US20190391152A1 - Markers selectively deregulated in tumor-infiltrating regulatory t cells - Google Patents

Markers selectively deregulated in tumor-infiltrating regulatory t cells Download PDF

Info

Publication number
US20190391152A1
US20190391152A1 US16/301,805 US201716301805A US2019391152A1 US 20190391152 A1 US20190391152 A1 US 20190391152A1 US 201716301805 A US201716301805 A US 201716301805A US 2019391152 A1 US2019391152 A1 US 2019391152A1
Authority
US
United States
Prior art keywords
tumor
marker
cells
molecule
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/301,805
Other languages
English (en)
Inventor
Sergio Abrignani
Massimiliano Pagani
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Checkmab SRL
Original Assignee
Checkmab SRL
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 Checkmab SRL filed Critical Checkmab SRL
Assigned to CHECKMAB S.R.L. reassignment CHECKMAB S.R.L. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABRIGNANI, SERGIO, PAGANI, MASSIMILIANO
Publication of US20190391152A1 publication Critical patent/US20190391152A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1135Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against oncogenes or tumor suppressor genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0637Immunosuppressive T lymphocytes, e.g. regulatory T cells or Treg
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Definitions

  • the present invention relates to a molecule able to modulate the expression and/or function of at least one marker that is selectively deregulated in tumor-infiltrating regulatory T cell or to a molecule capable of specifically binding to at least one marker that is selectively deregulated in tumor-infiltrating regulatory T cell and inducing antibody-dependent cell-mediated cytotoxicity (ADCC) for use in the prevention and/or treatment of cancer or for use in a method for in vivo depleting tumor-infiltrating regulatory T cell in a subject or for use in a method to enhance tumor immunity in a subject and relative pharmaceutical composition.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • T lymphocytes recognizing tumor derived peptides presented by major histocompatibility complex (MHC) molecules play a central role in immunotherapy and in conventional chemo-radiotherapy of cancer (Galluzzi et al., 2015).
  • MHC major histocompatibility complex
  • anti-tumor T cell responses arise in cancer patients but are disabled upon tumor progression by suppressive mechanisms triggered by the interplay between malignant cells and the tumor microenvironment (Munn and Bronte, 2015).
  • the tumor-dependent immunosuppressive mechanisms depend on the integrated action of infiltrating leukocytes and lymphocytes that upregulate a range of modulatory molecules, collectively called immune checkpoints, whose function is only partially characterized (Pardoll, 2012). Therefore, the search for agonists of co-stimulatory complexes or antagonists of inhibitory molecules to potentiate antigen-specific T cell responses is a primary goal of current anti-tumor research (Sharma and Allison, 2015; Zitvogel et al., 2013).
  • CTLA-4 has been one of the first to be translated into therapeutic applications.
  • Anti-CTLA-4 monoclonal antibodies showed remarkable success in metastatic melanoma, and more recently in non-small-cell lung cancer, prostate cancer, renal cell carcinoma, urothelial carcinoma and ovarian cancer (Carthon et al., 2010; Hodi et al., 2010; van den Eertwegh et al., 2012; Yang et al., 2007). However, the fraction of patients that do not respond remains high, prompting a deeper investigation of the mechanisms underpinning the modulation of immune responses by tumors.
  • Treg cells which are physiologically engaged in the maintenance of immunological self-tolerance and immune homeostasis (Josefowicz et al., 2012; Sakaguchi et al., 2008), are potent suppressors of effector cells and are found at high frequencies in various types of cancers (Fridman et al., 2012; Nishikawa and Sakaguchi, 2010). Interestingly, Treg cells adapt their transcriptional program to the various cytokines to which they are exposed in the inflammatory milieu (Campbell and Koch, 2011).
  • Treg cell populations with unique features and immunomodulatory functions (Duhen et al., 2012; Geginat et al., 2014).
  • Treg cells infiltrating non-lymphoid tissues are reported to exhibit unique phenotypes and transcriptional signatures, as they can display functions beyond their well-established suppressive roles, such as metabolic modulation in adipose tissue (Cipolletta et al., 2012) or regulation of tissue repair in skeletal muscle (Burzyn et al., 2013) and in lung tissue (Arpaia et al., 2015).
  • Treg cells depletion has been reported to increase anti-tumor specific immune responses and to reduce tumor burden (Marabelle et al., 2013; Teng et al., 2010; Walter et al., 2012). Although promising clinical results have been achieved with Treg cell depleting strategies, some relevant issues are to be addressed, for a safer, more effective and wider clinical application of these therapies.
  • severe autoimmunity can occur following systemic Treg cells depletion (Nishikawa and Sakaguchi, 2010), which could be avoided if selective depletion of tumor infiltrating Treg cells were feasible.
  • a second issue concerns the specificity of targeting, indeed Treg cells share with effector lymphocytes most of the molecules targeted for therapy, which can possibly deplete also the tumor-specific effector cells. Therefore, the molecular characterization of Treg cells at different tumor sites should help to better define therapeutic targets through a better description of their signature molecules and of the network that regulates Treg cell functions in the tumor microenvironment.
  • Non-small-cell lung cancer (NSCLC) and colorectal cancer (CRC) are the two most frequent cancers in both genders (Torre et al., 2015). NSCLC has the worst prognosis due to its high mortality rate even in early stages. Although CRC survival rate is highly dependent on the tumor stage at diagnosis, about 50% of patients will progress to metastatic cancer (Gonzalez-Pons and Cruz-Correa, 2015). Both tumors have been targeted with therapies based on monoclonal antibodies to checkpoint inhibitors, but the outcomes were different.
  • Tumor-infiltrating regulatory T lymphocytes can suppress effector T cells specific for tumor antigens. Since new anti-cancer immunotherapies aim at unleashing effector T cells by targeting immune-checkpoints, deeper molecular definitions of tumor-infiltrating-lymphocytes could offer new therapeutic opportunities.
  • Transcriptomes of T helper 1 (Th1), Th17 and Treg cells infiltrating colorectal or non-small-cell lung cancers were compared to transcriptomes of the same subsets from normal tissues, and validated at the single cell level.
  • Treg cells are highly suppressive, upregulate several immune-checkpoints, and express on the cell surface specific signature molecules such as interleukin-1 receptor 2 (IL1R2), programmed death (PD)-1 Ligand1, PD-1 Ligand2, and CCR8 chemokine which were not previously described on Treg cells.
  • IIL1R2 interleukin-1 receptor 2
  • PD programmed death
  • CCR8 chemokine which were not previously described on Treg cells.
  • high expression in whole tumor samples of Treg signature genes such as LAYN, MAGEH1 or CCR8, correlated with poor prognosis.
  • the invention provides new insights into the molecular identity and functions of human tumor-infiltrating Treg cells, and define new potential targets for tumor immunotherapy.
  • the inventors provide a comprehensive transcriptome analysis of human CD4 + Treg cells and effector cells (Th1 and Th17) infiltrating NSCLC or CRC and their matched normal tissues.
  • the inventors' findings provide new insights on the inhibitory mechanisms of Treg cells and offer precise targets for cancer immunotherapy.
  • the present invention provides a molecule able to modulate the expression and/or function of at least one marker that is selectively deregulated in tumor-infiltrating regulatory T cells for use in the prevention and/or treatment of said tumor.
  • the molecule according to the invention is capable of specifically binding to said at least one marker and inducing antibody-dependent cell-mediated cytotoxicity (ADCC).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • Said molecule is preferably able to selectively deplete tumor-infiltrating regulatory T cells.
  • Said molecule is preferably selected from the group consisting of:
  • a polynucleotide such as antisense construct, antisense oligonucleotide, RNA interference construct or siRNA,
  • the marker is selected from the group consisting of at least one marker disclosed in the following Table VIII.
  • each of said marker name is characterized by “Ensembl gene id” and includes all of therein disclosed isoform protein sequences.
  • Each gene of table VIII is characterized by its Ensembl Gene accession number (ENSG), retrievable in the public database EnsEMBL (http://www.ensembl.org) and by its Entrez Gene ID, retrievable in the public database NCBI (https://www.ncbi.nlm.nih.gov/), if present.
  • ENSG Ensembl Gene accession number
  • EnsEMBL http://www.ensembl.org
  • NCBI https://www.ncbi.nlm.nih.gov/
  • the marker is selected from the group consisting of: a transmembrane protein, a cytokine, an epigenetic factor, a kinase phosphatase or a transcription factor.
  • the marker is a transmembrane protein selected from the group of SEQ ID NO:1-661, even more preferably, the marker is selected from the group consisting of: LAYN (SEQ ID NOs:1-9), CCR8 (SEQ ID Nos:10-11), IL21R (SEQ ID Nos: 12-14), IL1R2 (SEQ ID Nos:206-209), LY75 (SEQ ID NO: 78), SIRPG (SEQ ID Nos:122-126), CD177 (SEQ ID Nos:651-653), CD7 (SEQ ID Nos:549-554), FCRL3 (SEQ ID Nos:452-457), CADM1 (SEQ ID Nos: 570-583), NTNG2 (SEQ ID Nos:621-622), CSF2RB (SEQ ID Nos:134-137), SECTM1 (SEQ ID Nos: 349-356), TSPAN5 (SEQ ID Nos:497-503), TMPRSS3 (SEQ ID Nos
  • LAYN (SEQ ID NOs: 1-9 [>ENSG00000204381_ENST00000375614_ ENSP00000364764_LAYN (SEQ ID NO: 1) MRPGTALQAVLLAVLLVGLRAATGRLLSGQPVCRGGTQRPCYKVIYFH DTSRRLNFEEAKEACRRDGGQLVSIESEDEQKLIEKFIENLLPSDGDF WIGLRRREEKQSNSTACQDLYAWTDGSISQFRNWYVDEPSCGSEVCVV MYHQPSAPAGIGGPYMFQWNDDRCNMKNNFICKYSDEKPAVPSREAEG EETELTTPVLPEETQEEDAKKTFKESREAALNLAYILIPSIPLLLLLV VTTVVCWVWICRKRKREQPDPSTKKQHTIWPSPHQGNSPDLEVYNVIR KQSEADLAETRPDLKNISFRVCSGEATPDDMSCDYDNMAVNPSESGFV TLVSVESGFVTNDIYEF
  • Said cytokine is preferably selected from the group of consisting of: IL32 (SEQ ID Nos: 19-30), IL7 (SEQ ID Nos: 168-174), EBI3 (SEQ ID NO: 175), SECTM1 (SEQ ID Nos: 349-356), CSF1 (SEQ ID Nos: 585-592) and LTA (SEQ ID Nos: 657-658).
