WO2016014148A1 - Targeting dna-pkcs and b7-h1 to treat cancer - Google Patents

Targeting dna-pkcs and b7-h1 to treat cancer Download PDF

Info

Publication number
WO2016014148A1
WO2016014148A1 PCT/US2015/032016 US2015032016W WO2016014148A1 WO 2016014148 A1 WO2016014148 A1 WO 2016014148A1 US 2015032016 W US2015032016 W US 2015032016W WO 2016014148 A1 WO2016014148 A1 WO 2016014148A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
dna
pkcs
composition
patient
Prior art date
Application number
PCT/US2015/032016
Other languages
French (fr)
Inventor
Haidong Dong
Original Assignee
Mayo Foundation For Medical Education And Research
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 Mayo Foundation For Medical Education And Research filed Critical Mayo Foundation For Medical Education And Research
Priority to US15/325,612 priority Critical patent/US10517875B2/en
Priority to EP15825450.8A priority patent/EP3171896A4/en
Publication of WO2016014148A1 publication Critical patent/WO2016014148A1/en
Priority to US16/681,154 priority patent/US11504376B2/en
Priority to US17/991,459 priority patent/US20230233571A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/11001Non-specific serine/threonine protein kinase (2.7.11.1), i.e. casein kinase or checkpoint kinase
    • 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
    • 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
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • 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
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70532B7 molecules, e.g. CD80, CD86
    • 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
    • G01N2333/70596Molecules with a "CD"-designation not provided for elsewhere in G01N2333/705
    • 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
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/912Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • G01N2333/91205Phosphotransferases in general

