WO2011071871A1 - Méthodes d'amplification d'un traitement à base d'anticorps anticancéreux - Google Patents

Méthodes d'amplification d'un traitement à base d'anticorps anticancéreux Download PDF

Info

Publication number
WO2011071871A1
WO2011071871A1 PCT/US2010/059221 US2010059221W WO2011071871A1 WO 2011071871 A1 WO2011071871 A1 WO 2011071871A1 US 2010059221 W US2010059221 W US 2010059221W WO 2011071871 A1 WO2011071871 A1 WO 2011071871A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
antibody
tumor
cell
day
Prior art date
Application number
PCT/US2010/059221
Other languages
English (en)
Inventor
Holbrook Kohrt
Roch Houot
Ronald Levy
Arash Ash Alizadeh
Matthew J. Goldstein
James Torchia
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
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
Priority to BR112012013736A priority Critical patent/BR112012013736A2/pt
Application filed by The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Priority to JP2012543196A priority patent/JP5950824B2/ja
Priority to US13/513,523 priority patent/US9005619B2/en
Priority to EP10836513.1A priority patent/EP2509627B1/fr
Priority to AU2010328347A priority patent/AU2010328347B2/en
Priority to KR1020127015865A priority patent/KR101853702B1/ko
Priority to SG2012041505A priority patent/SG181533A1/en
Priority to CA2781311A priority patent/CA2781311C/fr
Priority to CN201080063243XA priority patent/CN102753195A/zh
Priority to MX2012006420A priority patent/MX346912B/es
Publication of WO2011071871A1 publication Critical patent/WO2011071871A1/fr
Priority to US14/542,233 priority patent/US9758589B2/en
Priority to US15/669,418 priority patent/US10633450B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2893Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD52
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3015Breast
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3023Lung
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3046Stomach, Intestines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30 CD40 or CD95