  • Said epigenetic factor is preferably selected from the group of consisting of: TDRD3 (SEQ ID NO: 712-718), KAT2B (SEQ ID NO:719), FOXA1 (SEQ ID Nos: 720-721) and RCBTB1 (SEQ ID Nos: 722-723).
  • Said kinase phosphatase is preferably selected from the group of consisting of: GSK3B (SEQ ID Nos: 724-725), SSH1 (SEQ ID NOS:111-112), CDK6 (SEQ ID Nos: 726-727), MINPP1 (SEQ ID Nos:181-183), PTPRJ (SEQ ID Nos: 395-400), CALM3 (SEQ ID Nos: 728-734) and PTP4A3 (SEQ ID Nos: 593-598).
  • Said transcription factor is preferably selected from the group of consisting of:
  • VDR (SEQ ID NO:204), ZNF334 (SEQ ID Nos: 736-741), CREB3L2 (SEQ ID Nos: 565-567), ETV7 (SEQ ID NO:31 or 32), SOX4 (SEQ ID NO:735), TWIST1 (SEQ ID Nos: 743-745), TP73 (SEQ ID Nos: 746-756), FOXP3, NFE2L3 (SEQ ID NO:76), ARNTL2 (SEQ ID Nos: 757-764), BATF (SEQ ID Nos: 765-766), PTTG1 (SEQ ID Nos: 767-770), HIVEP3 (SEQ ID Nos: 771-772), FOXA1 (SEQ ID Nos: 720-721), ZBTB38 (SEQ ID NO:561), FOXM1 (SEQ ID Nos: 773-778), TADA3 (SEQ ID Nos: 779-782), NFAT5 (SEQ ID NO:160, 783-791
  • the marker is MAGEH1 (SEQ ID NO: 708 or 709)
  • the tumor is preferably a solid or liquid tumor.
  • the solid tumor is selected from the group consisting of: non-small cell lung cancer, colorectal cancer, breast cancer, gastric cancer.
  • the tumor is a metastasis, preferably a bone, a brain or a liver metastasis.
  • the metastasis derives from colon rectal cancer or non-small-cell lung cancer.
  • Another object of the invention is the above defined molecule for use in a method for in vivo depleting tumor-infiltrating regulatory T cells in a subject or for use in a method to enhance tumor immunity in a subject.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising the molecule as defined above and at least one pharmaceutically acceptable carrier.
  • a further object of the invention is a pharmaceutical composition comprising the molecule as above defined, for use in the prevention and/or treatment of tumor or for use in a method for in vivo depleting tumor-infiltrating regulatory T cell in a subject or for use in a method to enhance tumor immunity in a subject.
  • the pharmaceutical composition according to the invention may further comprise a therapeutic agent, preferably the therapeutic agent in an anti-tumoral agent.
  • Another object of the invention is an in vitro method for diagnosing and/or assessing the risk of developing and/or prognosing and/or for monitoring the progression and/or for monitoring the efficacy of a therapeutic treatment and/or for the screening of a therapeutic treatment of a tumour in a subject comprising the steps of:
  • Another object of the invention is an in vitro or ex-vivo method for diagnosing and/or assessing the risk of developing and/or prognosing and/or for monitoring the progression and/or for monitoring the efficacy of a therapeutic treatment and/or for the screening of a therapeutic treatment of a tumour in a subject as above defined, wherein the marker to be detected is at least one of the marker selected from the group consisting of: LAYN, MAGEH1 and CCR8.
  • the above method is for prognosing of colorectal cancer or non-small cell lung cancer in a subject and comprises the steps of:
  • an amount of said at least one marker in the isolated biological sample obtained from the subject higher than the control amount indicates that the subject has a poor prognosis.
  • step a) comprises measuring the amount of the marker or of fragments thereof or of the polynucleotide coding for said protein (DNA or mRNA) or of fragments thereof in said isolated biological sample obtained from the subject and step b) comprises comparing the measured amount of step a) with a proper control amount.
  • the in vitro method for monitoring the progression and/or for monitoring the efficacy of a therapeutic treatment of a tumour comprises the steps of:
  • step b) comparing the measured alteration of step a) with a proper control alteration.
  • Another object of the invention is a method for the treatment and/or prevention of tumor comprising administering to a subject the molecule as above defined.
  • a further object is a method for identifying a molecule acting as an anti-tumoral, comprising the steps of:
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • the biological sample is a fluid, a cell or a tissue sample, more preferably said sample is plasma or serum.
  • biological sample encompasses a clinical sample, and also includes tissue obtained by surgical resection, tissue obtained by biopsy, cells in culture, cell supernatants, cell lysates, tissue samples, organs, bone marrow, blood, plasma, serum, and the like.
  • sample in the context of the present teachings refers to any biological sample that is isolated from a subject.
  • a sample can include, without limitation an aliquot of body fluid, whole blood, serum, plasma, solid tissue samples such as tissue biopsies, or tissue cultures or cells derived therefrom and the progeny thereof, synovial fluid, lymphatic fluid, ascites fluid, and interstitial or extracellular fluid.
  • sample also encompasses the fluid in spaces between cells, including gingival crevicular fluid, bone marrow, cerebrospinal fluid (CSF), saliva, mucous, sputum, semen, sweat, urine, or any other bodily fluids.
  • Bood sample can refer to whole blood or any fraction thereof, including serum and plasma.
  • Samples can be obtained from a subject by means including but not limited to venipuncture, excretion, ejaculation, massage, biopsy, needle aspirate, lavage, scraping, surgical incision, or intervention or other means known in the art.
  • the definition also includes samples that have been manipulated in any way after their procurement, such as by treatment with reagents; washed; or enrichment for certain cell populations, such as cancer cells or samples in which regulatory T cells, are isolated and then analyzed.
  • the definition also includes sample that have been enriched for particular types of molecules, e.g., nucleic acids, polypeptides, etc.
  • Another object of the invention is a kit for carrying out the above methods, comprising
  • markers are LAYN and MAGEH1; LAYN and CCR8; CCR8 and MAGEH1; LAYN, MAGEH1 and CCR8.
  • the above polynucleotide is an RNAi inhibitor, preferably selected from the group consisting of: siRNA, miRNA, shRNA, stRNA, snRNA, and antisense nucleic acid, or a functional derivative thereof.
  • RNAi inhibitor preferably selected from the group consisting of: siRNA, miRNA, shRNA, stRNA, snRNA, and antisense nucleic acid, or a functional derivative thereof.
  • molecule able to modulate and “modulator” are herein interchangeable.
  • modulator it is meant a molecule that effects a change in the expression and/or function of at least one marker as above defined.
  • a “modulator” is a molecule which may suppress or inhibit the expression and/or function of at least one marker that is selectively deregulated in tumor-infiltrating regulatory T cell for use in the prevention and/or treatment of cancer.
  • suppressor or inhibitor or a “molecule which (selectively) suppresses or inhibits” it is meant a molecule that effects a change in the expression and/or function of the target.
  • a “modulator” is a molecule which may induce or activate the expression and/or function of at least one marker that is selectively deregulated in tumor-infiltrating regulatory T cell for use in the prevention and/or treatment of cancer.
  • the change is relative to the normal or baseline level of expression and/or function in the absence of the modulator, but otherwise under similar conditions, and it may represent an increase (e.g. by using an inducer or activator) or a decrease (e.g. by using a suppressor or inhibitor) in the normal/baseline expression and/or function.
  • the suppression or inhibition of the expression and/or function of the target may be assessed by any means known to the skilled in the art.
  • the assessment of the expression level or of the presence of the target is preferably performed using classical molecular biology techniques such as (real time Polymerase Chain Reaction) qPCR, microarrays, bead arrays, RNAse protection analysis or Northern blot analysis or cloning and sequencing.
  • the assessment of target function is preferably performed by in vitro suppression assay, whole transcriptome analysis, mass spectrometry analysis to identify proteins interacting with the target.
  • the target may be the gene, the mRNA, the cDNA, or the encoded protein thereof, including fragments, derivatives, variants, isoforms, etc.
  • the marker is characterized by its Accession numbers (i.e. NCBI Entrez ID; Ensembl Gene accession number (ENSG), Ensembl transcript accession number (ENST) and Ensembl protein accession number (ENSP), retrievable in the public database EnsEMBL (http://www.ensembl.org) and/or amino acid and nucleotide sequences, herein disclosed.
  • the term “treat” when referred to CD4+ T cell, means e.g. the exposure of the cell to an exogenous modulator as above defined.
  • the overexpression may be obtained e.g. by infecting the cells with a viral vector expressing the molecule of the invention.
  • the inhibition of marker expression may e.g. by obtained by transfection with polynucleotide, as e.g. with siRNAs.