Definitions

  • This document relates to materials and methods for treating potentially chemoresistant tumors, and particularly to materials and methods for treating such tumors with DNA-PKcs inhibitors.
  • Cancer therapies using checkpoint blockades can blunt the immune-suppressive function of ligands (e.g., B7-H1 on tumor cells) by blocking interaction with their receptors (e.g., PD-1 on T cells) (Zang and Allison, Clin Cancer Res 13:5271-5279, 2007; and Dong and Chen, JMol Med 81 :281-287, 2003). While such therapies can contribute to enhanced antitumor immunity, blocking the binding of B7-H1 to PD-1 may not overcome B7-H1 -mediated chemoresistance.
  • the therapeutic methods described herein can be used to target B7-H1 's intrinsic signaling pathway in relation to chemoresistance.
  • DNA-PKcs is a B7-H1 binding protein, and that DNA-PKcs can be targeted to reduce B7-H1 -mediated chemoresistance.
  • DNA-PKcs is a nuclear protein involved in DNA damage repair (Collis et al., Oncogene 24:949-961, 2005), while B7-H1 is an immunoregulatory molecule mainly expressed on the cell surface of tumor cells (Dong et al, Nat Med 8:793-800, 2002).
  • this document features a method for treating a cancer patient.
  • the method can include (a) identifying a cancer patient to be treated with a chemotherapeutic agent that causes DNA damage, (b) administering to the patient a molecule targeted to DNA-PKcs, wherein the molecule is administered in an amount sufficient to reduce the interaction of DNA-PKcs with B7-H1, and (c) administering the chemotherapeutic agent to the patient.
  • the patient can be a human.
  • the chemotherapeutic agent can be cisplatin, doxorubicin, SN38, paclitaxel, protein-bound paclitaxel, temozolomide, or carboplatin.
  • the molecule targeted to DNA-PKcs can be NU7026, NU7441, IC86621, IC87102, IC87361, OK-1035, SU11752, vanillin, or IC486241, or can be an anti-DNA-PKcs antibody.
  • the molecule targeted to DNA-PKcs and the chemotherapeutic agent can be administered simultaneously or sequentially.
  • this document features a method for treating a cancer patient, where the method can include (a) identifying the patient as having a tumor with cells that express B7-H1, and (b) administering to the patient a DNA-PKcs inhibitor and an anti- B7-H1 blocking antibody.
  • the cancer patient can be a human.
  • the cancer patient can be identified based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7-H1 mRNA in a sample obtained from the tumor.
  • the cancer can be a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer.
  • the DNA-PKcs inhibitor and the anti-B7-Hl blocking antibody can be administered to the cancer patient simultaneously or sequentially.
  • this document features a method for treating cancer, where the method can include administering a DNA-PKcs inhibitor and an anti-B7-Hl antibody to a mammal identified as having a tumor containing cells with an elevated level of B7-H1, where the DNA-PKcs inhibitor and the anti-B7-Hl antibody are administered under conditions in which the interaction of naturally-occurring B7-H1 with DNA-PKcs and the interaction of naturally-occurring B7-H1 with PD-1 or CD80 in the mammal is reduced after the administering.
  • the mammal can be a human.
  • the elevated level B7-H1 can be based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7-H1 m NA in a sample obtained from the tumor.
  • the cancer can be a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer.
  • the DNA-PKcs inhibitor and the anti-B7-Hl blocking antibody can be administered to the mammal simultaneously or sequentially.
  • this document features a composition containing a pharmaceutically acceptable carrier and a molecule targeted to DNA-PKcs for use in treating a cancer patient who also is to be treated with a chemotherapeutic agent that causes DNA damage, wherein the composition is to be administered in an amount sufficient to reduce the interaction of DNA-PKcs with B7-H1.
  • the patient can be a human.
  • the chemotherapeutic agent can be cisplatin, doxorubicin, SN38, paclitaxel, protein-bound paclitaxel, temozolomide, or carboplatin.
  • the molecule targeted to DNA- PKcs can be NU7026, NU7441, IC86621, IC87102, IC87361, OK-1035, SU11752, vanillin, or IC486241, or can be an anti-DNA-PKcs antibody.
  • the chemotherapeutic agent and the composition containing the molecule targeted to DNA-PKcs can be for simultaneous or sequential administration.
  • this document features a composition containing a
  • the cancer patient can be a human.
  • the cancer patient can be identified based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7-H1 m NA in a sample obtained from the tumor.
  • the cancer can be a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer.
  • the anti-B7-Hl blocking antibody and the composition containing the DNA-PKcs inhibitor can be for simultaneous or sequential administration.
  • this document features a composition containing a pharmaceutically acceptable carrier and a DNA-PKcs inhibitor for use in treating cancer in a mammal identified as having a tumor containing cells with an elevated level of B7- Hl, wherein the mammal also is to be treated with an anti-B7-Hl antibody, and wherein the composition containing the DNA-PKcs inhibitor and the anti-B7-Hl antibody are to be administered under conditions in which the interaction of naturally-occurring B7-H1 with DNA-PKcs and the interaction of naturally-occurring B7-H1 with PD-1 or CD80 in the mammal are reduced after the administering.
  • the mammal can be a human.
  • the elevated level B7-H1 can be based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7-H1 mRNA in a sample obtained from the tumor.
  • the cancer can be a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer.
  • composition containing the DNA-PKcs inhibitor can be for simultaneous or sequential administration.
  • FIGS. 1A-1C demonstrate that B7-H1 confers tumor cell resistance to cytotoxic drugs.
  • FIG. 1 A is a series of graphs plotting relative survival of Mock/624mel (triangles) and B7-Hl/624mel (squares) cells treated with the indicated drugs, as determined by MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H- tetrazolium) assay.
  • MTS 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H- tetrazolium
  • Apoptosis of tumor cells treated with doxorubicin was analyzed by tetramethylrhodamine ethyl ester (TMRE) and Annexin V staining (FIG. IB), and by intracellular staining for active caspase-3 (FIG. 1C). Numbers are percentages of gated population.
  • FIGS. 2A-2C are pictures from co-immunoprecipitation and Western blotting (WB) experiments, demonstrating that B7-H1 associates with DNA-PKcs.
  • FIG. 2A Anti-B7-Hl Ab, but not control Ab, co-precipitated a protein band in B7-H1 transfected 624mel cells.
  • FIG. 2B Co-immunoprecipitation (IP) and Western blotting (WB) using cell lysate as input.
  • FIG. 2C Human primary T cells were activated by
  • B7-H1 was pulled down by anti-B7-Hl antibody but not control antibody.
  • the association of B7-H1 with DNA-PKcs was detected by Western blotting using anti-DNA-PKcs antibody.
  • FIG. 3 is a graph plotting relative survival of tumor cells treated with a DNA- PKcs inhibitor, and showing that inhibition of DNA-PKcs reduced tumor drug resistance.
  • Cells were pre-treated with DMSO or NU7026 (10 uM) for 1 hour before treatment with doxorubicin (Dox). The relative survival of tumor cells was measured by MTS assay 72 hours after Dox treatment.
  • FIG. 4 is a diagram showing potential molecular mechanisms for B7-H1 -mediated tumor chemoresistance.
  • FIG. 5A is a cross-species alignment of the B7-H1 intracellular domains (ICD) from human (SEQ ID NO: l), chimpanzee (SEQ ID NO: l), Rhesus monkey (SEQ ID NO:2), marmoset (SEQ ID NO:3), sooty mangabey (SEQ ID NO:4), cow (SEQ ID NO:
  • FIG. 5B is a diagram of Flag-B7-Hl ICD domain fragments (SEQ ID NOS:9-13).
  • FIG. 6 is a series of pictures showing co-localization of B7-H1 and DNA-PKcs.
  • a human breast tumor cell line (MDA-MB-231) was treated with doxorubicin (2 ug/ml) for 2 hours. Co-localization of B7-H1 and DNA-PKcs was observed in the nuclei.
  • FIG. 7 is a picture of a blot showing B7-H1 or control (GAPDH) expression in human tumor cells (MDA-MB-231) following transfection with control or B7-H1 shRNA.
  • FIGS. 8A-8C demonstrate that B7-H1 enhances activation of the MAPK/ERK pathway.
  • FIG. 8A is a picture (top) and a graph (bottom) showing the results of an antibody array assay of phosphorylation of molecules in the MAPK/ERK pathway. The levels of phospho-ERK and total ERK were analyzed by Western blotting (FIG. 8B) and flow cytometry (FIG. 8C). Numbers are mean fluorescence intensity (MFI).
  • FIG. 9 is a series of graphs plotting the levels of phosphorylated ERK (top graphs) and total ERK (bottom graphs) in Mock/624mel (left) and B7-Hl/624mel (right) tumor cells treated with DMSO or NU7026. Cells were treated with DMSO or NU7026 (10 uM) for 24 hours before intracellular staining for phospho-ERK or total ERK.
  • FIG. 10 is a schematic showing a potential DNA-PKcs/ERK/Bim pathway that may be used by B7-H1 to counter tumor cell apoptosis.
  • FIG. 11 is a schematic showing a potential DNA-PKcs/ Akt/Bad pathway that may be used by B7-H1 to counter tumor cell apoptosis.
  • FIG. 12 contains representative nucleic acid (top) and amino acid (bottom) sequences for human B7-H1 (SEQ ID NOS: 14 and 15, respectively).
  • FIG. 13 contains representative nucleic acid (top) and amino acid (bottom) sequences for human PD-1 (SEQ ID NOS: 16 and 17, respectively).
  • FIGS. 14A and 14B contain representative nucleic acid (14A) and amino acid
  • FIGS. 15A and 15B show that DNA-PKcs activity is required for the association of DNA-PKcs with B7-H1.
  • FIG. 15A shows an immuno-precipitation assay, demonstrating that NU7026 abolished the association of B7-H1 with DNA-PKcs.
  • FIG. 15B is a series of pictures from an immunofluorescence assay in which the cells treated with doxorubicin, NU7026, or both, and stained for DNA-PKcs, DNA, or B7-H1. Doxorubicin-induced co-localization (arrow, upper right panel) of B7-H1 and DNA-PKcs at the nucleus was blocked by NU7026 (lower right panel).
  • This document provides methods and materials for treating cancer in patients with tumors that are chemoresistant or are at risk of becoming chemoresistant.
  • this document provides methods and materials for identifying a cancer patient (e.g., a mammal such as a human, non-human primate, cow, sheep, pig, dog, rabbit, rat, or mouse) as having a tumor that expresses B7-H1 at an elevated level, and treating the patient with a molecule that can interfere with the interaction between B7-H1 and DNA- PKcs.
  • a cancer patient e.g., a mammal such as a human, non-human primate, cow, sheep, pig, dog, rabbit, rat, or mouse
  • the methods provided herein also can include treating the patient with a molecule that can interfere with the interaction between B7-H1 and PD-1 , and/or the interaction between B7-H1 and CD80 (e.g., an antibody against B7-H1, PD-1, or CD80, or a fusion protein containing a portion of PD-1 or a portion of CD80 fused to an immunoglobulin (Ig) Fc domain).
  • a molecule that can interfere with the interaction between B7-H1 and PD-1 and/or the interaction between B7-H1 and CD80 (e.g., an antibody against B7-H1, PD-1, or CD80, or a fusion protein containing a portion of PD-1 or a portion of CD80 fused to an immunoglobulin (Ig) Fc domain).
  • a molecule that can interfere with the interaction between B7-H1 and PD-1 e.g., an antibody against B7-H1, PD-1, or CD80, or a fusion protein
  • the term "elevated level” as used herein with respect to a level of B7-H1 refers to a level that is greater (e.g., 50% greater, 2-fold greater, 3-fold greater, or more than 3-fold greater) than a reference level of B7-H1.
  • the term "reference level” as used herein with respect to B7-H1 can refer to the level of B7-H1 typically observed in cells from healthy subjects without cancer. In some embodiments, for example, a reference level of B7-H1 can be the average level of B7-H1 present in samples obtained from a random sampling of 50 humans free of cancer.
  • B7-H1 levels can be determined based on cell staining, and "elevated” and “reference” levels can set based on the percentage of evaluated cells that stain positive for B7-H1. For example, in some embodiments, samples (e.g., tumor samples) in which five percent or less (e.g., five percent, four percent, three percent, two percent, one percent, or less than one percent) of the cells stain positive for B7-H1 can be considered B7-H1 negative. In some
  • samples e.g., tumor samples
  • ten percent or more e.g., ten percent, 20 percent, 25 percent, 30 percent, 40 percent, 50 percent, or more than 50 percent
  • samples in which ten percent or more (e.g., ten percent, 20 percent, 25 percent, 30 percent, 40 percent, 50 percent, or more than 50 percent) of the cells stain positive for B7-H1 can be considered to have elevated levels of B7-H1 expression.
  • the presence of an elevated level of B7-H1 can be determined by measuring, for example, B7-H1 protein levels or B7-H1 nucleic acid levels.
  • the level of B7-H1 protein can be measured in a tumor sample from a mammal with cancer using cell staining, western blotting, or other immunological techniques.
  • the level of B7-H1 expression also can be measured at the nucleic acid level, using Northern blotting, or any other method suitable for determining mRNA levels of CD274, which encodes the B7-H1 protein.
  • B7-H1 protein or nucleic acid levels can be measured in ascites samples, or lymphoid organ samples. It will be appreciated that levels from comparable samples are used when determining whether or not a particular level is an elevated level.
  • a representative example of a human B7-H1 nucleic acid has the sequence set forth in GENBANK ® Accession No. AF177937 (GI No. 6708118) (SEQ ID NO: 14; Figure 12), and a representative human B7-H1 polypeptide has the sequence set forth in GENBANK ® Accession No. AAF25807 (GI No. 6708119) (SEQ ID NO: 15; Figure 12).
  • a representative example of a human PD-1 nucleic acid can have the sequence set forth in GENBANK ® Accession No. BC074740.2 (GI No. 50960296) (SEQ ID NO: 16; FIG. 13), and representative example of a human PD-1 polypeptide has the sequence set forth in GENBANK ® Accession No. AAH74740.1 (GI No. 49902307) (SEQ ID NO: 17; FIG. 13).
  • a representative example of a human CD80 nucleic acid has the sequence set forth in NCBI Reference No. NM_005191.3 (GI No. 113722122) (SEQ ID NO: 18; FIG. 14A), and a representative example of a human CD80 polypeptide has the sequence set forth in NCBI Reference No. NP 005182.1 (GI No. 4885123) (SEQ ID NO: 19; FIG. 14B).
  • the level of B7-H1 within a tumor sample from a mammal is determined, the level can be compared to a reference level, and the mammal can be classified as having or not having an elevated level of B7-H1. If the mammal is identified as having an elevated level of B7-H1, the mammal can be treated with a first molecule that inhibits the interaction between B7-H1 and DNA-PKcs.
  • a small molecule DNA- PKcs inhibitor such as NU7026, NU7441, IC86621, IC87102, IC87361, OK-1035, SU11752, vanillin, or IC486241 can be administered to the mammal.
  • an anti-DNA-PKcs antibody can be administered to block the interaction between B7-H1 and DNA-PKcs.
  • B7-H1 peptides can be useful. Such peptides can be fragments of B7-H1 (e.g., fragments containing about 10-20, about 20-50, or about 50-100 amino acids) that include the DNA-PKcs binding domain, such that they can inhibit the interaction between B7-H1 and DNA-PKcs. Such peptides can be referred to as "interfering B7-H1 small peptides.”
  • the mammal also can be treated with a second molecule that inhibits the interaction between B7-H1 and PD-1 and/or the interaction between B7- Hl and CD80.
  • second molecules include, without limitation, antibodies (e.g., anti-B7-Hl antibodies, anti-PD-1 antibodies, or anti-CD80 antibodies), and fusion proteins (e.g., PD-1 fusion proteins or CD80 fusion proteins).
  • fusion proteins can contain, for example, the extracellular domain of PD-1 fused to an IgG Fc domain, or the extracellular domain of CD80 fused to an IgG Fc domain.
  • binding of the fusion proteins to B7-H1 can reduce or block the ability of B7-H1 to interact with PD-1 and/or CD80.
  • antibody includes monoclonal antibodies, polyclonal antibodies, recombinant antibodies, humanized antibodies (Jones et al. (1986) Nature 321 :522-525; Riechmann et al. (1988) Nature 332:323-329; and Presta (1992) Curr. Op. Struct. Biol. 2:593-596), chimeric antibodies (Morrison et al. (1984) Proc. Natl. Acad. Sci.
  • antibody fragment comprises any portion of the afore -mentioned antibodies, such as their antigen binding or variable regions.
  • antibody fragments include Fab fragments, Fab' fragments, F(ab')2 fragments, Fv fragments, diabodies (Hollinger et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448), single chain antibody molecules (Pluckthun in: The Pharmacology of
  • anti-human B7-H1 antibodies examples include, without limitation, anti-human B7-H1 antibodies commercially available from Biolegend (e.g., Catalog No.
  • anti-human PD-1 antibodies examples include, without limitation, anti-human PD-1 antibodies commercially available from Biolegend (e.g., Catalog No. 329904 or 329905) or eBioscience (Catalog No. 12-2799-42; San Diego, CA).
  • anti-human CD80 antibodies include, without limitation, anti-human CD8 antibodies commercially available from Biolegend (e.g., Catalog No. 305201 or 305202) or eBioscience (e.g., Catalog No. 14-0809-80 or 14-0809-82).
  • antibody also includes antibody-like molecules that contain engineered sub-domains of antibodies or naturally occurring antibody variants. These antibody-like molecules may be single-domain antibodies such as VH-only or VL- only domains derived either from natural sources such as camelids (Muyldermans et al. (2001) Rev. Mol. Biotechnol. 14 211-1Q2) or through in vitro display of libraries from humans, camelids or other species (Holt et al. (2003) Trends Biotechnol. 21 :484-90).
  • the polypeptide structure of the antigen binding proteins can be based on antibodies, including, but not limited to, minibodies, synthetic antibodies (sometimes referred to as “antibody mimetics”), human antibodies, antibody fusions (sometimes referred to as “antibody conjugates”), and fragments thereof, respectively.
  • an “Fv fragment” is the minimum antibody fragment that contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy chain variable domain and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three CDR's of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six CDR's confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDR's specific for an antigen) has the ability to recognize and bind the antigen, although usually at a lower affinity than the entire binding site.
  • the “Fab fragment” also contains the constant domain of the light chain and the first constant domain (CHI) of the heavy chain.
  • the “Fab fragment” differs from the “Fab' fragment” by the addition of a few residues at the carboxy terminus of the heavy chain CHI domain, including one or more cysteines from the antibody hinge region.
  • the “F(ab')2 fragment” originally is produced as a pair of “Fab' fragments” which have hinge cysteines between them. Methods of preparing such antibody fragments, such as papain or pepsin digestion, are known to those skilled in the art.
  • An antibody can be of the IgA-, IgD-, IgE, IgG- or IgM-type, including IgG- or IgM-types such as, without limitation, IgGl-, IgG2-, IgG3-, IgG4-, IgMl- and IgM2- types.
  • an antibody can be of the IgGl-, IgG2- or IgG4- type.
  • antibodies as used in the methods described herein can be fully human or humanized antibodies.
  • Human antibodies can avoid certain problems associated with xenogeneic antibodies, such as antibodies that possess murine or rat variable and/or constant regions.
  • the effector portion is human, it can interact better with other parts of the human immune system, e.g., to destroy target cells more efficiently by complement-dependent cytotoxicity or antibody-dependent cellular cytotoxicity.
  • the human immune system should not recognize the antibody as foreign.
  • the half-life in human circulation will be similar to naturally occurring human antibodies, allowing smaller and less frequent doses to be given. Methods for preparing human antibodies are known in the art.
  • Humanized antibodies can be used, and can have many advantages.
  • Humanized antibodies generally are chimeric or mutant monoclonal antibodies from mouse, rat, hamster, rabbit or other species, bearing human constant and/or variable region domains or specific changes.
  • Techniques for generating humanized antibodies are well known to those of skill in the art. For example, controlled rearrangement of antibody domains joined through protein disulfide bonds to form new, artificial protein molecules or "chimeric" antibodies can be utilized (Konieczny et al. (1981) Haematologia (Budap.) 14:95).
  • Recombinant DNA technology can be used to construct gene fusions between DNA sequences encoding mouse antibody variable light and heavy chain domains and human antibody light and heavy chain constant domains (Morrison et al. (1984) Proc. Natl. Acad. Sci. USA 81 :6851).
  • DNA sequences encoding antigen binding portions or complementarity determining regions (CDR's) of murine monoclonal antibodies can be grafted by molecular means into DNA sequences encoding frameworks of human antibody heavy and light chains (Jones et al. (1986) Nature 321 :522; Riechmann et al. (1988) Nature 332:323). Expressed recombinant products are called "reshaped" or humanized antibodies, and comprise the framework of a human antibody light or heavy chain and antigen recognition portions, CDR's, of a murine monoclonal antibody.
  • Molecules that interfere with the interaction between B7-H1 and DNA-PKcs, the interaction between B7-H1 and PD-1, and/or the interaction between B7-H1 and CD80, as described herein can be incorporated into pharmaceutical compositions for treatment of cancer.
  • compositions further can include one or more pharmaceutically acceptable carriers, diluents and/or adjuvants.
  • a “pharmaceutically acceptable carrier” (also referred to as an “excipient” or a “carrier”) is a pharmaceutically acceptable solvent, suspending agent, stabilizing agent, or any other pharmacologically inert vehicle for delivering one or more therapeutic compounds to a subject, which is nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed.
  • Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, and other pertinent transport and chemical properties, when combined with one or more of therapeutic compounds and any other components of a given pharmaceutical composition.
  • Typical pharmaceutically acceptable carriers include, by way of example and not limitation: water, saline solution, binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose), fillers (e.g., lactose and other sugars, gelatin, or calcium sulfate), lubricants (e.g., starch, polyethylene glycol, or sodium acetate), disintegrates (e.g., starch or sodium starch glycolate), and wetting agents (e.g., sodium lauryl sulfate).
