a variety of factors such as subject-dependent variables (e.g., age, immune system health, etc.), the disease, and the treatment being effected.
  • Therapeutically effective amounts of PD-1 receptor antagonists and/or antibodies together with a potentiating agents cause an immune response to be activated or sustained.
  • the selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment desired. Generally dosage levels of 0.001 to 50 mg/kg of body weight daily are administered to mammals. Preferrably, said dose is 1 to 50 mg/kg, more preferably 1 to 40 mg/kg, or even 1 to 30 mg/kg, with a dose of 2 to 20 mg/kg being also a preferred dose. Examples of other dosages include 2 to 15 mg/kg, or 2 to 10 mg/kg or even 3 to 5 mg/kg, with a dose of about 4 mg/kg being a specific example.
  • dosages are commonly in the range of 0.1 to 100 mg/kg, with shorter ranges of 1 to 50 mg/kg preferred and ranges of 10 to 20 mg/kg being more preferred.
  • An appropriate dose for a human subject is between 5 and 15 mg/kg, with 10 mg/kg of antibody (for example, human anti-PD-1 antibody, like MDX-1106) most preferred (plus a suitable dose of cyclophosphamide or other potentiating agent given up to about 24 hours before the antibody).
  • dosage forms based on body weight for any of the signal transduction antagonists useful in the methods of the invention include doses in the range of 5-300 mg/kg, or 5-290 mg/kg, or 5- 280 mg/kg, or 5-270 mg/kg, or 5-260 mg/kg, or 5-250 mg/kg, or 5-240 mg/kg, or 5-230 mg/kg, or 5-220 mg/kg, or 5-210 mg/kg, or 20 to 180 mg/kg, or 30 to 170 mg/kg, or 40 to 160 mg/kg, or 50 to 150 mg/kg, or 60 to 140 mg/kg, or 70 to 130 mg/kg, or 80 to 120 mg/kg, or 90 to 110 mg/kg, or 95 to 105 mg/kg, with doses of 3 mg/kg, 5 mg/kg, 7 mg/kg, 10 mg/kg, 15 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 50 mg/kg and 100 mg/kg being specific examples of preferred doses.
  • doses may, of course, be repeated.
  • the dose will, of course, be correlated with the identity of the mammal receiving said dose.
  • Doses in the above-recited mg/kg ranges are convenient for mammals, including rodents, such as mice and rats, and primates, especially humans, with doses of about 5 mg/kg, about 10 mg/kg and about 15 mg/kg being especially preferred for treating humans.
  • the potentiating agent for example cyclophosphamide
  • the potentiating agent is administered in nontoxic doses that vary depending on the animal.
  • the potentiating agent is administered by any suitable means of administration, including parenteral or oral, the former including system administration, such as intravenous.
  • a potentiating agent like cyclophosphamide is normally administered orally.
  • Such administration may be at any convenient dosage, depending on the potentiating agent.
  • the dosage in each case may be based on body weight or may be administered as a unit dosage.
  • CTX While CTX itself is nontoxic, some of its metabolites are cytotoxic alkylating agents that induce DNA crosslinking and, at higher doses, strand breaks. Many cells are resistant to CTX because they express high levels of the detoxifying enzyme aldehyde dehydrogenase (ALDH). CTX targets proliferating lymphocytes, as lymphocytes (but not hematopoietic stem cells) express only low levels of ALDH, and cycling cells are most sensitive to DNA alkylation agents.
  • ALDH aldehyde dehydrogenase
  • CTX ⁇ 200 mg/kg
  • These low doses are sub-therapeutic and do not have a direct anti-tumor activity.
  • high doses of CTX inhibit the anti-tumor response.
  • CTX Several mechanisms may explain the role of CTX in potentiation of anti-tumor immune response: (a) depletion of CD4+CD25+FoxP3+ Treg (and specifically proliferating Treg, which may be especially suppressive), (b) depletion of B lymphocytes; (c) induction of nitric oxide (NO), resulting in suppression of tumor cell growth; (d) mobilization and expansion of CD11b+Gr-1+ MDSC. These primary effects have numerous secondary effects; for example following Treg depletion macrophages produce more IFN- ⁇ and less IL-10. CTX has also been shown to induce type I IFN expression and promote homeostatic proliferation of lymphocytes.
  • Treg depletion is most often cited as the mechanism by which CTX potentiates the anti-tumor immune response. This conclusion is based in part by the results of adoptive transfer experiments. In the AB1-HA tumor model,
  • CTX treatment at Day 9 gives a 75% cure rate.
  • Transfer of purified Treg at Day 9 gives a 75% cure rate.
  • CTX is a safe, well-tolerated, and effective agent for promoting anti-tumor immune responses
  • the optimal dose for CTX to potentiate an anti-tumor immune response is one that lowers overall T cell counts by lowering Treg levels below the normal range but is subtherapeutic (see Machiels et al. Cancer Res. 61 :3689-3697 (2001)).
  • 300 mg/m 2 In human clinical trials where CTX has been used as an immunopotentiating agent, a dose of 300 mg/m 2 has usually been used. For an average male (6 ft, 170 pound (78 kg) with a body surface area of 1.98 m 2 ), 300 mg/m 2 is 8 mg/kg, or 624 mg of total protein. In mouse models of cancer, efficacy has been seen at doses ranging from 15 - 150 mg/kg, which relates to 0.45 - 4.5 mg of total protein in a 3Og mouse (Machiels et al. Cancer Res. 61 :3689-3697 (2001), Hengst et al Cancer Res. 41 :2163-2167 (1981), Hengst Cancer Res. 40:2135-2141 (1980)).
  • mg/m 2 doses may be used but unit doses administered over a finite time interval may be preferred.
  • unit doses may be administered on a daily basis for a finite time period, such as up to 3 days, or up to 5 days, or up to 7 days, or up to 10 days, or up to 15 days or up to 20 days or up to 25 days, are all specifically contemplated by the invention.
  • the same regimen may be applied for the other potentiating agents recited herein.
  • administrations may occur before or after administration of a PD-1 binding molecule of the invention.
  • administration of one or more doses of a PD-1 binding molecule of the invention may be temprally staggered with the administration of potentiating agent to form a uniform or non-uniform course of treatment whereby one or more doses of potentiating agent are administered, followed by one or more doses of a PD-1 binding compound, followed by one or more doses of potentiating agent, all according to whatever schedule is selected or desired by the researcher or clinician administering said agents.
  • the treatment regimen includes multiple administrations of one or more PD-1 antagonists.
  • such multiple administrations of PD-1 antagonists are in conjunction with multiple administrations of the same or different potentiating agents.
  • the potentiating agent is administered at least 1 , 2, 3, 5, 10, 15, 20, 24 or 30 hours prior to or after administering of the PD-1 -antagonist.
  • compositions useful herein also contain a pharmaceutically acceptable carrier, including any suitable diluent or excipient, which includes any pharmaceutical agent that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity.
  • Pharmaceutically acceptable carriers include, but are not limited to, liquids such as water, saline, glycerol and ethanol, and the like, including carriers useful in forming sprays for nasal and other respiratory tract delivery or for delivery to the ophthalmic system.
  • Vaccine compositions may further incorporate additional substances to stabilize pH, or to function as adjuvants, wetting agents, or emulsifying agents, which can serve to improve the effectiveness of the vaccine.
  • Vaccines are generally formulated for parenteral administration and are injected either subcutaneously or intramuscularly. Such vaccines can also be formulated as suppositories or for oral administration, using methods known in the art, or for administration through nasal or respiratory routes.
  • Isolated PD-1 antagonist polypeptides including variants, homologs and fragments thereof, either wild-type or mutated, and fusion proteins comprising any of these, all contemplated for use in the invention, can be obtained by, for example, chemical synthesis or by recombinant production in a host cell.
  • a nucleic acid containing a nucleotide sequence encoding the polypeptide can be used to transform, transduce, or transfect a bacterial or eukaryotic host cell (e.g., an insect, yeast, or mammalian cell).
  • nucleotide sequences can be codon-optimized to increase levels of protein expression in a particular kind of host cell. Methods for codon optimization are well known in the art.
  • nucleic acid constructs include a regulatory sequence operably linked to a nucleotide sequence encoding a costimulatory polypeptide. Regulatory sequences (also referred to herein as expression control sequences) typically do not encode a gene product, but instead affect the expression of the nucleic acid sequences to which they are operably linked.
  • the signal peptides used to secrete proteins from a cell can be the endogenous signal peptides or any other signal peptide that facilitates secretion of the fusion protein from a host.
  • Diseases to be treated or prevented by administering a therapeutic combination provided by the present invention include a malignant tumor or a chronic infectious disease caused by a bacterium, virus, protozoan, helminth, or other microbial pathogen that enters intracellular ⁇ . Such diseases are often combatted through attack by cytotoxic T lymphocytes. Because the present invention provides combination therapes useful in enhancing T cell responses, through increased T cell activity, increased T cell proliferation and reduced T cell inhibitory signals, the combination therapies of the invention have unique advantage in treating (or even preventing) such diseases. In one embodiment, because viral infections are cleared primarily by T- cells, an increase in T-cell activity is therapeutically useful in enhancing clearance of an infective viral agent from an animal or primate, preferably human, subject.
  • the disclosed compounds of the invention with PD-1 receptor antagonist activity, together with a potentiating agent work in combination for the treatment of local or systemic viral infections.
  • Infections that are to be treated by the compounds of the invention include, but are not limited to, immunodeficiency (e.g., HIV), papilloma (e.g., HPV), herpes (e.g., HSV), encephalitis, influenza (e.g., human influenza virus A), hepatitis (e.g. HCV, HBV), and common cold (e.g., human rhinovirus) viral infections.
  • Pharmaceutical formulations of PD-1 receptor antagonists compositions can also be administered to treat systemic viral diseases, including, but not limited to, AIDS, influenza, the common cold, or encephalitis.
  • Non-viral infections treatable by the compounds of the invention include, but are not limited to, infections cause by microoganisms including, but not limited to, Actinomyces, Anabaena, Bacillus, Bacteroides, Bdellovibrio, Bordetella, Borrelia, Campylobacter, Caulobacter, Chlamydia, Chlorobium, Chromatium, Clostridium, Corynebacterium, Cytophaga, Deinococcus, Escherichia, Francisella, Halobacterium, Heliobacter, Haemophilus, Hemophilus influenza type B (HIB), Hyphomicrobium, Legionella, Leptspirosis, Listeria, Meningococcus A, B and C, Methanobacterium, Micrococcus, Myobacterium, Mycoplasma, Myxococcus, Neisseria, Nitrobacter, Oscillatoria, Prochloron, Proteus, Pseudomonas, Phodos
  • the present invention provides methods and compositions for inducing or enhancing an immune response in host for treating cancer.
  • the types of cancer that may be treated with the provided compositions and methods include, but are not limited to, the following: bladder, brain, breast, cervical, colo-rectal, esophageal, kidney, liver, lung, nasopharangeal, pancreatic, prostate, skin, stomach, uterine, ovarian testicular, and hematologic cancer.
  • Malignant tumors which may be treated are classified herein according to the embryonic origin of the tissue from which the tumor is derived.
  • Carcinomas are tumors arising from endodermal or ectodermal tissues such as skin or the epithelial lining of internal organs and glands.
  • Sarcomas which arise less frequently, are derived from mesodermal connective tissues such as bone, fat, and cartilage.
  • the leukemias and lymphomas are malignant tumors of hematopoietic cells of the bone marrow. Leukemias proliferate as single cells, whereas lymphomas tend to grow as tumor masses. Malignant tumors may show up at numerous organs or tissues of the body to establish a cancer.
  • the murine analog of B7-DC-lg (in which the mouse B7-DC ECD, which shares 72% sequence identity with the human protein, is fused to the Fc domain of mouse lgG 2a ) tested in syngeneic mouse tumor models for colon cancer, mastocytoma, and other tumor types incorporating a cyclophosphamide (CTX) pre-treatment as described herein.
  • CTX cyclophosphamide
  • CTX which acts as an immunopotentiating agent
  • murine B7-DC- IG eradicates established CT26 colon carcinoma tumors in up to 80% of the animals. Further, in CT26 colon carcinoma tumor re-challenge studies, no tumor re-growth was detected in mice that had previously eradicated tumor following CTX + murine B7-DC-lg treatment. These mice were also shown to have an increased tumor-specific CTL population relative to na ⁇ ve mice.
  • the present invention contemplates use of a compound that reduces inhibitory signal transduction in a T cell, as described elsewhere herein, in the manufacture of a medicament for increasing a T cell response by combination therapy wherein said compound is administered in conjunction with a potentiating agent.
  • the compound that reduces inhibitory signal transduction in a T cell and said potentiating agent are provided as separate medicaments for administration at different times, preferably where the potentiating agent is administered prior to the compound that reduces inhibitory signal transduction, for example, up to 24 hours prior to the inhibitory compound (or other time intervals recited herein).
  • the compound and potentiating agent are for use in the treatment of an infectious disease or cancer, including diseases caused by any of the infectious agents or cancers recited elsewhere herein.
  • a compound useful in these methods is a recombinant protein composed of the ECD of human B7-DC fused to the Fc domain of human IgGi, referred to herein as B7-DC-lg.
  • the present invention relates to a medical kit for administering a compound that reduces inhibitory signal transduction in a T cell, as disclosed herein, in combination with a potentiating agent, said kit comprising:
  • Vaccines require strong T cell response to eliminate cancer cells and infected cells or infectious agents.
  • PD-1 receptor antagonists described herein can be administered as a component of a vaccine, along with a potentiating agent, to provide a costimulatory signal to T cells.
  • Vaccines disclosed herein include antigens, a PD-1 receptor antagonist and optionally adjuvants and targeting molecules.
  • the antigens against which the T cell response is enhanced by the methods and composition of the invention includes peptides, proteins, polysaccharides, saccharides, lipids, nucleic acids, or combinations thereof.
  • the antigens, in the case of disease, are present due to the disease process.
  • the disclosed PD-1 receptor antagonists compositions may be administered in conjunction with prophylactic vaccines, which confer resistance in a subject to subsequent exposure to infectious agents, or in conjunction with therapeutic vaccines, which can be used to initiate or enhance a subject's immune response to a pre-existing antigen, such as a tumor antigen in a subject with cancer, or a viral antigen in a subject infected with a virus.
  • prophylactic vaccines which confer resistance in a subject to subsequent exposure to infectious agents
  • therapeutic vaccines which can be used to initiate or enhance a subject's immune response to a pre-existing antigen, such as a tumor antigen in a subject with cancer, or a viral antigen in a subject infected with a virus.
  • the desired outcome of a prophylactic, therapeutic or de-sensitized immune response may vary according to the disease, based on principles well known in the art.
  • an immune response against an infectious agent may completely prevent colonization and replication of an infectious agent, affecting "sterile immunity" and the absence of any disease symptoms.
  • a vaccine against infectious agents may be considered effective if it reduces the number, severity or duration of symptoms; if it reduces the number of individuals in a population with symptoms; or reduces the transmission of an infectious agent.
  • immune responses against cancer, allergens or infectious agents may completely treat a disease, may alleviate symptoms, or may be one facet in an overall therapeutic intervention against a disease.
  • the stimulation of an immune response against a cancer may be coupled with surgical, chemotherapeutic, radiologic, hormonal and other immunologic approaches in order to affect treatment.
  • the methods and products of the invention do not preclude use of an adjuvant in addition to the potentiating agent.
  • adjuvant may be administered, for example, along with the PD-1 antagonist.
  • the adjuvants useful in the compositions and methods of the invention include, but are not limited to, one or more of the following: oil emulsions (e.g., Freund's adjuvant); saponin formulations; virosomes and viral-like particles; bacterial and microbial derivatives; immunostimulatory oligonucleotides; ADP-ribosylating toxins and detoxified derivatives; alum; BCG; mineral-containing compositions (e.g., mineral salts, such as aluminium salts and calcium salts, hydroxides, phosphates, sulfates, etc.); bioadhesives and/or mucoadhesives; microparticles; liposomes; polyoxyethylene ether and polyoxyethylene ester formulations; muramyl peptides
  • Useful adjuvants also include immunomodulators such as cytokines, interleukins (e.g., IL-1 , IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc.), interferons (e.g., interferon- ⁇ ), macrophage colony stimulating factor, and tumor necrosis factor.
  • immunomodulators such as cytokines, interleukins (e.g., IL-1 , IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc.), interferons (e.g., interferon- ⁇ ), macrophage colony stimulating factor, and tumor necrosis factor.
  • PD-1 receptor antagonist including any of the polypeptides, fragments, variants, homologs and fusion proteins disclosed herein, from being administered to a subject in need thereof in combination with one or more additional therapeutic agents (in addition to the potentiating agent).
  • additional therapeutic agents are selected based on the condition, disorder or disease to be treated.
  • PD-1 receptor antagonists can be co-administered with one or more additional agents that function to enhance or promote an immune response, and which are considered herein as active agents.
  • Such agents include, but are not limited to, amsacrine, bleomycin, busulfan, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clofarabine, crisantaspase, cytarabine, dacarbazine, dactinomycin, daunorubicin, docetaxel, doxorubicin, epirubicin, etoposide, fludarabine, fluorouracil, gemcitabine, hydroxycarbamide, idarubicin, ifosfamide, irinotecan, leucovorin, liposomal doxorubicin, liposomal daunorubicin, lomustine, melphalan, mercaptopurine, mesna, methotrexate, mitomycin, mitoxantrone, oxaliplatin, paclitaxel, pemetrexed, pentostatin, pro
  • the therapies provided by the methods and compositions of the present invention may also be used in conjunction with other types of therapies, such as radiation treatments, surgery, and the like.
  • the present invention recites a number specific structures useful in practicing the methods of the invention.
  • Other compounds possessing antagonist activity and being useful in the methods of the invention may also be identified by reference to well known assay procedures for identifying chemical structures that bind to PD-1 , CTLA4, and ligands of any of these and that also possess the ability to reduce inhibitory signal transduction in T cells.
  • Some such assays are binding assays useful in determining if a selected chemical structure binds to receptors; these are well known in the art and need not be discussed in detail herein (see, for example, U.S. 2008/0274490, (pub. 6 November 2008) and U.S.
  • PD-1 binding activity of human B7-DC-lg was assessed by ELISA.
  • 96-well ELISA plates were coated with 100 uL 0.75 ug/mL recombinant human PD-1/Fc (R&D Systems) diluted in BupH Carbonate/Bicarbonate pH 9.4 buffer (Pierce) for 2 hours and then blocked with BSA solution (Jackson ImmunoResearch) for 90-120 minutes.
  • Serially diluted human B7-DC-lg wild type, as well as D111 S mutein, and K113S mutants that were selected for reduced binding to PD-1
  • human IgGI isotype control were allowed to bind for 90 minutes.
  • Bound B7-DC-lg was detected using 100 uL of 0.5 ug/mL biotin conjugated anti-human B7-DC clone MIH 18 (eBioscience) followed by 1 :1000 diluted HRP-Streptavidin (BD Bioscience) and TMB substrate (BioFX). Absorbance at 450 nm was read using a plate reader (Molecular Devices) and data were analyzed in SoftMax using a 4-parameter logistic fit. The data showed that human B7-DC-lg (wildtype) bound to PD-1 but the K113S and D111 S mutants do not bind to PD-1.
  • buffers, media, reagents, cells, culture conditions and the like are not intended to be limiting, but are to be read so as to include all related materials that one of ordinary skill in the art would recognize as being of interest or value in the particular context in which that discussion is presented. For example, it is often possible to substitute one buffer system or culture medium for another and still achieve similar, if not identical, results. Those of skill in the art will have sufficient knowledge of such systems and methodologies so as to be able, without undue experimentation, to make such substitutions as will optimally serve their purposes in using the methods and procedures disclosed herein.
  • B7-DC-lg binds to PD01 expressing CHO cells
  • B7-DC-lg was first conjugated with allophycocyanin (APC) and then incubated at various concentrations with a CHO cell line constitutively expressing PD-1 or parent CHO cells that do not express PD-1. Binding was analyzed by flow cytometry. Figure 1 shows the median fluorescence intensity
  • B7-DC-lg-APC binds to CHO.
  • PD-1 cells solid circle
  • untransfected CHO cells gray triangle
  • Example 2 B7-DC-lg competes with B7-H1 for binding to PD-1.
  • B7-DC-lg at various concentrations was first incubated with a CHO cell line constitutively expressing PD-1 before adding B7-H1 -Ig-APC to the probe and cell mixture.
  • Figure 2 shows the median fluorescence intensity (MFI) of B7- HI-Fc-APC is shown as a function of the concentration of unlabeled B7-DC-lg competitor (x-axis) added.
  • MFI median fluorescence intensity
  • Mouse colorectal tumor cell line, CT26 was obtained from ATCC.
  • a master cell bank at Passage 4 was generated following ATCC guidelines. Cells were tested and confirmed no mycoplasma and other pathogen contamination.
  • One vial of tumor cells was thawed from the cryopreserved stocks and grown for two passages prior to inoculation.
  • CT26 cells were split at 1 :5 dilution with 30 ml_ complete medium (RPMI + 10% FBS, 2 mM L-GIu 1 and 1x P/S) for two days culture or at
  • CT26 cells were harvested by aspirating medium, rinsing the flask with
  • Cell number and viability of the inoculated cells were analyzed by trypan blue dye staining with proper dilution (e.g. 1 :5 dilution, 10 ⁇ L cells + 40 ⁇ L trypan blue) and confirmed by NOVA cell count during the last wash step. Cell viability generally was greater than 95% for inoculation.
  • CT26 cells were diluted to 6.7x10 5 cells/mL for initial inoculation with plain RPMI and stored on ice. Typically each mouse was inoculated with 150 ⁇ L (1 x10 5 cells). On Day 9, all the tumor-bearing mice were first grouped into a rat cage and randomly divided the mice to experimental groups.
  • CTX solution was reconstituted by 1x PBS to 4 mg/mL Mice were intraperitoneal ⁇ (IP) injected with 0.5 ml_ of CTX solution resulting in 2 mg for a 20 gram mouse, i.e. 100 mg/kg.
  • IP intraperitoneal ⁇
  • mice were IP injected with 0.5 ml_ of B7-DC-lg (0.2 mg/mL) resulting in 0.1 mg for a 20 gram mouse, i.e. 5 mg/kg. The same dose was given 2 time a week for 4 weeks, total 8 doses. Tumor growth were monitored by measuring the tumor twice weekly, starting on the day when giving B7-DC-lg via a digital caliper. Tumor volume was calculated as following:
  • Tumor volume ⁇ (D short ) 2 * (D
  • Ong )/6 -0.52 x (D shO rt) 2 * (D
  • mice were euthanized and taken off the study if the tumor volume reached 2000 mm 3 or if there were skin ulcers and infections at the tumor inoculation site.
  • Example 4 Combination of cyclophosphamide and B7-DC-lg can eradicate established tumors.
  • mice at age of 9 to 11 weeks were implanted subcutaneously with 1.0 x 10 5 CT26 colorectal tumor cells as described above. On day 10 post tumor implantation, mice received 100 mg/kg of cyclophosphamide. B7- DC-Ig treatment started 1 day later, on day 11. Mice were treated with 100 ug of B7-DC-lg, 2 doses per week, for 4 weeks and total 8 doses. 75% of the mice that received the CTX + B7-DC-lg treatment regimen eradicated the established tumors by Day 44, whereas all mice in the control CTX alone group died as a result of tumor growth or were euthanized because tumors exceeded the sizes approved by IACUC (results shown in Figure 3). These results demonstrate the effectiveness of the treatment regimen on established tumors and not mere prophylaxis.
  • Combination of cyclophosphamide and B7-DC-lg can eradicate established tumors and protect against tumor re-challenge.
  • Combination of cyclophosphamide and B7-DC-lg can generate tumor specific, memory cytotoxic T lymphocytes
  • Mice eradiated established CT26 colorectal tumors from the above described experiment were rechallenged with 2.5x10 5 CT26 cells on Day 44. Seven days later, mouse spleens were isolated.
  • Mouse splenocytes were pulsed with 5 or 50 ug/ml_ of ovalbumin (OVA) or AH 1 peptides for 6 hours in the presence of a Golgi blocker (BD BioScience).
  • Memory T effector cells were analyzed by assessing CD8 + /IFN ⁇ + T cells. Results in Figure 5 show that there were significant amount of CT26 specific T effector cells in the CT26 tumor-eradicated mice.
  • mice at age of 9 to 11 weeks were implanted subcutaneously with 1.0 x 10 5 CT26 colorectal tumor cells.
  • mice received a single dose of cyclophosphamide (100 mg/kg) and started treatment on Day 10 with 30, 100 or 300 ⁇ g of B7-DC-lg, 2 doses per week for 4 weeks, total 8 doses.
  • Figure 6 shows there were 70% of the mice eradicated the tumors at 300 ⁇ g, 40% tumor eradication with 100 ⁇ g, and 30 ⁇ g dose gave rise to 10% tumor eradication.
  • Combination of cyclophosphamide and anti-PD-1 can eradicate established tumors.
  • mice at age of 9 to 11 weeks were challenged subcutaneously with 1.0 x 10 5 CT26 colorectal tumor cells.
  • mice received a single dose of cyclophosphamide (100 mg/kg) and started treatment with anti-PD-1 antibody (250 ug, Clone G4, Hirano F. et al., 2005 Cancer Research) which was administered 3 times per week for four weeks.
  • anti-PD-1 antibody 250 ug, Clone G4, Hirano F. et al., 2005 Cancer Research
  • 70% of the mice that received the CTX + anti-PD-1 regimen eradicated established CT26 tumors at day 50 after tumor challenge, whereas all mice in the control and anti-PD-1 alone groups died as a result of tumor growth or were euthanized because tumors exceeded the sizes approved by IACUC.
  • Combination of cyclophosphamide and anti-CTLA4 can eradicate established tumors.
  • mice at age of 9 to 11 weeks were challenged subcutaneously with 1.0 x 10 5 CT26 colorectal tumor cells. On day 11 post tumor challenge, mice received 100 mg/kg of cyclophosphamide.
  • Anti-CTLA4 an anti-mouse CTLA4 from hamster hybridoma - ATCC deposit UC10-4F10-11
  • Anti-CTLA4 an anti-mouse CTLA4 from hamster hybridoma - ATCC deposit UC10-4F10-11
  • Mice were treated with 100 ug of anti- CTLA4, 2 doses per week, for 4 weeks. 56% of the mice that received the CTX + anti-CTLA4 regimen were tumor free at day 50 after tumor challenge, whereas all mice in the control group died as a result of tumor growth or were euthanized because tumors exceeded the sizes approved by IACUC. Results are shown in Figure 8. These results show the effectiveness of the treatment regimen on established tumors and not mere prophylaxis.
  • Figure 9 shows the results of experiments wherein Balb/C mice at age of 9 to 11 weeks of age were implanted with 1 X 10 5 CT26 cells subcutaneously. On Day 9, mice were injected with 100 mg/kg of CTX, IP. Twenty four hours later, on Day 10, mice were treated with 100 ug of B7-DC- Ig. There were 5 groups: na ⁇ ve mice that did not receive any tumor cells, vehicle injected, CTX alone, CTX + B7-DC-lg or B7-DC-lg alone. Two naive mice and 4 mice from other groups were removed from the study on Day 11 (2 days post CTX) and Day 16 (7 days post CTX) for T cell analysis.
  • Example 11 Combination of cyclophosphamide and B7-DC-lg can promote mouse survival in a metastatic prostate tumor model B10.D2 mice at age of 9 to 11 weeks were injected intravenously with 3.0 x 10 5 SP-1 cells, which were isolated from lung metastasis post parent TRAMP cell injection.
  • the CTX mice received 3 doses of CTX, 50 mg/kg, on Day 5, 12 and 19.
  • the B7-DC-lg treated mice received 3 doses of B7-DC-lg, 5 mg/kg, on Day 6, 13 and 20.
  • On Day 100 17% of mice in the control groups, no-treated, CTX alone, B7-DC-lg alone survived while 43% of the mice received combination of CTX and B7-DC-lg survived. Results are shown in Figure 10.
  • Combination of Listeria cancer vaccine and B7-DC-lg can enhance mouse survival post CT26 liver implantation
  • mice received 1 injection of CTX at 50 mg/kg, IP.
  • mice were treated with recombinant
  • LD 50 (1x10 7 CFU), then on Day 14 and 17. Mice were also treated with B7- DC-Ig on Day 1 1 and then on Day 18. Figire 1 1 shows mice without any treatment or treated with CTX and Listeria cancer vaccine all died before
  • Cyclophosphamide, doxorubicin, and paclitaxel enhance the antitumor immune response of granulocyte/macrophage- colony stimulating factor-secreting whole-cell vaccines in HER-2/neu tolerized mice. Cancer Res. 2001 May 1 ;61 (9):3689-97

Abstract

Methods of treating cancer and infectious diseases utilizing a treatment regimen comprising administering a compound that reduces inhibitory signal transduction in T cells, in combination with a potentiating agent, such as cyclophosphamide, to produce potent T cell mediated responses, are described. Compositions comprising the PD-1 antagonists and potentiating agents useful in the methods of the invention are also disclosed.