Definitions

  • Monoclonal antibody technology is among the most notable scientific advances in the last quarter century. Rapid translation of this research has prolonged the survival of thousands of patients with cancer.
  • Monoclonal antibodies against HER2 (trastuzumab) and the EGF receptor (cetuximab) have similarly changed the natural history of select patients with breast cancer, and both colorectal and head and neck cancers, respectively.
  • NK cells natural killer cells
  • NK cells play a major role in the rejection of tumors and cells infected by viruses. The cells kill by releasing small cytoplasmic granules of proteins called perforin and granzyme that cause the target cell to die by apoptosis.
  • Natural killer cell activity is tightly regulated, and requires an activating signal.
  • activation of the Fc receptor by antibodies allows NK cells to lyse cells through antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular cytotoxicity
  • the NK cell releases granules containing granzymes and perforin.
  • Perforin forms pores in the cell membrane of the target cell, through which the granzymes and associated molecules can enter, inducing apoptosis.
  • ADCC is a primary mechanism by which tumor directed monoclonal antibody therapy works.
  • conventional cytotoxic chemotherapies induce myelosuppression, decreasing the population of NK cells, thereby reducing the efficacy of ADCC.
  • therapies which augment NK cell function might offer the ability to improve activity of monoclonal antibodies without increasing toxicity to non-cancer cells.
  • Clinically this is significant as an increasing population of cancer patients either due to older age, advanced disease, or prior therapies, are not candidates for conventional cytotoxic chemotherapy.
  • the present invention addresses this issue.
  • ADCC function is specifically augmented, which in turn enhances target cell killing, by sequential administration of an antibody directed against one or more tumor antigens, and an agonistic antibody against one or several inducible costimulatory molecules on NK cells.
  • An individual diagnosed with a tumor is first administered a tumor-directed antibody.
  • NK cells which are innate immune effector cells critical for ADCC upregulate expression of inducible costimulatory molecules such as CD137, OX40, GITR, CD30 or ICOS.
  • a second antibody is administered targeting the induced costimulatory molecule on NK cells (including but not limited to anti-CD137, anti-OX40, anti-GITR, anti-CD30 or anti-ICOS).
  • expression of the aforementioned costimulatory molecules is evaluated following administration of the tumor- directed antibody, in order to determine the optimal time for dosing the second agent. Alternatively a timing period is determined empirically, and generally applied.
  • the combination of agents and their sequential administration is shown to provide for a level of tumor-specific, therapeutic synergy that is not observed with administration of the single agents alone.
  • the method specifically enhances the anti-tumor function of monoclonal antibodies directed against tumor antigens. Because the second antibody targets costimulatory molecules that have been inducibly expressed on NK cells by the tumor- directed antibody, this methods allows specific stimulation of NK cells which are implicated in ADCC-mediated killing of the tumor cells, while sparing other NK cells, thereby limiting potential non specific side effects.
  • the inducible costimulatory molecule is
  • CD137 In such methods, an individual diagnosed with a tumor is first administered a tumor-selective antibody. After a period of time sufficient to upregulate expression of CD137 in immune system cells, a second agent is administered, which agent is an agonist of CD137. In some embodiments the level of CD137 expression on blood cells is determined prior to each administering step, where an increase in expression is indicative that the second agent may be administered.
  • FIG. 1 A-1 C Rituximab induces CD137 upregulation on human NK cells following incubation with CD20-positive tumor B cells.
  • Peripheral blood from three healthy donors was analyzed for CD137 expression on CD3 " CD56 + NK cells after 24 hour culture with lymphoma cell lines and trastuzumab or rituximab.
  • (A) shows the percentage of CD137 + cells among CD3 " CD56 + NK cells from three healthy donors cultured with CD20 " lymphoma cell line (OCI-Ly19) or CD20-positive lymphoma cell lines (Ramos, DHL-4, Raji) cell lines.
  • (B) shows CD20 surface expression on lymphoma cell lines (OCI-Ly19, Ramos, DHL-4 and Raji). Histograms were colored according to the log 10-fold increase in MFI of lymphoma cell lines relative to isotype.
  • (C) shows CD137 expression on NK cells subsets CD3 " CD56 bri9ht and CD3 " CD56 dim from a representative healthy donor after 24 hour culture with the CD20-positive lymphoma cell line, Ramos and rituximab.
  • FIGS. 2A-2F Anti-CD137 agonistic mAb increases rituximab-mediated NK cell cytotoxicity on tumor cells.
  • NK cell cytotoxicity on Raji, Ramos, and DHL-4 tumor cells was analyzed in chromium release assay (D-F).
  • Preactivated NK cells (as described in Material and Methods) were purified before being incubated with chromium labeled Raji, Ramos, and DHL-4 cells for 4 hours.
  • FIGS 3A-3D Anti-CD137 agonistic mAb enhances anti-lymphoma activity of murine anti-CD20 mAb in-vivo.
  • C57BL/6 mice were inoculated with 5x10 6 BL3750 lymphoma tumor cells, subcutaneously, on the abdomen.
  • A-B Post-tumor inoculation, mice then received either Rat IgG control on day 3( ⁇ ), anti-CD20 antibody on day 3(B), anti- CD137 antibody on day 4( ), or anti-CD20 antibody on day 3 and anti-CD137 antibody on day 4( A ).
  • C-D show tumor growth and survival with identical treatment sequence, however with treatment delayed until day 8 post tumor inoculation.
  • Mice received either Rat IgG control on day 8( ⁇ ), anti-CD20 antibody on day 8(B), anti-CD137 antibody on day 9( ), or anti-CD20 antibody on day 8 and anti-CD137 antibody on day 9( A ).
  • Mice (10 per group) were then monitored for tumor growth (C, *p ⁇ .001 ) and overall survival (D, */x.001 ).
  • FIGS. 4A-4B Anti-CD20 and anti-CD137 mAbs combination activity requires appropriate sequence of mAb administration.
  • FIGS 5A-5D Enhancement of the anti-lymphoma activity of anti-CD20 mAb by anti-CD137 agonistic mAb is dependent on NK cells and macrophages.
  • C-D C57BL/6 mice were inoculated with 5x10 6 BL3750 lymphoma tumor cells.
  • FIGS. 6A-6C Anti-CD137 agonistic mAb enhances anti-lymphoma activity of rituximab in vivo in a disseminated human lymphoma xenotransplant model.
  • FIGS 7A-7C Rituximab-coated, autologous lymphoma cells induce CD137 upregulation on NK cells from human patients with B cell malignancies.
  • Peripheral blood from patients with B cell malignancies and circulating tumor cells (CTC) were analyzed for CD137 expression on CD3 " CD56 + NK cells after 24 hour culture with media alone, trastuzumab, or rituximab (A and B).
  • A shows CD16 and CD137 expression on CD3 " CD56 + NK cells for a patient with marginal zone lymphoma (MZL) with 70% CTC.
  • (B) shows the percentage of CD137 + cells among CD3 " CD56 + NK cells in a cohort of 25 patients with follicular lymphoma (FL), chronic lymphocytic leukemia (CLL), MZL, mantle cell lymphoma (MCL), diffuse large B-cell lymphoma (DLBCL), and CD20-positive acute lymphoblastic leukemia (ALL).
  • FIGS 8A-8C CD137 induction and temporal expression on NK cells following preactivation.
  • Purified NK cells from healthy donors were analyzed for CD137 expression after 24 hour culture with media, rituximab, trastuzumab, lymphoma cell lines (Raji, Ramos, DHL-4, or OCI-Ly19) and rituximab (A).
  • Purified NK cells from a healthy donor were analyzed for CD137 expression after 0, 4, 16, 24, 48 and 72 hour culture with Raji cell line and rituximab (B).
  • peripheral blood mononuclear cells from a representative healthy donor were incubated with Raji, Ramos or DHL-4 and rituximab for 24 hours. Preactivated NK cells were then analyzed for CD137 expression (C) prior to performing the cytotoxicity assay.
  • FIGS 9A-9B Anti-CD137 agonistic mAb increases cytokine release and rituximab- mediated cytotoxicity of pre-activated NK cells.
  • NK cell interferon- ⁇ secretion purified NK cells were isolated from healthy PBMCs and cultured for 24 hours together with rituximab (10 g/mL) and irradiated (5,000 rads) lymphoma tumor cells (Raji) at a ratio of 1 :1 . After 24 hours, NK cells were isolated and assessed for purity (>90% purity as defined by CD3-CD56+ flow cytometry)(A-B).
  • NK cell cytotoxicity on Raji tumor cells was analyzed in chromium release assay with and without prior NK cell preactivation (B).
  • Preactivated, and non-preactivated, purified NK cells were incubated with chromium-labeled Raji for 4 hours.
  • Percent lysis of target cells by chromium release at varying effector preactivated NK cells depicted in continuous line, and non-preactivated NK cells depicted in dashed line
  • FIG. 10 Anti-CD137 agonistic mAb increases rituximab-mediated NK cell degranulation.
  • NK cells isolated and purified from the peripheral blood of healthy donors were analyzed for degranulation by CD107a mobilization after 24 hour culture with media alone, CD20-positive lymphoma cell line (Raji, Ramos, or DHL-4), tumor and rituximab, tumor and anti-CD137 antibody, or tumor, rituximab, and anti-CD137 agonistic antibody.
  • trastuzumab induces CD137 upregulation on human NK cells following incubation with HER2-positive tumor cells.
  • Peripheral blood from three healthy donors was analyzed for CD137 expression on CD3 " CD56 + NK cells after 24 hour culture with breast cancer cell lines and IgG control, trastuzumab or rituximab.
  • A shows the expression of CD69 + and CD137 + on CD3 " CD56 + NK cells from a representative healthy donor after 24 hour culture with HER2 + breast cancer cell line (BT474M1 ).
  • B shows HER2 surface expression on breast cancer cell lines (MCF7, BT474M1 , SKBR3, and HER18).
  • C shows CD137 expression from three healthy donors cultured on NK cells CD3 " CD56 + after 24 hour culture with variably expressing HER2 breast cancer cell lines (MCF7, BT474M1 , SKBR3, HER18).
  • FIGS 12A-12C Anti-CD137 agonistic mAb increases trastuzumab-mediated NK cell cytotoxicity on tumor cells as assayed by cell viability. Preactivated NK cells were purified before being incubated with MCF7, BT474M1 , and HER18 for 18 hours.
  • Anti-CD137 agonistic mAb increases trastuzumab-mediated NK cell cytotoxicity on tumor cells as assayed by chromium release.
  • NK cell cytotoxicity on BT474M1 tumor cells was analyzed in chromium release assay. Preactivated NK cells were purified before being incubated with chromium labeled BT474M1 cells for 4 hours.
  • Shown is percent lysis of target cells by chromium release at varying effector (activated NK cells) :target (BT474M1 ) cell ratios cultured with media alone(»), anti-CD137( T), rituximab(A ), or rituximab and anti-CD137(») antibodies (p .006).
  • FIGS 14A-14B Anti-CD137 agonistic mAb enhances anti-breast cancer activity of trastuzumab in-vivo.
  • Nu/nu nude mice were inoculated with 5x10 6 BT474M1 breast tumor cells, subcutaneously, on the abdomen 1 day after subcutaneous injection of 0.72 mg/60 day release beta-estradiol pellet.
  • FIG. 15A-15C Anti-CD137 agonistic mAb enhances anti-breast cancer activity of trastuzumab in-vivo while retaining HER2 specificity.
  • Nu/nu nude mice were inoculated with 5x10 6 MCF7 breast tumor cells, subcutaneously, on the left flank, and 5x10 6 HER18 breast tumor cells, subcutaneously, on the right flank 1 day after subcutaneous injection of 0.72mg/60 day release beta-estradiol pellet.
  • A-C Post-tumor inoculation, mice then received either trastuzumab on day 3, or trastuzumab on day 3 and anti-CD137 antibody on day 4 with each treatment repeated weekly for a total of three weeks.
  • A) Tumor model is a tumor model.
  • mice (3 of 10 per group) were then monitored for tumor growth.
  • FIG. 16 Anti-CD137 agonistic mAb enhances anti-breast cancer activity of trastuzumab in-vivo against HER2 + primary breast tumor.
  • SCID mice were inoculated with 1 x10 s HER2 + primary breast tumor cells by intramammary injection 24 hours after 200 cGy total body irradiation (TBI).
  • TBI total body irradiation
  • mice were randomized to one of four groups (5 mice per group) including IgG control with treatment on day 40 ( ⁇ ), trastuzumab on day 40 ( ⁇ ), anti-CD137 mAb on day 41 ( ⁇ ), or trastuzumab on day 40 and anti-CD137 mAb on day 41 ( A ).
  • FIGs 17A-17B Cetuximab induces CD137 upregulation on human NK cells following incubation with EGFR-positive tumor cells.
  • Peripheral blood from three healthy donors was analyzed for CD137 expression on CD3 " CD56 + NK cells after 24 hour culture with head and neck cancer cell lines and media alone, rituximab or cetuximab.
  • A shows EGFR surface expression on head and neck cancer cell lines (103, SCC4, PC1 , SCC6). Histograms were colored according to the log 10-fold increase in MFI of breast cancer cell lines relative to isotype.
  • B shows CD137 expression from three healthy donors cultured on NK cells CD3 " CD56 + after 24 hour culture with variably expressing EGFR head and neck cancer cell lines (SCC6, PC1 , and SCC4).
  • FIGS. 18A-18C Anti-CD137 agonistic mAb increases cetuximab-mediated NK cell cytotoxicity on tumor cells as assayed by cell viability. Preactivated NK cells were purified before being incubated with SCC6, PC1 , and SCC4 for 24 hours.
  • FIG. 19 Anti-CD137 agonistic mAb increases cetuximab-mediated NK cell cytotoxicity on tumor cells as assayed by chromium release. NK cell cytotoxicity on SCC6 tumor cells was analyzed in chromium release assay. Fresh and preactivated NK cells were purified before being incubated with chromium labeled SCC6 cells for 5 hours.
  • FIG. 20 Anti-CD137 agonistic mAb enhances anti-head and neck cancer activity of cetuximab in-vivo.
  • Nu/nu nude mice were inoculated with 3x10 6 SCC6 head and neck tumor cells, subcutaneously, on the abdomen. Post-tumor inoculation, mice then received either rat IgG control on day 21 ( ⁇ ), cetuximab on day 21 (B), anti-CD137 antibody on day 22( ), or cetuximab on day 21 and anti-CD137 antibody on day 22(A ) with each treatment repeated weekly for a total of three treatments. Mice (10 per group) were then monitored for tumor growth (A, * /x.001 ).
  • Figures 21 A-21 B Circulating NK cells upregulate CD137 following cetuximab infusion in patients with head and neck cancer.
  • Fresh peripheral blood from patient with head and neck cancer was analyzed for CD137 expression on CD3 " CD56 + NK cells.
  • (A) shows the phase 0, biomarker, trial schema (NCT01 1 14256).
  • (B) shows the percentage of CD137+ cells among fresh, peripheral blood CD3- CD56+ NK cells prior to and following cetuximab infusion.
  • ADCC function is augmented and target cell killing is enhanced by sequential administration of a combination of antibodies.
  • the combination of agents and sequential administration is shown to provide for synergistic effects, relative to the administration of the single agents.
  • Administration of a tumor-directed antibody up-regulates the expression of inducible costimulatory molecules such as CD137, OX40, GITR, CD30 or ICOS on NK cells which are innate immune effector cells critical for ADCC.
  • a second agonistic antibody is administered to target the induced costimulatory molecules (including but not limited to anti-CD137, -OX40, -GITR, -CD30 or -ICOS).
  • induced costimulatory molecules including but not limited to anti-CD137, -OX40, -GITR, -CD30 or -ICOS.
  • expression of the aforementioned costimulatory molecules following administration of the tumor-directed antibody is evaluated to determine the optimal time for dosing the second agent. Alternatively a timing period is determined empirically, and generally applied. Because the second antibody targets costimulatory molecules which have been inducibly expressed on NK cells by the tumor-directed antibody, this methods allows specific stimulation of NK cells that are implicated in ADCC- mediated killing of the tumor cells, while sparing other NK cells, thereby limiting potential non specific side effects.
  • an inducible costimulatory molecule is a polypeptide expressed on immune cells, including without limitation natural killer (NK) cells, which expression is induced or significantly upregulated during activation of NK cells. Activation of the costimulatory molecule enhanced the effector cell function, for example increasing ADCC mediated by the activated NK cells.
  • NK natural killer
  • Such inducible costimulatory molecules are known to those of skill in the art, and include, without limitation, CD137, OX40, GITR, CD30, ICOS, etc. Agonists of such molecules, including antibodies that bind to and activate the costimulatory molecule, are of interest for the methods of the invention. Many such costimulatory molecules are members of the tumor necrosis factor receptor family (TNFR). TNFR-related molecules do not have any known enzymatic activity and depend on the recruitment of cytoplasmic proteins for the activation of downstream signaling pathways.
  • TNFR tumor necrosis factor receptor family
  • CD137 which may also be referred to as Ly63, ILA or 4-1 BB is a member of the tumor necrosis factor (TNF) receptor family. Members of this receptor family and their structurally related ligands are important regulators of a wide variety of physiologic processes and play an important role in the regulation of immune responses.