  • binding agents e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose
  • fillers e.g., lactose and other sugars, gelatin, or calcium sulfate
  • lubricants e.g., starch, polyethylene glycol, or sodium acetate
  • disintegrates e.g., starch or sodium starch glycolate
  • wetting agents e.g., sodium lau
  • Pharmaceutically acceptable carriers also include aqueous pH buffered solutions or liposomes (small vesicles composed of various types of lipids, phospholipids and/or surfactants which are useful for delivery of a drug to a mammal).
  • Further examples of pharmaceutically acceptable carriers include buffers such as phosphate, citrate, and other organic acids, antioxidants such as ascorbic acid, low molecular weight (less than about ten residues) polypeptides, proteins such as serum albumin, gelatin, or immunoglobulins, hydrophilic polymers such as polyvinylpyrrolidone, amino acids such as glycine, glutamine, asparagine, arginine or lysine, monosaccharides, disaccharides, and other carbohydrates including glucose, mannose or dextrins, chelating agents such as EDTA, sugar alcohols such as mannitol or sorbitol, salt-forming counterions such as sodium, and/or nonionic surfactants such as TWEENTM
  • compositions can be formulated by mixing one or more active agents (e.g., a DNA-PKcs inhibitor or an anti-B7-Hl blocking antibody) with one or more physiologically acceptable carriers, diluents, and/or adjuvants, and optionally other agents that are usually incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • active agents e.g., a DNA-PKcs inhibitor or an anti-B7-Hl blocking antibody
  • physiologically acceptable carriers e.g., a DNA-PKcs inhibitor or an anti-B7-Hl blocking antibody
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. Any of the foregoing mixtures may be appropriate in treatments and therapies as described herein, provided that the active agent in the formulation is not inactivated by the formulation and the formulation is physiologically compatible and tolerable with the route of
  • compositions include, without limitation, solutions, emulsions, aqueous suspensions, and liposome-containing formulations. These compositions can be generated from a variety of components that include, for example, preformed liquids, self-emulsifying solids and self-emulsifying semisolids.
  • Emulsions are often biphasic systems comprising of two immiscible liquid phases intimately mixed and dispersed with each other; in general, emulsions are either of the water-in-oil (w/o) or oil-in-water (o/w) variety.
  • Emulsion formulations have been widely used for oral delivery of therapeutics due to their ease of formulation and efficacy of solubilization, absorption, and
  • compositions and formulations can include sterile aqueous solutions, which also can contain buffers, diluents and other suitable additives (e.g., penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers). Compositions additionally can contain other adjunct components conventionally found in
  • compositions also can include compatible, pharmaceutically active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or additional materials useful in physically formulating various dosage forms of the compositions provided herein, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • additional materials useful in physically formulating various dosage forms of the compositions provided herein, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings, and aromatic substances.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure,
  • a composition containing a DNA-PKcs inhibitor and/or an antibody or fusion protein as provided herein can be in the form of a solution or powder with or without a diluent to make an injectable suspension.
  • the composition may contain additional ingredients including, without limitation, pharmaceutically acceptable vehicles, such as saline, water, lactic acid, mannitol, or combinations thereof, for example.
  • Methods for using an agent e.g., a small molecule inhibitor of DNA-PKcs, an antibody against B7-H1, DNA-PKcs, PD-1, or CD80, or a PD-l-Fc or CD80-Fc fusion protein
  • an agent e.g., a small molecule inhibitor of DNA-PKcs, an antibody against B7-H1, DNA-PKcs, PD-1, or CD80, or a PD-l-Fc or CD80-Fc fusion protein
  • the methods can include, for example, administering an agent or composition to a subject identified as being in need thereof.
  • a method as provided herein can further include steps such as identifying a mammal (e.g., a human cancer patient) that is to be treated with a chemotherapeutic agent (e.g., cisplatin, doxorubicin, SN38, paclitaxel, protein-bound paclitaxel (e.g., ABRAXANE ® ), temozolomide (e.g., TEMODAR ® ), or carboplatin) that causes DNA damage, or identifying a mammal as having a tumor with cells that express B7-H1.
  • a chemotherapeutic agent e.g., cisplatin, doxorubicin, SN38, paclitaxel, protein-bound paclitaxel (e.g., ABRAXANE ® ), temozolomide (e.g., TEMODAR ® ), or carboplatin
  • a chemotherapeutic agent e.g., c
  • a method can include identifying a mammal to be treated with a chemotherapeutic agent that causes DNA damage, and treating the mammal with an agent that inhibits the interaction between B7-H1 and DNA-PKcs. In some cases, a method can further include the step of administering the chemotherapeutic agent that causes DNA damage.
  • Administration can be, for example, parenteral (e.g., by subcutaneous, intrathecal, intraventricular, intramuscular, or intraperitoneal injection, or by intravenous drip). Administration can be rapid (e.g., by injection) or can occur over a period of time (e.g., by slow infusion or administration of slow release formulations). In some embodiments, administration can be topical (e.g., transdermal, sublingual, ophthalmic, or intranasal), pulmonary (e.g., by inhalation or insufflation of powders or aerosols), or oral.
  • a composition containing an antibody or fusion protein as described herein can be administered prior to, after, or in lieu of surgical resection of a tumor.
  • compositions containing an antibody e.g., an anti-B7-Hl antibody, an anti- DNA-PKcs antibody, an anti-PD-1 antibody, or an anti-CD80 antibody
  • a small molecule e.g., NU7026
  • a fusion protein e.g., a PD-l-Fc fusion or a CD80-Fc fusion
  • a desired outcome e.g., to increase progression- free survival, or to reduce progression of the cancer.
  • Combination therapies in which a DNA-PKcs inhibitor and an anti-B7-Hl blocking antibody are administered to a mammal, can be particularly useful, as such therapies can target both chemoresistance and immunoresistance.
  • a first composition containing a DNA-PKcs inhibitor and a second composition containing an anti-B7-Hl antibody can be administered, either simultaneously (e.g., via simultaneous
  • a DNA-PKcs inhibitor and an anti-B7-Hl antibody can be administered to a mammal having cancer to reduce the progression rate of the cancer by 5, 10, 25, 50, 75, 100, or more percent. For example, the progression rate can be reduced such that no additional cancer progression is detected.
  • a DNA- PKcs inhibitor and an anti B7-H1 antibody can be administered to a mammal having cancer under conditions where progression-free survival is increased (e.g., by 5, 10, 25, 50, 75, 100, or more percent) as compared to the median progression-free survival of corresponding mammals having untreated cancer or the median progression-free survival of corresponding mammals having cancer and treated with other therapies (e.g., immune or chemotherapeutic agents alone).
  • Progression-free survival can be measured over any length of time (e.g., one month, two months, three months, four months, five months, six months, or longer). Any appropriate method can be used to determine whether or not the progression rate of cancer is reduced.
  • the progression rate can be assessed by imaging tissue at different time points and
  • determining the amount of cancer cells present The amounts of cancer cells determined within tissue at different times can be compared to determine the progression rate. After treatment as described herein, the progression rate can be determined again over another time interval. In some cases, the stage of cancer after treatment can be determined and compared to the stage before treatment to determine whether or not the progression rate has been reduced.
  • An effective amount of a composition containing an antibody as provided herein can be any amount that reduces a symptom of the condition being treated, without significant toxicity.
  • an effective amount can reduce the progression rate of the cancer, increase the progression-free survival rate, or increase the median time to progression.
  • Optimum dosages can vary depending on the relative potency of individual polypeptides (e.g., antibodies and fusion proteins), and can generally be estimated based on ECso found to be effective in in vitro and in vivo animal models. Typically, dosage is from 0.01 ⁇ g to 100 g per kg of body weight.
  • an effective amount of an antibody or fusion protein can be from about 1 mg/kg to about 100 mg/kg (e.g., about 5 mg/kg, about 10 mg/kg, about 20 mg/kg, about 50 mg/kg, or about 75 mg/kg). If a particular subject fails to respond to a particular amount, then the amount of the antibody can be increased by, for example, two fold. After receiving this higher concentration, the subject can be monitored for both responsiveness to the treatment and toxicity symptoms, and adjustments made accordingly. The effective amount can remain constant or can be adjusted as a sliding scale or variable dose depending on the mammal's response to treatment. Various factors can influence the actual effective amount used for a particular application. For example, the frequency of administration, duration of treatment, use of multiple treatment agents, route of administration, and severity of the clinical condition may require an increase or decrease in the actual effective amount administered.
  • the frequency of administration can be any frequency that reduces the
  • the frequency of administration can be once or more daily, biweekly, weekly, monthly, or even less.
  • the frequency of administration can remain constant or can be variable during the duration of treatment.
  • a course of treatment can include rest periods.
  • a composition containing one or more agents e.g., a small molecule, antibody, or fusion protein as provided herein
  • the effective amount various factors can influence the actual frequency of administration used for a particular application.
  • the effective amount, duration of treatment, use of multiple treatment agents, route of administration, and severity of the cancer may require an increase or decrease in administration frequency.
  • An effective duration for administering a composition provided herein can be any duration that reduces the progression rate of cancer, increases the progression-free survival rate, or increases the median time to progression without producing significant toxicity to the mammal.
  • the effective duration can vary from several days to several weeks, months, or years.
  • the effective duration for the treatment of cancer can range in duration from several weeks to several months.
  • an effective duration can be for as long as an individual mammal is alive. Multiple factors can influence the actual effective duration used for a particular treatment.
  • an effective duration can vary with the frequency of administration, effective amount, use of multiple treatment agents, route of administration, and severity of the cancer.
  • the mammal After administering one or more agents as provided herein to a mammal, the mammal can be monitored to determine whether or not the treatment was effective. For example, a mammal can be assessed after treatment to determine whether or not the progression rate of the cancer has been reduced. Any method, including those that are standard in the art, can be used to assess progression and survival rates.
  • Example 1 - B7-H1 renders tumor cells resistant to drugs that cause DNA damage.
  • B7-H1 -expressing tumors would show resistance to drugs that cause DNA damage.
  • cisplatin which crosslinks DNA
  • doxorubicin a topoisomerase II inhibitor that leads to generation of free radicals
  • SN38 a topoisomerase I inhibitor
  • Drug sensitivity in B7-H1- and Mock-transfected 624mel melanoma cells was examined in cultures with varying concentrations of the drugs. After 72 hours of culture, tumor cell survival was measured with a MTS assay (Pei et al, Cancer Cell 16:259-266, 2009). As shown in FIG.
  • B7- Hl/624mel cells were more resistant than Mock/624mel cells to cisplatin, doxorubicin, and SN38. Consistent with a previous pharmacogenetic analysis, these data supported the idea that B7-H1 renders tumor cells resistant to cytotoxic chemotherapeutic agents. Since a mechanism of action of most cytotoxic drugs is to induce apoptosis (Sedletska et al. , Curr Med Chem Anti-cancer Agents 5:251 -265 , 2005 ; and Wang et al.
  • apoptosis of B7-Hl/624mel and Mock/624mel tumor cells was measured and compared after treatment with doxorubicin. Specifically, apoptosis was measured based on the binding of Annexin V (AV) and the levels of
  • TMRE tetramethylrhodamine ethyl ester
  • FIG. IB B7- Hl/624mel cells had 5 -fold less apoptosis compared to Mock/624mel cells following treatment with doxorubicin.
  • the level of active caspase-3 an executive molecule of apoptosis also was lower in B7-Hl/624mel cells compared with Mock/624mel cells (FIG.1C).
  • B7-H1 may render tumor cells resistant to cytotoxic drugs by reducing their apoptotic potential to cytotoxic condition.
  • Example 2 - B7-H1 is associated with DNA-P cs
  • a co-immunoprecipitation (Co-IP) assay with tumor cell lysates using anti-B7-Hl monoclonal antibody resulted in identification of a 450-kDa protein band (FIG. 2A).
  • the protein band identified was larger than and distinct from the membrane proteins PD-1 (55kDa) and CD80 (65kDa) that also have been reported to interact with B7-H1.
  • the band was excised from the gel, and mass spectroscopy revealed that the major component was DNA-PKcs (DNA dependent protein kinase, catalytic subunit).
  • NU7026 is an ATP-competitive DNA-PKcs inhibitor, and 10 uM NU7026 completely inhibits DNA-PKcs activity (Veuger et al, Cancer Res 63:6008- 6015, 2003; and Willmore et al, Blood 103:4659-4665, 2004).
  • 10 uM 10 uM
  • B7-Hl/624mel cells demonstrated resistance to doxorubicin, compared with Mock/624mel cells.
  • B7-Hl/624mel cells lost their resistance to doxorubicin and had comparable drug sensitivity with Mock/624mel cells, suggesting that DNA-PKcs may contribute to B7-H1 -mediated drug resistance (FIG. 3).
  • NU7026 also increased the drug sensitivity of Mock/624mel cells, suggesting DNA-PKcs may be a downstream element of B7-H1 signaling pathway.
  • B7-H1 is a DNA damage checkpoint molecule that, association with DNA-PKcs, promotes tumor survival via DNA damage repair and activation of pro-survival signaling pathways, thus countering cytotoxic effects of chemotherapeutic agents (FIG. 4).
  • experiments are conducted to (1) define the role of B7-H1 interaction with DNA-PKcs in DNA damage repair and (2) define the role of B7-H1 interaction with DNA-PKcs in the activation of pro-survival signaling pathways.
  • these studies extend the role of B7-H1, originally defined as an immune checkpoint molecule, to a DNA damage checkpoint molecule of tumor cells upon treatment with cytotoxic drugs.
  • Example 4 Identifying the binding sites(s) of B7-H1 in association with DNA-PKcs
  • the anti-apoptosis function of B7-H1 has been identified in its intracellular domain (ICD) in tumor cells (Azuma et al, Blood 111 :3635-3643, 2008). It is possible that B7-H1 uses its ICD in association with DNA-PKcs to achieve its anti-apoptosis function in tumor cells.
  • the B7-H1 ICD has eight amino acid residues that are conserved across species (FIG. 5A, red font). Based on their distribution and locations, these amino acids can be grouped in three regions (I, II, and III).
  • B7-H1 -negative 624mel tumor cells are transfected with Flag-B7-Hl ICD, and are used in Co-IP (using anti-Flag antibody in pull-down assays) to assess the association of mutant or truncated B7-H1 ICD with DNA-PKcs. It is noted that the transmembrane domain of B7-H1 may be needed if the association with DNA-PKcs requires anchoring of B7-H1 to the cytoplasm or nuclear membrane. In this case, the transmembrane domain of B7-H1 is included in the Flag-B7-Hl fusion protein for the Co-IP assay.
  • Example 5 Identifying the effects of B7-H1 on DNA repair function of DNA-PKcs B7-H1 undergoes redistribution from the cell surface into the nucleus in human breast tumor cells upon treatment with the cytotoxic drug, doxorubicin (Ghebeh et al, supra).
  • doxorubicin the cytotoxic drug
  • studies were conducted to examine whether translocation of B7-H1 results in close association with DNA-PKcs in the nucleus following drug treatment.
  • the results shown in FIG. 6 demonstrate that before treatment with cytotoxic drug, DNA-PKcs was mainly localized in nuclei and B7-H1 was mainly localized in cytoplasm of tumor cells (arrows).
  • B7-H1 was enriched in the nuclei, and co-localization of B7-H1 and DNA- PKcs was observed in the nuclei (arrow heads in FIG. 6), suggesting that B7-H1 may be recruited to the nucleus to regulate DNA-PKcs activity in responses to DNA damage caused by chemotherapeutic drugs.
  • Nuclear ⁇ 2 ⁇ is measured by anti- phospho-H2AX (Serl39) (Cell Signaling, clone 20E3) in confocal immunofluorescence (IF) assays.
  • IF confocal immunofluorescence
  • a time course examination is conducted, with comparison of the distribution (nuclear foci or nuclear periphery) of ⁇ 2 ⁇ between B7-H1 positive and B7-H1 negative (knockdown) tumor cells upon treatment with cytotoxic drug (doxorubicin or cisplatin).
  • cytotoxic drug doxorubicin or cisplatin
  • a flow cytometry-based assay is used to measure the nuclear levels of ⁇ 2 ⁇ (Kataoka et al, J Rad Res 47:245-257, 2006) while measuring DNA content with propidium iodide (Solier et al, Mol Cell Biol 29:68-82, 2009).
  • DNA-PKcs activity anti-phospho-DNA-PKcs (Thr2609) antibody is used (BioLegend, clone 10B1) to detect auto-phosphorylated DNA-PKcs by Western blotting following treatment with cytotoxic agents over a course of time.
  • an EJ5GFP-based chromosomal break reporter (from the Addgene plasmid repository) is used to measure DNA-PKcs mediated non-homologous end joining (NHEJ; Bennardo et al, PLoS Genet 4:el000110, 2008; and Gunn et al, J Biol Chem 286:42470-42482, 2011).
  • EJ5GFP contains a promoter that is separated from a GFP coding cassette by a puro gene that is flanked by two I-Scel sites that are in the same orientation. Once the puro gene is excised by NHEJ repair of the two I-Scel-induced DSBs, the promoter is joined to the rest of the expression cassette, leading to restoration of the GFP+ gene. By measuring the frequency of GFP+ cells, the function of DNA-PKcs in DNA repair is determined. Briefly, reporter plasmids are transfected into B7-H1 -positive or -negative tumor cells, followed by treatment with one or more cytotoxic agents. The frequency of GFP+ cells is determined and compared between B7- Hl -positive and negative-tumor cells using flow cytometry.
  • DNA-PKcs expression of ⁇ 2 ⁇ (a sign of DNA DSBs) will decrease more slowly in B7-H1 -positive tumor cells than in B7-H1 -negative tumor cells, as a result of sufficient DNA repair by DNA-PKcs.
  • Nuclear foci distributed throughout the nucleus are the most common distribution of ⁇ 2 ⁇ , but ⁇ 2 ⁇ expression at the nuclear periphery has been reported in early stage apoptotic cells (Solier et al, supra).
  • DNA-PKcs as a kinase activates Akt and ERK pro-survival signaling pathways (Dragoi et al, EMBO J 24 ⁇ 9- ⁇ %9, 2005; and (Yotsumoto et al, J Immunol 180:809- 816, 2008).
  • the direct effects of DNA-PKcs on ERK activation are not clear, however.
  • NU7026 DNA-PKcs inhibitor
  • FIG. 7 endogenous B7-H1 -positive MDA-MB-231 human tumor cells (FIG. 7) following treatment with cytotoxic drugs.
  • the IC50 for NU7026 to inhibit DNA-PKcs is 0.23 uM (while the IC50 for inhibition of PI3K is 13 uM).
  • NU7026 is titrated gradually from 10 uM to 0.23 uM (using the same volume of DMSO solvent as a control).
  • DNA-PKcs knockout cell lines (Wu et al., J Immunol 174:934-941, 2005) or DNA-PKcs knockdown cell lines are used to measure the levels of ERK1/2 activation in the absence of DNA-PKcs proteins.
  • the ERK1/2 inhibitor U0126 (which inhibits MEK, an upstream kinase of ERK1/2) is used to confirm that phosphorylation of Bim is mediated by ERK1/2 activation.
  • phosphorylated Bim migrates more slowly than non-phosphorylated Bim.
  • lysates are incubated with lambda phosphatase ( ⁇ -PPase, 15 ug/200U for 1 h), which de- phosphorylates modified serine, threonine and tyrosine residues.
  • ⁇ -PPase lambda phosphatase
  • an anti-phospho-Bim (Ser69 in human) antibody (Cell Signaling Tech. #4581) is used in a Western blotting assay, since phosphorylation of Bim at Ser69 by ERK/1/2 promotes Bim degradation (Luciano et al., supra).
  • Total Bim also is measured to determine to what degree Bim is undergoing degradation. Accordingly, the apoptosis of tumor cells is measured by flow cytometry using antibody against activated caspase-3 as described in FIG. 1.
  • DNA-PKcs enhances Akt activity (Dragoi et al., supra; and Feng et al., J Biol Chem 279:41189-41196, 2004).
  • the Akt inhibitor MK2206 (Merck) is used to directly inhibit the enzyme activity of activated Akt (Merck data sheet) in the studies.
  • anti-phospho-Bad (Serl36) antibody (Cell Signaling Technology, clone 185D10) is used in a Western blotting assay, since Akt preferentially phosphorylates Bad at Serl36 in tumor cells (Hayakawa et al., Cancer Res 60:5988-5994, 2000).
  • Phosphorylation of Bad could be mediated by activated ERK1/2, which phosphorylates Bad at Serl22; an anti-phospho-Bad (Serl22) antibody (Pierce- Antibodies) is used to test this possibility. Total Bad is measured to determine the degree of Bad degradation. If DNA-PKcs functions as an upstream regulator of Bad, reduced phosphorylation of Bad is identified in cells treated with DNA-PKcs inhibitor NU7026 or in cells without DNA-PKcs.
  • Example 8 DNA-P cs activity is required for association of DNA-P cs with B7-H1 B7-H1 positive human breast cancer cells (MBA-MD-231) were used to examine the association of DNA-PKcs and B7-H1.
  • Immuno-precipitation assays showed that NU7026, an inhibitor of DNA-PKCs, abolished the association of B7-H1 with DNA- PKcs (FIG. 15 A), while immunofluorescence staining showed that doxorubicin-induced co-localization of B7-H1 and DNA-PKcs at the nucleus (FIG. 15B, arrow, upper right panel) was blocked by 1 ⁇ NU7026 (lower right panel).
  • the IC50 for inhibiting DNA- PKcs activity is 0.23 ⁇ .