Description

Compositions of PD-1 Antagonists and Methods of Use
This application claims priority of U.S. Provisional Application Serial No. 61/211 ,697, filed 2 April 2009, 61/ 091 ,694, filed 25 August 2008, 61/091 ,709 filed 25 August 2008, and 61/091 ,705 filed 25 August 2008, the disclosures of which are hereby incorporated by reference in their entirety.
FIELD OF THE INVENTION
The present invention relates to therapeutic compositions containing a compound that prevents inhibitory signal transduction on T cells in combination with potentiating agents and the use of said components together or separately for the induction of T cell responses valuable in disease therapy.
BACKGROUND OF THE INVENTION
The response of T lymphocytes to disease states, such as infection and chronic diseases like cancer, is complicated and involves intercellular interactions and the production of soluble mediators (called cytokines or lymphokines). Activation of T cells normally depends on an antigen-specific signal following contact of the T cell receptor (TCR) with an antigenic peptide presented via the major histocompatibility complex (MHC) while the extent of this reaction is controlled by positive and negative antigen-independent signals eminating from a variety of co-stimulatory molecules. The latter are commonly members of the CD28/B7 family. Conversely, Programmed Death-
1 (PD-1 ) is a member of the CD28 family of receptors that delivers a negative immune response when induced on T cells. Contact between PD-1 and one of its ligands (B7-H1 or B7-DC) induces an inhibitory response that decreases T cell multiplication and/or the strength and/or duration of a T cell response.
Thus, the T lymphocyte response is regulated by various factors, including cell surface molecules that act as receptors, where the latter include both the TCR complex as well as other surface molecules.
In sum, an antigen specific T cell response is mediated by two signals: 1) engagement of the TCR with antigenic peptide presented in the context of HC (signal 1), and 2) a second antigen-independent signal delivered by contact between different receptor/ligand pairs (signal 2). This "second signal" is critical in determining the type of T cell response (activation vs tolerance) as well as the strength and duration of that response, and is regulated by both positive and negative signals from costimulatory molecules, such as the B7 family of proteins.
The most extensively characterized T cell costimulatory pathway is B7- CD28, in which B7-1 (CD80) and B7-2 (CD86) each can engage the stimulatory CD28 receptor and the inhibitory CTLA-4 (CD152) receptor. In conjunction with signaling through the T cell receptor, CD28 ligation increases antigen-specific proliferation of T cells, enhances production of cytokines, stimulates differentiation and effector function, and promotes survival of T cells (Lenshow, et al., Annu. Rev. Immunol., 14:233-258 (1996); Chambers and Allison, Curr. Opin. Immunol., 9:396-404 (1997); and Rathmell and Thompson, Annu. Rev. Immunol., 17:781-828 (1999)). In contrast, signaling through CTLA-4 is thought to deliver a negative signal that inhibits T cell proliferation, IL-2 production, and cell cycle progression (Krummel and Allison, J. Exp. Med., 183:2533-2540 (1996); and Walunas, et al., J. Exp. Med., 183:2541-2550 (1996)). Other members of the B7 family of costimulatory molecules include B7-H1 (Dong, et al., Nature Med., 5:1365-1369 (1999); and Freeman, et al., J. Exp. Med., 192:1-9 (2000)), B7-DC (Tseng, et al., J. Exp. Med., 193:839-846 (2001); and Latchman, et al., Nature Immunol., 2:261-268 (2001 )), B7-H2 (Wang, et al., Blood, 96:2808-2813 (2000); Swallow, et al., Immunity, 11 :423- 432 (1999); and Yoshinaga, et al., Nature, 402:827-832 (I999)), B7-H3 (Chapoval, et al., Nature Immunol., 2:269-274 (2001)) and B7-H4 (Choi, et al., J. Immunol., 171 :4650-4654 (2003); Sica, et al., Immunity, 18:849-861 (2003); Prasad, et al., Immunity, 18:863-873 (2003); and Zang, et al., Proc. Natl. Acad. Sci. U.S.A., 100:10388-10392 (2003)). B7-H5 (described in WO 2006/012232) is a newly discovered member of the B7 family.
B7 family molecules have a membrane proximal IgC (constant) domain and a membrane distal IgV (variable) domain. The CD28-like family of receptors for these ligands share a common extracellular IgV-like domain. Interactions of receptor-ligand pairs are mediated predominantly through residues in the IgV domains of the ligands and receptors (Schwartz, et al., Nature Immunol., 3:427-434 (2002)). In general, IgV domains are described as having two sheets that each contains a layer of β-strands (Williams and Barclay, Annu. Rev. Immunol., 6:381-405 (1988)). The front and back sheets of CTLA-4 contain strands A'GFC'C and ABEDC, respectively (Ostrov, et al., Science, 290:816-819 (2000)), whereas the front and back sheets of the B7 IgV domains are composed of strands AGFCCC" and BED, respectively (Schwartz, et al., Nature, 410:604-608 (2001); Stamper, et al., Nature, 410:608-611 (2001); and Ikemizu, et al., Immunity, 12:51-60 (2000)). Crystallographic analysis revealed that the CTLA-4/B7 binding interface is dominated by the interaction of the CDR3-analogous loop from CTLA-4, composed of a MYPPPY motif, with a surface on B7 formed predominately by the G, F, C, C and C" strands (Schwartz, et al., Nature, 410:604-608 (2001 ); and Stamper, et al., Nature, 410:608-611 (2001)). Data from amino acid homologies, mutation, and computer modeling provide support for the concept that this motif also is a major B7-binding site for CD28 (Bajorath, et al., J. MoI. Graph. Model., 15:135-139 (1997)). Although the MYPPPY motif is not conserved in ICOS, the receptor for B7-H2, studies have indicated that a related motif having the sequence FDPPPF and located at the analogous position is a major determinant for binding of ICOS to B7-H2 (Wand, et al., J. Exp. Med., 195:1033-1041 (2002)). B7-DC (also called PD-L2 or CD273) is a relatively new member of the
B7 family, and has an amino acid sequence that is about 34% identical to B7-
H1 (also called PD-L1). Human and mouse B7-DC orthologues share about
70% amino acid identity. While B7-H1 and B7-DC transcripts are found in various tissues (Dong, et al., Nature Med., 5:1365-1369 (1999); Latchman, et al., Nature Immunol., 2:261-268 (2001); and Tamura, Blood, 97:1809-1816
(2001)), the expression profiles of the proteins are quite distinct. B7-H1 is broadly expressed on a wide variety of tissue and cell types, while B7-DC expression is predominantly restricted to activated dendritic cells (DC) and macrophages.
It has been shown that both B7-H1 and B7-DC bind to PD-1 (Freeman, et al., J. Exp. Med., 192:1027-1034 (2000)), a distant member of the CD28 family with an immunoreceptor tyrosine-based inhibitory motif (ITIM) in its cytoplasmic domain (Ishida, et al., EMBO J., 11 :3887-3895 (1992)). PD-1 , a member of the CD28 family of receptors, is inducibly expressed on activated T cells, B cells, natural killer (NK) cells, monocytes, DC, and macrophages (Keir, et al Curr. Opin. Immunol. 19:309-314 (2007)).
The primary result of PD-1 ligation by its ligands is to inhibit signaling downstream of the T cell Receptor (TCR). Therefore, signal transduction via PD-1 usually provides a suppressive or inhibitory signal to the T cell that results in decreased T cell proliferation or other reduction in T cell activation. B7-H1 is the predominant PD-1 ligand causing inhibitory signal transduction in T cells. The present invention solves the problem of undesired T cell inhibition by providing agents that bind to PD-1 and thus prevent inhibitory signal transduction, or else bind to ligands of PD-1 such as B7-H1 , thereby preventing the ligand from binding to PD-1 to deliver an inhibitory signal. In either case, T cell responses, such as T cell proliferation or activation, are stimulated.
B7-H1 is the predominant PD-1 ligand, likely due to its broader distribution and higher expression levels. PD-1 inhibition occurs only when PD-1 and TCR are ligated in close proximity to each other, in the context of the immune synapse. PD-1 and its ligands have been the topic of several review articles.
B7-H1 is also over expressed in many cancers (including breast cancer, colon cancer, esophageal cancer, gastric cancer, glioma, leukemia, lung cancer, melanoma, multiple myeloma, ovarian cancer, pancreatic cancer, renal cell carcinoma, and urothelial cancer), and has been linked to poor prognosis. B7-H1 is expressed by many tumor cell lines, especially following stimulation with interferon gamma (IFN-γ), and is also upregulated on tumor infiltrating myeloid derived suppressor cells (MDSC). For example, PD-1 is up-regulated on tumor specific CD8 T cells and is associated with functional impairment, anergy, exhaustion, and apoptosis. PD-1 upregulation has also been associated with dysfunctional and/or suppressive phenotypes on additional cell types, such as regulatory T cells (Treg) and natural killer T (NKT) cells.
The present invention makes use of such molecular functions by providing treatment regimens for treating diseases through increased T cell activity, especially cancer and infectious diseases.
BRIEF SUMMARY OF THE INVENTION
In one aspect, the present invention relates to a method of increasing T cell responses, for example, to an antigen, in a mammal in need of such increase, comprising administering to said mammal a compound that reduces inhibitory signal transduction in immune cells, especially T cells, and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal.
Compounds useful in the treatment regimen of the invention include those that bind to and block PD-1 receptors on T cells without triggering inhibitory signal transduction, compounds that bind to PD-1 ligands to prevent their binding to PD-1 , compounds that do both and compounds that prevent expression of genes that encode either PD-1 or natural ligands of PD-1. Such compounds are referred to herein as "PD-1 antagonists." Compounds that bind to natural ligands of PD-1 include PD-1 itself, as well as active fragments of PD-1 , and in the case of the B7-H1 ligand, B7.1 proteins and fragments. Such antagonists include proteins, antibodies, anti-sense molecules and small organics. In a preferred embodiment, said T cell response is greater than that produced by either of said PD-1 antagonist or said potentiating agent when either is administered without the other.
In another embodiment, compounds useful in the methods of the invention are those that bind to T cell surface molecules such as CTLA4 to prevent the inhibitory signals triggered by binding of natural ligands thereof or that bind to said natural ligands. Such antagonists include proteins, antibodies, anti-sense molecules and small organics.
In a general embodiment, compounds useful in treatment regimens and compositions of the present invention include those that bind to PD-1 without triggering, inducing, increasing, facilitating and/or permitting co- ligation of PD-1 with TCR.
Preferred compounds that prevent inhibitory signal transduction through PD-1 and thus act as PD-1 antagonists include, but are not limited to, B7-DC polypeptides, especially soluble portions of these, including active fragments of these, variants and homologs of these, as well as fusion proteins incorporating any of the foregoing, that bind to PD-1 without triggering inhibitory signal transduction. In preferred embodiments, B7-DC comprises the amino acid sequence of SEQ ID NO: 1 , 2, 3 or 4. Preferred such compounds are those incorporating the soluble domain of B7-DC (i.e., without transmembrane sequence). Suitable fragments of B7-DC polypeptides include fragments containing the IgV and/or IgC domains or fragments containing only the IgV domain, with the latter being a preferred embodiment, with amino acids 20-121 of SEQ ID NO: 1 being a preferred example of an IgV domain.
Preferred PD-1 antagonists also include, but are not limited to, active fragments of natural ligands of PD-1 , such as B7-H1 polypeptides (disclosed in U.S. Patent No. 6,803,192, incorporated by reference herein in its entirety), especially soluble portions of these, including variants and homologs of these, as well as fusion proteins incorporating any of the foregoing, that bind to PD-1 without triggering inhibitory signal transduction.
Preferred compounds of the invention also include, but are not limited to, compounds, including active fragments, variants and homologs, that bind to natural ligands of PD-1 , such as fragments of B7-1 that bind to B7-H1 , as well as fusion proteins incorporating any of the foregoing, that bind to ligands of PD-1 to prevent the latter from binding to PD-1 to trigger inhibitory signal transduction.
In another embodiment, the compositions and methods of use thereof, include a combination of a PD-1 receptor antagonist that binds to and blocks the PD-1 receptor, and a separate PD-1 receptor antagonist that binds to and blocks PD-1 receptor ligands. Another embodiment of the present invention provides PD-1 receptor antagonists that bind to the PD-1 receptor without triggering inhibitory signal transduction through the PD-1 receptor and also have the ability to bind and antagonize PD-1 receptor ligands, such as B7-H1 , that would otherwise trigger inhibitory signal transduction through the PD-1 receptor. Other contemplated PD-1 receptor antagonists include bi-specific antibodies that can bind both the PD-1 receptor and PD-1 receptor ligands.
Preferred embodiments of compounds useful in the present invention also include antibodies that bind to PD-1 or CTLA4, thereby reducing, or abolishing, inhibitory signal transduction mediated by these sources. Preferred compounds for use in the methods of the invention also include, but are not limited to, active fragments of ligands of CTLA4 (such as B7-1 and B7-2) that bind to CTLA4 to reduce subsequent inhibitory signals yet do not bind to CD28 or otherwise inhibit positive signal transduction by CD28.
Preferred compounds that prevent inhibitory signal transduction through PD-1 and thus act as PD-1 antagonists include, but are not limited to,
B7-DC antagonists, especially soluble portions of these, including active fragments of these, variants and homologs of these, as well as fusion proteins incorporating any of the foregoing, that bind to B7-DC.
In one embodiment, B7-DC polypeptides, fragments or variants thereof are coupled to other polypeptides to form fusion proteins that antagonize the PD-1 receptor by binding to the PD-1 receptor without causing inhibitory signal transduction through PD-1 , thereby reducing, or interfering with, ligand binding to PD-1 , particularly B7-H1 binding, and thereby interfering with inhibitory signal transduction through the PD-1 receptor. Examples of such fusion proteins are polypeptides comprising the amino acid sequence of SEQ ID NO: 9, 10, 12 or 13, as well as homologs thereof. In one preferred embodiment, all or a portion of the extracellular domain (ECD) of B7-DC is part of a fusion protein wherein it is linked to a second polypeptide containing an Fc portion of an immunoglobulin. A preferred example of this is B7-DC-lg, especially where this structure is part of a homodimer wherein two B7-DC-lg molecules are linked to each other, such as by a disulfide linkage.
In specific embodiments, fragments useful in the compounds of the invention consist of at least 10, 15, 25, 50, 75, 100, 150, 200 or more contiguous amino acids of a polypeptide having the desired antagonist activity. Such fragments are also commonly part of fusion proteins for use in the invention.
In another aspect, the present invention relates to a method of increasing T cell responses in a mammal in need thereof, comprising administering to said mammal an effective treatment regimen comprising an anti-PD-1 antibody and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal.
In another aspect, the present invention relates to a method of increasing T cell responses in a mammal in need thereof, comprising administering to said mammal an effective treatment regimen comprising an immunomodulator, and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal. Such immunomodulators include molecules that antagonize other CD28 family receptors (such as CTLA4) that inhibit T cell responses. A preferred embodiment uses an anti-CTLA4 antibody and a potentiating agent. Additional immunomodulators include: molecules that agonize CD28 family receptors (such as CD28 and ICOS) that activate T cell responses; molecules that antagonize B7 family ligands (such as B7-H1 , B7-DC, B7-H4) that inhibit T cell responses; and molecules that agonize B7 family ligands (such as B7.1 and B7.2) that activate T cell responses.
In additional embodiments of any of the methods of the invention, the treatment regimen of a PD-1 antagonist compound and a potentiating agent further comprises at least one additional therapeutic agent. Additional therapeutic agents contemplated include immunomodulatory agents. Exemplary immunomodulating agents for such methods include anti-PD-1 and anti-CTLA4 antibodies.
In one embodiment, the potentiating agent is selected from cyclophosphamide and analogs of cyclophosphamide, Sunitinib (Sutent), anti- TGFβ and lmatinib (Gleevac), a mitosis inhibitor, such as paclitaxel, an aromatase inhibitor, such as letrozole, an A2a adenosine receptor (A2AR) antagonist, an angiogenesis inhibitor, anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists. Some of these agents reduce the number of Tregs (i.e., regulatory T lymphocytes or T-regs) within the tumor microenvironment. In another embodiment, the methods and/or compositions of the invention specifically contemplate use of any suitable adjuvant as part of said method and/or composition.
In accordance with the invention, T cells can be contacted with PD-1 receptor antagonist and/or compositions thereof containing a potentiating agent in vitro, ex vivo or in vivo. Contacting T cells using PD-1 receptor antagonists and/or compositions thereof containing a potentiating agent can occur before, during or after activation of the T cell.
In a specific embodiment, a molecule that prevents or reduces inhibitory signal transduction through PD-1 and the potentiating agent are administered at different times, such as where the potentiating agent is administered prior to administering the PD-1 antagonist. Such administration may be in conjunction with an additional therapeutic agent.
In specific embodiments of any of the methods of the invention, the treatment regimen includes administration of the potentiating agent at least 1 hour, or at least 2 hours, or at least 3 hours, or at least 5 hours, or at least 10 hours, or at least 15 hours, or at least 20 hours, or at least 24 hours, or at least 30 hours or even longer before administering any or all of the PD-1 antagonist, the anti-PD-1 antibody, the anti-CTLA4 antibody, and/or additional therapeutic agents. Administration of the potentiating agent may also occur after administering any or all of the PD-1 antagonist, the anti-PD-1 antibody, the anti-CTLA4 antibody and/or additional therapeutic agents, such as no more than 1 hour, 2 hours, 3 hours, 5 hours, 10 hours, 15 hours, 20 hours, 24 hours, or even up to 30 hours after administering a PD-1 antagonist, or may occur in conjunction with administering the PD-1 antagonist.
The increased T cell response achieved as a result of the methods of the invention is sufficient to treat a disease, including one or more of cancer, viral infection, bacterial infection and parasitic infection. Where the disease is cancer, such cancer is any one or more of bladder, brain, breast, cervical, colo-rectal, esophageal, kidney, liver, lung, nasopharangeal, pancreatic, prostate, skin, stomach, uterine, ovarian, testicular, or hematologic cancer.
In another aspect, the present invention includes compositions of the antagonists used in the methods of the invention, in a pharmaceutically acceptable carrier and wherein said PD-1 binding molecule and said potentiating agent are each present in an amount effective to produce increased T cell stimulation.
In one preferred embodiment, the invention includes medical kits comprising containers holding one or more of the agents for use in the invention together with pharmaceutical carriers for dilution thereof and instructions for administration. In addition, both of said PD-1 receptor antagonist and potentiating agent may be present as components in a single container, in a pharmaceutically acceptable carrier, when said components are to be administered at the same time.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows that B7-DC-lg binds to PD-1. Labeled B7-DC-lg was incubated at various concentrations with a CHO cell line constitutively expressing PD-1 or parent CHO cells that do not express PD-1. Binding was analyzed by flow cytometry. The median fluorescence intensity (MFI) of B7- DC-Ig (y-axis) is shown as a function of the concentration of probe (x-axis). B7-DC-lg binds to CHO.PD-1 cells (solid circle) but not untransfected CHO cells (gray triangle).
Figure 2 shows that B7-DC-lg competes with B7-H1 for binding to PD- 1. Unlabeled B7-DC-lg at various concentrations was first incubated with a CHO cell line constitutively expressing PD-1 before adding labeled B7-H1-lg to the cell mixture. The median fluorescence intensity (MFI) of B7-H1-lg (y- axis) is shown as a function of the concentration of unlabeled B7-DC-lg competitor (x-axis) added. As the concentration of unlabeled B7-DC-lg is increased the amount of labeled B7-H1-lg bound to CHO cells decreases, demonstrating that B7-DC-lg competes with B7-H1 for binding to PD-1.
Figure 3 shows the results of experiments wherein the combination of cyclophosphamide (CTX or Cytoxan®) and dimeric murine B7-DC-lg resulted in eradication of established CT26 tumors (colon carcinoma) in mice. Graph A shows tumor volume (mm3) versus days post tumor challenge in mice treated with 100 mg/kg of CTX on Day 10 while Graph B shows tumor volume (mm3) versus days post tumor challenge in mice treated with CTX on Day 10 followed a day later by the first B7-DC-lg administration. Each line in each graph represents one mouse. Black arrow stands for B7-DC-lg administration. Graph C shows average tumor volume.
Figure 4 shows the results of experiments wherein the combination of CTX and dimeric murine B7-DC-lg eradicated established CT26 tumors (colon carcinoma) in mice and protected against re-challenge with CT26. Mice that were treated with CTX and B7-DC-lg and found to be free of tumor growth on day 44 following tumor inoculation were rechallenged with tumors. The mice were later rechallenged again on on Day 70. None of the mice displayed tumor growth by day 100.
Figure 5 shows CTX and B7-DC-lg treatment resulted in generation of tumor specific memory CTL. Mice eradicated established CT26 subcutenous tumors post CTX and B7-DC-lg treatment were re-challenged with CT26 cells. Seven days later, splenocytes were isolated and pulsed with either ovalbumin, an irrelevant peptide, or AH1 , a CT26 specific peptide. Cells were stained with anti-CD8 antibody first followed by intracellular staining with anti- IFNγ antibody prior to FACS analysis. Figure 6 shows the effects of different doses of B7-DC-lg in combination with CTX on the eradication of established CT26 tumors in mice. Balb/C mice at age of 9 to 11 weeks were implanted subcutaneously with 1 E05 CT26 cells. On Day 9, mice were injected IP with 100 mg/kg of CTX. Twenty four hours later, on Day 10, mice were treated with 30, 100, or 300 ug of B7-DC-lg followed by 2 injections every week up to total 8 treatments. Tumor growth was measured two times per week.
Figure 7 shows the results of experiments wherein the combination of CTX and anti-PD-1 antibody resulted in eradication of established CT26 tumors (colon carcinoma) in mice. Graph A shows tumor volume (mm3) versus days post tumor challenge in untreated mice (i.e., mice treated with vehicle alone), Graph B shows tumor volume (mm3) versus days post tumor challenge in mice treated with anti-PD-1 alone starting on Day 11 at 300μg per injection, 3 times per week, up to 12 injections and Graph C shows tumor volume (mm3) versus days post tumor challenge in mice treated with CTX on Day 11 and the first anti-PD-1 administration on Day 12 at 300μg per injection, 3 times per week, up to 12 injections. Each line in each graph represents one mouse. Black arrow stands for anti-PD-1 administration.
Figure 8 shows the results of experiments wherein the combination of CTX and anti-CTLA4 antibody resulted in eradication of established CT26 tumors (colon carcinoma) in mice. Here, Graph A shows tumor volume (mm3) versus days post tumor challenge in mice treated with 100 mg/kg of CTX on Day 11 while Graph B shows tumor volume (mm3) versus days post tumor challenge in mice treated with CTX on Day 11 and anti-CTLA4 on Day 12 at 100μg per injection, 2 times per week, up to 8 injections. Each line in each graph represents one mouse. Black arrow stands for anti-CTLA-4 administration.
Figure 9 shows the results of experiments wherein Balb/C mice at age of 9 to 11 weeks of age were implanted with 1 X 105 CT26 cells subcutaneously. On Day 9, mice were injected with 100 mg/kg of CTX, IP. Twenty four hours later, on Day 10, mice were treated with 100 ug of B7-DC- Ig. There were 5 groups: naive mice that did not receive any tumor cells, vehicle injected, CTX alone, CTX + B7-DC-lg or B7-DC-lg alone. Two naive mice and 4 mice from other groups were removed from the study on Day 11 (2 days post CTX) and Day 16 (7 days post CTX) for T cell analysis. Left panel shows on Day 11 , 2 days post CTX injection, Treg in the spleen of the mice with CTX treatment was significantly lower than the one in the mice with tumor implantation and injected with vehicle. Right panel shows that on Day 16, 7 days post CTX and 6 days post B7-DC-lg treatment, B7-DC-lg significantly lowered the CD4+ T cells expressing high PD-1. This was observed in both the B7-DC-lg treated and CTX + B7-DC-lg treated mice. Mice implanted with tumor cells intended to have more PD-1 +/CD4+ T cells in the draining LN compared with naϊve mice.
Figure 10 shows the results of experiments wherein the combination of
CTX and B7-DC-lg resulted in increased survival in mice with tail vein injection of a mouse prostate tumor cell line. SP-1 cells were isolated from mouse lungs that were metastasized from TRAMP prostate tumor cell injection. B10.D2 mice were first injected with 3x105 SP-1 cells via tail vein injection. On Day 5, 12 and 19, mice were injected with 50 mg/kg of CTX where was indicated. On Day 6, 13 and 20, mice were administered with 5 mg/kg of B7-DC-lg were it was indicated. Here, "NT" refers to "not treated".
Figure 11. Balb/C mice at age of 11-13 weeks were given isolated hepatic metastases using a hemispleen injection technique. The spleens of anesthetized mice were divided into two halves and the halves were clipped.
CT26 cells (1 E05) were injected into one hemispleen, and after 30 seconds, that hemispleen was resected and the splenic draining vein was clipped. On
Day 10, mice received 1 injection of CTX at 50 mg/kg, IP. Twenty four hours later, on Day 11 , mice were treated with recombinant Listeria carrying AH 1 peptide, an immunodominant epitope of CT26, at 0.1 x LD5O (1 x107 CFU), then on Day 14 and 17. Mice were also treated with B7-DC-lg on Day 11 and then on Day 18. Mouse overall survival was monitored. DEFINITIONS
Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of skill in the art to which the disclosed invention belongs. In particular, the following terms and phrases have the following meaning.
The term "inhibitory signal transduction" is intended to mean any signal transduction having the effect of abolishing, or otherwise reducing, T cell responses against an antigen, whether by reducing T cell proliferation or by any other inhibitory mechanism, whereby the extent or duration of an immunogenic T cell response is decreased. Such inhibitory signal transduction may be due to PD-1 binding to a natural ligand, such as binding of PD-1 by B7-H1 or some other member of this class of ligands, B7-DC, or may be due to binding of CTLA4 to ligands, such as B7-1 or B7-2. In general, compounds of the invention reduce such inhibitory signal transduction and include, but are not limited to, PD-1 antagonists and CTLA4 antagonists.
The term "PD-1 antagonist" means any molecule that attenuates inhibitory signal transduction mediated by PD-1 , found on the surface of T cells, B cells, natural killer (NK) cells, monocytes, DC, and macrophages.
Such an antagonist includes a molecule that disrupts any inhibitory signal generated by a PD-1 molecule on a T cell. In specific examples of the invention, a PD-1 antagonist is a molecule that inhibits, reduces, abolishes or otherwise reduces inhibitory signal transduction through the PD-1 receptor signaling pathway. Such decrease may result where: (i) the PD-1 antagonist of the invention binds to a PD-1 receptor without triggering signal transduction, to reduce or block inhibitory signal transduction,; (ii) the PD-1 antagonist binds to a ligand (e.g. an agonist) of the PD-1 receptor, preventing its binding thereto (for example, where said agonist is B7-H1); (iii) the PD-1 antagonist binds to, or otherwise inhibits the activity of, a molecule that is part of a regulatory chain that, when not inhibited, has the result of stimulating or otherwise facilitating PD-1 inhibitory signal transduction; or (iv) the PD-1 antagonist inhibits expression of a PD-1 receptor or expression ligand thereof, especially by reducing or abolishing expression of one or more genes encoding PD-1 or one or more of its natural ligands. Thus, a PD-1 antagonist of the invention is a molecule that effects a decrease in PD-1 inhibitory signal transduction, thereby increasing T cell response to one or more antigens.
As used herein, the term "CTLA4 antagonist" means a compound that reduces CTLA4-mediated inhibition of T cell reactions. For example, in an T cell, CTLA4 delivers an inhibitory impulse upon binding of B7 ligands, such B7-1 and B7-2. A CTLA4 antagonist is one that dirupts binding of said ligands to CTLA4 on activated T cells. In one embodiment, the antagonist is an anti- CTLA4 antibody that binds CTLA4 to prevent ligand binding. I
As used herein, the term "active fragment" refers to a portion of a natural polypeptide, or a polypeptide with high sequence homology (for example, at least 80%, 85%, 90%, 95%, 98%, or 99% amino acid sequence identity) to a natural polypeptide and that exhibits PD-1 antagonist activity, for example, by binding PD-1 or by binding to a ligand of PD-1. In preferred embodiments, such a fragment would consist of the extracellular domain (ECD) of a B7-DC protein that binds to PD-1 , such as SEQ ID NO: 3, preferably amino acids 20 to 221 thereof. In the case of PD-1 polypeptide, an active fragment would be a portion of said polypeptide comprising a binding domain that binds to a natural ligand of PD-1 to prevent stimulation of PD-1 mediated inhibitory signal transduction by said ligand. Active fragments may be identified by their ability to compete with the molecule they are derived from for binding to a natural binding site. For example, active fragments will compete with wild-type B7-DC for binding to PD-1.
With respect to an antibody, the term "active fragment" means an antigen binding portion of an antibody that is less than an entire immunoglobulin. Such fragments include Fab and F(ab2)' fragments, capable of reacting with and binding to any of the polypeptides disclosed herein as being receptors or ligands. These Fab and F(ab')2 fragments lack the Fc portion of an intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody (Wahl et al., J. Nuc. Med. 24:316-325 (1983)). Also included are Fv fragments (Hochman, J. et al. (1973) Biochemistry 12:1130-1135; Sharon, J. et al.(1976) Biochemistry 15:1591-1594). These various fragments are produced using conventional techniques such as protease cleavage or chemical cleavage (see, e.g., Rousseaux et al., Meth. Enzymol., 121 :663-69 (1986)).
As used herein, the term "soluble portion" of a PD-1 antagonist means that portion of the full length polypeptide that does not include any part of the transmembrane portion or segment. For example, with respect to B7-DC, a soluble portion would include the extracellular portion (with or without the N- terminal signal sequence) but would not include any part of the transmembrane portion (or, at least, not enough to reduce solubility). Thus, the ECD of human B7-DC is shown as SEQ ID NO: 3 and consists of both the
IgV-like and IgC-like domains of the full length molecule (i.e., amino acids 20-
221 of the full length sequence (SEQ ID NO: 1).
As used herein, a "co-stimulatory polypeptide" is a polypeptide that, upon interaction with a cell-surface molecule on T cells, modulates the activity of the T cell. Thus, the response of the T cell can be an effector (e.g., CTL or antibody-producing B cell) response, a helper response providing help for one or more effector (e.g., CTL or antibody-producing B cell) responses, or a suppressive response.
As used herein, the term "treatment regimen" refers to a treatment of a disease or a method for achieving a desired physiological change, such as increased or decreased response of the immune system to an antigen or immunogen, such as an increase or decrease in the number or activity of one or more cells, or cell types, that are involved in such response, wherein said treatment or method comprises administering to an animal, such as a mammal, especially a human being, a sufficient amount of two or more chemical agents or components of said regimen to effectively treat a disease or to produce said physiological change, wherein said chemical agents or components are administered together, such as part of the same composition, or administered separately and independently at the same time or at different times (i.e., administration of each agent or component is separated by a finite period of time from one or more of the agents agents or components) and where administration of said one or more agents or components achieves a result greater than that of any of said agents or components when administered alone or in isolation.
As used herein the term "isolated" is meant to describe a compound of interest (e.g., either a polynucleotide or a polypeptide) that is in an environment different from that in which the compound naturally occurs e.g. separated from its natural milieu such as by concentrating a peptide to a concentration at which it is not found in nature. "Isolated" is meant to include compounds that are within samples that are substantially enriched for the compound of interest and/or in which the compound of interest is partially or substantially purified.
As used herein, the term "polypeptide" refers to a chain of amino acids of any length, regardless of modification (e.g., phosphorylation or glycosylation). A polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide.
As used herein, a "variant" polypeptide contains at least one amino acid sequence alteration as compared to the amino acid sequence of the corresponding wild-type polypeptide.
As used herein, an "amino acid sequence alteration" can be, for example, a substitution, a deletion, or an insertion of one or more amino acids. As used herein, the terms "portion," "segment," and "fragment," when used in relation to polypeptides, refer to a continuous sequence of residues, such as amino acid residues, which sequence forms a subset of a larger sequence. For example, if a polypeptide were subjected to treatment with any of the common endopeptidases, such as trypsin or chymotrypsin, the oligopeptides resulting from such treatment would represent portions, segments or fragments of the starting polypeptide. A "fragment" of a polypeptide thus refers to any subset of the polypeptide that is a shorter polypeptide of the full length protein. Generally, fragments will be five or more amino acids in length.
A derivative, analog or homolog, of a polypeptide (or fragment thereof) of the invention may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the mature polypeptide or a proprotein sequence. Such derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
As used herein, "valency" refers to the number of binding sites available per molecule.
In accordance with the present invention, the term "percent identity" or "percent identical," when referring to a sequence, means that a sequence is compared to a claimed or described sequence after alignment of the sequence to be compared (the "Compared Sequence") with the described or claimed sequence (the "Reference Sequence"). The Percent Identity is then determined according to the following formula:
Percent Identity = 100 [1-(CYR)]
wherein C is the number of differences between the Reference Sequence and the Compared Sequence over the length of alignment between the Reference Sequence and the Compared Sequence wherein (i) each base or amino acid in the Reference Sequence that does not have a corresponding aligned base or amino acid in the Compared Sequence and (ii) each gap in the Reference Sequence and (iii) each aligned base or amino acid in the Reference Sequence that is different from an aligned base or amino acid in the Compared Sequence, constitutes a difference; and R is the number of bases or amino acids in the Reference Sequence over the length of the alignment with the Compared Sequence with any gap created in the Reference Sequence also being counted as a base or amino acid. If an alignment exists between the Compared Sequence and the Reference Sequence for which the percent identity as calculated above is about equal to or greater than a specified minimum Percent Identity then the Compared Sequence has the specified minimum percent identity to the Reference Sequence even though alignments may exist in which the hereinabove calculated Percent Identity is less than the specified Percent Identity.
As used herein, the term "conservative amino acid substitution" means a substitution wherein the substituted amino acid has similar structural or chemical properties, and "non-conservative" amino acid substitutions are those in which the charge, hydrophobicity, or bulk of the substituted amino acid is significantly altered. Non-conservative substitutions will differ more significantly in their effect on maintaining (a) the structure of the peptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Examples of conservative amino acid substitutions include those in which the substitution is within one of the five following groups: 1) small aliphatic, nonpolar or slightly polar residues (AIa1 Ser, Thr, Pro, GIy); 2) polar, negatively charged residues and their amides (Asp, Asn, GIu, GIn); polar, positively charged residues (His, Arg, Lys); large aliphatic, nonpolar residues (Met, Leu, Me, VaI, Cys); and large aromatic resides (Phe, Tyr, Trp). Examples of non-conservative amino acid substitutions are those where 1 ) a hydrophilic residue, e.g., seryl or threonyl, is substituted for (or by) a hydrophobic residue, e.g., leucyl, isoleucyl, phenylalanyl, valyl, or alanyl; 2) a cysteine or proline is substituted for (or by) any other residue; 3) a residue having an electropositive side chain, e.g., lysyl, arginyl, or histidyl, is substituted for (or by) an electronegative residue, e.g., glutamyl or aspartyl; or 4) a residue having a bulky side chain, e.g., phenylalanine, is substituted for (or by) a residue that does not have a side chain, e.g., glycine.
The terms "individual", "host", "subject", and "patient" are used interchangeably herein, and refer to a mammal, including, but not limited to, primates, for example, human beings, as well as rodents, such as mice and rats, and other laboratory animals.
As used herein the term "effective amount" or "therapeutically effective amount" means a dosage sufficient to treat, inhibit, or alleviate one or more symptoms of a disease state being treated or to otherwise provide a desired pharmacologic and/or physiologic effect, especially enhancing T cell response to a selected antigen. The precise dosage will vary according to a variety of factors such as subject-dependent variables (e.g., age, immune system health, etc.), the disease, and the treatment being administered.
As used herein "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the therapeutic compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
The term "antibody" is meant to include both intact molecules as well as fragments thereof that include the antigen-binding site. Whole antibody structure is often given as H2L2 and refers to the fact that antibodies commonly comprise 2 light (L) amino acid chains and 2 heavy (H) amino acid chains. Both chains have regions capable of interacting with a structurally complementary antigenic target. The regions interacting with the target are referred to as "variable" or "V" regions and are characterized by differences in amino acid sequence from antibodies of different antigenic specificity. The variable regions of either H or L chains contains the amino acid sequences capable of specifically binding to antigenic targets. Within these sequences are smaller sequences dubbed "hypervariable" because of their extreme variability between antibodies of differing specificity. Such hypervariable regions are also referred to as "complementarity determining regions" or "CDR" regions. These CDR regions account for the basic specificity of the antibody for a particular antigenic determinant structure. The CDRs represent non-contiguous stretches of amino acids within the variable regions but, regardless of species, the positional locations of these critical amino acid sequences within the variable heavy and light chain regions have been found to have similar locations within the amino acid sequences of the variable chains. The variable heavy and light chains of all antibodies each have 3 CDR regions, each non-contiguous with the others (termed L1 , L2, L3, H1 , H2, H3) for the respective light (L) and heavy (H) chains. The accepted CDR regions have been described by Kabat et al, J. Biol. Chem. 252:6609-6616 (1977). The antibodies disclosed according to the invention may also be wholly synthetic, wherein the polypeptide chains of the antibodies are synthesized and, possibly, optimized for binding to the polypeptides disclosed herein as being receptors. Such antibodies may be chimeric or humanized antibodies and may be fully tetrameric in structure, or may be dimeric and comprise only a single heavy and a single light chain. DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a treatment regimen, or combination therapy, for treating disease in mammals comprising a compound that reduces or abolishes inhibitory signal transduction in T cells, preferably human T cells, administered in conjunction with a potentiating agent to increase an immune response.
The methods of the invention also relate to the use of broad spectrum immunomodulators and compositions of these. In general, the increased T cell response resulting from these methods is greater than any increased T cell response resulting from administering the same dose of either of said PD-
1 antagonist or said potentiating agent alone.
The disclosed compositions and regimens are useful to stimulate or enhance immune responses involving T cells. Thus, the methods of the invention are most useful in treating a disease condition that would benefit from an increase in T cell activity and where the increased T cell response is necessary or sufficient to treat said disease, even though the disease is not specifically caused or aggravated by a reduced T cell response. In a preferred embodiment, the type of disease to be treated or prevented is a malignant tumor or a chronic infectious disease caused by a bacterium, virus, protozoan, helminth, or other intracellular microbial pathogen that is attacked, i.e., by cytotoxic T lymphocytes. Activation of T cells using the disclosed compositions is also advantageous to treat or prevent conditions characterized by immunosuppression.
In accordance with the present invention, the T cell response can be regulated by molecules that bind to receptors on the T cell surface and molecules that bind to ligands of such receptors. In the case of PD-1 , molecules that bind PD-1 to reduce its inhibitory effect and/or molecules that bind one or more PD-1 ligands to reduce their ability to bind PD-1 have the effect of reducing the ability of PD-1 to inhibit T cell response, thereby increasing this response and the immunological effects thereof.
A. PD-1 RECEPTOR ANTAGONISTS
Compositions containing antagonists of PD-1 receptors are provided and include compounds or agents that either bind to and block a ligand of PD- 1 to interfere with or inhibit the binding of the ligand to the PD-1 receptor, or bind directly to and block the PD-1 receptor without inducing inhibitory signal transduction through the PD-1 receptor. In another embodiment, the PD-1 receptor antagonist binds directly to the PD-1 receptor without triggering inhibitory signal transduction and also binds to a ligand of the PD-1 receptor to reduce or inhibit the ligand from triggering signal transduction through the PD-1 receptor. By reducing the number and/or amount of ligands that bind to PD-1 receptor and trigger the transduction of an inhibitory signal, fewer cells are attenuated by the negative signal delivered by PD-1 signal transduction and a more robust immune response can be achieved.
In accordance with the present invention, PD-1 signaling requires binding to a PD-1 ligand (such as B7-H1 or B7-DC) in close proximity to a peptide antigen presented by major histocompatibility complex (MHC) (see, for example, Freeman Proc. Natl. Acad. Sci. U. S. A 105:10275-10276 (2008)). Therefore, proteins, antibodies or small molecules that prevent co- ligation of PD-1 and TCR on the T cell membrane are useful PD-1 antagonists contemplated by this invention.
Exemplary PD-1 receptor antagonists include, but are not limited to B7- DC polypeptides, including homologs and variants of these, as well as active fragments of any of the foregoing, and fusion proteins that incorporate any of these. In a preferred embodiment, the fusion protein comprises the soluble portion of B7-DC coupled to the Fc portion of an antibody, such as human IgG, and does not incorporate all or part of the transmembrane portion of human B7-DC. The PD-1 receptor antagonists can also be small molecule antagonists or antibodies that reduce or interfere with PD-1 receptor signal transduction by binding to ligands of PD-1 or to PD-1 itself, especially where co-ligation of PD-1 with TCR does not follow such binding, thereby not triggering inhibitory signal transduction through the PD-1 receptor.
The PD-1 receptor antagonists provided herein are generally useful in vivo and ex vivo as immune response-stimulating therapeutics. In general, the disclosed antagonist compositions are useful for treating a subject having or being predisposed to any disease or disorder to which the subject's immune system mounts an immune response.
1. B7-DC Polypeptides
In certain embodiments, B7-DC proteins can be used as PD-1 receptor antagonists. B7-DC is a natural ligand of PD-1 and binds to PD-1 with higher affinity than B7-H1 , and can thus inhibit B7-H1 :PD-1 interactions. Suitable B7- DC polypeptides, including variants, homologs and fragments thereof, can be obtained from the following full length human B7-DC polypeptides with (SEQ ID NO:1) or without (SEQ ID NO:2) the endogenous signal peptide.
MIFLLLMLSL ELQLHQIAAL FTVTVPKELY IIEHGSNVTL ECNFDTGSHV NLGAITASLQ 60
KVENDTSPHR ERATLLEEQL PLGKASFHIP QVQVRDEGQY QCIIIYGVAW DYKYLTLKVK 120
ASYRKINTHI LKVPETDEVE LTCQATGYPL AEVSWPNVSV PANTSHSRTP EGLYQVTSVL 180 RLKPPPGRNF SCVFWNTHVR ELTLASIDLQ SQMEPRTHPT WLLHIFIPFC IIAFIFIATV 240
IALRKQLCQK LYSSKDTTKR PVTTTKREVN SAI 273
(SEQ ID NO:1)
LFTVTVPKEL YIIEHGSNVT LECNFDTGSH VNLGAITASL QKVENDTSPH RERATLLEEQ 60 LPLGKASFHI PQVQVRDEGQ YQCIIIYGVA WDYKYLTLKV KASYRKINTH ILKVPETDEV 120
ELTCQATGYP LAEVSWPNVS VPANTSHSRT PEGLYQVTSV LRLKPPPGRN FSCVFWNTHV 180
RELTLASIDL QSQMEPRTHP TWLLHIFIPF CIIAFIFIAT VIALRKQLCQ KLYSSKDTTK 240
RPVTTTKREV NSAI 254
(SEQ ID NO: 2) The B7 family of molecules, including B7-DC, are expressed at the cell surface with a membrane proximal constant IgC domain and a membrane distal IgV domain. Receptors for these ligands share a common extracellular IgV-like domain. Interactions of receptor-ligand pairs are mediated predominantly through residues in the IgV domains of the ligands and receptors. In general, IgV domains are described as having two sheets that each contains a layer of β-strands. These β-strands are referred to as A', B, C, C, C", D, E, F and G. The structure of such polypeptides has been described in the literature (See Molnar et al., Crystal structure of the complex between programmed death-1 (PD-1) and its ligand PD-L2, PNAS, Vol. 105, pp. 10483-10488 (29 July 2008)).
B7-DC, a transmembrane protein, in its monomeric form, comprises IgV and IgC domains that make up the extracellular portion of the molecule (the extracellular domain, or ECD), with the IgV-like domain being responsible, in whole or in part, for PD-1 binding as well as other functions recited in the methods of the invention. For the human protein, the IgV domain is characterized in that it possesses a disulfide bond linking the B and F strands (referred to above), which appears to be characteristic of many IgV domains and possesses a similar three-dimensional structure with the IgV domains of both B7-1 and B7-2 (see Molnar et al.(2008), supra).
In one embodiment the B7-DC variant polypeptides contain amino acid alterations (i.e., substitutions, deletions or insertions) within one or more of these β-strands in any possible combination. In another embodiment, B7-DC variants contain one or more amino acid alterations (i.e., substitutions, deletions or insertions) within the A', C, C, C", D, E, F or G β-strands. In a preferred embodiment B7-DC variants contain one or more amino acid alterations in the G β-strand. In another embodiment, variant B7-DC polypeptide fragments include the IgC and IgV domains of B7-DC. In another embodiment, variant B7-DC polypeptide fragments include the IgV domain of B7-DC. Human and mouse B7-DC proteins contain a short intracytoplasmic domain, a single transmembrane domain and an extracellular domain. The extracellular domain contains two Ig domains; a membrane proximal IgC domain and a membrane distal IgV domain. Useful fragments of variant B7- DC polypeptides include soluble fragments. Soluble B7-DC fragments are fragments of B7-DC that may be shed, secreted or otherwise extracted from the producing cells. In one embodiment, variant B7-DC polypeptide fragments include the entire extracellular domain of B7-DC. The extracellular domain of B7-DC includes amino acids from about 20 to about amino acid 221 of murine or human B7-DC or active fragments thereof. In another embodiment, variant B7-DC polypeptide fragments include the IgC and IgV domains of B7-DC. In another embodiment, variant B7-DC polypeptide fragments include the IgV domain of B7-DC.