  • CD137 is expressed by activated NK cells, T and B lymphocytes and monocytes/macrophages. The gene encodes a 255-amino acid protein with 3 cysteine-rich motifs in the extracellular domain (characteristic of this receptor family), a transmembrane region, and a short N- terminal cytoplasmic portion containing potential phosphorylation sites. Expression in primary cells is strictly activation dependent.
  • the ligand for the receptor is TNFSF9.
  • Human CD137 is reported to bind only to its ligand.
  • Agonists include the native ligand (TNFSF9), aptamers (see McNamara et al. (2008) J. Clin. Invest. 1 18: 376-386), and antibodies.
  • OX40 CD134
  • OX40L CD252
  • OX40 and OX40L regulate cytokine production from T cells, antigen-presenting cells, natural killer cells, and natural killer T cells, and modulate cytokine receptor signaling.
  • GITR Glucocorticoid-lnduced TNFR-Related (GITR) protein belongs to tumor necrosis factor receptor/tumor necrosis factor superfamily and stimulates both the acquired and innate immunity. It is expressed in several cells and tissues, including T and Natural Killer (NK) cells and is activated by its ligand, GITRL, mainly expressed on antigen presenting cells and endothelial cells. GITR/GITRL system participates in the development of autoimmune/inflammatory responses and potentiates response to infection and tumors by mechanisms including NK-cell co-activation.
  • NK Natural Killer
  • CD30 The transmembrane receptor CD30 (TNFRSF8) and its ligand CD30L
  • CD153, TNFSF8 are members of the tumor necrosis factor (TNF) superfamily and display restricted expression in subpopulations of activated immune cells.
  • CD30 is a type I transmembrane glycoprotein of the TNF receptor superfamily.
  • the ligand for CD30 is CD30L (CD153). The binding of CD30 to CD30L mediates pleiotropic effects including cell proliferation, activation, differentiation, and apoptotic cell death.
  • ICOS Inducible costimulator
  • ICOS is a member of the CD28 family. ICOS expression, may be readily detectable resting, but it upregulated upon activation. ICOS and ICOS-L appear to be a monogamous pair. ICOS costimulation enhances effector functions.
  • inducible costimulatory molecule agonist includes the native ligands, as described above, aptamers, antibodies specific for an inducible costimulatory molecule that activate the receptor, and derivatives, variants, and biologically active fragments of antibodies that selectively bind to an inducible costimulatory molecule.
  • a "variant” polypeptide means a biologically active polypeptide as defined below having less than 100% sequence identity with a native sequence polypeptide.
  • Such variants include polypeptides wherein one or more amino acid residues are added at the N- or C-terminus of, or within, the native sequence; from about one to forty amino acid residues are deleted, and optionally substituted by one or more amino acid residues; and derivatives of the above polypeptides, wherein an amino acid residue has been covalently modified so that the resulting product has a non-naturally occurring amino acid.
  • a biologically active variant will have an amino acid sequence having at least about 90% amino acid sequence identity with a native sequence polypeptide, preferably at least about 95%, more preferably at least about 99%.
  • the variant polypeptides can be naturally or non-naturally glycosylated, i.e., the polypeptide has a glycosylation pattern that differs from the glycosylation pattern found in the corresponding naturally occurring protein.
  • Fragments of the ligand or antibodies specific for an inducible costimulatory molecule are of interest. Fragments of interest will typically be at least about 10 aa to at least about 15 aa in length, usually at least about 50 aa in length, but will usually not exceed about 200 aa in length, where the fragment will have a contiguous stretch of amino acids that is identical to the polypeptide from which it is derived.
  • a fragment "at least 20 aa in length,” for example, is intended to include 20 or more contiguous amino acids from, for example, an antibody specific for CD137, or from TNFSF9.
  • the protein variants described herein are encoded by polynucleotides that are within the scope of the invention.
  • the genetic code can be used to select the appropriate codons to construct the corresponding variants.
  • the polynucleotides may be used to produce polypeptides, and these polypeptides may be used to produce antibodies by known methods.
  • a "fusion" polypeptide is a polypeptide comprising a polypeptide or portion (e.g., one or more domains) thereof fused or bonded to heterologous polypeptide.
  • the inducible costimulatory molecule agonist is an antibody.
  • antibody or “antibody moiety” is intended to include any polypeptide chain- containing molecular structure with a specific shape that fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope.
  • Antibodies utilized in the present invention may be polyclonal antibodies, although monoclonal antibodies are preferred because they may be reproduced by cell culture or recombinantly, and can be modified to reduce their antigenicity.
  • Polyclonal antibodies can be raised by a standard protocol by injecting a production animal with an antigenic composition. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988.
  • antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Freund's, Freund's complete, oil-in- water emulsions, etc.)
  • a suitable adjuvant e.g., Freund's, Freund's complete, oil-in- water emulsions, etc.
  • conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH).
  • BSA bovine serum albumin
  • KLH keyhole limpet hemocyanin
  • peptides derived from the full sequence may be utilized.
  • a superior immune response may be elicited if the polypeptide is joined to a carrier protein, such as ovalbumin, BSA or KLH.
  • hybridomas may be formed by isolating the stimulated immune cells, such as those from the spleen of the inoculated animal.
  • immortalized cells such as myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line.
  • immortalized cells such as myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line.
  • the antibodies or antigen binding fragments may be produced by genetic engineering. Humanized, chimeric, or xenogeneic human antibodies, which produce less of an immune response when administered to humans, are preferred for use in the present invention.
  • immunoglobulin fragments comprising the epitope binding site (e.g., Fab', F(ab') 2 , or other fragments) are useful as antibody moieties in the present invention.
  • Such antibody fragments may be generated from whole immunoglobulins by ricin, pepsin, papain, or other protease cleavage. "Fragment,” or minimal immunoglobulins may be designed utilizing recombinant immunoglobulin techniques.
  • Fv immunoglobulins for use in the present invention may be produced by linking a variable light chain region to a variable heavy chain region via a peptide linker (e.g., poly-glycine or another sequence which does not form an alpha helix or beta sheet motif).
  • a peptide linker e.g., poly-glycine or another sequence which does not form an alpha helix or beta sheet motif.
  • enhancing efficacy is meant an increase in ADCC-mediated apoptosis of tumor cells compared to level of apoptosis observed with a single agent, e.g. a monoclonal antibody specific for a tumor cell.
  • a single agent e.g. a monoclonal antibody specific for a tumor cell.
  • synergistic it is meant that a combination of agents provides for an effect greater than a single agent, which effect may be greater than the additive effect of the combined agents.
  • Tumor directed antibodies A number of antibodies are currently in clinical use for the treatment of cancer, and others are in varying stages of clinical development. Antibodies of interest for the methods of the invention act through ADCC, and are typically selective for tumor cells, although one of skill in the art will recognize that some clinically useful antibodies do act on non-tumor cells, e.g. CD20.
  • CD20 which is found on B cell malignancies.
  • Rituximab is a chimeric unconjugated monoclonal antibody directed at the CD20 antigen.
  • CD20 has an important functional role in B cell activation, proliferation, and differentiation.
  • the CD52 antigen is targeted by the monoclonal antibody alemtuzumab, which is indicated for treatment of chronic lymphocytic leukemia.
  • CD22 is targeted by a number of antibodies, and has recently demonstrated efficacy combined with toxin in chemotherapy-resistant hairy cell leukemia.
  • FDA Food and Drug Administration
  • Monoclonal antibodies useful in the methods of the invention include without limitation edrecolomab and trastuzumab (herceptin).
  • Edrecolomab targets the 17-1 A antigen seen in colon and rectal cancer, and has been approved for use in Europe for these indications. Its antitumor effects are mediated through ADCC, CDC, and the induction of an anti-idiotypic network.
  • Trastuzumab targets the HER- 2/neu antigen. This antigen is seen on 25% to 35% of breast cancers.
  • Trastuzumab is thought to work in a variety of ways: downregulation of HER-2 receptor expression, inhibition of proliferation of human tumor cells that overexpress HER-2 protein, enhancing immune recruitment and ADCC against tumor cells that overexpress HER-2 protein, and downregulation of angiogenesis factors.
  • Alemtuzumab (Campath) is used in the treatment of chronic lymphocytic leukemia; colon cancer and lung cancer; Gemtuzumab (Mylotarg) finds use in the treatment of acute myelogenous leukemia; Ibritumomab (Zevalin) finds use in the treatment of non-Hodgkin's lymphoma; Panitumumab (Vectibix) finds use in the treatment of colon cancer.
  • Cetuximab (Erbitux) is also of interest for use in the methods of the invention.
  • the antibody binds to the EGF receptor (EGFR), and has been used in the treatment of solid tumors including colon cancer and squamous cell carcinoma of the head and neck (SCCHN).
  • EGFR EGF receptor
  • SCCHN squamous cell carcinoma of the head and neck
  • biological sample encompasses a variety of sample types obtained from an organism and can be used in a diagnostic or monitoring assay.
  • the term encompasses blood and other liquid samples of biological origin, solid tissue samples, such as a biopsy specimen or tissue cultures or cells derived therefrom and the progeny thereof.
  • the term encompasses samples that have been manipulated in any way after their procurement, such as by treatment with reagents, solubilization, or enrichment for certain components.
  • the term encompasses a clinical sample, and also includes cells in cell culture, cell supernatants, cell lysates, serum, plasma, biological fluids, and tissue samples.
  • cancer neoplasm
  • tumor tumor
  • tumor tumor
  • tumor tumor-associated phenotype
  • cancer tumor-associated phenotype
  • cancer tumor-associated phenotype
  • tumor tumor-associated phenotype
  • tumor tumor-associated phenotype
  • tumor tumor-associated phenotype
  • tumor tumor-associated phenotype
  • tumor tumor-associated phenotype
  • tumor tumor-associated phenotype
  • tumor tumor-associated phenotype
  • cancerous cells e.g., tumor cells
  • non-metastatic e.g., tumor cells, and non-metastatic cells. Detection of cancerous cells is of particular interest.
  • normal as used in the context of "normal cell,” is meant to refer to a cell of an untransformed phenotype or exhibiting a morphology of a non-transformed cell of the tissue type being examined.
  • Cancerous phenotype generally refers to any of a variety of biological phenomena that are characteristic of a cancerous cell, which phenomena can vary with the type of cancer.
  • the cancerous phenotype is generally identified by abnormalities in, for example, cell growth or proliferation (e.g., uncontrolled growth or proliferation), regulation of the cell cycle, cell mobility, cell-cell interaction, or metastasis, etc.
  • Cancers of interest include, without limitation, hematopoietic cancers including leukemias, lymphomas (Hodgkins and non-Hodgkins), myelomas and myeloproliferative disorders; sarcomas, melanomas, adenomas, carcinomas of solid tissue, squamous cell carcinomas of the mouth, throat, larynx, and lung, liver cancer, genitourinary cancers such as cervical, bladder cancer and renal cell carcinomas, head and neck cancers, gastro intestinal track cancers and nervous system cancers, benign lesions such as papillomas, and the like.
  • solid tumor refers to an abnormal mass of tissue that usually does not contain cysts or liquid areas. Solid tumors may be benign or malignant. Different types of solid tumors are named for the type of cells that form them. Examples of solid tumors are sarcomas, carcinomas, lymphomas etc.
  • Monoclonal antibodies directed against a specific cancer epitope, or combination of epitopes allows the targeting and/or depletion of cancer cell populations expressing the marker.
  • Various techniques can be utilized using monoclonal antibodies to screen for cellular populations expressing the marker(s), and include magnetic separation using antibody-coated magnetic beads, "panning" with antibody attached to a solid matrix (i.e., plate), and flow cytometry (See, e.g., U.S. Pat. No. 5,985,660; and Morrison et al. Cell, 96:737-49 (1999)). These techniques allow for the screening of particular populations of cells; in immunohistochemistry of biopsy samples; in detecting the presence of markers shed by cancer cells into the blood and other biologic fluids, and the like.
  • treatment used herein to generally refer to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete stabilization or cure for a disease and/or adverse effect attributable to the disease.
  • Treatment covers any treatment of a disease in a mammal, particularly a human, and includes: (a) preventing the disease or symptom from occurring in a subject which may be predisposed to the disease or symptom but has not yet been diagnosed as having it; (b) inhibiting the disease symptom, i.e., arresting its development; or (c) relieving the disease symptom, i.e., causing regression of the disease or symptom.
  • Therapeutic target refers to molecules expressed by the tumor cells and/or non tumor (immune) cells that can be targeted to induce or enhance antitumor activity.
  • Methods are provided to enhance the efficacy of cell killing induced by the administration of antibodies directed against tumor cells.
  • An effective dose of a primary tumor-directed antibody is administered to a patient, which induces the upregulation of inducible costimulatory molecules such as CD137, OX40, GITR, CD30 or ICOS on NK cells, which are innate immune effector cells critical for ADCC.
  • an effective dose of a second agonistic antibody against one of these molecules including but not limited to CD137, OX40, GITR, CD30 or ICOS
  • the second antibody is sufficient to enhance ADCC killing of tumor cells targeted by the first antibody.
  • the second antibody targets costimulatory molecules that have been inducibly expressed on NK cells by the tumor-directed antibody, this methods allows specific stimulation of NK cells that are implicated in ADCC-mediated killing of the tumor cells, while sparing other NK cells, thereby limiting potential non-specific side effects.
  • the level of costimulatory molecules (including but not limited to CD137, OX40, GITR, CD30 or ICO) induced by the tumor-directed antibody is determined in a patient sample, usually a patient blood sample or cellular fraction thereof.
  • the level of costimulatory molecules may be determined in a sample prior to administering the tumor-directed antibody, and the increase in expression following administration of the tumor-directed antibody determined.
  • an effective dose of a tumor-selective antibody is administered to a patient, following which sufficient time is elapsed for an upregulation of CD137 expression on immune system cells, particularly NK cells.
  • the sufficient time is usually at least about 12 hours, more usually at least about 18 hours, and usually at least about 24 hours, and may be at least about 2 days, at least about 3 days, and not more than about 5 days, usually not more than about 4 days.
  • an effective dose of a CD137 agonist is administered to said individual, where the agonist is sufficient to enhance ADCC killing of tumor cells targeted by the first antibody.
  • the level of CD137 is determined in a patient sample, usually a patient blood sample or cellular fraction thereof.
  • the level of CD137 may be determined in a sample prior to administering the tumor-selective antibody, and the increase in expression following administration of the tumor-selective antibody determined.
  • a desirable increase in expression of CD137 on NK cells is at least about 1 .5-fold, at least about 2-fold, or higher.
  • An increase in expression when measured in overall blood cells may be lower due to the number of contaminating non-responsive cells.
  • Reducing growth of cancer cells includes, but is not limited to, reducing proliferation of cancer cells, and increasing apoptosis of tumor cells. Whether a reduction in cancer cell growth has been achieved can be readily determined using any known assay, including, but not limited to, [ 3 H]-thymidine incorporation; counting cell number over a period of time; detecting and/or measuring a marker associated with the cancer of interest, etc.
  • Whether a substance, or a specific amount of the substance, is effective in treating cancer can be assessed using any of a variety of known diagnostic assays for cancer, including, but not limited to biopsy, contrast radiographic studies, CAT scan, and detection of a tumor marker associated with cancer in the blood of the individual.