Landscapes

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

Abstract

Materials and methods for treating potentially chemoresistant tumors (e.g., using DNA-PKcs inhibitors and anti-B7-H1 antibodies) are provided herein.

Description

TARGETING DNA-PKcs AND B7-H1 TO TREAT CANCER
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims benefit of priority from U.S. Provisional Application No. 62/027,841, filed on July 23, 2014.
TECHNICAL FIELD
This document relates to materials and methods for treating potentially chemoresistant tumors, and particularly to materials and methods for treating such tumors with DNA-PKcs inhibitors.
BACKGROUND
The development of resistance to chemotherapy and immunotherapy is a major obstacle in prolonging survival of cancer patients. The emergence of chemoresistance and immunoresistance were traditionally viewed as parallel and unrelated events, but more recent evidence indicates that overexpression of some immune checkpoint molecules may negatively influence antitumor immunity and also render tumor cells resistant to chemotherapeutic agents (Tamura et al., Leukemia : Official Journal of the Leukemia Society of America, Leukemia Research Fund, UK 2 ':464-472, 2013; Ghebeh et al, Breast Cancer Res 12:R48, 2010; and Liu et al, Molecular Cancer Ther 10:960- 971, 2011).
SUMMARY
Cancer therapies using checkpoint blockades can blunt the immune-suppressive function of ligands (e.g., B7-H1 on tumor cells) by blocking interaction with their receptors (e.g., PD-1 on T cells) (Zang and Allison, Clin Cancer Res 13:5271-5279, 2007; and Dong and Chen, JMol Med 81 :281-287, 2003). While such therapies can contribute to enhanced antitumor immunity, blocking the binding of B7-H1 to PD-1 may not overcome B7-H1 -mediated chemoresistance. The therapeutic methods described herein can be used to target B7-H1 's intrinsic signaling pathway in relation to chemoresistance. These methods are based on the results of experiments that elucidated a molecular mechanism underlying B7-H1 -mediated tumor chemoresistance, as described below, thus providing new therapeutic targets to defuse this mechanism. For example, this document is based at least in part on the discovery that DNA-PKcs is a B7-H1 binding protein, and that DNA-PKcs can be targeted to reduce B7-H1 -mediated chemoresistance. This discovery was unexpected, as DNA-PKcs is a nuclear protein involved in DNA damage repair (Collis et al., Oncogene 24:949-961, 2005), while B7-H1 is an immunoregulatory molecule mainly expressed on the cell surface of tumor cells (Dong et al, Nat Med 8:793-800, 2002).
In one aspect, this document features a method for treating a cancer patient. The method can include (a) identifying a cancer patient to be treated with a chemotherapeutic agent that causes DNA damage, (b) administering to the patient a molecule targeted to DNA-PKcs, wherein the molecule is administered in an amount sufficient to reduce the interaction of DNA-PKcs with B7-H1, and (c) administering the chemotherapeutic agent to the patient. The patient can be a human. The chemotherapeutic agent can be cisplatin, doxorubicin, SN38, paclitaxel, protein-bound paclitaxel, temozolomide, or carboplatin. The molecule targeted to DNA-PKcs can be NU7026, NU7441, IC86621, IC87102, IC87361, OK-1035, SU11752, vanillin, or IC486241, or can be an anti-DNA-PKcs antibody. The molecule targeted to DNA-PKcs and the chemotherapeutic agent can be administered simultaneously or sequentially.
In another aspect, this document features a method for treating a cancer patient, where the method can include (a) identifying the patient as having a tumor with cells that express B7-H1, and (b) administering to the patient a DNA-PKcs inhibitor and an anti- B7-H1 blocking antibody. The cancer patient can be a human. The cancer patient can be identified based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7-H1 mRNA in a sample obtained from the tumor. The cancer can be a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer. The DNA-PKcs inhibitor and the anti-B7-Hl blocking antibody can be administered to the cancer patient simultaneously or sequentially.
In another aspect, this document features a method for treating cancer, where the method can include administering a DNA-PKcs inhibitor and an anti-B7-Hl antibody to a mammal identified as having a tumor containing cells with an elevated level of B7-H1, where the DNA-PKcs inhibitor and the anti-B7-Hl antibody are administered under conditions in which the interaction of naturally-occurring B7-H1 with DNA-PKcs and the interaction of naturally-occurring B7-H1 with PD-1 or CD80 in the mammal is reduced after the administering. The mammal can be a human. The elevated level B7-H1 can be based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7-H1 m NA in a sample obtained from the tumor. The cancer can be a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer. The DNA-PKcs inhibitor and the anti-B7-Hl blocking antibody can be administered to the mammal simultaneously or sequentially.
In still another aspect, this document features a composition containing a pharmaceutically acceptable carrier and a molecule targeted to DNA-PKcs for use in treating a cancer patient who also is to be treated with a chemotherapeutic agent that causes DNA damage, wherein the composition is to be administered in an amount sufficient to reduce the interaction of DNA-PKcs with B7-H1. The patient can be a human. The chemotherapeutic agent can be cisplatin, doxorubicin, SN38, paclitaxel, protein-bound paclitaxel, temozolomide, or carboplatin. The molecule targeted to DNA- PKcs can be NU7026, NU7441, IC86621, IC87102, IC87361, OK-1035, SU11752, vanillin, or IC486241, or can be an anti-DNA-PKcs antibody. The chemotherapeutic agent and the composition containing the molecule targeted to DNA-PKcs can be for simultaneous or sequential administration.
In another aspect, this document features a composition containing a
pharmaceutically acceptable carrier and a DNA-PKcs inhibitor for use in treating cancer in a patient identified as having a tumor with cells that express B7-H1, wherein the patient also is to be treated with an anti-B7-Hl blocking antibody. The cancer patient can be a human. The cancer patient can be identified based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7-H1 m NA in a sample obtained from the tumor. The cancer can be a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer. The anti-B7-Hl blocking antibody and the composition containing the DNA-PKcs inhibitor can be for simultaneous or sequential administration.
In still another aspect, this document features a composition containing a pharmaceutically acceptable carrier and a DNA-PKcs inhibitor for use in treating cancer in a mammal identified as having a tumor containing cells with an elevated level of B7- Hl, wherein the mammal also is to be treated with an anti-B7-Hl antibody, and wherein the composition containing the DNA-PKcs inhibitor and the anti-B7-Hl antibody are to be administered under conditions in which the interaction of naturally-occurring B7-H1 with DNA-PKcs and the interaction of naturally-occurring B7-H1 with PD-1 or CD80 in the mammal are reduced after the administering. The mammal can be a human. The elevated level B7-H1 can be based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7-H1 mRNA in a sample obtained from the tumor. The cancer can be a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer. The anti-B7-Hl blocking antibody and the
composition containing the DNA-PKcs inhibitor can be for simultaneous or sequential administration.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used to practice the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
DESCRIPTION OF DRAWINGS
FIGS. 1A-1C demonstrate that B7-H1 confers tumor cell resistance to cytotoxic drugs. FIG. 1 A is a series of graphs plotting relative survival of Mock/624mel (triangles) and B7-Hl/624mel (squares) cells treated with the indicated drugs, as determined by MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H- tetrazolium) assay. The p-value for area under the curve (dose-response curves) was significant (p < 0.01) for treatment with cisplatin (left), doxorubicin (center), and SN38 (right). One of three experiments is shown. Apoptosis of tumor cells treated with doxorubicin (1.5 ug/ml, 48 hr.) was analyzed by tetramethylrhodamine ethyl ester (TMRE) and Annexin V staining (FIG. IB), and by intracellular staining for active caspase-3 (FIG. 1C). Numbers are percentages of gated population.
FIGS. 2A-2C are pictures from co-immunoprecipitation and Western blotting (WB) experiments, demonstrating that B7-H1 associates with DNA-PKcs. FIG. 2A: Anti-B7-Hl Ab, but not control Ab, co-precipitated a protein band in B7-H1 transfected 624mel cells. FIG. 2B: Co-immunoprecipitation (IP) and Western blotting (WB) using cell lysate as input. FIG. 2C: Human primary T cells were activated by
phytohemagglutinin (PHA) for 48 hours. B7-H1 was pulled down by anti-B7-Hl antibody but not control antibody. The association of B7-H1 with DNA-PKcs was detected by Western blotting using anti-DNA-PKcs antibody.
FIG. 3 is a graph plotting relative survival of tumor cells treated with a DNA- PKcs inhibitor, and showing that inhibition of DNA-PKcs reduced tumor drug resistance. Cells were pre-treated with DMSO or NU7026 (10 uM) for 1 hour before treatment with doxorubicin (Dox). The relative survival of tumor cells was measured by MTS assay 72 hours after Dox treatment. FIG. 4 is a diagram showing potential molecular mechanisms for B7-H1 -mediated tumor chemoresistance.
FIG. 5A is a cross-species alignment of the B7-H1 intracellular domains (ICD) from human (SEQ ID NO: l), chimpanzee (SEQ ID NO: l), Rhesus monkey (SEQ ID NO:2), marmoset (SEQ ID NO:3), sooty mangabey (SEQ ID NO:4), cow (SEQ ID
NO:5), pig (SEQ ID NO:6), mouse (SEQ ID NO:7), and rat (SEQ ID NO:8). Conserved residues are in red. FIG. 5B is a diagram of Flag-B7-Hl ICD domain fragments (SEQ ID NOS:9-13).
FIG. 6 is a series of pictures showing co-localization of B7-H1 and DNA-PKcs. A human breast tumor cell line (MDA-MB-231) was treated with doxorubicin (2 ug/ml) for 2 hours. Co-localization of B7-H1 and DNA-PKcs was observed in the nuclei.
Arrows or arrowheads indicate B7-H1 in plasma or in nuclei, respectively.
FIG. 7 is a picture of a blot showing B7-H1 or control (GAPDH) expression in human tumor cells (MDA-MB-231) following transfection with control or B7-H1 shRNA.
FIGS. 8A-8C demonstrate that B7-H1 enhances activation of the MAPK/ERK pathway. FIG. 8A is a picture (top) and a graph (bottom) showing the results of an antibody array assay of phosphorylation of molecules in the MAPK/ERK pathway. The levels of phospho-ERK and total ERK were analyzed by Western blotting (FIG. 8B) and flow cytometry (FIG. 8C). Numbers are mean fluorescence intensity (MFI).
FIG. 9 is a series of graphs plotting the levels of phosphorylated ERK (top graphs) and total ERK (bottom graphs) in Mock/624mel (left) and B7-Hl/624mel (right) tumor cells treated with DMSO or NU7026. Cells were treated with DMSO or NU7026 (10 uM) for 24 hours before intracellular staining for phospho-ERK or total ERK.
Numbers are MFI. Inhibition of DNA-PKcs reduced ERK activation, as indicated in the diagram on the right.
FIG. 10 is a schematic showing a potential DNA-PKcs/ERK/Bim pathway that may be used by B7-H1 to counter tumor cell apoptosis.
FIG. 11 is a schematic showing a potential DNA-PKcs/ Akt/Bad pathway that may be used by B7-H1 to counter tumor cell apoptosis. FIG. 12 contains representative nucleic acid (top) and amino acid (bottom) sequences for human B7-H1 (SEQ ID NOS: 14 and 15, respectively).
FIG. 13 contains representative nucleic acid (top) and amino acid (bottom) sequences for human PD-1 (SEQ ID NOS: 16 and 17, respectively).
FIGS. 14A and 14B contain representative nucleic acid (14A) and amino acid
(14B) sequences for human CD80 (SEQ ID NOS:18 and 19, respectively).
FIGS. 15A and 15B show that DNA-PKcs activity is required for the association of DNA-PKcs with B7-H1. MBA-MD-231 human breast cancer cells, which are positive for B7-H1, were incubated with NU7026, an inhibitor of DNA-PKCs. FIG. 15A shows an immuno-precipitation assay, demonstrating that NU7026 abolished the association of B7-H1 with DNA-PKcs. FIG. 15B is a series of pictures from an immunofluorescence assay in which the cells treated with doxorubicin, NU7026, or both, and stained for DNA-PKcs, DNA, or B7-H1. Doxorubicin-induced co-localization (arrow, upper right panel) of B7-H1 and DNA-PKcs at the nucleus was blocked by NU7026 (lower right panel).
DETAILED DESCRIPTION
This document provides methods and materials for treating cancer in patients with tumors that are chemoresistant or are at risk of becoming chemoresistant. For example, this document provides methods and materials for identifying a cancer patient (e.g., a mammal such as a human, non-human primate, cow, sheep, pig, dog, rabbit, rat, or mouse) as having a tumor that expresses B7-H1 at an elevated level, and treating the patient with a molecule that can interfere with the interaction between B7-H1 and DNA- PKcs. In some embodiments, the methods provided herein also can include treating the patient with a molecule that can interfere with the interaction between B7-H1 and PD-1 , and/or the interaction between B7-H1 and CD80 (e.g., an antibody against B7-H1, PD-1, or CD80, or a fusion protein containing a portion of PD-1 or a portion of CD80 fused to an immunoglobulin (Ig) Fc domain).
The term "elevated level" as used herein with respect to a level of B7-H1 refers to a level that is greater (e.g., 50% greater, 2-fold greater, 3-fold greater, or more than 3-fold greater) than a reference level of B7-H1. The term "reference level" as used herein with respect to B7-H1 can refer to the level of B7-H1 typically observed in cells from healthy subjects without cancer. In some embodiments, for example, a reference level of B7-H1 can be the average level of B7-H1 present in samples obtained from a random sampling of 50 humans free of cancer. In some embodiments, B7-H1 levels can be determined based on cell staining, and "elevated" and "reference" levels can set based on the percentage of evaluated cells that stain positive for B7-H1. For example, in some embodiments, samples (e.g., tumor samples) in which five percent or less (e.g., five percent, four percent, three percent, two percent, one percent, or less than one percent) of the cells stain positive for B7-H1 can be considered B7-H1 negative. In some
embodiments, samples (e.g., tumor samples) in which ten percent or more (e.g., ten percent, 20 percent, 25 percent, 30 percent, 40 percent, 50 percent, or more than 50 percent) of the cells stain positive for B7-H1 can be considered to have elevated levels of B7-H1 expression.
The presence of an elevated level of B7-H1 can be determined by measuring, for example, B7-H1 protein levels or B7-H1 nucleic acid levels. For example, the level of B7-H1 protein can be measured in a tumor sample from a mammal with cancer using cell staining, western blotting, or other immunological techniques. The level of B7-H1 expression also can be measured at the nucleic acid level, using Northern blotting, or any other method suitable for determining mRNA levels of CD274, which encodes the B7-H1 protein. In some cases, B7-H1 protein or nucleic acid levels can be measured in ascites samples, or lymphoid organ samples. It will be appreciated that levels from comparable samples are used when determining whether or not a particular level is an elevated level.
A representative example of a human B7-H1 nucleic acid has the sequence set forth in GENBANK® Accession No. AF177937 (GI No. 6708118) (SEQ ID NO: 14; Figure 12), and a representative human B7-H1 polypeptide has the sequence set forth in GENBANK® Accession No. AAF25807 (GI No. 6708119) (SEQ ID NO: 15; Figure 12).
A representative example of a human PD-1 nucleic acid can have the sequence set forth in GENBANK® Accession No. BC074740.2 (GI No. 50960296) (SEQ ID NO: 16; FIG. 13), and representative example of a human PD-1 polypeptide has the sequence set forth in GENBANK® Accession No. AAH74740.1 (GI No. 49902307) (SEQ ID NO: 17; FIG. 13).
A representative example of a human CD80 nucleic acid has the sequence set forth in NCBI Reference No. NM_005191.3 (GI No. 113722122) (SEQ ID NO: 18; FIG. 14A), and a representative example of a human CD80 polypeptide has the sequence set forth in NCBI Reference No. NP 005182.1 (GI No. 4885123) (SEQ ID NO: 19; FIG. 14B).
After the level of B7-H1 within a tumor sample from a mammal is determined, the level can be compared to a reference level, and the mammal can be classified as having or not having an elevated level of B7-H1. If the mammal is identified as having an elevated level of B7-H1, the mammal can be treated with a first molecule that inhibits the interaction between B7-H1 and DNA-PKcs. For example, a small molecule DNA- PKcs inhibitor such as NU7026, NU7441, IC86621, IC87102, IC87361, OK-1035, SU11752, vanillin, or IC486241 can be administered to the mammal. For the structures of these molecules, see, e.g., Davidson et al, Front Pharmacol 4: 1-7, 2013. In some embodiments, an anti-DNA-PKcs antibody can be administered to block the interaction between B7-H1 and DNA-PKcs. Further, B7-H1 peptides can be useful. Such peptides can be fragments of B7-H1 (e.g., fragments containing about 10-20, about 20-50, or about 50-100 amino acids) that include the DNA-PKcs binding domain, such that they can inhibit the interaction between B7-H1 and DNA-PKcs. Such peptides can be referred to as "interfering B7-H1 small peptides."
In some embodiments, the mammal also can be treated with a second molecule that inhibits the interaction between B7-H1 and PD-1 and/or the interaction between B7- Hl and CD80. Examples of such second molecules include, without limitation, antibodies (e.g., anti-B7-Hl antibodies, anti-PD-1 antibodies, or anti-CD80 antibodies), and fusion proteins (e.g., PD-1 fusion proteins or CD80 fusion proteins). Such fusion proteins can contain, for example, the extracellular domain of PD-1 fused to an IgG Fc domain, or the extracellular domain of CD80 fused to an IgG Fc domain. Binding of the fusion proteins to B7-H1 can reduce or block the ability of B7-H1 to interact with PD-1 and/or CD80. The term "antibody" includes monoclonal antibodies, polyclonal antibodies, recombinant antibodies, humanized antibodies (Jones et al. (1986) Nature 321 :522-525; Riechmann et al. (1988) Nature 332:323-329; and Presta (1992) Curr. Op. Struct. Biol. 2:593-596), chimeric antibodies (Morrison et al. (1984) Proc. Natl. Acad. Sci. USA 81 :6851-6855), multispecific antibodies (e.g., bispecific antibodies) formed from at least two antibodies, and antibody fragments. The term "antibody fragment" comprises any portion of the afore -mentioned antibodies, such as their antigen binding or variable regions. Examples of antibody fragments include Fab fragments, Fab' fragments, F(ab')2 fragments, Fv fragments, diabodies (Hollinger et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448), single chain antibody molecules (Pluckthun in: The Pharmacology of
Monoclonal Antibodies 113, Rosenburg and Moore, eds., Springer Verlag, N.Y. (1994), 269-315) and other fragments as long as they exhibit the desired capability of binding to their target (e.g., B7-H1, PD-1, or CD80).
Examples of anti-human B7-H1 antibodies include, without limitation, anti- human B7-H1 antibodies commercially available from Biolegend (e.g., Catalog No.
329701 or 329702; San Diego, CA) or eBioscience (e.g., Catalog No. 14-5983-80 or 14- 5983-82).
Examples of anti-human PD-1 antibodies include, without limitation, anti-human PD-1 antibodies commercially available from Biolegend (e.g., Catalog No. 329904 or 329905) or eBioscience (Catalog No. 12-2799-42; San Diego, CA).
Examples of anti-human CD80 antibodies include, without limitation, anti-human CD8 antibodies commercially available from Biolegend (e.g., Catalog No. 305201 or 305202) or eBioscience (e.g., Catalog No. 14-0809-80 or 14-0809-82).
The term "antibody," as used herein, also includes antibody-like molecules that contain engineered sub-domains of antibodies or naturally occurring antibody variants. These antibody-like molecules may be single-domain antibodies such as VH-only or VL- only domains derived either from natural sources such as camelids (Muyldermans et al. (2001) Rev. Mol. Biotechnol. 14 211-1Q2) or through in vitro display of libraries from humans, camelids or other species (Holt et al. (2003) Trends Biotechnol. 21 :484-90). In certain embodiments, the polypeptide structure of the antigen binding proteins can be based on antibodies, including, but not limited to, minibodies, synthetic antibodies (sometimes referred to as "antibody mimetics"), human antibodies, antibody fusions (sometimes referred to as "antibody conjugates"), and fragments thereof, respectively.
An "Fv fragment" is the minimum antibody fragment that contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy chain variable domain and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three CDR's of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six CDR's confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDR's specific for an antigen) has the ability to recognize and bind the antigen, although usually at a lower affinity than the entire binding site. The "Fab fragment" also contains the constant domain of the light chain and the first constant domain (CHI) of the heavy chain. The "Fab fragment" differs from the "Fab' fragment" by the addition of a few residues at the carboxy terminus of the heavy chain CHI domain, including one or more cysteines from the antibody hinge region. The "F(ab')2 fragment" originally is produced as a pair of "Fab' fragments" which have hinge cysteines between them. Methods of preparing such antibody fragments, such as papain or pepsin digestion, are known to those skilled in the art.