PD-1 signaling is thought to require binding to a PD-1 ligand (typically
B7-H1) in close proximity to a peptide antigen presented by major histocompatibility complex (MHC) (Freeman Proc. Natl. Acad. Sci. U. S. A 105:10275-10276 (2008)). Therefore, proteins, antibodies or small molecules that prevent co-ligation of PD-1 and TCR on the T cell membrane are useful PD-1 antagonists comntemplated by this invention.
The PD-1 antagonist useful in the methods and compositions of the invention include fragments of the B7-DC protein incorporating the ECD. Alternatively, the fragments of B7-DC include part of the extracellular domain that comprise the an IgV or IgV-like domain, preferably amino acids 20-221 , more preferably 20-121 , that are sufficient to bind to the PD-1 receptor to interfere with, or prevent, or otherwise reduce inhibitory signal transduction through the PD-1 receptor. In a preferred embodiment the B7-DC fragment competes with B7-H1 for binding to PD-1 receptors.
In one embodiment, variant B7-DC polypeptide fragments may contain a region of the polypeptide that is important for binding to PD-1. These polypeptide fragments may be useful to compete for binding to PD-1 and to prevent native B7-DC from binding to PD-1. By competing for binding to PD- 1 , these fragments may be useful to enhance an immune response, as inhibiting interactions of B7-H1 and B7-DC with PD-1 inhibits the suppression of immune responses that would otherwise occur. A polypeptide fragment of mouse or human B7-DC that could competitively bind to PD-1 can contain, for example, amino acids 101-108 or 110-114. The binding of wild-type B7-DC to PD-1 typically is inhibited by at least 50 percent, 60 percent, 70 percent, 75 percent, 80 percent, 90 percent, 95 percent, or more than 95 percent as compared to the level of binding of wild-type B7-DC to PD-1 in the absence of a fragment of said wild-type B7-DC. Exemplary B7-DC fragments useful in the methods and/or compositions of the invention include, but are in no way limited to, the following B7-DC extracellular domains:
Human B7-DC extracellular domain (ECD):
LFTVTVPKEL YIIEHGSNVT LECNFDTGSH VNLGAITASL QKVENDTSPH RERATLLEEQ 60
LPLGKASFHI PQVQVRDEGQ YQCIIIYGVA WDYKYLTLKV KASYRKINTH ILKVPETDEV 120
ELTCQATGYP LAEVSWPNVS VPANTSHSRT PEGLYQVTSV LRLKPPPGRN FSCVFWNTHV 180
RELTLASIDL QSQMEPRTHP TW 202 (SEQ ID NO:3)
and murine B7-DC ECD:
LFTVTAPKEV YTVDVGSSVS LECDFDRREC TELEGIRASL QKVENDTSLQ SERATLLEEQ 60 LPLGKALFHI PSVQVRDSGQ YRCLVICGAA WDYKYLTVKV KASYMRIDTR ILEVPGTGEV 120
QLTCQARGYP LAEVSWQNVS VPANTSHIRT PEGLYQVTSV LRLKPQPSRN FSCMFWNAHM 180
KELTSAIIDP LSRMEPKVPR TW 202
(SEQ ID NO:4)
Cynomolgus monkey B7-DC ECD:
LFTVTVPKEL YIIEHGSNVT LECNFDTGSH VNLGAITASL QKVENDTSPH RERATLLEEQ 60
LPLGKASFHI PQVQVRDEGQ YQCIIIYGVA WDYKYLTLKV KASYRKINTH ILKVPETDEV 120
ELTCQATGYP LAEVSWPNVS VPANTSHSRT PEGLYQVTSV LRLKPPPGRN FSCVFWNTHV 180 RELTLASIDL QSQMEPRTHP TW 202
(SeqlDNO: 15) Numerous other primate sequences useful in the methods and compositions of the invention are provided in Onlamoon et al., Immunology, Vol. 124, pp. 277-293 (2008).
A PD-1 antagonist useful in the compositions and methods of the invention also includes a fusion protein (as described below) that comprises first and second polypeptide portions, wherein said fusion protein, or at least the first polypeptide portion thereof, possesses PD-1 antagonist activity, especially where said fusion protein binds to and blocks PD-1 or binds to and blocks a ligand of PD-1. The first polypeptide portion of such fusion protein can comprise, or consist of, any of the PD-1 antagonistic polypeptides, or PD- 1 binding fragments thereof, otherwise recited herein for use as PD-1 antagonists in the methods of the invention. In a preferred embodiment of such a fusion protein, the recited first polypeptide portion is N-terminal to the recited second polypeptide portion. In a separate embodiment, the recited first polypeptide portion is linked to the recited second polypeptide portion by an oligopeptide in addition to the amino acids composing the recited first and second polypeptide portions, where said linking amino acids do not substantially decrease the PD-1 antagonist activity of said fusion protein.
In a preferred dimeric fusion protein, the dimer results from the covalent bonding of Cys residues in the CH regions of two of the Ig heavy chains that are the same Cys residues that are disulfide linked in dimerized normal Ig heavy chains.
A large number of polypeptide sequences that are routinely used as fusion protein binding partners are well known in the art. Examples of useful polypeptide binding partners include, but are not limited to, green fluorescent protein (GFP)1 glutathione S-transferase (GST), polyhistidine, myc, hemaglutinin, Flag™ tag (Kodak, New Haven, CT), maltose E binding protein and protein A. Still another embodiment provides a tetramer construct having a BirA substrate fused to the extracellular domain of a variant B7-DC polypeptide. Methods for making tetramer constructs are known in the art (see Pertovas, et al., J. Exp. Med., 203:2281 (2006)).
Exemplary murine B7-DC fusion proteins contain amino acids 20-221 of murine B7-DC fused to amino acids 237-469 of murine lgG2a (CAA49868). In one non-limiting example, human B7-DC fusion proteins contain amino acids 20-221 of human B7-DC fused to amino acids 245-476 of human IgGI (AAA02914). The signal peptides for B7-DC fusion proteins include the endogenous signal peptides or any other signal peptide that facilitates secretion of the fusion protein from a host. In another embodiment, the first polypeptide would include only the IgV domain. Other embodiments may comprise the hinge and Fc domain of an IgG antibody, such IgGI , with none of the variable region present. Other embodiments include use of the hinge and Fc region of lgG2 or lgG4, especially having an N297Q or other mutation that reduces effector function.
In accordance with the methods and compositions of the invention, the polypeptide useful as a PD-1 antagonist, or the first polypeptide portion of a fusion protein useful as a PD-1 antagonist, comprises an amino acid sequence that has at least 60%, or at least 65%, or at least 70%, or at least
75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or at least 99%, identity to amino acids 1-221 of SEQ ID NO: 1 , preferably amino acids 20-221 of SEQ ID NO: 1 , or amino acids 26-221 of SEQ ID NO: 1 , or amino acids 1 -202 of SEQ ID NO: 3 or 4, more preferably amino acids 20-121 of SEQ ID NO: 1 or amino acids 1 -102 of SEQ ID NO: 3 or 4.
In one embodiment, a polypeptide useful as a PD-1 antagonist, or the first polypeptide portion of a fusion protein useful as a PD-1 antagonist, consists of amino acids 1-221 of SEQ ID NO: 1 , or consists of amino acids
20-221 of SEQ ID NO: 1 , or consists of amino acids 26-221 of SEQ ID NO: 1 , or consists of amino acids 1-202 of SEQ ID NO: 3 or 4. In one embodiment (SEQ ID NO: 2), it does not comprise amino acids 1-19 of SEQ ID NO: 1.
In other specific examples, a PD-1 antagonist polypeptide, or first polypeptide portion of a PD-1 antagonist fusion protein, comprises the amino acid sequence 20-121 of SEQ ID NO: 1 , preferably where it comprises the amino acid sequence WDYKY at residues 110-114 thereof, or where it comprises amino acids 1-102 of SEQ ID NO: 3, preferably where it comprises the amino acid sequence WDYKY at residues 91-95 thereof.
In a preferred embodiment, such percent identities are achieved by reliance on conservative amino acid substitutions as defined elsewhere herein.
In one such embodiment, the PD-1 antagonist polypeptide, or first polypeptide portion of a PD-1 antagonist fusion protein, does not comprise amino acids 1-19 of SEQ ID NO: 1 , or does not comprise any portion of a transmembrane domain, especially not the entire such domain, or does not comprise any portion of the intracellular (or soluble) domain, especially not the entire such domain, of a PD-1 ligand or other PD-1 antagonist protein. In a preferred embodiment, such antagonist, or first polypeptide portion, comprises only the extracellular domain (ECD) of SEQ ID NO:1 and is thus comprised only of a soluble portion of the polypeptide of said sequence, or a fragment of said soluble portion.
In other such embodiments, the PD-1 antagonist polypeptide, or first polypeptide portion of a PD-1 antagonist fusion protein, comprises the IgV domain, or IgV-like domain, or PD-1 binding fragment thereof, of a PD-1 ligand, or consists of the IgV domain, or IgV-like domain, or PD-1 binding fragment thereof, of a PD-1 ligand. In specific examples, such PD-1 ligand is a wild-type B7-DC or B7-H1 molecule, preferably mouse or primate, preferably human, wild-type B7-DC or B7-H1 molecule. In other such embodiments, the PD-1 antagonist polypeptide, or first polypeptide portion of a PD-1 antagonist fusion protein, a PD-1 binding fragment of the IgV domain, or IgV-like domain, of a PD-1 ligand, especially where IgV domain, or IgV-like domain, consists of amino acids 20 - 121 of SEQ ID NO: 1 or amino acids 1 - 102 of SEQ ID NO: 3.
A PD-1 antagonist of the invention also includes a PD-1 binding fragment of amino acids 20-121 of SEQ ID NO: 1 (human full length), or amino acids 1-102 of SEQ ID NO: 3 (extracellular domain or ECD).
In specific embodiments thereof, the polypeptide or PD-1 binding fragment also incorporates amino acids WDYKY at residues 110-114 of SEQ ID NO: 1 or WDYKY at residues 91-95 of SEQ ID NO: 3. By way of non- limiting examples, such a PD-1 binding fragment comprises at least 10 , or at least 20, or at least 30, or at least 40, or at least 50, or at least 60, or at least 70, or at least 75, or at least 80, or at least 85, or at least 90, or at least 95, or at least 100 contiguous amino acids of the sequence of amino acids 20-121 of SEQ ID NO: 1 , wherein a preferred embodiment of each such PD-1 binding fragment would comprise as a sub-fragment the amino acids WDYKY found at residues 110-114 of SEQ ID NO: 1 or WDYKY at residues 91-95 of SEQ ID NO: 3.
Other preferred polypeptides and PD-1 binding fragments specifically contemplated by the invention include the polypeptide sequence of amino acids 20-121 of SEQ ID NO: 1 (human full length) and PD-1 binding fragments thereof, wherein, in such polypeptide or PD-1 binding fragment, a cysteine is present at residues 42 and/or 102, with a cysteine at both positions being preferred, and/or wherein a phenylalanine is present at residue 21 , and/or wherein a glutamic acid is present at residue 28, and/or wherein a threonine, and/or wherein a glutamine is present at residue 60, and/or wherein a glutamic acid is present at residue 101 , and/or wherein isoleucine is present at residue 103, and/or wherein an isoleucine is present at residue 105, and/or wherein a glycine is present at residue 107, and/or wherein valine is present at residue 108, and/or wherein a tryptophan is present at residue 110, and/or wherein aspartic acid is present at residue 111 , and/or wherein a tyrosine is present at residue 112, and/or wherein a lysine is present at residue 113, and/or wherein a tyrosine is present at residue 114, provided that, in the case of PD-1 binding fragments, said fragment is large enough to include such amino acid positions.
Additional preferred polypeptides and PD-1 binding fragments specifically contemplated by the invention include the polypeptide sequence of amino acids 1-102 of SEQ ID NO: 3 (human ECD) or SEQ ID NO: 4 (murine ECD) and PD-1 binding fragments thereof, wherein, in such polypeptide or PD-1 binding fragment, a cysteine is present at residues 23 and/or 83, with a cysteine at both positions being preferred, and/or wherein a phenylalanine is present at residue 2, and/or wherein a glutamic acid is present at residue 9, and/or wherein a threonine or arginine is present at residue 37, with threonine preferred, and/or wherein a glutamine is present at residue 41 , and/or wherein arginine is present at residue 82, and/or wherein a leucine is present at residue 84, and/or wherein an isoleucine is present at residue 86, and/or wherein a glycine is present at residue 88, and/or wherein an alanine is present at residue 89, and/or wherein a tryptophan is present at residue 91 , and/or wherein a aspartic acid is present at residue 92, and/or wherein a tyrosine is present at residue 93, and/or wherein a lysine is present at residue 94, and/or wherein a tyrosine is present at residue 95, provided that, in the case of PD-1 binding fragments, said fragment is large enough to include such amino acid positions.
In additional embodiments, any of the above polypeptides may also incorporate portions or fragments, for example, from 1 to 10 contiguous amino acids, drawn from the signal, transmembrane or C-terminal domains of the B7-DC or B7-H1 polypeptide, such as that of mouse or primate, preferably human. Such polypeptides and/or PD-1 binding fragments can also be present in any of the fusion proteins of the invention, for example, where such polypeptide or PD-1 binding fragment represents the "first polypeptide" of such fusion protein.
In specific examples, the molecule, combined with a potentiating agent for use in a treatment regimen of the invention, comprises a PD-1 binding fragment of amino acids 20-221 of SEQ ID NO: 1. In one such embodiment, the fragment is from amino acids 20 - 121 of SEQ ID NO: 1 , preferably where the fragment contains amino acids 110-114 of SEQ ID NO: 1. In some ebodiments, more than one such fragment is present (as described elsewhere herein) and the molecule comprises at least 2, 3, 4, 5 or more fragments of a B7-DC protein, especially where the fragment is part of, or contains part of, amino acids 20-221 of SEQ ID NO: 1. In a preferred embodiment thereof, at least one said fragment is from amino acids 20 - 121 of SEQ ID NO: 1 , more preferrably wherein at least one said fragment includes amino acids 110-114 of SEQ ID NO: 1 (i.e., the sequence WDYKY (SEQ ID NO: 14)). In preferred embodiments, the PD-1 binding fragment comprises at least 10, or at least 25, or at least 50, or at least 75, or at least 100 contiguous amino acids in length.
The endogenous human signal peptide has the following sequence MIFLLLMLSL ELQLHQIAA (SEQ ID NO:5) and represents the first 19 amino acids of SEQ ID NO: 1. In certain embodiments, the polypeptide fragments of B7-DC can include 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 contiguous amino acids of the endogenous or heterologous signal peptide (which can be used to produce a recombinant B7-DC polypeptide by expression in and secretion from a transformed cell). It will also be appreciated that a useful B7-DC polypeptide can include 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 contiguous amino acids of the transmembrane domain of B7-DC, and/or 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 contiguous amino acids of the cytoplasmic domain, or combinations thereof provided the B7-DC fragment retains the ability to antagonize the PD-1 receptor. The phenotypes of PD-1-/- mice provide direct evidence for PD-1 being a negative regulator of immune responses in vivo. In the absence of PD-1 , mice on the C57BL/6 background slowly develop a lupus-like glomerulonephritis and progressive arthritis (Nishimura, et al., Immunity, 11 :141-151 (1999)). PD-1-/- mice on the BALB/c background rapidly develop a fatal autoimmune dilated cardiomyopathy (Nishimura, et al., Science. 291 :319-322 (2001)). However, substantial evidence indicates that B7-DC can function to costimulate activate T cell responses. In the presence of suboptimal TCR signals, B7-DC causes increased proliferation and production of cytokines in vitro (Tseng, et al., J. Exp. Med. 193:839-846 (2001)). On the other hand, in vitro studies indicate a negative regulatory role for B7-DC in T cell responses. These seemingly contradictory data are best interpreted by expression of additional receptors for B7-DC on T cells other than PD-1.
Therefore, B7-DC proteins, variants, fragments and fusions thereof, may have the advantage of directly enhancing T cell responses by binding to an unknown receptor that activates the T cell, in addition to enhancing T cell responses by preventing the PD-1 mediated inhibitory signal transduction.
2. B7-H1 Polypeptides
In another embodiment, the compound for use in combination with a potentiating agent in the treatment regimen of the invention, is, or comprises, a fragment of a mammalian B7-H1 , preferably from mouse or primate, preferably human, wherein said fragment binds to and blocks PD-1 but does not result in inhibitory signal transduction through PD-1 and said fragment is at least 10, or at least 20, or at least 30, or at least 40, or at least 50, or at least 60, or at least 70, or at least 80, or at least 90, or at least 100 contiguous amino acids in length. In other embodiments, the fragment can be of variable length so long as it has the function of binding to PD-1 but does not produce inhibitory signal transduction that results in reduced T cell proliferation. Such B7-H1 fragments also find use as part of the first polypeptide portion of fusion proteins of the invention. B7-H1 sequences are as follows:
Human B7-H1 Polypeptide (SEQ ID NO. 16): MRIFAVFIFM TYWHLLNAFT VTVPKDLYW EYGSNMTIEC KFPVEKQLDL AALIVYWEME 60
DKNIIQFVHG EEDLKVQHSS YRQRARLLKD QLSLGNAALQ ITDVKLQDAG VYRCMISYGG 120
ADYKRITVKV NAPYNKINQR ILWDPVTSE HELTCQAEGY PKAEVIWTSS DHQVLSGKTT 180
TTNSKREEKL FNVTSTLRIN TTTNEIFYCT FRRLDPEENH TAELVIPELP LAHPPNERTH 240
LVILGAILLC LGVALTFIFR LRKGRMMDVK KCGIQDTNSK KQSDTHLEET 290
Murine B7-H1 (SEQ ID NO: 17)
MRIFAGIIFT ACCHLLRAFT ITAPKDLYW EYGSNVTMEC RFPVERELDL LALWYWEKE 60
DEQVIQFVAG EEDLKPQHSN FRGRASLPKD QLLKGNAALQ ITDVKLQDAG VYCCIISYGG 120
ADYKRITLKV NAPYRKINQR ISVDPATSEH ELICQAEGYP EAEVIWTNSD HQPVSGKRSV 280 TTSRTEGMLL NVTSSLRVNA TANDVFYCTF WRSQPGQNHT AELIIPELPA THPPQNRTHW 240
VLLGSILLFL IWSTVLLFL RKQVRMLDVE KCGVEDTSSK NRNDTQFEET 290
Macaca mulatta PD-L1 (SEQ ID NO: 18) MRIFAVFIFT IYWHLLNAFT VTVPKDLYW EYGSNMTIEC RFPVEKQLGL 60
TSLIVYWEME DKNIIQFVHG EEDLKVQHSN YRQRAQLLKD QLSLGNAALR 120
ITDVKLQDAG VYRCMISYGG ADYKRITVKV NAPYNKINQR ILWDPVTSE 180
HELTCQAEGY PKAEVIWTSS DHQVLSGKTT TTNSKREEKL LNVTSTLRIN 240
TTANEIFYCI FRRLGPEENH TAELVIPELP LALPPNERTH LVILGAIFLL 300 LGVALTFIFY LRKGRMMDMK KSGIRVTNSK KQRDTQLEET 340
B7-H1-lg proteins are described in WO/2001/014557 (pub. 1 March 2001 ) and in WO/2002/079499 (pub. 10 October 2002).
3. PD-1 and Other Polypeptides
Other useful polypeptides of the invention include those that bind to the ligands of the PD-1 receptor. These include the PD-1 receptor protein, or soluble fragments thereof, which can bind to the PD-1 ligands, such as B7-H1 or B7-DC, and prevent binding to the endogenous PD-1 receptor, thereby preventing inhibitory signal transduction. B7-H1 has also been shown to bind the protein B7.1 (Butte et al., Immunity, Vol. 27, pp. 111-122, (2007)). Such fragments also include the soluble ECD portion of the PD-1 protein that includes mutations, such as the A99L mutation, that increases binding to the natural ligands (Molnar et al., Crystal structure of the complex between programmed death-1 (PD-1) and its ligand PD-L2, PNAS, Vol. 105, pp. 10483-10488 (29 July 2008)),. B7-1 or soluble fragments thereof, which can bind to the B7-H1 ligand and prevent binding to the endogenous PD-1 receptor, thereby preventing inhibitory signal transduction, are also useful.
PD-1 polypeptides useful in the methods of the invention are as follows:.
Human PD-1 (SEQ ID NO: 19)
MQIPQAPWPV VWAVLQLGWR PGWFLDSPDR PWNPPTFFPA LLWTEGDNA TFTCSFSNTS 60
ESFVLNWYRM SPSNQTDKLA AFPEDRSQPG QDCRFRVTQL PNGRDFHMSV VRARRNDSGT 120
YLCGAISLAP KAQIKESLRA ELRVTERRAE VPTAHPSPSP RPAGQFQTLV VGWGGLLGS 180 LVLLVWVLAV ICSRAARGTI GARRTGQPLK EDPSAVPVFS VDYGELDFQW REKTPEPPVP 240
CVPEQTEYAT IVFPSGMGTS SPARRGSADG PRSAQPLRPE DGHCSWPL 288
Cynomolgus monkey PD-1 (SEQ ID NO: 20)
MQIPQAPWPV VWAVLQLGWR PGWFLESPDR PWNAPTFSPA LLLVTEGDNA TFTCSFSNAS 60
ESFVLNWYRM SPSNQTDKLA AFPEDRSQPG QDCRFRVTRL PNGRDFHMSV VRARRNDSGT 120
YLCGAISLAP KAQIKESLRA ELRVTERRAE VPTAHPSPSP RPAGQFQTLV VGWGGLLGS 180
LVLLVWVLAV ICSRAARGTI GARRTGQPLK EDPSAVPVFS VDYGELDFQW REKTPEPPVP 240 CVPEQTEYAT IVFPSGMGTS SPARRGSADG PRSAQPLRPE DGHCSWPL 288
In accordance with the invention, because B7-1 and fragments thereof can also bind to B7-H1 and send inhibitory transducstion to T cells through B7-H1 , blocking of this interaction can also reduce inhibitory signal transduction that occurs through B7-H1. Compounds for use in the invention include those molecules that block this type of interaction. Such molecules have been disclosed in Butte et al (2007), supra, and include anti-B7-H1 antibodies with dual-specificity that block either the B7-H1 :B7-1 and B7- H1 :PD-1 interaction as well as antibodies exhibiting mono-specificity that block the PD-L1 :B7-1 interaction. Compounds that block this interaction by blocking B7-1 are also useful, and include anti-B7-1 antibodies.
4. Variant polypeptides
Polypeptides useful in the invention, as described, include those that are mutated to contain one or more amino acid substitutions, deletions, or insertions. Methods for mutagenesis are known in the art. The mutated or variant polypeptides inhibit or reduce inhibitory signal transduction through PD-1 receptors by binding to ligands of PD-1. Alternatively, the variants (e.g. B7-DC polypeptides) can bind to the PD-1 receptor and inhibit, reduce, or block inhibitory signal transduction through the PD-1 receptor. The variant polypeptides may be of any species of origin. In one embodiment, the variant polypeptide is from a mammalian species. In a preferred embodiment, the variant polypeptide is of murine or primate, preferably human, origin.
In one embodiment the variant polypeptide is a B7-DC polypeptide that has the same binding affinity to PD-1 as wildtype or non-variant B7-DC but does not have or has less than 10% ability to trigger inhibitory signal transduction through the PD-1 receptor relative to a non-mutated B7-DC polypeptide. In other embodiments, the variant B7-DC polypeptide has 10%, 20%, 30%, 40%, 50%, or 60% more binding affinity to PD-1 than wildtype B7- DC without triggering PD-1 inhibitory signaling transduction.
A variant polypeptide (e.g. a variant B7-DC polypeptide) includes those having any combination of amino acid substitutions, deletions or insertions so long as the PD-1 antagonizing activity is not substantially reduced versus the wild type. However, where there is such a reduction, this should be by no more than half that of the wild type so that said variant has at least 50% of the PD-1 antagonist activity of the wild type protein, preferably at least 60%, more preferably at least 80%, most preferably at least 90% or 95%, with at least 100% being especially preferred. Increases in such activity resulting from said variant is even more desirable. In one embodiment, isolated B7-DC variant polypeptides have amino acid alterations such that their amino acid sequence shares at least 60, 70, 80, 85, 90, 95, 97, 98, 99, 99.5 or 100% identity with an amino acid sequence of a wild type B7-DC polypeptide, especially that from a mammal, preferably wild type murine or wild type primate, preferably human, B7-DC polypeptide.
Polypeptide sequence identity can be calculated using the definition of % identity provided hereinabove.
Amino acid substitutions in polypeptides may be "conservative" or
"non-conservative".
B7 family molecules, including B7-DC, are expressed at the cell surface with a membrane proximal constant IgC domain and a membrane distal IgV domain. Receptors for these ligands share a common extracellular IgV-like domain. Interactions of receptor-ligand pairs are mediated predominantly through residues in the IgV domains of the ligands and receptors. In general, IgV domains are described as having two sheets that each contains a layer of β-strands. These β-strands are referred to as A', B, C, C, C", D, E, F and G. In one embodiment the B7-DC variant polypeptides contain amino acid alterations (i.e., substitutions, deletions or insertions) within one or more of these β-strands in any possible combination. In another embodiment, B7-DC variants contain one or more amino acid alterations (i.e., substitutions, deletions or insertions) within the A', C, C1 C", D, E, F or G β-strands. In one embodiment, B7-DC variants contain one or more amino acid alterations in the G β-strand.
With respect to murine or primate, preferably human, B7-DC, a variant B7-DC polypeptide can contain, without limitation, substitutions, deletions or insertions at positions that do not substantially reduce binding to PD-1 relative to non-mutated B7-DC. It is understood, however, that substitutions at the recited amino acid positions can be made using any amino acid or amino acid analog. For example, the substitutions at the recited positions can be made with any of the naturally-occurring amino acids (e.g., alanine, aspartic acid, asparagine, arginine, cysteine, glycine, glutamic acid, glutamine, histidine, leucine, valine, isoleucine, lysine, methionine, proline, threonine, serine, phenylalanine, tryptophan, or tyrosine).
While the substitutions described herein are with respect to mouse and primate, especially human, B7-DC, it is noted that one of ordinary skill in the art could readily make equivalent alterations in the corresponding polypeptides from other species (e.g., rat, hamster, guinea pig, gerbil, rabbit, dog, cat, horse, pig, sheep, cow or non-human primate).
Preferred fragments include all or part of the extracellular domain of
B7-DC effective to bind to PD-1.
In one embodiment, variant B7-DC polypeptide fragments are those that retain the ability to bind to PD-1 without triggering PD-1 inhibitory signal transduction. One embodiment provides a variant B7-DC polypeptide that is a fragment of full-length B7-DC and typically has at least 20 percent, 30 percent, 40 percent, 50 percent, 60 percent, 70 percent, 80 percent, 90 percent, 95 percent, 98 percent, 99 percent, 100 percent, or even more than 100 percent of the PD-1 antagonist activity of the full-length variant B7-DC polypeptide.
Useful fragments of variant B7-DC polypeptides include soluble fragments. Soluble B7-DC fragments are fragments of B7-DC that may be shed, secreted or otherwise extracted from the producing cells. In one embodiment, variant B7-DC polypeptide fragments include the entire extracellular domain of B7-DC. The extracellular domain of B7-DC includes amino acids from about 20 to about amino acid 221 of murine or primate, preferably human, B7-DC. In another embodiment, variant B7-DC polypeptide fragments include the IgC and IgV domains of B7-DC. In another embodiment, variant B7-DC polypeptide fragments include the IgV domain of B7-DC.
In one embodiment, variant B7-DC polypeptide fragments contain a region of the polypeptide that is important for binding affinity for PD-1. These polypeptide fragments are useful to bind to and block the PD-1 receptor to prevent native ligands from binding to PD-1 receptor, thereby enhancing an immune response. Inhibiting interactions of native B7-H1 or B7-DC with PD-1 inhibits the suppression of immune responses that would otherwise occur. A polypeptide fragment of mouse or primate, preferably human, B7-DC that binds to PD-1 contains, by way of non-limiting example, amino acids 101-105, or 111-113. The binding of B7-H1 to PD-1 receptor typically is inhibited by at least 50 percent, or by at least 60 percent, or by at least 70 percent, or by at least 75 percent, or by at least 80 percent, or by at least 90 percent, or by at least 95 percent, or more compared to the level of binding of B7-H1 to PD-1 in the absence of the fragment.
Human PD-1 mutant A99L binds B7-DC and B7-H1 with higher affinity than unmutated human PD-1 (Lazar Molnar et al PNAS 105 p. 10483-10488
(2008)). In one embodiment of the invention, the compound acting to reduce inhibitory signal transduction is a soluble protein, such as the ECD of PD-1 incorporating this mutation.
5. Modified Polypeptides
Polypeptides useful in the invention, as decribed, including variants, homologs and fragments thereof, can be modified by chemical moieties found associated with polypeptides in the normal cellular environment, for example, by phosphorylation, methylation, amidation, sulfation, acylation, glycosylation, sumoylation and ubiquitylation of the polypeptide. Such polypeptides may also be modified by chemical moieties that are not normally part of polypeptides in a cellular environment. Such modifications can be introduced into the molecule by reacting targeted amino acid residues of the polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or terminal residues. Another useful modification is cyclization of the protein. Such modifications also include introduction of a label capable of providing a detectable signal, either directly or indirectly, including, but not limited to, radioisotopes and fluorescent compounds.
Examples of chemical derivatives of the polypeptides include lysinyl and amino terminal residues derivatized with succinic or other carboxylic acid anhydrides. Derivatization with a cyclic carboxylic anhydride has the effect of reversing the charge of the lysinyl residues. Other suitable reagents for derivatizing amino-containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylisourea; 2,4 pentanedione; and transaminase-catalyzed reaction with glyoxylate. Carboxyl side groups, aspartyl or glutamyl, may be selectively modified by reaction with carbodiimides (R — N=C=N-R') such as 1-cyclohexyl-3-(2-morpholinyl-(4- ethyl)carbodiimide or 1-ethyl-3-(4-azonia-4,4-dimethylpentyl) carbodiimide. Furthermore, aspartyl and glutamyl residues can be converted to asparaginyl and glutaminyl residues by reaction with ammonia. Polypeptides of the invention can also include one or more D-amino acids that are substituted for one or more L-amino acids.
In other embodiments, the potentiating agent, such as CTX1 may be itself part of the compound that reduces inhibitory signal transduction, such as where the potentiating agent is chemically linked to a PD-1 antagonist of the invention. 6. Fusion Proteins
Fusion polypeptides having a first fusion partner, or polypeptide portion, comprising all or a part of a PD-1 antagonist protein, a B7-DC polypeptide for example, (including variants, homologs and fragments thereof) fused (i) directly to a second polypeptide or, (ii) optionally, fused to a linker peptide sequence that is fused to the second polypeptide are also provided. The presence of the fusion partner can alter, for example, the solubility, affinity and/or valency of the PD-1 antagonist polypeptide. The disclosed fusion proteins include any combination of amino acid alteration (i.e., substitution, deletion or insertion), fragment, and/or modification of a PD-1 antagonist polypeptide as described above. In one embodiment, B7-DC fusion proteins include the extracellular domain of a B7-DC protein as the first binding partner. In another embodiment, such B7-DC fusion proteins include the IgV and IgC domain of a B7-DC protein as the first binding partner. In another embodiment, variant B7-DC fusion proteins include the IgV domain of a B7-DC protein as the first binding partner.
Representative first fusion partners include primate, preferably human, or murine B7-DC polypeptide, fragments thereof, and variants thereof disclosed hereinabove. Preferred fragments include the extracellular domain of B7-DC. As recited, the extracellular domain can include 1-10 contiguous amino acids of a signal peptide, B7-DC transmembrane domain, or both.
In one embodiment, the compositions and/or products and/or methods of the invention utilize PD-1 receptor antagonist, especially polypeptides, including variants, homologs and fragments thereof, that are coupled to other polypeptides to form fusion proteins that antagonize the PD-1 receptor by binding a PD-1 ligand, such as B7-H1 , thereby inhibiting the ligand from interacting with PD-1. In another embodiment, PD-1 receptor antagonist polypeptides, or variants thereof, are coupled to other polypeptides to form fusion proteins that antagonize the PD-1 receptor by binding to and blocking the PD-1 receptor and inhibit or reduce inhibitory signal transduction through PD-1.
The second polypeptide binding partner, or second polypeptide portion, may be N-terminal or C-terminal relative to the PD-1 antagonist polypeptide. In a preferred embodiment, the second polypeptide is C-terminal to the PD-1 antagonist polypeptide.
In a preferred embodiment, the fusion protein contemplated for use in the methods and compositions and/or products of the invention comprises at least a portion of an antibody. With the advent of methods of molecular biology and recombinant technology, it is now possible to produce antibody molecules by recombinant means and thereby generate gene sequences that code for specific amino acid sequences found in the polypeptide structure of the antibodies. Such antibodies can be produced by either cloning the gene sequences encoding the polypeptide chains of said antibodies or by direct synthesis of said polypeptide chains, with in vitro assembly of the synthesized chains to form active tetrameric (H2L2) structures with affinity for specific epitopes and antigenic determinants. This has permitted the ready production of antibodies having sequences characteristic of neutralizing antibodies from different species and sources.
Regardless of the source of the antibodies, or how they are recombinant^ constructed, or how they are synthesized, in vitro or in vivo, using transgenic animals, such as cows, goats and sheep, using large cell cultures of laboratory or commercial size, in bioreactors or by direct chemical synthesis employing no living organisms at any stage of the process, all antibodies have a similar overall 3 dimensional structure. This structure is often given as H2L2 and refers to the fact that antibodies commonly comprise 2 light (L) amino acid chains and 2 heavy (H) amino acid chains. Both chains have regions capable of interacting with a structurally complementary antigenic target. The regions interacting with the target are referred to as "variable" or "V" regions and are characterized by differences in amino acid sequence from antibodies of different antigenic specificity.
In preferred embodiments, the PD-1 receptor antagonist polypeptides, including fragments, mutants and other variants, have a first fusion partner having all or a part of a B7-DC protein or variant thereof fused (i) directly to a second polypeptide or, (ii) optionally, fused to a linker peptide sequence that is fused to the second polypeptide. The presence of the fusion partner can alter the solubility, affinity and/or valency of the B7-DC polypeptide. In more preferred embodiments, B7-DC polypeptides are fused to one or more domains of an Ig heavy chain constant region, more preferably an amino acid sequence corresponding to the hinge, CH2 and CH3 regions of a human immunoglobulin Cγ1 chain or to the hinge, CH2 and CH3 regions of a murine immunoglobulin Cγ2a chain. In a preferred embodiment, the constant region preferably includes a mutation (for example N297Q) to eliminate or reduce Fc receptor binding.
The hinge, CH2 and CH3 regions of a human immunoglobulin Cγ1 chain has the following amino acid sequence:
EPKSCDKTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCVWD VSHEDPEVKF 60
NWYVDGVEVH NAKTKPREEQ YNSTYRWSV LTVLHQDWLN GKEYKCKVSN KALPAPIEKT 120
ISKAKGQPRE PQVYTLPPSR DELTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP 180
PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GK 232 (SEQ ID NO:6).
The hinge, CH2 and CH3 regions of a murine immunoglobulin Cγ2a chain has the following amino acid sequence:
EPRGPTIKPC PPCKCPAPNL LGGPSVFIFP PKIKDVLMIS LSPIVTCVW DVSEDDPDVQ 60
ISWFVNNVEV HTAQTQTHRE DYNSTLRWS ALPIQHQDWM SGKEFKCKVN NKDLPAPIER 120
TISKPKGSVR APQVYVLPPP EEEMTKKQVT LTCMVTDFMP EDIYVEWTNN GKTELNYKNT 180
EPVLDSDGSY FMYSKLRVEK KNWVERNSYS CSWHEGLHN HHTTKSFSRT PGK 233
(SEQ ID NO:7) Exemplary murine B7-DC fusion proteins contain amino acids 20-221 of murine B7-DC fused to amino acids 237-469 of murine lgG2a (CAA49868). Human B7-DC fusion proteins can contain amino acids 20-221 of human B7-DC fused to amino acids 245-476 of human IgGI (AAA02914). The signal peptides for B7-DC fusion proteins can be the endogenous signal peptides or any other signal peptide that facilitates secretion of the fusion protein from a host.
A representative murine B7-DC-lg fusion protein is encoded by the nucleic acid sequence of SEQ ID NO:8.
It will be appreciated that the disclosed nucleic acid sequences can be codon-optimized to increase levels of expression for synthesizing the fusion proteins useful in the methods and compositions of the present invention. Methods for codon optimization are known in the art.
The murine B7-DC-lg fusion protein encoded by SEQ ID NO:8 has the following amino acid sequence:
MLLLLPILNL SLQLHPVAAL FTVTAPKEVY TVDVGSSVSL ECDFDRRECT ELEGIRASLQ 60
KVENDTSLQS ERATLLEEQL PLGKALFHIP SVQVRDSGQY RCLVICGAAW DYKYLTVKVK 120
ASYMRIDTRI LEVPGTGEVQ LTCQARGYPL AEVSWQNVSV PANTSHIRTP EGLYQVTSVL 180
RLKPQPSRNF SCMFWNAHMK ELTSAIIDPL SRMEPKVPRT WEPRGPTIKP CPPCKCPAPN 240
LLGGPSVFIF PPKIKDVLMI SLSPIVTCW VDVSEDDPDV QISWFVNNVE VHTAQTQTHR 300 EDYNSTLRW SALPIQHQDW MSGKEFKCKV NNKDLPAPIE RTISKPKGSV RAPQVYVLPP 360
PEEEMTKKQV TLTCMVTDFM PEDIYVEWTN NGKTELNYKN TEPVLDSDGS YFMYSKLRVE 420
KKNWVERNSY SCSWHEGLH NHHTTKSFSR TPGK 454
( SEQ ID NO : 9 )
SEQ ID NO: 10 provides the amino acid sequence for murine B7-DC-lg fusion protein without the signal sequence.
LFTVTAPKEV YTVDVGSSVS LECDFDRREC TELEGIRASL QKVENDTSLQ SERATLLEEQ 60
LPLGKALFHI PSVQVRDSGQ YRCLVICGAA WDYKYLTVKV KASYMRIDTR ILEVPGTGEV 120 QLTCQARGYP LAEVSWQNVS VPANTSHIRT PEGLYQVTSV LRLKPQPSRN FSCMFWNAHM 180
KELTSAIIDP LSRMEPKVPR TWEPRGPTIK PCPPCKCPAP NLLGGPSVFI FPPKIKDVLM 240
ISLSPIVTCV WDVSEDDPD VQISWFVNNV EVHTAQTQTH REDYNSTLRV VSALPIQHQD 300 WMSGKEFKCK VNNKDLPAPI ERTISKPKGS VRAPQVYVLP PPEEEMTKKQ VTLTCMVTDF 360 MPEDIYVEWT NNGKTELNYK NTEPVLDSDG SYFMYSKLRV EKKNWVERNS YSCSWHEGL 420 HNHHTTKSFS RTPGK 435 (SEQ ID NO: 10)
In one embodiment human B7-DC-lg is encoded by the nucleic acid sequence of SEQ ID NO:11 , encoding the amino acid sequence for human B7-DC-lg:
MIFLLLMLSL ELQLHQIAAL FTVTVPKELY IIEHGSNVTL ECNFDTGSHV NLGAITASLQ 60
KVENDTSPHR ERATLLEEQL PLGKASFHIP QVQVRDEGQY QCIIIYGVAW DYKYLTLKVK 120
ASYRKINTHI LKVPETDEVE LTCQATGYPL AEVSWPNVSV PANTSHSRTP EGLYQVTSVL 180
RLKPPPGRNF SCVFWNTHVR ELTLASIDLQ SQMEPRTHPT WEPKSCDKTH TCPPCPAPEL 240
LGGPSVFLFP PKPKDTLMIS RTPEVTCVW DVSHEDPEVK FNWYVDGVEV HNAKTKPREE 300 QYNSTYRWS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS 360
RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK 420
SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK 453
(SEQ ID NO:12)
The present invention specifically contemplates embodiments where the mature fusion protein useful in the methods and compositions of the invention have the signal sequence removed. In a preferred embodiment, the signal sequence is completely removed.
SEQ ID NO:13 provides the amino acid sequence for human B7-DC-lg without the signal sequence.
LFTVTVPKEL YIIEHGSNVT LECNFDTGSH VNLGAITASL QKVENDTSPH RERATLLEEQ 60
LPLGKASFHI PQVQVRDEGQ YQCIIIYGVA WDYKYLTLKV KASYRKINTH ILKVPETDEV 120 ELTCQATGYP LAEVSWPNVS VPANTSHSRT PEGLYQVTSV LRLKPPPGRN FSCVFWNTHV 180
RELTLASIDL QSQMEPRTHP TWEPKSCDKT HTCPPCPAPE LLGGPSVFLF PPKPKDTLMI 240
SRTPEVTCW VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE EQYNSTYRW SVLTVLHQDW 300
LNGKEYKCKV SNKALPAPIE KTISKAKGQP REPQVYTLPP SRDELTKNQV SLTCLVKGFY 360
PSDIAVEWES NGQPENNYKT TPPVLDSDGS FFLYSKLTVD KSRWQQGNVF SCSVMHEALH 420 NHYTQKSLSL SPGK 434
(SEQIDNO:13). The present invention specifically contemplates embodiments where the disclosed B7-DC-lg fusion proteins used in the methods and compositions disclosed herein have at least about 80%, 85%, 90%, 99% or 100% sequence identity to SEQ ID NO: 9, 10, 12, or 13.
In another embodiment of the invention, the fusion polypeptide may have bi-specific function whereby the first fusion partner binds to a ligand of
PD-1 , such as B7-H1 , and the second fusion partner binds to the PD-1 receptor without triggering inhibitory signal transduction through the PD-1 receptor.
While a polypeptide useful in the invention may be monomeric or dimeric, the fusion proteins themselves may be present in a monomeric or an oligomeric form, preferably as a dimer. In specific embodiments, the fusion proteins useful as PD-1 antagonists in the methods and compositions of the invention may assemble spontaneously into oligomeric, especially dimeric, forms or may be chemically linked to form such oligomers by means well known in the art. For example, a fusion protein useful in practicing the invention may itself comprise a portion of a B7-DC polypeptide fused to a portion of an antibody and these may be further assembled into a dimer. In one such example, a polypeptide for use in the invention is fused as a single amino acid chain to the Fc region of an antibody (such as where this construct is expressed from a single recombinant polynucleotide), after which two such fusion products are linked to each other to form a homodimer, such as by a disulfide linkage between the respective Fc regions.
Such dimeric products may be homodimers (where both monomeric fusion proteins are identical) or may be heterodimers (where two different fusion proteins are linked to each other). The individual monomers of such dimers may be linked by any means known in the art, such as by covalent linkage (e.g., a disulfide bond) or by non-covalent linkage (such as an ionic interaction). The B7-DC-lg used in the examples of the invention were present in the form of a homodimer having 2 copies of SEQ ID NO: 10 linked together by a disulfide linkage. In addition, the heterodimers of the invention include bispecific proteins and fusion proteins wherein one monomeric portion binds to PD-1 and the other binds to a natural ligand of PD-1. Such heterodimers are formed by coupling of polypeptides and fusion proteins fully described elsewhere herein.
In another useful embodiment of the invention, the PD-1 antagonist is a heterodimer, such as where two fusion proteins are linked together but they are not of identical amino acid sequence. In a specific example, each monomer may comprise an Fc portion of an antibody linked to an active fragment of a B7-DC polypeptide where these active fragments are from different portions of the B7-DC polypeptide or where a fusion protein comprising an Fc portion of an antibody fused to a full length native B7-DC polypeptide is linked (for example, cross-linked) to a fusion protein comprising an Fc portion of an antibody and an active fragment of a full length native BY- DC polypeptide. In each such case, the portion of the antibody used in forming each monomeric fusion protein may be different between the two monomeric units. Any such dimeric combination is specifically contemplated by the methods and compositions of the invention.
In a preferred dimeric fusion protein, the dimer results from the covalent bonding of Cys residue in the CH regions of two of the Ig heavy chains that are the same Cys residues that are disulfide linked in dimerized normal Ig heavy chains.
Still another embodiment provides a tetramer construct having a BirA substrate fused to the extracellular domain of a variant B7-DC polypeptide. Methods for making tetramer constructs are known in the art (see Pertovas, et al., J. Exp. Med., 203:2281 (2006)). 7. Anti-PD-1 and Other Antibodies
Other PD-1 antagonists contemplated by the methods of this invention include antibodies that bind to PD-1 or ligands of PD-1 , and other antibodies.
In one aspect, the present invention relates to a method of increasing a
T cell response in a mammal in need thereof, comprising administering to said mammal an effective treatment regimen comprising an anti-PD-1 antibody and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal to said antigen.
Anti-PD-1 antibodies useful in the treatment regimens(s) of the invention include, but are not limited to, those described in the following publications:
PCT/IL03/00425 (Hardy et al., WO/2003/099196)
PCT/JP2006/309606 (Korman et al., WO/2006/121168)
PCT/US2008/008925 (Li et al., WO/2009/014708)
PCT/JP03/08420 (Honjo et al., WO/2004/004771) PCT/JP04/00549 (Honjo et al., WO/2004/072286)
PCT/IB2003/006304 (Collins et al., WO/2004/056875)
PCT/US2007/088851 (Ahmed et al., WO/2008/083174)
PCT/US2006/026046 (Korman et al., WO/2007/005874)
PCT/US2008/084923 (Terrett et al., WO/2009/073533) Berger et al., Clin. Cancer Res., Vol. 14, pp. 30443051 (2008).
A specific example of an anti-PD-1 antibody useful in the methods of the invention is MDX-1106 (see Kosak, US 20070166281 (pub. 19 July 2007) at par. 42), a human anti-PD-1 antibody, preferably administered at a dose of 3 mg/kg.
In another aspect, the present invention relates to a method of increasing a T cell response in a mammal in need thereof, comprising administering to said mammal an effective treatment regimen comprising an anti-PD-1 ligand antibody, an anti-B7-H1 antibody for example, and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal to said antigen.
Anti-B7-H1 antibodies useful in the treatment regimens(s) of the invention include, but are not limited to, those described in the following publications:
PCT/US06/022423 (WO/2006/133396, pub. 14 December 2006) PCT/US07/088851 (WO/2008/083174, pub. 10 July 2008) US 2006/0110383 (pub. 25 May 2006)
A specific example of an anti-B7-H1 antibody useful in the methods of the invention is MDX-1105 (WO/2007/005874, published 11 January 2007)), a human anti-B7-H1 antibody.
For anti-B7-DC antibodies see 7,411 ,051 , 7,052,694, 7,390,888, 20060099203
Another embodiment of the invention includes a bi-specific antibody that comprises an antibody that binds to the PD-1 receptor bridged to an antibody that binds to a ligand of PD-1 , such as B7-H1. In a preferred embodiment, the PD-1 binding portion reduces or inhibits signal transduction through the PD-1 receptor.
The antibody for use in the invention need not be an anti-PD-1 or anti- PD-1 ligand antibody but may be another antibody useful in mediating the effects of T cells in an immune response. In this aspect, the present invention relates to a method of increasing a T cell response to an antigen in a mammal in need thereof, comprising administering to said mammal an effective treatment regimen comprising an anti-CTLA4 antibody and a potentiating agent, wherein said treatment regimen is effective to increase the T cell response of said mammal to said antigen. An example of an anti-CTLA4 antibody contemplated for use in the methods of the invention includes an antibody as described in PCT/US2006/043690 (Fischkoff et al., WO/2007/056539).
Specific examples of an anti-CTLA4 antibody useful in the methods of the invention are Ipilimumab, also known as MDX-010 or MDX-101 , a human anti- CTLA4 antibody, preferably administered at a dose of 10 mg/kg, and Tremelimumab a human anti-CTLA4 antibody, preferably administered at a dose of 15 mg/kg.
8. Small Molecule PD-1 Antagonists
The PD-1 receptor antagonists can also be small molecule antagonists. The term "small molecule" refers to small organic compounds having a molecular weight of more than 100 and less than about 2,500 daltons, preferably between 100 and 2000, more preferably between about 100 and about 1250, more preferably between about 100 and about 1000, more preferably between about 100 and about 750, more preferably between about 200 and about 500 daltons. The small molecules often include cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more functional groups. The small molecule antagonists reduce or interfere with PD-1 receptor signal transduction by binding to ligands of PD-1 such as B7-H1 and B7-DC and preventing the ligand from interacting with PD-1 or by binding directly to and blocking the PD- 1 receptor without triggering signal transduction through the PD-1 receptor.
In one embodiment, such a small molecule may be administered in combination with another PD-1 antagonist or CTLA4 antagonist, such as an antibody specific for PD-1 or one of its ligands or an antibody specific for
CTLA4 or one of its ligands. Thus, such small molecules may be administered as compounds in one or more of the methods of the invention or may be administered in combination with other compounds useful in the methods of the invention. For example, a series of small organic compounds have been shown to bind to the B7-1 ligand to prevent binding to CTLA4 (see Erbe et al., J. Biol. Chem., Vol. 277, pp. 7363-7368 (2002). Such small organics could be administered alone or together with an anti-CTLA4 antibody, in combination with CTX administration, to reduce inhibitory signal transduction of T cells.
In one embodiment, PD-1 antagonists or CTLA4 antagonists contemplated for use in the methods of the invention include anti-sense nucleic acids, both DNA and RNA, as well as siRNA molecules. Such anti- sense molecules prevent expression of PD-1 on T cells as well as production of T cell ligands, such as B7-H1 , PD-L1 and PD-L2. For example, siRNA (for example, of about 21 nucleotides in length, which is specific for the gene encoding PD-1 , or encoding a PD-1 ligand, and which oligonucleotides can be readily purchased commercially) complexed with carriers, such as polyethyleneimine (see Cubillos-Ruiz et al., J. CHn. Invest. 119(8): 2231-2244 (2009), are readily taken up by cells that express PD-1 as well as ligands of PD-1 and reduce expression of these receptors and ligands to achieve a decrease in inhibitory signal transduction in T cells, thereby activating T cells.
B. POTENTIATING AGENTS
In accordance with the invention, the activity of the PD-1 antagonist is increased, preferably synergistically, by the presence of a potentiating agent. The potentiating agent acts to increase the efficacy of the PD-1 receptor antagonist, possibly by more than one mechanism, although the precise mechanism of action is not essential to the broad practice of the present invention.
