US20100086550A1 - Anticancer agent comprising anti-pd-1 antibody or anti-pd-l1 antibody - Google Patents

Anticancer agent comprising anti-pd-1 antibody or anti-pd-l1 antibody Download PDF

Info

Publication number
US20100086550A1
US20100086550A1 US12/410,732 US41073209A US2010086550A1 US 20100086550 A1 US20100086550 A1 US 20100086550A1 US 41073209 A US41073209 A US 41073209A US 2010086550 A1 US2010086550 A1 US 2010086550A1
Authority
US
United States
Prior art keywords
cancer
antibody
inkt
cells
alpha
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/410,732
Inventor
Chang Yuil Kang
Woo Sung Chang
Ji Yeon Kim
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ANTICANCER AGENT COMPRISING ANTI-PD-1 ANTIBODY OR ANTI-PD-L1 ANTIBODY
Seoul National University Industry Foundation
Original Assignee
Seoul National University Industry Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Seoul National University Industry Foundation filed Critical Seoul National University Industry Foundation
Assigned to ANTICANCER AGENT COMPRISING ANTI-PD-1 ANTIBODY OR ANTI-PD-L1 ANTIBODY reassignment ANTICANCER AGENT COMPRISING ANTI-PD-1 ANTIBODY OR ANTI-PD-L1 ANTIBODY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHANG, WOO SUNG, KANG, CHANG YUIL, KIM, JI YEON
Publication of US20100086550A1 publication Critical patent/US20100086550A1/en
Priority to US13/398,915 priority Critical patent/US8617546B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates, in general, to an anticancer agent and, more particularly, to an anticancer agent comprising an anti-PD-1 antibody or an anti-PD-L1 antibody as an active ingredient, functioning to restore the responsiveness of iNKT cells in which anergy has been induced by administration with an iNKT cell ligand.
  • Natural killer T (NKT) cells co-expressing a T cell receptor and NK cell markers, are essential for several aspects of immunity, such as immunomodulation and immunuopotentiation, in various immune diseases including autoimmune diseases, infectious diseases, cancer, etc.
  • NKT cells exist at high levels in the thymus, the liver, and the bone marrow, but at low levels in the spleen, lymph nodes and blood.
  • NKT cells recognize glycolipid antigens presented by CD1d, a MHC class 1-like molecule.
  • a major subset of NKT cells called type 1 NKT cells or invariant natural T (iNKT) cells, express an invariant natural T cell receptor (TCR) composed of V ⁇ 14-J ⁇ 18 chains in mice (V ⁇ 24-J ⁇ 18 in humans).
  • TCR invariant natural T cell receptor
  • iNKT cells Upon TCR stimulation with a ligand, such as ⁇ -galactosylceramide ( ⁇ -GC), iNKT cells rapidly produce a wide range of cytokines including IL-4, IFN- ⁇ , L-12, and GM-CSF.
  • iNKT cells This rapid and potent response to a ligand enables iNKT cells to enhance or regulate the activity of various immune cells in innate and acquired immunity. Found in diverse diseases and promoting tumor rejection or regulating autoimmune disorders, these immunomodulatory roles of iNKT cells are studied for use in immunotherapy treatments for cancer and autoimmune diseases.
  • iNKT cells tend to greatly decrease in responsiveness following repeated stimulation after a first stimulation with their ligands via the T cell receptor.
  • iNKT cells that have been stimulated in vivo with a-GC have reduced proliferation and cytokine production upon secondary stimulation with the same ligand. This iNKT cell anergy is a major obstacle in immunotherapeutic trials targeting iNKT cells.
  • T cells are known to become anergic when they receive a TCR signal with insufficient co-stimulatory signals.
  • Co-stimulatory molecules such as CD28, CD40L and ICOS are known to be involved in the development and activation of iNKT cells.
  • 4-1BB contributes to promote the activation of iNKT cells as a co-stimulatory molecule and can affect iNKT cell-mediated allergic lung inflammation (Kim, D. H., W. S. Chang, Y. S. Lee, K. A. Lee, Y. K. Kim, B. S. Kwon, and C. Y. Kang. 2008.
  • PD-1 is a 55 KDa type 1 transmebrane protein of the immunoglobulin superfamily, and is known as a co-inhibitory molecule on T cells. That is, PD-1 is a member of the co-inhibitory molecules of the CD28 family (e.g., CD28, CTLA4, ICOS and BTLA) expressed on activated B cells, T cells and bone marrow cells. Two ligands for PD-1, PD-L1 and PD-L2, have been identified thus far. The interaction of PD-1 with the PD ligands can transduce inhibitory or co-stimulatory signals into the T cells.
  • PD-1 is not expressed on naive T cells, but is inducibly expressed after T cell activation.
  • PD-L1 it is expressed to some degree on naive T cells and its level is increased on activated T cells.
  • PD-L1 is found at high levels in various human cancers and interacts with PD-1 to transduce inhibitory or co-stimulatory signals from entering into the T cells. For example, the interaction between PD-1 and PD-L1 induces a decrease in the level of tumor invasive lymphocytes and in T cell receptor-mediated proliferation and causes the immune evasion of tumor cells.
  • PD-1 develops various autoimmune phenotypes, such as autoimmune cardiac infarction and lupus-like syndromes with arthritis and nephritis, and plays an important role in the development of autoimmune encephalomyelitis, systemic lupus erythematosus, graft-versus-host disease (GVHD), type 1 diabetes and rheumatic arthritis.
  • GVHD graft-versus-host disease
  • Aged PD-1-deficient mice develop autoimmune diseases, indicating that PD-1 plays a critical role in the regulation of autoimmunity and immune tolerance.
  • PD-1 signals are essential for inducing T cell exhaustion during chronic infection.
  • the NKT cell ligand ⁇ -GC has conventionally been used as an anticancer agent.
  • repeated stimulation of iNKT cells with ⁇ -GC induces anergy leading to a great decrease in responsiveness, it cannot achieve effective anticancer effects. Therefore, there is a pressing need for an anticancer agent that can restore the responsiveness of iNKT cells even in the state of anergy caused by stimulation with iNKT cell ligands.
  • the anti-PD1 or anti-PD-L1 antibody was found to induce potent anti-tumor activity of iNKT cells as demonstrated by a significant reduction in the number of metastatic nodules in B16F10 melanoma metastasis models in vivo.
  • FIG. 1 is of flow histograms showing the expression levels of PD-1, PD-L1 and PD-L2 on iNKT cells of the splenocytes isolated from ⁇ -GC-treated C57BL/6 mice at indicated time points (0 hr, 6 hrs, 72 hrs, 7 days, 1 month and 2 months) after treatment.
  • FIG. 2 is of histograms showing IFN- ⁇ and IL-4 levels in the supernatants obtained after incubating splenocytes isolated from na ⁇ ve C57BL/6 mice with ⁇ -GC in the presence of control rat IgG, anti-PD-1 monoclonal antibody, anti-PD-L1 monoclonal antibody or anti-PD-L2 monoclonal antibody as analyzed by ELISA (in vitro).
  • FIG. 3 is of plots showing IFN- ⁇ and IL-4 levels in the sera obtained from the C57BL/6 mice, which were intraperitoneally injected with control rat IgG, anti-PD-1 monoclonal antibody, anti-PD-L1 monoclonal antibody or anti-PD-L2 monoclonal antibody 24 hrs before ⁇ -GC treatment, at 0, 2, 6, 12, 24, 48 and 72 hrs after the ⁇ -GC treatment as analyzed by ELISA (in vivo).
  • FIG. 4 shows FACS results of IFN- ⁇ + or IL- 4 + iNKT cells on which intracellular cytokine staining was performed to examine if the increased production of IFN- ⁇ and IL-4 in the sera of FIG. 3 comes from iNKT cells.
  • FIG. 5 is of histograms showing IFN- ⁇ and IL-4 levels in the supernatants obtained after the splenocytes of C57BL/6 mice, in which iNKT cell anergy was induced by ⁇ -GC treatment 7 days and one month before splenocyte isolation therefrom, were incubated with ⁇ -GC in the presence of control rat IgG, anti-PD-1 monoclonal antibody, anti-PD-L1 monoclonal antibody or anti-PD-L2 monoclonal antibody, as analyzed by ELISA (in vitro).
  • FIG. 6 is of histograms showing IFN- ⁇ and L-4 levels in sera from C57BL/6 mice, as analyzed by ELISA.
  • the mice were intraperitoneally injected with control rat IgG, anti-PD-1 monoclonal antibody, anti-PD-L1 monoclonal antibody or anti-PD-L2 monoclonal antibody, followed by double injection with ⁇ -GC 24 hrs and 14 days later.
  • the sera were obtained 2 and 12 hrs after the secondary injection of ⁇ -GC (in vivo).
  • FIG. 7 shows FACS results of IFN- ⁇ + or IL-4 + iNKT cells after intracellular cytokine staining was performed on the splenocytes isolated 2 hrs after the secondary injection (in vivo).
  • FIG. 8 shows CD69 expression levels on iNKT and NK cells as measured by flow cytometry using the splenocytes isolated 12 hrs after the secondary injection (in vivo).
  • FIG. 9 is a histogram showing weights of normal and metastatic lungs of C57BL/6 mice.
  • the mice were intraperitoneally injected with control rat IgG, anti-PD-1 monoclonal antibody or anti-PD-L1 antibody 24 hrs before intravenous injection with skin tumor cells.
  • the mice On Days 0, 4 and 8, the mice were co-administered with ⁇ -GC and the control rat IgG, the anti-PD-1 antibody or the anti-PD-L1 antibody.
  • the lungs were excised 14 days after the co-administration.
  • FIG. 10 shows effects of the blockage of PD-1/PD-L1 interaction on the antitumor activity of iNKT cells in terms of the number of metastatic pulmonary nodules (A) and optical microscopic views (B).
  • C57BL/6 mice were intraperitoneally injected with control rat IgG, anti-PD-1 antibody or anti-PD-L1 antibody 24 hrs before ⁇ -GC treatment. 7 days later, the mice were intravenously injected with skin tumor cells and re-injected with ⁇ -GC on Days 0, 4 and 8. 14 days later, the lungs were excised.
  • the present invention provides an anticancer agent, comprising an iNKT cell ligand as a first anticancer factor and an anti-PD-1 antibody or anti-PD-L1 antibody as a second anticancer factor, having a function of reversing unresponsiveness of iNKT cells in which anergy is induced by administration with the iNKT cell ligand.
  • the present invention provides a method for reversing the unresponsiveness of iNKT cells with anergy induced therein by iNKT cell ligand treatment, comprising treating the anergic iNKT cells with an anti-PD-1 antibody or an anti-PD-L1 antibody.
  • the iNKT cell ligand may be selected from a group consisting of alpha-galactosyl ceramide, alpha-glucuronosyl ceramide, phosphatidylinositoltetramannoside, isoglobotrihexosylceramide, ganglioside GD3, phsphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, sulfatide, beta-galactosylceramide, lipophosphoglycan, glycoinositol phospholipid, alpha-galactosylceramide analogs including beta-anomer galactoceramide and alpha-anomer galactosylceramide, and bacterial lipid antigens.
  • the anti-PD-1 antibody or the anti-PD-L1 antibody may be a monoclonal antibody or a polyclonal antibody.
  • the anti-PD-1 antibody or anti-PD-L1 antibody according to the present invention can block the signaling of PD-1 or PD-L1 to prevent the iNKT cell ligand-induced anergy of iNKT cells and can provide cytokine secretion ability for even anergic iNKT cells to restore their responsiveness.
  • the anti-PD-1 antibody or anti-PD-L1 antibody of the present invention inhibits the anergy induction of iNKT cells to significantly decrease the number of pulmonary nodules in a lung metastasis model of B16F10 melanoma, thus effectively eliciting anticancer immune responses of iNKT cells and showing anti-tumor effects of iNKT cells against cancer metastasis. Therefore, an anticancer agent comprising the anti-PD-1 antibody or anti-PD-L 1 antibody of the present invention can be very useful in the treatment of cancer, particularly, metastatic cancer.
  • the cancer to which the anticancer agent according to the present invention is therapeutically applicable may be gynecologic tumor, endocrine gland cancer, CNS (central nervous system) tumor or ureter cancer.
  • the cancer include lung cancer, stomach cancer, liver cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, melanoma, uterine cancer, ovarian cancer, rectal cancer, colorectal cancer, colon cancer, breast cancer, uterine sarcoma, Fallopian tube carcinoma, endometrial carcinoma, uterine cervical carcinoma, vaginal carcinoma, vulva carcinoma, esophageal cancer, larynx cancer, small intestine cancer, thyroid cancer, parathyroid cancer, soft tissue sarcoma, uterine cancer, penis cancer, prostate cancer, chronic or acute leukemia, pediatric solid tumor, differentiated lymphoma, bladder cancer, kidney cancer, renal cell carcinoma, renal pelvic carcinoma, primary CNS lymphoma, spinal cord tumor, brainstem glioma, and pitu
  • the anticancer agent of the present invention may be formulated into a pharmaceutical composition with at least one conventional anticancer ingredient.
  • the pharmaceutical composition may further comprise a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier include saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol, etc.
  • conventional additives such as antioxidants, buffers, bacteriostatic agents, etc., may be added to the composition.
  • the active ingredients may be admixed with a diluent, a dispersant, a surfactant, a binder and/or a lubricant.
  • composition of the present invention may be administered via oral routes or parenteral routes (e.g., intravenous, subcutaneous, intraperitoneal, topical, etc.).
  • parenteral routes e.g., intravenous, subcutaneous, intraperitoneal, topical, etc.
  • the effective dosage of the anticancer agent in accordance with the present invention depends on various factors, including the patient's weight, age, gender, state of health, diet, the time of administration, route of administration, excretion rate, severity of diseases, etc. In general, it may be administered in a single dose, and preferably in multiple doses per day at a daily dose ranging from 0.01 to 1000 mg/day, and preferably from 0.1 to 100 mg/kg of the anti-PD-1 antibody or anti-PD-L1 antibody.