An antibody can be of the IgA-, IgD-, IgE, IgG- or IgM-type, including IgG- or IgM-types such as, without limitation, IgGl-, IgG2-, IgG3-, IgG4-, IgMl- and IgM2- types. For example, in some cases, an antibody can be of the IgGl-, IgG2- or IgG4- type.
In some embodiments, antibodies as used in the methods described herein can be fully human or humanized antibodies. Human antibodies can avoid certain problems associated with xenogeneic antibodies, such as antibodies that possess murine or rat variable and/or constant regions. First, because the effector portion is human, it can interact better with other parts of the human immune system, e.g., to destroy target cells more efficiently by complement-dependent cytotoxicity or antibody-dependent cellular cytotoxicity. Second, the human immune system should not recognize the antibody as foreign. Third, the half-life in human circulation will be similar to naturally occurring human antibodies, allowing smaller and less frequent doses to be given. Methods for preparing human antibodies are known in the art.
In addition to human antibodies, "humanized" antibodies can be used, and can have many advantages. Humanized antibodies generally are chimeric or mutant monoclonal antibodies from mouse, rat, hamster, rabbit or other species, bearing human constant and/or variable region domains or specific changes. Techniques for generating humanized antibodies are well known to those of skill in the art. For example, controlled rearrangement of antibody domains joined through protein disulfide bonds to form new, artificial protein molecules or "chimeric" antibodies can be utilized (Konieczny et al. (1981) Haematologia (Budap.) 14:95). Recombinant DNA technology can be used to construct gene fusions between DNA sequences encoding mouse antibody variable light and heavy chain domains and human antibody light and heavy chain constant domains (Morrison et al. (1984) Proc. Natl. Acad. Sci. USA 81 :6851).
DNA sequences encoding antigen binding portions or complementarity determining regions (CDR's) of murine monoclonal antibodies can be grafted by molecular means into DNA sequences encoding frameworks of human antibody heavy and light chains (Jones et al. (1986) Nature 321 :522; Riechmann et al. (1988) Nature 332:323). Expressed recombinant products are called "reshaped" or humanized antibodies, and comprise the framework of a human antibody light or heavy chain and antigen recognition portions, CDR's, of a murine monoclonal antibody.
Other methods for designing heavy and light chains and for producing humanized antibodies are described in, for example, U.S. Patent Nos. 5,530,101; 5,565,332;
5,585,089; 5,639,641; 5,693,761; 5,693,762; and 5,733,743. Yet additional methods for humanizing antibodies are described in U.S. Patent Nos. 4,816,567; 4,935,496;
5,502,167; 5,558,864; 5,693,493; 5,698,417; 5,705,154; 5,750,078; and 5,770,403, for example.
Molecules that interfere with the interaction between B7-H1 and DNA-PKcs, the interaction between B7-H1 and PD-1, and/or the interaction between B7-H1 and CD80, as described herein (e.g., small molecule inhibitors of DNA-PKcs, antibodies against B7- HI, DNA-PKcs, PD-1, and CD80, and fusion proteins containing portions of PD-1 or CD80 linked to an Ig Fc domain), can be incorporated into pharmaceutical compositions for treatment of cancer. Thus, this document also provides the use of such molecules in the manufacture of medicaments for treating cancer. The compositions further can include one or more pharmaceutically acceptable carriers, diluents and/or adjuvants.
A "pharmaceutically acceptable carrier" (also referred to as an "excipient" or a "carrier") is a pharmaceutically acceptable solvent, suspending agent, stabilizing agent, or any other pharmacologically inert vehicle for delivering one or more therapeutic compounds to a subject, which is nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed. Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, and other pertinent transport and chemical properties, when combined with one or more of therapeutic compounds and any other components of a given pharmaceutical composition. Typical pharmaceutically acceptable carriers include, by way of example and not limitation: water, saline solution, binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose), fillers (e.g., lactose and other sugars, gelatin, or calcium sulfate), lubricants (e.g., starch, polyethylene glycol, or sodium acetate), disintegrates (e.g., starch or sodium starch glycolate), and wetting agents (e.g., sodium lauryl sulfate). Pharmaceutically acceptable carriers also include aqueous pH buffered solutions or liposomes (small vesicles composed of various types of lipids, phospholipids and/or surfactants which are useful for delivery of a drug to a mammal). Further examples of pharmaceutically acceptable carriers include buffers such as phosphate, citrate, and other organic acids, antioxidants such as ascorbic acid, low molecular weight (less than about ten residues) polypeptides, proteins such as serum albumin, gelatin, or immunoglobulins, hydrophilic polymers such as polyvinylpyrrolidone, amino acids such as glycine, glutamine, asparagine, arginine or lysine, monosaccharides, disaccharides, and other carbohydrates including glucose, mannose or dextrins, chelating agents such as EDTA, sugar alcohols such as mannitol or sorbitol, salt-forming counterions such as sodium, and/or nonionic surfactants such as TWEEN™, polyethylene glycol (PEG), and PLURONICS™. Pharmaceutical compositions can be formulated by mixing one or more active agents (e.g., a DNA-PKcs inhibitor or an anti-B7-Hl blocking antibody) with one or more physiologically acceptable carriers, diluents, and/or adjuvants, and optionally other agents that are usually incorporated into formulations to provide improved transfer, delivery, tolerance, and the like. Pharmaceutical compositions can be formulated, e.g., in lyophilized formulations, aqueous solutions, dispersions, or solid preparations, such as tablets, dragees or capsules. A multitude of appropriate formulations can be found in the formulary known to pharmaceutical chemists: Remington's Pharmaceutical Sciences (18th ed, Mack Publishing Company, Easton, PA (1990)), particularly Chapter 87 by Block, Lawrence, therein. These formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTIN™), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. Any of the foregoing mixtures may be appropriate in treatments and therapies as described herein, provided that the active agent in the formulation is not inactivated by the formulation and the formulation is physiologically compatible and tolerable with the route of
administration. See, also, Baldrick (2000) Regul. Toxicol. Pharmacol. 32:210-218; Wang (2000) Int. J. Pharm. 203:1-60; Charman (2000) J. Pharm. Sci. 89:967-978; and Powell et al. (1998) PDA J. Pharm. Sci. Technol. 52:238-311), and the citations therein for additional information related to formulations, excipients and carriers well known to pharmaceutical chemists.
Pharmaceutical compositions include, without limitation, solutions, emulsions, aqueous suspensions, and liposome-containing formulations. These compositions can be generated from a variety of components that include, for example, preformed liquids, self-emulsifying solids and self-emulsifying semisolids. Emulsions are often biphasic systems comprising of two immiscible liquid phases intimately mixed and dispersed with each other; in general, emulsions are either of the water-in-oil (w/o) or oil-in-water (o/w) variety. Emulsion formulations have been widely used for oral delivery of therapeutics due to their ease of formulation and efficacy of solubilization, absorption, and
bioavailability.
Compositions and formulations can include sterile aqueous solutions, which also can contain buffers, diluents and other suitable additives (e.g., penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers). Compositions additionally can contain other adjunct components conventionally found in
pharmaceutical compositions. Thus, the compositions also can include compatible, pharmaceutically active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or additional materials useful in physically formulating various dosage forms of the compositions provided herein, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. Furthermore, the composition can be mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings, and aromatic substances. When added, however, such materials should not unduly interfere with the biological activities of the polypeptide components within the compositions provided herein. The formulations can be sterilized if desired.
In some embodiments, a composition containing a DNA-PKcs inhibitor and/or an antibody or fusion protein as provided herein (e.g., an anti-B7-H7, anti-DNA-PKcs, anti- PD-1, or anti-CD80 antibody, or a PD-l-Fc or CD80-Fc fusion protein) can be in the form of a solution or powder with or without a diluent to make an injectable suspension. The composition may contain additional ingredients including, without limitation, pharmaceutically acceptable vehicles, such as saline, water, lactic acid, mannitol, or combinations thereof, for example.
Methods for using an agent (e.g., a small molecule inhibitor of DNA-PKcs, an antibody against B7-H1, DNA-PKcs, PD-1, or CD80, or a PD-l-Fc or CD80-Fc fusion protein) or a composition containing such an agent to treat cancer patients also are provided herein. The methods can include, for example, administering an agent or composition to a subject identified as being in need thereof. In some embodiments, a method as provided herein can further include steps such as identifying a mammal (e.g., a human cancer patient) that is to be treated with a chemotherapeutic agent (e.g., cisplatin, doxorubicin, SN38, paclitaxel, protein-bound paclitaxel (e.g., ABRAXANE®), temozolomide (e.g., TEMODAR®), or carboplatin) that causes DNA damage, or identifying a mammal as having a tumor with cells that express B7-H1. For example, a method can include identifying a mammal to be treated with a chemotherapeutic agent that causes DNA damage, and treating the mammal with an agent that inhibits the interaction between B7-H1 and DNA-PKcs. In some cases, a method can further include the step of administering the chemotherapeutic agent that causes DNA damage.
Any appropriate method can be used to administer a molecule as described herein to a mammal. Administration can be, for example, parenteral (e.g., by subcutaneous, intrathecal, intraventricular, intramuscular, or intraperitoneal injection, or by intravenous drip). Administration can be rapid (e.g., by injection) or can occur over a period of time (e.g., by slow infusion or administration of slow release formulations). In some embodiments, administration can be topical (e.g., transdermal, sublingual, ophthalmic, or intranasal), pulmonary (e.g., by inhalation or insufflation of powders or aerosols), or oral. In addition, a composition containing an antibody or fusion protein as described herein can be administered prior to, after, or in lieu of surgical resection of a tumor.
Compositions containing an antibody (e.g., an anti-B7-Hl antibody, an anti- DNA-PKcs antibody, an anti-PD-1 antibody, or an anti-CD80 antibody), a small molecule (e.g., NU7026) or a fusion protein (e.g., a PD-l-Fc fusion or a CD80-Fc fusion) can be administered to a mammal in any appropriate amount, at any appropriate frequency, and for any appropriate duration effective to achieve a desired outcome (e.g., to increase progression- free survival, or to reduce progression of the cancer).
Combination therapies, in which a DNA-PKcs inhibitor and an anti-B7-Hl blocking antibody are administered to a mammal, can be particularly useful, as such therapies can target both chemoresistance and immunoresistance. For example, a first composition containing a DNA-PKcs inhibitor and a second composition containing an anti-B7-Hl antibody can be administered, either simultaneously (e.g., via simultaneous
administration of separate compositions, or via administration of a composition containing both agents), or sequentially. In some embodiments, a DNA-PKcs inhibitor and an anti-B7-Hl antibody can be administered to a mammal having cancer to reduce the progression rate of the cancer by 5, 10, 25, 50, 75, 100, or more percent. For example, the progression rate can be reduced such that no additional cancer progression is detected. In some embodiments, a DNA- PKcs inhibitor and an anti B7-H1 antibody can be administered to a mammal having cancer under conditions where progression-free survival is increased (e.g., by 5, 10, 25, 50, 75, 100, or more percent) as compared to the median progression-free survival of corresponding mammals having untreated cancer or the median progression-free survival of corresponding mammals having cancer and treated with other therapies (e.g., immune or chemotherapeutic agents alone). Progression-free survival can be measured over any length of time (e.g., one month, two months, three months, four months, five months, six months, or longer). Any appropriate method can be used to determine whether or not the progression rate of cancer is reduced. For skin cancer (e.g., melanoma), for example, the progression rate can be assessed by imaging tissue at different time points and
determining the amount of cancer cells present. The amounts of cancer cells determined within tissue at different times can be compared to determine the progression rate. After treatment as described herein, the progression rate can be determined again over another time interval. In some cases, the stage of cancer after treatment can be determined and compared to the stage before treatment to determine whether or not the progression rate has been reduced.
An effective amount of a composition containing an antibody as provided herein can be any amount that reduces a symptom of the condition being treated, without significant toxicity. With cancer, for example, an effective amount can reduce the progression rate of the cancer, increase the progression-free survival rate, or increase the median time to progression. Optimum dosages can vary depending on the relative potency of individual polypeptides (e.g., antibodies and fusion proteins), and can generally be estimated based on ECso found to be effective in in vitro and in vivo animal models. Typically, dosage is from 0.01 μg to 100 g per kg of body weight. For example, an effective amount of an antibody or fusion protein can be from about 1 mg/kg to about 100 mg/kg (e.g., about 5 mg/kg, about 10 mg/kg, about 20 mg/kg, about 50 mg/kg, or about 75 mg/kg). If a particular subject fails to respond to a particular amount, then the amount of the antibody can be increased by, for example, two fold. After receiving this higher concentration, the subject can be monitored for both responsiveness to the treatment and toxicity symptoms, and adjustments made accordingly. The effective amount can remain constant or can be adjusted as a sliding scale or variable dose depending on the mammal's response to treatment. Various factors can influence the actual effective amount used for a particular application. For example, the frequency of administration, duration of treatment, use of multiple treatment agents, route of administration, and severity of the clinical condition may require an increase or decrease in the actual effective amount administered.
The frequency of administration can be any frequency that reduces the
progression rate of cancer, increases the progression-free survival rate, or increases the median time to progression without producing significant toxicity to the mammal. For example, the frequency of administration can be once or more daily, biweekly, weekly, monthly, or even less. The frequency of administration can remain constant or can be variable during the duration of treatment. A course of treatment can include rest periods. For example, a composition containing one or more agents (e.g., a small molecule, antibody, or fusion protein as provided herein) can be administered over a two week period followed by a two week rest period, and such a regimen can be repeated multiple times. As with the effective amount, various factors can influence the actual frequency of administration used for a particular application. For example, the effective amount, duration of treatment, use of multiple treatment agents, route of administration, and severity of the cancer may require an increase or decrease in administration frequency.
An effective duration for administering a composition provided herein can be any duration that reduces the progression rate of cancer, increases the progression-free survival rate, or increases the median time to progression without producing significant toxicity to the mammal. Thus, the effective duration can vary from several days to several weeks, months, or years. In general, the effective duration for the treatment of cancer can range in duration from several weeks to several months. In some cases, an effective duration can be for as long as an individual mammal is alive. Multiple factors can influence the actual effective duration used for a particular treatment. For example, an effective duration can vary with the frequency of administration, effective amount, use of multiple treatment agents, route of administration, and severity of the cancer.
After administering one or more agents as provided herein to a mammal, the mammal can be monitored to determine whether or not the treatment was effective. For example, a mammal can be assessed after treatment to determine whether or not the progression rate of the cancer has been reduced. Any method, including those that are standard in the art, can be used to assess progression and survival rates.
The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.
EXAMPLES
Example 1 - B7-H1 renders tumor cells resistant to drugs that cause DNA damage Experiments were conducted to test whether B7-H1 -expressing tumors would show resistance to drugs that cause DNA damage. Along with cisplatin, which crosslinks DNA, doxorubicin (a topoisomerase II inhibitor that leads to generation of free radicals) and SN38 (a topoisomerase I inhibitor) were tested. Drug sensitivity in B7-H1- and Mock-transfected 624mel melanoma cells was examined in cultures with varying concentrations of the drugs. After 72 hours of culture, tumor cell survival was measured with a MTS assay (Pei et al, Cancer Cell 16:259-266, 2009). As shown in FIG. 1A, B7- Hl/624mel cells were more resistant than Mock/624mel cells to cisplatin, doxorubicin, and SN38. Consistent with a previous pharmacogenetic analysis, these data supported the idea that B7-H1 renders tumor cells resistant to cytotoxic chemotherapeutic agents. Since a mechanism of action of most cytotoxic drugs is to induce apoptosis (Sedletska et al. , Curr Med Chem Anti-cancer Agents 5:251 -265 , 2005 ; and Wang et al. , J Biol Chem 279:25535-25543, 2004), apoptosis of B7-Hl/624mel and Mock/624mel tumor cells was measured and compared after treatment with doxorubicin. Specifically, apoptosis was measured based on the binding of Annexin V (AV) and the levels of
tetramethylrhodamine ethyl ester (TMRE), a marker for mitochondria membrane potential (Jayaraman, J Immunolog Meth 306:68-79, 2005). As shown in FIG. IB, B7- Hl/624mel cells had 5 -fold less apoptosis compared to Mock/624mel cells following treatment with doxorubicin. The level of active caspase-3 (an executive molecule of apoptosis) also was lower in B7-Hl/624mel cells compared with Mock/624mel cells (FIG.1C). Thus, B7-H1 may render tumor cells resistant to cytotoxic drugs by reducing their apoptotic potential to cytotoxic condition.
Example 2 - B7-H1 is associated with DNA-P cs
A co-immunoprecipitation (Co-IP) assay with tumor cell lysates using anti-B7-Hl monoclonal antibody resulted in identification of a 450-kDa protein band (FIG. 2A). The protein band identified was larger than and distinct from the membrane proteins PD-1 (55kDa) and CD80 (65kDa) that also have been reported to interact with B7-H1. The band was excised from the gel, and mass spectroscopy revealed that the major component was DNA-PKcs (DNA dependent protein kinase, catalytic subunit). To confirm the association of B7-H1 with DNA-PKcs, Co-IP assays were performed using anti-DNA- PKcs and anti-B7-Hl monoclonal antibodies in a tumor cell line that constitutively expresses B7-H1 (Karpas299) and in a B7-Hl-transfected tumor cell line (B7- Hl/624mel). Both DNA-PKcs and B7-H1 were pulled down by their respective antibodies in B7-H1 transfected 624mel cells, as well as in the endogenous B7-H1- positive Karpas299 cells (FIG. 2B), suggesting that DNA-PKcs and B7-H1 indeed associate in vivo. Significantly, an association of DNA-PKcs and B7-H1 was detected in activated (known to up-regulate B7-H1 expression) but not resting human primary T cells (FIG. 2C), suggesting the association of B7-H1 with DNA-PKcs may be a general biological interaction that is not limited to tumor cells.
Example 3 - Inhibition of DNA-PKcs activity abolishes B7-H1 -mediated
chemoresistance
To test whether the association of B7-H1 with DNA-PKcs is a mechanism of drug resistance, the effects of NU7026 on drug sensitivity of B7-H1- or Mock- transfected 624mel cells were tested. NU7026 is an ATP-competitive DNA-PKcs inhibitor, and 10 uM NU7026 completely inhibits DNA-PKcs activity (Veuger et al, Cancer Res 63:6008- 6015, 2003; and Willmore et al, Blood 103:4659-4665, 2004). Using the same dose (10 uM) of NU7026, experiments were conducted to determine whether NU7026 would affect the drug sensitivity of B7-Hl/624mel cells. As shown in FIG. 3, B7-Hl/624mel cells demonstrated resistance to doxorubicin, compared with Mock/624mel cells. After pretreatment with NU7026, however, B7-Hl/624mel cells lost their resistance to doxorubicin and had comparable drug sensitivity with Mock/624mel cells, suggesting that DNA-PKcs may contribute to B7-H1 -mediated drug resistance (FIG. 3). NU7026 also increased the drug sensitivity of Mock/624mel cells, suggesting DNA-PKcs may be a downstream element of B7-H1 signaling pathway.