In the preferred embodiment, the potentiating agent is cyclophosphamide. Cyclophosphamide (CTX, Cytoxan®, or Neosar®) is an oxazahosphorine drug and analogs include ifosfamide (IFO, Ifex), perfosfamide, trophosphamide (trofosfamide; Ixoten), and pharmaceutically acceptable salts, solvates, prodrugs and metabolites thereof (US patent application 20070202077 which is incorporated in its entirety), lfosfamide (MITOXANA®) is a structural analog of cyclophosphamide and its mechanism of action is considered to be identical or substantially similar to that of cyclophosphamide. Perfosfamide (4-hydroperoxycyclophosphamide) and trophosphamide are also alkylating agents, which are structurally related to cyclophosphamide. For example, perfosfamide alkylates DNA, thereby inhibiting DNA replication and RNA and protein synthesis. New oxazaphosphorines derivatives have been designed and evaluated with an attempt to improve the selectivity and response with reduced host toxicity (Liang J, Huang M, Duan W, Yu XQ, Zhou S. Design of new oxazaphosphorine anticancer drugs. Curr Pharm Des. 2007;13(9):963-78. Review). These include mafosfamide (NSC 345842), glufosfamide (D19575, beta-D-glucosylisophosphoramide mustard), S-(-)-bromofosfamide (CBM-11), NSC 612567 (aldophosphamide perhydrothiazine) and NSC 613060 (aldophosphamide thiazolidine). Mafosfamide is an oxazaphosphorine analog that is a chemically stable 4-thioethane sulfonic acid salt of 4-hydroxy-CPA. Glufosfamide is IFO derivative in which the isophosphoramide mustard, the alkylating metabolite of IFO, is glycosidically linked to a beta-D-glucose molecule. Additional cyclophosphamide analogs are described in US patent 5,190,929 entitled "Cyclophosphamide analogs useful as anti-tumor agents" which is incorporated herein by reference in its entirety.
In other embodiments, the potentiating agent is an agent that reduces activity and/or number of regulatory T lymphocytes (T-regs), preferably Sunitinib (SUTENT®), anti-TGFβ or lmatinib (GLEEVAC®). The recited treatment regimen may also include administering an adjuvant.
Useful potentiating agents also include mitosis inhibitors, such as paclitaxol, aromatase inhibitors (e.g. Letrozole) and angiogenesis inhibitors
(VEGF inhibitors e.g. Avastin, VEGF-Trap) (see, for example, Li et al.,
Vascular endothelial growth factor blockade reduces intratumoral regulatory T cells and enhances the efficacy of a GM-CSF-secreting cancer immunotherapy. Clin Cancer Res. 2006 Nov 15;12(22):6808-16.), anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
C. PHARMACEUTICAL COMPOSITIONS
In one aspect, the invention relates to a therapeutic composition, comprising a molecule that prevents inhibitory signal transduction through PD- 1 , or a CTLA4 antagonist, and a potentiating agent in a pharmaceutically acceptable carrier. The components of said composition are present in an amount effective to increase a T cell response in a mammal. In specific embodiments, the potentiating agent is cyclophosphamide or an analog of cyclophosphamide, examples of such analogs having been recited above.
In other specific examples, the potentiating agent is an agent that reduces activity of regulatory T lymphocytes (T-regs), preferably where the activity is reduced due to a decrease in the number of said T-regs. In preferred non-limiting embodiments, the agent is Sunitinib (SUTENT®), anti- TGFβ or lmatinib (GLEEVAC®).
The potentiating agent useful in formulating compositions of the invention also include mitosis inhibitors, such as paclitaxol, aromatase inhibitors (e.g. Letrozole), agniogenesis inhibitors (VEGF inhibitors e.g. Avastin, VEGF-Trap), anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
A therapeutic composition of the invention also optionally comprises at least one additional agent that may be one or more of an anti-PD-1 antibody, an anti-CTLA4 antibody, a mitosis inhibitor, an aromatase inhibitor, an A2a adenosine receptor (A2AR) antagonist, or an angiogenesis inhibitor. Any of the therapeutic compositions of the invention may also contain one or more adjuvants as described herein.
A PD-1 antagonist useful as a component of a therapeutic composition of the invention includes any of the PD-1 antagonists recited herein for use in any of the methods of the invention. For example, such PD-1 antagonist includes any of the fusion proteins recited herein. Such antagonist can also be any of the polypeptides or PD-1 binding fragments recited herein for use as the first polypeptide portion of any of the fusion proteins described for use in any of the methods of the invention. Such antagonist can further be an antibody, such as any of the known anti-PD-1 , -B7-DC or -B7-H1 antibodies mentioned herein.
A therapeutic composition of the invention also includes, in addition to or in place of the aforementioned PD-1 antagonist, an anti-CTLA4 antibody. Such a composition would therefore contain such an anti-CTLA4 antibody and a potentiating agent of the kind already described herein.
A therapeutic composition of the invention finds use in any of the methods of the invention disclosed herein. Such composition, while intended for use as an active treatment of a disease condition, may also find use as prophylactic compositions to prevent any of the diseases recited herein.
In one aspect, the present invention contemplates a therapeuitc composition comprising a PD-1 antagonist and a potentiating agent in a pharmaceutically acceptable carrier, wherein the PD-1 antagonist and the potentiating agent are together present in an amount effective to increase a T cell response in a mammal.
Therapeutic compositions within the scope of the invention include compositions comprising any and all combinations of the PD-1 antagonists and/or antibodies disclosed herein with any of the recited potentiating agents. By way of non-limiting examples, a therapeutic composition of the invention includes a composition comprising an effective amount of one or more PD-1 antagonists, such as a combination of any or all of the full length polypeptides enumerated herein as specific SEQ ID NOs. or homologs thereof together with one or more fragments of any of said polypeptides, including where any or all of these are fused to other proteins, such as being fused to one or more immunoglobulins recited herein, or not so fused, and comprising one or more potentiating agents, such as cyclophosphamide alone, or cyclophosphamide plus one or more analogs thereof, of just one or more analogs of cyclophosphamide, or the potentiating agent may consist of cyclophosphamide and an agent that reduces T reg number in a mammal receiving the composition, or may consist of a cyclophosphamide analog plus an agent that reduces T reg number or the potentiating agent may consist only of one or more agents that reduce T reg number or other Treg activity. All such combinations are contemplated by the invention so long as the composition comprises at least one PD-1 anatgonist and/or antibody mediating T cell activity and at least one potentiating agent.
The compositions of the invention may also include additional active agents. In preferred embodiments of any of the compositions of the invention, the pharmaceutical or therapeutic composition further comprises at least one additional agent selected from the group consisting of an anti-PD-1 antibody, an anti-CTLA4 antibody, a mitosis inhibitor, such as paclitaxel, an aromatase inhibitor, such as letrozole, an A2AR antagonist, an angiogenesis inhibitor, anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
The PD-1 antagonist and/or potentiating agent may be administered by any suitable means. In a preferred embodiment, the PD-1 antagonist and/or potentiating agent is administered in an aqueous solution, by parenteral injection. The formulation may also be in the form of a suspension or emulsion. In general, pharmaceutical compositions are provided including effective amounts of a peptide or polypeptide, and optionally include pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers. Such compositions include diluents sterile water, buffered saline of various buffer content (e.g., Tris-HCI, acetate, phosphate), pH and ionic strength; and optionally, additives such as detergents and solubilizing agents (e.g., TWEEN 20, TWEEN 80, Polysorbate 80), anti- oxidants (e.g., ascorbic acid, sodium metabisulfite), and preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol). Examples of non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate. The formulations may be lyophilized and redissolved/resuspended immediately before use. The formulation may be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions.
Pharmaceutical compositions of the invention may be administered by parenteral (intramuscular, intraperitoneal, intravenous (IV) or subcutaneous injection), transdermal (either passively or using iontophoresis or electroporation), or transmucosal (nasal, vaginal, rectal, or sublingual) routes of administration. The methods of the invention do not preclude administering the PD-1 antagonist and the potentiating agent by separate and different routes (e.g. topically).
The PD-1 antagonist and the potentiating agent may be administered at the same time, or at different times, with the potentiating agent being administered before or after the PD-1 antagonist. In one embodiment, a potentiating agent is administered both before and after the PD-1 antagonist. In one such embodiment, the same potentiating agent is administered before and after the PD-1 antagonist. In another embodiment, the potentiating agent administered before the PD-1 antagonist.
As used herein the term "effective amount" or "therapeutically effective amount" means a dosage sufficient to treat, inhibit, or alleviate one or more symptoms of the disorder being treated or to otherwise provide a desired pharmacologic and/or physiologic effect. The precise dosage will vary according to a variety of factors such as subject-dependent variables (e.g., age, immune system health, etc.), the disease, and the treatment being effected. Therapeutically effective amounts of PD-1 receptor antagonists and/or antibodies together with a potentiating agents cause an immune response to be activated or sustained.
The selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment desired. Generally dosage levels of 0.001 to 50 mg/kg of body weight daily are administered to mammals. Preferrably, said dose is 1 to 50 mg/kg, more preferably 1 to 40 mg/kg, or even 1 to 30 mg/kg, with a dose of 2 to 20 mg/kg being also a preferred dose. Examples of other dosages include 2 to 15 mg/kg, or 2 to 10 mg/kg or even 3 to 5 mg/kg, with a dose of about 4 mg/kg being a specific example.
For treatment regimens using a potentiating agent and an antibody, such as an anti-PD-1 antibody or an anti-CTLA4 antibody, dosages are commonly in the range of 0.1 to 100 mg/kg, with shorter ranges of 1 to 50 mg/kg preferred and ranges of 10 to 20 mg/kg being more preferred. An appropriate dose for a human subject is between 5 and 15 mg/kg, with 10 mg/kg of antibody (for example, human anti-PD-1 antibody, like MDX-1106) most preferred (plus a suitable dose of cyclophosphamide or other potentiating agent given up to about 24 hours before the antibody).
In general, by way of example only, dosage forms based on body weight for any of the signal transduction antagonists useful in the methods of the invention include doses in the range of 5-300 mg/kg, or 5-290 mg/kg, or 5- 280 mg/kg, or 5-270 mg/kg, or 5-260 mg/kg, or 5-250 mg/kg, or 5-240 mg/kg, or 5-230 mg/kg, or 5-220 mg/kg, or 5-210 mg/kg, or 20 to 180 mg/kg, or 30 to 170 mg/kg, or 40 to 160 mg/kg, or 50 to 150 mg/kg, or 60 to 140 mg/kg, or 70 to 130 mg/kg, or 80 to 120 mg/kg, or 90 to 110 mg/kg, or 95 to 105 mg/kg, with doses of 3 mg/kg, 5 mg/kg, 7 mg/kg, 10 mg/kg, 15 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 50 mg/kg and 100 mg/kg being specific examples of preferred doses. Such doses may, of course, be repeated. The dose will, of course, be correlated with the identity of the mammal receiving said dose. Doses in the above-recited mg/kg ranges are convenient for mammals, including rodents, such as mice and rats, and primates, especially humans, with doses of about 5 mg/kg, about 10 mg/kg and about 15 mg/kg being especially preferred for treating humans.
In accordance with the treatment regimen of the invention, the potentiating agent, for example cyclophosphamide, is administered in nontoxic doses that vary depending on the animal. In specific embodiments, the potentiating agent is administered by any suitable means of administration, including parenteral or oral, the former including system administration, such as intravenous. For example, a potentiating agent like cyclophosphamide is normally administered orally. Such administration may be at any convenient dosage, depending on the potentiating agent. The dosage in each case may be based on body weight or may be administered as a unit dosage.
While CTX itself is nontoxic, some of its metabolites are cytotoxic alkylating agents that induce DNA crosslinking and, at higher doses, strand breaks. Many cells are resistant to CTX because they express high levels of the detoxifying enzyme aldehyde dehydrogenase (ALDH). CTX targets proliferating lymphocytes, as lymphocytes (but not hematopoietic stem cells) express only low levels of ALDH, and cycling cells are most sensitive to DNA alkylation agents.
Low doses of CTX (< 200 mg/kg) can have immune stimulatory effects, including stimulation of anti-tumor immune responses in humans and mouse models of cancer (Brode & Cooke Crit Rev. Immunol. 28:109-126 (2008)). These low doses are sub-therapeutic and do not have a direct anti-tumor activity. In contrast, high doses of CTX inhibit the anti-tumor response. Several mechanisms may explain the role of CTX in potentiation of anti-tumor immune response: (a) depletion of CD4+CD25+FoxP3+ Treg (and specifically proliferating Treg, which may be especially suppressive), (b) depletion of B lymphocytes; (c) induction of nitric oxide (NO), resulting in suppression of tumor cell growth; (d) mobilization and expansion of CD11b+Gr-1+ MDSC. These primary effects have numerous secondary effects; for example following Treg depletion macrophages produce more IFN-γ and less IL-10. CTX has also been shown to induce type I IFN expression and promote homeostatic proliferation of lymphocytes.
Treg depletion is most often cited as the mechanism by which CTX potentiates the anti-tumor immune response. This conclusion is based in part by the results of adoptive transfer experiments. In the AB1-HA tumor model,
CTX treatment at Day 9 gives a 75% cure rate. Transfer of purified Treg at
Day 12 almost completely inhibited the CTX response (van der Most et al.
Cancer Immunol. Immunother. 58:1219-1228 (2009). A similar result was observed in the HHD2 tumor model: adoptive transfer of CD4+CD25+ Treg after CTX pretreatment eliminated therapeutic response to vaccine (Taieb.J.
J. Immunol. 176:2722-2729 (2006)).
Numerous human clinical trials have demonstrated that low dose CTX is a safe, well-tolerated, and effective agent for promoting anti-tumor immune responses (Bas, & Mastrangelo Cancer Immunol. Immunother. 47:1-12 (1998)).
The optimal dose for CTX to potentiate an anti-tumor immune response, is one that lowers overall T cell counts by lowering Treg levels below the normal range but is subtherapeutic (see Machiels et al. Cancer Res. 61 :3689-3697 (2001)).
In human clinical trials where CTX has been used as an immunopotentiating agent, a dose of 300 mg/m2 has usually been used. For an average male (6 ft, 170 pound (78 kg) with a body surface area of 1.98 m2), 300 mg/m2 is 8 mg/kg, or 624 mg of total protein. In mouse models of cancer, efficacy has been seen at doses ranging from 15 - 150 mg/kg, which relates to 0.45 - 4.5 mg of total protein in a 3Og mouse (Machiels et al. Cancer Res. 61 :3689-3697 (2001), Hengst et al Cancer Res. 41 :2163-2167 (1981), Hengst Cancer Res. 40:2135-2141 (1980)).
For larger mammals, such as a primate, preferably human, patient, such mg/m2 doses may be used but unit doses administered over a finite time interval may be preferred. Such unit doses may be administered on a daily basis for a finite time period, such as up to 3 days, or up to 5 days, or up to 7 days, or up to 10 days, or up to 15 days or up to 20 days or up to 25 days, are all specifically contemplated by the invention. The same regimen may be applied for the other potentiating agents recited herein.
All such administrations may occur before or after administration of a PD-1 binding molecule of the invention. Alternatively, administration of one or more doses of a PD-1 binding molecule of the invention may be temprally staggered with the administration of potentiating agent to form a uniform or non-uniform course of treatment whereby one or more doses of potentiating agent are administered, followed by one or more doses of a PD-1 binding compound, followed by one or more doses of potentiating agent, all according to whatever schedule is selected or desired by the researcher or clinician administering said agents.
In other specific embodiments, the treatment regimen includes multiple administrations of one or more PD-1 antagonists. In some embodiments, such multiple administrations of PD-1 antagonists are in conjunction with multiple administrations of the same or different potentiating agents.
As in other embodiments of the invention, here the potentiating agent is administered at least 1 , 2, 3, 5, 10, 15, 20, 24 or 30 hours prior to or after administering of the PD-1 -antagonist.
The pharmaceutical compositions useful herein also contain a pharmaceutically acceptable carrier, including any suitable diluent or excipient, which includes any pharmaceutical agent that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which may be administered without undue toxicity. Pharmaceutically acceptable carriers include, but are not limited to, liquids such as water, saline, glycerol and ethanol, and the like, including carriers useful in forming sprays for nasal and other respiratory tract delivery or for delivery to the ophthalmic system. A thorough discussion of pharmaceutically acceptable carriers, diluents, and other excipients is presented in REMINGTON'S PHARMACEUTICAL SCIENCES (Mack Pub. Co., N.J. current edition).
Vaccine compositions (as discussed below) may further incorporate additional substances to stabilize pH, or to function as adjuvants, wetting agents, or emulsifying agents, which can serve to improve the effectiveness of the vaccine.
Vaccines are generally formulated for parenteral administration and are injected either subcutaneously or intramuscularly. Such vaccines can also be formulated as suppositories or for oral administration, using methods known in the art, or for administration through nasal or respiratory routes.
D. METHODS OF MANUFACTURE
Isolated PD-1 antagonist polypeptides, including variants, homologs and fragments thereof, either wild-type or mutated, and fusion proteins comprising any of these, all contemplated for use in the invention, can be obtained by, for example, chemical synthesis or by recombinant production in a host cell. To recombinantly produce a costimulatory polypeptide, a nucleic acid containing a nucleotide sequence encoding the polypeptide can be used to transform, transduce, or transfect a bacterial or eukaryotic host cell (e.g., an insect, yeast, or mammalian cell). It will be appreciated that the nucleotide sequences can be codon-optimized to increase levels of protein expression in a particular kind of host cell. Methods for codon optimization are well known in the art. In general, nucleic acid constructs include a regulatory sequence operably linked to a nucleotide sequence encoding a costimulatory polypeptide. Regulatory sequences (also referred to herein as expression control sequences) typically do not encode a gene product, but instead affect the expression of the nucleic acid sequences to which they are operably linked. The signal peptides used to secrete proteins from a cell can be the endogenous signal peptides or any other signal peptide that facilitates secretion of the fusion protein from a host.
For general molecular biology procedures useful in practicing the present invention, a number of standard references are available that contain procedures well known in the art of molecular biology and genetic engineering and which procedures need not be further described herein. Useful references include Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), Wu et al, Methods in Gene Biotechnology (CRC Press, New York, NY, 1997), and Recombinant Gene Expression Protocols, in Methods in Molecular Biology, Vol. 62, (Tuan, ed., Humana Press, Totowa, NJ, 1997), the disclosures of which are hereby incorporated by reference.
E. DISEΞASE TREATMENT
Diseases to be treated or prevented by administering a therapeutic combination provided by the present invention include a malignant tumor or a chronic infectious disease caused by a bacterium, virus, protozoan, helminth, or other microbial pathogen that enters intracellular^. Such diseases are often combatted through attack by cytotoxic T lymphocytes. Because the present invention provides combination therapes useful in enhancing T cell responses, through increased T cell activity, increased T cell proliferation and reduced T cell inhibitory signals, the combination therapies of the invention have unique advantage in treating (or even preventing) such diseases. In one embodiment, because viral infections are cleared primarily by T- cells, an increase in T-cell activity is therapeutically useful in enhancing clearance of an infective viral agent from an animal or primate, preferably human, subject. Thus, the disclosed compounds of the invention, with PD-1 receptor antagonist activity, together with a potentiating agent work in combination for the treatment of local or systemic viral infections. Infections that are to be treated by the compounds of the invention include, but are not limited to, immunodeficiency (e.g., HIV), papilloma (e.g., HPV), herpes (e.g., HSV), encephalitis, influenza (e.g., human influenza virus A), hepatitis (e.g. HCV, HBV), and common cold (e.g., human rhinovirus) viral infections. Pharmaceutical formulations of PD-1 receptor antagonists compositions can also be administered to treat systemic viral diseases, including, but not limited to, AIDS, influenza, the common cold, or encephalitis.
Non-viral infections treatable by the compounds of the invention include, but are not limited to, infections cause by microoganisms including, but not limited to, Actinomyces, Anabaena, Bacillus, Bacteroides, Bdellovibrio, Bordetella, Borrelia, Campylobacter, Caulobacter, Chlamydia, Chlorobium, Chromatium, Clostridium, Corynebacterium, Cytophaga, Deinococcus, Escherichia, Francisella, Halobacterium, Heliobacter, Haemophilus, Hemophilus influenza type B (HIB), Hyphomicrobium, Legionella, Leptspirosis, Listeria, Meningococcus A, B and C, Methanobacterium, Micrococcus, Myobacterium, Mycoplasma, Myxococcus, Neisseria, Nitrobacter, Oscillatoria, Prochloron, Proteus, Pseudomonas, Phodospirillum, Rickettsia, Salmonella, Shigella, Spirillum, Spirochaeta, Staphylococcus, Streptococcus, Streptomyces, Sulfolobus, Thermoplasma, Thiobacillus, Treponema, Vibrio, Yersinia, Cryptococcus neoformans, Histoplasma sp. (such as Histoplasma capsulatum), Candida albicans, Candida tropicalis, Nocardia asteroides, Rickettsia ricketsii, Rickettsia typhi, Leishmania, Mycoplasma pneumoniae, Chlamydial psittaci, Chlamydial trachomatis, Plasmodium sp. (such as Plasmodium falciparum), Trypanosoma brucei, Entamoeba histolytica, Toxoplasma gondii, Trichomonas vaginalis and Schistosoma mansoni. In one embodiment, the present invention provides methods and compositions for inducing or enhancing an immune response in host for treating cancer. The types of cancer that may be treated with the provided compositions and methods include, but are not limited to, the following: bladder, brain, breast, cervical, colo-rectal, esophageal, kidney, liver, lung, nasopharangeal, pancreatic, prostate, skin, stomach, uterine, ovarian testicular, and hematologic cancer.
Malignant tumors which may be treated are classified herein according to the embryonic origin of the tissue from which the tumor is derived. Carcinomas are tumors arising from endodermal or ectodermal tissues such as skin or the epithelial lining of internal organs and glands. Sarcomas, which arise less frequently, are derived from mesodermal connective tissues such as bone, fat, and cartilage. The leukemias and lymphomas are malignant tumors of hematopoietic cells of the bone marrow. Leukemias proliferate as single cells, whereas lymphomas tend to grow as tumor masses. Malignant tumors may show up at numerous organs or tissues of the body to establish a cancer.
As a demonstration of the value of the treatment regimens of the invention, the murine analog of B7-DC-lg (in which the mouse B7-DC ECD, which shares 72% sequence identity with the human protein, is fused to the Fc domain of mouse lgG2a) tested in syngeneic mouse tumor models for colon cancer, mastocytoma, and other tumor types incorporating a cyclophosphamide (CTX) pre-treatment as described herein.
The results showed that treatment with a single subtherapeutic dose of
CTX, which acts as an immunopotentiating agent, followed by murine B7-DC- IG eradicates established CT26 colon carcinoma tumors in up to 80% of the animals. Further, in CT26 colon carcinoma tumor re-challenge studies, no tumor re-growth was detected in mice that had previously eradicated tumor following CTX + murine B7-DC-lg treatment. These mice were also shown to have an increased tumor-specific CTL population relative to naϊve mice.
In one embodiment, the present invention contemplates use of a compound that reduces inhibitory signal transduction in a T cell, as described elsewhere herein, in the manufacture of a medicament for increasing a T cell response by combination therapy wherein said compound is administered in conjunction with a potentiating agent. Further, the compound that reduces inhibitory signal transduction in a T cell and said potentiating agent are provided as separate medicaments for administration at different times, preferably where the potentiating agent is administered prior to the compound that reduces inhibitory signal transduction, for example, up to 24 hours prior to the inhibitory compound (or other time intervals recited herein). Preferably, the compound and potentiating agent are for use in the treatment of an infectious disease or cancer, including diseases caused by any of the infectious agents or cancers recited elsewhere herein.
In a preferred embodiment, a compound useful in these methods is a recombinant protein composed of the ECD of human B7-DC fused to the Fc domain of human IgGi, referred to herein as B7-DC-lg.
In one embodiment, the present invention relates to a medical kit for administering a compound that reduces inhibitory signal transduction in a T cell, as disclosed herein, in combination with a potentiating agent, said kit comprising:
(a) a dosage supply of a compound that reduces inhibitory signal transduction in a T cell,
(b) a supply of a potentiating agent;
(c) a supply of pharmaceutically acceptable carrier; and (d) printed instructions for administering the compound in a use as described above. F. COMBINATION THERAPIES
Vaccines require strong T cell response to eliminate cancer cells and infected cells or infectious agents. PD-1 receptor antagonists described herein can be administered as a component of a vaccine, along with a potentiating agent, to provide a costimulatory signal to T cells. Vaccines disclosed herein include antigens, a PD-1 receptor antagonist and optionally adjuvants and targeting molecules.
The antigens against which the T cell response is enhanced by the methods and composition of the invention includes peptides, proteins, polysaccharides, saccharides, lipids, nucleic acids, or combinations thereof. The antigens, in the case of disease, are present due to the disease process.
The disclosed PD-1 receptor antagonists compositions may be administered in conjunction with prophylactic vaccines, which confer resistance in a subject to subsequent exposure to infectious agents, or in conjunction with therapeutic vaccines, which can be used to initiate or enhance a subject's immune response to a pre-existing antigen, such as a tumor antigen in a subject with cancer, or a viral antigen in a subject infected with a virus.
The desired outcome of a prophylactic, therapeutic or de-sensitized immune response may vary according to the disease, based on principles well known in the art. For example, an immune response against an infectious agent may completely prevent colonization and replication of an infectious agent, affecting "sterile immunity" and the absence of any disease symptoms. However, a vaccine against infectious agents may be considered effective if it reduces the number, severity or duration of symptoms; if it reduces the number of individuals in a population with symptoms; or reduces the transmission of an infectious agent. Similarly, immune responses against cancer, allergens or infectious agents may completely treat a disease, may alleviate symptoms, or may be one facet in an overall therapeutic intervention against a disease. For example, the stimulation of an immune response against a cancer may be coupled with surgical, chemotherapeutic, radiologic, hormonal and other immunologic approaches in order to affect treatment.
The methods and products of the invention do not preclude use of an adjuvant in addition to the potentiating agent. Such adjuvant may be administered, for example, along with the PD-1 antagonist. The adjuvants useful in the compositions and methods of the invention include, but are not limited to, one or more of the following: oil emulsions (e.g., Freund's adjuvant); saponin formulations; virosomes and viral-like particles; bacterial and microbial derivatives; immunostimulatory oligonucleotides; ADP-ribosylating toxins and detoxified derivatives; alum; BCG; mineral-containing compositions (e.g., mineral salts, such as aluminium salts and calcium salts, hydroxides, phosphates, sulfates, etc.); bioadhesives and/or mucoadhesives; microparticles; liposomes; polyoxyethylene ether and polyoxyethylene ester formulations; muramyl peptides; polyphosphazene; imidazoquinolone compounds; and surface active substances (e.g. lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol). Useful adjuvants also include immunomodulators such as cytokines, interleukins (e.g., IL-1 , IL-2, IL-4, IL-5, IL-6, IL-7, IL-12, etc.), interferons (e.g., interferon-γ), macrophage colony stimulating factor, and tumor necrosis factor.
Nothing herein precludes the disclosed PD-1 receptor antagonist, including any of the polypeptides, fragments, variants, homologs and fusion proteins disclosed herein, from being administered to a subject in need thereof in combination with one or more additional therapeutic agents (in addition to the potentiating agent). The additional therapeutic agents are selected based on the condition, disorder or disease to be treated. For example, PD-1 receptor antagonists can be co-administered with one or more additional agents that function to enhance or promote an immune response, and which are considered herein as active agents. Such agents include, but are not limited to, amsacrine, bleomycin, busulfan, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clofarabine, crisantaspase, cytarabine, dacarbazine, dactinomycin, daunorubicin, docetaxel, doxorubicin, epirubicin, etoposide, fludarabine, fluorouracil, gemcitabine, hydroxycarbamide, idarubicin, ifosfamide, irinotecan, leucovorin, liposomal doxorubicin, liposomal daunorubicin, lomustine, melphalan, mercaptopurine, mesna, methotrexate, mitomycin, mitoxantrone, oxaliplatin, paclitaxel, pemetrexed, pentostatin, procarbazine, raltitrexed, satraplatin, streptozocin, tegafur-uracil, temozolomide, teniposide, thiotepa, tioguanine, topotecan, treosulfan, vinblastine, vincristine, vindesine, vinorelbine, or a combination thereof. Representative pro- apoptotic agents include, but are not limited to fludarabinetaurosporine, cycloheximide, actinomycin D, lactosylceramide, 15d-PGJ(2) and combinations thereof.
The therapies provided by the methods and compositions of the present invention may also be used in conjunction with other types of therapies, such as radiation treatments, surgery, and the like.
G. ASSAYS FOR ANTAGONIST ACTIVITY
The present invention recites a number specific structures useful in practicing the methods of the invention. Other compounds possessing antagonist activity and being useful in the methods of the invention may also be identified by reference to well known assay procedures for identifying chemical structures that bind to PD-1 , CTLA4, and ligands of any of these and that also possess the ability to reduce inhibitory signal transduction in T cells. Some such assays are binding assays useful in determining if a selected chemical structure binds to receptors; these are well known in the art and need not be discussed in detail herein (see, for example, U.S. 2008/0274490, (pub. 6 November 2008) and U.S. 7,105,328 (issued 12 September 2006), each showing assays for PD-1 signaling modulators using T cells) the disclosures of which are hereby incorporated by reference in its entirety. Other assays are used to determine the effects of agents of the invention, such as active fragments, to activate T cells by increasing proliferation and/or production of cytokines. Such assays are also well known in the art. For example, increased proliferation of cells can be demonstrated by increased 3H-thymidine incorporation (due to increased DNA synthesis needed for cellular mulitplication) or ELISA and/or RIA for detecting increased production of cytokines by T cells in culture.
In one such experiment, PD-1 binding activity of human B7-DC-lg was assessed by ELISA. 96-well ELISA plates were coated with 100 uL 0.75 ug/mL recombinant human PD-1/Fc (R&D Systems) diluted in BupH Carbonate/Bicarbonate pH 9.4 buffer (Pierce) for 2 hours and then blocked with BSA solution (Jackson ImmunoResearch) for 90-120 minutes. Serially diluted human B7-DC-lg (wild type, as well as D111 S mutein, and K113S mutants that were selected for reduced binding to PD-1 ) as well as human IgGI isotype control were allowed to bind for 90 minutes. Bound B7-DC-lg was detected using 100 uL of 0.5 ug/mL biotin conjugated anti-human B7-DC clone MIH 18 (eBioscience) followed by 1 :1000 diluted HRP-Streptavidin (BD Bioscience) and TMB substrate (BioFX). Absorbance at 450 nm was read using a plate reader (Molecular Devices) and data were analyzed in SoftMax using a 4-parameter logistic fit. The data showed that human B7-DC-lg (wildtype) bound to PD-1 but the K113S and D111 S mutants do not bind to PD-1.
In carrying out the procedures of the present invention it is of course to be understood that reference to particular buffers, media, reagents, cells, culture conditions and the like are not intended to be limiting, but are to be read so as to include all related materials that one of ordinary skill in the art would recognize as being of interest or value in the particular context in which that discussion is presented. For example, it is often possible to substitute one buffer system or culture medium for another and still achieve similar, if not identical, results. Those of skill in the art will have sufficient knowledge of such systems and methodologies so as to be able, without undue experimentation, to make such substitutions as will optimally serve their purposes in using the methods and procedures disclosed herein.
The invention is described in more detail in the following non-limiting examples. It is to be understood that these methods and examples in no way limit the invention to the embodiments described herein and that other embodiments and uses will no doubt suggest themselves to those skilled in the art.
Examples
Example 1
B7-DC-lg binds to PD01 expressing CHO cells
B7-DC-lg was first conjugated with allophycocyanin (APC) and then incubated at various concentrations with a CHO cell line constitutively expressing PD-1 or parent CHO cells that do not express PD-1. Binding was analyzed by flow cytometry. Figure 1 shows the median fluorescence intensity
(MFI) of B7-DC-lg-APC as a function of the concentration of probe (x-axis).
B7-DC-lg-APC binds to CHO. PD-1 cells (solid circle) but not untransfected CHO cells (gray triangle).
Example 2 B7-DC-lg competes with B7-H1 for binding to PD-1.
B7-H1-lg was first conjugated with allophycocyanin (APC). Unlabeled
B7-DC-lg at various concentrations was first incubated with a CHO cell line constitutively expressing PD-1 before adding B7-H1 -Ig-APC to the probe and cell mixture. Figure 2 shows the median fluorescence intensity (MFI) of B7- HI-Fc-APC is shown as a function of the concentration of unlabeled B7-DC-lg competitor (x-axis) added. As the concentration of unlabeled B7-DC-lg is increased the amount of B7-H1 -Ig-APC bound to CHO cells decreases, demonstrating that B7-DC-lg competes with B7-H1 for binding to PD-1.
Example 3 CT26 Tumor Model
Mouse colorectal tumor cell line, CT26, was obtained from ATCC. A master cell bank at Passage 4 was generated following ATCC guidelines. Cells were tested and confirmed no mycoplasma and other pathogen contamination. One vial of tumor cells was thawed from the cryopreserved stocks and grown for two passages prior to inoculation.
CT26 cells were split at 1 :5 dilution with 30 ml_ complete medium (RPMI + 10% FBS, 2 mM L-GIu1 and 1x P/S) for two days culture or at
1 :10 dilution with 30 ml complete medium for 3 days culture.
CT26 cells were harvested by aspirating medium, rinsing the flask with
5 ml_ PBS, adding 5 ml_ trypsin, incubating at 37 0C for 2 min, and then neutralizing with 10 ml_ complete medium. After centrifuge at 600 x g (~1000 rpm) for 5 min, media was sspirateed and the cell pellet was resuspended by pipetting with 10 ml plain RPMI. This wash step was repeated for three times.
Cell number and viability of the inoculated cells were analyzed by trypan blue dye staining with proper dilution (e.g. 1 :5 dilution, 10 μL cells + 40 μL trypan blue) and confirmed by NOVA cell count during the last wash step. Cell viability generally was greater than 95% for inoculation.
CT26 cells were diluted to 6.7x105 cells/mL for initial inoculation with plain RPMI and stored on ice. Typically each mouse was inoculated with 150 μL (1 x105 cells). On Day 9, all the tumor-bearing mice were first grouped into a rat cage and randomly divided the mice to experimental groups. CTX solution was reconstituted by 1x PBS to 4 mg/mL Mice were intraperitoneal^ (IP) injected with 0.5 ml_ of CTX solution resulting in 2 mg for a 20 gram mouse, i.e. 100 mg/kg.
On Day 10, mice were IP injected with 0.5 ml_ of B7-DC-lg (0.2 mg/mL) resulting in 0.1 mg for a 20 gram mouse, i.e. 5 mg/kg. The same dose was given 2 time a week for 4 weeks, total 8 doses. Tumor growth were monitored by measuring the tumor twice weekly, starting on the day when giving B7-DC-lg via a digital caliper. Tumor volume was calculated as following:
Tumor volume = π(Dshort)2 * (D|Ong)/6= -0.52 x (DshOrt)2 * (D|Ong)
Mice were euthanized and taken off the study if the tumor volume reached 2000 mm3 or if there were skin ulcers and infections at the tumor inoculation site.
Example 4 Combination of cyclophosphamide and B7-DC-lg can eradicate established tumors.
Balb/C mice at age of 9 to 11 weeks were implanted subcutaneously with 1.0 x 105 CT26 colorectal tumor cells as described above. On day 10 post tumor implantation, mice received 100 mg/kg of cyclophosphamide. B7- DC-Ig treatment started 1 day later, on day 11. Mice were treated with 100 ug of B7-DC-lg, 2 doses per week, for 4 weeks and total 8 doses. 75% of the mice that received the CTX + B7-DC-lg treatment regimen eradicated the established tumors by Day 44, whereas all mice in the control CTX alone group died as a result of tumor growth or were euthanized because tumors exceeded the sizes approved by IACUC (results shown in Figure 3). These results demonstrate the effectiveness of the treatment regimen on established tumors and not mere prophylaxis. Example 5
Combination of cyclophosphamide and B7-DC-lg can eradicate established tumors and protect against tumor re-challenge.
Mice eradicated established CT26 colorectal tumors from the above described experiment were rechallenged with 1x105 CT26 cells on Day 44 and Day 70. No tumors grew out from the rechallenge suggesting they had developed long term anti-tumor immunity from the cyclophosphamide and B7- DC-Ig combination treatment. All mice in the vehicle control group developed tumors (results shown in Figure 4). These results show the effectiveness of the treatment regimen on established tumors and that the cyclophosphamide and B7-DCIg combination treatment resulted in memory responses to tumor antigens.
Example 6
Combination of cyclophosphamide and B7-DC-lg can generate tumor specific, memory cytotoxic T lymphocytes Mice eradiated established CT26 colorectal tumors from the above described experiment were rechallenged with 2.5x105 CT26 cells on Day 44. Seven days later, mouse spleens were isolated. Mouse splenocytes were pulsed with 5 or 50 ug/ml_ of ovalbumin (OVA) or AH 1 peptides for 6 hours in the presence of a Golgi blocker (BD BioScience). Memory T effector cells were analyzed by assessing CD8+/IFNγ+ T cells. Results in Figure 5 show that there were significant amount of CT26 specific T effector cells in the CT26 tumor-eradicated mice.
Example 7 Effect of B7-DC-lg dose dependent on tumor eradication
Balb/C mice at age of 9 to 11 weeks were implanted subcutaneously with 1.0 x 105 CT26 colorectal tumor cells. On day 9 post tumor implantation, mice received a single dose of cyclophosphamide (100 mg/kg) and started treatment on Day 10 with 30, 100 or 300 μg of B7-DC-lg, 2 doses per week for 4 weeks, total 8 doses. Figure 6 shows there were 70% of the mice eradicated the tumors at 300 μg, 40% tumor eradication with 100 μg, and 30 μg dose gave rise to 10% tumor eradication.
Example 8
Combination of cyclophosphamide and anti-PD-1 can eradicate established tumors.
Balb/C mice at age of 9 to 11 weeks were challenged subcutaneously with 1.0 x 105 CT26 colorectal tumor cells. On day 11 post tumor challenge, mice received a single dose of cyclophosphamide (100 mg/kg) and started treatment with anti-PD-1 antibody (250 ug, Clone G4, Hirano F. et al., 2005 Cancer Research) which was administered 3 times per week for four weeks. 70% of the mice that received the CTX + anti-PD-1 regimen eradicated established CT26 tumors at day 50 after tumor challenge, whereas all mice in the control and anti-PD-1 alone groups died as a result of tumor growth or were euthanized because tumors exceeded the sizes approved by IACUC. These results show the effectiveness of the treatment regimen on established tumors and not mere prophylaxis. Results are shown in Figure 7.
Example 9
Combination of cyclophosphamide and anti-CTLA4 can eradicate established tumors.
Balb/C mice at age of 9 to 11 weeks were challenged subcutaneously with 1.0 x 105 CT26 colorectal tumor cells. On day 11 post tumor challenge, mice received 100 mg/kg of cyclophosphamide. Anti-CTLA4 (an anti-mouse CTLA4 from hamster hybridoma - ATCC deposit UC10-4F10-11) treatment was started 1 day later, on day 12. Mice were treated with 100 ug of anti- CTLA4, 2 doses per week, for 4 weeks. 56% of the mice that received the CTX + anti-CTLA4 regimen were tumor free at day 50 after tumor challenge, whereas all mice in the control group died as a result of tumor growth or were euthanized because tumors exceeded the sizes approved by IACUC. Results are shown in Figure 8. These results show the effectiveness of the treatment regimen on established tumors and not mere prophylaxis.
Example 10
Combination of cyclophosphamide and B7-DC-lg Regimen Leads to Reduction of Tregs in the Tumor Microenvironment
Figure 9 shows the results of experiments wherein Balb/C mice at age of 9 to 11 weeks of age were implanted with 1 X 105 CT26 cells subcutaneously. On Day 9, mice were injected with 100 mg/kg of CTX, IP. Twenty four hours later, on Day 10, mice were treated with 100 ug of B7-DC- Ig. There were 5 groups: naϊve mice that did not receive any tumor cells, vehicle injected, CTX alone, CTX + B7-DC-lg or B7-DC-lg alone. Two naive mice and 4 mice from other groups were removed from the study on Day 11 (2 days post CTX) and Day 16 (7 days post CTX) for T cell analysis. Left panel shows on Day 11 , 2 days post CTX injection, Treg in the spleen of the mice with CTX treatment was significantly lower than the one in the mice with tumor implantation and injected with vehicle. Right panel shows that on Day 16, 7 days post CTX and 6 days post B7-DC-lg treatment, B7-DC-lg significantly lowered the CD4+ T cells expressing high PD-1. This was observed in both the B7-DC-lg treated and CTX + B7-DC-lg treated mice. Mice implanted with tumor cells intended to have more PD-1+/CD4+ T cells in the draining LN compared with naive mice.
Example 11 Combination of cyclophosphamide and B7-DC-lg can promote mouse survival in a metastatic prostate tumor model B10.D2 mice at age of 9 to 11 weeks were injected intravenously with 3.0 x 105 SP-1 cells, which were isolated from lung metastasis post parent TRAMP cell injection. The CTX mice received 3 doses of CTX, 50 mg/kg, on Day 5, 12 and 19. The B7-DC-lg treated mice received 3 doses of B7-DC-lg, 5 mg/kg, on Day 6, 13 and 20. On Day 100, 17% of mice in the control groups, no-treated, CTX alone, B7-DC-lg alone survived while 43% of the mice received combination of CTX and B7-DC-lg survived. Results are shown in Figure 10.
Example 12
Combination of Listeria cancer vaccine and B7-DC-lg can enhance mouse survival post CT26 liver implantation
Balb/C mice at age of 1 1-13 weeks were implanted with CT26 cells using a hemispleen injection technique (Yoshimura K et al., 2007, Cancer
Research). On Day 10, mice received 1 injection of CTX at 50 mg/kg, IP.
Twenty four hours later, on Day 1 1 , mice were treated with recombinant
Listeria carrying AH1 peptide, an immunodominant epitope of CT26, at 0.1
LD50 (1x107 CFU), then on Day 14 and 17. Mice were also treated with B7- DC-Ig on Day 1 1 and then on Day 18. Figire 1 1 shows mice without any treatment or treated with CTX and Listeria cancer vaccine all died before
Dady 45. There were 60% of the mice received triple combination, CTX +
Listeria cancer vaccine and B7-DC-lg survived.
1. Reference List 1 . Brode S, Cooke A. Immune-potentiating effects of the chemotherapeutic drug cyclophosphamide. Crit Rev.lmmunol. 2008;28(2): 109-26
2. van der Most RG, Currie AJ, Mahendran S, Prosser A, Darabi A, Robinson BW, Nowak AK, Lake RA. Tumor eradication after cyclophosphamide depends on concurrent depletion of regulatory T 9e'!s.: a r0'er cycling TNFR2-expressing effector-suppressor T cells in limiting effective chemotherapy. Cancer Immunol. Immunother. 2009 Aug;58(8): 1219-28
3. Taieb J, Chaput N, Schartz N1 Roux S, Novault S, Menard C, Ghiringhelli F, Terme M, Carpentier AF, Darrasse-Jeze G, et al. Chemoimmunotherapy of tumors: cyclophosphamide synergizes with exosome based vaccines. J.Immunol. 2006 Mar 1 ; 176(5):2722-9 4. Machiels JP, Reilly RT, Emens LA1 Ercolini AM, Lei RY, Weintraub D, Okoye Fl, Jaffee EM. Cyclophosphamide, doxorubicin, and paclitaxel enhance the antitumor immune response of granulocyte/macrophage- colony stimulating factor-secreting whole-cell vaccines in HER-2/neu tolerized mice. Cancer Res. 2001 May 1 ;61 (9):3689-97
5. Bass KK, Mastrangelo MJ. lmmunopotentiation with low-dose cyclophosphamide in the active specific immunotherapy of cancer. Cancer Immunol. Immunother. 1998 Sep;47(1 ):1-12
6. Hengst JC, Mokyr MB1 Dray S. Cooperation between cyclophosphamide tumoricidal activity and host antitumor immunity in the cure of mice bearing large MOPC-315 tumors. Cancer Res. 1981 Jun;41 (6):2163-7
7. Hengst JC1 Mokyr MB, Dray S. Importance of timing in cyclophosphamide therapy of MOPC-315 tumor-bearing mice. Cancer Res. 1980 Jul;40(7):2135-41 8. Tsung K1 Meko JB1 Tsung YL, Peplinski GR, Norton JA. Immune response against large tumors eradicated by treatment with cyclophosphamide and IL-12. J.Immunol. 1998 Feb 1 ;160(3): 1369-77
9. Honeychurch J, Glennie MJ, lllidge TM. Cyclophosphamide inhibition of anti-CD40 monoclonal antibody-based therapy of B cell lymphoma is dependent on CD1 1 b+ cells. Cancer Res. 2005 Aug 15;65(16):7493-501
10. Wada S, Yoshimura K, Hipkiss EL1 Harris TJ1 Yen HR1 Goldberg MV,
Grosso JF, Getnet D, Demarzo AM, Netto GJ, Anders R, Pardoll DM,
Drake CG. Cyclophosphamide augments antitumor immunity: studies in an autochthonous prostate cancer model. Cancer Res. 2009 May 15;69(10):4309-18.
1 1 . Freeman, G. J. Structures of PD-1 with its ligands: sideways and dancing cheek to cheek. Proc. Natl. Acad. Sci. U. S. A 105, 10275-10276 (2008).
12. Brode.S. & Cooke.A. Immune-potentiating effects of the chemotherapeutic drug cyclophosphamide. Crit Rev. Immunol. 28, 109-
126 (2008).
13. van der Most.R.G. et al. Tumor eradication after cyclophosphamide depends on concurrent depletion of regulatory T cells: a role for cycling TNFR2-expressing effector-suppressor T cells in limiting effective chemotherapy. Cancer Immunol. Immunother. 58, 1219-1228 (2009).
14. Taieb.J. et al. Chemoimmunotherapy of tumors: cyclophosphamide synergizes with exosome based vaccines. J. Immunol. 176, 2722-2729 (2006).
15. Bass.K.K. & Mastrangelo, M. J. lmmunopotentiation with low-dose cyclophosphamide in the active specific immunotherapy of cancer. Cancer Immunol. Immunother. 47, 1-12 (1998). 16. Machiels.J.P. et al. Cyclophosphamide, doxorubicin, and paclitaxel enhance the antitumor immune response of granulocyte/macrophage- colony stimulating factor-secreting whole-cell vaccines in HER-2/neu tolerized mice. Cancer Res. 61 , 3689-3697 (2001 ). 17. Hengst.J.C, Mokyr.M.B., & Dray.S. Cooperation between cyclophosphamide tumoricidal activity and host antitumor immunity in the cure of mice bearing large MOPC-315 tumors. Cancer Res. 41 , 2163- 2167 (1981). 18. Hengst.J.C., Mokyr.M.B., & Dray.S. Importance of timing in cyclophosphamide therapy of MOPC-315 tumor-bearing mice. Cancer Res. 40. 2135-2141 (1980).
19. Tsung.K., Meko.J.B., Tsung.Y.L, Peplinski.G.R., & Norton, J.A. Immune response against large tumors eradicated by treatment with cyclophosphamide and 11-12. J. Immunol. 160, 1369-1377 (1998).
20. Honeychurch.J., Glennie.M.J., & lllidge.T.M. Cyclophosphamide inhibition of anti-CD40 monoclonal antibody-based therapy of B cell lymphoma is dependent on CD11b+ cells. Cancer Res. 65, 7493-7501
(2005).
21. Wada S, Yoshimura K, Hipkiss EL, Harris TJ, Yen HR, Goldberg MV, Grosso JF, Getnet D, Demarzo AM, Netto GJ, Anders R, Pardoll DM, Drake CG. Cyclophosphamide augments antitumor immunity: studies in an autochthonous prostate cancer model. Cancer Res. 2009 May 15;69(10):4309-18.