  • composition according to the present invention may be used alone or in combination with surgical operation, hormonal therapy, chemotherapy, and/or biological response controllers.
  • mice Six- to eight-week-old female C56BL/6 mice were purchased from Orient Bio. All mice were bred and maintained in specific pathogen-free conditions.
  • Hybridoma clones producing antibodies to mouse PD-1 (RMP1-14, rat IgG2a), PD-L1 (MIH-5, rat IgG2a), and PD-L2 (Ty25, rat IgG2a) were produced according to methods well known in the art [Yamazaki, T., H. Akiba, H. Iwai, H. Matsuda, M. Aoki, Y. Tanno, T. Shin, H. Tsuchiya, D. M. Pardoll, K. Okumura, M. Azuma, and H. Yagita. 2002.
  • Splenocytes were isolated from C57BL/6 mice administered with 2 ⁇ g of ⁇ -GC at different time points (0 hr, 6 hrs, 72 hrs, 7 days, one month and two months) after the administration.
  • the cells were stained with a PE-conjugated anti-PD-1 monoclonal antibody, a PE-conjugated anti-PD-L1 monoclonal antibody, a PE-conjugated anti-PD-L2 monoclonal antibody and a PE-conjugated isotype control monoclonal antibody, respectively.
  • iNKT cells were gated on B220 ⁇ TCR- ⁇ int ⁇ -GC/CD1d:Ig + population.
  • PD-1, PD-L1 and PD-L2 expression (open histograms) was analyzed by FACS. The results are given in FIG. 1 . Shaded histograms indicate staining with isotype control niAbs.
  • PD-1 was constitutively expressed on iNKT cells at a low level and its expression was upregulated after ⁇ -GC stimulation, followed by the persistence of the upregulated level for two months.
  • PD-L1 expression was temporarily increased on ⁇ -GC-stimulated iNKT cells but declined toward na ⁇ ve state levels within 72 hrs.
  • PD-L2 its expression was not found on iNKT cells regardless of their activation status.
  • spelenocytes After being prepared from na ⁇ ve C57BL/6 mice, 5 ⁇ 10 5 spelenocytes were incubated for 3 days with 100 ng/ml of ⁇ -GC in the presence of 50 ⁇ g/ml of the control rat IgG, the anti-PD-1 mAb, the anti-PD-L1 mAb, or the anti-PD-L2 mAb. Then, the supernatants were obtained and assayed for IFN- ⁇ and IL-4 levels by ELISA.
  • anti-PD-1 mAb significantly increased the production of IFN- ⁇ and, to a lesser extent, IL-4.
  • Anti-PD-L1 mAb also induced greater production of IFN- ⁇ , but anti-PD-L2 mAb did not.
  • C57BL/6 mice was injected with 200 ⁇ g of the control rat IgG, the anti-PD-1 mAb, the anti-PD-L1 mAb or the anti-PD-L2 mAb 24 hrs before treatment with 2 ⁇ g of ⁇ -GC.
  • Sera were obtained at 0, 2, 6, 12, 24, 48 and 72 hrs after the treatment, followed by ELISA analysis for IFN- ⁇ and IL-4 levels.
  • cytokine staining was followed by flow cytometry analysis.
  • splenocytes were isolated 2 hrs after ⁇ -GC treatment and 5 ⁇ 10 6 cells were incubated with Golgi plug for 2 hrs to accumulate cytokines.
  • Intracellular cytokine staining was performed using BD Cytofix/Cytoperm Plus with Golgiplug kit according to the manufacturer's protocol (BD Biosciences).
  • iNKT cells were gated on B220 ⁇ TCR- ⁇ int ⁇ -GC/CD1d:Ig + population and IFN- ⁇ + or IL-4 + iNKT cells were analyzed by flow cytometry.
  • IFN- ⁇ and IL-4 levels in blood are depicted in FIG. 3 and flow histograms of IFN- ⁇ + and IL-4 + iNKT cells are given in FIG. 4 .
  • the blood IFN- ⁇ and IL-4 levels were observed to peak, respectively, within 12 hrs and 2 hrs after ⁇ -GC treatment, indicating iNKT cell activation.
  • IFN- ⁇ levels in the sera of anti-PD-1 or anti-PD-L1 mAb-treated mice were significantly enhanced compared with levels in the group treated with control IgG. Slightly different L-4 production was observed after treatment with the blocking Abs, but with no significance.
  • iNKT cells producing IFN- ⁇ increased in number because they blocked PD-1/PD-L1 interaction with the anti-PD-1 mAb or anti-PD-L1 mAb in comparison with control cells.
  • no significant differences were found between populations of IL-4 + iNKT cells and control cells after mAb treatment.
  • ⁇ -GC-induced unresponsiveness of iNKT cells was detected as early as 3 days after primary stimulation and observed to persist until 7 ⁇ 30 days after ⁇ -GC stimulation.
  • C57BL/6 mice were injected with 2 ⁇ g of ⁇ -GC to induce iNKT cell anergy.
  • splenocytes were isolated from the mice and 5 ⁇ 10 5 cells were incubated for 3 days with 100 ng/ml of ⁇ -GC in the presence of 50 ⁇ g/ml of the control rat IgG, the anti-PD-1 mAb, the anti-PD-L1 mAb, or the anti-PD-L2 mAb.
  • mice splenocytes isolated from the mice were incubated for 3 days with 10 ng/ml of ⁇ -GC without mAb, and these were represented by ‘activation’. The supernatants were then assayed for IFN- ⁇ and IL-4 levels by ELISA [*:p ⁇ 0.05 and **:p ⁇ 0.01 (vs. control rat IgG)].
  • 200 ⁇ g of the control rat IgG, the anti-PD-1 antibody or the anti-PD-L1 antibody was intraperitoneally injected into C57BL/6 mice 24 hrs before treatment with 2 ⁇ g of ⁇ -GC. 14 days later, 2 ⁇ g of ⁇ -GC was injected again, followed by the preparation of sera 2 and 12 hrs after the re-injection for ELISA assay of IL-4 and IFN- ⁇ levels, respectively.
  • Splenocytes (5 ⁇ 10 6 cells), prepared two and twelve hours after the second injection of ⁇ -GC, were incubated with Golgi plug for 2 hrs to accumulate cytokines. Intracellular cytokine staining was performed on the splenocytes prepared 2 hrs after the secondary GC treatment, using BD Cytofix/Cytoperm Plus with Golgiplug kit according to the manufacturer's protocol (BD Biosciences). The splenocytes prepared 12 hrs later were used in an assay for CD69 expression in iNKT and NK cells.
  • iNKT cells were gated on B220 ⁇ TCR- ⁇ int ⁇ -GC/CD1d:1g + population and NK cells were gated on B220 ⁇ TCR- ⁇ ⁇ NK1.1 high population.
  • IFN- ⁇ + or IL-4 + iNKT cells were analyzed by flow cytometry.
  • CD69 expression on iNKT and NK cells was analyzed by flow cytometry.
  • IFN- ⁇ and L-4 levels in blood are graphed in FIG. 6 and FACS results of IFN- ⁇ + or L-4 + iNKT cells and flow histograms of CD69 expression on iNKT and NK cells are given in FIGS. 7 and 8 , respectively.
  • mice pretreated with ⁇ -GC produced significantly low IFN- ⁇ and L-4 upon secondary ⁇ -GC injection.
  • mice treated with the anti-PD-1 antibody during the primary ⁇ -GC injection produced remarkably higher IFN- ⁇ and L-4 upon the secondary ⁇ -GC injection than did the control IgG-treated group.
  • CD69 expression upon secondary ⁇ -GC injection was increased on iNKT and NK cells in a similar manner by the treatment with anti-PD-1 mAbs, as shown in FIG. 8 .
  • PD-1/PD-L1 interaction is essential for the induction of iNKT cell anergy in vivo.
  • Skin tumor cells (B16F10, ATCC) were cultured in DMEM media supplemented with 10% FBS and 1% penicillin/streptomycin.
  • C56BL/6 mice were intraperitoneally injected with 200 g of the control rat IgG, the anti-PD-1 mAb orthe anti-PD-L1 mAb 24 hrs before i.v. inoculation with 2 ⁇ 10 5 tumor cells.
  • the mice On Days zero, 4 and 8, the mice were treated with 500 ng of ⁇ -GC plus 200 ⁇ g of the control rat IgG, the anti-PD-1 mAb or the anti-PD-L1 mAb.
  • the lungs were weighed and weight differences between metastatic and normal lungs are graphed in FIG. 9 .
  • the B16F10 metastatic lungs from the group treated with ⁇ -GC plus the anti-PD-1 mAb were significantly reduced in weight compared with those from the group treated with ⁇ -GC plus the control rat IgG.
  • mice were intraperitoneally injected with 200 ⁇ g of the control rat IgG, the anti-PD-1 antibody or the anti-PD-L1 antibody 24 hrs after which injection with 2 ⁇ g of ⁇ -GC induced iNKT cell anergy. 7 days later, 5 ⁇ 10 5 B16F10 tumor cells were i.v. injected.
  • the mice were treated with 500 ng of ⁇ -GC to induce anti-tumor activity of iNKT cells. 14 days later, the lungs were excised and the nodules formed by cancer metastasis were counted (A). The isolated lungs were observed under an optical microscope (B). The results are given in FIG. 10 .
  • the data obtained above indicates that the blockage of PD-1/PD-L1 interaction by treatment with the anti-PD-1 antibody or the anti-PD-L1 antibody during the induction phase of iNKT cell anergy leads to the persistence of the anti-tumor effects of iNKT cells.
  • Formulation examples are given to illustrate dosage preparations containing the anticancer agent of the present invention.
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g Lactose 1.5 g Talc 0.5 g
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g Lactose 7.9 g Crystalline cellulose 1.5 g Magnesium stearate 0.5 g
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g Corn starch 5 g Carboxycellulose 4.9 g
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g Sterile water for injection proper quantity pH Adjuster proper quantity
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g Isomerized sugar 10 g Mannitol 5 g Purified water proper quantity
  • the anti-PD-1 or anti-PD-L1 antibody blocks the PD-1/PD-L1-mediated signaling pathway not only to prevent the iNKT cell ligand-induced iNKT cell anergy, but also to reverse the unresponsiveness of already anergic iNKT cells to produce cytokines.
  • the anti-PD1 or anti-PD-L1 antibody ensures the potent anti-tumor activity of iNKT cells as demonstrated by a significant reduction in the number of metastatic nodules in B16F10 melanoma metastasis models in vivo.
  • the anticancer agent comprising an anti-PD-1 or anti-PD-L1 antibody in accordance with the present invention can be very useful in the treatment of cancer, particularly metastatic cancer.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Provided is an anticancer agent which comprises an anti-PD-1 antibody or an anti-PD-L1 antibody as an active ingredient, functioning to reverse the unresponsiveness of iNKT cells in which anergy has been induced by administration with an iNKT cell ligand. The anti-PD-1 or anti-PD-L1 antibody blocks the PD-1/PD-L1-mediated signaling pathway not only to prevent the iNKT cell ligand-induced iNKT cell anergy, but also to reverse the unresponsiveness of already anergic iNKT cells to produce cytokines. In addition, the anti-PD1 or anti-PD-L1 antibody ensures the potent anti-tumor activity of iNKT cells as demonstrated by a significant reduction in the number of metastatic nodules in B16F10 melanoma metastasis models in vivo. Collectively, the anticancer agent can be very useful in the treatment of cancer, particularly metastatic cancer.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims priority from Korean Patent Application No.10-2008-0097236, filed Oct. 2, 2008, the entire disclosure of which is incorporated herein by reference in its entirety.
  • TECHNICAL FIELD
  • The present invention relates, in general, to an anticancer agent and, more particularly, to an anticancer agent comprising an anti-PD-1 antibody or an anti-PD-L1 antibody as an active ingredient, functioning to restore the responsiveness of iNKT cells in which anergy has been induced by administration with an iNKT cell ligand.
  • BACKGROUND OF THE INVENTION
  • Natural killer T (NKT) cells, co-expressing a T cell receptor and NK cell markers, are essential for several aspects of immunity, such as immunomodulation and immunuopotentiation, in various immune diseases including autoimmune diseases, infectious diseases, cancer, etc. NKT cells exist at high levels in the thymus, the liver, and the bone marrow, but at low levels in the spleen, lymph nodes and blood.
  • Unlike conventional T cells that recognize small peptide antigens presented by major histocompatibility complex MHC class 1 or MHC class 2, NKT cells recognize glycolipid antigens presented by CD1d, a MHC class 1-like molecule. A major subset of NKT cells, called type 1 NKT cells or invariant natural T (iNKT) cells, express an invariant natural T cell receptor (TCR) composed of Vα14-Jα18 chains in mice (Vα24-Jα18 in humans). Upon TCR stimulation with a ligand, such as α-galactosylceramide (α-GC), iNKT cells rapidly produce a wide range of cytokines including IL-4, IFN-γ, L-12, and GM-CSF. This rapid and potent response to a ligand enables iNKT cells to enhance or regulate the activity of various immune cells in innate and acquired immunity. Found in diverse diseases and promoting tumor rejection or regulating autoimmune disorders, these immunomodulatory roles of iNKT cells are studied for use in immunotherapy treatments for cancer and autoimmune diseases.
  • However, iNKT cells tend to greatly decrease in responsiveness following repeated stimulation after a first stimulation with their ligands via the T cell receptor. For instance, iNKT cells that have been stimulated in vivo with a-GC have reduced proliferation and cytokine production upon secondary stimulation with the same ligand. This iNKT cell anergy is a major obstacle in immunotherapeutic trials targeting iNKT cells.
  • Conventional T cells are known to become anergic when they receive a TCR signal with insufficient co-stimulatory signals. Co-stimulatory molecules such as CD28, CD40L and ICOS are known to be involved in the development and activation of iNKT cells. Recently, it has been reported that 4-1BB contributes to promote the activation of iNKT cells as a co-stimulatory molecule and can affect iNKT cell-mediated allergic lung inflammation (Kim, D. H., W. S. Chang, Y. S. Lee, K. A. Lee, Y. K. Kim, B. S. Kwon, and C. Y. Kang. 2008. 4-1BB engagement co-stimulates NKT cell activation and exacerbates NKT cell ligand-induced airway hyperresponsiveness and inflammation. J Immunol 180:2062-2068.). On the other hand, it has recently been suggested that coinhibitory molecules, such as PD-1, B7H3, and B7H4, may actively anergize or tolerize T cells by delivering inhibitory signals into TCR-stimulated T cells. In a lymphocytic choriomeningitis virus (LCMV) infected model, CD8 T cells are tolerized by LCMV epitope-presenting dendritic cells. However, the blockade of the PD-1 signal can reverse the anergic phenotype of CD8 T cells. It has also been reported that the inhibition of PD-1/PD-L1 restores the function of exhausted CD8 T cells in a chronic infection model.