  • the term “treat” may also mean that the cells are manipulated in order to overexpress or silence the marker.
  • the overexpression or the silencing may be obtained e.g. by genetically modifying the cells.
  • Control means can be used to compare the amount or the increase of amount of the marker defined to a proper control.
  • the proper control may be obtained for example, with reference to known standard, either from a normal subject or from normal population, or from T cells different from tumour infiltrating regulatory T cells or regulatory T cells.
  • the means to measure the amount of at least one marker as above defined are preferably at least one antibody, functional analogous or derivatives thereof. Said antibody, functional analogous or derivatives thereof are specific for said marker.
  • the antibody is preferably selected from the group consisting of an intact immunoglobulin, a Fv, a scFv (single chain Fv fragment), a Fab, a F(ab′)2, an “antibody-like” domain, an “antibody-mimetic domain”, a single antibody domain (VH domain or VL domains), a multimeric antibody, recombinant or synthetic antigen-binding fragments, a peptide or a proteolytic fragment containing the epitope binding region.
  • antibody and “immunoglobulin” can be used interchangeably and are herein used in the broadest sense and encompass various antibodies and antibody mimetics structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), chimeric antibodies, nanobodies, antibody derivatives, antibody fragments, anticalins, DARPins, affibodies, affilins, affimers, affitins, alphabodies, avimers, fynomers, monobodies and other binding domains, so long as they exhibit the desired antigen-binding activity.
  • monoclonal antibodies polyclonal antibodies
  • multispecific antibodies e.g., bispecific antibodies
  • chimeric antibodies nanobodies, antibody derivatives, antibody fragments, anticalins, DARPins, affibodies, affilins, affimers, affitins, alphabodies, avimers, fynomers, monobodies and other
  • immunoglobulin also includes “conjugate” thereof.
  • conjugated in relation to the antibody of the invention includes antibodies (or fragments thereof) conjugated with a substance (a compound, etc.) having a therapeutic activity, e.g. anti-tumor activity and/or cell-killing activity or a cytotoxic agents such as various A chain toxins, ribosomes inactivating proteins, and ribonucleases; bispecific antibodies designed to induce cellular mechanisms for killing tumors (see, for example, U.S. Pat. Nos. 4,676,980 and 4,954,617).
  • the conjugate may be formed by previously preparing each of the aforementioned antibody molecule and the aforementioned substance having anti-tumor activity and/or cell-killing activity, separately, and then combining them (immunoconjugate) or by ligating a protein toxin used as such a substance having anti-tumor activity and/or cell-killing activity to an antibody gene on a gene according to a genetic recombination technique, so as to allow it to express as a single protein (a fusion protein) (immunotoxin).
  • antibody fragment refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab′, Fab′-SH, F(ab′)2; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
  • VH or VL Fvs are also called “Nanobodies”.
  • antibody mimetics refers to those organic compounds or binding domains that are not antibody derivatives but that can bind specifically an antigen like antibodies do. They include anticalins, DARPins, affibodies, affilins, affimers, affitins, alphabodies, avimers, fynomers, monobodies and others.
  • chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.
  • full length antibody “intact antibody,” and “whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
  • the kit of the invention comprises:
  • the reagents can be provided as a kit comprising reagents in a suspension or suspendable form, e.g. reagents bound to beads suitable for flow cytometry, preferably magnetic beads coated with antibody capture.
  • the instructions may comprise instructions for conducting an antibody-based flow cytometry assay.
  • Detection means are preferably means able to detect and/or measure the amount of the described markers, e.g. means able to detect the complex antigen-antibody, as enzyme conjugated secondary antibodies, luminescent substrates, magnetic beads coated with antibody capture, customized dried antibody cocktails and/or columns with size filter cartridges and/or combined with specific antibody filter (SAF).
  • SAF specific antibody filter
  • the method further comprises selecting a therapeutic regimen based on the analysis. In an embodiment, the method further comprises determining a treatment course for the subject based on the analysis. Other means may be e.g. specific primers and probes for RT PCR.
  • the kits according to the invention can further comprise customary auxiliaries, such as buffers, carriers, markers, etc. and/or instructions for use. In the context of the present invention the term “detecting” may be intended also as “measuring the amount” or “measuring the alteration”.
  • the proper control may be a sample taken from a healthy patient or from a patient affected by another disorder or pathology, and the proper control amount or activity may be the amount or activity of the same protein or polynucleotide measured in a sample taken from a healthy patient or from a patient affected by another disorder or pathology.
  • the progress of the cancer is monitored and the proper control may be a sample taken from the same subject at various times or from another patient, and the proper control amount or activity may by the amount or activity of the same protein or polynucleotide measured in a sample taken from the same subject at various times or from another patient.
  • the proper control may by a sample taken from the same subject before initiation of the therapy or taken at various times during the course of the therapy and the proper control amount or activity may be the amount or activity of the same protein or polynucleotide measured in a sample taken from the same subject before initiation of the therapy or taken at various times during the course of the therapy.
  • the proper control may be a sample taken from subjects without treatment and from subjects treated with a substance that is to be assayed or from subjects treated with a reference treatment and the proper control amount or activity may be the average of the amounts or activity of the same protein or polynucleotide measured in samples taken from subjects without treatment and from subjects treated with a substance that is to be assayed or from subjects treated with a reference treatment.
  • the amount or activity of MAGEH1 and/or LAYN and/or CCR8 or polynucleotides thereof in the isolated biological sample obtained from the subject is lower or equal than the control amount or activity, it may indicate that the tested substance is effective for the treatment of the tumour.
  • the expression “measuring the amount” can be intended as measuring the amount (or the activity) or concentration or level of the respective protein and/or mRNA thereof and/or DNA thereof, preferably semi-quantitative or quantitative.
  • Measurement of a protein can be performed directly or indirectly.
  • Direct measurement refers to the amount or concentration measure of the marker, based on a signal obtained directly from the protein, and which is directly correlated with the number of protein molecules present in the sample. This signal—which can also be referred to as intensity signal—can be obtained, for example, by measuring an intensity value of a chemical or physical property of the marker.
  • Indirect measurements include the measurement obtained from a secondary component (e.g., a different component from the gene expression product) and a biological measurement system (e.g. the measurement of cellular responses, ligands, “tags” or enzymatic reaction products).
  • amount refers but is not limited to the absolute or relative amount of proteins and/or mRNA thereof and/or DNA thereof, and any other value or parameter associated with the same or which may result from these.
  • values or parameters comprise intensity values of the signal obtained from either physical or chemical properties of the protein, obtained by direct measurement, for example, intensity values in an immunoassay, mass spectroscopy or a nuclear magnetic resonance. Additionally, these values or parameters include those obtained by indirect measurement, for example, any of the measurement systems described herein. Methods of measuring mRNA and DNA in samples are known in the art.
  • the cells in a test sample can be lysed, and the levels of mRNA in the lysates or in RNA purified or semi-purified from lysates can be measured by any variety of methods familiar to those in the art. Such methods include hybridization assays using detectably labeled DNA or RNA probes (i.e., Northern blotting) or quantitative or semi-quantitative RT-PCR methodologies using appropriate oligonucleotide primers. Alternatively, quantitative or semi-quantitative in situ hybridization assays can be carried out using, for example, tissue sections, or unlysed cell suspensions, and detectably labeled (e.g., fluorescent, or enzyme-labeled) DNA or RNA probes. Additional methods for quantifying mRNA include RNA protection assay (RPA), cDNA and oligonucleotide microarrays, representation difference analysis (RDA), differential display, EST sequence analysis, and serial analysis of gene expression (SAGE).
  • RPA RNA protection assay
  • RDA representation difference
  • the subject may present cancer or go towards an aggravation of said disease.
  • the subject may be not affected by cancer or go toward an amelioration of cancer, respectively.
  • the expression “detecting” or “measuring the amount” is intended as measuring the alteration of the molecule.
  • Said alteration can reflect an increase or a decrease in the amount or activity of the molecules as above defined.
  • An increase of the protein or of the activity of the marker or of the polynucleotide coding for said marker can be correlated to an aggravation of cancer.
  • a decrease of the protein or of the activity of said marker or of the polynucleotide coding for said protein can be correlated to an amelioration of cancer or to recovery of the subject.
  • marker is intended to include also the corresponding protein encoded from said marker orthologous or homologous genes, functional mutants, functional derivatives, functional fragments or analogues, isoforms, splice variants thereof.
  • markers When the expression “marker” is referred to genes, it is intended to include also the corresponding orthologous or homologous genes, functional mutants, functional derivatives, functional fragments or analogues, isoforms thereof.
  • fragments refers to polynucleotides having preferably a length of at least 1000 nucleotides, 1100 nucleotide, 1200 nucleotides, 1300 nucleotides, 1400 nucleotides, 1500 nucleotides.
  • fragments refers to polypeptides having preferably a length of at least 10 amino acids, more preferably at least 15, at least 17 amino acids or at least 20 amino acids, even more preferably at least 25 amino acids or at least 37 or 40 amino acids, and more preferably of at least 50, or 100, or 150 or 200 or 250 or 300 or 350 or 400 or 450 or 500 amino acids.
  • polynucleotide also refers to modified polynucleotides.
  • vector refers to an expression vector, and may be for example in the form of a plasmid, a viral particle, a phage, etc.
  • vectors may include bacterial plasmids, phage DNA, baculovirus, yeast plasmids, vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, lentivirus, fowl pox virus, and pseudorabies. Large numbers of suitable vectors are known to those of skill in the art and are commercially available.
  • the polynucleotide sequence preferably the DNA sequence in the vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis.
  • promoter an appropriate expression control sequence(s)
  • prokaryotic or eukaryotic promoters such as CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I.