Claims

What is claimed is:
1. A method of increasing a T cell response in a mammal, comprising an effective treatment regimen, said regimen comprising administering to a mammal a compound that reduces inhibitory signal transduction in a T cell and a potentiating agent, wherein said treatment regimen is effective to increase T cell response in said mammal.
2. The method of claim 1 , wherein said compound is a fusion protein comprising first and second peptide portions, wherein said first peptide portion comprises an active fragment of a PD-1 antagonist polypeptide and said second peptide portion comprises a portion of an immunoglobulin (Ig).
3. The method of claim 2, wherein said PD-1 antagonist polypeptide is a wild type B7-DC polypeptide.
4. The method of claim 3, wherein said B7-DC is a human B7-DC.
5. The method of claim 4, wherein said active fragment does not comprise any substantial portion of the transmembrane portion of said B7-DC polypeptide.
6. The method of claim 5, wherein said active fragment comprises the soluble portion of said B7-DC polypeptide and said second peptide portion comprises the Fc region of an antibody but does not comprise any of the variable region of said antibody.
7. The method of claim 5, wherein said active fragment comprises the amino acid sequence of SEQ ID NO: 3 and said second polypeptide portion comprises the Fc region of an antibody but does not comprise any of the variable region of said antibody.
8. The method of claim 4, wherein said first peptide portion consists of an amino acid sequence having at least 80% identity to amino acids 20-221 or 20-121 of SEQ ID NO: 1.
9. The method of claim 4, wherein said first peptide portion consists of the amino acid sequence of amino acids 20-221 or 20-121 of SEQ ID NO: 1.
10. The method of claim 5, wherein said PD-1 binding fragment comprises at least 10 contiguous amino acids, or at least 25 contiguous amino acids, or at least 50 contiguous amino acids, or at least 75 contiguous amino acids, or at least 90 contiguous amino acids, or at least 100 contiguous amino acids of SEQ ID NO: 1.
11. The method of claim 2, wherein said fusion protein comprises an amino acid sequence having at least 80% identity to the sequence of SEQ ID
NO: 9, 10, 12 or 13.
12. The method of claim 2, wherein said fusion protein comprises the amino acid sequence of SEQ ID 9, 10, 12, or 13.
13. The method of claim 2, wherein said fusion protein is a monomer.
14. The method of claim 2, wherein said fusion protein is part of a dimer.
15. The method of claim 14, wherein said dimer is a homodimer.
16. The method of claim 14, wherein said dimer is a heterodimer.
17. The method of claim 1 , wherein said potentiating agent is cyclophosphamide or an analog of cyclophosphamide.
18. The method of claim 1 , wherein said potentiating agent is an agent that reduces activity of regulatory T lymphocytes (T-regs).
19. The method of claim 1 , wherein said potentiating agent is administered before administering said compound.
20. The method of claim 19, wherein said potentiating agent is administered at least X hours before administering said compound, wherein X is selected from 1 , 2, 3, 5, 10, 15, 20, 24 and 30.
21. The method of claim 1 , wherein said treatment regimen further comprises administering at least one additional agent selected from the group consisting of an anti-PD-1 antibody, an anti-CTLA4 antibody, a mitosis inhibitor, an aromatase inhibitor, an A2AR antagonist, and an angiogenesis inhibitor.
22. The method of claim 1 , wherein said compound is a polypeptide that comprises an active fragment of a wild-type B7-DC polypeptide.
23. The method of claim 22, wherein said active fragment does not comprise any portion of the transmembrane portion of such polypeptide.
24. The method of claim 22, wherein said B7-DC polypeptide is human B7-DC polypeptide.
25. The method of claim 24, wherein said active fragment comprises an amino acid sequence having at least 80% identity to amino acids 20-221 or 20-121 Of SEQ ID NO: 1.
26. The method of claim 24, wherein said active fragment consists of the amino acid sequence of amino acids 20-221 or 20-121 of SEQ ID NO: 1.
27. The method of claim 23, wherein said PD-1 binding fragment comprises at least 10 contiguous amino acids, or at least 25 contiguous amino acids, or at least 50 contiguous amino acids, or at least 75 contiguous amino acids, or at least 90 contiguous amino acids, or at least 100 contiguous amino acids of SEQ ID NO: 1 or 19.
28. The method of claim 1 , wherein said compound is a polypeptide that comprises an active fragment of a wild-type B7-H1 polypeptide.
29. The method of claim 28, wherein said active fragment does not comprise any portion of the transmembrane portion of such polypeptide.
30. The method of claim 28, wherein said B7-H1 polypeptide is human B7-H1 polypeptide.
31. The method of claim 28, wherein said active fragment comprises at least 10 contiguous amino acids, or at least 25 contiguous amino acids, or at least 50 contiguous amino acids, or at least 75 contiguous amino acids, or at least 90 contiguous amino acids, or at least 100 contiguous amino acids of SEQ ID NO: 16.
32. The method of claim 1 , wherein said compound is a polypeptide that comprises an active fragment of a wild-type PD-1 polypeptide.
33. The method of claim 32, wherein said active fragment does not comprise any portion of the transmembrane portion of such polypeptide.
34. The method of claim 32, wherein said PD-1 polypeptide is human PD-1 polypeptide.
35. The method of claim 32, wherein said active fragment comprises at least 10 contiguous amino acids, or at least 25 contiguous amino acids, or at least 50 contiguous amino acids, or at least 75 contiguous amino acids, or at least 90 contiguous amino acids, or at least 100 contiguous amino acids of SEQ ID NO: 17 or 18.
36. The method of claim 1 , wherein said compound is an anti-PD-1 antibody or an active fragment thereof.
37. The method of claim 36, wherein said potentiating agent is cyclophosphamide or an analog of cyclophosphamide.
38. The method of claim 36, wherein said potentiating agent is administered before administering said compound.
39. The method of claim 38, wherein said potentiating agent is administered at least X hours before administering said compound, wherein X is selected from 1 , 2, 3, 5, 10, 15, 20, 24 and 30.
40. The method of claim 1 , wherein said compound is an anti-CTLA4 antibody or an active fragment thereof.
41. The method of claim 40, wherein said potentiating agent is cyclophosphamide or an analog of cyclophosphamide.
42. The method of claim 40, wherein said potentiating agent is administered before administering said compound.
43. The method of claim 42, wherein said potentiating agent is administered at least X hours before administering said compound, wherein X is selected from 1 , 2, 3, 5, 10, 15, 20, 24 and 30.
44. The method of claim 1 , wherein said compound consists of the extracellular domain of B7-DC, B7-H1 , PD-1 , B7.1 , or an active fragment of any of these.
45. The method of claim 44, wherein said active fragment comprises at least 10 contiguous amino acids, or at least 25 contiguous amino acids, or at least 50 contiguous amino acids, or at least 75 contiguous amino acids, or at least 90 contiguous amino acids, or at least 100 contiguous amino acids.
46. The method of claim 1 , wherein said compound is present in an amount sufficient to treat a disease amendable to treatment by an increased T cell-mediated immune response.
47. The method of claim 46, wherein said compound is selected from the group consisting of:
(a) a fusion protein comprising first and second peptide portions, wherein said first peptide portion comprises an active fragment of a PD-1 antagonist polypeptide and said second peptide portion comprises a portion of an immunoglobulin (Ig);
(b) an anti-PD-1 antibody, including active fragments thereof;
(c) an anti-CTLA4 antibody, including active fragments thereof;
(d) a PD-1 -binding polypeptide or PD-1 ligand-binding polypeptide; and
(e) a combination of any 2 or more of (a), (b), (c) and (d).
48. The method of claim 47, wherein said potentiating agent is cyclophosphamide or an analog of cyclophosphamide.
49. The method of claim 47, wherein said potentiating agent is an agent that reduces activity of regulatory T lymphocytes (T-regs).
50. The method of claim 47, wherein said potentiating agent is Sunitinib (SUTENT®), anti-TGNFβ or lmatinib (GLEEVAC®), anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
51. The method of claim 47, further comprising administering at least one additional agent selected from the group consisting of an anti-PD-1 antibody, an anti-CTLA4 antibody, a mitosis inhibitor, an aromatase inhibitor, an A2AR antagonist, and an angiogenesis inhibitor.
52. The method of claim 46, wherein said disease is an infectious disease.
53. The method of claim 46, wherein said disease is cancer.
54. The method of claim 53, wherein said cancer is bladder cancer, brain cancer, breast cancer, cervical cancer, colo-rectal cancer, esophageal cancer, kidney cancer, liver cancer, lung cancer, nasopharangeal cancer, pancreatic cancer, prostate cancer, skin cancer, stomach cancer, uterine cancer, ovarian cancer, testicular cancer, or hematologic cancer.
55. A therapeutic composition, comprising a compound that prevents inhibitory signal transduction through Programmed Cell Death-1 (PD-1) on T cells and a potentiating agent in a pharmaceutically acceptable carrier, wherein said compound and said potentiating agent are together present in an amount effective to increase a T cell response in a mammal.
56. The composition of claim 55, wherein said compound is selected from the group consisting of:
(a) a fusion protein comprising first and second peptide portions, wherein said first peptide portion comprises an active fragment of a PD-1 antagonist polypeptide and said second peptide portion comprises a portion of an immunoglobulin (Ig);
(b) an anti-PD-1 antibody, including active fragments thereof;
(c) an anti-CTLA4 antibody, including active fragments thereof;
(d) a PD-1 -binding polypeptide, PD-1 ligand-binding polypeptide, a PD- 1 polypeptide, or an active fragment thereof; and
(e) a combination of any 2 or more of (a), (b), (c) and (d).
57. The composition of claim 55, wherein said potentiating agent is cyclophosphamide or an analog of cyclophosphamide.
58. The composition of claim 55, wherein said potentiating agent is Sunitinib (SUTENT®), anti-TGNFβ or lmatinib (GLEEVAC®), anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists.
59. The composition of claim 47, further comprising at least one additional agent selected from the group consisting of an anti-PD-1 antibody, an anti-CTLA4 antibody, a mitosis inhibitor, an aromatase inhibitor, an A2AR antagonist, and an angiogenesis inhibitor.
60. The use of a compound that reduces inhibitory signal transduction in a T cell in the manufacture of a medicament for increasing a T cell response by combination therapy wherein said compound is administered in conjunction with a potentiating agent.
61. The use of claim 60, wherein said compound that reduces inhibitory signal transduction in a T cell and said potentiating agent are provided as separate medicaments for administration at different times.
62. The use according to claim 61 , wherein the potentiating agent is administered up to 24 hours prior to the inhibitory compound.
63. Use of the compound and potentiating agent of claim 60 in the treatment of an infectious disease.
64. Use of the compound and potentiating agent of claim 60 in the treatment of cancer.
65. The use of claim 64, wherein said cancer is bladder cancer, brain cancer, breast cancer, cervical cancer, colo-rectal cancer, esophageal cancer, kidney cancer, liver cancer, lung cancer, nasopharangeal cancer, pancreatic cancer, prostate cancer, skin cancer, stomach cancer, uterine cancer, ovarian cancer, testicular cancer, or hematologic cancer.
66. Use of the compound and potentiating agent of claim 60 in the treatment of an infectious disease.
67. A medical kit for administering a compound that reduces inhibitory signal transduction in a T cell in combination with a potentiating agent, said kit comprising: (a) a dosage supply of a compound that reduces inhibitory signal transduction in a T cell,
(b) a supply of a potentiating agent;
(c) a supply of pharmaceutically acceptable carrier; and
(d) printed instructions for administering the compound in a use according to claims 59-64.
68. The method of claim 1 , wherein said compounds consists of an extracellular domain of B7-DC, B7-H1 , PD-1 , or B7-1 , or a polypeptide differing therefrom by only conservative amino acid substitutions, and fragments of these.
PCT/US2009/004825 2008-08-25 2009-08-25 Compositions of pd-1 antagonists and methods of use WO2010027423A2 (en)

Priority Applications (20)

Application Number Priority Date Filing Date Title
UAA201103619A UA106050C2 (en) 2008-08-25 2009-08-25 Normal;heading 1;heading 2;heading 3;COMPOSITION OF PD-1 ANTAGONIST AND A POTENTIATING AGENT FOR SIMULATING A T CELL RESPONSE
DK09811805.2T DK2350129T3 (en) 2008-08-25 2009-08-25 PREPARATIONS WITH PD-1 ANTAGONISTS AND PROCEDURES FOR USE THEREOF
PL09811805T PL2350129T3 (en) 2008-08-25 2009-08-25 Compositions of pd-1 antagonists and methods of use
EA201170373A EA023148B1 (en) 2008-08-25 2009-08-25 Compositions of pd-1 antagonists and use thereof
MX2011002252A MX2011002252A (en) 2008-08-25 2009-08-25 Compositions of pd-1 antagonists and methods of use.
CN2009801422563A CN102203132A (en) 2008-08-25 2009-08-25 Compositions of PD-1 antagonists and methods of use
SI200931266T SI2350129T1 (en) 2008-08-25 2009-08-25 Compositions of pd-1 antagonists and methods of use
ES09811805.2T ES2545609T3 (en) 2008-08-25 2009-08-25 PD-1 antagonist compositions and methods of use
CA2734908A CA2734908A1 (en) 2008-08-25 2009-08-25 Compositions of pd-1 antagonists and methods of use
AU2009288730A AU2009288730B2 (en) 2008-08-25 2009-08-25 Compositions of PD-1 antagonists and methods of use
NZ591130A NZ591130A (en) 2008-08-25 2009-08-25 Compositions comprising a PD-1 antagonists and cyclophosphamide and methods of use thereof
EP09811805.2A EP2350129B1 (en) 2008-08-25 2009-08-25 Compositions of pd-1 antagonists and methods of use
BRPI0917320A BRPI0917320A2 (en) 2008-08-25 2009-08-25 pd-1 antagonist compositions and methods of use
JP2011524986A JP5798919B2 (en) 2008-08-25 2009-08-25 Compositions and methods of use of PD-1 antagonists
RS20150547A RS54233B1 (en) 2008-08-25 2009-08-25 Compositions of pd-1 antagonists and methods of use
ZA2011/01120A ZA201101120B (en) 2008-08-25 2011-02-11 Compositions of pd-1 antagonists and methods of use
IL211298A IL211298A0 (en) 2008-08-25 2011-02-17 Compositions of pd-1 antagonists and methods of use
MA33714A MA32646B1 (en) 2008-08-25 2011-03-18 Anti-PD compositions and methods of use
HRP20150933TT HRP20150933T1 (en) 2008-08-25 2015-09-07 Compositions of pd-1 antagonists and methods of use
SM201500212T SMT201500212B (en) 2008-08-25 2015-09-09 Pd-1 antagonist compositions and methods of use

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US9170508P 2008-08-25 2008-08-25
US9170908P 2008-08-25 2008-08-25
US9169408P 2008-08-25 2008-08-25
US61/091,709 2008-08-25
US61/091,705 2008-08-25
US61/091,694 2008-08-25
US21169709P 2009-04-02 2009-04-02
US61/211,697 2009-04-02

Publications (2)

Publication Number Publication Date
WO2010027423A2 true WO2010027423A2 (en) 2010-03-11
WO2010027423A3 WO2010027423A3 (en) 2010-05-06

Family

ID=41725778

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/004825 WO2010027423A2 (en) 2008-08-25 2009-08-25 Compositions of pd-1 antagonists and methods of use

Country Status (28)

Country Link
US (4) US8114845B2 (en)
EP (2) EP2350129B1 (en)
JP (1) JP5798919B2 (en)
KR (1) KR20110050529A (en)
CN (1) CN102203132A (en)
AU (1) AU2009288730B2 (en)
CA (1) CA2734908A1 (en)
CL (1) CL2011000423A1 (en)
CO (1) CO6341574A2 (en)
CY (1) CY1116816T1 (en)
DK (1) DK2350129T3 (en)
DO (1) DOP2011000059A (en)
EA (2) EA201500417A1 (en)
ES (1) ES2545609T3 (en)
HR (1) HRP20150933T1 (en)
HU (1) HUE027525T2 (en)
IL (1) IL211298A0 (en)
MA (1) MA32646B1 (en)
MX (1) MX2011002252A (en)
NZ (1) NZ591130A (en)
PE (1) PE20110435A1 (en)
PL (1) PL2350129T3 (en)
PT (1) PT2350129E (en)
RS (1) RS54233B1 (en)
SI (1) SI2350129T1 (en)
SM (1) SMT201500212B (en)
WO (1) WO2010027423A2 (en)
ZA (1) ZA201101120B (en)

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8039589B1 (en) 2002-10-04 2011-10-18 Mayo Foundation For Medical Education And Research B7-DC variants
CN102298053A (en) * 2011-05-20 2011-12-28 中山大学肿瘤防治中心 Composite antibody kit used in postoperative recurrence risk assessment of primary hepatocellular carcinoma
US8114845B2 (en) 2008-08-25 2012-02-14 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
EP2504028A2 (en) * 2009-11-24 2012-10-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
WO2012168944A1 (en) * 2011-06-08 2012-12-13 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
US8445447B2 (en) 2007-07-13 2013-05-21 The Johns Hopkins University B7-DC variants immunogenic compositions and methods of use thereof
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
WO2013176915A1 (en) * 2012-05-25 2013-11-28 Roman Galetto Methods for engineering allogeneic and immunosuppressive resistant t cell for immunotherapy
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
US8747833B2 (en) 2004-10-06 2014-06-10 Mayo Foundation For Medical Education And Research B7-H1 and methods of diagnosis, prognosis, and treatment of cancer
JP2014525918A (en) * 2011-08-01 2014-10-02 ジェネンテック, インコーポレイテッド Method for treating cancer using PD-1 axis binding antagonist and MEK inhibitor
WO2014194293A1 (en) 2013-05-30 2014-12-04 Amplimmune, Inc. Improved methods for the selection of patients for pd-1 or b7-h4 targeted therapies, and combination therapies thereof
EP2910572A1 (en) * 2010-11-11 2015-08-26 The University of Hong Kong Soluble pd-1 variants, fusion constructs, and uses thereof
US9370565B2 (en) 2000-04-28 2016-06-21 The Johns Hopkins University Dendritic cell co-stimulatory molecules
WO2017025498A1 (en) 2015-08-07 2017-02-16 Pieris Pharmaceuticals Gmbh Novel fusion polypeptide specific for lag-3 and pd-1
WO2017055443A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-pd1 antibodies and methods of use
WO2017055404A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific antibodies specific for pd1 and tim3
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US9683048B2 (en) 2014-01-24 2017-06-20 Novartis Ag Antibody molecules to PD-1 and uses thereof
EP3087099A4 (en) * 2013-12-23 2017-07-19 Oncomed Pharmaceuticals, Inc. Immunotherapy with binding agents
EP3243832A1 (en) 2016-05-13 2017-11-15 F. Hoffmann-La Roche AG Antigen binding molecules comprising a tnf family ligand trimer and pd1 binding moiety
US9895441B2 (en) 2012-05-31 2018-02-20 Genentech, Inc. Methods of treating cancer using PD-L1 axis binding antagonists and VEGF antagonists
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
WO2018083204A1 (en) 2016-11-02 2018-05-11 Engmab Sàrl Bispecific antibody against bcma and cd3 and an immunological drug for combined use in treating multiple myeloma
WO2018134279A1 (en) 2017-01-18 2018-07-26 Pieris Pharmaceuticals Gmbh Novel fusion polypeptides specific for lag-3 and pd-1
WO2018185043A1 (en) 2017-04-05 2018-10-11 F. Hoffmann-La Roche Ag Bispecific antibodies specifically binding to pd1 and lag3
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US10188729B2 (en) 2013-08-20 2019-01-29 Merck Sharp & Dohme Corp. Modulation of tumor immunity
EP3444271A1 (en) 2013-08-08 2019-02-20 Cytune Pharma Il-15 and il-15raplha sushi domain based modulokines
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
WO2019149716A1 (en) 2018-01-31 2019-08-08 F. Hoffmann-La Roche Ag Bispecific antibodies comprising an antigen-binding site binding to lag3
US10457725B2 (en) 2016-05-13 2019-10-29 Regeneron Pharmaceuticals, Inc. Methods of treating skin cancer by administering a PD-1 inhibitor
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
WO2019229699A1 (en) 2018-05-31 2019-12-05 Novartis Ag Hepatitis b antibodies
WO2019234576A1 (en) 2018-06-03 2019-12-12 Lamkap Bio Beta Ltd. Bispecific antibodies against ceacam5 and cd47
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
WO2020021061A1 (en) 2018-07-26 2020-01-30 Pieris Pharmaceuticals Gmbh Humanized anti-pd-1 antibodies and uses thereof
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
WO2020043683A1 (en) 2018-08-27 2020-03-05 Pieris Pharmaceuticals Gmbh Combination therapies comprising cd137/her2 bispecific agents and pd-1 axis inhibitors and uses thereof
WO2020053742A2 (en) 2018-09-10 2020-03-19 Novartis Ag Anti-hla-hbv peptide antibodies
EP3659622A1 (en) 2013-08-08 2020-06-03 Cytune Pharma Combined pharmaceutical composition
US10730951B2 (en) 2014-03-31 2020-08-04 Genentech, Inc. Anti-OX40 antibodies and methods of use
US10737113B2 (en) 2014-01-23 2020-08-11 Regeneron Pharmaceuticals, Inc. Human antibodies to PD-1
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1
US10946093B2 (en) 2014-07-15 2021-03-16 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
WO2021053587A1 (en) 2019-09-18 2021-03-25 Klaus Strein Bispecific antibodies against ceacam5 and cd3
EP3831849A1 (en) 2019-12-02 2021-06-09 LamKap Bio beta AG Bispecific antibodies against ceacam5 and cd47
US11065285B2 (en) 2012-01-25 2021-07-20 Dnatrix, Inc. Biomarkers and combination therapies using oncolytic virus and immunomodulation
US11077144B2 (en) 2013-05-13 2021-08-03 Cellectis CD19 specific chimeric antigen receptor and uses thereof
US11078282B2 (en) 2016-04-15 2021-08-03 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11096988B2 (en) 2017-03-16 2021-08-24 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
CN113354710A (en) * 2016-12-09 2021-09-07 复诺健生物科技加拿大有限公司 Compositions and methods for inhibiting CD279 interactions
US11117970B2 (en) 2014-01-23 2021-09-14 Regeneron Pharmaceuticals, Inc. Human antibodies to PD-L1
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11236175B2 (en) 2012-10-10 2022-02-01 Sangamo Therapeutics, Inc. T cell modifying compounds and uses thereof
US11319359B2 (en) 2015-04-17 2022-05-03 Alpine Immune Sciences, Inc. Immunomodulatory proteins with tunable affinities
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
WO2022130348A1 (en) 2020-12-18 2022-06-23 Lamkap Bio Beta Ag Bispecific antibodies against ceacam5 and cd47
US11413331B2 (en) 2017-04-03 2022-08-16 Hoffmann-La Roche Inc. Immunoconjugates
US11447537B2 (en) 2016-10-27 2022-09-20 Io Biotech Aps PDL2 compounds
WO2023012147A1 (en) 2021-08-03 2023-02-09 F. Hoffmann-La Roche Ag Bispecific antibodies and methods of use
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
WO2023242351A1 (en) 2022-06-16 2023-12-21 Lamkap Bio Beta Ag Combination therapy of bispecific antibodies against ceacam5 and cd47 and bispecific antibodies against ceacam5 and cd3