Taken together, these preliminary data strongly suggest that DNA-PKcs plays a key role in B7-H1 -mediated tumor chemoresistance, and that B7-H1 is a DNA damage checkpoint molecule that, association with DNA-PKcs, promotes tumor survival via DNA damage repair and activation of pro-survival signaling pathways, thus countering cytotoxic effects of chemotherapeutic agents (FIG. 4). To test this hypothesis, experiments are conducted to (1) define the role of B7-H1 interaction with DNA-PKcs in DNA damage repair and (2) define the role of B7-H1 interaction with DNA-PKcs in the activation of pro-survival signaling pathways. Conceptually, these studies extend the role of B7-H1, originally defined as an immune checkpoint molecule, to a DNA damage checkpoint molecule of tumor cells upon treatment with cytotoxic drugs.
Example 4 - Identifying the binding sites(s) of B7-H1 in association with DNA-PKcs The anti-apoptosis function of B7-H1 has been identified in its intracellular domain (ICD) in tumor cells (Azuma et al, Blood 111 :3635-3643, 2008). It is possible that B7-H1 uses its ICD in association with DNA-PKcs to achieve its anti-apoptosis function in tumor cells. The B7-H1 ICD has eight amino acid residues that are conserved across species (FIG. 5A, red font). Based on their distribution and locations, these amino acids can be grouped in three regions (I, II, and III).
To test which region is required for B7-H1 to associate with DNA-PKcs, individual region deletions or truncations are made for the B7-H1 ICD, as shown in FIG. 5B. Next, single residue mutations of each conserved amino acid in the required region are generated to test which amino acid is required for binding. Candidate amino acids are changed to Ala in this mutagenesis assay. Flag-B7-Hl ICD fusion proteins carrying regional truncations or individual mutations are produced. B7-H1 -negative 624mel tumor cells are transfected with Flag-B7-Hl ICD, and are used in Co-IP (using anti-Flag antibody in pull-down assays) to assess the association of mutant or truncated B7-H1 ICD with DNA-PKcs. It is noted that the transmembrane domain of B7-H1 may be needed if the association with DNA-PKcs requires anchoring of B7-H1 to the cytoplasm or nuclear membrane. In this case, the transmembrane domain of B7-H1 is included in the Flag-B7-Hl fusion protein for the Co-IP assay.
Example 5 - Identifying the effects of B7-H1 on DNA repair function of DNA-PKcs B7-H1 undergoes redistribution from the cell surface into the nucleus in human breast tumor cells upon treatment with the cytotoxic drug, doxorubicin (Ghebeh et al, supra). Using the same model (MDA-MB-231 , B7-H1 positive cell line), studies were conducted to examine whether translocation of B7-H1 results in close association with DNA-PKcs in the nucleus following drug treatment. The results shown in FIG. 6 demonstrate that before treatment with cytotoxic drug, DNA-PKcs was mainly localized in nuclei and B7-H1 was mainly localized in cytoplasm of tumor cells (arrows). After treatment, B7-H1 was enriched in the nuclei, and co-localization of B7-H1 and DNA- PKcs was observed in the nuclei (arrow heads in FIG. 6), suggesting that B7-H1 may be recruited to the nucleus to regulate DNA-PKcs activity in responses to DNA damage caused by chemotherapeutic drugs.
The phosphorylation of DNA-PKcs is required for rejoining of DNA double- strand breaks (DSBs) (Chan et al, Genes Dev 16:2333-2338, 2002), raising the possibility that B7-H1 stimulates DNA-PKcs activity to repair DNA DSBs. To test this, gamma-H2AX (γΗ2ΑΧ) is used. γΗ2ΑΧ has been widely used as a sensitive marker for DSBs (Banath et al, BMC Cancer 10:4, 2010; Mah et al, Leukemia 24:679-686, 2010; and Yuan et al, FEBS Lett 584:3717-3724, 2010). Nuclear γΗ2ΑΧ is measured by anti- phospho-H2AX (Serl39) (Cell Signaling, clone 20E3) in confocal immunofluorescence (IF) assays. A time course examination is conducted, with comparison of the distribution (nuclear foci or nuclear periphery) of γΗ2ΑΧ between B7-H1 positive and B7-H1 negative (knockdown) tumor cells upon treatment with cytotoxic drug (doxorubicin or cisplatin). As shown in FIG. 7, a method to knockdown B7-H1 in human tumor cells has been established. Five different fields are scored for γΗ2ΑΧ distribution and expression. Fisher's exact test is used to calculate the p-value. To quantify γΗ2ΑΧ expression and its relation with a specific cell cycle phase, a flow cytometry-based assay is used to measure the nuclear levels of γΗ2ΑΧ (Kataoka et al, J Rad Res 47:245-257, 2006) while measuring DNA content with propidium iodide (Solier et al, Mol Cell Biol 29:68-82, 2009).
To measure DNA-PKcs activity, anti-phospho-DNA-PKcs (Thr2609) antibody is used (BioLegend, clone 10B1) to detect auto-phosphorylated DNA-PKcs by Western blotting following treatment with cytotoxic agents over a course of time. To directly measure the DNA repair function of DNA-PKcs, an EJ5GFP-based chromosomal break reporter (from the Addgene plasmid repository) is used to measure DNA-PKcs mediated non-homologous end joining (NHEJ; Bennardo et al, PLoS Genet 4:el000110, 2008; and Gunn et al, J Biol Chem 286:42470-42482, 2011). EJ5GFP contains a promoter that is separated from a GFP coding cassette by a puro gene that is flanked by two I-Scel sites that are in the same orientation. Once the puro gene is excised by NHEJ repair of the two I-Scel-induced DSBs, the promoter is joined to the rest of the expression cassette, leading to restoration of the GFP+ gene. By measuring the frequency of GFP+ cells, the function of DNA-PKcs in DNA repair is determined. Briefly, reporter plasmids are transfected into B7-H1 -positive or -negative tumor cells, followed by treatment with one or more cytotoxic agents. The frequency of GFP+ cells is determined and compared between B7- Hl -positive and negative-tumor cells using flow cytometry.
If B7-H1 in association with DNA-PKcs stimulates the DNA repair function of
DNA-PKcs, expression of γΗ2ΑΧ (a sign of DNA DSBs) will decrease more slowly in B7-H1 -positive tumor cells than in B7-H1 -negative tumor cells, as a result of sufficient DNA repair by DNA-PKcs. Nuclear foci distributed throughout the nucleus are the most common distribution of γΗ2ΑΧ, but γΗ2ΑΧ expression at the nuclear periphery has been reported in early stage apoptotic cells (Solier et al, supra). Since DNA-PKcs also phosphorylates H2AX during apoptotic DNA fragmentation (Mukherjee et al., DNA Repair 5:575-590, 2006), the pattern of γΗ2ΑΧ distribution predicts whether B7-H1 regulates the function of DNA-PKcs in DNA repair or in DNA fragmentation. Example 6 - Defining the role of B7-H1 interaction with DNA-PKcs in the ER /Bim
pathway
DNA-PKcs as a kinase activates Akt and ERK pro-survival signaling pathways (Dragoi et al, EMBO J 24 ΊΊ9-Ί%9, 2005; and (Yotsumoto et al, J Immunol 180:809- 816, 2008). The direct effects of DNA-PKcs on ERK activation are not clear, however. When the relative phosphorylation levels of proteins involved in the MAPK/ERK pathway in B7-Hl/624mel cells and Mock /624mel cells was evaluated using antibody arrays (R&D Systems, Minneapolis, MN), phosphorylation of mTOR (an element in Akt pathway) and RSK1/2 (an element in ERK pathway) were significantly increased among B7-Hl/624mel cells compared with Mock/624mel cells (FIG. 8A). Accordingly, phosphorylation of ERKl/2 and GSK-3 (a downstream element of Akt; Bodine, Med Science Sports Exercise 38: 1950-1957, 2006) also were increased. Since increased phosphorylation of Akt in the nucleus is associated with B7-H1 re-distribution to the nucleus (Ghebeh et al, supra), and because B7-H1 and DNA-PKcs co-localize in the nucleus (FIG. 6), it is possible that activation of Akt pathway could be regulated by DNA-PKcs in association with B7-H1. Increased ERKl/2 activation was confirmed by Western blotting (FIG. 8B) and an intracellular flow cytometry assay that showed a >2- fold increase of phospho-ERKl/2 (FIG. 8C). In both assays, total ERK levels remained comparable between B7-Hl/624mel and Mock/624mel, suggesting that B7-H1 regulates the activation of ERKl/2 rather than the ERKl/2 protein level. Based on these data, it is possible that upon treatment with cytotoxic drugs, B7-H1 is recruited to the nucleus where it functions as a platform for DNA-PKcs to activate ERK and Akt signaling pathways thereby suppressing tumor cell apoptosis.
To test the role of DNA-PKcs in activation of the MAPK signal cascade, tumor cells were treated with NU7026, a DNA-PKcs specific inhibitor. The results shown in FIG. 9 demonstrate that NU7026 dramatically decreased (~2-fold) the activation of ER 1/2 in B7-Hl/624mel cells but not Mock/624mel cells, suggesting DNA-PKcs may be a positive regulator of ERK1/2, and that DNA-PKcs may require B7-H1 in activation of ERK1/2. As a mechanism of action of ERK1/2 in drug resistance, ERK1/2
phosphorylates Bim and enhances degradation of Bim in tumor cells (Gillings, et al., FEBS J 276:6050-6062, 2009; and Luciano et al, Oncogene 22:6785-6793, 2003). The findings shown in FIG. 9 suggested that B7-H1 in association with DNA-PKcs enhances the activation of ERK1/2 that promotes Bim degradation by phosphorylation, thus countering tumor cell apoptosis (diagrammed in FIG.10).
To test this, the phosphorylation of Bim in the presence of DNA-PKcs inhibitor (NU7026) is examined in endogenous B7-H1 -positive MDA-MB-231 human tumor cells (FIG. 7) following treatment with cytotoxic drugs. The IC50 for NU7026 to inhibit DNA-PKcs is 0.23 uM (while the IC50 for inhibition of PI3K is 13 uM). To specifically examine the effects of NU7026 on Bim phosphorylation, NU7026 is titrated gradually from 10 uM to 0.23 uM (using the same volume of DMSO solvent as a control). To further confirm the role of DNA-PKcs in Bim phosphorylation, DNA-PKcs knockout cell lines (Wu et al., J Immunol 174:934-941, 2005) or DNA-PKcs knockdown cell lines are used to measure the levels of ERK1/2 activation in the absence of DNA-PKcs proteins. In both cases, the ERK1/2 inhibitor U0126 (which inhibits MEK, an upstream kinase of ERK1/2) is used to confirm that phosphorylation of Bim is mediated by ERK1/2 activation. To specifically determine whether DNA-PKcs requires B7-H1 to regulate ERK1/2 activation and Bim phosphorylation, the effects of DNA-PKcs inhibitor are examined in B7-H1 knockdown tumor cells (FIG. 7). To measure phosphorylation of Bim, an electrophoretic mobility shift assay of Bim in Western blot is used (Luciano et al, supra; and O'Reilly et al, J Immunol 183:261-269, 2009). In this assay,
phosphorylated Bim migrates more slowly than non-phosphorylated Bim. To confirm that the slower migrating band of Bim is due to phosphorylation of Bim, lysates are incubated with lambda phosphatase (λ-PPase, 15 ug/200U for 1 h), which de- phosphorylates modified serine, threonine and tyrosine residues. To specifically identify the phosphorylation of Bim, an anti-phospho-Bim (Ser69 in human) antibody (Cell Signaling Tech. #4581) is used in a Western blotting assay, since phosphorylation of Bim at Ser69 by ERK/1/2 promotes Bim degradation (Luciano et al., supra). Total Bim also is measured to determine to what degree Bim is undergoing degradation. Accordingly, the apoptosis of tumor cells is measured by flow cytometry using antibody against activated caspase-3 as described in FIG. 1.
Example 7 - Defining the role of B7-H1 interaction with DNA-PKcs in Akt/Bad pathway
DNA-PKcs enhances Akt activity (Dragoi et al., supra; and Feng et al., J Biol Chem 279:41189-41196, 2004). The increase of mTOR activation in B7-Hl/624mel cells (FIG. 8) and the association of B7-H1 with DNA-PKcs (FIG. 2) suggest that Akt could be a downstream element in the B7-Hl/DNA-PKcs pathway. Since activated Akt promotes cell survival by phosphorylating Bad, and blocks Bad-induced death (Datta et al, Cell 91 :231-241, 1997; and del Peso et al, Science 278:687-689, 1997), it is possible that B7- Hl in association with DNA-PKcs enhances the activation of Akt that decreases Bad via phosphorylation, thus countering tumor cell apoptosis (FIG. 11). To test this hypothesis, the phosphorylation of Bad is examined in B7-H1 positive or negative tumor cells following treatment with cytotoxic agents in the presence of DNA-PKcs inhibitor (NU7026), or in DNA-PKcs knockout cells. In both cases, to confirm that activated Akt mediates the phosphorylation of Bad, the Akt inhibitor MK2206 (Merck) is used to directly inhibit the enzyme activity of activated Akt (Merck data sheet) in the studies. To specifically identify the phosphorylation of Bad, anti-phospho-Bad (Serl36) antibody (Cell Signaling Technology, clone 185D10) is used in a Western blotting assay, since Akt preferentially phosphorylates Bad at Serl36 in tumor cells (Hayakawa et al., Cancer Res 60:5988-5994, 2000). Phosphorylation of Bad could be mediated by activated ERK1/2, which phosphorylates Bad at Serl22; an anti-phospho-Bad (Serl22) antibody (Pierce- Antibodies) is used to test this possibility. Total Bad is measured to determine the degree of Bad degradation. If DNA-PKcs functions as an upstream regulator of Bad, reduced phosphorylation of Bad is identified in cells treated with DNA-PKcs inhibitor NU7026 or in cells without DNA-PKcs.
Taken together, these studies provide new insights for overcoming tumor chemoresistance, and a new direction for future combined chemotherapy and immunotherapy targeting B7-H1 expressed by aggressive or refractory human tumor cells.
Example 8 - DNA-P cs activity is required for association of DNA-P cs with B7-H1 B7-H1 positive human breast cancer cells (MBA-MD-231) were used to examine the association of DNA-PKcs and B7-H1. Immuno-precipitation assays showed that NU7026, an inhibitor of DNA-PKCs, abolished the association of B7-H1 with DNA- PKcs (FIG. 15 A), while immunofluorescence staining showed that doxorubicin-induced co-localization of B7-H1 and DNA-PKcs at the nucleus (FIG. 15B, arrow, upper right panel) was blocked by 1 μΜ NU7026 (lower right panel). The IC50 for inhibiting DNA- PKcs activity is 0.23 μΜ.
OTHER EMBODIMENTS
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A method for treating a cancer patient, comprising:
(a) identifying a cancer patient to be treated with a chemotherapeutic agent that causes DNA damage;
(b) administering to the patient a molecule targeted to DNA-PKcs, wherein the molecule is administered in an amount sufficient to reduce the interaction of DNA-PKcs with B7-Hl; and
(c) administering the chemotherapeutic agent to the patient.
2. The method of claim 1 , wherein the patient is a human.
3. The method of claim 1, wherein the chemotherapeutic agent comprises cisplatin, doxorubicin, SN38, paclitaxel, protein-bound paclitaxel, temozolomide, or carboplatin.
4. The method of claim 1, wherein the molecule targeted to DNA-PKcs comprises NU7026, NU7441, IC86621, IC87102, IC87361, OK-1035, SU11752, vanillin, or IC486241.
5. The method of claim 1, wherein the molecule targeted to DNA-PKcs is an anti- DNA-PKcs antibody.
6. The method of claim 1 , wherein the molecule targeted to DNA-PKcs and the chemotherapeutic agent are administered simultaneously.
7. The method of claim 1 , wherein the molecule targeted to DNA-PKcs and the chemotherapeutic agent are administered sequentially.
8. A method for treating a cancer patient, comprising:
(a) identifying the patient as having a tumor with cells that express B7-H1 ; and
(b) administering to the patient a DNA-PKcs inhibitor and an anti-B7-Hl blocking antibody.
9. The method of claim 8, wherein the cancer patient is a human.
10. The method of claim 8, wherein the cancer patient is identified based on the level of B7-H1 protein in a sample obtained from the tumor.
11. The method of claim 8, wherein the cancer patient is identified based on the level of B7-H1 mRNA in a sample obtained from the tumor.
12. The method of claim 8, wherein the cancer is a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer.
13. The method of claim 8, wherein the DNA-PKcs inhibitor and the anti-B7-Hl blocking antibody are administered to the cancer patient simultaneously.
14. The method of claim 8, wherein the DNA-PKcs inhibitor and the anti-B7-Hl blocking antibody are administered to the cancer patient sequentially.
15. A method for treating cancer, comprising administering a DNA-PKcs inhibitor and an anti-B7-Hl antibody to a mammal identified as having a tumor containing cells with an elevated level of B7-H1, wherein the DNA-PKcs inhibitor and the anti-B7-Hl antibody are administered under conditions wherein the interaction of naturally-occurring B7-H1 with DNA-PKcs and the interaction of naturally-occurring B7-H1 with PD-1 or CD80 in the mammal is reduced after the administering.
16. The method of claim 15, wherein the mammal is a human.
17. The method of claim 15, wherein the elevated level B7-H1 is based on the level of B7-H1 protein in a sample obtained from the tumor.
18. The method of claim 15, wherein the elevated level of B7-H1 is based on the level of B7-H1 mRNA in a sample obtained from the tumor.
19. The method of claim 15, wherein said the cancer is a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer.
20. The method of claim 15, wherein the DNA-PKcs inhibitor and the anti-B7-Hl blocking antibody are administered to the mammal simultaneously.
21. The method of claim 15, wherein the DNA-PKcs inhibitor and the anti-B7-Hl blocking antibody are administered to the mammal sequentially.
22. A composition comprising a pharmaceutically acceptable carrier and a molecule targeted to DNA-PKcs for use in treating cancer in a patient, wherein the patient also is to be treated with a chemotherapeutic agent that causes DNA damage, and wherein the composition is for administration in an amount sufficient to reduce the interaction of DNA-PKcs with B7-H1.
23. The composition of claim 22, wherein the patient is a human.
24. The composition of claim 22, wherein the chemotherapeutic agent is cisplatin, doxorubicin, SN38, paclitaxel, protein-bound paclitaxel, temozolomide, or carboplatin.
25. The composition of claim 22, wherein the molecule targeted to DNA-PKcs is NU7026, NU7441, IC86621, IC87102, IC87361, OK-1035, SU11752, vanillin, or IC486241.
26. The composition of claim 22, wherein the molecule targeted to DNA-PKcs is an anti-DNA-PKcs antibody.
27. The composition of claim 22, wherein the chemotherapeutic agent and the composition comprising the molecule targeted to DNA-PKcs are for simultaneous administration.
28. The composition of claim 22, wherein the chemotherapeutic agent and the composition comprising the molecule targeted to DNA-PKcs are for sequential administration.
29. A composition comprising a pharmaceutically acceptable carrier and a DNA- PKcs inhibitor for treating cancer in a patient, wherein the patient is identified as having a tumor with cells that express B7-H1, and wherein the patient also is to be treated with an anti-B7-Hl blocking antibody.
30. The composition of claim 29, wherein the cancer patient is a human.
31. The composition of claim 29, wherein the cancer patient is identified based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7- Hl mPvNA in a sample obtained from the tumor.
32. The composition of claim 29, wherein the cancer is a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer.
33. The composition of claim 29, wherein the anti-B7-Hl blocking antibody and the composition containing the DNA-PKcs inhibitor are for simultaneous administration.
34. The composition of claim 29, wherein the anti-B7-Hl blocking antibody and the composition containing the DNA-PKcs inhibitor are for sequential administration.
35. A composition comprising a pharmaceutically acceptable carrier and a DNA- PKcs inhibitor for use in treating cancer in a mammal identified as having a tumor containing cells with an elevated level of B7-H1, wherein the mammal also is to be treated with an anti-B7-Hl antibody, and wherein the anti-B7-Hl antibody and the composition comprising the DNA-PKcs inhibitor are for administration under conditions in which the interaction of naturally-occurring B7-H1 with DNA-PKcs and the interaction of naturally-occurring B7-H1 with PD-1 or CD80 in the mammal is reduced after the administering.
36. The composition of claim 35, wherein the mammal is a human.
37. The composition of claim 35, wherein the elevated level B7-H1 was determined based on the level of B7-H1 protein in a sample obtained from the tumor, or based on the level of B7-H1 m NA in a sample obtained from the tumor.
38. The composition of claim 35, wherein the cancer is a melanoma cancer, a breast cancer, a lung cancer, a renal cell carcinoma cancer, a pancreas cancer, a prostate cancer, a colon cancer, a brain cancer, a liver cancer, or an ovarian cancer.
39. The composition of claim 35, wherein the anti-B7-Hl blocking antibody and the composition containing the DNA-PKcs inhibitor are for simultaneous administration.
40. The composition of claim 35, wherein the anti-B7-Hl blocking antibody and the composition containing the DNA-PKcs inhibitor are for sequential administration.
PCT/US2015/032016 2014-07-23 2015-05-21 Targeting dna-pkcs and b7-h1 to treat cancer WO2016014148A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US15/325,612 US10517875B2 (en) 2014-07-23 2015-05-21 Targeting DNA-PKcs and B7-H1 to treat cancer
EP15825450.8A EP3171896A4 (en) 2014-07-23 2015-05-21 Targeting dna-pkcs and b7-h1 to treat cancer
US16/681,154 US11504376B2 (en) 2014-07-23 2019-11-12 Targeting DNA-PKCS and B7-H1 to treat cancer
US17/991,459 US20230233571A1 (en) 2014-07-23 2022-11-21 Targeting dna-pkcs and b7-h1 to treat cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462027841P 2014-07-23 2014-07-23
US62/027,841 2014-07-23