  • the expression vector may also contain a ribosome binding site for translation initiation and a transcription vector.
  • the vector may also include appropriate sequences for amplifying expression.
  • the vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydro folate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E. coli.
  • the term “host cell genetically engineered” relates to host cells which have been transduced, transformed or transfected with the polynucleotide or with the vector described previously.
  • appropriate host cells one can cite bacterial cells, such as E. coli, Streptomyces, Salmonella typhimurium , fungal cells such as yeast, insect cells such as Sf9, animal cells such as CHO or COS, plant cells, etc.
  • bacterial cells such as E. coli, Streptomyces, Salmonella typhimurium
  • fungal cells such as yeast
  • insect cells such as Sf9
  • animal cells such as CHO or COS, plant cells, etc.
  • said host cell is an animal cell, and most preferably a human cell.
  • the introduction of the polynucleotide or of the vector described previously into the host cell can be effected by method well known from one of skill in the art such as calcium phosphate transfection, DEAE-Dextran mediated transfection, electroporation, lipofection, microinjection, viral infection, thermal shock, transformation after chemical permeabilisation of the membrane or cell fusion.
  • the polynucleotide may be a vector such as for example a viral vector.
  • polynucleotides as above defined can be introduced into the body of the subject to be treated as a nucleic acid within a vector which replicates into the host cells and produces the polynucleotides or the proteins.
  • Suitable administration routes of the pharmaceutical composition of the invention include, but are not limited to, oral, rectal, transmucosal, intestinal, enteral, topical, suppository, through inhalation, intrathecal, intraventricular, intraperitoneal, intranasal, intraocular and parenteral (e.g., intravenous, intramuscular, intramedullary, and subcutaneous).
  • An additional suitable administration route includes chemoembolization.
  • Other suitable administration methods include injection, viral transfer, use of liposomes, e.g. cationic liposomes, oral intake and/or dermal application.
  • a pharmaceutical composition of the present invention is administered in the form of a dosage unit (e.g., tablet, capsule, bolus, etc.).
  • a dosage unit e.g., tablet, capsule, bolus, etc.
  • the composition may be in the form of a solution, e.g. an injectable solution, emulsion, suspension or the like.
  • the carrier may be any suitable pharmaceutical carrier.
  • a carrier is used which is capable of increasing the efficacy of the RNA molecules to enter the target cells. Suitable examples of such carriers are liposomes.
  • the modulator as above defined is administered in a pharmaceutically effective dosage, which in the case of polynucleotides may be in the range of 0.001 pg/kg body weight to 10 mg/kg body weight depending on the route of administration and the type or severity of the disease.
  • composition refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • effective amount shall mean an amount which achieves a desired effect or therapeutic effect as such effect is understood by those of ordinary skill in the art.
  • the antibody may be administered simultaneously or sequentially with another therapeutic treatment, that may be a chemotherapy or radiotherapy.
  • the invention provides formulations comprising a therapeutically effective amount of an antibody as disclosed herein, a buffer maintaining the pH in the range from about 4.5 to about 8.5, and, optionally, a surfactant.
  • the formulations are typically for an antibody as disclosed herein, recombinant or synthetic antigen-binding fragments thereof of the invention as active principle concentration from about 0.1 mg/ml to about 100 mg/ml.
  • the antibody, recombinant or synthetic antigen-binding fragments thereof concentration is from about 0.1 mg/ml to 1 mg/ml; preferably from 1 mg/ml to 10 mg/ml, preferably from 10 to 100 mg/ml.
  • Therapeutic formulations of the antibody/antibodies can be prepared by mixing the antibody having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed., 1980), in the form of lyophilized formulations or aqueous solutions.
  • compositions containing the antibody of the present invention may be manufactured by processes well known in the art, e.g., using a variety of well-known mixing, dissolving, granulating, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes. Proper formulation is dependent upon the route of administration chosen. Parenteral routes are preferred in many aspects of the invention.
  • the compounds of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as physiological saline buffer or polar solvents including, without limitation, a pyrrolidone or dimethylsulfoxide.
  • physiologically compatible buffers such as physiological saline buffer or polar solvents including, without limitation, a pyrrolidone or dimethylsulfoxide.
  • Formulations for injection may be presented in unit dosage form, e.g. in ampoules or in multi-dose containers.
  • Useful compositions include, without limitation, suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain adjuncts such as suspending, stabilizing and/or dispersing agents.
  • Pharmaceutical compositions for parenteral administration include aqueous solutions of a water soluble form, such as, without limitation, a salt of the active compound.
  • suspensions of the active compounds may be prepared in a lipophilic vehicle. Suitable lipophilic vehicles include fatty oils such as sesame oil, synthetic fatty acid esters such as ethyl oleate and triglycerides, or materials such as liposomes.
  • Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers and/or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • the antibody of the present invention can conveniently be delivered in the form of an aerosol spray using a pressurized pack or a nebulizer and a suitable propellant.
  • the antibody may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
  • the antibody may also be formulated as depot preparations.
  • Such long acting formulations may be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds of this invention may be formulated for this route of administration with suitable polymeric or hydrophobic materials (for instance, in an emulsion with a pharmacologically acceptable oil), with ion exchange resins, or as a sparingly soluble derivative such as, without limitation, a sparingly soluble salt.
  • the antibody may be delivered using a sustained-release system, such as semi-permeable matrices of solid hydrophobic polymers containing the therapeutic agent. Other delivery systems such as liposomes and emulsions can also be used.
  • a therapeutically effective amount refers to an amount of compound effective to prevent, alleviate or ameliorate cancer or cancer recurrence symptoms. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the disclosure herein.
  • the therapeutically effective amount can be estimated initially from in vitro assays. Then, the dosage can be formulated for use in animal models so as to achieve a circulating concentration range that includes the effective dosage. Such information can then be used to more accurately determine dosages useful in patients.
  • the amount of the composition that is administered will depend upon the parent molecule included therein. Generally, the amount used in the treatment methods is that amount which effectively achieves the desired therapeutic result in mammals.
  • the dosages of the various compounds can vary somewhat depending upon the compound, rate of in vivo hydrolysis, etc.
  • the dosage can vary depending upon the dosage form and route of administration.
  • the range set forth above is illustrative and those skilled in the art will determine the optimal dosing of the compound selected based on clinical experience and the treatment indication.
  • the exact formulation, route of administration and dosage can be selected by the individual physician in view of the patient's condition and of the most effective route of administration (e.g., intravenous, subcutaneous, intradermal).
  • toxicity and therapeutic efficacy of the antibody and other therapeutic agent described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals using methods well-known in the art.
  • the treatment will be given for one or more cycles until the desired clinical and biological result is obtained.
  • the exact amount, frequency and period of administration of the compound of the present invention will vary, of course, depending upon the sex, age and medical condition of the patient as well as the severity and type of the disease as determined by the attending clinician.
  • the modulator of the present invention may comprise a single type of modulator or a plurality of different modulators.
  • the function of a regulatory T-cell may be inhibited by inhibiting markers activity and/or expression or by decreasing the number of cells positive for such markers in a T-cell population (for example by binding at least one of the above marker and inducing antibody-dependent cell-mediated cytotoxicity (ADCC)). Inhibiting the function of regulatory T-cells in an organism may be used to enhance the immune T-cell response in those circumstances where such a response is desirable, such as in a patient suffering from cancer.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • an anti-tumor vaccine or therapy When treating a cancer patient with an inhibitory agent that binds to marker protein or mRNA, one may optionally co-administer an anti-tumor vaccine or therapy.
  • Such vaccines may be directed to isolated antigens or to groups of antigens or to whole tumor cells. It may be desirable to administer the inhibitory agent with chemotherapeutic agents or together with radiotherapy.
  • Treatment with multiple agents need not be done using a mixture of agents but may be done using separate pharmaceutical preparations.
  • the preparations need not be delivered at the same exact time, but may be coordinated to be delivered to a patient during the same period of treatment, i.e. within a week or a month or each other.
  • compositions comprising two active ingredients may be constituted in the body of the patient.
  • Any suitable anti-tumor treatment can be coordinated with the treatments of the present invention targeted to the markers.
  • other anti-infection agents can be coordinated with the treatment of the present invention targeted to the markers.
  • agents may be small molecule drugs, vaccines, antibodies, etc.
  • the number of marker+ cells in a T-cell population can be modified by using an antibody or other agent that selectively binds to the marker.
  • marker+ cells represent an enriched population of regulatory T-cells that can be introduced back into the original source of the T-cells or into another compatible host to enhance regulatory T-cell function.
  • the marker-cells represent a population of T-cells deficient in regulatory T-cell activity that can be reintroduced into the original source of the T-cells or another compatible host to inhibit or reduce regulatory T-cell function while retaining general T-cell activity.
  • Any desired means for either increasing or decreasing (modulating) marker activity can be used in the methods of the invention. This includes directly modulating the function of marker protein, modulating marker signal transduction, and modulating expression of marker in T-cells by modulating either transcription or translation or both. Those means which selectively modulate marker activity are preferred over nonselective modulators. Also, those inhibitory means which create a transient marker deficiency in a population of T-cells which then return to normal levels of marker activity may be preferred for treating a temporary T-cell deficiency. The transiently deficient T-cells may be used to reconstitute a diminished T-cell population with T-cells that will be genetically normal with respect to the marker.
  • Modulation of marker activity can be performed on cells in vitro or in whole animals, in vivo.
  • Cells which are treated in vitro can be administered to a patient, either the original source of the cells or an unrelated individual.
  • marker antibodies or small molecule inhibitors can be used.
  • Antibodies or antibody fragments that are useful for this purpose will be those that can bind to the marker and block its ability to function.