Families Citing this family (480)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2161336T4 (en) * 2005-05-09 2017-04-24 Ono Pharmaceutical Co Human monoclonal antibodies for programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapies
CA2631812C (en) 2005-12-02 2023-08-29 Peter Palese Chimeric viruses presenting non-native surface proteins and uses thereof
EP2347775B1 (en) 2005-12-13 2020-04-15 President and Fellows of Harvard College Scaffolds for cell transplantation
US20110223188A1 (en) * 2008-08-25 2011-09-15 Solomon Langermann Targeted costimulatory polypeptides and methods of use to treat cancer
EP2542590B2 (en) 2010-03-05 2020-04-01 The Johns Hopkins University Compositions and methods for targeted immunomodulatory antibodies and fusion proteins
US8906374B2 (en) 2010-04-20 2014-12-09 Cedars-Sinai Medical Center Combination therapy with CD4 lymphocyte depletion and mTOR inhibitors
US9783578B2 (en) 2010-06-25 2017-10-10 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
US8907053B2 (en) 2010-06-25 2014-12-09 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
WO2012018538A2 (en) * 2010-07-26 2012-02-09 Schering Corporation Bioassays for determining pd-1 modulation
KR102157971B1 (en) 2010-10-06 2020-09-18 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Injectable, pore-forming hydrogels for materials-based cell therapies
CA2834548C (en) 2011-04-28 2021-06-01 The Broad Institute, Inc. Inhibitors of histone deacetylase
US20140234320A1 (en) * 2011-06-20 2014-08-21 La Jolla Institute For Allergy And Immunology Modulators of 4-1bb and immune responses
US10081684B2 (en) 2011-06-28 2018-09-25 Whitehead Institute For Biomedical Research Using sortases to install click chemistry handles for protein ligation
GB201120527D0 (en) * 2011-11-29 2012-01-11 Ucl Business Plc Method
WO2013155493A1 (en) 2012-04-12 2013-10-17 Yale University Methods of treating inflammatory and autoimmune diseases and disorders
AU2013249366B2 (en) 2012-04-16 2018-02-01 President And Fellows Of Harvard College Mesoporous silica compositions for modulating immune responses
EP2877444B1 (en) 2012-07-27 2020-09-02 The Broad Institute, Inc. Inhibitors of histone deacetylase
US20150250837A1 (en) * 2012-09-20 2015-09-10 Morningside Technology Ventures Ltd. Oncolytic virus encoding pd-1 binding agents and uses of the same
US9657082B2 (en) 2013-01-31 2017-05-23 Thomas Jefferson University PD-L1 and PD-L2-based fusion proteins and uses thereof
WO2014122271A1 (en) * 2013-02-07 2014-08-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for predicting the survival time of patients suffering from diffuse large b-cell lymphomas
ES2814962T3 (en) 2013-02-20 2021-03-29 Novartis Ag Efficient targeting of primary human leukemia using anti-CD123 chimeric antigen receptor modified T cells
CN111139256A (en) 2013-02-20 2020-05-12 诺华股份有限公司 Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptors
EP2958588B1 (en) 2013-02-22 2017-08-23 CureVac AG Combination of vaccination and inhibition of the pd-1 pathway
SG11201506052PA (en) * 2013-02-22 2015-09-29 Curevac Gmbh Combination of vaccination and inhibition of the pd-1 pathway
US20160017011A1 (en) * 2013-02-26 2016-01-21 Rongfu Wang Phf20 and jmjd3 compositions and methods of use in cancer immunotherapy
MY180687A (en) 2013-03-14 2020-12-07 Icahn School Med Mount Sinai Newcastle disease viruses and uses thereof
UY35468A (en) 2013-03-16 2014-10-31 Novartis Ag CANCER TREATMENT USING AN ANTI-CD19 CHEMERIC ANTIGEN RECEIVER
US10532050B2 (en) 2013-04-09 2020-01-14 Lixte Biotechnology, Inc. Formulations of oxabicycloheptanes and oxabicycloheptenes
WO2014179664A2 (en) 2013-05-02 2014-11-06 Anaptysbio, Inc. Antibodies directed against programmed death-1 (pd-1)
EP3546484B1 (en) 2013-05-10 2021-09-08 Whitehead Institute for Biomedical Research In vitro production of red blood cells with sortaggable proteins
EP2994530A4 (en) 2013-05-10 2016-11-16 Whitehead Biomedical Inst Protein modification of living cells using sortase
US11311575B2 (en) 2013-05-13 2022-04-26 Cellectis Methods for engineering highly active T cell for immunotherapy
US9452217B2 (en) 2013-06-22 2016-09-27 Nitor Therapeutics Methods for potentiating immune response for the treatment of infectious diseases and cancer
ES2728578T3 (en) 2013-09-20 2019-10-25 Bristol Myers Squibb Co Combination of anti-LAG-3 antibodies and anti-PD-1 antibodies to treat tumors
WO2015066413A1 (en) 2013-11-01 2015-05-07 Novartis Ag Oxazolidinone hydroxamic acid compounds for the treatment of bacterial infections
CA2987519A1 (en) 2013-11-01 2015-05-07 Yale University Delivery vehicles
US10556024B2 (en) 2013-11-13 2020-02-11 Whitehead Institute For Biomedical Research 18F labeling of proteins using sortases
US20150140036A1 (en) 2013-11-13 2015-05-21 Novartis Institutes For Biomedical Research, Inc. Low, immune enhancing, dose mtor inhibitors and uses thereof
SI3081576T1 (en) 2013-12-12 2019-12-31 Shanghai Hengrui Pharmaceutical Co., Ltd., Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
US20150190506A1 (en) * 2013-12-17 2015-07-09 Genentech, Inc. Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists
JP6779785B2 (en) 2013-12-19 2020-11-04 ノバルティス アーゲー Human mesothelin chimeric antigen receptor and its use
EP4070818A3 (en) 2014-01-06 2023-01-11 The Trustees of the University of Pennsylvania Pd1 and pdl1 antibodies and vaccine combinations and use of same for immunotherapy
JP2017503503A (en) * 2014-01-06 2017-02-02 エクスプレッション、パソロジー、インコーポレイテッド SRM assay for PD-L1
JO3517B1 (en) 2014-01-17 2020-07-05 Novartis Ag N-azaspirocycloalkane substituted n-heteroaryl compounds and compositions for inhibiting the activity of shp2
US11213583B2 (en) * 2014-02-05 2022-01-04 Cedars-Sinai Medical Center Methods and compositions for treating cancer and infectious diseases
AU2015222685A1 (en) 2014-02-27 2016-09-08 Viralytics Limited Combination method for treatment of cancer
EP3114144A1 (en) * 2014-03-05 2017-01-11 Bristol-Myers Squibb Company Treatment of renal cancer using a combination of an anti-pd-1 antibody and another anti-cancer agent
US10519237B2 (en) 2014-03-12 2019-12-31 Yeda Research And Development Co. Ltd Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
US10618963B2 (en) 2014-03-12 2020-04-14 Yeda Research And Development Co. Ltd Reducing systemic regulatory T cell levels or activity for treatment of disease and injury of the CNS
EP3590529A1 (en) 2014-03-12 2020-01-08 CureVac AG Combination of vaccination and ox40 agonists
NZ725006A (en) 2014-03-12 2019-11-29 Yeda Res & Dev Reducing systemic regulatory t cell levels or activity for treatment of disease and injury of the cns
US9394365B1 (en) 2014-03-12 2016-07-19 Yeda Research And Development Co., Ltd Reducing systemic regulatory T cell levels or activity for treatment of alzheimer's disease
CN113583129A (en) 2014-03-14 2021-11-02 诺华股份有限公司 Antibody molecules against LAG-3 and uses thereof
EP3119423B1 (en) 2014-03-15 2022-12-14 Novartis AG Treatment of cancer using chimeric antigen receptor
PE20161371A1 (en) 2014-03-24 2016-12-21 Novartis Ag ORGANIC MONOBACTAM COMPOUNDS FOR THE TREATMENT OF BACTERIAL INFECTIONS
RU2718542C2 (en) 2014-04-07 2020-04-08 Новартис Аг Treating a malignant tumor using a chimeric antigen cd19 receptor
JP7348708B2 (en) 2014-04-30 2023-09-21 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Combination vaccine device and method for killing cancer cells
MX2016014753A (en) 2014-05-15 2017-03-06 Bristol Myers Squibb Co Treatment of lung cancer using a combination of an anti-pd-1 antibody and another anti-cancer agent.
SG11201609721WA (en) 2014-05-28 2016-12-29 Agenus Inc Anti-gitr antibodies and methods of use thereof
TWI693232B (en) 2014-06-26 2020-05-11 美商宏觀基因股份有限公司 Covalently bonded diabodies having immunoreactivity with pd-1 and lag-3, and methods of use thereof
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
ES2805475T3 (en) 2014-07-21 2021-02-12 Novartis Ag Cancer treatment using a chimeric CD33 antigen receptor
US20170209492A1 (en) 2014-07-31 2017-07-27 Novartis Ag Subset-optimized chimeric antigen receptor-containing t-cells
CN107001316A (en) 2014-08-06 2017-08-01 诺华股份有限公司 It is used as the Carbostyril derivative of antiseptic
KR101940430B1 (en) 2014-08-07 2019-01-18 가꼬우호우징 효고 이카다이가쿠 Therapeutic agent for cancer which comprises combination of il-18 and molecule-targeting antibody
US10800830B2 (en) * 2014-08-08 2020-10-13 The Board Of Trustees Of The Leland Stanford Junior University High affinity PD-1 agents and methods of use
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
TW202140557A (en) 2014-08-19 2021-11-01 瑞士商諾華公司 Treatment of cancer using a cd123 chimeric antigen receptor
WO2016041945A1 (en) * 2014-09-16 2016-03-24 Innate Pharma Neutralization of inhibitory pathways in lymphocytes
CA2961636A1 (en) 2014-09-17 2016-03-24 Boris ENGELS Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
AU2015327868A1 (en) 2014-10-03 2017-04-20 Novartis Ag Combination therapies
US10053683B2 (en) 2014-10-03 2018-08-21 Whitehead Institute For Biomedical Research Intercellular labeling of ligand-receptor interactions
KR20170068504A (en) 2014-10-08 2017-06-19 노파르티스 아게 Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
MA41044A (en) 2014-10-08 2017-08-15 Novartis Ag COMPOSITIONS AND METHODS OF USE FOR INCREASED IMMUNE RESPONSE AND CANCER TREATMENT
CN107001478B (en) 2014-10-14 2022-01-11 诺华股份有限公司 Antibody molecules against PD-L1 and uses thereof
NZ732211A (en) * 2014-10-24 2020-04-24 Calidi Biotherapeutics Inc Combination immunotherapy approach for treatment of cancer
SG11201702723VA (en) 2014-10-29 2017-05-30 Five Prime Therapeutics Inc Combination therapy for cancer
ES2875338T3 (en) * 2014-11-06 2021-11-10 Hibercell Inc Beta-glucan methods and compositions that affect the tumor microenvironment
KR20170080697A (en) 2014-11-13 2017-07-10 더 존스 홉킨스 유니버시티 Checkpoint blockade and microsatellite instability
AU2015347015B2 (en) 2014-11-14 2019-02-14 Novartis Ag Antibody drug conjugates
BR112017011536A2 (en) * 2014-12-02 2018-02-27 Celgene Corp combination therapies
US20180334490A1 (en) 2014-12-03 2018-11-22 Qilong H. Wu Methods for b cell preconditioning in car therapy
WO2016094273A1 (en) * 2014-12-08 2016-06-16 Dana-Farber Cancer Institute, Inc. Methods for upregulating immune responses using combinations of anti-rgmb and anti-pd-1 agents
TWI595006B (en) 2014-12-09 2017-08-11 禮納特神經系統科學公司 Anti-pd-1 antibodies and methods of use thereof
CA2969338A1 (en) * 2014-12-16 2016-06-23 Bristol-Myers Squibb Company Use of immune checkpoint inhibitors in central nervous systems neoplasms
PE20171044A1 (en) 2014-12-16 2017-07-19 Novartis Ag ISOXAZOLE HYDROXAMIC ACID COMPOUNDS AS LpxC INHIBITORS
WO2016100882A1 (en) 2014-12-19 2016-06-23 Novartis Ag Combination therapies
CN105983097B (en) * 2015-01-28 2021-06-08 华中科技大学同济医学院附属协和医院 Anti-tumor preparation and preparation method thereof
CA3012602A1 (en) * 2015-01-30 2016-08-04 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
EP3256165B1 (en) * 2015-02-13 2021-07-14 Sorrento Therapeutics, Inc. Antibody therapeutics that bind ctla4
CA2977767A1 (en) * 2015-02-26 2016-09-01 Merck Patent Gmbh Pd-1 / pd-l1 inhibitors for the treatment of cancer
BR112017018908A2 (en) 2015-03-10 2018-04-17 Aduro Biotech, Inc. compositions and methods for activating interferon gene stimulator-dependent signaling
HUE048111T2 (en) 2015-03-23 2020-05-28 Bayer Pharma AG Anti-ceacam6 antibodies and uses thereof
WO2016164580A1 (en) 2015-04-07 2016-10-13 Novartis Ag Combination of chimeric antigen receptor therapy and amino pyrimidine derivatives
EP3280464A4 (en) 2015-04-10 2018-09-26 President and Fellows of Harvard College Immune cell trapping devices and methods for making and using the same
GB201506411D0 (en) 2015-04-15 2015-05-27 Bergenbio As Humanized anti-axl antibodies
MY188749A (en) 2015-04-17 2021-12-28 Bristol Myers Squibb Co Compositions comprising a combination of nivolumab and ipilimumab
JP7114457B2 (en) 2015-04-17 2022-08-08 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Methods for Improving Efficacy and Growth of Chimeric Antigen Receptor-Expressing Cells
EP3286211A1 (en) 2015-04-23 2018-02-28 Novartis AG Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
KR20170140316A (en) 2015-04-28 2017-12-20 브리스톨-마이어스 스큅 컴퍼니 Treatment of PD-L1-positive melanoma with anti-PD-1 antibody
KR20170138555A (en) 2015-04-28 2017-12-15 브리스톨-마이어스 스큅 컴퍼니 Treatment of PD-L1-negative melanoma with anti-PD-1 antibody and anti-CTLA-4 antibody
WO2016191751A1 (en) 2015-05-28 2016-12-01 Bristol-Myers Squibb Company Treatment of pd-l1 positive lung cancer using an anti-pd-1 antibody
WO2016196389A1 (en) 2015-05-29 2016-12-08 Bristol-Myers Squibb Company Treatment of renal cell carcinoma
CN107849144B (en) 2015-05-29 2021-09-17 艾吉纳斯公司 anti-CTLA-4 antibodies and methods of use thereof
TW201709929A (en) 2015-06-12 2017-03-16 宏觀基因股份有限公司 Combination therapy for the treatment of cancer
EP3310813A1 (en) 2015-06-17 2018-04-25 Novartis AG Antibody drug conjugates
JP7014706B2 (en) 2015-07-13 2022-02-01 サイトメックス セラピューティクス インコーポレイテッド Anti-PD-1 antibody, activating anti-PD-1 antibody, and how to use it
WO2017011666A1 (en) 2015-07-14 2017-01-19 Bristol-Myers Squibb Company Method of treating cancer using immune checkpoint inhibitor
EP3322448A4 (en) 2015-07-16 2019-03-06 Bioxcel Therapeutics, Inc. A novel approach for treatment of cancer using immunomodulation
US10786547B2 (en) 2015-07-16 2020-09-29 Biokine Therapeutics Ltd. Compositions, articles of manufacture and methods for treating cancer
WO2017015427A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
KR20180034426A (en) 2015-07-29 2018-04-04 노파르티스 아게 Combination of anti-PD-1 and anti-M-CSF antibodies in the treatment of cancer
BR112018001640A2 (en) 2015-07-29 2018-09-18 Novartis Ag combination of pd-1 antagonist with an egfr inhibitor
ES2878188T3 (en) 2015-07-29 2021-11-18 Novartis Ag Combination therapies comprising antibody molecules against LAG-3
EP3878465A1 (en) 2015-07-29 2021-09-15 Novartis AG Combination therapies comprising antibody molecules to tim-3
PL3328419T3 (en) 2015-07-30 2021-12-27 Macrogenics, Inc. Pd-1-binding molecules and methods of use thereof
US10660954B2 (en) 2015-07-31 2020-05-26 University Of Florida Research Foundation, Incorporated Hematopoietic stem cells in combinatorial therapy with immune checkpoint inhibitors against cancer
EA201800148A1 (en) 2015-08-11 2019-01-31 Калиди Биотерапьютикс, Инк. CHALLENGE VACCINE FOR CANCER TREATMENT
EP3334431B9 (en) 2015-08-11 2020-03-04 Novartis AG 5-bromo-2,6-di-(1h-pyrazol-l-yl)pyrimidin-4-amine for use in the treatment of cancer
US11747346B2 (en) 2015-09-03 2023-09-05 Novartis Ag Biomarkers predictive of cytokine release syndrome
EP4218833A1 (en) 2015-10-01 2023-08-02 Whitehead Institute for Biomedical Research Labeling of antibodies
WO2017062619A2 (en) 2015-10-08 2017-04-13 Macrogenics, Inc. Combination therapy for the treatment of cancer
WO2017066561A2 (en) 2015-10-16 2017-04-20 President And Fellows Of Harvard College Regulatory t cell pd-1 modulation for regulating t cell effector immune responses
EP3365062A4 (en) 2015-10-19 2019-07-17 Cold Genesys, Inc. Methods of treating solid or lymphatic tumors by combination therapy
MA44334A (en) 2015-10-29 2018-09-05 Novartis Ag ANTIBODY CONJUGATES INCLUDING A TOLL-TYPE RECEPTOR AGONIST
MX2018005517A (en) 2015-11-02 2018-11-09 Five Prime Therapeutics Inc Cd80 extracellular domain polypeptides and their use in cancer treatment.
CA3003969A1 (en) 2015-11-06 2017-05-11 Orionis Biosciences Nv Bi-functional chimeric proteins and uses thereof
KR20180071386A (en) 2015-11-18 2018-06-27 브리스톨-마이어스 스큅 컴퍼니 Treatment of lung cancer using a combination of anti-PD-1 antibody and anti-CTLA-4 antibody
EP3380523A1 (en) 2015-11-23 2018-10-03 Five Prime Therapeutics, Inc. Fgfr2 inhibitors alone or in combination with immune stimulating agents in cancer treatment
KR20180083944A (en) 2015-12-02 2018-07-23 아게누스 인코포레이티드 Antibodies and methods for their use
AR107781A1 (en) 2015-12-14 2018-06-06 Macrogenics Inc BISPECIFIC MOLECULES THAT HAVE IMMUNORREACTIVITY WITH PD-1 AND CTLA-4, AND METHODS OF USE OF THE SAME
EP3389712B1 (en) 2015-12-17 2024-04-10 Novartis AG Antibody molecules to pd-1 and uses thereof
US10392442B2 (en) 2015-12-17 2019-08-27 Bristol-Myers Squibb Company Use of anti-PD-1 antibody in combination with anti-CD27 antibody in cancer treatment
WO2017103895A1 (en) 2015-12-18 2017-06-22 Novartis Ag Antibodies targeting cd32b and methods of use thereof
EP3393504A1 (en) 2015-12-22 2018-10-31 Novartis AG Mesothelin chimeric antigen receptor (car) and antibody against pd-l1 inhibitor for combined use in anticancer therapy
ES2954153T3 (en) 2015-12-22 2023-11-20 Regeneron Pharma Combination of anti-PD-1 antibodies and anti-CD20/anti-CD3 bispecific antibodies to treat cancer
WO2017112943A1 (en) 2015-12-23 2017-06-29 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
EP3403091B1 (en) 2016-01-11 2021-12-22 Technion Research & Development Foundation Limited Methods of determining prognosis of sepsis and treating same
CA3009001A1 (en) 2016-01-11 2017-07-20 Universitat Zurich Immune-stimulating humanized monoclonal antibodies against human interleukin-2, and fusion proteins thereof
CA3013554A1 (en) 2016-02-05 2017-08-10 Orionis Biosciences Nv Targeted therapeutic agents and uses thereof
CN115531609A (en) 2016-02-06 2022-12-30 哈佛学院校长同事会 Remodeling hematopoietic niches to reconstitute immunity
WO2017140821A1 (en) 2016-02-19 2017-08-24 Novartis Ag Tetracyclic pyridone compounds as antivirals
EP3423488A4 (en) 2016-02-29 2019-11-06 Foundation Medicine, Inc. Methods of treating cancer
CN109153714A (en) 2016-03-04 2019-01-04 诺华股份有限公司 Express the cell and application thereof of multiple Chimeric antigen receptor (CAR) molecule
WO2017155981A1 (en) 2016-03-07 2017-09-14 Massachusetts Institute Of Technology Protein-chaperoned t-cell vaccines
EP3426271A4 (en) 2016-03-10 2019-10-16 Cold Genesys, Inc. Methods of treating solid or lymphatic tumors by combination therapy
US20210309965A1 (en) 2016-03-21 2021-10-07 Dana-Farber Cancer Institute, Inc. T-cell exhaustion state-specific gene expression regulators and uses thereof
DK3433257T3 (en) 2016-03-24 2024-01-02 Novartis Ag ALKYNYL NUCLEOSIDE ANALOGUES AS INHIBITORS OF HUMAN RHINOVIRUS
US11209441B2 (en) 2016-04-05 2021-12-28 Bristol-Myers Squibb Company Cytokine profiling analysis
AU2017249698B2 (en) 2016-04-13 2023-03-09 Vivia Biotech, S.L Ex vivo bite-activated T cells
EP3454887B1 (en) 2016-05-13 2021-01-20 Orionis Biosciences BV Targeted mutant interferon-beta and uses thereof
CA3023881A1 (en) 2016-05-13 2017-11-16 Orionis Biosciences Nv Therapeutic targeting of non-cellular structures
CA3024470A1 (en) 2016-05-18 2017-11-23 Modernatx, Inc. Polynucleotides encoding interleukin-12 (il12) and uses thereof
CN109476718B (en) 2016-05-18 2023-07-04 莫得纳特斯公司 Combination of MRNA encoding immunomodulatory polypeptides and uses thereof
CA3024509A1 (en) 2016-05-18 2017-11-23 Modernatx, Inc. Mrna combination therapy for the treatment of cancer
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
JP7194022B2 (en) 2016-05-20 2022-12-21 イーライ リリー アンド カンパニー Combination therapy with Notch inhibitors and PD-1 or PD-L1 inhibitors
KR20230091191A (en) 2016-05-27 2023-06-22 아게누스 인코포레이티드 Anti-tim-3 antibodies and methods of use thereof
KR20190008962A (en) 2016-06-02 2019-01-25 브리스톨-마이어스 스큅 컴퍼니 Use of anti-PD-1 antibodies in combination with anti-CD30 antibodies in the treatment of lymphoma
ES2954139T3 (en) 2016-06-02 2023-11-20 Bristol Myers Squibb Co PD-1 Blockade With Nivolumab in Refractory Hodgkin Lymphoma
CN109476754A (en) 2016-06-03 2019-03-15 百时美施贵宝公司 Purposes of the anti-PD-1 antibody in treatment colorectal cancer patients
KR20190015407A (en) 2016-06-03 2019-02-13 브리스톨-마이어스 스큅 컴퍼니 Anti-PD-1 antibody for use in the treatment of recurrent small cell lung cancer
EP3464369A1 (en) 2016-06-03 2019-04-10 Bristol-Myers Squibb Company Anti-pd-1 antibody for use in a method of treating a tumor
WO2017216705A1 (en) 2016-06-14 2017-12-21 Novartis Ag Crystalline form of (r)-4-(5-(cyclopropylethynyl)isoxazol-3-yl)-n-hydroxy-2-methyl-2-(methylsulfonyl)butanamide as an antibacterial agent
WO2017216686A1 (en) 2016-06-16 2017-12-21 Novartis Ag 8,9-fused 2-oxo-6,7-dihydropyrido-isoquinoline compounds as antivirals
WO2017216685A1 (en) 2016-06-16 2017-12-21 Novartis Ag Pentacyclic pyridone compounds as antivirals
WO2017223422A1 (en) 2016-06-24 2017-12-28 Infinity Pharmaceuticals, Inc. Combination therapies
US11098077B2 (en) 2016-07-05 2021-08-24 Chinook Therapeutics, Inc. Locked nucleic acid cyclic dinucleotide compounds and uses thereof
WO2018013797A1 (en) 2016-07-13 2018-01-18 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same
KR102565885B1 (en) 2016-07-20 2023-08-09 유니버시티 오브 유타 리서치 파운데이션 CD229 CAR T Cells and Methods of Using The Same
WO2018023025A1 (en) * 2016-07-28 2018-02-01 Novartis Ag Combination therapies of chimeric antigen receptors adn pd-1 inhibitors
CN109562282A (en) 2016-07-29 2019-04-02 伊莱利利公司 MERESTINIB and anti-PD-L1 or the combination treatment of anti-PD-1 inhibitor are used for treating cancer
EP3490548A4 (en) 2016-08-01 2020-04-15 Molecular Templates, Inc. Administration of hypoxia activated prodrugs in combination with immune modulatory agents for treating cancer
AU2016419048B2 (en) * 2016-08-11 2024-02-15 The Council Of The Queensland Institute Of Medical Research Immune-modulating compounds
JP7198666B2 (en) * 2016-08-26 2023-01-04 哲治 奥野 Microvascular blood flow reducing agent and its use
US20190218294A1 (en) 2016-09-09 2019-07-18 Bristol-Myers Squibb Company Use of an anti-pd-1 antibody in combination with an anti-mesothelin antibody in cancer treatment
TW201811788A (en) 2016-09-09 2018-04-01 瑞士商諾華公司 Polycyclic pyridone compounds as antivirals
AU2017322501A1 (en) 2016-09-09 2019-03-28 Laboratoire Francais Du Fractionnement Et Des Biotechnologies Combination of an anti-CD20 antibody, PI3 kinase-delta inhibitor, and anti-PD-1 or anti-PD-L1 antibody for treating hematological cancers
UA124631C2 (en) * 2016-09-14 2021-10-20 Еббві Байотерапьютікс Інк. Anti-pd-1(cd279) antibodies
WO2018057585A1 (en) 2016-09-21 2018-03-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptor (car) that targets chemokine receptor ccr4 and its use
RU2759334C2 (en) 2016-09-21 2021-11-12 Нексткьюр, Инк. Antibodies against siglec-15 and their application methods
JP7274413B2 (en) 2016-09-23 2023-05-16 マレンゴ・セラピューティクス,インコーポレーテッド Multispecific antibody molecules containing lambda and kappa light chains
JOP20190061A1 (en) 2016-09-28 2019-03-26 Novartis Ag Beta-lactamase inhibitors
US10525083B2 (en) 2016-10-07 2020-01-07 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
MA46529A (en) 2016-10-11 2019-08-21 Agenus Inc ANTI-LAG-3 ANTIBODIES AND PROCESSES FOR USE
WO2018071576A1 (en) 2016-10-14 2018-04-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Treatment of tumors by inhibition of cd300f
WO2018073753A1 (en) 2016-10-18 2018-04-26 Novartis Ag Fused tetracyclic pyridone compounds as antivirals
WO2018077893A1 (en) 2016-10-24 2018-05-03 Orionis Biosciences Nv Targeted mutant interferon-gamma and uses thereof
MA49863A (en) 2016-11-01 2020-06-17 Anaptysbio Inc ANTIBODIES DIRECTED AGAINST PROGRAMMED DEATH 1 (PD -1)
PL3535298T3 (en) 2016-11-02 2021-12-27 Jounce Therapeutics, Inc. Antibodies to pd-1 and uses thereof
WO2018101448A1 (en) 2016-11-30 2018-06-07 Kyowa Hakko Kirin Co., Ltd. Method of treating cancer using anti-ccr4 antibody and anti-pd-1 antibody
EP3548083A1 (en) 2016-12-03 2019-10-09 Juno Therapeutics, Inc. Methods for modulation of car-t cells
IL266918B2 (en) 2016-12-07 2024-03-01 Agenus Inc Anti-ctla-4 antibodies and methods of use thereof
MA50948A (en) 2016-12-07 2020-10-14 Agenus Inc ANTIBODIES AND METHODS OF USING THE SAME
MX2019008207A (en) 2017-01-09 2019-12-11 Tesaro Inc Methods of treating cancer with anti-pd-1 antibodies.
WO2018129497A1 (en) 2017-01-09 2018-07-12 Bioxcel Therapeutics, Inc. Predictive and diagnostic methods for prostate cancer
EA201991696A1 (en) 2017-01-13 2020-02-05 Эйдженус Инк. T-CELL RECEPTORS RELATING TO NY-ESO-1 AND METHODS OF APPLICATION
EP3570870A1 (en) 2017-01-20 2019-11-27 Novartis AG Combination therapy for the treatment of cancer
TWI787230B (en) 2017-01-20 2022-12-21 法商賽諾菲公司 Anti-tgf-beta antibodies and their use
AR110755A1 (en) 2017-01-20 2019-05-02 Genzyme Corp BONE DIRECTED ANTIBODIES
JOP20190187A1 (en) 2017-02-03 2019-08-01 Novartis Ag Anti-ccr7 antibody drug conjugates
JP2020506727A (en) 2017-02-06 2020-03-05 オリオンズ バイオサイエンス エヌブイ Targeted chimeric proteins and uses thereof
US10906985B2 (en) 2017-02-06 2021-02-02 Orionis Biosciences, Inc. Targeted engineered interferon and uses thereof
WO2018151820A1 (en) 2017-02-16 2018-08-23 Elstar Therapeutics, Inc. Multifunctional molecules comprising a trimeric ligand and uses thereof
CA3054289A1 (en) 2017-02-21 2018-08-30 Regeneron Pharmaceuticals, Inc. Anti-pd-1 antibodies for treatment of lung cancer
JP7132232B2 (en) 2017-02-24 2022-09-06 マクロジェニクス,インコーポレーテッド Bispecific binding molecules capable of binding CD137 and tumor antigens and uses thereof
EP3589308A1 (en) 2017-02-28 2020-01-08 Sanofi Therapeutic rna
US11179413B2 (en) 2017-03-06 2021-11-23 Novartis Ag Methods of treatment of cancer with reduced UBB expression
WO2018167778A1 (en) 2017-03-12 2018-09-20 Yeda Research And Development Co. Ltd. Methods of diagnosing and prognosing cancer
WO2018167780A1 (en) 2017-03-12 2018-09-20 Yeda Research And Development Co. Ltd. Methods of prognosing and treating cancer
US20200031944A1 (en) 2017-03-31 2020-01-30 Five Prime Therapeutics, Inc. Combination therapy for cancer using anti-gitr antibodies
WO2018185618A1 (en) 2017-04-03 2018-10-11 Novartis Ag Anti-cdh6 antibody drug conjugates and anti-gitr antibody combinations and methods of treatment
MX2019012223A (en) 2017-04-13 2019-12-09 Agenus Inc Anti-cd137 antibodies and methods of use thereof.
WO2018195283A1 (en) 2017-04-19 2018-10-25 Elstar Therapeutics, Inc. Multispecific molecules and uses thereof
JP7408396B2 (en) 2017-04-20 2024-01-05 アーデーセー セラピューティクス ソシエテ アノニム combination therapy
US11596696B2 (en) 2017-04-20 2023-03-07 Adc Therapeutics Sa Combination therapy with an anti-CD25 antibody-drug conjugate
AR111419A1 (en) 2017-04-27 2019-07-10 Novartis Ag INDAZOL PIRIDONA FUSIONED COMPOUNDS AS ANTIVIRALS
UY37695A (en) 2017-04-28 2018-11-30 Novartis Ag BIS 2’-5’-RR- (3’F-A) (3’F-A) CYCLE DINUCLEOTIDE COMPOUND AND USES OF THE SAME
AU2018260545B2 (en) 2017-04-28 2023-11-23 Marengo Therapeutics, Inc. Multispecific molecules comprising a non-immunoglobulin heterodimerization domain and uses thereof
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
AR111651A1 (en) 2017-04-28 2019-08-07 Novartis Ag CONJUGATES OF ANTIBODIES THAT INCLUDE TOLL TYPE RECEIVER AGONISTS AND COMBINATION THERAPIES
US20200179511A1 (en) 2017-04-28 2020-06-11 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
KR20190139216A (en) 2017-04-28 2019-12-17 파이브 프라임 테라퓨틱스, 인크. Therapeutic Methods Using CD80 Extracellular Domain Polypeptides
PL3618863T3 (en) 2017-05-01 2023-11-06 Agenus Inc. Anti-tigit antibodies and methods of use thereof
AR111658A1 (en) 2017-05-05 2019-08-07 Novartis Ag 2-TRICYCLINAL CHINOLINONES AS ANTIBACTERIAL AGENTS
SG10202107880XA (en) 2017-05-12 2021-09-29 Harpoon Therapeutics Inc Mesothelin binding proteins
US11685787B2 (en) 2017-05-16 2023-06-27 Bristol-Myers Squibb Company Treatment of cancer with anti-GITR agonist antibodies
JP7285220B2 (en) 2017-05-18 2023-06-01 モデルナティエックス インコーポレイテッド Lipid nanoparticles comprising linked interleukin-12 (IL12) polypeptide-encoding polynucleotides
WO2018215937A1 (en) 2017-05-24 2018-11-29 Novartis Ag Interleukin-7 antibody cytokine engrafted proteins and methods of use in the treatment of cancer
US20200362058A1 (en) 2017-05-24 2020-11-19 Novartis Ag Antibody-cytokine engrafted proteins and methods of use
AU2018274216A1 (en) 2017-05-24 2019-12-12 Novartis Ag Antibody-cytokine engrafted proteins and methods of use in the treatment of cancer
AU2018277559A1 (en) 2017-05-30 2019-10-17 Bristol-Myers Squibb Company Compositions comprising a combination of an anti-LAG-3 antibody, a PD-1 pathway inhibitor, and an immunotherapeutic agent
DK3631454T3 (en) 2017-05-30 2023-12-04 Bristol Myers Squibb Co TREATMENT OF LAYER-3 POSITIVE TUMORS
JP7301002B2 (en) 2017-05-30 2023-06-30 ブリストル-マイヤーズ スクイブ カンパニー Compositions comprising anti-LAG-3 antibodies or anti-LAG-3 antibodies and anti-PD-1 or anti-PD-L1 antibodies
EP3630836A1 (en) 2017-05-31 2020-04-08 Elstar Therapeutics, Inc. Multispecific molecules that bind to myeloproliferative leukemia (mpl) protein and uses thereof
WO2018222685A1 (en) 2017-05-31 2018-12-06 Stcube & Co., Inc. Methods of treating cancer using antibodies and molecules that immunospecifically bind to btn1a1
CA3065929A1 (en) 2017-06-01 2018-12-06 Michael Wayne SAVILLE Bispecific antibodies that bind cd123 and cd3
WO2018223004A1 (en) 2017-06-01 2018-12-06 Xencor, Inc. Bispecific antibodies that bind cd20 and cd3
WO2018222989A1 (en) * 2017-06-02 2018-12-06 The Penn State Research Foundation Ceramide nanoliposomes, compositions and methods of using for immunotherapy
MX2019014268A (en) 2017-06-02 2020-08-03 Juno Therapeutics Inc Articles of manufacture and methods for treatment using adoptive cell therapy.
KR20200026209A (en) 2017-06-06 2020-03-10 주식회사 에스티큐브앤컴퍼니 How to treat cancer using antibodies and molecules that bind BTN1A1 or BTN1A1-ligand
WO2018229222A1 (en) 2017-06-14 2018-12-20 Adc Therapeutics Sa Dosage regimes for the administration of an anti-cd19 adc
CN110785187B (en) 2017-06-22 2024-04-05 诺华股份有限公司 Antibody molecules against CD73 and uses thereof
WO2018237173A1 (en) 2017-06-22 2018-12-27 Novartis Ag Antibody molecules to cd73 and uses thereof
MX2019015155A (en) 2017-06-29 2020-08-03 Juno Therapeutics Inc Mouse model for assessing toxicities associated with immunotherapies.
EP3658914A1 (en) 2017-07-28 2020-06-03 Bristol-Myers Squibb Company Predictive peripheral blood biomarker for checkpoint inhibitors
WO2019023525A1 (en) * 2017-07-28 2019-01-31 Dana-Farber Cancer Institute, Inc. Enhanced immunotherapy of cancer using targeted transcriptional modulators
WO2019036438A1 (en) * 2017-08-14 2019-02-21 Phosphorex, Inc. Microparticle formulations of adenosine receptor antagonists for treating cancer
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. Multispecific molecules that bind to bcma and uses thereof
IL272697B2 (en) 2017-08-18 2023-12-01 Adastra Pharmaceuticals Inc Polymorphic form of tg02
AU2018323462A1 (en) 2017-08-28 2020-03-26 Bristol-Myers Squibb Company TIM-3 antagonists for the treatment and diagnosis of cancers
JP7387585B2 (en) 2017-09-04 2023-11-28 アジェナス インコーポレイテッド T-cell receptor that binds mixed lineage leukemia (MLL)-specific phosphopeptide and methods of use thereof
MX2020002612A (en) 2017-09-07 2020-07-13 Univ Res Inst Inc Augusta Antibodies to programmed cell death protein 1.
CN109662966A (en) * 2017-10-16 2019-04-23 北京莱科金基因科技有限责任公司 Purposes of the istradefylline in the drug that preparation is used for oncotherapy
EP3697434A1 (en) 2017-10-18 2020-08-26 Vivia Biotech, S.L. Bite-activated car-t cells
KR20200116077A (en) 2017-11-01 2020-10-08 주노 쎄러퓨티크스 인코퍼레이티드 Chimeric antigen receptor and coding polynucleotide specific for B cell maturation antigen
US20210132042A1 (en) 2017-11-01 2021-05-06 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
CA3080904A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for b-cell maturation antigen
TW201922291A (en) 2017-11-16 2019-06-16 瑞士商諾華公司 Combination therapies
AR113696A1 (en) 2017-11-17 2020-06-03 Merck Sharp & Dohme SPECIFIC ANTIBODIES FOR IMMUNOGLOBULIN TYPE 3 (ILT3) SIMILAR TRANSCRIPT AND ITS USES
US20210079015A1 (en) 2017-11-17 2021-03-18 Novartis Ag Novel dihydroisoxazole compounds and their use for the treatment of hepatitis b
MX2020005651A (en) 2017-11-30 2020-10-28 Novartis Ag Bcma-targeting chimeric antigen receptor, and uses thereof.
WO2019113464A1 (en) 2017-12-08 2019-06-13 Elstar Therapeutics, Inc. Multispecific molecules and uses thereof
US11946094B2 (en) 2017-12-10 2024-04-02 Augusta University Research Institute, Inc. Combination therapies and methods of use thereof
WO2019118937A1 (en) 2017-12-15 2019-06-20 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
JP2021507906A (en) 2017-12-20 2021-02-25 ノバルティス アーゲー Fusion tricyclic pyrazolo-dihydropyrazinyl-pyridone compound as an antiviral agent
US11324774B2 (en) 2018-01-05 2022-05-10 Augusta University Research Institute, Inc. Compositions of oral alkaline salts and metabolic acid inducers and uses thereof
JP2021510733A (en) 2018-01-12 2021-04-30 ケーディーエーシー セラピューティクス,インコーポレーテッドKdac Therapeutics, Inc. Combination of selective histone deacetylase 3 (HDAC3) inhibitor and immunotherapeutic agent for the treatment of cancer
BR112020013144A2 (en) 2018-01-12 2020-12-08 Bristol-Myers Squibb Company COMBINATION THERAPY WITH ANTI-IL-8 ANTIBODIES AND ANTI-PD-1 ANTIBODIES FOR CANCER TREATMENT
EP3743061A1 (en) 2018-01-22 2020-12-02 Pascal Biosciences Inc. Cannabinoids and derivatives for promoting immunogenicity of tumor and infected cells
BR112020014929A2 (en) 2018-01-23 2020-12-08 Nextcure, Inc. PHARMACEUTICAL COMPOSITION, METHODS FOR INCREASING, REDUCING, INTENSIFYING OR INDUCING AN IMMUNE RESPONSE IN AN INDIVIDUAL, MOLECULE, MONOCLONAL ANTIBODY OR FRAGMENT OF BINDING THE ANTIGEN OF THE SAME, NUCLEUS ANTIGEN ANTIGEN, ANTICORUS , ANTI-B7H4 ANTIBODY OR ANTIGEN BINDING FRAGMENT OF THE SAME, ANTI-B7H4 ANTIBODY LIGHT AND HEAVY CHAINS, AND LIGHT AND HEAVY ANTIBODY CHAINS
US20210069246A1 (en) 2018-01-31 2021-03-11 Celgene Corporation Combination therapy using adoptive cell therapy and checkpoint inhibitor
US20200405806A1 (en) 2018-02-08 2020-12-31 Bristol-Myers Squibb Company Combination of a tetanus toxoid, anti-ox40 antibody and/or anti-pd-1 antibody to treat tumors
JP2021514982A (en) 2018-02-28 2021-06-17 ノバルティス アーゲー Indole-2-carbonyl compounds and their use for the treatment of hepatitis B
US20210002373A1 (en) 2018-03-01 2021-01-07 Nextcure, Inc. KLRG1 Binding Compositions and Methods of Use Thereof
CA3093036A1 (en) 2018-03-07 2019-09-12 Pfizer Inc. Anti-pd-1 antibody compositions
JP2021517589A (en) 2018-03-12 2021-07-26 アンセルム(アンスティチュート・ナシオナル・ドゥ・ラ・サンテ・エ・ドゥ・ラ・ルシェルシュ・メディカル) Use of calorie restriction mimics to enhance chemoimmunotherapy for the treatment of cancer
CN113754768B (en) 2018-03-14 2023-01-06 表面肿瘤学公司 Antibodies that bind CD39 and uses thereof
US20210238280A1 (en) 2018-03-14 2021-08-05 Elstar Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
EP3765516A2 (en) 2018-03-14 2021-01-20 Elstar Therapeutics, Inc. Multifunctional molecules and uses thereof
US20210361734A1 (en) 2018-03-19 2021-11-25 9 Meters Biopharma, Inc. Compositions and methods for potentiating immune checkpoint inhibitor therapy
WO2019195658A1 (en) 2018-04-05 2019-10-10 Dana-Farber Cancer Institute, Inc. Sting levels as a biomarker for cancer immunotherapy
JP2021521182A (en) 2018-04-12 2021-08-26 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Combination therapy of CD73 antagonist and PD-1 / PD-L1 axis antagonist
KR20200144116A (en) 2018-04-16 2020-12-28 온퀄리티 파마슈티컬스 차이나 리미티드 Methods for preventing or treating side effects of cancer treatment
SG11202010011RA (en) 2018-04-17 2020-11-27 Celldex Therapeutics Inc Anti-cd27 and anti-pd-l1 antibodies and bispecific constructs
EP3781599A1 (en) 2018-04-18 2021-02-24 Xencor, Inc. Pd-1 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and pd-1 antigen binding domains and uses thereof
KR20210003170A (en) 2018-04-18 2021-01-11 젠코어 인코포레이티드 IL-15/IL-15Rα heterodimer Fc fusion protein and uses thereof
JP2021522239A (en) 2018-04-26 2021-08-30 アジェナス インコーポレイテッド Heat shock protein-binding peptide composition and how to use it
US20210047405A1 (en) 2018-04-27 2021-02-18 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
EP3796942A1 (en) 2018-05-23 2021-03-31 ADC Therapeutics SA Molecular adjuvant
AR126019A1 (en) 2018-05-30 2023-09-06 Novartis Ag ANTIBODIES AGAINST ENTPD2, COMBINATION THERAPIES AND METHODS OF USE OF ANTIBODIES AND COMBINATION THERAPIES
US20210214459A1 (en) 2018-05-31 2021-07-15 Novartis Ag Antibody molecules to cd73 and uses thereof
US20210205449A1 (en) 2018-06-01 2021-07-08 Novartis Ag Dosing of a bispecific antibody that bind cd123 and cd3
JP7398396B2 (en) 2018-06-01 2023-12-14 ノバルティス アーゲー Binding molecules for BCMA and their uses
US11505782B2 (en) 2018-06-04 2022-11-22 Calidi Biotherapeutics, Inc. Cell-based vehicles for potentiation of viral therapy
BR112020025048A2 (en) 2018-06-13 2021-04-06 Novartis Ag BCMA CHEMICAL ANTIGEN RECEPTORS AND USES OF THE SAME
TW202005985A (en) 2018-06-21 2020-02-01 美商再生元醫藥公司 Methods for treating cancer with bispecific anti-CD3xMUC16 antibodies and anti-PD-1 antibodies
EP3818083A2 (en) 2018-07-03 2021-05-12 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
CA3103385A1 (en) 2018-07-10 2020-01-16 Novartis Ag 3-(5-hydroxy-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of ikaros family zinc finger 2 (ikzf2)-dependent diseases
AR116109A1 (en) 2018-07-10 2021-03-31 Novartis Ag DERIVATIVES OF 3- (5-AMINO-1-OXOISOINDOLIN-2-IL) PIPERIDINE-2,6-DIONA AND USES OF THE SAME
US20210277135A1 (en) 2018-07-13 2021-09-09 Bristol-Myers Squibb Company Ox-40 agonist, pd-1 pathway inhibitor and ctla-4 inhibitor combination for use in a method of treating a cancer or a solid tumor
KR20210040080A (en) 2018-07-26 2021-04-12 브리스톨-마이어스 스큅 컴퍼니 LAG-3 combination therapy for cancer treatment
CN109053895B (en) * 2018-08-30 2020-06-09 中山康方生物医药有限公司 Bifunctional antibody for resisting PD-1-VEGFA, pharmaceutical composition and application thereof
WO2020047345A1 (en) 2018-08-31 2020-03-05 Yale University Compositions and methods of using cell-penetrating antibodies in combination with immune checkpoint modulators
WO2020048942A1 (en) 2018-09-04 2020-03-12 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for enhancing cytotoxic t lymphocyte-dependent immune responses
KR20210057726A (en) 2018-09-07 2021-05-21 화이자 인코포레이티드 Anti-αvβ8 antibodies and compositions and uses thereof
KR20230170813A (en) 2018-09-12 2023-12-19 노파르티스 아게 Antiviral pyridopyrazinedione compounds
JP2022511337A (en) 2018-09-19 2022-01-31 インサーム (インスティテュート ナショナル デ ラ サンテ エ デ ラ ルシェルシェ メディカル) Methods and Pharmaceutical Compositions for the Treatment of Cancers Resistant to Immune Checkpoint Treatment
JP2022501361A (en) 2018-09-19 2022-01-06 アルパイン イミューン サイエンシズ インコーポレイテッド Methods and uses of variant CD80 fusion proteins and related constructs
MX2021003554A (en) 2018-09-25 2021-05-27 Harpoon Therapeutics Inc Dll3 binding proteins and methods of use.
AU2019346645A1 (en) 2018-09-27 2021-04-29 Marengo Therapeutics, Inc. CSF1R/CCR2 multispecific antibodies
EP3856779A1 (en) 2018-09-28 2021-08-04 Novartis AG Cd22 chimeric antigen receptor (car) therapies
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
JP2022502385A (en) 2018-09-29 2022-01-11 ノバルティス アーゲー Method for producing a compound for inhibiting the activity of SHP2
US20220040183A1 (en) 2018-10-01 2022-02-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of inhibitors of stress granule formation for targeting the regulation of immune responses
MA53822A (en) 2018-10-03 2021-08-11 Xencor Inc IL-12 HETERODIMER FC FUSION PROTEINS
US11377477B2 (en) 2018-10-12 2022-07-05 Xencor, Inc. PD-1 targeted IL-15/IL-15RALPHA fc fusion proteins and uses in combination therapies thereof
WO2020079692A1 (en) 2018-10-17 2020-04-23 Biolinerx Ltd. Treatment of metastatic pancreatic adenocarcinoma
EP3873532A1 (en) 2018-10-31 2021-09-08 Novartis AG Dc-sign antibody drug conjugates
EP3873943A2 (en) 2018-11-01 2021-09-08 Juno Therapeutics, Inc. Methods for treatment using chimeric antigen receptors specific for b-cell maturation antigen
WO2020092854A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Chimeric antigen receptors specific for g protein-coupled receptor class c group 5 member d (gprc5d)
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
WO2020102375A1 (en) 2018-11-14 2020-05-22 Regeneron Pharmaceuticals, Inc. Intralesional administration of pd-1 inhibitors for treating skin cancer
MX2021005686A (en) 2018-11-14 2021-07-07 Bayer Ag Pharmaceutical combination of anti-ceacam6 and either anti-pd-1 or anti-pd-l1 antibodies for the treatment of cancer.
AU2019379325A1 (en) 2018-11-16 2021-06-03 Genexine, Inc. Method of treating a tumor with a combination of IL-7 protein and an immune checkpoint inhibitor
JP2022513062A (en) 2018-11-16 2022-02-07 ジュノー セラピューティクス インコーポレイテッド Methods of Dosing Manipulated T Cells to Treat B-Cell Malignancies
JP2022511437A (en) 2018-11-26 2022-01-31 デバイオファーム インターナショナル エス.エー. Combined treatment of HIV infection
US20220018828A1 (en) 2018-11-28 2022-01-20 Inserm (Institut National De La Santé Et La Recherche Médicale Methods and kit for assaying lytic potential of immune effector cells
US20220088070A1 (en) 2018-11-30 2022-03-24 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
WO2020115262A1 (en) 2018-12-07 2020-06-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of cd26 and cd39 as new phenotypic markers for assessing maturation of foxp3+ t cells and uses thereof for diagnostic purposes
US20220047556A1 (en) 2018-12-17 2022-02-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of sulconazole as a furin inhibitor
CN113271945A (en) 2018-12-20 2021-08-17 诺华股份有限公司 Dosing regimens and pharmaceutical combinations comprising 3- (1-oxoisoindolin-2-yl) piperidine-2, 6-dione derivatives
US11618776B2 (en) 2018-12-20 2023-04-04 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains
CR20210324A (en) 2018-12-21 2021-07-14 Novartis Ag Antibodies to pmel17 and conjugates thereof
WO2020127965A1 (en) 2018-12-21 2020-06-25 Onxeo New conjugated nucleic acid molecules and their uses
MX2021008525A (en) 2019-01-15 2021-11-12 Univ Nantes Mutated interleukin-34 (il-34) polypeptides and uses thereof in therapy.
JP2022518207A (en) 2019-01-17 2022-03-14 ジョージア テック リサーチ コーポレイション Drug delivery system containing oxidized cholesterol
CA3126110A1 (en) 2019-01-21 2020-07-30 Sanofi Therapeutic rna and anti-pd1 antibodies for advanced stage solid tumor cancers
US20220096651A1 (en) 2019-01-29 2022-03-31 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for receptor tyrosine kinase like orphan receptor 1 (ror1)
AU2020222346B2 (en) 2019-02-15 2021-12-09 Novartis Ag Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
US20220107320A1 (en) 2019-02-15 2022-04-07 Incelldx, Inc. Assaying Bladder-Associated Samples, Identifying and Treating Bladder-Associated Neoplasia, and Kits for Use Therein
MX2021009763A (en) 2019-02-15 2021-09-08 Novartis Ag 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof.
WO2020169472A2 (en) 2019-02-18 2020-08-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods of inducing phenotypic changes in macrophages
US20220088075A1 (en) 2019-02-22 2022-03-24 The Trustees Of The University Of Pennsylvania Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
CA3137361A1 (en) 2019-02-28 2020-09-03 Regeneron Pharmaceuticals, Inc. Administration of pd-1 inhibitors for treating skin cancer
SG11202109003QA (en) 2019-03-06 2021-09-29 Regeneron Pharma Il-4/il-13 pathway inhibitors for enhanced efficacy in treating cancer
SG11202110449YA (en) 2019-03-26 2021-10-28 Univ Michigan Regents Small molecule degraders of stat3
US20220185831A1 (en) 2019-03-29 2022-06-16 The Regents Of The University Of Michigan Stat3 protein degraders
JP2022520287A (en) * 2019-04-02 2022-03-29 ボード オブ リージェンツ,ザ ユニバーシティ オブ テキサス システム Combination of Transcription Inhibitors and Immune Checkpoint Inhibitors for Treatment of Disease
JP2022527972A (en) 2019-04-02 2022-06-07 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル How to predict and prevent cancer in patients with premalignant lesions
EP3952850A1 (en) 2019-04-09 2022-02-16 Institut National de la Santé et de la Recherche Médicale (INSERM) Use of sk2 inhibitors in combination with immune checkpoint blockade therapy for the treatment of cancer
EP3956446A1 (en) 2019-04-17 2022-02-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treatment of nlrp3 inflammasome mediated il-1beta dependent disorders
EP3725370A1 (en) 2019-04-19 2020-10-21 ImmunoBrain Checkpoint, Inc. Modified anti-pd-l1 antibodies and methods and uses for treating a neurodegenerative disease
CA3136568A1 (en) 2019-05-13 2020-11-19 Regeneron Pharmaceuticals, Inc. Combination of pd-1 inhibitors and lag-3 inhibitors for enhanced efficacy in treating cancer
WO2020236253A1 (en) 2019-05-20 2020-11-26 Massachusetts Institute Of Technology Boronic ester prodrugs and uses thereof
US20220241412A1 (en) 2019-05-24 2022-08-04 Pfizer Inc. Combination therapies using cdk inhibitors
CN114222729A (en) 2019-06-12 2022-03-22 范德比尔特大学 Amino acid transport inhibitors and uses thereof
EP3983377A1 (en) 2019-06-12 2022-04-20 Vanderbilt University Dibenzylamines as amino acid transport inhibitors
CA3143680A1 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and anti-pd-1 or anti-pd-l1 antibody
WO2020255009A2 (en) 2019-06-18 2020-12-24 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and anti-pd-1 antibody
JP2022541436A (en) 2019-07-16 2022-09-26 ザ・リージェンツ・オブ・ザ・ユニバーシティ・オブ・ミシガン Imidazopyrimidines and their use as EED inhibitors
MX2022001321A (en) * 2019-08-02 2022-05-20 Akeso Pharmaceuticals Inc Anti-ctla4-anti-pd-1 bispecific antibody and uses thereof.
CN114514032A (en) 2019-08-02 2022-05-17 兰提欧派普有限公司 Angiotensin type 2 (AT2) receptor agonists for the treatment of cancer
MX2022001732A (en) 2019-08-12 2022-05-06 Purinomia Biotech Inc Methods and compositions for promoting and potentiating t-cell mediated immune responses through adcc targeting of cd39 expressing cells.
GB201912107D0 (en) 2019-08-22 2019-10-09 Amazentis Sa Combination
US20220305048A1 (en) 2019-08-26 2022-09-29 Dana-Farber Cancer Institute, Inc. Use of heparin to promote type 1 interferon signaling
JP2022545735A (en) 2019-08-27 2022-10-28 ザ リージェンツ オブ ザ ユニヴァシティ オブ ミシガン cereblon E3 ligase inhibitor
KR20220053007A (en) 2019-08-30 2022-04-28 아게누스 인코포레이티드 Anti-CD96 antibodies and methods of use thereof
WO2021055329A1 (en) 2019-09-16 2021-03-25 Surface Oncology, Inc. Anti-cd39 antibody compositions and methods
WO2021053559A1 (en) 2019-09-18 2021-03-25 Novartis Ag Entpd2 antibodies, combination therapies, and methods of using the antibodies and combination therapies
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
CR20220107A (en) 2019-09-18 2022-04-25 Novartis Ag Nkg2d fusion proteins and uses thereof
AU2020348849A1 (en) 2019-09-19 2022-04-07 The Regents Of The University Of Michigan Spirocyclic androgen receptor protein degraders
JP2022549495A (en) 2019-09-25 2022-11-25 シージェン インコーポレイテッド Combination of anti-CD30 ADC, anti-PD-1, and chemotherapeutic agents for treatment of hematopoietic cancer
EP4034559A1 (en) 2019-09-25 2022-08-03 Surface Oncology, Inc. Anti-il-27 antibodies and uses thereof
US11667613B2 (en) 2019-09-26 2023-06-06 Novartis Ag Antiviral pyrazolopyridinone compounds
AU2020358726A1 (en) 2019-10-01 2022-04-07 Silverback Therapeutics, Inc. Combination therapy with immune stimulatory conjugates
EP3800201A1 (en) 2019-10-01 2021-04-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Cd28h stimulation enhances nk cell killing activities
EP4037700A2 (en) 2019-10-03 2022-08-10 Xencor, Inc. Targeted il-12 heterodimeric fc-fusion proteins
US20220363776A1 (en) 2019-10-04 2022-11-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for the treatment of ovarian cancer, breast cancer or pancreatic cancer
TW202128757A (en) 2019-10-11 2021-08-01 美商建南德克公司 Pd-1 targeted il-15/il-15ralpha fc fusion proteins with improved properties
US20220395553A1 (en) 2019-11-14 2022-12-15 Cohbar, Inc. Cxcr4 antagonist peptides
WO2021101271A1 (en) * 2019-11-20 2021-05-27 주식회사 지아이셀 Medium composition for culturing t cells and method for culturing t cells using same
AU2020394441A1 (en) 2019-11-26 2022-06-02 Novartis Ag CD19 and CD22 chimeric antigen receptors and uses thereof
WO2021108025A1 (en) 2019-11-26 2021-06-03 Massachusetts Institute Of Technology Cell-based cancer vaccines and cancer therapies
GB201917254D0 (en) 2019-11-27 2020-01-08 Adc Therapeutics Sa Combination therapy
AU2020397956A1 (en) 2019-12-04 2022-07-07 Orna Therapeutics, Inc. Circular RNA compositions and methods
US11897950B2 (en) 2019-12-06 2024-02-13 Augusta University Research Institute, Inc. Osteopontin monoclonal antibodies
AU2020399976A1 (en) 2019-12-09 2022-06-30 Merck Sharp & Dohme B.V. Combination therapy with LIV1-ADC and PD-1 antagonist
KR20220116257A (en) 2019-12-20 2022-08-22 노파르티스 아게 Combination of anti-TIM-3 antibody MBG453 and anti-TGF-beta antibody NIS793 with or without decitabine or anti-PD-1 antibody spartalizumab for treating myelofibrosis and myelodysplastic syndrome
EP4084821A2 (en) 2020-01-03 2022-11-09 Marengo Therapeutics, Inc. Multifunctional molecules that bind to cd33 and uses thereof
CN111110655B (en) * 2020-01-20 2020-12-25 山东大学 Nano composite and preparation method and application thereof
BR112022014849A2 (en) 2020-01-28 2022-10-11 Genentech Inc METHODS OF TREATMENT OF A SOLID TUMOR, METHODS TO INDUCE T-CELL PROLIFERATION, TO INDUCE NK CELL PROLIFERATION AND TO INDUCE IFNY PRODUCTION
IL295093A (en) 2020-01-30 2022-09-01 Ona Therapeutics S L Combination therapy for treatment of cancer and cancer metastasis
WO2021171264A1 (en) 2020-02-28 2021-09-02 Novartis Ag Dosing of a bispecific antibody that binds cd123 and cd3
AU2021232158A1 (en) 2020-03-06 2022-09-29 Ona Therapeutics, S.L. Anti-CD36 antibodies and their use to treat cancer
CA3172423A1 (en) 2020-03-20 2021-03-22 Alex WESSELHOEFT Circular rna compositions and methods
WO2021195481A1 (en) 2020-03-26 2021-09-30 The Regents Of The University Of Michigan Small molecule stat protein degraders
CA3179800A1 (en) 2020-04-10 2021-10-14 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
US20230181756A1 (en) 2020-04-30 2023-06-15 Novartis Ag Ccr7 antibody drug conjugates for treating cancer
PE20221894A1 (en) 2020-05-01 2022-12-13 Ngm Biopharmaceuticals Inc ILT BINDING AGENTS AND METHODS FOR THEIR USE
CA3178260A1 (en) 2020-05-13 2021-11-18 Massachusetts Institute Of Technology Compositions of polymeric microdevices and their use in cancer immunotherapy
MX2022014943A (en) 2020-05-26 2023-03-08 Inst Nat Sante Rech Med Severe acute respiratory syndrome coronavirus 2 (sars-cov-2) polypeptides and uses thereof for vaccine purposes.
CA3168738A1 (en) 2020-05-26 2021-12-02 Matthew G. Fury Methods of treating cervical cancer by administering a pd-1 inhibitor
US20230233474A1 (en) 2020-05-28 2023-07-27 Modernatx, Inc. Use of mrnas encoding ox40l, il-23 and il-36gamma for treating cancer
US11767353B2 (en) 2020-06-05 2023-09-26 Theraly Fibrosis, Inc. Trail compositions with reduced immunogenicity
AU2021288224A1 (en) 2020-06-11 2023-01-05 Novartis Ag ZBTB32 inhibitors and uses thereof
AR122644A1 (en) 2020-06-19 2022-09-28 Onxeo NEW CONJUGATED NUCLEIC ACID MOLECULES AND THEIR USES
KR20230027056A (en) 2020-06-23 2023-02-27 노파르티스 아게 Dosage regimen comprising 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives
US20230293530A1 (en) 2020-06-24 2023-09-21 Yeda Research And Development Co. Ltd. Agents for sensitizing solid tumors to treatment
US20230233690A1 (en) 2020-07-10 2023-07-27 The Regents Of The University Of Michigan Androgen receptor protein degraders
WO2022011204A1 (en) 2020-07-10 2022-01-13 The Regents Of The University Of Michigan Small molecule androgen receptor protein degraders
EP4188549A1 (en) 2020-08-03 2023-06-07 Novartis AG Heteroaryl substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
CN116134155A (en) 2020-08-26 2023-05-16 瑞泽恩制药公司 Methods of treating cancer by administering PD-1 inhibitors
AU2021333779A1 (en) 2020-08-26 2023-04-13 Marengo Therapeutics, Inc. Methods of detecting TRBC1 or TRBC2
CA3189987A1 (en) 2020-09-02 2022-03-10 Pharmabcine Inc. Combination therapy of a pd-1 antagonist and an antagonist for vegfr-2 for treating patients with cancer
JP2023542490A (en) 2020-09-03 2023-10-10 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Method of treating cancer pain by administering a PD-1 inhibitor
JP2023541853A (en) * 2020-09-08 2023-10-04 ユーティレックス カンパニー リミテッド PD-1 polypeptide variants
BR112023005377A2 (en) 2020-09-24 2023-04-25 Merck Sharp & Dohme Llc STABLE FORMULATIONS OF PROGRAMMED DEATH 1 (PD-1) RECEPTOR ANTIBODIES AND HYALURONIDASE VARIANTS AND FRAGMENTS THEREOF, AND METHODS OF USE THEREOF
US20230374160A1 (en) 2020-10-02 2023-11-23 Regeneron Pharmaceuticals, Inc. Combination of antibodies for treating cancer with reduced cytokine release syndrome
CA3199095A1 (en) 2020-11-06 2022-05-12 Novartis Ag Cd19 binding molecules and uses thereof
CA3200878A1 (en) 2020-11-12 2022-05-19 Inserm (Institut National De La Sante Et De La Recherche Medicale) Antibodies conjugated or fused to the receptor-binding domain of the sars-cov-2 spike protein and uses thereof for vaccine purposes
WO2022101463A1 (en) 2020-11-16 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of the last c-terminal residues m31/41 of zikv m ectodomain for triggering apoptotic cell death
WO2022118197A1 (en) 2020-12-02 2022-06-09 Pfizer Inc. Time to resolution of axitinib-related adverse events
EP4259149A1 (en) 2020-12-08 2023-10-18 Infinity Pharmaceuticals, Inc. Eganelisib for use in the treatment of pd-l1 negative cancer
KR20220082558A (en) 2020-12-10 2022-06-17 재단법인 의약바이오컨버젼스연구단 Novel fragmented cysteinyl-tRNA synthetase peptide with immune-enhancing activity and use thereof
WO2022162569A1 (en) 2021-01-29 2022-08-04 Novartis Ag Dosage regimes for anti-cd73 and anti-entpd2 antibodies and uses thereof
WO2022165403A1 (en) 2021-02-01 2022-08-04 Yale University Chemotherapeutic bioadhesive particles with immunostimulatory molecules for cancer treatment
TW202241494A (en) 2021-02-10 2022-11-01 大陸商同潤生物醫藥(上海)有限公司 A method and combination for treating tumors
GB202102396D0 (en) 2021-02-19 2021-04-07 Adc Therapeutics Sa Molecular adjuvant
WO2022187419A1 (en) 2021-03-03 2022-09-09 The Regents Of The University Of Michigan Small molecule degraders of androgen receptor
WO2022187423A1 (en) 2021-03-03 2022-09-09 The Regents Of The University Of Michigan Cereblon ligands
JP2024510947A (en) 2021-03-05 2024-03-12 レアダルティス、ソシエダッド リミターダ Trimeric polypeptides and their use in cancer therapy
KR20230159590A (en) 2021-03-23 2023-11-21 리제너론 파아마슈티컬스, 인크. Method for treating cancer in immunosuppressed or immunocompromised patients by administering PD-1 inhibitors
KR20240005700A (en) 2021-03-29 2024-01-12 주노 쎄러퓨티크스 인코퍼레이티드 Dosing and Treatment Methods Using Combination of Checkpoint Inhibitor Therapy and CAR T Cell Therapy
TW202304979A (en) 2021-04-07 2023-02-01 瑞士商諾華公司 USES OF ANTI-TGFβ ANTIBODIES AND OTHER THERAPEUTIC AGENTS FOR THE TREATMENT OF PROLIFERATIVE DISEASES
GB2623199A (en) 2021-04-08 2024-04-10 Marengo Therapeutics Inc Multifunctional molecules binding to TCR and uses thereof
EP4322938A1 (en) 2021-04-14 2024-02-21 Institut National de la Santé et de la Recherche Médicale (INSERM) New method to improve nk cells cytotoxicity
BR112023021475A2 (en) 2021-04-16 2023-12-19 Novartis Ag ANTIBODY-DRUG CONJUGATES AND METHODS FOR PRODUCING THEM
AR125874A1 (en) 2021-05-18 2023-08-23 Novartis Ag COMBINATION THERAPIES
WO2022242737A1 (en) 2021-05-21 2022-11-24 天津立博美华基因科技有限责任公司 Pharmaceutical combination and use thereof
CN117412767A (en) 2021-05-25 2024-01-16 雪绒花免疫公司 C-X-C motif chemokine receptor 6 (CXCR 6) binding molecules and methods of use thereof
BR112023024775A2 (en) 2021-05-26 2024-02-15 Ct Inmunologia Molecular USE OF A THERAPEUTIC COMPOSITION, AND PATIENT STRATIFICATION METHOD
WO2022254337A1 (en) 2021-06-01 2022-12-08 Novartis Ag Cd19 and cd22 chimeric antigen receptors and uses thereof
TW202313117A (en) 2021-06-03 2023-04-01 美商欣爍克斯公司 Head and neck cancer combination therapy comprising an il-2 conjugate and cetuximab
WO2023280790A1 (en) 2021-07-05 2023-01-12 INSERM (Institut National de la Santé et de la Recherche Médicale) Gene signatures for predicting survival time in patients suffering from renal cell carcinoma
WO2023004287A1 (en) 2021-07-19 2023-01-26 Regeneron Pharmaceuticals, Inc. Combination of checkpoint inhibitors and an oncolytic virus for treating cancer
AU2022320051A1 (en) 2021-07-30 2024-01-25 ONA Therapeutics S.L. Anti-cd36 antibodies and their use to treat cancer
WO2023015198A1 (en) 2021-08-04 2023-02-09 Genentech, Inc. Il15/il15r alpha heterodimeric fc-fusion proteins for the expansion of nk cells in the treatment of solid tumours
IL310773A (en) 2021-09-02 2024-04-01 Deutsches Krebsforschungszentrum Stiftung Des ?Ffentlichen Rechts Anti-cecam6 antibodies with reduced side-effects
AU2022341239A1 (en) 2021-09-08 2024-03-21 Redona Therapeutics, Inc. Papd5 and/or papd7 inhibiting 4-oxo-1,4-dihydroquinoline-3-carboxylic acid derivatives
TW202327595A (en) 2021-10-05 2023-07-16 美商輝瑞大藥廠 Combinations of azalactam compounds for the treatment of cancer
WO2023077034A1 (en) 2021-10-28 2023-05-04 Lyell Immunopharma, Inc. Methods for culturing immune cells
WO2023079428A1 (en) 2021-11-03 2023-05-11 Pfizer Inc. Combination therapies using tlr7/8 agonist
WO2023088968A1 (en) 2021-11-17 2023-05-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Universal sarbecovirus vaccines
WO2023111203A1 (en) 2021-12-16 2023-06-22 Onxeo New conjugated nucleic acid molecules and their uses
WO2023122573A1 (en) 2021-12-20 2023-06-29 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
WO2023130081A1 (en) 2021-12-30 2023-07-06 Neoimmunetech, Inc. Method of treating a tumor with a combination of il-7 protein and vegf antagonist
WO2023133280A1 (en) 2022-01-07 2023-07-13 Regeneron Pharmaceuticals, Inc. Methods of treating recurrent ovarian cancer with bispecific anti-muc16 x anti-cd3 antibodies alone or in combination with anti-pd-1 antibodies
CN114432441B (en) * 2022-01-11 2023-04-25 中国人民解放军陆军军医大学第一附属医院 PD-1 modified gold composite copper selenide nanoparticle and application thereof in mediating photothermal targeting treatment of cancer
WO2023154799A1 (en) 2022-02-14 2023-08-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Combination immunotherapy for treating cancer
TW202342474A (en) 2022-02-14 2023-11-01 美商基利科學股份有限公司 Antiviral pyrazolopyridinone compounds
WO2023159102A1 (en) 2022-02-17 2023-08-24 Regeneron Pharmaceuticals, Inc. Combinations of checkpoint inhibitors and oncolytic virus for treating cancer
WO2023161453A1 (en) 2022-02-24 2023-08-31 Amazentis Sa Uses of urolithins
WO2023177772A1 (en) 2022-03-17 2023-09-21 Regeneron Pharmaceuticals, Inc. Methods of treating recurrent epithelioid sarcoma with bispecific anti-muc16 x anti-cd3 antibodies alone or in combination with anti-pd-1 antibodies
WO2023192478A1 (en) 2022-04-01 2023-10-05 Bristol-Myers Squibb Company Combination therapy with anti-il-8 antibodies and anti-pd-1 antibodies for treating cancer
WO2023196988A1 (en) 2022-04-07 2023-10-12 Modernatx, Inc. Methods of use of mrnas encoding il-12
WO2023214325A1 (en) 2022-05-05 2023-11-09 Novartis Ag Pyrazolopyrimidine derivatives and uses thereof as tet2 inhibitors
WO2023224912A1 (en) 2022-05-16 2023-11-23 Regeneron Pharmaceuticals, Inc. Methods of treating metastatic castration-resistant prostate cancer with bispecific anti-psma x anti-cd3 antibodies alone or in combination with anti-pd-1 antibodies
WO2023230554A1 (en) 2022-05-25 2023-11-30 Pfizer Inc. Combination of a braf inhibitor, an egfr inhibitor, and a pd-1 antagonist for the treatment of braf v600e-mutant, msi-h/dmmr colorectal cancer
WO2023250400A1 (en) 2022-06-22 2023-12-28 Juno Therapeutics, Inc. Treatment methods for second line therapy of cd19-targeted car t cells
WO2024015803A2 (en) 2022-07-11 2024-01-18 Autonomous Therapeutics, Inc. Encrypted rna and methods of its use
US20240043560A1 (en) 2022-08-02 2024-02-08 Regeneron Pharmaceuticals, Inc. Methods of Treating Metastatic Castration-Resistant Prostate Cancer with Bispecific Anti-PSMA x Anti-CD28 Antibodies in Combination with Anti-PD-1 Antibodies
US20240041929A1 (en) 2022-08-05 2024-02-08 Juno Therapeutics, Inc. Chimeric antigen receptors specific for gprc5d and bcma
WO2024040175A1 (en) 2022-08-18 2024-02-22 Pulmatrix Operating Company, Inc. Methods for treating cancer using inhaled angiogenesis inhibitor
WO2024040264A1 (en) 2022-08-19 2024-02-22 Massachusetts Institute Of Technology Compositions and methods for targeting dendritic cell lectins
WO2024052356A1 (en) 2022-09-06 2024-03-14 Institut National de la Santé et de la Recherche Médicale Inhibitors of the ceramide metabolic pathway for overcoming immunotherapy resistance in cancer