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/325,612 A-371-Of-International US10517875B2 (en) 2014-07-23 2015-05-21 Targeting DNA-PKcs and B7-H1 to treat cancer
US16/681,154 Continuation US11504376B2 (en) 2014-07-23 2019-11-12 Targeting DNA-PKCS and B7-H1 to treat cancer

Publications (1)

Publication Number Publication Date
WO2016014148A1 true WO2016014148A1 (en) 2016-01-28

Family

ID=55163494

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/032016 WO2016014148A1 (en) 2014-07-23 2015-05-21 Targeting dna-pkcs and b7-h1 to treat cancer

Country Status (3)

Country Link
US (3) US10517875B2 (en)
EP (1) EP3171896A4 (en)
WO (1) WO2016014148A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018178040A1 (en) 2017-03-30 2018-10-04 Merck Patent Gmbh Combination of an anti-pd-l1 antibody and a dna-pk inhibitor for the treatment of cancer
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US10344090B2 (en) 2013-12-12 2019-07-09 Shanghai Hangrui Pharmaceutical Co., Ltd. PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
EP3518931A4 (en) * 2016-09-27 2020-05-13 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of dna-damaging agents and dna-pk inhibitors
US10800846B2 (en) 2015-02-26 2020-10-13 Merck Patent Gmbh PD-1/PD-L1 inhibitors for the treatment of cancer
US10869924B2 (en) 2015-06-16 2020-12-22 Merck Patent Gmbh PD-L1 antagonist combination treatments
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1
CN112512576A (en) * 2018-05-04 2021-03-16 默克专利有限公司 Combined inhibition of PD-1/PD-L1, TGF beta and DNA-PK for the treatment of cancer
US11274154B2 (en) 2016-10-06 2022-03-15 Pfizer Inc. Dosing regimen of avelumab for the treatment of cancer
US11504376B2 (en) 2014-07-23 2022-11-22 Mayo Foundation For Medical Education And Research Targeting DNA-PKCS and B7-H1 to treat cancer

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110200973A (en) * 2019-06-19 2019-09-06 南京市儿童医院 Purposes of the DNA dependent protein kinase specific inhibitor in preparation prevention and treatment kidney fibrosis drug
WO2023158911A2 (en) * 2022-02-15 2023-08-24 The Regents Of The University Of California Methods to prevent therapy resistance in melanoma via blocking genomic instability
WO2024043946A1 (en) * 2022-08-25 2024-02-29 Arima Genomics, Inc. Methods of selecting and treating cancer subjects having a genetic structural variant associated with ptprd

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004085418A2 (en) * 2003-03-24 2004-10-07 Luitpold Pharmaceuticals, Inc. Xanthones, thioxanthones and acridinones as dna-pk inhibitors
WO2006042237A2 (en) * 2004-10-06 2006-04-20 Mayo Foundation For Medical Education And Research B7-h1 and methods of diagnosis, prognosis, and treatment of cancer
US20130273656A1 (en) * 2010-10-08 2013-10-17 Regents Of The University Of Minnesota Method to increase gene targeting frequency