  • Programmed death-1 (PD-1) is a 55 KDa type 1 transmebrane protein of the immunoglobulin superfamily, and is known as a co-inhibitory molecule on T cells. That is, PD-1 is a member of the co-inhibitory molecules of the CD28 family (e.g., CD28, CTLA4, ICOS and BTLA) expressed on activated B cells, T cells and bone marrow cells. Two ligands for PD-1, PD-L1 and PD-L2, have been identified thus far. The interaction of PD-1 with the PD ligands can transduce inhibitory or co-stimulatory signals into the T cells. In conventional T cells, PD-1 is not expressed on naive T cells, but is inducibly expressed after T cell activation. As for PD-L1, it is expressed to some degree on naive T cells and its level is increased on activated T cells. PD-L1 is found at high levels in various human cancers and interacts with PD-1 to transduce inhibitory or co-stimulatory signals from entering into the T cells. For example, the interaction between PD-1 and PD-L1 induces a decrease in the level of tumor invasive lymphocytes and in T cell receptor-mediated proliferation and causes the immune evasion of tumor cells. In PD-1-deficient animals, PD-1 develops various autoimmune phenotypes, such as autoimmune cardiac infarction and lupus-like syndromes with arthritis and nephritis, and plays an important role in the development of autoimmune encephalomyelitis, systemic lupus erythematosus, graft-versus-host disease (GVHD), type 1 diabetes and rheumatic arthritis. Aged PD-1-deficient mice develop autoimmune diseases, indicating that PD-1 plays a critical role in the regulation of autoimmunity and immune tolerance. In particular, PD-1 signals are essential for inducing T cell exhaustion during chronic infection.
  • With the ability thereof to stimulate the T cell receptor to rapidly produce various cytokines of iNKT cells, the NKT cell ligand α-GC has conventionally been used as an anticancer agent. However, since repeated stimulation of iNKT cells with α-GC induces anergy leading to a great decrease in responsiveness, it cannot achieve effective anticancer effects. Therefore, there is a pressing need for an anticancer agent that can restore the responsiveness of iNKT cells even in the state of anergy caused by stimulation with iNKT cell ligands.
  • Leading to the present invention, intensive and thorough research into an anticancer agent taking advantage of the responsiveness of iNKT cells, conducted by the present inventors, resulted in the finding that PD-1 expressed on iNKT cells is upregulated after stimulation and that blocking of the PD-1/PD-L1 signaling pathway by an anti-PD-1 or anti-PD-L1 antibody allows iNKT cells under an iNKT cell ligand-induced anergy condition to recover their responsiveness, such as the production of cytokines. Also, the anti-PD1 or anti-PD-L1 antibody was found to induce potent anti-tumor activity of iNKT cells as demonstrated by a significant reduction in the number of metastatic nodules in B16F10 melanoma metastasis models in vivo.
  • SUMMARY OF THE INVENTION
  • It is therefore an object of the present invention to provide an anticancer agent which is based on the activity of iNKT cells, featuring the recovery of iNKT cell responsiveness.
  • It is another object of the present invention to provide a method for the treatment of cancer using the same.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The above and other objects, features and advantages of the present invention will be more clearly understood from the following detailed description taken in conjunction with the accompanying drawings, in which:
  • FIG. 1 is of flow histograms showing the expression levels of PD-1, PD-L1 and PD-L2 on iNKT cells of the splenocytes isolated from α-GC-treated C57BL/6 mice at indicated time points (0 hr, 6 hrs, 72 hrs, 7 days, 1 month and 2 months) after treatment.
  • FIG. 2 is of histograms showing IFN-γ and IL-4 levels in the supernatants obtained after incubating splenocytes isolated from naïve C57BL/6 mice with α-GC in the presence of control rat IgG, anti-PD-1 monoclonal antibody, anti-PD-L1 monoclonal antibody or anti-PD-L2 monoclonal antibody as analyzed by ELISA (in vitro).
  • FIG. 3 is of plots showing IFN-γ and IL-4 levels in the sera obtained from the C57BL/6 mice, which were intraperitoneally injected with control rat IgG, anti-PD-1 monoclonal antibody, anti-PD-L1 monoclonal antibody or anti-PD-L2 monoclonal antibody 24 hrs before α-GC treatment, at 0, 2, 6, 12, 24, 48 and 72 hrs after the α-GC treatment as analyzed by ELISA (in vivo).
  • FIG. 4 shows FACS results of IFN-γ+ or IL-4 + iNKT cells on which intracellular cytokine staining was performed to examine if the increased production of IFN-γ and IL-4 in the sera of FIG. 3 comes from iNKT cells.
  • FIG. 5 is of histograms showing IFN-γ and IL-4 levels in the supernatants obtained after the splenocytes of C57BL/6 mice, in which iNKT cell anergy was induced by α-GC treatment 7 days and one month before splenocyte isolation therefrom, were incubated with α-GC in the presence of control rat IgG, anti-PD-1 monoclonal antibody, anti-PD-L1 monoclonal antibody or anti-PD-L2 monoclonal antibody, as analyzed by ELISA (in vitro).
  • FIG. 6 is of histograms showing IFN-γ and L-4 levels in sera from C57BL/6 mice, as analyzed by ELISA. The mice were intraperitoneally injected with control rat IgG, anti-PD-1 monoclonal antibody, anti-PD-L1 monoclonal antibody or anti-PD-L2 monoclonal antibody, followed by double injection with α-GC 24 hrs and 14 days later. The sera were obtained 2 and 12 hrs after the secondary injection of α-GC (in vivo).
  • FIG. 7 shows FACS results of IFN-γ+ or IL-4+ iNKT cells after intracellular cytokine staining was performed on the splenocytes isolated 2 hrs after the secondary injection (in vivo).
  • FIG. 8 shows CD69 expression levels on iNKT and NK cells as measured by flow cytometry using the splenocytes isolated 12 hrs after the secondary injection (in vivo).
  • FIG. 9 is a histogram showing weights of normal and metastatic lungs of C57BL/6 mice. The mice were intraperitoneally injected with control rat IgG, anti-PD-1 monoclonal antibody or anti-PD-L1 antibody 24 hrs before intravenous injection with skin tumor cells. On Days 0, 4 and 8, the mice were co-administered with α-GC and the control rat IgG, the anti-PD-1 antibody or the anti-PD-L1 antibody. The lungs were excised 14 days after the co-administration.
  • FIG. 10 shows effects of the blockage of PD-1/PD-L1 interaction on the antitumor activity of iNKT cells in terms of the number of metastatic pulmonary nodules (A) and optical microscopic views (B). C57BL/6 mice were intraperitoneally injected with control rat IgG, anti-PD-1 antibody or anti-PD-L1 antibody 24 hrs before α-GC treatment. 7 days later, the mice were intravenously injected with skin tumor cells and re-injected with α-GC on Days 0, 4 and 8. 14 days later, the lungs were excised.
  • DETAILED DESCRIPTION OF THE INVENTION
  • In accordance with an aspect thereof, the present invention provides an anticancer agent, comprising an iNKT cell ligand as a first anticancer factor and an anti-PD-1 antibody or anti-PD-L1 antibody as a second anticancer factor, having a function of reversing unresponsiveness of iNKT cells in which anergy is induced by administration with the iNKT cell ligand.
  • In accordance with another aspect thereof, the present invention provides a method for reversing the unresponsiveness of iNKT cells with anergy induced therein by iNKT cell ligand treatment, comprising treating the anergic iNKT cells with an anti-PD-1 antibody or an anti-PD-L1 antibody.
  • The iNKT cell ligand may be selected from a group consisting of alpha-galactosyl ceramide, alpha-glucuronosyl ceramide, phosphatidylinositoltetramannoside, isoglobotrihexosylceramide, ganglioside GD3, phsphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, sulfatide, beta-galactosylceramide, lipophosphoglycan, glycoinositol phospholipid, alpha-galactosylceramide analogs including beta-anomer galactoceramide and alpha-anomer galactosylceramide, and bacterial lipid antigens.
  • The anti-PD-1 antibody or the anti-PD-L1 antibody may be a monoclonal antibody or a polyclonal antibody.
  • The anti-PD-1 antibody or anti-PD-L1 antibody according to the present invention can block the signaling of PD-1 or PD-L1 to prevent the iNKT cell ligand-induced anergy of iNKT cells and can provide cytokine secretion ability for even anergic iNKT cells to restore their responsiveness. In addition, the anti-PD-1 antibody or anti-PD-L1 antibody of the present invention inhibits the anergy induction of iNKT cells to significantly decrease the number of pulmonary nodules in a lung metastasis model of B16F10 melanoma, thus effectively eliciting anticancer immune responses of iNKT cells and showing anti-tumor effects of iNKT cells against cancer metastasis. Therefore, an anticancer agent comprising the anti-PD-1 antibody or anti-PD-L 1 antibody of the present invention can be very useful in the treatment of cancer, particularly, metastatic cancer.
  • The cancer to which the anticancer agent according to the present invention is therapeutically applicable may be gynecologic tumor, endocrine gland cancer, CNS (central nervous system) tumor or ureter cancer. Concrete examples of the cancer include lung cancer, stomach cancer, liver cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, melanoma, uterine cancer, ovarian cancer, rectal cancer, colorectal cancer, colon cancer, breast cancer, uterine sarcoma, Fallopian tube carcinoma, endometrial carcinoma, uterine cervical carcinoma, vaginal carcinoma, vulva carcinoma, esophageal cancer, larynx cancer, small intestine cancer, thyroid cancer, parathyroid cancer, soft tissue sarcoma, uterine cancer, penis cancer, prostate cancer, chronic or acute leukemia, pediatric solid tumor, differentiated lymphoma, bladder cancer, kidney cancer, renal cell carcinoma, renal pelvic carcinoma, primary CNS lymphoma, spinal cord tumor, brainstem glioma, and pituitary adenoma.
  • The anticancer agent of the present invention may be formulated into a pharmaceutical composition with at least one conventional anticancer ingredient.
  • In addition to the active ingredients, the pharmaceutical composition may further comprise a pharmaceutically acceptable carrier. Examples of the pharmaceutically acceptable carrier include saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol, etc. Optionally, conventional additives, such as antioxidants, buffers, bacteriostatic agents, etc., may be added to the composition. For the preparation of dosage forms including injections, such as aqueous solutions, suspensions and emulsions, pills, capsules, granules and tablets, the active ingredients may be admixed with a diluent, a dispersant, a surfactant, a binder and/or a lubricant. Reference may be made to literature (Remington's Pharmaceutical Science (recent edition), Mack Publishing Company, Easton Pa.) upon the formulation of the pharmaceutical composition into suitable dosage forms.
  • The composition of the present invention may be administered via oral routes or parenteral routes (e.g., intravenous, subcutaneous, intraperitoneal, topical, etc.). The effective dosage of the anticancer agent in accordance with the present invention depends on various factors, including the patient's weight, age, gender, state of health, diet, the time of administration, route of administration, excretion rate, severity of diseases, etc. In general, it may be administered in a single dose, and preferably in multiple doses per day at a daily dose ranging from 0.01 to 1000 mg/day, and preferably from 0.1 to 100 mg/kg of the anti-PD-1 antibody or anti-PD-L1 antibody.
  • For the effective prophylaxis and treatment of cancer, the composition according to the present invention may be used alone or in combination with surgical operation, hormonal therapy, chemotherapy, and/or biological response controllers.
  • A better understanding of the present invention may be obtained through the following examples which are set forth to illustrate, but are not to be construed as limiting the present invention.
  • EXAMPLES Example 1 Expression of PD-1 and PD Ligands in iNKT Cells
  • 1. Experimental Animals
  • Six- to eight-week-old female C56BL/6 mice were purchased from Orient Bio. All mice were bred and maintained in specific pathogen-free conditions.
  • 2. Reagents and Antibodies
  • α-GC was dissolved in PBS containing 0.5% Tween 20. Hybridoma clones producing antibodies to mouse PD-1 (RMP1-14, rat IgG2a), PD-L1 (MIH-5, rat IgG2a), and PD-L2 (Ty25, rat IgG2a) were produced according to methods well known in the art [Yamazaki, T., H. Akiba, H. Iwai, H. Matsuda, M. Aoki, Y. Tanno, T. Shin, H. Tsuchiya, D. M. Pardoll, K. Okumura, M. Azuma, and H. Yagita. 2002. Expression of programmed death 1 ligands by murine T cells and APC. J Immunol 169:5538-5545.; Tsushima, F., H. Iwai, N. Otsuki, M. Abe, S. Hirose, T. Yamazaki, H. Akiba, H. Yagita, Y. Takahashi, K. Omura, K. Okumura, and M. Azuma. 2003. Preferential contribution of B7-H1 to programmed death-1-mediated regulation of hapten-specific allergic inflammatory responses. Eur J Immunol 33:2773-2782.; Yamazaki, T., H. Akiba, A. Koyanagi, M. Azuma, H. Yagita, and K. Okumura. 2005. Blockade of B7-H1 on macrophages suppresses CD4+ T cell proliferation by augmenting IFN-gamma-induced nitric oxide production. J Immunol 175:1586-1592.]. All clones were cultured in RPMI 1640 (Gibco) with 10% FBS (Gibco) and 1% penicillin/streptomycin (BioWhittaker). All antibodies were prepared from the ascites of nude mice using caprylic acid purification. Control rat IgG was also prepared by caprylic acid purification from sera of naïve rats.