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5190929A (en) 1988-05-25 1993-03-02 Research Corporation Technologies, Inc. Cyclophosphamide analogs useful as anti-tumor agents
WO2001014557A1 (en) 1999-08-23 2001-03-01 Dana-Farber Cancer Institute, Inc. Pd-1, a receptor for b7-4, and uses therefor
WO2002079499A1 (en) 2001-04-02 2002-10-10 Wyeth Pd-1, a receptor for b7-4, and uses therefor
WO2003099196A2 (en) 2002-05-23 2003-12-04 Cure Tech Ltd. Humanized immunomodulatory monoclonal antibodies for the treatment of neoplastic disease or immunodeficiency
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
WO2004072286A1 (en) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
US6803192B1 (en) 1999-11-30 2004-10-12 Mayo Foundation For Medical Education And Research B7-H1, a novel immunoregulatory molecule
WO2006012232A1 (en) 2004-06-24 2006-02-02 Mayo Foundation For Medical Education And Research B7-h5, a costimulatory polypeptide
US20060099203A1 (en) 2004-11-05 2006-05-11 Pease Larry R B7-DC binding antibody
US7052694B2 (en) 2002-07-16 2006-05-30 Mayo Foundation For Medical Education And Research Dendritic cell potentiation
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
WO2006133396A2 (en) 2005-06-08 2006-12-14 Dana-Farber Cancer Institute Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (pd-1) pathway
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2007056539A2 (en) 2005-11-08 2007-05-18 Medarex, Inc. Prophylaxis and treatment of enterocolitis associated with anti-ctla-4 antibody therapy
US20070166281A1 (en) 2004-08-21 2007-07-19 Kosak Kenneth M Chloroquine coupled antibodies and other proteins with methods for their synthesis
US20070202077A1 (en) 2005-12-02 2007-08-30 Brodsky Robert A Use of High-Dose Oxazaphosphorine Drugs for Treating Immune Disorders
WO2008083174A2 (en) 2006-12-27 2008-07-10 Emory University Compositions and methods for the treatment of infections and tumors
US7411051B2 (en) 1997-03-07 2008-08-12 Human Genome Sciences, Inc. Antibodies to HDPPA04 polypeptide
WO2009014708A2 (en) 2007-07-23 2009-01-29 Cell Genesys, Inc. Pd-1 antibodies in combination with a cytokine-secreting cell and methods of use thereof
WO2009073533A2 (en) 2007-11-30 2009-06-11 Medarex, Inc. Anti-b7h4 monoclonal antibody-drug conjugate and methods of use