  • the substance can be administered systemically or locally, usually systemically.
  • Therapeutic formulations comprising one or more antibodies utilized in the methods of the invention may be prepared for storage by mixing the antibody having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • the antibody composition will be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular cancer being treated, the clinical condition of the individual patient, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the "therapeutically effective amount" of the antibody to be administered will be governed by such considerations.
  • the therapeutic dose may be at least about 0.01 ⁇ g kg body weight, at least about
  • 0.05 ⁇ g kg body weight at least about 0.1 ⁇ g kg body weight, at least about 0.5 ⁇ g kg body weight, at least about 1 ⁇ g kg body weight, at least about 2.5 ⁇ g kg body weight, at least about 5 ⁇ g kg body weight, and not more than about 100 ⁇ g kg body weight. It will be understood by one of skill in the art that such guidelines will be adjusted for the molecular weight of the active agent, e.g. in the use of antibody fragments, or in the use of antibody conjugates. The dosage may also be varied for route of administration.
  • Acceptable carriers, excipients, or stabilizers are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyidimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, hist
  • an article of manufacture containing materials useful for the treatment of the disorders described above comprises a container and a label.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the active agent in the composition is one or more antibodies as described above.
  • the label on, or associated with, the container indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • a pharmaceutically-acceptable buffer such as phosphate-buffered saline, Ringer's solution and dextrose solution.
  • It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • kits and agonists suitable for use in the methods of the invention may be provided in a kit form, for example presented in a pack or dispenser device containing one or more unit dosage forms containing the active ingredients.
  • a pack or device may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • Compositions comprising agents useful in the methods of the invention may be formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Suitable conditions indicated on the label may include treatment of cancer.
  • Kits may also comprise a unit suitable for measuring expression of an inducible costimulatory molecule on NK cells, e.g. including detectable labeled reagent that specifically binds to an inducible costimulatory molecule, and references for expression, and the like as known in the art.
  • trastuzumab a humanized lgG1 antibody against HER2 (human epidermal growth factor receptor 2)
  • HER2 + breast cancer a humanized lgG1 antibody against HER2 + breast cancer.
  • cetuximab a chimeric lgG1 antibody targeting the EGFR (epidermal growth factor receptor)
  • cetuximab was also approved for treatment of head and neck cancers.
  • cancer therapies are now focused on discovery of both novel and known targets to which new or current antibodies can be aimed.
  • ADCC antibody dependent cell- mediated cytotoxicity
  • NK natural killer cells or macrophage/monocytes bearing an Fc receptor which binds to the antibody- targeted tumor cell.
  • NK cell Fc receptor binding to antibody activates the NK cell resulting in release of cytokines and cytotoxic granules which trigger apoptosis in the antibody-targeted tumor cell.
  • Increased NK cell function augments ADCC and results in improved antitumor activity.
  • the present invention identifies costimulatory molecules that are inducibly expressed on NK cells, which are innate effector cells critical for ADCC following recognition of antibody-coated tumor cells. Following up-regulation by the tumor directed antibody, these costimulatory molecules (including but not limited to CD137, OX40, GITR, CD30 or ICOS) can subsequently be targeted with agonistic antibodies to enhance ADCC mediated by these effector cells. Given the increasing number of tumor target-specific antibodies, this has therapeutic implications for any cancer type directly targeted by a monoclonal antibody.
  • agonistic antibodies that enhance NK cell function is limited only by the number of targets expressed by tumor cells and target-specific antibodies. Given the impact of rituximab, trastuzumab and cetuximab over the prior decade, target and antibody discovery efforts are likely to continue to be an area of active research. Though application of agonistic NK cell antibodies will extend beyond non-Hodgkin lymphoma, breast cancer, colorectal and head and neck cancers, these four demonstrate the magnitude of clinical benefit.
  • Antibody coated tumor cells trigger the activation of NK cells, whose killing activity can then be stimulated by a second activating antibody against CD137.
  • the addition of agonistic anti-CD137 antibody is a general approach to enhance the therapeutic effect of any anti-tumor antibody.
  • CD137 Increased NK cell expression of CD137 occurs following NK cell exposure to CD20 + lymphoma coated with rituximab.
  • Targeting CD137 with an agonistic antibody to enhance antitumor ADCC is dependent upon its increased surface expression on NK cells following their exposure to antibody coated tumor cells.
  • the second antibody targets a costimulatory molecule (CD137) which is inducibly expressed on NK cells by the tumor- directed antibody (rituximab, trastuzumab)
  • this methods allows specific stimulation of NK cells which are implicated in ADCC-mediated killing of the tumor cells (lymphoma, breast cancer), while sparing other NK cells, thereby limiting potential non specific side effects.
  • PBMCs peripheral blood mononuclear cells
  • CLL chronic lymphocytic leukemia
  • MZL marginal zone lymphoma
  • ALL CD20 + acute lymphoblastic leukemia
  • PBMCs were analyzed by flow cytometry after culture with trastuzumab or rituximab.
  • Percent CD137 + NK cells increased from 1 -2% at baseline to 22-43% with concurrent downregulation of CD16 (the Fc receptor) following culture with rituximab. This appeared antibody dependent since no activation occurred following culture with trastuzumab.
  • Rituximab induces CD137 upregulation on human NK cells following incubation with CD20- positive tumor B cells.
  • Peripheral blood from three healthy donors was analyzed for CD137 expression on CD3 " CD56 + NK cells after 24 hour culture with lymphoma cell lines and trastuzumab or rituximab.
  • Anti-CD137 agonistic mAb increases rituximab-mediated NK cell cytotoxicity on tumor cells, as shown in Figures 2A-2F.
  • NK cells isolated and purified from the peripheral blood of healthy donors were analyzed for degranulation by CD107a mobilization after 24 hour culture with five conditions: media alone; CD20-positive lymphoma cell line (Raji, Ramos, or DHL-4); tumor and rituximab; tumor and anti-CD137 antibody; or tumor, rituximab, and anti-CD137 agonistic antibody.
  • media alone CD20-positive lymphoma cell line (Raji, Ramos, or DHL-4)
  • tumor and rituximab tumor and anti-CD137 antibody
  • tumor, rituximab, and anti-CD137 agonistic antibody Shown in Figures 3A-3D, there is also an enhancement of anti-lymphoma activity with anti-CD137 agonistic mAb.
  • FIG. 5A-5D peripheral blood cell subsets from lymphoma- bearing C57BL/6 mice 4 days post-tumor inoculation treated on day 3 with either IgG control or anti-CD20 antibody were analyzed for CD137 expression on CD3-NK1 .1 + NK cells (NK), F4/80 + macrophages ( ⁇ ), CD3 + CD8 + T cells (CD8), and CD3 + CD4 + T cells (CD4); Tumor-infiltrating lymphocytes from lymphoma-bearing C57BL/6 mice 7 days post- tumor inoculation treated on day 3 with either IgG control or anti-CD20 antibody were analyzed for CD137 expression on CD3-NK1 .1 + NK cells (NK), F4/80 + macrophages ( ⁇ ), CD3 + CD8
  • mice are inoculated on day 0 with 3x10 6 Raji cells transfected with Firefly Luciferase, and treated with rituximab (10 ⁇ g/g weight, IP) on day 3 followed by 150 ⁇ g of rat anti-mouse anti-CD137Ab IP on day 4.
  • rituximab 10 ⁇ g/g weight, IP
  • Blood is collected on day 3 prior to rituximab treatment, on day 4 prior to anti-CD137Ab treatment, and on day 5 to assay NK cell expression of CD137, CD69, and CD16.
  • Bioluminescent imaging is performed following IP luciferin injection (200 ⁇ _) on day 3 prior to Ab therapy and repeated weekly.
  • the effect of CD137 with rituximab is shown in a disseminated human lymphoma xenotransplant model, shown in Figures 6A-6C.
  • Rituximab-coated, autologous lymphoma cells induce CD137 upregulation on NK cells from human patients with B cell malignancies, shown in Figures 7A-7C.
  • Peripheral blood from patients with B cell malignancies and circulating tumor cells (CTC) were analyzed for CD137 expression on CD3 " CD56 + NK cells after 24 hour culture with media alone, trastuzumab, or rituximab.
  • the kinetics of CD137 induction and temporal expression on NK cells following preactivation are analyzed in Figures 8A-8C.
  • CD137Ab monotherapy CD137Ab monotherapy. However, all mice treated with anti-CD20Ab died before 60 days, and only 50% of mice treated with anti-CD137Ab were alive 100 days post-tumor inoculation. Treatment with anti-CD20Ab on day 3 followed by anti-CD137Ab on day 4 resulted in complete regression of tumor and survival at 100 days in 90% of mice.
  • NK cells were depleted with anti-asialo-GM1 on day -1 , 0, and every 5 days thereafter till 20 days at which point a clear separation of treatment groups was observed. NK cell depletion with anti-asialo-GM1 abrogated the benefit of combination therapy.
  • Anti-CD137 agonistic mAb increases cytokine release and rituximab-mediated cytotoxicity of pre-activated NK cells, shown in Figures 9A-9B.
  • NK cell interferon- ⁇ secretion purified NK cells were isolated from healthy PBMCs and cultured for 24 hours together with rituximab (10 g/mL) and irradiated (5,000 rads) lymphoma tumor cells (Raji) at a ratio of 1 :1 . After 24 hours, NK cells were isolated and assessed for purity.
  • NK cells Preactivated, purified NK cells were then cultured for 4 hours in media alone, or with anti- CD137 mAb alone, rituximab alone, or rituximab plus anti-CD137 mAbs and supernatant was harvested and analyzed by ELISA for interferon- ⁇ .
  • NK cell cytotoxicity on Raji tumor cells was analyzed in chromium release assay with and without prior NK cell preactivation.
  • Anti-CD137 agonistic mAb also increases rituximab-mediated NK cell degranulation, shown in Figure 10.
  • NK cell expression of CD137 occurs following NK cell exposure to HER2 + breast cancer coated with trastuzumab.
  • MCF7 a non- HER2 expressing breast cancer cell line
  • SKBR3 a HER2 overexpressing breast cancer cell line
  • NK cell expression of CD137 was determined following co-culture with appropriate tumor cell lines coated with cetuximab (10 ⁇ )/ ⁇ -), rituximab (10 ⁇ g mL) or trastuzumab (10 ⁇ g/mL) as detailed above for breast cancer cell lines.
  • Flow cytometry for CD3 and CD56 was performed to evaluate purity of the NK cell isolation. Additional markers of activation including CD69, CD107, and CD16 were included in the flow cytometry panel.
  • the colon cancer cell lines include HCT-8 (EGFR + ) and SW620 (EGFR), and the squamous head and neck cancer cell lines include TE3 (EGFR + HER2 ) and TE4 (EGFR " HER2 + ).
  • anti-CD137 agonistic mAb increases trastuzumab- mediated NK cell cytotoxicity on tumor cells as assayed by cell viability.
  • Preactivated NK cells were purified before being incubated with MCF7, BT474M1 , and HER18 for 18 hours, and apoptosis evaluated
  • NK cells isolated from normal blood were activated by co- culture with SKBR3 (HER2 + ) and trastuzumab. These exposed NK cells were tested by flow cytometry for CD137 and CD69 expression to evaluate their activation.
  • Activated NK cells are added to chromium labeled SKBR3 target cells at ratios of 12.5:1 , 25:1 , 50:1 , and 100:1 together with trastuzumab (10 ⁇ g mL), anti-CD137Ab (10 ⁇ g mL), or trastuzumab + anti- CD137Ab (both at 10 ⁇ g mL).
  • trastuzumab 10 ⁇ g mL
  • anti-CD137Ab 10 ⁇ g mL
  • trastuzumab + anti- CD137Ab both at 10 ⁇ g mL.
  • a similar activation and killing assay is performed to investigate in-vitro functional activity against EGFR + colon cancer with HCT-8 (EGFR + ) cell line and cetuximab, and EGFR + head and neck cancers with TE3 (EGFR + HER2 ) cell line and cetuximab.
  • anti-CD137 agonistic mAb increases trastuzumab- mediated
  • mice Post-tumor inoculation, mice then received either rat IgG control on day 3, trastuzumab antibody on day 3, anti-CD137 antibody on day 4, or trastuzumab on day 3 and anti-CD137 antibody on day 4 with each treatment repeated weekly for a total of three weeks. Mice were then monitored for tumor growth and overall survival.
  • anti-CD137 agonistic mAb enhances anti-breast cancer activity of trastuzumab in-vivo while retaining HER2 specificity.
  • mice were inoculated with 5x10 6 MCF7 breast tumor cells, subcutaneously, on the left flank, and 5x10 6 HER18 breast tumor cells, subcutaneously, on the right flank 1 day after subcutaneous injection of 0.72mg/60 day release beta-estradiol pellet. Post-tumor inoculation, mice then received either trastuzumab on day 3, or trastuzumab on day 3 and anti-CD137 antibody on day 4 with each treatment repeated weekly for a total of three weeks. Representative mice were then monitored for tumor growth
  • Anti-CD137 agonistic mAb enhances anti-breast cancer activity of trastuzumab in- vivo against HER2 + primary breast tumor, as shown in Figure 16.
  • SCID mice were inoculated with 1 x10 s HER2 + primary breast tumor cells by intramammary injection 24 hours after 200 cGy total body irradiation (TBI).
  • TBI total body irradiation
  • mice were randomized to one of four groups including IgG control with treatment on day 40, trastuzumab on day 40, anti-CD137 mAb on day 41 , or trastuzumab on day 40 and anti-CD137 mAb on day 41 .
  • Treatment was repeated weekly in each group for a total of three treatments. Mice were monitored for tumor growth.
  • Cetuximab induces CD137 upregulation on human NK cells following incubation with
  • EGFR-positive tumor cells as shown in Figures 17A-17B.
  • Peripheral blood from three healthy donors was analyzed for CD137 expression on CD3 " CD56 + NK cells after 24 hour culture with head and neck cancer cell lines and media alone, rituximab or cetuximab, and shown to have increased expression.
  • anti-CD137 agonistic mAb increases cetuximab- mediated NK cell cytotoxicity on tumor cells as assayed by cell viability.
  • Preactivated NK cells were purified before being incubated with SCC6, PC1 , and SCC4 for 24 hours.
  • the percent of apoptotic target cells was determined by annexin and 7AAD viability staining after culture with tumor alone or tumor and NK cells with media, cetuximab, anti-CD137, cetuximab and anti-CD137 antibodies.
  • Anti-CD137 agonistic mAb increases cetuximab-mediated NK cell cytotoxicity on tumor cells as assayed by chromium release.
  • NK cell cytotoxicity on SCC6 tumor cells was analyzed in chromium release assay. Fresh and preactivated NK cells were purified before being incubated with chromium labeled SCC6 cells for 5 hours. Shown in Figure 19 is percent lysis of target cells by chromium release at varying effector (fresh NK cells) :target (SCC6) cell ratios.
  • Anti-CD137 agonistic mAb enhances anti-head and neck cancer activity of cetuximab in-vivo.
  • Nu/nu nude mice were inoculated with 3x10 6 SCC6 head and neck tumor cells, subcutaneously, on the abdomen, shown in Figure 20. Post-tumor inoculation, mice received either rat IgG control on day 21 , cetuximab on day 21 , anti-CD137 antibody on day 22, or cetuximab on day 21 and anti-CD137 antibody on day 22 with each treatment repeated weekly for a total of three treatments. Mice were then monitored for tumor growth.
  • NK cell function is dramatically increased when both antibodies are combined.
  • the present invention is of high importance as it has the opportunity to demonstrate the clinical value of anti-CD137 combination antibody therapy, despite minimal single agent activity in solid tumors.
  • Therapy based on synergy through immunomodulation is an innovative and paradigm changing approach to improving the survival of patients with many types of cancer.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Biomedical Technology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Microbiology (AREA)
  • Reproductive Health (AREA)
  • Pregnancy & Childbirth (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Hospice & Palliative Care (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention a pour objet des méthodes d'amplification de l'efficacité de la cytotoxicité cellulaire dirigée par un anticorps (ADCC) pour un traitement orienté vers la destruction des cellules tumorales. Les antigènes de la surface cellulaire spécifiques d'un cancer sont liés par des anticorps monoclonaux, stimulant ainsi une réponse des lymphocytes T cytotoxiques caractérisée par une régulation à la hausse de l'expression par la surface cellulaire de protéines costimulatrices sur le lymphocyte T. La réponse ADCC augmente par l'administration subséquente d'un second anticorps qui est un agoniste de la molécule costimulatrice.
PCT/US2010/059221 2009-12-07 2010-12-07 Méthodes d'amplification d'un traitement à base d'anticorps anticancéreux WO2011071871A1 (fr)