  • 3. Expression of PD-1 and PD Ligands on iNKT Cells After α-GC Administration
  • Splenocytes were isolated from C57BL/6 mice administered with 2 μg of α-GC at different time points (0 hr, 6 hrs, 72 hrs, 7 days, one month and two months) after the administration. The cells were stained with a PE-conjugated anti-PD-1 monoclonal antibody, a PE-conjugated anti-PD-L1 monoclonal antibody, a PE-conjugated anti-PD-L2 monoclonal antibody and a PE-conjugated isotype control monoclonal antibody, respectively. iNKT cells were gated on B220 TCR-βintα-GC/CD1d:Ig+ population. PD-1, PD-L1 and PD-L2 expression (open histograms) was analyzed by FACS. The results are given in FIG. 1. Shaded histograms indicate staining with isotype control niAbs.
  • As depicted in FIG. 1, PD-1 was constitutively expressed on iNKT cells at a low level and its expression was upregulated after α-GC stimulation, followed by the persistence of the upregulated level for two months. On the other hand, PD-L1 expression was temporarily increased on α-GC-stimulated iNKT cells but declined toward naïve state levels within 72 hrs. As for PD-L2, its expression was not found on iNKT cells regardless of their activation status.
  • Example 2 Effects of PD-1, PD-L1 and PD-L2 on iNKT Cell Activation
  • The following in vitro and in vivo experiments were conducted to examine the effects of PD-1, PD-L1 and PD-L2 on iNKT cell activation.
  • 1. iNKT Cell Activation In Vitro
  • After being prepared from naëve C57BL/6 mice, 5×105 spelenocytes were incubated for 3 days with 100 ng/ml of α-GC in the presence of 50 μg/ml of the control rat IgG, the anti-PD-1 mAb, the anti-PD-L1 mAb, or the anti-PD-L2 mAb. Then, the supernatants were obtained and assayed for IFN-γ and IL-4 levels by ELISA.
  • The results are given in FIG. 2.
  • Compared with control IgG treatment, as is apparent from FIG. 2, anti-PD-1 mAb significantly increased the production of IFN-γ and, to a lesser extent, IL-4. Anti-PD-L1 mAb also induced greater production of IFN-γ, but anti-PD-L2 mAb did not.
  • 2. iNKT Cell Activation in Vivo
  • C57BL/6 mice was injected with 200 μg of the control rat IgG, the anti-PD-1 mAb, the anti-PD-L1 mAb or the anti-PD-L2 mAb 24 hrs before treatment with 2 μg of α-GC. Sera were obtained at 0, 2, 6, 12, 24, 48 and 72 hrs after the treatment, followed by ELISA analysis for IFN-γ and IL-4 levels.
  • In order to determine whether the increased production of IFN-γ was distinctively attributed to iNKT cells, intracellular cytokine staining was followed by flow cytometry analysis. In this regard, splenocytes were isolated 2 hrs after α-GC treatment and 5×106 cells were incubated with Golgi plug for 2 hrs to accumulate cytokines. Intracellular cytokine staining was performed using BD Cytofix/Cytoperm Plus with Golgiplug kit according to the manufacturer's protocol (BD Biosciences). iNKT cells were gated on B220TCR-βint α-GC/CD1d:Ig+ population and IFN-γ+ or IL-4+ iNKT cells were analyzed by flow cytometry.
  • IFN-γ and IL-4 levels in blood are depicted in FIG. 3 and flow histograms of IFN-γ+ and IL-4+ iNKT cells are given in FIG. 4.
  • As shown in FIG. 3, the blood IFN-γ and IL-4 levels were observed to peak, respectively, within 12 hrs and 2 hrs after α-GC treatment, indicating iNKT cell activation. IFN-γ levels in the sera of anti-PD-1 or anti-PD-L1 mAb-treated mice were significantly enhanced compared with levels in the group treated with control IgG. Slightly different L-4 production was observed after treatment with the blocking Abs, but with no significance.
  • As shown in FIG. 4, iNKT cells producing IFN-γ increased in number because they blocked PD-1/PD-L1 interaction with the anti-PD-1 mAb or anti-PD-L1 mAb in comparison with control cells. On the other hand, no significant differences were found between populations of IL-4+ iNKT cells and control cells after mAb treatment. These data indicate that the blockage of PD-1/PD-L1 interaction allows the delivery of the co-inhibitory signal during iNKT activation, resulting in increased IFN-γ secretion from iNKT cells.
  • Example 3 Effects of Blockage of PD-1/PD-L1 Interaction on Responsiveness of Anergic iNKT Cells
  • The following in vitro and in vivo experiments were performed in order to examine whether the blockage of PD-1/PD-L 1 interaction reverses iNKT cell anergy.
  • 1. Recovery of Responsiveness of Anergic iNKT Cells in Vitro
  • The α-GC-induced unresponsiveness of iNKT cells was detected as early as 3 days after primary stimulation and observed to persist until 7˜30 days after α-GC stimulation. Thus, C57BL/6 mice were injected with 2 μg of α-GC to induce iNKT cell anergy. One week and one month later, splenocytes were isolated from the mice and 5×105 cells were incubated for 3 days with 100 ng/ml of α-GC in the presence of 50 μg/ml of the control rat IgG, the anti-PD-1 mAb, the anti-PD-L1 mAb, or the anti-PD-L2 mAb. Also, splenocytes isolated from the mice were incubated for 3 days with 10 ng/ml of α-GC without mAb, and these were represented by ‘activation’. The supernatants were then assayed for IFN-γ and IL-4 levels by ELISA [*:p<0.05 and **:p<0.01 (vs. control rat IgG)].
  • The results are depicted in FIG. 5.
  • As seen in the graphs of FIG. 5, when restimulated with α-GC in the presence of the control IgG, the splenocytes of the mice in which iNKT cell anergy was induced by pre-treatment with α-GC 7 days before the restimulation were found to greatly decrease in the production of IFN-γ and IL-4. In contrast, the restimulation of iNKT cell anergy-induced splenocytes with α-GC in the presence of the anti-PD-1 antibody or anti-PD-L1 antibody resulted in a great increase in the production of IFN-γ and L-4 (A). This upregulation was observed to persist for one to two months after α-GC treatment (B). Therefore, the blockage of PD-1/PD-L1 interaction during restimulation with α-GC reverses the established anergic phenotype of iNKT cells.
  • 2. Recovery of Responsiveness of Anergic iNKT Cells in Vivo
  • 200 μg of the control rat IgG, the anti-PD-1 antibody or the anti-PD-L1 antibody was intraperitoneally injected into C57BL/6 mice 24 hrs before treatment with 2 μg of α-GC. 14 days later, 2 μg of α-GC was injected again, followed by the preparation of sera 2 and 12 hrs after the re-injection for ELISA assay of IL-4 and IFN-γ levels, respectively.
  • Splenocytes (5×106 cells), prepared two and twelve hours after the second injection of α-GC, were incubated with Golgi plug for 2 hrs to accumulate cytokines. Intracellular cytokine staining was performed on the splenocytes prepared 2 hrs after the secondary GC treatment, using BD Cytofix/Cytoperm Plus with Golgiplug kit according to the manufacturer's protocol (BD Biosciences). The splenocytes prepared 12 hrs later were used in an assay for CD69 expression in iNKT and NK cells. iNKT cells were gated on B220TCR-βintα-GC/CD1d:1g+ population and NK cells were gated on B220 TCR-βNK1.1high population. IFN-γ+ or IL-4+ iNKT cells were analyzed by flow cytometry. Also, CD69 expression on iNKT and NK cells was analyzed by flow cytometry.
  • IFN-γ and L-4 levels in blood are graphed in FIG. 6 and FACS results of IFN-γ+ or L-4+ iNKT cells and flow histograms of CD69 expression on iNKT and NK cells are given in FIGS. 7 and 8, respectively.
  • Like mice treated with control rat IgG, as seen in FIG. 6, mice pretreated with α-GC produced significantly low IFN-γ and L-4 upon secondary α-GC injection. In contrast, mice treated with the anti-PD-1 antibody during the primary α-GC injection produced remarkably higher IFN-γ and L-4 upon the secondary α-GC injection than did the control IgG-treated group.
  • As seen in FIG. 7, the increased cytokine production was attributed to iNKT cells.
  • It was also found that CD69 expression upon secondary α-GC injection was increased on iNKT and NK cells in a similar manner by the treatment with anti-PD-1 mAbs, as shown in FIG. 8. Thus, PD-1/PD-L1 interaction is essential for the induction of iNKT cell anergy in vivo.
  • Example 4 Effects of Anti-PD-1 mAb or Anti-PD-L1 mAb on Anti-Tumor Activity of Activated iNKT Cells
  • The following experiments were performed to investigate the effects of the anti-PD-1 antibody or the anti-PD-L1 antibody on the anticancer activity of activated iNKT cells in a B16F10 melanoma metastasis model.
  • 1. Weight of Lung
  • Skin tumor cells (B16F10, ATCC) were cultured in DMEM media supplemented with 10% FBS and 1% penicillin/streptomycin. C56BL/6 mice were intraperitoneally injected with 200 g of the control rat IgG, the anti-PD-1 mAb orthe anti-PD-L1 mAb 24 hrs before i.v. inoculation with 2×105 tumor cells. On Days zero, 4 and 8, the mice were treated with 500 ng of α-GC plus 200 μg of the control rat IgG, the anti-PD-1 mAb or the anti-PD-L1 mAb. On Day 14, the lungs were weighed and weight differences between metastatic and normal lungs are graphed in FIG. 9.
  • As depicted in FIG. 9, the B16F10 metastatic lungs from the group treated with α-GC plus the anti-PD-1 mAb were significantly reduced in weight compared with those from the group treated with α-GC plus the control rat IgG.
  • 2. Number of Pulmonary Nodules
  • Anergy-induced iNKT cells were assayed for the recovery of anti-tumor activity by the antibodies of the present invention. In this regard, naive C57BL/6 mice were intraperitoneally injected with 200 μg of the control rat IgG, the anti-PD-1 antibody or the anti-PD-L1 antibody 24 hrs after which injection with 2 μg of α-GC induced iNKT cell anergy. 7 days later, 5×105 B16F10 tumor cells were i.v. injected. On Day 0, 4 and 8, the mice were treated with 500 ng of α-GC to induce anti-tumor activity of iNKT cells. 14 days later, the lungs were excised and the nodules formed by cancer metastasis were counted (A). The isolated lungs were observed under an optical microscope (B). The results are given in FIG. 10.
  • As seen in FIG. 10, when induced by treatment with the control IgG and α-GC, iNKT cell anergy was unchanged with time to show a metastasis result comparable to that of the normal metastasis control group, indicating that α-GC treatment during tumor inoculation did not suppress tumor growth. In contrast, when the anergy induction of α-GC was prevented by treatment with the anti-PD-1 antibody or the anti-PD-L1, the numbers of tumor nodules in the B16F10 melanoma metastasis model were remarkably reduced compared with those upon treatment with the control IgG, demonstrating the superior anti-tumor activity of the antibodies of the present invention. Taken together, the data obtained above indicates that the blockage of PD-1/PD-L1 interaction by treatment with the anti-PD-1 antibody or the anti-PD-L1 antibody during the induction phase of iNKT cell anergy leads to the persistence of the anti-tumor effects of iNKT cells.
  • Formulation examples are given to illustrate dosage preparations containing the anticancer agent of the present invention.
  • Formulation Example 1 Preparation of Powder
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g
    Lactose 1.5 g
    Talc 0.5 g
  • These ingredients were mixed and loaded into an airtight sac to give a powder.
  • Formulation Example 2 Preparation of Tablet
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g
    Lactose 7.9 g
    Crystalline cellulose 1.5 g
    Magnesium stearate 0.5 g
  • These ingredients were mixed and directly compressed into a tablet.
  • Formulation Example 3 Preparation of Capsule
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g
    Corn starch   5 g
    Carboxycellulose 4.9 g
  • These ingredients were admixed together and the admixture was loaded into a conventional capsule using a suitable device.
  • Formulation Example 4 Preparation of Injection
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g
    Sterile water for injection proper quantity
    pH Adjuster proper quantity
  • Using a conventional method, these ingredients were put into an ampule (2 ml) to give an injection.
  • Formulation Example 5 Preparation of Liquid Medicine
  • Anti-PD-1 or Anti-PD-L1 Antibody 0.1 g
    Isomerized sugar  10 g
    Mannitol   5 g
    Purified water proper quantity
  • Each ingredient was dissolved in purified water and flavored with lemon before admixing together. Purified water was added to the admixture to form a final volume of 100 ml which was then loaded into a brown vial and sterilized.
  • As described hitherto, the anti-PD-1 or anti-PD-L1 antibody according to the present invention blocks the PD-1/PD-L1-mediated signaling pathway not only to prevent the iNKT cell ligand-induced iNKT cell anergy, but also to reverse the unresponsiveness of already anergic iNKT cells to produce cytokines. In addition, the anti-PD1 or anti-PD-L1 antibody ensures the potent anti-tumor activity of iNKT cells as demonstrated by a significant reduction in the number of metastatic nodules in B16F10 melanoma metastasis models in vivo. Collectively, the anticancer agent comprising an anti-PD-1 or anti-PD-L1 antibody in accordance with the present invention can be very useful in the treatment of cancer, particularly metastatic cancer.
  • Although the preferred embodiments of the present invention have been disclosed for illustrative purposes, those skilled in the art will appreciate that various modifications, additions and substitutions are possible, without departing from the scope and spirit of the invention as disclosed in the accompanying claims.