  • Such antibodies may be polyclonal antibodies, monoclonal antibodies, chimeric antibodies, humanized antibodies, single-chain antibodies, soluble MHC class II molecules, antibody fragments, etc.
  • Antibodies generated against marker polypeptides can be obtained by direct injection of the marker polypeptides into an animal or by administering marker polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the marker polypeptides itself. In this manner, even a sequence encoding only a fragment of the marker polypeptide can be used to generate antibodies binding the whole native marker polypeptide.
  • any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler and Milstein, 1975, Nature, 256: 495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4: 72), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole, et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). Techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778) can be readily used to produce single chain antibodies to marker polypeptides.
  • transgenic mice may be used to express humanized antibodies to immunogenic marker polypeptides.
  • any agent which increases the level of the marker or the activity of existing marker in the T-cell may be used. Such agents may be identified using the screening assays described below.
  • Expression vectors encoding the marker can also be administered to increase the gene dosage.
  • the expression vectors can be plasmid vectors or viral vectors, as are known in the art. Any vector can be chosen by the skilled in the art for particularly desirable properties.
  • polynucleotide includes DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA, siRNA, shRNA) and analogues of the DNA or RNA generated using nucleotide analogues.
  • the polynucleotide may be single-stranded or double-stranded.
  • the polynucleotide may be synthesized using oligonucleotide analogues or derivatives (e.g., inosine or phosphorothioate nucleotides).
  • RNAi inhibitors as above defined are preferably capable of hybridizing to all or part of specific target sequence. Therefore, RNAi inhibitors may be fully or partly complementary to all of or part of the target sequence
  • RNAi inhibitors may hybridize to the specified target sequence under conditions of medium to high stringency.
  • RNAi inhibitors may be defined with reference to a specific sequence identity to the reverse complement of the sequence to which it is intended to target.
  • the antisense sequences will typically have at least about 75%, preferably at least about 80%, at least about 85%, at least about 90%, at least about 95% or at least about 99% sequence identity with the reverse complements of their target sequences.
  • polynucleotide and polypeptide also includes derivatives and functional fragments thereof.
  • the at least one gene or marker as above defined is preferably characterized by at least one of the sequence identified by its Ensembl Gene ID or NCBI Accession Numbers, as disclosed in Tables VIII or VI, or by at least one of the SEQ ID No. 1-709.
  • gene herein also includes corresponding orthologous or homologous genes, isoforms, variants, allelic variants, functional derivatives, functional fragments thereof.
  • protein is intended to include also the corresponding protein encoded from a corresponding orthologous or homologous genes, functional mutants, functional derivatives, functional fragments or analogues, isoforms thereof.
  • analogue as used herein referring to a protein means a modified peptide wherein one or more amino acid residues of the peptide have been substituted by other amino acid residues and/or wherein one or more amino acid residues have been deleted from the peptide and/or wherein one or more amino acid residues have been deleted from the peptide and or wherein one or more amino acid residues have been added to the peptide.
  • Such addition or deletion of amino acid residues can take place at the N-terminal of the peptide and/or at the C-terminal of the peptide.
  • a “derivative” may be a nucleic acid molecule, as a DNA molecule, coding the polynucleotide as above defined, or a nucleic acid molecule comprising the polynucleotide as above defined, or a polynucleotide of complementary sequence.
  • the term “derivatives” also refers to longer or shorter polynucleotides and/or polypeptides having e.g. a percentage of identity of at least 41%, 50%, 60%, 65%, 70% or 75%, more preferably of at least 85%, as an example of at least 90%, and even more preferably of at least 95% or 100% with the sequences herein mentioned or with their complementary sequence or with their DNA or RNA corresponding sequence.
  • the term “derivatives” and the term “polynucleotide” also include modified synthetic oligonucleotides.
  • the modified synthetic oligonucleotide are preferably LNA (Locked Nucleic Acid), phosphoro-thiolated oligos or methylated oligos, morpholinos, 2′-O-methyl, 2′-O-methoxyethyl oligonucleotides and cholesterol-conjugated 2′-O-methyl modified oligonucleotides (antagomirs).
  • derivative may also include nucleotide analogues, i.e. a naturally occurring ribonucleotide or deoxyribonucleotide substituted by a non-naturally occurring nucleotide.
  • derivatives also includes nucleic acids or polypeptides that may be generated by mutating one or more nucleotide or amino acid in their sequences, equivalents or precursor sequences.
  • derivatives also includes at least one functional fragment of the polynucleotide.
  • the vector as above defined is preferably selected from the group consisting of: plasmids, viral vectors and phages, more preferably the viral vector is a lentiviral vector.
  • the host cell as above defined is preferably selected from the group consisting of: bacterial cells, fungal cells, insect cells, animal cells, plant cells, preferably being an animal cell.
  • compositions comprising a mixture of antibodies which specifically bind to the marker(s); and an anti-cancer vaccine can be made in vitro.
  • the composition is made under conditions which render it suitable for use as a pharmaceutical composition.
  • Pharmaceutical compositions may be sterile and pyrogen-free.
  • the components of the composition can also be administered separately to a patient within a period of time such that they are both within the patient's body at the same time. Such a time-separated administration leads to formation of the mixture of antibodies and vaccine within the patient's body. If the antibody and vaccine are to be administered in a time-separated fashion, they may be supplied together in a kit. Within the kit the components may be separately packaged or contained.
  • kits Other components such as excipients, carriers, other immune modulators or adjuvants, instructions for administration of the antibody and the vaccine, and injection devices can be supplied in the kit as well. Instructions can be in a written, video, or audio form, can be contained on paper, an electronic medium, or even as a reference to another source, such as a website or reference manual.
  • Anti-marker antibodies of the invention can be used to increase the magnitude of anti-cancer response of the cancer patient to the anti-cancer vaccine or anti-cancer therapy. It can also be used to increase the number of responders in a population of cancer patients. Thus the antibodies can be used to overcome immune suppression found in patients refractory to anti-cancer vaccines or treatment.
  • the anti-cancer vaccines can be any that are known in the art, including, but not limited to whole tumor cell vaccines, isolated tumor antigens or polypeptides comprising one or more epitopes of tumor antigens.
  • Expression of marker in T-cells can be modulated at the transcriptional or translational level. Agents which are capable of such modulation can be identified using the screening assays described below.
  • RNA small interference RNA
  • Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA.
  • the 5′coding portion of the polynucleotide sequence which codes for the mature polypeptides of the present invention, is used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length.
  • a DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription (triple helix—see Lee et al., Nucl. Acids Res., 6: 3073 (1979); Cooney et al, Science, 241: 456 (1988); and Dervan et al., Science, 251: 1360 (1991)), thereby preventing transcription and the production of the marker.
  • the antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the marker polypeptide (Antisense—Okano, J. Neurochem., 56: 560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988)).
  • the oligonucleotides described above can also be delivered to cells by antisense expression constructs such that the antisense RNA or DNA may be expressed in vivo to inhibit production of the marker. Such constructs are well known in the art.
  • Antisense constructs, antisense oligonucleotides, RNA interference constructs or siRNA duplex RNA molecules can be used to interfere with expression of the marker.
  • at least 15, 17, 19, or 21 nucleotides of the complement of marker mRNA sequence are sufficient for an antisense molecule.
  • at least 19, 21, 22, or 23 nucleotides of marker are sufficient for an RNA interference molecule.
  • an RNA interference molecule will have a 2 nucleotide 3′overhang. If the RNA interference molecule is expressed in a cell from a construct, for example from a hairpin molecule or from an inverted repeat of the desired marker sequence, then the endogenous cellular machinery will create the overhangs.
  • siRNA molecules can be prepared by chemical synthesis, in vitro transcription, or digestion of long dsRNA by Rnase III or Dicer. These can be introduced into cells by transfection, electroporation, or other methods known in the art. (See Hannon, G J, 2002, RNA Interference, Nature 418:244-251; Bernstein E et al., 2002, The rest is silence. RNA 7:1509-1521; Hutvagner G et al. 9 RNAi: Nature harbors a double-strand. Curr. Opin. Genetics & Development 12: 225-232, 2002, A system for stable expression of short interfering RNAs in mammalian cells.
  • Short hairpin RNAs induce sequence-specific silencing in mammalian cells. Genes & Dev. 16: 948-958; Paul C P, Good P D, Winer I, and Engelke D R. (2002). Effective expression of small interfering RNA in human cells. Nature Biotechnol. 20: 505-508; Sui G, Soohoo C, Affar E-B, Gay F, Shi Y, Forester W C, and Shi Y. (2002). A DNA vector-based RNAi technology to suppress gene expression in mammalian cells. Proc. Natl. Acad. Sci. USA 99 (6): 5515-5520; Yu J-Y, DeRuiter S L, and Turner D L. (2002). RNA interference by expression of short-interfering RNAs and hairpin RNAs in mammalian cells. Proc. Natl. Acad. Sci. USA 99 (9): 6047-6052).
  • modulators of markers activity that are useful in the methods of the invention can be identified using two-hybrid screens, conventional biochemical approaches, and cell-based screening techniques, such as screening candidate molecules for an ability to bind to marker or screening for compounds which inhibit marker activity in cell culture.
  • the method may identify agents that directly interact with and modulate the marker, as well as agents that indirectly modulate marker activity by affecting a step in the marker signal transduction pathway.
  • Cell-based assays employing cells which express the marker can employ cells which are isolated from mammals and which naturally express the marker. Alternatively, cells which have been genetically engineered to express the marker can be used. Preferably the genetically engineered cells are T-cells.
  • Agents which modulate the marker activity by modulating the marker gene expression can be identified in cell based screening assays by measuring amounts of the marker protein in the cells in the presence and absence of candidate agents.