Priority Applications (12)

Application Number Priority Date Filing Date Title
KR1020127015865A KR101853702B1 (ko) 2009-12-07 2010-12-07 항-종양 항체 치료를 향상시키는 방법
JP2012543196A JP5950824B2 (ja) 2009-12-07 2010-12-07 抗腫瘍抗体療法を増強するための方法
US13/513,523 US9005619B2 (en) 2009-12-07 2010-12-07 Methods for enhancing anti-tumor antibody therapy
EP10836513.1A EP2509627B1 (fr) 2009-12-07 2010-12-07 Méthodes d'amplification d'un traitement à base d'anticorps anticancéreux
AU2010328347A AU2010328347B2 (en) 2009-12-07 2010-12-07 Methods for enhancing anti-tumor antibody therapy
BR112012013736A BR112012013736A2 (pt) 2009-12-07 2010-12-07 processo para intesificação de terapia com anticorpos antitumor
SG2012041505A SG181533A1 (en) 2009-12-07 2010-12-07 Methods for enhancing anti-tumor antibody therapy
MX2012006420A MX346912B (es) 2009-12-07 2010-12-07 Metodos para mejorar terapia con anticuerpos antitumor.
CN201080063243XA CN102753195A (zh) 2009-12-07 2010-12-07 用于增强抗肿瘤抗体疗法的方法
CA2781311A CA2781311C (fr) 2009-12-07 2010-12-07 Methodes d'amplification d'un traitement a base d'anticorps anticancereux
US14/542,233 US9758589B2 (en) 2009-12-07 2014-11-14 Methods for enhancing anti-tumor antibody therapy
US15/669,418 US10633450B2 (en) 2009-12-07 2017-08-04 Methods for enhancing anti-tumor antibody therapy