Claims (7)

1. An anticancer agent, comprising an iNKT cell ligand as a first anticancer factor and an anti-PD-1 antibody or anti-PD-L1 antibody as a second anticancer factor, having a function of reversing unresponsiveness of iNKT cells in which anergy is induced by administration with the iNKT cell ligand.
2. The anticancer agent as defined in claim 1, wherein the iNKT cell ligand is selected from a group consisting of alpha-galactosyl ceramide, alpha-glucuronosyl ceramide, phosphatidylinositoltetramannoside, isoglobotrihexosylceramide, ganglioside GD3, phsphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, sulfatide, beta-galactosylceramide, lipophosphoglycan, glycoinositol phospholipid, alpha-galactosylceramide analogs including beta-anomer galactoceramide and alpha-anomer galactosylceramide, and bacterial lipid antigens.
3. The anticancer agent as defined in claim 1, wherein the anti-PD-1 antibody or the anti-PD-L1 antibody is a monoclonal antibody or a polyclonal antibody.
4. The anticancer agent as defined in claim 1, wherein the cancer is selected from a group consisting of lung cancer, stomach cancer, liver cancer, bone cancer, pancreatic cancer, skin cancer, head and neck cancer, melanoma, uterine cancer, ovarian cancer, rectal cancer, colorectal cancer, colon cancer, breast cancer, uterine sarcoma, Fallopian tube carcinoma, endometrial carcinoma, uterine cervical carcinoma, vaginal carcinoma, vulva carcinoma, esophageal cancer, larynx cancer, small intestine cancer, thyroid cancer, parathyroid cancer, soft tissue sarcoma, uterine cancer, penis cancer, prostate cancer, chronic or acute leukemia, pediatric solid tumor, differentiated lymphoma, bladder cancer, kidney cancer, renal cell carcinoma, renal pelvic carcinoma, primary CNS lymphoma, spinal cord tumor, brainstem glioma, and pituitary adenoma.
5. A method for reversing the unresponsiveness of iNKT cells with anergy induced therein by iNKT cell ligand treatment, comprising treating the anergic iNKT cells with an anti-PD-1 antibody or an anti-PD-L1 antibody.
6. The method as defined in claim 5, wherein the iNKT cell ligand is selected from a group consisting of alpha-galactosyl ceramide, alpha-glucuronosyl ceramide, phosphatidylinositoltetramannoside, isoglobotrihexosylceramide, ganglioside GD3, phsphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, sulfatide, beta-galactosylceramide, lipophosphoglycan, glycoinositol phospholipid, alpha-galactosylceramide analogs including beta-anomer galactoceramide and alpha-anomer galactosylceramide, and bacterial lipid antigens.
7. The method as defined in claim 5, wherein the anti-PD-1 antibody or the anti-PD-L1 antibody is a monoclonal antibody or a polyclonal antibody.
US12/410,732 2008-10-02 2009-03-25 Anticancer agent comprising anti-pd-1 antibody or anti-pd-l1 antibody Abandoned US20100086550A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/398,915 US8617546B2 (en) 2008-10-02 2012-02-17 Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2008-0097236 2008-10-02
KR1020080097236A KR101050829B1 (en) 2008-10-02 2008-10-02 Anticancer agents comprising an anti-PD-1 antibody or an anti-PD-L1 antibody