  • the marker protein can be detected and measured, for example, by flow cytometry using anti-marker specific monoclonal antibodies.
  • Marker mRNA can also be detected and measured using techniques known in the art, including but not limited to Northern blot, RT-PCR, and array hybridization.
  • marker inhibitors may be administered to an organism to increase the number of T-cells in the organism. This method may be useful for treating organisms suffering from conditions resulting in a low T-cell population. Such conditions include disorders involving unwanted cellular invasion or growth, such as tumor growth or cancer. Marker inhibitors may also be useful when administered in combination with conventional therapeutics to treat T-cell proliferation sensitive disorders. For instance, a tumor, which is a T-cell proliferation sensitive disorder, is conventionally treated with a chemotherapeutic agent which functions by killing rapidly dividing cells.
  • marker inhibitors of the invention when administered in conjunction with a chemotherapeutic agent enhance the tumoricidal effect of the chemotherapeutic agent by stimulating T-cell proliferation to enhance the immunological rejection of the tumor cells.
  • marker activators (agonists) or expression enhancers may be administered to an organism to decrease the number of T-cells, in particular tumor-infiltrating regulatory T cells, in the organism and thereby decrease deleterious T-cell activity.
  • the methods of the invention may be applied to any organism which contains T-cells that express the marker. This includes, but is not limited to, any mammal and particularly includes humans and mice.
  • the effective amount of the marker modulator used will vary with the particular modulator being used, the particular condition being treated, the age and physical condition of the subject being treated, the severity of the condition, the duration of the treatment, the nature of the concurrent therapy (if any), the specific route of administration and similar factors within the knowledge and expertise of the health practitioner.
  • an effective amount can depend upon the degree to which an individual has abnormally depressed levels of T cells.
  • the pharmaceutical preparations of the invention are applied in pharmaceutically-acceptable amounts and in pharmaceutically-acceptably compositions.
  • Such preparations may routinely contain salt, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents.
  • the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically-acceptable salts thereof and are not excluded from the scope of the invention.
  • Such pharmacologically and pharmaceutically-acceptable salts include, but are not limited to those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic, and the like.
  • pharmaceutically-acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts. Marker modulators may be combined, optionally, with a pharmaceutically-acceptable carrier.
  • pharmaceutically-acceptable carrier means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration into a human.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • the components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
  • the pharmaceutical compositions may contain suitable buffering agents, including: acetic acid in a salt; citric acid in a salt; boric acid in a salt; and phosphoric acid in a salt.
  • suitable buffering agents including: acetic acid in a salt; citric acid in a salt; boric acid in a salt; and phosphoric acid in a salt.
  • the pharmaceutical compositions also may contain, optionally, suitable preservatives, such as: benzalkonium chloride; chlorobutanol; parabens and thimerosal.
  • Compositions suitable for parenteral administration conveniently comprise a sterile aqueous preparation of the anti-inflammatory agent, which is preferably isotonic with the blood of the recipient. This aqueous preparation may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation also may be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in butane diol.
  • a non-toxic parenterally-acceptable diluent or solvent for example, as a solution in butane diol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • fatty acids such as oleic acid may be used in the preparation of injectables.
  • Carrier formulation suitable for oral, subcutaneous, intravenous, intramuscular, etc. administrations can be found in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa.
  • a variety of administration routes are available. The particular mode selected will depend, of course, upon the particular drug selected, the severity of the condition being treated and the dosage required for therapeutic efficacy.
  • the methods of the invention may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects.
  • modes of administration include oral, rectal, topical, nasal, interdermal, or parenteral routes.
  • parenteral includes subcutaneous, intravenous, intramuscular, or infusion. Intravenous or intramuscular routes are not particularly suitable for long-term therapy and prophylaxis. They could, however, be preferred in emergency situations. Oral administration will be preferred because of the convenience to the patient as well as the dosing schedule.
  • compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well-known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the active agent into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • Compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active agent.
  • Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as a syrup, elixir or an emulsion.
  • Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the active agent, increasing convenience to the subject and the physician.
  • Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer base systems such as poly (lactide-glycolide), copolyoxalates, polycaprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid, and polyanhydrides. Microcapsules of the foregoing polymers containing drugs are described in, for example, U.S. Pat. No. 5,075,109.
  • Delivery systems also include non-polymer systems that are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides; hydrogel release systems; sylastic systems; peptide based systems; wax coatings; compressed tablets using conventional binders and excipients; partially fused implants; and the like.
  • lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides
  • hydrogel release systems such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides
  • sylastic systems such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di- and tri-glycerides
  • peptide based systems such as fatty acids
  • wax coatings such as those described in U.S. Pat. Nos.
  • a long-term sustained release implant may be particularly suitable for treatment of chronic conditions.
  • Long-term release are used herein, means that the implant is constructed and arranged to deliver therapeutic levels of the active ingredient for at least 30 days, and preferably 60 days.
  • Long-term sustained release implants are well-known to those of ordinary skill in the art and include some of the release systems described above.
  • FIG. 1 Purification, functional characterization and expression of immune checkpoints in tumor infiltrating cells.
  • C Z-score normalized RNA-seq expression values of immune checkpoints genes are represented as a heatmap. Cell populations are reported in the upper part of the graph, while gene names have been assigned to heatmap rows. Hierarchical clustering results are shown as a dendrogram drawn on the left side of the matrix. Colon tissues are indicated as C, lung tissues as L and peripheral blood as B. See also FIG. 6 .
  • FIG. 2 Differential expression analysis identifies co-regulated genes in tumor infiltrating Treg cells
  • FIG. 3 Single cell analysis of tumor infiltrating Treg cells
  • FIG. 4 Expression of tumor-infiltrating Treg cells protein signatures in CRC and NSCLC samples.
  • FIG. 5 Prognostic value of signature transcripts of tumor infiltrating Treg cells.
  • FIG. 6 related to FIG. 1 . Transcriptome analysis of tumor infiltrating lymphocytes.
  • RNA-seq expression values (normalized counts) of FOXP3, TBX21 and RORC in CD4+ Th1, Th17 and Treg cells from CRC (C), NSCLC (L) or peripheral blood (PB) of healthy donors.
  • RNA-seq normalized counts data for selected immune checkpoints and their ligands are shown as histogram plot. Cell population names are reported in the lower part of each graph, while gene names are shown in the upper part.
  • FIG. 7 related to FIG. 3 . Single-cell analysis of tumor infiltrating Treg cells.
  • CD4+ Treg, Th1, Th17, Th2, CD8+ T cells and B cell markers expression (percentage of expressing cells) in single Treg cells purified from NSCLC and CRC.
  • FIG. 8 related to FIG. 4 . Comparison of BATF expression in CD4+ Treg vs Th17 cells.
  • FIG. 9 related to FIG. 5 . Expression levels of tumour-infiltrating Treg signature genes.
  • RNA-seq normalized counts data of three tumour-infiltrating Treg signature genes (MAGEH1 (panel A), LAYN (panel B) and CCR8 (panel C)) across listed cell populations.
  • Non-small-cell lung cancer were cut into pieces and single-cell suspensions were prepared by using the Tumor Dissociation Kit, human and the gentleMACSTM Dissociator (Miltenyi Biotech cat. 130-095-929) according to the accompanying standard protocol. Cell suspensions were than isolated by ficoll-hypaque density-gradient centrifugation (Amersham Bioscience). Colorectal cancer (CRC) specimens were cut into pieces and incubated in DTT 0.1 mM (Sigma-Aldrich) for 10 min, then extensively washed in HBSS (Thermo Scientific) and incubated in 1 mM EDTA (Sigma-Aldrich) for 50 min at 37° C. in the presence of 5% CO2.
  • DTT 0.1 mM Sigma-Aldrich
  • HBSS Thermo Scientific
  • 1 mM EDTA Sigma-Aldrich
  • CD4 T cell subsets were purified by FACS sorting using the following fluorochrome conjugated antibodies: anti-CD4 APC/Cy7 (Biolegend clone OKT4), anti-CD27 Pacific Blue (Biolegend, clone M-T271), anti-IL7R PE (Milteniy, clone MB15-18C9), anti-CD25 PE/Cy7 (eBioscience, clone BC96), anti-CXCR3 PE/Cy5 (BD, clone 1C6/CXCR3), anti-CCR6 APC (Biolegend, clone G034E3) and anti-CCRS FITC (Biolegend, clone j418F1) using a FACSAria II (BD).
  • anti-CD4 APC/Cy7 Biolegend clone OKT4
  • anti-CD27 Pacific Blue Biolegend, clone M-T271
  • anti-IL7R PE Milteniy,
  • Intracellular staining was performed using eBioscience Foxp3 staining kit according to the manufactured's protocol (eBioscience cat 00-5523-00). Briefly cells were harvested and fixed for 30 min in fixation/permeabilization buffer at 4° C., and than stained with anti-FOXP3 antibody (eBioscience, clone 236A/E7) and anti-BATF (eBioscience clone MBM7C7) in permeabilisation buffer for 30 min at 4° C. Cells were then washed two times, resuspended in FACS washing buffer and analyzed by flow cytometry.
  • CFSE carboxyfluorescein diacetate succinimidyl ester
  • RNA from tumor-infiltrating lymphocytes was isolated using mirVana Isolation Kit. Residual contaminating genomic DNA was removed from the total RNA fraction using Turbo DNA-free (Thermo Fisher). The RNA yields were quantified using the QuantiFluor RNA System (Promega) and the RNA quality was assessed by the Agilent 2100 Bioanalyzer (Agilent). Libraries for Illumina sequencing were constructed from 50 ng of total RNA with the Illumina TruSeq RNA Sample Preparation Kit v2 (Set A). The generated libraries were loaded on to the cBot (Illumina) for clustering on a HiSeq Flow Cell v3. The flow cell was then sequenced using a HiSeq 2500 in High Output mode (Illumina). A paired-end (2 ⁇ 125) run was performed.