Family Cites Families (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2011464A (en) 1933-03-23 1935-08-13 Winkler Alfred Device for removing from a pile of sheets the sheet lowermost at the time
US4272398A (en) 1978-08-17 1981-06-09 The United States Of America As Represented By The Secretary Of Agriculture Microencapsulation process
US4376110A (en) 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US5155020A (en) 1989-03-08 1992-10-13 Health Research Inc. Recombinant poxvirus host range selection system
US4769330A (en) 1981-12-24 1988-09-06 Health Research, Incorporated Modified vaccinia virus and methods for making and using the same
US4650764A (en) 1983-04-12 1987-03-17 Wisconsin Alumni Research Foundation Helper cell
US4861719A (en) 1986-04-25 1989-08-29 Fred Hutchinson Cancer Research Center DNA constructs for retrovirus packaging cell lines
NL8720442A (en) 1986-08-18 1989-04-03 Clinical Technologies Ass DELIVERY SYSTEMS FOR PHARMACOLOGICAL AGENTS.
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4980289A (en) 1987-04-27 1990-12-25 Wisconsin Alumni Research Foundation Promoter deficient retroviral vector
US4861627A (en) 1987-05-01 1989-08-29 Massachusetts Institute Of Technology Preparation of multiwall polymeric microcapsules
US6699475B1 (en) 1987-09-02 2004-03-02 Therion Biologics Corporation Recombinant pox virus for immunization against tumor-associated antigens
US5567584A (en) 1988-01-22 1996-10-22 Zymogenetics, Inc. Methods of using biologically active dimerized polypeptide fusions to detect PDGF
US5750375A (en) 1988-01-22 1998-05-12 Zymogenetics, Inc. Methods of producing secreted receptor analogs and biologically active dimerized polypeptide fusions
ATE140963T1 (en) 1988-01-22 1996-08-15 Zymogenetics Inc METHOD FOR PRODUCING SECRETED RECEPTOR ANALOGUES
US6018026A (en) 1988-01-22 2000-01-25 Zymogenetics, Inc. Biologically active dimerized and multimerized polypeptide fusions
US5278056A (en) 1988-02-05 1994-01-11 The Trustees Of Columbia University In The City Of New York Retroviral packaging cell lines and process of using same
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5124263A (en) 1989-01-12 1992-06-23 Wisconsin Alumni Research Foundation Recombination resistant retroviral helper cell and products produced thereby
US5225538A (en) 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
US5225336A (en) 1989-03-08 1993-07-06 Health Research Incorporated Recombinant poxvirus host range selection system
US5175099A (en) 1989-05-17 1992-12-29 Research Corporation Technologies, Inc. Retrovirus-mediated secretion of recombinant products
US5240846A (en) 1989-08-22 1993-08-31 The Regents Of The University Of Michigan Gene therapy vector for cystic fibrosis
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5283173A (en) 1990-01-24 1994-02-01 The Research Foundation Of State University Of New York System to detect protein-protein interactions
US5204243A (en) 1990-02-14 1993-04-20 Health Research Incorporated Recombinant poxvirus internal cores
US6641809B1 (en) 1990-03-26 2003-11-04 Bristol-Myers Squibb Company Method of regulating cellular processes mediated by B7 and CD28
WO1992016192A1 (en) 1991-03-15 1992-10-01 Amgen Inc. Pulmonary administration of granulocyte colony stimulating factor
US5637481A (en) 1993-02-01 1997-06-10 Bristol-Myers Squibb Company Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
CA2120234A1 (en) * 1991-10-21 1993-04-29 Jack B. Jiang 2-aminopropan-1,3-diol chemotherapeutic agents
US5932448A (en) 1991-11-29 1999-08-03 Protein Design Labs., Inc. Bispecific antibody heterodimers
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
TW225528B (en) 1992-04-03 1994-06-21 Ciba Geigy Ag
US5861310A (en) 1993-11-03 1999-01-19 Dana-Farber Cancer Institute Tumor cells modified to express B7-2 with increased immunogenicity and uses therefor
US5942607A (en) 1993-07-26 1999-08-24 Dana-Farber Cancer Institute B7-2: a CTLA4/CD28 ligand
US5451569A (en) 1994-04-19 1995-09-19 Hong Kong University Of Science And Technology R & D Corporation Limited Pulmonary drug delivery system
US5632983A (en) 1994-11-17 1997-05-27 University Of South Florida Method for treating secondary immunodeficiency
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5675848A (en) 1995-10-18 1997-10-14 Mallinckrodt Medical, Inc. Inflatable blanket having perforations of different sizes
US6750334B1 (en) 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
AU737910B2 (en) 1997-01-31 2001-09-06 Regents Of The University Of California, The Chimeric antibody fusion proteins for the recruitment and stimulation of an antitumor immune response
US20060223088A1 (en) 1997-03-07 2006-10-05 Rosen Craig A Human secreted proteins
US20070224663A1 (en) 1997-03-07 2007-09-27 Human Genome Sciences, Inc. Human Secreted Proteins
US7368531B2 (en) 1997-03-07 2008-05-06 Human Genome Sciences, Inc. Human secreted proteins
US6468546B1 (en) 1998-12-17 2002-10-22 Corixa Corporation Compositions and methods for therapy and diagnosis of ovarian cancer
WO2001001137A1 (en) 1999-06-30 2001-01-04 Children's Medical Center Corporation Fusion protein and uses thereof
EP1074617A3 (en) 1999-07-29 2004-04-21 Research Association for Biotechnology Primers for synthesising full-length cDNA and their use
AR034118A1 (en) 2000-02-15 2004-02-04 Sugen Inc COMPOUNDS OF 2-INDOLINONES REPLACED WITH PROTEINQUINASE INHIBITING PIRROLS; YOUR PHARMACEUTICAL AND INTERMEDIARY SYNTHESIS COMPOSITIONS
WO2001079442A2 (en) 2000-04-12 2001-10-25 Human Genome Sciences, Inc. Albumin fusion proteins
US7030219B2 (en) * 2000-04-28 2006-04-18 Johns Hopkins University B7-DC, Dendritic cell co-stimulatory molecules
US20030031675A1 (en) * 2000-06-06 2003-02-13 Mikesell Glen E. B7-related nucleic acids and polypeptides useful for immunomodulation
MXPA02012106A (en) 2000-06-06 2003-06-06 Bristol Myers Squibb Co B7-related nucleic acids and polypeptides useful for immunomodulation.
JP2003535907A (en) * 2000-06-22 2003-12-02 ユニバーシティ オブ アイオワ リサーチ ファウンデーション Method for promoting antibody-induced cell lysis and treating cancer
CA2414331C (en) 2000-06-28 2011-11-29 Genetics Institute, Llc. Pd-l2 molecules: novel pd-1 ligands and uses therefor
EP1301524A1 (en) 2000-06-30 2003-04-16 Human Genome Sciences, Inc. B7-like polynucleotides, polypeptides, and antibodies
US6635750B1 (en) 2000-07-20 2003-10-21 Millennium Pharmaceuticals, Inc. B7-H2 nucleic acids, members of the B7 family
MXPA03002413A (en) 2000-09-20 2003-06-19 Amgen Inc B7-like molecules and uses thereof.
US7182942B2 (en) 2000-10-27 2007-02-27 Irx Therapeutics, Inc. Vaccine immunotherapy for immune suppressed patients
US7408041B2 (en) 2000-12-08 2008-08-05 Alexion Pharmaceuticals, Inc. Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
US6562576B2 (en) 2001-01-04 2003-05-13 Myriad Genetics, Inc. Yeast two-hybrid system and use thereof
US6743619B1 (en) 2001-01-30 2004-06-01 Nuvelo Nucleic acids and polypeptides
AR036993A1 (en) 2001-04-02 2004-10-20 Wyeth Corp USE OF AGENTS THAT MODULATE THE INTERACTION BETWEEN PD-1 AND ITS LINKS IN THE SUBMODULATION OF IMMUNOLOGICAL ANSWERS
US20060084794A1 (en) 2001-04-12 2006-04-20 Human Genome Sciences, Inc. Albumin fusion proteins
WO2002086083A2 (en) 2001-04-20 2002-10-31 Mayo Foundation For Medical Education And Research Methods of enhancing cell responsiveness
GB0121285D0 (en) * 2001-09-03 2001-10-24 Cancer Res Ventures Ltd Anti-cancer combinations
JP2005506073A (en) 2001-10-19 2005-03-03 ザイモジェネティクス,インコーポレイティド Dimerized growth factors and materials and methods for producing the same
EP1456652A4 (en) 2001-11-13 2005-11-02 Dana Farber Cancer Inst Inc Agents that modulate immune cell activation and methods of use thereof
US7164500B2 (en) 2002-01-29 2007-01-16 Hewlett-Packard Development Company, L.P. Method and apparatus for the automatic generation of image capture device control marks
NZ538628A (en) 2002-08-12 2008-06-30 Dynavax Tech Corp Immunomodulatory compositions, methods of making, and methods of use thereof
US7432351B1 (en) 2002-10-04 2008-10-07 Mayo Foundation For Medical Education And Research B7-H1 variants
CA2516834C (en) 2003-02-27 2013-07-16 Theravision Gmbh Polypeptides and methods for making the same
EP1668031B1 (en) 2003-08-07 2008-03-12 ZymoGenetics, Inc. Homogeneous preparations of il-29
WO2005087810A2 (en) 2004-03-08 2005-09-22 Zymogenetics, Inc. Dimeric fusion proteins and materials and methods for producing them
EP1773318A1 (en) * 2004-07-23 2007-04-18 OM Pharma Combination anticancer therapy and pharmaceutical compositions therefor
SI1810026T1 (en) * 2004-10-06 2018-08-31 Mayo Foundation For Medical Education And Research B7-h1 and pd-1 in treatment of renal cell carcinoma
CA2585776A1 (en) 2004-10-29 2006-05-11 University Of Southern California Combination cancer immunotherapy with co-stimulatory molecules
GB0519303D0 (en) 2005-09-21 2005-11-02 Oxford Biomedica Ltd Chemo-immunotherapy method
US20070231344A1 (en) * 2005-10-28 2007-10-04 The Brigham And Women's Hospital, Inc. Conjugate vaccines for non-proteinaceous antigens
EP1954311A4 (en) 2005-12-07 2009-12-23 Medarex Inc Ctla-4 antibody dosage escalation regimens
JP5564181B2 (en) 2005-12-08 2014-07-30 シルワン,ハヴァル Immune stimulating compositions and methods
WO2008085562A2 (en) 2006-09-20 2008-07-17 The Johns Hopkins University Combinatorieal therapy of cancer and infectious diseases with anti-b7-h1 antibodies
WO2008037080A1 (en) 2006-09-29 2008-04-03 Universite De Montreal Methods and compositions for immune response modulation and uses thereof
WO2008100562A2 (en) * 2007-02-14 2008-08-21 Medical College Of Georgia Research Institute, Inc. Indoleamine 2,3-dioxygenase, pd-1/pd-l pathways, and ctla4 pathways in the activation of regulatory t cells
AU2008293885A1 (en) 2007-07-13 2009-03-05 The John Hopkins University B7-DC variants
WO2009023566A2 (en) 2007-08-09 2009-02-19 Genzyme Corporation Method of treating autoimmune disease with mesenchymal stem cells
US20110008332A1 (en) 2007-10-31 2011-01-13 The Scripps Research Institute Combination Therapy to Treat Persistent Viral Infections
RU2531758C2 (en) 2008-02-11 2014-10-27 Куретек Лтд. Monoclonal antibodies for tumour treatment
AU2009223784A1 (en) 2008-03-08 2009-09-17 Immungene, Inc. Engineered fusion molecules immunotherapy in cancer and inflammatory diseases
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
DK2113253T3 (en) 2008-04-30 2010-07-19 Immatics Biotechnologies Gmbh New Composition of Tumor-Associated Peptides Binding to Human Leukocyte Antigen (HLA) Class I or II Molecules for Vaccine Use
US20110223188A1 (en) 2008-08-25 2011-09-15 Solomon Langermann Targeted costimulatory polypeptides and methods of use to treat cancer
AU2009288730B2 (en) 2008-08-25 2013-06-20 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
PL2376535T3 (en) 2008-12-09 2017-09-29 F.Hoffmann-La Roche Ag Anti-pd-l1 antibodies and their use to enhance t-cell function
JP2013512251A (en) 2009-11-24 2013-04-11 アンプリミューン、インコーポレーテッド Simultaneous inhibition of PD-L1 / PD-L2

Patent Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5190929A (en) 1988-05-25 1993-03-02 Research Corporation Technologies, Inc. Cyclophosphamide analogs useful as anti-tumor agents
US7411051B2 (en) 1997-03-07 2008-08-12 Human Genome Sciences, Inc. Antibodies to HDPPA04 polypeptide
WO2001014557A1 (en) 1999-08-23 2001-03-01 Dana-Farber Cancer Institute, Inc. Pd-1, a receptor for b7-4, and uses therefor
US6803192B1 (en) 1999-11-30 2004-10-12 Mayo Foundation For Medical Education And Research B7-H1, a novel immunoregulatory molecule
WO2002079499A1 (en) 2001-04-02 2002-10-10 Wyeth Pd-1, a receptor for b7-4, and uses therefor
US20080274490A1 (en) 2001-04-02 2008-11-06 Dana-Farber Cancer Institute, Inc. PD-1, a receptor for B7-4, and uses therefor
US7105328B2 (en) 2001-04-02 2006-09-12 Dana-Farber Cancer Institute Methods for screening for compounds that modulate pd-1 signaling
WO2003099196A2 (en) 2002-05-23 2003-12-04 Cure Tech Ltd. Humanized immunomodulatory monoclonal antibodies for the treatment of neoplastic disease or immunodeficiency
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
US20060110383A1 (en) 2002-07-03 2006-05-25 Tasuku Honjo Immunopotentiative composition
US7390888B2 (en) 2002-07-16 2008-06-24 Mayo Foundation For Medical Education And Research Dendritic cell potentiation
US7052694B2 (en) 2002-07-16 2006-05-30 Mayo Foundation For Medical Education And Research Dendritic cell potentiation
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
WO2004072286A1 (en) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
WO2006012232A1 (en) 2004-06-24 2006-02-02 Mayo Foundation For Medical Education And Research B7-h5, a costimulatory polypeptide
US20070166281A1 (en) 2004-08-21 2007-07-19 Kosak Kenneth M Chloroquine coupled antibodies and other proteins with methods for their synthesis
US20060099203A1 (en) 2004-11-05 2006-05-11 Pease Larry R B7-DC binding antibody
WO2006121168A1 (en) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
WO2006133396A2 (en) 2005-06-08 2006-12-14 Dana-Farber Cancer Institute Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (pd-1) pathway
WO2007005874A2 (en) 2005-07-01 2007-01-11 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
WO2007056539A2 (en) 2005-11-08 2007-05-18 Medarex, Inc. Prophylaxis and treatment of enterocolitis associated with anti-ctla-4 antibody therapy
US20070202077A1 (en) 2005-12-02 2007-08-30 Brodsky Robert A Use of High-Dose Oxazaphosphorine Drugs for Treating Immune Disorders
WO2008083174A2 (en) 2006-12-27 2008-07-10 Emory University Compositions and methods for the treatment of infections and tumors
WO2009014708A2 (en) 2007-07-23 2009-01-29 Cell Genesys, Inc. Pd-1 antibodies in combination with a cytokine-secreting cell and methods of use thereof
WO2009073533A2 (en) 2007-11-30 2009-06-11 Medarex, Inc. Anti-b7h4 monoclonal antibody-drug conjugate and methods of use

Non-Patent Citations (80)

* Cited by examiner, † Cited by third party
Title
BAJORATH ET AL., J. MOL. GRAPH. MODEL., vol. 15, 1997, pages 135 - 139
BAS; MASTRANGELO, CANCER IMMUNOL. IMMUNOTHER, vol. 47, 1998, pages 1 - 12
BASS KK; MASTRANGELO MJ: "Immunopotentiation with low-dose cyclophosphamide in the active specific immunotherapy of cancer", CANCER IMMUNOL.IMMUNOTHER, vol. 47, no. 1, September 1998 (1998-09-01), pages 1 - 12
BASS,K.K.; MASTRANGELO,M.J.: "Immunopotentiation with low-dose cyclophosphamide in the active specific immunotherapy of cancer", CANCER IMMUNOL. IMMUNOTHER., vol. 47, 1998, pages 1 - 12
BERGER ET AL., CLIN. CANCER RES., vol. 14, 2008, pages 3044 - 3051
BRODE S; COOKE A: "Immune-potentiating effects of the chemotherapeutic drug cyclophosphamide", CRIT REV.LMMUNOL., vol. 28, no. 2, 2008, pages 109 - 26
BRODE,S.; COOKE,A: "Immune-potentiating effects of the chemotherapeutic drug cyclophosphamide", CRIT REV. IMMUNOL., vol. 28, 2008, pages 109 - 126
BRODE; COOKE, CRIT REV. IMMUNOL., vol. 28, 2008, pages 109 - 126
BUTTE ET AL., IMMUNITY, vol. 27, 2007, pages 111 - 122
CHAMBERS; ALLISON, CURR. OPIN. IMMUNOL., vol. 9, 1997, pages 396 - 404
CHAPOVAL ET AL., NATURE IMMUNOL., vol. 2, 2001, pages 269 - 274
CHOI ET AL., J. IMMUNOL., vol. 171, 2003, pages 4650 - 4654
CUBILLOS-RUIZ ET AL., J. CLIN. INVEST., vol. 119, no. 8, 2009, pages 2231 - 2244
DONG ET AL., NATURE MED., vol. 5, 1999, pages 1365 - 1369
ERBE ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 7363 - 7368
FREEMAN ET AL., J. EXP. MED., vol. 192, 2000, pages 1 - 9
FREEMAN ET AL., J. EXP. MED., vol. 192, 2000, pages 1027 - 1034
FREEMAN, PROC. NATL. ACAD. SCI. U. S. A, vol. 105, 2008, pages 10275 - 10276
FREEMAN,G.J.: "Structures of PD-1 with its ligands: sideways and dancing cheek to cheek", PROC. NATL. ACAD. SCI. U. S. A, vol. 105, 2008, pages 10275 - 10276
HENGST ET AL., CANCER RES., vol. 41, 1981, pages 2163 - 2167
HENGST JC; MOKYR MB; DRAY S: "Cooperation between cyclophosphamide tumoricidal activity and host antitumor immunity in the cure of mice bearing large MOPC-315 tumors", CANCER RES., vol. 41, no. 6, June 1981 (1981-06-01), pages 2163 - 7
HENGST JC; MOKYR MB; DRAY S: "Importance of timing in cyclophosphamide therapy of MOPC-315 tumor-bearing mice", CANCER RES., vol. 40, no. 7, July 1980 (1980-07-01), pages 2135 - 41
HENGST, CANCER RES., vol. 40, 1980, pages 2135 - 2141
HENGST,J.C.; MOKYR,M.B.; DRAY,S.: "Cooperation between cyclophosphamide tumoricidal activity and host antitumor immunity in the cure of mice bearing large MOPC-315 tumors", CANCER RES., vol. 41, 1981, pages 2163 - 2167
HENGST,J.C.; MOKYR,M.B.; DRAY,S.: "Importance of timing in cyclophosphamide therapy of MOPC-315 tumor-bearing mice", CANCER RES., vol. 40, 1980, pages 2135 - 2141
HIRANO F. ET AL., CANCER RESEARCH, 2005
HOCHMAN, J. ET AL., BIOCHEMISTRY, vol. 12, 1973, pages 1130 - 1135
HONEYCHURCH J; GLENNIE MJ; ILLIDGE TM: "Cyclophosphamide inhibition of anti-CD40 monoclonal antibody-based therapy of B cell lymphoma is dependent on CD11b+ cells", CANCER RES., vol. 65, no. 16, 15 August 2005 (2005-08-15), pages 7493 - 501
HONEYCHURCH,J.; GLENNIE,M.J.; ILIIDGE,T.M.: "Cyclophosphamide inhibition of anti-CD40 monoclonal antibody-based therapy of B cell lymphoma is dependent on CD11 b+ cells", CANCER RES., vol. 65, 2005, pages 7493 - 7501
IKEMIZU ET AL., IMMUNITY, vol. 12, 2000, pages 51 - 60
ISHIDA ET AL., EMBO J., vol. 11, 1992, pages 3887 - 3895
KABAT ET AL., J. BIOL. CHEM., vol. 252, 1977, pages 6609 - 6616
KEIR ET AL., CURR. OPIN. IMMUNOL., vol. 19, 2007, pages 309 - 314
KRUMMEL; ALLISON, J. EXP. MED., vol. 183, 1996, pages 2533 - 2540
LATCHMAN ET AL., NATURE IMMUNOL., vol. 2, 2001, pages 261 - 268
LAZAR MOLNAR ET AL., PNAS, vol. 105, 2008, pages 10483 - 10488
LENSHOW ET AL., ANNU. REV. IMMUNOL., vol. 14, 1996, pages 233 - 258
LI ET AL.: "Vascular endothelial growth factor blockade reduces intratumoral regulatory T cells and enhances the efficacy of a GM-CSF-secreting cancer immunotherapy", CLIN CANCER RES., vol. 12, no. 22, 15 November 2006 (2006-11-15), pages 6808 - 16
LIANG J; HUANG M; DUAN W; YU XQ; ZHOU S: "Design of new oxazaphosphorine anticancer drugs", CURR PHARM DES., vol. 13, no. 9, 2007, pages 963 - 78
MACHIELS ET AL., CANCER RES., vol. 61, 2001, pages 3689 - 3697
MACHIELS JP; REILLY RT; EMENS LA; ERCOLINI AM; LEI RY; WEINTRAUB D; OKOYE FI; JAFFEE EM: "Cyclophosphamide, doxorubicin, and paclitaxel enhance the antitumor immune response of granulocyte/macrophage-colony stimulating factor-secreting whole-cell vaccines in HER-2/neu tolerized mice", CANCER RES., vol. 61, no. 9, 1 May 2001 (2001-05-01), pages 3689 - 97
MACHIELS,J.P. ET AL.: "Cyclophosphamide, doxorubicin, and paclitaxel enhance the antitumor immune response of granulocyte/macrophage-colony stimulating factor-secreting whole-cell vaccines in HER-2/neu tolerized mice", CANCER RES., vol. 61, 2001, pages 3689 - 3697
MOLNAR ET AL.: "Crystal structure of the complex between programmed death-1 (PD-1) and its ligand PD-L2", PNAS, vol. 105, 29 July 2008 (2008-07-29), pages 10483 - 10488
NISHIMURA ET AL., IMMUNITY, vol. 11, 1999, pages 141 - 151
NISHIMURA ET AL., SCIENCE, vol. 291, 2001, pages 319 - 322
ONLAMOON ET AL., IMMUNOLOGY, vol. 124, 2008, pages 277 - 293
OSTROV ET AL., SCIENCE, vol. 290, 2000, pages 816 - 819
PERTOVAS ET AL., J. EXP. MED., vol. 203, 2006, pages 2281
PRASAD ET AL., IMMUNITY, vol. 18, 2003, pages 863 - 873
RATHMELL; THOMPSON, ANNU. REV. IMMUNOL., vol. 17, 1999, pages 781 - 828
ROUSSEAUX ET AL., METH. ENZYMOL., vol. 121, 1986, pages 663 - 69
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual, Second Edition,", 1989, COLD SPRING HARBOR
SCHWARTZ ET AL., NATURE IMMUNOL., vol. 3, 2002, pages 427 - 434
SCHWARTZ ET AL., NATURE, vol. 410, 2001, pages 604 - 608
See also references of EP2350129A4
SHARON, J. ET AL., BIOCHEMISTRY, vol. 15, 1976, pages 1591 - 1594
SICA ET AL., IMMUNITY, vol. 18, 2003, pages 849 - 861
STAMPER ET AL., NATURE, vol. 410, 2001, pages 608 - 611
SWALLOW ET AL., IMMUNITY, vol. 11, 1999, pages 423 - 432
TAIEB J; CHAPUT N; SCHARTZ N; ROUX S; NOVAULT S; MENARD C; GHIRINGHELLI F; TERME M; CARPENTIER AF; DARRASSE-JEZE G ET AL.: "Chemoimmunotherapy of tumors: cyclophosphamide synergizes with exosome based vaccines", J.IMMUNOL., vol. 176, no. 5, 1 March 2006 (2006-03-01), pages 2722 - 9
TAIEB,J. ET AL.: "Chemoimmunotherapy of tumors: cyclophosphamide synergizes with exosome based vaccines", J. IMMUNOL., vol. 176, 2006, pages 2722 - 2729
TAIEB,J., J. IMMUNOL., vol. 176, 2006, pages 2722 - 2729
TAMURA, BLOOD, vol. 97, 2001, pages 1809 - 1816
TSENG ET AL., J. EXP. MED., vol. 193, 2001, pages 839 - 846
TSUNG K; MEKO JB; TSUNG YL; PEPLINSKI GR; NORTON JA: "Immune response against large tumors eradicated by treatment with cyclophosphamide and IL-12", J.IMMUNOL., vol. 160, no. 3, 1 February 1998 (1998-02-01), pages 1369 - 77
TSUNG,K.; MEKO,J.B.; TSUNG,Y.L.; PEPLINSKI,G.R.; NORTON,J.A.: "Immune response against large tumors eradicated by treatment with cyclophosphamide and IL-12", J. IMMUNOL., vol. 160, 1998, pages 1369 - 1377
TUAN,: "Methods in Molecular Biology", vol. 62, 1997, HUMANA PRESS, article "Recombinant Gene Expression Protocols"
VAN DER MOST ET AL., CANCER IMMUNOL. IMMUNOTHER, vol. 58, 2009, pages 1219 - 1228
VAN DER MOST RG; CURRIE AJ; MAHENDRAN S; PROSSER A; DARABI A; ROBINSON BW; NOWAK AK; LAKE RA: "Tumor eradication after cyclophosphamide depends on concurrent depletion of regulatory T cells: a role for cycling TNFR2-expressing effector-suppressor T cells in limiting effective chemotherapy", CANCER LMMUNOL. LMMUNOTHER., vol. 58, no. 8, August 2009 (2009-08-01), pages 1219 - 28
VAN DER MOST,R.G. ET AL.: "Tumor eradication after cyclophosphamide depends on concurrent depletion of regulatory T cells: a role for cycling TNFR2-expressing effector-suppressor T cells in limiting effective chemotherapy", CANCER IMMUNOL. IMMUNOTHER., vol. 58, 2009, pages 1219 - 1228
WADA S; YOSHIMURA K; HIPKISS EL; HARRIS TJ; YEN HR; GOLDBERG MV; GROSSO JF; GETNET D; DEMARZO AM; NETTO GJ: "Cyclophosphamide augments antitumor immunity: studies in an autochthonous prostate cancer model", CANCER RES., vol. 69, no. 10, 15 May 2009 (2009-05-15), pages 4309 - 18
WAHL ET AL., J. NUC. MED., vol. 24, 1983, pages 316 - 325
WALUNAS ET AL., J. EXP. MED., vol. 183, 1996, pages 2541 - 2550
WAND ET AL., J. EXP. MED., vol. 195, 2002, pages 1033 - 1041
WANG ET AL., BLOOD, vol. 96, 2000, pages 2808 - 2813
WILLIAMS; BARCLAY, ANNU. REV. IMMUNOL., vol. 6, 1988, pages 381 - 405
WU ET AL.: "Methods in Gene Biotechnology", 1997, CRC PRESS
YOSHIMURA K ET AL., CANCER RESEARCH, 2007
YOSHINAGA ET AL., NATURE, vol. 402, 1999, pages 827 - 832
ZANG ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 100, 2003, pages 10388 - 10392

Cited By (118)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
US9370565B2 (en) 2000-04-28 2016-06-21 The Johns Hopkins University Dendritic cell co-stimulatory molecules
US8039589B1 (en) 2002-10-04 2011-10-18 Mayo Foundation For Medical Education And Research B7-DC variants
US8273864B2 (en) 2002-10-04 2012-09-25 Mayo Foundation For Medical Education And Research Nucleic acid molecules encoding B7-DC variants
US11939378B2 (en) 2004-10-06 2024-03-26 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US8747833B2 (en) 2004-10-06 2014-06-10 Mayo Foundation For Medical Education And Research B7-H1 and methods of diagnosis, prognosis, and treatment of cancer
US11242387B2 (en) 2004-10-06 2022-02-08 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US9803015B2 (en) 2004-10-06 2017-10-31 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US8445447B2 (en) 2007-07-13 2013-05-21 The Johns Hopkins University B7-DC variants immunogenic compositions and methods of use thereof
US8609089B2 (en) 2008-08-25 2013-12-17 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
US8114845B2 (en) 2008-08-25 2012-02-14 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
EP2504028A4 (en) * 2009-11-24 2014-04-09 Amplimmune Inc Simultaneous inhibition of pd-l1/pd-l2
EP2504028A2 (en) * 2009-11-24 2012-10-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
EP2910572A1 (en) * 2010-11-11 2015-08-26 The University of Hong Kong Soluble pd-1 variants, fusion constructs, and uses thereof
CN102298053A (en) * 2011-05-20 2011-12-28 中山大学肿瘤防治中心 Composite antibody kit used in postoperative recurrence risk assessment of primary hepatocellular carcinoma
US9096642B2 (en) 2011-06-08 2015-08-04 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
WO2012168944A1 (en) * 2011-06-08 2012-12-13 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
CN103732238A (en) * 2011-06-08 2014-04-16 奥瑞基尼探索技术有限公司 Therapeutic compounds for immunomodulation
JP2014525918A (en) * 2011-08-01 2014-10-02 ジェネンテック, インコーポレイテッド Method for treating cancer using PD-1 axis binding antagonist and MEK inhibitor
US10646567B2 (en) 2011-08-01 2020-05-12 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
US9724413B2 (en) 2011-08-01 2017-08-08 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
US11065285B2 (en) 2012-01-25 2021-07-20 Dnatrix, Inc. Biomarkers and combination therapies using oncolytic virus and immunomodulation
WO2013176915A1 (en) * 2012-05-25 2013-11-28 Roman Galetto Methods for engineering allogeneic and immunosuppressive resistant t cell for immunotherapy
US11007224B2 (en) 2012-05-25 2021-05-18 Cellectis CD19 specific chimeric antigen receptor and uses thereof
US11274316B2 (en) 2012-05-25 2022-03-15 Cellectis Use of pre T alpha or functional variant thereof for expanding TCR alpha deficient T cells
US11414674B2 (en) 2012-05-25 2022-08-16 Cellectis Use of pre T alpha or functional variant thereof for expanding TCR alpha deficient T cells
US10874693B2 (en) 2012-05-25 2020-12-29 Cellectis CD19 specific chimeric antigen receptor and uses thereof
US11603539B2 (en) 2012-05-25 2023-03-14 Cellectis Methods for engineering allogeneic and immunosuppressive resistant T cell for immunotherapy
US11891614B2 (en) 2012-05-25 2024-02-06 Cellectis Methods for engineering allogeneic and immunosuppressive resistant T cell for immunotherapy
US10342829B2 (en) 2012-05-25 2019-07-09 Cellectis Multi-chain chimeric antigen receptor and uses thereof
US10517896B2 (en) 2012-05-25 2019-12-31 Cellectis Use of pre T alpha or functional variant thereof for expanding TCR alpha deficient T cells
US10363270B2 (en) 2012-05-25 2019-07-30 Cellectis Methods for engineering allogeneic and immunosuppressive resistant T cell immunotherapy
US9895441B2 (en) 2012-05-31 2018-02-20 Genentech, Inc. Methods of treating cancer using PD-L1 axis binding antagonists and VEGF antagonists
EP3275899A1 (en) 2012-07-02 2018-01-31 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
EP3795592A1 (en) 2012-07-02 2021-03-24 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
US11236175B2 (en) 2012-10-10 2022-02-01 Sangamo Therapeutics, Inc. T cell modifying compounds and uses thereof
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US11077144B2 (en) 2013-05-13 2021-08-03 Cellectis CD19 specific chimeric antigen receptor and uses thereof
WO2014194293A1 (en) 2013-05-30 2014-12-04 Amplimmune, Inc. Improved methods for the selection of patients for pd-1 or b7-h4 targeted therapies, and combination therapies thereof
EP3444271A1 (en) 2013-08-08 2019-02-20 Cytune Pharma Il-15 and il-15raplha sushi domain based modulokines
EP3995507A1 (en) 2013-08-08 2022-05-11 Cytune Pharma Il-15 and il-15ralpha sushi domain based on modulokines
EP3659622A1 (en) 2013-08-08 2020-06-03 Cytune Pharma Combined pharmaceutical composition
EP4269441A2 (en) 2013-08-08 2023-11-01 Cytune Pharma Il-15 and il-15ralpha sushi domain based on modulokines
US10994008B2 (en) 2013-08-20 2021-05-04 Merck Sharp & Dohme Corp. Modulation of tumor immunity
US10188729B2 (en) 2013-08-20 2019-01-29 Merck Sharp & Dohme Corp. Modulation of tumor immunity
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
US11708412B2 (en) 2013-09-26 2023-07-25 Novartis Ag Methods for treating hematologic cancers
US11136393B2 (en) 2013-10-01 2021-10-05 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of Bim
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
EP3087099A4 (en) * 2013-12-23 2017-07-19 Oncomed Pharmaceuticals, Inc. Immunotherapy with binding agents
US11117970B2 (en) 2014-01-23 2021-09-14 Regeneron Pharmaceuticals, Inc. Human antibodies to PD-L1
US10737113B2 (en) 2014-01-23 2020-08-11 Regeneron Pharmaceuticals, Inc. Human antibodies to PD-1
US11827704B2 (en) 2014-01-24 2023-11-28 Novartis Ag Antibody molecules to PD-1 and uses thereof
US9815898B2 (en) 2014-01-24 2017-11-14 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10752687B2 (en) 2014-01-24 2020-08-25 Novartis Ag Antibody molecules to PD-1 and uses thereof
US9683048B2 (en) 2014-01-24 2017-06-20 Novartis Ag Antibody molecules to PD-1 and uses thereof
US10472419B2 (en) 2014-01-31 2019-11-12 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US10981990B2 (en) 2014-01-31 2021-04-20 Novartis Ag Antibody molecules to TIM-3 and uses thereof
US11155620B2 (en) 2014-01-31 2021-10-26 Novartis Ag Method of detecting TIM-3 using antibody molecules to TIM-3
US10730951B2 (en) 2014-03-31 2020-08-04 Genentech, Inc. Anti-OX40 antibodies and methods of use
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US10946093B2 (en) 2014-07-15 2021-03-16 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US11504376B2 (en) 2014-07-23 2022-11-22 Mayo Foundation For Medical Education And Research Targeting DNA-PKCS and B7-H1 to treat cancer
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
US11813295B1 (en) 2014-09-18 2023-11-14 Theobald Therapeutics LLC Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US11633435B1 (en) 2014-09-18 2023-04-25 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US10828356B1 (en) 2014-09-18 2020-11-10 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US10729731B1 (en) 2014-09-18 2020-08-04 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
US11319359B2 (en) 2015-04-17 2022-05-03 Alpine Immune Sciences, Inc. Immunomodulatory proteins with tunable affinities
WO2017025498A1 (en) 2015-08-07 2017-02-16 Pieris Pharmaceuticals Gmbh Novel fusion polypeptide specific for lag-3 and pd-1
WO2017055404A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific antibodies specific for pd1 and tim3
US10287352B2 (en) 2015-10-02 2019-05-14 Hoffman-La Roche Inc. Bispecific antibodies specific for PD1 and TIM3
WO2017055443A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Anti-pd1 antibodies and methods of use
US11130810B2 (en) 2015-10-02 2021-09-28 Hoffmann-La Roche Inc. Bispecific antibodies specific for PD1 and TIM3
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1
US11359022B2 (en) 2016-04-15 2022-06-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11479609B2 (en) 2016-04-15 2022-10-25 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11498967B2 (en) 2016-04-15 2022-11-15 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11078282B2 (en) 2016-04-15 2021-08-03 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US10457725B2 (en) 2016-05-13 2019-10-29 Regeneron Pharmaceuticals, Inc. Methods of treating skin cancer by administering a PD-1 inhibitor
EP3243832A1 (en) 2016-05-13 2017-11-15 F. Hoffmann-La Roche AG Antigen binding molecules comprising a tnf family ligand trimer and pd1 binding moiety
US11505600B2 (en) 2016-05-13 2022-11-22 Regeneron Pharmaceuticals, Inc. Methods of treating skin cancer by administering a PD-1 inhibitor
US11447537B2 (en) 2016-10-27 2022-09-20 Io Biotech Aps PDL2 compounds
EP4295918A2 (en) 2016-11-02 2023-12-27 Bristol-Myers Squibb Company Bispecific antibody against bcma and cd3 and an immunological drug for combined use in treating multiple myeloma
WO2018083204A1 (en) 2016-11-02 2018-05-11 Engmab Sàrl Bispecific antibody against bcma and cd3 and an immunological drug for combined use in treating multiple myeloma
US11124577B2 (en) 2016-11-02 2021-09-21 Engmab Sàrl Bispecific antibody against BCMA and CD3 and an immunological drug for combined use in treating multiple myeloma
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
CN113354710A (en) * 2016-12-09 2021-09-07 复诺健生物科技加拿大有限公司 Compositions and methods for inhibiting CD279 interactions
WO2018134279A1 (en) 2017-01-18 2018-07-26 Pieris Pharmaceuticals Gmbh Novel fusion polypeptides specific for lag-3 and pd-1
US11096988B2 (en) 2017-03-16 2021-08-24 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11639375B2 (en) 2017-03-16 2023-05-02 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11230588B2 (en) 2017-03-16 2022-01-25 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11117949B2 (en) 2017-03-16 2021-09-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11117950B2 (en) 2017-03-16 2021-09-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11117948B2 (en) 2017-03-16 2021-09-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
US11413331B2 (en) 2017-04-03 2022-08-16 Hoffmann-La Roche Inc. Immunoconjugates
US11285207B2 (en) 2017-04-05 2022-03-29 Hoffmann-La Roche Inc. Bispecific antibodies specifically binding to PD1 and LAG3
WO2018185043A1 (en) 2017-04-05 2018-10-11 F. Hoffmann-La Roche Ag Bispecific antibodies specifically binding to pd1 and lag3
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
WO2019149716A1 (en) 2018-01-31 2019-08-08 F. Hoffmann-La Roche Ag Bispecific antibodies comprising an antigen-binding site binding to lag3
US11932681B2 (en) 2018-05-31 2024-03-19 Novartis Ag Hepatitis B antibodies
WO2019229699A1 (en) 2018-05-31 2019-12-05 Novartis Ag Hepatitis b antibodies
WO2019234576A1 (en) 2018-06-03 2019-12-12 Lamkap Bio Beta Ltd. Bispecific antibodies against ceacam5 and cd47
US11555071B2 (en) 2018-06-03 2023-01-17 Lamkap Bio Beta Ltd. Bispecific antibodies against CEACAM5 and CD47
WO2020021061A1 (en) 2018-07-26 2020-01-30 Pieris Pharmaceuticals Gmbh Humanized anti-pd-1 antibodies and uses thereof
WO2020043683A1 (en) 2018-08-27 2020-03-05 Pieris Pharmaceuticals Gmbh Combination therapies comprising cd137/her2 bispecific agents and pd-1 axis inhibitors and uses thereof
WO2020053742A2 (en) 2018-09-10 2020-03-19 Novartis Ag Anti-hla-hbv peptide antibodies
WO2021053587A1 (en) 2019-09-18 2021-03-25 Klaus Strein Bispecific antibodies against ceacam5 and cd3
EP3831849A1 (en) 2019-12-02 2021-06-09 LamKap Bio beta AG Bispecific antibodies against ceacam5 and cd47
WO2021110647A1 (en) 2019-12-02 2021-06-10 Lamkap Bio Beta Ag Bispecific antibodies against ceacam5 and cd47
US11753481B2 (en) 2020-12-18 2023-09-12 Lamkap Bio Beta Ltd Bispecific antibodies against CEACAM5 and CD47
WO2022130348A1 (en) 2020-12-18 2022-06-23 Lamkap Bio Beta Ag Bispecific antibodies against ceacam5 and cd47
WO2023012147A1 (en) 2021-08-03 2023-02-09 F. Hoffmann-La Roche Ag Bispecific antibodies and methods of use
WO2023242351A1 (en) 2022-06-16 2023-12-21 Lamkap Bio Beta Ag Combination therapy of bispecific antibodies against ceacam5 and cd47 and bispecific antibodies against ceacam5 and cd3

Also Published As

Publication number Publication date
KR20110050529A (en) 2011-05-13
PT2350129E (en) 2015-10-02
JP2012500847A (en) 2012-01-12
MX2011002252A (en) 2011-06-24
US8709416B2 (en) 2014-04-29
MA32646B1 (en) 2011-09-01
US20120114649A1 (en) 2012-05-10
SMT201500212B (en) 2015-10-30
SI2350129T1 (en) 2015-11-30
CN102203132A (en) 2011-09-28
US20100055102A1 (en) 2010-03-04
ES2545609T3 (en) 2015-09-14
PL2350129T3 (en) 2015-12-31
IL211298A0 (en) 2011-04-28
EP2350129B1 (en) 2015-06-10
EA201500417A1 (en) 2015-11-30
US8609089B2 (en) 2013-12-17
AU2009288730B2 (en) 2013-06-20
AU2009288730A1 (en) 2010-03-11
WO2010027423A3 (en) 2010-05-06
PE20110435A1 (en) 2011-07-20
EA023148B1 (en) 2016-04-29
US20120114648A1 (en) 2012-05-10
US8114845B2 (en) 2012-02-14
CO6341574A2 (en) 2011-11-21
CY1116816T1 (en) 2017-03-15
CL2011000423A1 (en) 2012-07-20
EA201170373A1 (en) 2011-10-31
DOP2011000059A (en) 2011-05-15
JP5798919B2 (en) 2015-10-21
EP2350129A4 (en) 2012-04-25
DK2350129T3 (en) 2015-08-31
ZA201101120B (en) 2012-02-29
HRP20150933T1 (en) 2015-10-09
CA2734908A1 (en) 2010-03-11
EP2350129A2 (en) 2011-08-03
HUE027525T2 (en) 2016-11-28
EP2927240A1 (en) 2015-10-07
US20130230514A1 (en) 2013-09-05
RS54233B1 (en) 2015-12-31
NZ591130A (en) 2012-09-28

Similar Documents

Publication Publication Date Title
AU2009288730B2 (en) Compositions of PD-1 antagonists and methods of use
AU2009288289B2 (en) PD-1 antagonists and methods of use thereof
EP1294391B1 (en) Combination of agents for inhibiting transplant rejection
US7601813B2 (en) Anti-GL50 antibodies
JP2012110333A (en) New polypeptide involved in immune response
AU2001275411A1 (en) Methods for regulating a cell-mediated immune response by blocking lymphocytic signals and by blocking LFA-1 mediated adhesion
RU2757394C2 (en) Compositions and methods for modulating the transmission of a lair signal
CA3089469A1 (en) B7-h4 antibodies and methods of use thereof
WO2004073732A1 (en) Modulators of notch signalling and of immune cell costimulatory activity for immunotherapy
AU2013227994A1 (en) Compositions of PD-1 antagonists and methods of use
EP1900816A2 (en) GL50 molecules and uses therefor
RU2812915C2 (en) Antibodies to programmed cell death protein 1
WO2017004167A1 (en) Methods and compositions for treating cancer

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980142256.3

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09811805

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 591130

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2009288730

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 211298

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 12011500378

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2734908

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2011020292

Country of ref document: EG

ENP Entry into the national phase

Ref document number: 2011524986

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 11023516

Country of ref document: CO

Ref document number: 000200-2011

Country of ref document: PE

Ref document number: MX/A/2011/002252

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2009288730

Country of ref document: AU

Date of ref document: 20090825

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2169/DELNP/2011

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 20117006754

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2009811805

Country of ref document: EP

Ref document number: 201170373

Country of ref document: EA

Ref document number: DZP2011000215

Country of ref document: DZ

WWE Wipo information: entry into national phase

Ref document number: CR2014-000015

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: P-2015/0547

Country of ref document: RS

ENP Entry into the national phase

Ref document number: PI0917320

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110223