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/398,915 Continuation US8617546B2 (en) 2008-10-02 2012-02-17 Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody

Publications (1)

Publication Number Publication Date
US20100086550A1 true US20100086550A1 (en) 2010-04-08

Family

ID=41171109

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/410,732 Abandoned US20100086550A1 (en) 2008-10-02 2009-03-25 Anticancer agent comprising anti-pd-1 antibody or anti-pd-l1 antibody
US13/398,915 Active US8617546B2 (en) 2008-10-02 2012-02-17 Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/398,915 Active US8617546B2 (en) 2008-10-02 2012-02-17 Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody

Country Status (4)

Country Link
US (2) US20100086550A1 (en)
EP (1) EP2172219B1 (en)
JP (1) JP5054057B2 (en)
KR (1) KR101050829B1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013043647A1 (en) * 2011-09-19 2013-03-28 The Johns Hopkins University Cancer immunotherapy
WO2016196173A1 (en) * 2015-05-29 2016-12-08 Merck Sharp & Dohme Corp. Combination of a pd-1 antagonist and cpg-c type oligonucleotide for treating cancer
US9624298B2 (en) 2011-11-28 2017-04-18 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US9815897B2 (en) 2013-05-02 2017-11-14 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US10280223B2 (en) 2014-07-09 2019-05-07 Nippon Zenyaku Kogyo Co., Ltd. Anti-canine PD-1 antibody or anti-canine PD-L1 antibody
US10416160B2 (en) 2014-07-25 2019-09-17 Riken Memory invariant NKT cell marker
CN111920948A (en) * 2020-09-25 2020-11-13 北京广未生物科技有限公司 Pharmaceutical composition comprising immune cells for treating cancer
RU2744880C1 (en) * 2014-02-04 2021-03-16 Инсайт Корпорейшн Combination of pd-1 antagonist and ido1 inhibitor for treating ancer
US11155624B2 (en) 2016-11-01 2021-10-26 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
US11407830B2 (en) 2017-01-09 2022-08-09 Tesaro, Inc. Methods of treating cancer with anti-PD-1 antibodies
US11685782B2 (en) 2017-10-23 2023-06-27 Children's Medical Center Corporation Methods of treating cancer using LSD1 inhibitors in combination with immunotherapy
WO2024137442A1 (en) 2022-12-21 2024-06-27 Gilead Sciences, Inc. Combination therapy for treating cancer