  • Adapter1 (SEQ ID NO: 710) AGATCGGAAGAGCACACGTCTGAACTCCAGTCACNNNNNNATCTCGTA TGCCGTCTTCTGCTTG Adapter2: (SEQ ID NO: 711) AGATCGGAAGAGCGTCGTGTAGGGAAAGAGTGTAGATCTCGGTGGTCG CCGTATCATT
  • Trimming was performed on raw reads using Trimmomatic (Bolger et al., 2014): standard parameters for phred33 encoding were used: ILLUMINACLIP (LEADING:3 TRAILING:3 SLIDINGWINDOW:4:15), MINLEN parameter was set to 50.
  • Differential expression analysis differential expression analyses of tumor-infiltrating CD4+ Treg/Th1/Th17 subsets vs. CD4+ Treg/Th1/Th17 from PBMC were performed using DESeq2. Upregulated/downregulated genes were selected for subsequent analyses if their expression values were found to exceed the threshold of 0.05 FDR (Benjamini-Hochberg correction).
  • Treg cells from 5 CRC and 5 NSCLC specimens were isolated as previously described (See also Table II). Single cells were captured on a microfluidic chip on the C1 System (Fluidigm) and whole-transcriptome amplified. cDNA was prepared on chip using the SMARTer Ultra Low RNA kit (Clontech). Cells were loaded onto the chip at a concentration of 3-5E5 cells/ml, stained for viability (LIVE/DEAD cell viability assay; Thermo Fisher) and imaged by phase-contrast and fluorescence microscopy to assess the number and viability of cells per capture site. Only single, live cells were included in the analysis.
  • harvested cDNA was pre-amplified using a 0.2 ⁇ pool of primers prepared from the same gene expression assays to be used for qPCR. Pre-amplification allows for multiplex sequence-specific amplification 78 targets.
  • a 1.25 ⁇ l aliquot of single cell cDNA was pre-amplified in a final volume of 5 ⁇ l using 1 ⁇ l of PreAmp Master Mix (Fluidigm) and 1.25 ⁇ l pooled TaqMan assay mix (0.2 ⁇ ).
  • cDNA went through amplification by denaturing at 95° C. for 15 s, and annealing and amplification at 60° C. for 4 min for 20 cycles.
  • pre-amplified cDNA was diluted 1:5 by adding 20 ⁇ l TE Buffer to the final 5 ⁇ l reaction volume for a total volume of 25 ⁇ l.
  • the Quality Threshold in the BioMarkTM Analysis software is a qualitative tool designed to measure the “quality” of each amplification curve. Basically, each amplification curve is compared to an ideal exponential curve and as the quality score approaches 1 the closer it is to ideal. The further the curve is from ideal, its quality score approaches 0.
  • the default cutoff of 0.65 is an arbitrary value set by Fluidigm. Any curve above 0.65 passes. Any curve below, fails. Baseline correction was set on Linear (Derivative)[default].
  • Ct Threshold Method was set on Auto (Detectors). This method independently calculates a threshold for each detector on a chip.
  • Single-cell qPCR data are inherently noisy, and due the limitations of current technologies the expression patterns of a certain number of genes may be affected by the ‘dropout effect’.
  • Inventors performed a gene selection procedure in order to take into account this ‘dropout’ effect and discard those genes whose expression values cannot be reliably used in a binary comparison (tumor-peripheral vs blood).
  • Inventors fitted a number of parametric distributions to the ratios of detected genes on the total number of tumor cells (both NSCLC and CRC) and selected the reciprocal inverse Gaussian continuous random variable as best fit.
  • x ⁇ i , j x i , j x CD ⁇ ⁇ 3 ⁇ ⁇ G , j :
  • a threshold on the is required and defined as
  • T ( Upper , Lower ) median ⁇ ( x ⁇ i , j ) ⁇ ⁇ ⁇ ( x ⁇ i , j ) 10
  • T Upper,Lower
  • Inventors examined the prognostic significance of tumor Treg cells transcripts by using log-rank statistics; a p-value of less than 0.05 was considered statistically significant. Since the log-rank test resulted in a p-value of less than 0.05, a post stage comparison by means of box plot representation was performed in order to evaluate the correlation degree between the expression level of the transcripts and tumor stages in the cohort of CRC patients. The annotation was normalized to four tumor stages (1, 2, 3, 4).
  • accession numbers for the present data are as follows: ENA: PRJEB11844 for RNA-seq tumor and tissue infiltrating lymphocytes; ArrayExpress: E-MTAB-2319 for RNA-seq human lymphocytes datasets; ArrayExpress: E-MTAB-513 for Illumina Human BodyMap 2.0 project; GEO: GSE50760 for RNA-seq datasets CRC; GEO: GSE40419 for RNA-seq datasets NSCLC; GEO: GSE17536 for CRC expression profiling by array; and GEO: GSE41271 for NSCLC expression profiling by array.
  • the probability of surface exposure of the proteins encoded by the genes of interest was determined by a combination of four different cell localization prediction algorithms: Yloc (Briesemeister et al, 2010), TMHMM (http://www.cbs.dtu.dk/services/TMHMM/), SignalP (http://www.cbs.dtu.dk/services/SignalP/) and Phobius (Käll et. al, 2007).
  • Yloc is a interpretable system offering multiple predictive models in animal version; inventors used both YLoc-LowRes predicting into 4 location (nucleus, cytoplasm, mitochondrion, secretory pathway) and Yloc-HighRes predicting into 9 locations (extracellular space, plasma membrane, nucleus, cytoplasm, mitochondrion, endoplasmic reticulum, peroxisome, Golgi apparatus, and lysosome).
  • TMHMM and SignalP were developed by the bioinformatic unit of the technical University of Denmark for the prediction of transmembrane helices and the presence and location of signal peptide cleavage sites in amino acid sequences, respectively.
  • Phobius is a combined transmembrane topology and signal peptide predictor.
  • CD4 + lymphocytes subsets from two different tumors, NSCLC and CRC, from the adjacent normal tissues, and from peripheral blood samples. From all these tissues, the inventors purified by flow cytometry ( FIGS. 1A and 6A and 6B ) CD4 + Treg (36 samples from 18 individuals), Th1 (30 samples from 21 individuals) and Th17 (22 samples from 14 individuals) cells (Table I and Table II).
  • NSCLC PATIENTS LIST RNA SEQUENCING
  • T)Th1 (T)Th17 (T)Treg H)Th1 (H)Th17 (H)Treg
  • SMOKE HABIT STATUS PATIENT1 SQ_0342 PREVIOUS SMOKER > 15 y ALIVE PATIENT2 SQ_0339 PREVIOUS SMOKER ⁇ 15 y ALIVE PATIENT3 SQ_0365 SQ_0375 PREVIOUS SMOKER ⁇ 15 y ALIVE PATIENT4 SQ_0366 SQ_0374 SQ_0341 PREVIOUS SMOKER > 15 y ALIVE PATIENT5 SQ_0364 SQ_0373 SQ_0350 SQ_0351 SMOKER DEAD PATIENT6 SQ_0358 SQ_0336 SQ_0350 SQ_0351 PREVIOUS SMOKER ⁇ 15 y ALIVE PATIENT7 SQ_0363 SQ_0376 SQ_0334 SQ_03
  • Treg cell function inventors tested their suppressor activity and showed that Treg cells infiltrating either type of tumor tissues have a remarkably stronger suppressive activity in vitro compared to Treg cells isolated from the adjacent normal tissue and peripheral blood of the same patients ( FIG. 1B ).
  • RNA-sequencing data of CD4+ T cells infiltrating both CRC and NSCLC and their matched normal tissues, to quantitate mRNA expression of known immune checkpoints and their ligands.
  • inventors analyzed RNA-seq data of CRC and NSCLC, as well as of normal colon and lung samples.
  • PBMCs peripheral blood mononuclear cells
  • RNA-seq normalized counts data for selected immune checkpoints genes and their ligands in all the subsets analyzed.
  • the inventors then asked whether tumor infiltrating Treg cells could be defined by specific gene expression patterns.
  • the inventors included in the expression pattern analyses the transcriptome dataset they previously obtained from different T and B lymphocyte subsets purified from PBMCs (Ranzani et al., 2015). In so doing, the inventors obtained a signature of 328 transcripts whose expression is higher in tumor infiltrating Treg cells (Wilcoxon Mann Whitney test p ⁇ 2.2 ⁇ 10-16) ( FIG. 2 , and Table IV compared to the other lymphocyte subsets purified from non-tumoral tissues and from PBMCs of healthy or neoplastic patients.
  • Treg cells display the most pronounced differences in transcripts expression among CD4 + T cell subsets infiltrating normal and tumor tissues.
  • the inventors defined a subset of signature genes that describe the specific gene expression profile of tumor infiltrating Treg cells.
  • the inventors then looked at the single cell level for the differential expression profile of signature genes of tumor infiltrating Treg cells.
  • the inventors isolated CD4 + T cells from 5 CRC and 5 NSCLC tumor samples as well as from 5 PBMCs of healthy individuals (Table II), purified Treg cells, and using an automated microfluidic system (C1 Fluidigm) captured single cells (a total of 858 Treg cells: 320 from CRC and 286 from NSCLC; 252 from PBMCs of healthy individuals).