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US26733709P 2009-12-07 2009-12-07
US61/267,337 2009-12-07
US28706709P 2009-12-16 2009-12-16
US61/287,067 2009-12-16
US35830310P 2010-06-24 2010-06-24
US61/358,303 2010-06-24

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/513,523 A-371-Of-International US9005619B2 (en) 2009-12-07 2010-12-07 Methods for enhancing anti-tumor antibody therapy
US14/542,233 Continuation US9758589B2 (en) 2009-12-07 2014-11-14 Methods for enhancing anti-tumor antibody therapy

Publications (1)

Publication Number Publication Date
WO2011071871A1 true WO2011071871A1 (fr) 2011-06-16

Family

ID=44145876

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/059221 WO2011071871A1 (fr) 2009-12-07 2010-12-07 Méthodes d'amplification d'un traitement à base d'anticorps anticancéreux

Country Status (11)

Country Link
US (3) US9005619B2 (fr)
EP (1) EP2509627B1 (fr)
JP (1) JP5950824B2 (fr)
KR (1) KR101853702B1 (fr)
CN (1) CN102753195A (fr)
AU (3) AU2010328347B2 (fr)
BR (1) BR112012013736A2 (fr)
CA (1) CA2781311C (fr)
MX (1) MX346912B (fr)
SG (2) SG10201501784YA (fr)
WO (1) WO2011071871A1 (fr)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2012224631A (ja) * 2011-04-19 2012-11-15 Pfizer Inc 癌の治療のための抗−4−1bb抗体及びadcc誘導抗体の組合せ
US8821867B2 (en) 2010-09-09 2014-09-02 Pfizer Inc 4-1BB binding molecules
US9006399B2 (en) 2010-08-23 2015-04-14 Board Of Regents, The University Of Texas System Anti-OX40 antibodies and methods of using the same
US20160235842A1 (en) * 2013-07-15 2016-08-18 The Board Of Trustees Of The Leland Stanford Junior University Medical uses of cd38 agonists
US9464139B2 (en) 2013-08-30 2016-10-11 Amgen Inc. GITR antigen binding proteins and methods of use thereof
EP3091031A1 (fr) * 2015-05-04 2016-11-09 Affimed GmbH Combinaison d'un anticorps bispécifique avec une molécule modulatrice immunitaire pour le traitement d'une tumeur
US9644032B2 (en) 2015-05-29 2017-05-09 Bristol-Myers Squibb Company Antibodies against OX40 and uses thereof
US9701751B2 (en) 2009-09-03 2017-07-11 Merck Sharp & Dohme Corp. Anti-GITR antibodies
WO2018020273A1 (fr) * 2016-07-29 2018-02-01 University Of Southampton Thérapie contre le cancer et les maladies liées aux lymphocytes b
US9957323B2 (en) 2016-06-20 2018-05-01 Kymab Limited Anti-ICOS antibodies
US9975957B2 (en) 2014-03-31 2018-05-22 Genentech, Inc. Anti-OX40 antibodies and methods of use
US10526413B2 (en) 2015-10-02 2020-01-07 Hoffmann-La Roche Inc. Bispecific antibodies specific for OX40
WO2020033851A1 (fr) * 2018-08-09 2020-02-13 Notable Labs, Inc. Compositions comprenant des agents d'amorçage et leur utilisation dans le traitement du cancer
US10662247B2 (en) 2014-10-08 2020-05-26 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy
WO2020150496A1 (fr) * 2019-01-16 2020-07-23 Compass Therapeutics Llc Formulations d'anticorps qui se lient au cd137 humain et leurs utilisations
US11046776B2 (en) 2016-08-05 2021-06-29 Genentech, Inc. Multivalent and multiepitopic antibodies having agonistic activity and methods of use
US11440960B2 (en) 2017-06-20 2022-09-13 Kymab Limited TIGIT antibodies, encoding nucleic acids and methods of using said antibodies in vivo
US11459394B2 (en) 2017-02-24 2022-10-04 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
US11629189B2 (en) 2017-12-19 2023-04-18 Kymab Limited Bispecific antibody for ICOS and PD-L1
US11753479B2 (en) 2014-03-04 2023-09-12 Kymab Limited Nucleic acids encoding anti-OX40L antibodies
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
US11858996B2 (en) 2016-08-09 2024-01-02 Kymab Limited Anti-ICOS antibodies
EP4045018A4 (fr) * 2019-10-18 2024-01-10 The University of North Carolina at Chapel Hill Procédés et compositions pour le traitement du cancer utilisant des nanoparticules conjuguées à de multiples ligands pour se lier à des récepteurs sur des cellules tueuses naturelles