Families Citing this family (116)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX359551B (en) 2009-11-24 2018-10-02 Medimmune Ltd Targeted binding agents against b7-h1.
KR20200079568A (en) * 2012-05-31 2020-07-03 제넨테크, 인크. Methods of treating cancer using pd-l1 axis binding antagonists and vegf antagonists
MX367042B (en) * 2012-10-02 2019-08-02 Bristol Myers Squibb Co Combination of anti-kir antibodies and anti-pd-1 antibodies to treat cancer.
WO2014194293A1 (en) * 2013-05-30 2014-12-04 Amplimmune, Inc. Improved methods for the selection of patients for pd-1 or b7-h4 targeted therapies, and combination therapies thereof
CA2913490A1 (en) 2013-06-06 2014-12-11 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of cancer using pd-l1 isoforms
CN104250302B (en) * 2013-06-26 2017-11-14 上海君实生物医药科技股份有限公司 The anti-antibody of PD 1 and its application
AU2014320343B2 (en) * 2013-09-11 2020-05-28 Medimmune Limited Anti-B7-H1 antibodies for treating tumors
PL3044234T3 (en) 2013-09-13 2020-10-19 Beigene Switzerland Gmbh Anti-pd1 antibodies and their use as therapeutics and diagnostics
US10570204B2 (en) 2013-09-26 2020-02-25 The Medical College Of Wisconsin, Inc. Methods for treating hematologic cancers
CN104698059B (en) * 2013-12-04 2017-07-21 苏州中赢医疗科技有限公司 A kind of glioma tumor markers and its application
PE20160953A1 (en) 2013-12-12 2016-09-26 Shanghai hengrui pharmaceutical co ltd ANTIBODY PD-1, ANTIGEN BINDING FRAGMENT OF THIS AND MEDICAL USE OF THIS
RU2687209C1 (en) 2013-12-20 2019-05-07 Интервет Интернэшнл Б.В. Antibodies of dogs with modified ch2-ch3 sequences
TWI681969B (en) 2014-01-23 2020-01-11 美商再生元醫藥公司 Human antibodies to pd-1
TWI680138B (en) * 2014-01-23 2019-12-21 美商再生元醫藥公司 Human antibodies to pd-l1
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
JOP20200096A1 (en) 2014-01-31 2017-06-16 Children’S Medical Center Corp Antibody molecules to tim-3 and uses thereof
US9987258B2 (en) 2014-04-06 2018-06-05 H. Lee Moffitt Cancer Center And Research Institute, Inc. Histone deacetylase as a modulator of PDL1 expression and activity
EP3134085A1 (en) 2014-05-23 2017-03-01 Eisai R&D Management Co., Ltd. Combination therapies for the treatment of cancer
WO2016000619A1 (en) * 2014-07-03 2016-01-07 Beigene, Ltd. Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
JP6986965B2 (en) 2014-07-22 2021-12-22 アポロミクス インコーポレイテッド Anti-PD-1 antibody
CN118388646A (en) 2014-08-05 2024-07-26 中美冠科生物技术(太仓)有限公司 Anti-PD-L1 antibodies
CN107074949B (en) 2014-08-07 2021-06-15 索隆-基特林癌症研究协会 Anti-ceramide antibodies
US11344620B2 (en) 2014-09-13 2022-05-31 Novartis Ag Combination therapies
MA40035A (en) 2014-10-14 2016-04-21 Dana Farber Cancer Inst Inc PD-L1 ANTIBODY MOLECULES AND THEIR USES
CN105622753B (en) * 2014-11-04 2019-04-09 博生吉医药科技(苏州)有限公司 A kind of PD-1 monoclonal antibody and its application
JP7305300B2 (en) 2014-11-05 2023-07-10 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Combination immunotherapy
WO2016089610A1 (en) 2014-12-06 2016-06-09 H. Lee Moffitt Cancer Center And Research Institute, Inc. Bispecific antibody for cancer immunotherapy
CA2976377A1 (en) * 2015-02-12 2016-08-18 The Johns Hopkins University Inhibition of yap for breaking tumor immune tolerance
BR112017019559B1 (en) 2015-03-13 2020-08-04 Cytomx Therapeutics, Inc ANTI-PDL1 ANTIBODIES, ACTIVABLE ANTI-PDL1 ANTIBODIES, AND METHODS OF USE OF THESE
US11071775B2 (en) 2015-04-03 2021-07-27 H. Lee Moffitt Cancer Center And Research Institute, Inc. Combination immunotherapy for treating cancer
US10167334B2 (en) 2015-04-03 2019-01-01 Xoma Technology Ltd. Treatment of cancer using anti-TGF-BETA and PD-1 antibodies
US10995140B2 (en) 2015-06-05 2021-05-04 H. Lee Moffitt Cancer Center And Research Institute, Inc. GM-CSF/CD40L vaccine and checkpoint inhibitor combination therapy
MA42447A (en) 2015-07-13 2018-05-23 Cytomx Therapeutics Inc ANTI-PD-1 ANTIBODIES, ACTIVABLE ANTI-PD-1 ANTIBODIES, AND METHODS OF USE THEREOF
AR105654A1 (en) 2015-08-24 2017-10-25 Lilly Co Eli ANTIBODIES PD-L1 (LINKING 1 OF PROGRAMMED CELL DEATH)
US10323091B2 (en) 2015-09-01 2019-06-18 Agenus Inc. Anti-PD-1 antibodies and methods of use thereof
US10947598B2 (en) 2015-09-29 2021-03-16 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods for determining the metabolic status of lymphomas
JP6654694B2 (en) 2015-10-02 2020-02-26 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Anti-PD1 antibody and method of use
BR112018008867A8 (en) 2015-11-03 2019-02-26 Janssen Biotech Inc antibodies that specifically bind to pd-1 and their uses
US10815304B2 (en) 2015-11-17 2020-10-27 Suzhou Suncadia Biopharmaceuticals Co., Ltd. PD-L1 antibody, antigen-binding fragment thereof and medical application thereof
RS64588B1 (en) 2015-12-22 2023-10-31 Regeneron Pharma Combination of anti-pd-1 antibodies and bispecific anti-cd20/anti-cd3 antibodies to treat cancer
EP3400443B1 (en) 2016-01-04 2020-09-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of pd-1 and tim-3 as a measure for cd8+ cells in predicting and treating renal cell carcinoma
ES2801873T3 (en) * 2016-03-04 2021-01-14 Sichuan Kelun Biotech Biopharmaceutical Co Ltd PDL-1 antibody, its pharmaceutical composition and its uses
TW202408578A (en) 2016-05-13 2024-03-01 美商再生元醫藥公司 Methods of treating skin cancer by administering a pd-1 inhibitor
BR112018075737A2 (en) * 2016-06-13 2019-03-26 I-Mab antibody, composition, isolated cell, and methods for treating cancer or infection in a patient and for detecting pd-11 expression in a sample.
EP3481393B1 (en) 2016-07-05 2021-04-14 Beigene, Ltd. Combination of a pd-1 antagonist and a raf inhibitor for treating cancer
EP4353747A3 (en) 2016-08-19 2024-06-26 BeiGene Switzerland GmbH Combination of zanubrutinib with an anti-cd20 or an anti-pd-1 antibody for use in treating cancer
US10323248B2 (en) 2016-09-01 2019-06-18 Chimera Bioengineering, Inc. Gold optimized CAR T-cells
CN110831629B (en) 2016-09-14 2022-08-23 Abivax公司 Combinations comprising ABX196 for the treatment of cancer
US11524988B2 (en) 2016-09-19 2022-12-13 H. Lee Moffitt Cancer Center And Research Institute, Inc. Artificial antigen presenting cells for genetic engineering of immune cells
US10342785B2 (en) 2016-11-04 2019-07-09 Askat Inc. Use of EP4 receptor antagonists for the treatment of NASH-associated liver cancer
KR102603681B1 (en) 2016-12-07 2023-11-17 아게누스 인코포레이티드 Antibodies and methods of using them
TWI774726B (en) 2017-01-25 2022-08-21 英屬開曼群島商百濟神州有限公司 Crystalline forms of (s)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
CN106699891B (en) * 2017-01-25 2019-04-09 北京天广实生物技术股份有限公司 A kind of anti-PD-L1 antibody, its medical composition and its use
WO2018156434A1 (en) 2017-02-22 2018-08-30 H. Lee Moffitt Cancer Center And Research Institute Inc. Tim3-binding chimeric antigen receptors
US12077590B2 (en) 2017-02-22 2024-09-03 H. Lee Moffitt Cancer Center And Research Institute, Inc. Bispecific antibody for cancer immunotherapy
EP3600427A1 (en) 2017-03-24 2020-02-05 INSERM - Institut National de la Santé et de la Recherche Médicale Methods and compositions for treating melanoma
US11603407B2 (en) 2017-04-06 2023-03-14 Regeneron Pharmaceuticals, Inc. Stable antibody formulation
CA3060554A1 (en) 2017-04-18 2018-10-25 Tempest Therapeutics, Inc. Bicyclic compounds and their use in the treatment of cancer
BR112019025188A2 (en) 2017-06-01 2020-06-23 Cytomx Therapeutics, Inc. ACTIVABLE ANTI-PDL1 ANTIBODIES AND METHODS OF USE OF THE SAME
EP3645569A4 (en) 2017-06-26 2021-03-24 BeiGene, Ltd. Immunotherapy for hepatocellular carcinoma
CN111801334B (en) 2017-11-29 2023-06-09 百济神州瑞士有限责任公司 Treatment of indolent or invasive B-cell lymphomas using combinations comprising BTK inhibitors
EP3735590A1 (en) 2018-01-04 2020-11-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma resistant
CN112040957A (en) 2018-01-08 2020-12-04 H·李·莫菲特癌症中心和研究所公司 Compositions and methods for targeting CD 99-expressing cancers
MX2020007338A (en) 2018-01-09 2020-11-06 H Lee Moffitt Cancer Ct & Res Compositions and methods for targeting clec12a-expressing cancers.
CA3087105A1 (en) 2018-01-10 2019-07-18 Jiangsu Hengrui Medicine Co., Ltd. Pd-l1 antibody, antigen-binding fragment thereof, and pharmaceutical use thereof
US20200405719A1 (en) * 2018-02-17 2020-12-31 Apollomics Inc. Cancer treatment using combination of neutrophil modulator with modulator of immune checkpoint
US20210080467A1 (en) 2018-02-21 2021-03-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of sk1 as biomarker for predicting response to immunecheckpoint inhibitors
BR112020021042A2 (en) 2018-04-17 2021-01-19 Tempest Therapeutics, Inc. BICYCLIC CARBOXAMIDES AND METHODS OF USE OF THE SAME
WO2020020307A1 (en) 2018-07-25 2020-01-30 I-Mab Biopharma Co., Ltd. Anti-cd73 anti-pd-l1 bispecific antibodies
WO2020104479A1 (en) 2018-11-20 2020-05-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating cancers and resistant cancers with anti transferrin receptor 1 antibodies
US20230183379A1 (en) 2018-11-20 2023-06-15 INSERM (Institut National de la Santé et de la Recherche Médicale) Bispecific antibody targeting transferrin receptor 1 and soluble antigen
WO2020115261A1 (en) 2018-12-07 2020-06-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
WO2020120592A1 (en) 2018-12-12 2020-06-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating melanoma
EP3898699A1 (en) 2018-12-19 2021-10-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating cancers by immuno-modulation using antibodies against cathespin-d
WO2020127885A1 (en) 2018-12-21 2020-06-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Compositions for treating cancers and resistant cancers
WO2020157131A1 (en) 2019-01-30 2020-08-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for identifying whether a subject suffering from a cancer will achieve a response with an immune-checkpoint inhibitor
CA3128104A1 (en) 2019-02-03 2020-08-06 Jiangsu Hengrui Medicine Co., Ltd. Anti-pd-1 antibody, antigen-binding fragment thereof and pharmaceutical use thereof
WO2020161083A1 (en) 2019-02-04 2020-08-13 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for modulating blood-brain barrier
EP3924520A1 (en) 2019-02-13 2021-12-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for selecting a cancer treatment in a subject suffering from cancer
EP3963109A1 (en) 2019-04-30 2022-03-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
US20220233685A1 (en) 2019-06-18 2022-07-28 Janssen Sciences Ireland Unlimited Company Combination of hepatitis b virus (hbv) vaccines and anti-pd-1 antibody
AU2020295012A1 (en) 2019-06-18 2022-02-17 Janssen Sciences Ireland Unlimited Company Combination of hepatitis B virus (HBV) vaccines and anti-PD-1 or anti-PD-L1 antibody
EP4011389A4 (en) * 2019-08-09 2024-09-04 Riken Combined use of artificial adjuvant vector cells and an immunostimulant
WO2021048292A1 (en) 2019-09-11 2021-03-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
WO2021064180A1 (en) 2019-10-03 2021-04-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for modulating macrophages polarization
US20240301497A1 (en) 2019-10-17 2024-09-12 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosing nasal intestinal type adenocarcinomas
WO2021083959A1 (en) 2019-10-29 2021-05-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating uveal melanoma
AU2020408198A1 (en) 2019-12-19 2022-07-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and vaccine compositions to treat cancers
WO2021144426A1 (en) 2020-01-17 2021-07-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
US20230072528A1 (en) 2020-02-05 2023-03-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for discontinuing a treatment with a tyrosine kinase inhibitor (tki)
WO2021170777A1 (en) 2020-02-28 2021-09-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for diagnosing, prognosing and managing treatment of breast cancer
JP2023525020A (en) 2020-05-05 2023-06-14 リジェネロン・ファーマシューティカルズ・インコーポレイテッド CARs containing CD28zeta and CD3zeta
CA3181538A1 (en) 2020-05-05 2021-11-11 Teon Therapeutics, Inc. Cannabinoid receptor type 2 (cb2) modulators and uses thereof
JP2023531493A (en) 2020-06-26 2023-07-24 アムジエン・インコーポレーテツド IL-10 muteins and fusion proteins thereof
WO2022023379A1 (en) 2020-07-28 2022-02-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for preventing and treating a cancer
AU2021357841A1 (en) 2020-10-08 2023-06-15 Affimed Gmbh Trispecific binders
WO2022084531A1 (en) 2020-10-23 2022-04-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating glioma
WO2022101484A1 (en) 2020-11-16 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating uveal melanoma
EP4244391A1 (en) 2020-11-16 2023-09-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for predicting and treating uveal melanoma
TW202227089A (en) 2020-11-30 2022-07-16 大陸商杭州阿諾生物醫藥科技有限公司 Combination therapy for the treatment of pik3ca mutant cancer
EP4308118A1 (en) 2021-03-17 2024-01-24 Institut National de la Santé et de la Recherche Médicale (INSERM) Methods and compositions for treating melanoma
WO2022223791A1 (en) 2021-04-23 2022-10-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating cell senescence accumulation related disease
WO2023007023A1 (en) 2021-07-30 2023-02-02 Affimed Gmbh Duplexbodies
WO2023012147A1 (en) 2021-08-03 2023-02-09 F. Hoffmann-La Roche Ag Bispecific antibodies and methods of use
WO2023034530A1 (en) 2021-09-02 2023-03-09 Teon Therapeutics, Inc. Methods of improving growth and function of immune cells
WO2023078900A1 (en) 2021-11-03 2023-05-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating triple negative breast cancer (tnbc)
WO2023081730A1 (en) 2021-11-03 2023-05-11 Teon Therapeutics, Inc. 4-hydroxy-2-oxo-1,2-dihydro-1,8-naphthyridine-3-carboxamide derivatives as cannabinoid cb2 receptor modulators for the treatment of cancer
CA3238283A1 (en) 2021-11-15 2023-05-19 Yangxin Fu Fusion protein construct taking interleukin 15 as active ingredient and use thereof
WO2023097211A1 (en) 2021-11-24 2023-06-01 The University Of Southern California Methods for enhancing immune checkpoint inhibitor therapy
KR20230092466A (en) 2021-12-17 2023-06-26 한국과학기술연구원 composition comprising PD-L1-inhibiting prodrug for the prevention or treatment of cancer
WO2023118165A1 (en) 2021-12-21 2023-06-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating melanoma
WO2024015372A1 (en) 2022-07-14 2024-01-18 Teon Therapeutics, Inc. Adenosine receptor antagonists and uses thereof
WO2024033399A1 (en) 2022-08-10 2024-02-15 Institut National de la Santé et de la Recherche Médicale Sigmar1 ligand for the treatment of pancreatic cancer
WO2024033400A1 (en) 2022-08-10 2024-02-15 Institut National de la Santé et de la Recherche Médicale Sk2 inhibitor for the treatment of pancreatic cancer
WO2024056716A1 (en) 2022-09-14 2024-03-21 Institut National de la Santé et de la Recherche Médicale Methods and pharmaceutical compositions for the treatment of dilated cardiomyopathy
WO2024084034A1 (en) 2022-10-21 2024-04-25 Institut National de la Santé et de la Recherche Médicale Methods and pharmaceutical compositions for the treatment of osteoarthritis