Family Cites Families (173)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4034074A (en) 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US4036945A (en) 1976-05-03 1977-07-19 The Massachusetts General Hospital Composition and method for determining the size and location of myocardial infarcts
US4098876A (en) 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
US4233402A (en) 1978-04-05 1980-11-11 Syva Company Reagents and method employing channeling
US4272398A (en) 1978-08-17 1981-06-09 The United States Of America As Represented By The Secretary Of Agriculture Microencapsulation process
US4257774A (en) 1979-07-16 1981-03-24 Meloy Laboratories, Inc. Intercalation inhibition assay for compounds that interact with DNA or RNA
US4331647A (en) 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
US4376110A (en) 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US4769330A (en) 1981-12-24 1988-09-06 Health Research, Incorporated Modified vaccinia virus and methods for making and using the same
US5155020A (en) 1989-03-08 1992-10-13 Health Research Inc. Recombinant poxvirus host range selection system
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4650764A (en) 1983-04-12 1987-03-17 Wisconsin Alumni Research Foundation Helper cell
JPS61134325A (en) 1984-12-04 1986-06-21 Teijin Ltd Expression of hybrid antibody gene
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5618920A (en) 1985-11-01 1997-04-08 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
US4861719A (en) 1986-04-25 1989-08-29 Fred Hutchinson Cancer Research Center DNA constructs for retrovirus packaging cell lines
WO1988001213A1 (en) 1986-08-18 1988-02-25 Clinical Technologies Associates, Inc. Delivery systems for pharmacological agents
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4987071A (en) 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US4980289A (en) 1987-04-27 1990-12-25 Wisconsin Alumni Research Foundation Promoter deficient retroviral vector
US4861627A (en) 1987-05-01 1989-08-29 Massachusetts Institute Of Technology Preparation of multiwall polymeric microcapsules
US5254678A (en) 1987-12-15 1993-10-19 Gene Shears Pty. Limited Ribozymes
DE68926888T2 (en) 1988-01-22 1997-01-09 Zymogenetics Inc Process for the production of secreted receptor analogs
US5750375A (en) 1988-01-22 1998-05-12 Zymogenetics, Inc. Methods of producing secreted receptor analogs and biologically active dimerized polypeptide fusions
US6018026A (en) 1988-01-22 2000-01-25 Zymogenetics, Inc. Biologically active dimerized and multimerized polypeptide fusions
US5567584A (en) 1988-01-22 1996-10-22 Zymogenetics, Inc. Methods of using biologically active dimerized polypeptide fusions to detect PDGF
US5278056A (en) 1988-02-05 1994-01-11 The Trustees Of Columbia University In The City Of New York Retroviral packaging cell lines and process of using same
US5750666A (en) 1988-05-26 1998-05-12 Competitve Technologies, Inc. Polynucleotide phosphorodithioate compounds
US6303121B1 (en) 1992-07-30 2001-10-16 Advanced Research And Technology Method of using human receptor protein 4-1BB
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5124263A (en) 1989-01-12 1992-06-23 Wisconsin Alumni Research Foundation Recombination resistant retroviral helper cell and products produced thereby
US5225538A (en) 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
US5225336A (en) 1989-03-08 1993-07-06 Health Research Incorporated Recombinant poxvirus host range selection system
US5175099A (en) 1989-05-17 1992-12-29 Research Corporation Technologies, Inc. Retrovirus-mediated secretion of recombinant products
US5240846A (en) 1989-08-22 1993-08-31 The Regents Of The University Of Michigan Gene therapy vector for cystic fibrosis
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
DK0710719T3 (en) 1990-01-12 2007-07-09 Amgen Fremont Inc Generation of xenogenic antibodies
EP0438803B1 (en) 1990-01-26 1997-03-12 Immunomedics, Inc. Vaccines against cancer and infectious diseases
US5204243A (en) 1990-02-14 1993-04-20 Health Research Incorporated Recombinant poxvirus internal cores
US5214136A (en) 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
US6641809B1 (en) 1990-03-26 2003-11-04 Bristol-Myers Squibb Company Method of regulating cellular processes mediated by B7 and CD28
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
EP0537293A4 (en) 1990-07-02 1993-09-08 Bristol-Myers Company Ligand for cd28 receptor on b cells and methods
EP0585287B1 (en) 1990-07-10 1999-10-13 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
CA2092002A1 (en) 1990-09-20 1992-03-21 Mark Matteucci Modified internucleoside linkages
US5296347A (en) 1991-02-08 1994-03-22 Ciba Corning Diagnostics Corp. Bridge immunoassay
JP3854306B2 (en) 1991-03-06 2006-12-06 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Humanized and chimeric monoclonal antibodies
JP3507486B2 (en) 1991-03-15 2004-03-15 アムジエン・インコーポレーテツド Intrapulmonary administration of granulocyte colony-stimulating factor
CA2112578C (en) 1991-07-03 2000-10-10 Yasuhiro Ogawa Thermoplastic polyurethane elastomer, process for producing same, apparatus for producing same and elastomer fibers made from same
CA2078539C (en) 1991-09-18 2005-08-02 Kenya Shitara Process for producing humanized chimera antibody
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
GB9125979D0 (en) 1991-12-06 1992-02-05 Wellcome Found Antibody
NZ249677A (en) 1992-02-19 1996-08-27 Schering Corp Monoclonal antibodies and compositions useful for treating il-4 diseases and intermediates for making such antibodies
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
AU6827094A (en) 1993-05-07 1994-12-12 Immunex Corporation Cytokine designated 4-1bb ligand and human receptor that binds thereto
US7211259B1 (en) 1993-05-07 2007-05-01 Immunex Corporation 4-1BB polypeptides and DNA encoding 4-1BB polypeptides
US5861310A (en) 1993-11-03 1999-01-19 Dana-Farber Cancer Institute Tumor cells modified to express B7-2 with increased immunogenicity and uses therefor
US5942607A (en) 1993-07-26 1999-08-24 Dana-Farber Cancer Institute B7-2: a CTLA4/CD28 ligand
WO1995005464A1 (en) 1993-08-16 1995-02-23 Arch Development Corporation B7-2: ctla4/cd28 counter receptor
CN1135181A (en) 1993-09-14 1996-11-06 Cytel有限公司 Alteration of immune response using pan Dr-binding peptides
KR100244960B1 (en) 1993-09-16 2000-02-15 왈터 엘 쿤, 주니어 Human receptor h4-1bb
US5451569A (en) 1994-04-19 1995-09-19 Hong Kong University Of Science And Technology R & D Corporation Limited Pulmonary drug delivery system
US5972703A (en) 1994-08-12 1999-10-26 The Regents Of The University Of Michigan Bone precursor cells: compositions and methods
US5705154A (en) 1995-03-08 1998-01-06 Schering Corporation Humanized monoclonal antibodies against human interleukin-4
WO1996029348A1 (en) 1995-03-23 1996-09-26 Indiana University Foundation Monoclonal antibody against human receptor protein 4-1bb and methods of its use for treatment of diseases
ATE226641T1 (en) 1995-04-08 2002-11-15 Lg Chemical Ltd HUMAN 4-1BB SPECIFIC HUMAN ANTIBODIES AND CELL LINE PRODUCING THEM
US5675848A (en) 1995-10-18 1997-10-14 Mallinckrodt Medical, Inc. Inflatable blanket having perforations of different sizes
PT859959E (en) 1995-11-10 2003-12-31 Elan Corp Plc PEPTIDES THAT INCREASE TRANSPORTATION THROUGH TISSUES AND METHODS FOR THEIR IDENTIFICATION AND USES
WO1997024447A1 (en) 1996-01-02 1997-07-10 Chiron Corporation Immunostimulation mediated by gene-modified dendritic cells
US5874240A (en) 1996-03-15 1999-02-23 Human Genome Sciences, Inc. Human 4-1BB receptor splicing variant
DE69718268D1 (en) 1996-10-03 2003-02-13 Canon Kk Methods for the detection of target nucleic acid, methods for their quantification and pyrylium compounds for chemiluminescence analysis
IL129138A0 (en) 1996-10-11 2000-02-17 Bristol Myers Squibb Co Methods and compositions for immunomodulation
ATE414149T1 (en) 1996-11-20 2008-11-15 Univ Yale SURVIVIN, A PROTEIN THAT INHIBITS CELLULAR APOPTOSIS AND ITS MODULATION
AUPO390396A0 (en) 1996-11-29 1996-12-19 Csl Limited Novel promiscuous T helper cell epitopes
EP1012275A1 (en) 1997-01-31 2000-06-28 University Of Rochester Chimeric antibody fusion proteins for the recruitment and stimulation of an antitumor immune response
AU6534898A (en) 1997-02-14 1998-09-08 E.I. Du Pont De Nemours And Company Detection of double-stranded dna in a homogeneous solution
US20070224663A1 (en) 1997-03-07 2007-09-27 Human Genome Sciences, Inc. Human Secreted Proteins
US20060223088A1 (en) 1997-03-07 2006-10-05 Rosen Craig A Human secreted proteins
US7411051B2 (en) 1997-03-07 2008-08-12 Human Genome Sciences, Inc. Antibodies to HDPPA04 polypeptide
US7368531B2 (en) 1997-03-07 2008-05-06 Human Genome Sciences, Inc. Human secreted proteins
EP1015578A4 (en) 1997-09-17 2004-12-01 Walker And Eliza Hall Inst Of Therapeutic molecules
EP1086224B1 (en) 1998-06-10 2006-03-29 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES B2 microglobulin fusion proteins and high affinity variants
WO2000026342A1 (en) 1998-10-30 2000-05-11 Northwestern University Method of interfering with cell proliferation
KR20000034847A (en) 1998-11-17 2000-06-26 성재갑 Humanized Antibody Specific for Human 4-1BB Molecule and Pharmaceutical Composition Comprising Same
US6734172B2 (en) 1998-11-18 2004-05-11 Pacific Northwest Research Institute Surface receptor antigen vaccines
AU3209300A (en) 1999-01-15 2000-08-01 Mount Sinai School Of Medicine Of The City University Of New York, The Combination therapy of cancer by the activation of co-stimulatory molecules expressed by immune cells and cytokines
US20040010134A1 (en) 2000-04-12 2004-01-15 Rosen Craig A. Albumin fusion proteins
WO2000055375A1 (en) 1999-03-17 2000-09-21 Alphagene, Inc. Secreted proteins and polynucleotides encoding them
EP2028190A1 (en) 1999-04-02 2009-02-25 Corixa Corporation Compounds and methods for therapy and diagnosis of lung cancer
EP1074617A3 (en) 1999-07-29 2004-04-21 Research Association for Biotechnology Primers for synthesising full-length cDNA and their use
US6423885B1 (en) 1999-08-13 2002-07-23 Commonwealth Scientific And Industrial Research Organization (Csiro) Methods for obtaining modified phenotypes in plant cells
US20020177551A1 (en) 2000-05-31 2002-11-28 Terman David S. Compositions and methods for treatment of neoplastic disease
JP5583308B2 (en) 1999-11-12 2014-09-03 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド 21 human secreted proteins
CA2392477A1 (en) 1999-11-30 2001-06-07 Mayo Foundation For Medical Education And Research B7-h1, a novel immunoregulatory molecule
US6803192B1 (en) 1999-11-30 2004-10-12 Mayo Foundation For Medical Education And Research B7-H1, a novel immunoregulatory molecule
JP2003523768A (en) 2000-02-25 2003-08-12 イミュネックス・コーポレーション Integrin antagonist
US20030165831A1 (en) 2000-03-21 2003-09-04 John Lee Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of ovarian cancer
US7030219B2 (en) 2000-04-28 2006-04-18 Johns Hopkins University B7-DC, Dendritic cell co-stimulatory molecules
CA2412377A1 (en) 2000-06-06 2001-12-13 Bristol-Myers Squibb Company B7-related nucleic acids and polypeptides and their uses for immunomodulation
US20030031675A1 (en) 2000-06-06 2003-02-13 Mikesell Glen E. B7-related nucleic acids and polypeptides useful for immunomodulation
US20020164600A1 (en) 2000-06-28 2002-11-07 Gordon Freeman PD-L2 molecules: novel PD-1 ligands and uses therefor
JP2004502414A (en) 2000-06-30 2004-01-29 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド B7-like polynucleotides, polypeptides and antibodies
EP1299606B1 (en) 2000-07-05 2006-10-04 Vogue Pool Products Above ground swimming pool support structure
US20020076409A1 (en) 2000-07-12 2002-06-20 March Carl J. Method for treating cancer
US6635750B1 (en) 2000-07-20 2003-10-21 Millennium Pharmaceuticals, Inc. B7-H2 nucleic acids, members of the B7 family
US6891030B2 (en) 2000-07-27 2005-05-10 Mayo Foundation For Medical Education And Research T-cell immunoregulatory molecule
ES2292619T3 (en) 2000-09-15 2008-03-16 Ortho-Mcneil Pharmaceutical, Inc. COMPOSITIONS AND METHODS TO INDUCE SPECIFIC CYTOLOGICAL ANSWERS OF CELL T.
US20020107363A1 (en) 2000-09-20 2002-08-08 Amgen, Inc. B7-Like molecules and uses thereof
US20040247563A1 (en) 2000-11-02 2004-12-09 Lynch David H. Method of enhancing lymphocyte-mediated immune responses
WO2002046449A2 (en) 2000-12-07 2002-06-13 The Penn State Research Foundation Selection of catalytic nucleic acids targeted to infectious agents
CA2433313A1 (en) 2000-12-19 2002-07-25 Curagen Corporation Polypetides and nucleic acids encoding same
WO2002058535A2 (en) 2000-12-26 2002-08-01 Oxigene, Inc. Use of combretastatin a4 and its prodrugs as an immune enhancing therapy
US20030180309A1 (en) 2001-01-08 2003-09-25 Baum Peter R. Human B7 polypeptides
CA2437305A1 (en) 2001-01-12 2003-07-25 Yale University Detection of survivin in urine of bladder cancer patients
US6743619B1 (en) 2001-01-30 2004-06-01 Nuvelo Nucleic acids and polypeptides
WO2003008583A2 (en) 2001-03-02 2003-01-30 Sagres Discovery Novel compositions and methods for cancer
AR036993A1 (en) 2001-04-02 2004-10-20 Wyeth Corp USE OF AGENTS THAT MODULATE THE INTERACTION BETWEEN PD-1 AND ITS LINKS IN THE SUBMODULATION OF IMMUNOLOGICAL ANSWERS
US20060084794A1 (en) 2001-04-12 2006-04-20 Human Genome Sciences, Inc. Albumin fusion proteins
AU2002258941A1 (en) 2001-04-20 2002-11-05 Mayo Foundation For Medical Education And Research Methods of enhancing cell responsiveness
US20060276422A1 (en) 2001-05-18 2006-12-07 Nassim Usman RNA interference mediated inhibition of B7-H1 gene expression using short interfering nucleic acid (siNA)
WO2003006632A2 (en) 2001-07-12 2003-01-23 Canvac Methods and compisitions for activation human t cells in vitro
WO2003049755A1 (en) 2001-10-09 2003-06-19 Mayo Foundation For Medical Education And Research Enhancement of immune responses by 4-1bb-binding agents
WO2003033677A2 (en) 2001-10-19 2003-04-24 Zymogenetics, Inc. Dimerized growth factor and materials and methods for producing it
CA2466279A1 (en) 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents that modulate immune cell activation and methods of use thereof
US7164500B2 (en) 2002-01-29 2007-01-16 Hewlett-Packard Development Company, L.P. Method and apparatus for the automatic generation of image capture device control marks
AU2003222249A1 (en) 2002-03-04 2003-09-22 Fidelity Systems, Inc., Et Al. The complete genome and protein sequence of the hyperthermophile methanopyrus kandleri av19 and monophyly of archael methanogens and methods of use thereof
US20030223989A1 (en) 2002-04-18 2003-12-04 Pluenneke John D. CD137 agonists to treat patients with IgE-mediated conditions
ATE481985T1 (en) 2002-07-03 2010-10-15 Ono Pharmaceutical Co IMMUNOPOTENTATING COMPOSITIONS
NZ538216A (en) 2002-07-15 2008-09-26 Mayo Foundation Treatment and prophylaxis with 4-1BB-binding agents
US7432351B1 (en) 2002-10-04 2008-10-07 Mayo Foundation For Medical Education And Research B7-H1 variants
US7449300B2 (en) 2002-11-21 2008-11-11 Mayo Foundation For Medical Education And Research Detection of antibodies specific for B7-H1 in subjects with diseases or pathological conditions mediated by activated T cells
EP1591527B1 (en) 2003-01-23 2015-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
CA2558811A1 (en) 2004-03-08 2005-09-22 Zymogenetics, Inc. Dimeric fusion proteins and materials and methods for producing them
EP2366717A3 (en) 2004-10-29 2011-12-14 University of Southern California Combination Cancer Immunotherapy with Co-Stimulatory Molecules
CN104436190A (en) 2005-06-08 2015-03-25 达纳-法伯癌症研究院公司 Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (PD-1) pathway
US20070231344A1 (en) 2005-10-28 2007-10-04 The Brigham And Women's Hospital, Inc. Conjugate vaccines for non-proteinaceous antigens
US20100015642A1 (en) 2006-01-05 2010-01-21 Kwon Eugene D B7-h1 and survivin in cancer
WO2007082154A2 (en) 2006-01-05 2007-07-19 Mayo Foundation For Medical Education And Research B7-h1 and b7-h4 in cancer
WO2007124361A2 (en) 2006-04-20 2007-11-01 Mayo Foundation For Medical Education And Research Soluble b7-h1
WO2007134384A2 (en) 2006-05-19 2007-11-29 Children Youth And Women's Health Service Selective modulation of receptor signalling
AU2007342338A1 (en) 2006-09-20 2008-07-17 The Johns Hopkins University Combinatorial therapy of cancer and infectious diseases with anti-B7-H1 antibodies
WO2008037080A1 (en) 2006-09-29 2008-04-03 Universite De Montreal Methods and compositions for immune response modulation and uses thereof
NZ578650A (en) 2006-12-27 2011-12-22 Harvard College Pd-1 and activated t-cell compositions and methods for the treatment of viral infections and tumors
EP2514762B1 (en) 2007-07-13 2015-04-08 The Johns Hopkins University B7-DC variants
US20090324609A1 (en) 2007-08-09 2009-12-31 Genzyme Corporation Method of treating autoimmune disease with mesenchymal stem cells
US20100285039A1 (en) 2008-01-03 2010-11-11 The Johns Hopkins University B7-H1 (CD274) Antagonists Induce Apoptosis of Tumor Cells
WO2009111315A2 (en) 2008-02-29 2009-09-11 Mayo Foundation For Medical Education And Research Methods for reducing granulomatous inflammation
US20090226435A1 (en) 2008-03-08 2009-09-10 Sanjay Khare Engineered fusion molecules immunotherapy in cancer and inflammatory diseases
JP2012510429A (en) 2008-08-25 2012-05-10 アンプリミューン、インコーポレーテッド PD-1 antagonist and method of use thereof
PE20110435A1 (en) 2008-08-25 2011-07-20 Amplimmune Inc ANTAGONIST COMPOSITIONS OF PD-1
US8577949B2 (en) 2009-07-07 2013-11-05 L-3 Communications Integrated Systems, L.P. System for conjugate gradient linear iterative solvers
US20130017199A1 (en) 2009-11-24 2013-01-17 AMPLIMMUNE ,Inc. a corporation Simultaneous inhibition of pd-l1/pd-l2
ES2646863T3 (en) 2009-11-24 2017-12-18 Medimmune Limited B7-H1 specific binding agents
WO2012012710A2 (en) 2010-07-22 2012-01-26 The Johns Hopkins University Radiation sensitization agents for prostate cancer
US9670547B2 (en) * 2010-09-15 2017-06-06 Almac Diagnostics Limited Molecular diagnostic test for cancer
JP2014527036A (en) 2011-06-27 2014-10-09 ザ ジャクソン ラボラトリー Methods and compositions for the treatment of cancer and autoimmune diseases
CA2843595C (en) 2011-08-01 2022-10-18 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
CN107586340B (en) 2011-08-23 2022-01-21 罗切格利卡特公司 Bispecific antibodies specific for T cell activating antigens and tumor antigens and methods of use
US20150004175A1 (en) 2011-12-13 2015-01-01 Yale University Compositions and Methods for Reducing CTL Exhaustion
AR090263A1 (en) 2012-03-08 2014-10-29 Hoffmann La Roche COMBINED ANTIBODY THERAPY AGAINST HUMAN CSF-1R AND USES OF THE SAME
WO2014131711A1 (en) 2013-02-26 2014-09-04 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
US9302005B2 (en) 2013-03-14 2016-04-05 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
WO2015179654A1 (en) 2014-05-22 2015-11-26 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti b7-h1 antibodies
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US9535074B2 (en) 2014-09-08 2017-01-03 Merck Sharp & Dohme Corp. Immunoassay for soluble PD-L1
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004085418A2 (en) * 2003-03-24 2004-10-07 Luitpold Pharmaceuticals, Inc. Xanthones, thioxanthones and acridinones as dna-pk inhibitors
WO2006042237A2 (en) * 2004-10-06 2006-04-20 Mayo Foundation For Medical Education And Research B7-h1 and methods of diagnosis, prognosis, and treatment of cancer
US20130273656A1 (en) * 2010-10-08 2013-10-17 Regents Of The University Of Minnesota Method to increase gene targeting frequency

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3171896A4 *

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US11136393B2 (en) 2013-10-01 2021-10-05 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of Bim
US11365255B2 (en) 2013-12-12 2022-06-21 Suzhou Suncadia Biopharmaceuticals Co., Ltd. PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
US10344090B2 (en) 2013-12-12 2019-07-09 Shanghai Hangrui Pharmaceutical Co., Ltd. PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US11504376B2 (en) 2014-07-23 2022-11-22 Mayo Foundation For Medical Education And Research Targeting DNA-PKCS and B7-H1 to treat cancer
US10800846B2 (en) 2015-02-26 2020-10-13 Merck Patent Gmbh PD-1/PD-L1 inhibitors for the treatment of cancer
US10869924B2 (en) 2015-06-16 2020-12-22 Merck Patent Gmbh PD-L1 antagonist combination treatments
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1
EP3518931A4 (en) * 2016-09-27 2020-05-13 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of dna-damaging agents and dna-pk inhibitors
AU2017335648B2 (en) * 2016-09-27 2022-02-17 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA-damaging agents and DNA-PK inhibitors
US11110108B2 (en) 2016-09-27 2021-09-07 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA-damaging agents and DNA-PK inhibitors
US11980633B2 (en) 2016-09-27 2024-05-14 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of DNA-damaging agents and DNA-PK inhibitors
US11274154B2 (en) 2016-10-06 2022-03-15 Pfizer Inc. Dosing regimen of avelumab for the treatment of cancer
CN110494161A (en) * 2017-03-30 2019-11-22 默克专利股份有限公司 The combination of anti-PD-L1 antibody and DNA-PK inhibitor for treating cancer
RU2765997C2 (en) * 2017-03-30 2022-02-07 Мерк Патент Гмбх Combination of anti-pd-l1 antibody and dna-pk inhibitor for treating malignant neoplasm
JP2020515609A (en) * 2017-03-30 2020-05-28 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Combination of anti-PD-L1 antibody and DNA-PK inhibitor for treatment of cancer
JP7166278B2 (en) 2017-03-30 2022-11-07 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Combination of anti-PD-L1 antibody and DNA-PK inhibitor for cancer treatment
KR20190135028A (en) * 2017-03-30 2019-12-05 메르크 파텐트 게엠베하 Combination of anti-PD-L1 antibodies and DNA-PK inhibitors for the treatment of cancer
KR102644408B1 (en) * 2017-03-30 2024-03-07 메르크 파텐트 게엠베하 Combination of anti-PD-L1 antibody and DNA-PK inhibitor for treatment of cancer
WO2018178040A1 (en) 2017-03-30 2018-10-04 Merck Patent Gmbh Combination of an anti-pd-l1 antibody and a dna-pk inhibitor for the treatment of cancer
CN112512576A (en) * 2018-05-04 2021-03-16 默克专利有限公司 Combined inhibition of PD-1/PD-L1, TGF beta and DNA-PK for the treatment of cancer

Also Published As

Publication number Publication date
US20200061077A1 (en) 2020-02-27
US10517875B2 (en) 2019-12-31
US20230233571A1 (en) 2023-07-27
EP3171896A1 (en) 2017-05-31
US11504376B2 (en) 2022-11-22
EP3171896A4 (en) 2018-03-21
US20170173030A1 (en) 2017-06-22

Similar Documents

Publication Publication Date Title
US11504376B2 (en) Targeting DNA-PKCS and B7-H1 to treat cancer
US11136393B2 (en) Methods for treating cancer in patients with elevated levels of Bim
JP6861418B2 (en) Antibodies specific for human T cell immunoglobulin and ITIM domain (TIGIT)
US20190361033A1 (en) Distinguishing antagonistic and agonistic anti b7-h1 antibodies
EP3423495B1 (en) Antibodies specific to human poliovirus receptor (pvr)
MX2011000236A (en) Notch-binding agents and antagonists and methods of use thereof.
JP2018538263A (en) Method for treating lung cancer using a combination of anti-PD-1 antibody and anti-CTLA-4 antibody
US20180044422A1 (en) Treating solid tumor by targeting dectin-1 signaling
US11110171B2 (en) PD-1 related cancer therapy
TW201716434A (en) Single domain antibodies directed against intracellular antigens
Cicatiello et al. Powerful anti-tumor and anti-angiogenic activity of a new anti-vascular endothelial growth factor receptor 1 peptide in colorectal cancer models
US20180028566A1 (en) Gamma delta t cells as a target for treatment of solid tumors
KR20180058824A (en) Reasonable combination therapy for cancer treatment
Milito et al. NKG2D engagement on human NK cells leads to DNAM‐1 hypo‐responsiveness through different converging mechanisms
US20200024347A1 (en) Or10h1 antigen binding proteins and uses thereof
US20240101652A1 (en) Therapeutic single domain antibody
US11897950B2 (en) Osteopontin monoclonal antibodies
Bader Targeting Cell Surface GRP78 for Specific Nanoparticle Mediated Drug Delivery to Breast Cancer
Sun et al. Platelet-mediated circulating tumor cell evasion from natural killer cell killing via immune checkpoint CD155-TIGIT
US20210340274A1 (en) Targeting CLPTMIL for Treatment and Prevention of Cancer
WO2021140173A1 (en) Methods and uses for treating fibrotic solid tumors with bags inhibitors
WO2024026019A1 (en) Methods for treating chronic myelomonocytic leukemia with anti-ilt3 antibodies
AU2022318734A1 (en) Methods for treating acute myeloid leukemia with anti-ilt3 antibodies
Bernardini et al. Erythropoietin receptor modulates breast cancer cell response to tamoxifen treatment

Legal Events

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

Ref document number: 15825450

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 15325612

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015825450

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015825450

Country of ref document: EP