Families Citing this family (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011071871A1 (fr) 2009-12-07 2011-06-16 The Board Of Trustees Of The Leland Stanford Junior University Méthodes d'amplification d'un traitement à base d'anticorps anticancéreux
TW202132337A (zh) 2014-05-28 2021-09-01 美商艾吉納斯公司 抗糖皮質素誘導性tnfr家族相關性受體(gitr)抗體類及使用彼等之方法
WO2015188047A1 (fr) * 2014-06-06 2015-12-10 University Of Maryland, Baltimore Anticorps monoclonaux anti-cd-137 présentant des capacités de liaison distinctes au fcγr pour le traitement d'un cancer ou d'une auto-immunité
WO2016054638A1 (fr) * 2014-10-03 2016-04-07 Dana-Farber Cancer Institute, Inc. Anticorps dirigés contre le récepteur du facteur de nécrose tumorale induit par glucocorticoïdes (gitr) et leurs procédés d'utilisation
MA41414A (fr) * 2015-01-28 2017-12-05 Centre Nat Rech Scient Protéines de liaison agonistes d' icos
HUE049938T2 (hu) 2015-03-23 2020-11-30 Jounce Therapeutics Inc Icos elleni antitestek
WO2016167809A1 (fr) * 2015-04-17 2016-10-20 The Board Of Trustees Of The Leland Stanford Junior University Traitement par t-dm1 amélioré
CN115109158A (zh) 2015-05-07 2022-09-27 阿吉纳斯公司 抗ox40抗体及其使用方法
JP2018524404A (ja) * 2015-07-07 2018-08-30 インセルム(インスティチュート ナショナル デ ラ サンテ エ デ ラ リシェルシェ メディカル) Nk細胞殺傷活性を増強する方法および医薬組成物
MX2018000948A (es) 2015-07-23 2018-09-27 Inhibrx Inc Proteinas de fusion que se unen a gitir multivalentes y multiespecificas.
WO2017040374A1 (fr) * 2015-08-31 2017-03-09 The Trustees Of The University Of Pennsylvania Compositions et procédés d'amélioration de la réponse antitumorale à l'aide des neutrophiles hybrides
TW201723190A (zh) 2015-10-22 2017-07-01 永斯醫療股份有限公司 用於測定icos表現之基因印記
JP7089470B2 (ja) 2015-12-02 2022-06-22 アジェナス インコーポレイテッド 抗体およびその使用方法
JP2019519536A (ja) 2016-07-01 2019-07-11 ファイブ プライム セラピューティクス, インコーポレイテッド Gitrアゴニストおよびcpgを用いた組み合わせ抗腫瘍療法
CA3041340A1 (fr) 2016-11-09 2018-05-17 Agenus Inc. Anticorps anti-ox40, anticorps anti-gitr, et leurs procedes d'utilisation
BR112019017241A2 (pt) 2017-04-13 2020-04-14 Agenus Inc anticorpos anti-cd137 e métodos de uso dos mesmos
SG10201913144TA (en) 2017-07-11 2020-03-30 Compass Therapeutics Llc Agonist antibodies that bind human cd137 and uses thereof
US11698367B2 (en) 2017-09-15 2023-07-11 Beckman Coulter, Inc. Flow based assays for therapeutics
US11718679B2 (en) 2017-10-31 2023-08-08 Compass Therapeutics Llc CD137 antibodies and PD-1 antagonists and uses thereof
WO2019100052A2 (fr) 2017-11-20 2019-05-23 Compass Therapeutics Llc Anticorps cd137 et anticorps ciblant un antigène tumoral et leurs utilisations
KR20200131282A (ko) * 2018-03-13 2020-11-23 에프. 호프만-라 로슈 아게 표적화된 4-1bb(cd137) 작용제에 의한 조합 치료
CA3103629A1 (fr) 2018-06-15 2019-12-19 Flagship Pioneering Innovations V, Inc. Augmentation de l'activite immunitaire par modulation de facteurs de signalisation post-cellulaires
KR20200030337A (ko) * 2018-09-12 2020-03-20 주식회사 녹십자랩셀 종양 치료를 위한 항-cd 19 항체 및 자연살해세포를 포함하는 약학적 조합물
EP3958908A1 (fr) 2019-04-24 2022-03-02 Heidelberg Pharma Research GmbH Conjugués anticorps-médicaments d'amatoxine et leurs utilisations
WO2020227159A2 (fr) 2019-05-03 2020-11-12 Flagship Pioneering Innovations V, Inc. Métodes de modulation de l'activité immunitaire
EP4076434A1 (fr) 2019-12-17 2022-10-26 Flagship Pioneering Innovations V, Inc. Polythérapies anticancéreuses ayant des inducteurs de désassemblage cellulaire dépendant du fer
CN113491768A (zh) * 2020-04-01 2021-10-12 深圳市罗湖区人民医院 Cd137抗体在制备促进nk细胞表达cd16分子的药物中的应用
WO2021202917A1 (fr) * 2020-04-01 2021-10-07 The Board Of Trustees Of The Leland Stanford Junior University Approche multiparamétrique non invasive pour l'identification précoce d'un bénéfice thérapeutique à partir de l'inhibition du point de contrôle immunitaire pour le cancer du poumon
EP4172323A1 (fr) 2020-06-29 2023-05-03 Flagship Pioneering Innovations V, Inc. Virus modifiés pour favoriser la thanotransmission et leur utilisation dans le traitement du cancer
US11661459B2 (en) 2020-12-03 2023-05-30 Century Therapeutics, Inc. Artificial cell death polypeptide for chimeric antigen receptor and uses thereof
EP4263600A1 (fr) 2020-12-18 2023-10-25 Century Therapeutics, Inc. Systèmes récepteurs antigéniques chimériques ayant une spécificité de récepteur adaptable
JP2024512669A (ja) 2021-03-31 2024-03-19 フラグシップ パイオニアリング イノベーションズ ブイ,インコーポレーテッド タノトランスミッションポリペプチド及び癌の処置におけるそれらの使用
KR20240026507A (ko) 2021-06-29 2024-02-28 플래그쉽 파이어니어링 이노베이션스 브이, 인크. 타노트랜스미션을 촉진시키도록 엔지니어링된 면역 세포 및 이의 용도
WO2024077191A1 (fr) 2022-10-05 2024-04-11 Flagship Pioneering Innovations V, Inc. Molécules d'acide nucléique codant pour des trif et des polypeptides supplémentaires et leur utilisation dans le traitement du cancer

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040018194A1 (en) * 2000-11-28 2004-01-29 Francisco Joseph A. Recombinant anti-CD30 antibodies and uses thereof
US20070071675A1 (en) * 2005-08-19 2007-03-29 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
US20080003225A1 (en) * 2006-06-29 2008-01-03 Henri Vie Method for enhancing the antibody-dependent cellular cytotoxicity (ADCC) and uses of T cells expressing CD16 receptors
US20090214544A1 (en) * 2005-04-25 2009-08-27 Medarex Method of treating cd30 positive lymphomas

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030035790A1 (en) * 1999-01-15 2003-02-20 Shu-Hsia Chen Combination therapy for the prevention or treatment of cancer, inflammatory disorders or infectious diseases in a subject
TW200303759A (en) 2001-11-27 2003-09-16 Schering Corp Methods for treating cancer
FR2844513B1 (fr) 2002-09-13 2007-08-03 Lab Francais Du Fractionnement Anticorps pour adcc et induisant la production de cytokines.
US7288638B2 (en) 2003-10-10 2007-10-30 Bristol-Myers Squibb Company Fully human antibodies against human 4-1BB
WO2006029275A2 (fr) 2004-09-08 2006-03-16 Genentech, Inc. Methodes d'utilisation de ligands de recepteurs de mort, ainsi que d'anticorps cd20
EP2495257A3 (fr) * 2005-08-19 2012-10-17 Abbott Laboratories Immunoglobuline à double domaine variable et ses utilisations
US8475790B2 (en) 2008-10-06 2013-07-02 Bristol-Myers Squibb Company Combination of CD137 antibody and CTLA-4 antibody for the treatment of proliferative diseases
WO2011071871A1 (fr) * 2009-12-07 2011-06-16 The Board Of Trustees Of The Leland Stanford Junior University Méthodes d'amplification d'un traitement à base d'anticorps anticancéreux
WO2011146382A1 (fr) 2010-05-17 2011-11-24 Bristol-Myers Squibb Company Régimes posologiques immunothérapeutiques améliorés et combinaisons de ceux-ci
WO2012027536A1 (fr) 2010-08-26 2012-03-01 Bristol-Myers Squibb Company Combinaison d'anticorps anti-ctla4 avec des inhibiteurs de braf pour le traitement synergique de maladies prolifératives
CN103476407A (zh) 2010-12-09 2013-12-25 霍夫曼-拉罗奇有限公司 用帕利他赛和曲妥珠单抗-mcc-dm1治疗her2阳性癌症
CN112587658A (zh) 2012-07-18 2021-04-02 博笛生物科技有限公司 癌症的靶向免疫治疗
CA2887027C (fr) 2012-10-02 2021-11-16 Bristol-Myers Squibb Company Combinaison d'anticorps anti-kir et d'anticorps anti-pd-1 pour le traitement du cancer
WO2014066532A1 (fr) 2012-10-23 2014-05-01 Bristol-Myers Squibb Company Association d'anticorps anti-kir et anti-ctla-4 pour le traitement du cancer