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100549866B1 (en) * 2001-08-22 2006-02-08 고려대학교 산학협력단 Pharmaceutic ingredient for medical treatment and prevention of cancer
EP2206517B1 (en) 2002-07-03 2023-08-02 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions comprising anti-PD-L1 antibodies
JP4713638B2 (en) * 2006-04-27 2011-06-29 ソウル ナショナル ユニバーシティー インダストリー ファウンデーション B-cell-mediated vaccine loaded with natural killer T cell ligand and antigen
EP2061504A4 (en) * 2006-09-20 2010-01-27 Univ Johns Hopkins Combinatorieal therapy of cancer and infectious diseases with anti-b7-h1 antibodies

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
CN103957939A (en) * 2011-09-19 2014-07-30 约翰霍普金斯大学 Cancer immunotherapy
US9308253B2 (en) 2011-09-19 2016-04-12 The Johns Hopkins University Cancer immunotherapy
WO2013043647A1 (en) * 2011-09-19 2013-03-28 The Johns Hopkins University Cancer immunotherapy
US10487147B2 (en) 2011-11-28 2019-11-26 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US11884724B2 (en) 2011-11-28 2024-01-30 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US9624298B2 (en) 2011-11-28 2017-04-18 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US10759856B2 (en) 2011-11-28 2020-09-01 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
US10738117B2 (en) 2013-05-02 2020-08-11 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
US9815897B2 (en) 2013-05-02 2017-11-14 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
RU2744880C1 (en) * 2014-02-04 2021-03-16 Инсайт Корпорейшн Combination of pd-1 antagonist and ido1 inhibitor for treating ancer
US10280223B2 (en) 2014-07-09 2019-05-07 Nippon Zenyaku Kogyo Co., Ltd. Anti-canine PD-1 antibody or anti-canine PD-L1 antibody
US10416160B2 (en) 2014-07-25 2019-09-17 Riken Memory invariant NKT cell marker
US10751412B2 (en) 2015-05-29 2020-08-25 Merck Sharp & Dohme Corp. Combination of a PD-1 antagonist and CPG-C type oligonucleotide for treating cancer
WO2016196173A1 (en) * 2015-05-29 2016-12-08 Merck Sharp & Dohme Corp. Combination of a pd-1 antagonist and cpg-c type oligonucleotide for treating cancer
US11918648B2 (en) 2015-05-29 2024-03-05 Merck Sharp & Dohme Llc Combination of a PD-1 antagonist and CpG-C type oligonucleotide for treating cancer
US11155624B2 (en) 2016-11-01 2021-10-26 Anaptysbio, Inc. Antibodies directed against programmed death-1 (PD-1)
US11407830B2 (en) 2017-01-09 2022-08-09 Tesaro, Inc. Methods of treating cancer with anti-PD-1 antibodies
US11685782B2 (en) 2017-10-23 2023-06-27 Children's Medical Center Corporation Methods of treating cancer using LSD1 inhibitors in combination with immunotherapy
CN111920948A (en) * 2020-09-25 2020-11-13 北京广未生物科技有限公司 Pharmaceutical composition comprising immune cells for treating cancer
WO2024137442A1 (en) 2022-12-21 2024-06-27 Gilead Sciences, Inc. Combination therapy for treating cancer

Also Published As

Publication number Publication date
JP5054057B2 (en) 2012-10-24
US8617546B2 (en) 2013-12-31
KR101050829B1 (en) 2011-07-20
EP2172219A1 (en) 2010-04-07
US20120237522A1 (en) 2012-09-20
EP2172219B1 (en) 2013-09-11
KR20100037899A (en) 2010-04-12
JP2010083863A (en) 2010-04-15

Similar Documents

Publication Publication Date Title
US8617546B2 (en) Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody
US11866509B2 (en) Humanized antibodies against CEACAM1
EP2218461B1 (en) CD40 antibody formulation and methods
US8475790B2 (en) Combination of CD137 antibody and CTLA-4 antibody for the treatment of proliferative diseases
KR100510604B1 (en) Identification of unique binding interactions between certain antibodies and the human b7.1 and b7.2 co-stimulatory antigens
RU2155190C2 (en) Immunostimulatory monoclonal antibodies
JP2022033899A (en) Treatment of cancer with combinations of immunoregulatory agents
WO2013169693A1 (en) Methods of treating cancer using an il-21 polypeptide and an anti-pd-1 antibody
US20110038855A1 (en) Trail and methods of modulating t cell activity and adaptive immune responses using trail
EP3156072A1 (en) Method for reducing side effects of immune checkpoint control agent
CN109476752A (en) Purposes of the combination of anti-PD-1 antibody and anti-CD30 antibody in lymphoma treating
JP2021534088A (en) Combination of immunotherapy and MDM2 inhibitor
WO1995020605A9 (en) Immuno-stimulatory monoclonal antibodies
TWI793325B (en) Antibodies specific for cd3 and uses thereof
CN110312525A (en) The combination and application thereof of T cell redirection multipurpose antibody and immunologic test point regulator
WO2018101448A1 (en) Method of treating cancer using anti-ccr4 antibody and anti-pd-1 antibody
US10005843B2 (en) Anti-TNFRSF25 antibodies
US20210277135A1 (en) Ox-40 agonist, pd-1 pathway inhibitor and ctla-4 inhibitor combination for use in a method of treating a cancer or a solid tumor
US11427647B2 (en) Polynucleotides encoding humanized antibodies against CEACAM1
EP4342492A1 (en) Pharmaceutical combination and use thereof
MXPA06006153A (en) Cd40 antibody formulation and methods

Legal Events

Date Code Title Description
AS Assignment

Owner name: ANTICANCER AGENT COMPRISING ANTI-PD-1 ANTIBODY OR

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KANG, CHANG YUIL;CHANG, WOO SUNG;KIM, JI YEON;REEL/FRAME:022448/0914

Effective date: 20090316

STCB Information on status: application discontinuation

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