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040018194A1 (en) * 2000-11-28 2004-01-29 Francisco Joseph A. Recombinant anti-CD30 antibodies and uses thereof
US20090214544A1 (en) * 2005-04-25 2009-08-27 Medarex Method of treating cd30 positive lymphomas
US20070071675A1 (en) * 2005-08-19 2007-03-29 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
US20080003225A1 (en) * 2006-06-29 2008-01-03 Henri Vie Method for enhancing the antibody-dependent cellular cytotoxicity (ADCC) and uses of T cells expressing CD16 receptors

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
MELERO ET AL.: "Immunostimulatory monoclonal antibodies for cancer therapy", NATURE REVIEWS CANCER, vol. 7, February 2007 (2007-02-01), pages 95 - 106, XP002716002, DOI: doi:10.1038/NRC2051 *
See also references of EP2509627A4 *
TERME M, ET AL.: "Natura killer cell-directed therapies: moving from unexpected results to successful strategies", NATURE IMMUNOLOGY, vol. 9, no. 5, May 2008 (2008-05-01), pages 486 - 494, XP008159145 *

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9701751B2 (en) 2009-09-03 2017-07-11 Merck Sharp & Dohme Corp. Anti-GITR antibodies
US10400040B2 (en) 2009-09-03 2019-09-03 Merck Sharp & Dohme Corp. Anti-GITR antibodies
US9163085B2 (en) 2010-08-23 2015-10-20 Board Of Regents, The University Of Texas System Anti-OX40 antibodies and methods of treating cancer
US10196450B2 (en) 2010-08-23 2019-02-05 Board Of Regents, The University Of Texas System Anti-OX40 antibodies and methods of using the same
US9006399B2 (en) 2010-08-23 2015-04-14 Board Of Regents, The University Of Texas System Anti-OX40 antibodies and methods of using the same
US10851173B2 (en) 2010-08-23 2020-12-01 Board Of Regents, The University Of Texas System Anti-OX40 antibodies and methods of using the same
US9695246B2 (en) 2010-08-23 2017-07-04 Board Of Regents, The University Of Texas System Anti-OX40 antibodies and methods of using the same
US9527917B2 (en) 2010-08-23 2016-12-27 Board Of Regents, The University Of Texas System Nucleic acid encoding anti-OX40 antibodies
US10640568B2 (en) 2010-09-09 2020-05-05 Pfizer Inc. 4-1BB binding molecules
US9468678B2 (en) 2010-09-09 2016-10-18 Pfizer Inc. Method of producing 4-1BB binding molecules and associated nucleic acids
US8821867B2 (en) 2010-09-09 2014-09-02 Pfizer Inc 4-1BB binding molecules
WO2012145183A3 (fr) * 2011-04-19 2013-03-14 Pfizer Inc. Combinaisons d'anticorps anti-4-1bb et d'anticorps induisant une cytotoxicité à médiation cellulaire dépendante d'un anticorps (adcc) pour le traitement du cancer
JP2018115192A (ja) * 2011-04-19 2018-07-26 ファイザー・インク 癌の治療のための抗−4−1bb抗体及びadcc誘導抗体の組合せ
JP2012224631A (ja) * 2011-04-19 2012-11-15 Pfizer Inc 癌の治療のための抗−4−1bb抗体及びadcc誘導抗体の組合せ
US20160235842A1 (en) * 2013-07-15 2016-08-18 The Board Of Trustees Of The Leland Stanford Junior University Medical uses of cd38 agonists
US9464139B2 (en) 2013-08-30 2016-10-11 Amgen Inc. GITR antigen binding proteins and methods of use thereof
US11753479B2 (en) 2014-03-04 2023-09-12 Kymab Limited Nucleic acids encoding anti-OX40L antibodies
US11773175B2 (en) 2014-03-04 2023-10-03 Kymab Limited Antibodies, uses and methods
US9975957B2 (en) 2014-03-31 2018-05-22 Genentech, Inc. Anti-OX40 antibodies and methods of use
US10730951B2 (en) 2014-03-31 2020-08-04 Genentech, Inc. Anti-OX40 antibodies and methods of use
US10662247B2 (en) 2014-10-08 2020-05-26 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy
EP3091031A1 (fr) * 2015-05-04 2016-11-09 Affimed GmbH Combinaison d'un anticorps bispécifique avec une molécule modulatrice immunitaire pour le traitement d'une tumeur
US10683357B2 (en) 2015-05-29 2020-06-16 Bristol-Myers Squibb Company Antibodies against OX40 and uses thereof
US9644032B2 (en) 2015-05-29 2017-05-09 Bristol-Myers Squibb Company Antibodies against OX40 and uses thereof
US11466092B2 (en) 2015-05-29 2022-10-11 Bristol-Myers Squibb Company Antibodies against OX-40 and uses thereof
US10526413B2 (en) 2015-10-02 2020-01-07 Hoffmann-La Roche Inc. Bispecific antibodies specific for OX40
US11965026B2 (en) 2016-06-20 2024-04-23 Kymab Limited Anti-PD-L1 and IL-2 cytokines
US10604576B2 (en) 2016-06-20 2020-03-31 Kymab Limited Antibodies and immunocytokines
US9957323B2 (en) 2016-06-20 2018-05-01 Kymab Limited Anti-ICOS antibodies
WO2018020273A1 (fr) * 2016-07-29 2018-02-01 University Of Southampton Thérapie contre le cancer et les maladies liées aux lymphocytes b
US11046776B2 (en) 2016-08-05 2021-06-29 Genentech, Inc. Multivalent and multiepitopic antibodies having agonistic activity and methods of use
US11858996B2 (en) 2016-08-09 2024-01-02 Kymab Limited Anti-ICOS antibodies
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
US11942149B2 (en) 2017-02-24 2024-03-26 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
US11459394B2 (en) 2017-02-24 2022-10-04 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
US11440960B2 (en) 2017-06-20 2022-09-13 Kymab Limited TIGIT antibodies, encoding nucleic acids and methods of using said antibodies in vivo
US11629189B2 (en) 2017-12-19 2023-04-18 Kymab Limited Bispecific antibody for ICOS and PD-L1
WO2020033851A1 (fr) * 2018-08-09 2020-02-13 Notable Labs, Inc. Compositions comprenant des agents d'amorçage et leur utilisation dans le traitement du cancer
CN113474371A (zh) * 2019-01-16 2021-10-01 指南针制药有限责任公司 与人cd137结合的抗体的制剂及其用途
WO2020150496A1 (fr) * 2019-01-16 2020-07-23 Compass Therapeutics Llc Formulations d'anticorps qui se lient au cd137 humain et leurs utilisations
EP4045018A4 (fr) * 2019-10-18 2024-01-10 The University of North Carolina at Chapel Hill Procédés et compositions pour le traitement du cancer utilisant des nanoparticules conjuguées à de multiples ligands pour se lier à des récepteurs sur des cellules tueuses naturelles

Also Published As

Publication number Publication date
CA2781311A1 (fr) 2011-06-16
AU2016201190A1 (en) 2016-03-17
AU2010328347B2 (en) 2015-12-17
EP2509627A4 (fr) 2013-12-25
BR112012013736A2 (pt) 2018-08-14
US9005619B2 (en) 2015-04-14
CA2781311C (fr) 2019-03-12
US9758589B2 (en) 2017-09-12
US10633450B2 (en) 2020-04-28
KR20120102714A (ko) 2012-09-18
JP5950824B2 (ja) 2016-07-13
EP2509627A1 (fr) 2012-10-17
SG10201501784YA (en) 2015-05-28
CN102753195A (zh) 2012-10-24
EP2509627B1 (fr) 2017-01-25
KR101853702B1 (ko) 2018-05-03
US20160083474A1 (en) 2016-03-24
AU2010328347A1 (en) 2012-06-21
JP2013512958A (ja) 2013-04-18
SG181533A1 (en) 2012-07-30
US20120321646A1 (en) 2012-12-20
US20180208670A1 (en) 2018-07-26
MX346912B (es) 2017-04-05
AU2017268492A1 (en) 2017-12-14
MX2012006420A (es) 2012-12-05

Similar Documents

Publication Publication Date Title
US10633450B2 (en) Methods for enhancing anti-tumor antibody therapy
US11078295B2 (en) Use of semaphorin-4D inhibitory molecules with an immune modulating therapy to inhibit tumor growth and metastases
US11608377B2 (en) Enhanced depletion of targeted cells with CD47 blockade and an immune costimulatory agonist
JP2016534090A (ja) Cd38アゴニストの医学的使用
US12030944B2 (en) Enhanced depletion of targeted cells with CD47 blockade and an immune costimulatory agonist

Legal Events

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

Ref document number: 201080063243.X

Country of ref document: CN

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

Ref document number: 10836513

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2781311

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2010328347

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: MX/A/2012/006420

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2012543196

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20127015865

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2010328347

Country of ref document: AU

Date of ref document: 20101207

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 5576/CHENP/2012

Country of ref document: IN

REEP Request for entry into the european phase

Ref document number: 2010836513

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2010836513

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13513523

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012013736

Country of ref document: BR

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112012013736

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012013736

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20120606