  • C1 Fluidigm automated microfluidic system
  • the inventors then assessed by high throughput RT-qPCR (Biomark HD, Fluidigm) the expression of 79 genes selected among the highly expressed (>10 FKPM) tumor Treg cell signature genes ( FIGS. 3A, 3C and 7 ).
  • Treg cell markers i.e., FOXP3 + and IL2RA
  • FIG. 3B The percentage of co-expression between these Treg cell markers and the 79 genes selected among the tumor-infiltrating-Treg-cell signature genes ranged between 81% of TIGIT and 0.59% of CGA ( FIG. 3B ).
  • the expression of Treg signature genes in the RNA-seq of the whole Treg cell population correlated with the percentage of single cells expressing the different genes ( FIG. 3C ).
  • the forty-five signature transcripts of tumor infiltrating Treg cells detected above this threshold were in most cases significantly over-expressed in Treg cells from both tumors (39 over 45, 87%; Wilcoxon Mann Whitney test p ⁇ 0.05) or in one tumor type (43 over 45, 96%; FIG. 3D ).
  • Treg cells Homogeneity of the purified tissue infiltrating Treg cells can be affected by the carry-over of cells from other lymphocyte subsets.
  • the single cell RT-qPCR analyses of Treg cells was performed including markers specific for other lymphocytes subsets (i.e., Th1, Th2, Th17, Tfh, CD8 T cells, B cells) ( FIG. 7 ).
  • Our data showed that only a very low fraction of the purified single cells displayed markers of lymphocytes subsets different from Treg cells ( FIG. 7 ).
  • the inventors then assessed at the single cell level by flow cytometry the protein expression of ten representative signature genes present in CRC and NSCLC infiltrating Treg cells, adjacent normal tissues, and patients PBMCs.
  • ten proteins two are proteins (OX40 and TIGIT) whose relevance for Treg cells biology has been demonstrated (Joller et al., 2014; Voo et al., 2013), seven are proteins (BATF, CCR8, CD30, IL-1R2, IL-21R, PDL-1 and PDL-2) whose expression has never been described in tumor-infiltrating Treg cells, and one protein, 4-1BB, is a co-stimulatory receptor expressed on several hematopoietic cells, whose expression on Treg cells has been shown to mark antigen-activated cells (Schoenbrunn et al., 2012).
  • Our findings showed that all these proteins were upregulated ( FIGS. 4A and 4B ), at different extent, in tumor infiltrating Treg cells compared to the Tre
  • the inventors asked whether the expression of the tumor-Treg signature transcripts correlated with disease prognosis in CRC and NSCLC patients.
  • the inventors therefore interrogated for expression of Treg signature genes transcriptomic datasets obtained from resected tumor tissues of a cohort of 177 CRC patients (GSE17536 (Smith et al., 2010) and of a cohort of 263 NSCLC patients (GSE41271—(Sato et al., 2013), and correlated high and low gene expression levels with the 5-years survival data.
  • FIG. 9A-C Among those genes whose expression is highly enriched in tumor infiltrating Treg cells, LAYN, MAGEH1 and CCR8 were selected as they are the three genes more selectively expressed ( FIG. 9A-C ). To normalize for differences in T cell densities within the resected tumor tissues, the inventors used the ratio between expression of the selected signature genes and CD3G. Remarkably, it was found that high expression of the three signature genes is in all cases correlated with a significantly reduced survival ( FIG. 5A ). Interestingly, it was also observed that expressions of the three signature genes increased with tumor staging of CRC patients ( FIG. 5B ).
  • Genes of table VI are characterized by their Ensembl Gene accession number (ENSG), retrievable in the public database EnsEMBL (http://www.ensembl.org). Each related protein isoform is characterized by an Ensembl transcript accession number (ENST) and an Ensembl protein accession number (ENSP).
  • ENSG Ensembl Gene accession number
  • ENST Ensembl transcript accession number
  • ENSP Ensembl protein accession number
  • tumor-T reg An important aspect to be verified in the selection of potential targets of tumor-T reg is that the protein isoforms predicted to be surface exposed/membrane associated by the cell localization algorithms are indeed expressed in tumor Treg cells.
  • total RNA was extracted from tumor Treg cells isolated from NSCLC or CRC samples and subjected to RT-PCR using specific primer pairs able to discriminate the different isoforms annotated for each gene.
  • Exemplificative results of protein isoforms predicted to be surface exposed and detected in tumor T reg cells is reported in Table VII.
  • an example of RT-PCR analysis carried out for SIRPG is reported in FIG. 10 .
  • transcripts detected in tumor-infiltrating Treg cells GENE Surface predicted isoform detected SYMBOL in Tumor Treg cells CCR8 ENST00000326306 LAYN ENST00000375614 and/or ENST00000533265 and/or ENST00000375615 and/or ENST00000525126 CD7 ENST00000312648 and/or ENST00000584284 CXCL13 ENST00000286758 FCRL3 ENST00000492769 and/or ENST00000368184 and/or ENST00000368186 and/or ENST00000485028 IL1R2 ENST00000332549 and/or ENST00000393414 IL21R ENST00000337929 and/or ENST00000395754 and/or ENST00000564089 NTNG2 ENST00000393229 SIRPG ENST00000303415 and/or ENST00000216927 and/or ENST00000344103 and/or ENST00000381580 and/
  • Treg cells Diversity of tumor infiltrating Treg cells should be fully elucidated to understand their functional relevance and prognostic significance in different types of cancer, and to possibly improve the therapeutic efficacy of Treg cell modulation through the selective depletion of tumor infiltrating Treg cells.
  • the transcriptome analysis performed on CRC- and NSCLC-infiltrating T cells showed that tumor-infiltrating Treg cells are different from both circulating and normal tissue-infiltrating Tregs, suggesting that the tumor microenvironment influences specific gene expression in Treg cells.
  • Our findings further support the view that Treg cells from different tissues are instructed by environmental factors to display different gene expression profiles (Panduro et al., 2016).
  • tumor-infiltrating-Treg signature genes are not only largely shared between CRC and NSCLC infiltrating cells, but are also conserved in breast and gastric cancers as well as in CRC and NSCLC metastatic tumors (in liver and brain respectively) suggesting that expression of these genes is a common feature of tumor infiltrating Treg cells that may correlate with Treg cells specific function within the tumor microenvironment.
  • checkpoints Although our knowledge on the function of immune checkpoints on lymphocytes is still incomplete, agonist or antagonist monoclonal antibodies targeting checkpoints are in clinical development. Interestingly, it has been found that some of these checkpoints (such as GITR, OX40, TIGIT, LAG-3 and TIM-3) and some of their ligands (such as OX40LG, Galectin-9, CD70) are upregulated also in tumor infiltrating Treg cells, and this fact should be taken into account in interpreting clinical results with checkpoint inhibitors. Indeed, it is likely that assessment of the expression of checkpoints and of their ligands on the various subsets of tumor infiltrating lymphocytes will help to elucidate conflicting results and provide the rationale for combination therapies.
  • checkpoints such as GITR, OX40, TIGIT, LAG-3 and TIM-3
  • ligands such as OX40LG, Galectin-9, CD70
  • tumor infiltrating lymphocytes Single-cell analysis on selected tumor Treg signature genes confirmed the whole transcriptomic data and provided information on the expression frequency of these genes.
  • Tumor infiltrating Treg cells express with high frequency genes that are associated with increased suppressor activity, such as the well characterized OX40, CTLA4 and GITR.
  • IL-1R2 upregulation could be another mechanism that tumor resident Treg cells employ to dampen anti-tumor immune responses through the neutralization of IL-1 ⁇ function on effector cells.
  • PD-L1 and PD-L2 expression has been recently reported on activated T cells or APCs (Boussiotis et al., 2014; Lesterhuis et al., 2011; Messal et al., 2011) but, to the best of our knowledge, neither PD-L2 nor PD-L1 expression has ever been reported in Treg cells, and our finding that they are overexpressed in tumor infiltrating Treg cells adds an additional level of complexity to the PD1/PD-Ls immunomodulatory axis within the tumor microenvironment.
  • BATF is a transcription factor that has been mainly associated to Th17 development and CD8 + T cells differentiation (Murphy et al., 2013).
  • Treg cells express high amounts of 4-1BB (CD137) a marker of TcR mediated activation (Schoenbrunn et al., 2012) and have shown they display very high suppressor function on effector T cell proliferation. It could be that expression of the signature genes correlated with the enhanced suppressive ability and so contributed to the establishment of a strong immunosuppressive environment at tumor sites. A corollary to our findings would have that increased number of Treg cells in the tumor environment should associate with a worst clinical outcome.
  • 4-1BB CD137
  • CCL18 the ligand of CCR8 (Islam et al., 2013), is highly expressed in different tumors including NSCLC (Chen et al., 2011; Schutyser et al., 2005).
  • NSCLC Chotyser et al., 2005.
  • the high specificity of CCR8 expression on tumor infiltrating Treg cells suggests it could be a new interesting therapeutic target to inhibit Treg cells trafficking to tumor sites, without disturbing recruitment of other effector T cells that do not express CCR8.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Wood Science & Technology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physics & Mathematics (AREA)
  • Oncology (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Urology & Nephrology (AREA)
  • Virology (AREA)
  • Developmental Biology & Embryology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
US16/301,805 2016-05-16 2017-05-15 Markers selectively deregulated in tumor-infiltrating regulatory t cells Abandoned US20190391152A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP16169791 2016-05-16
EP16169791.7 2016-05-16
EP16198724 2016-11-14
EP16198724.3 2016-11-14
PCT/EP2017/061642 WO2017198631A1 (fr) 2016-05-16 2017-05-15 Marqueurs à dérégulation sélective présents dans des lymphocytes t régulateurs infiltrant la tumeur

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/061642 A-371-Of-International WO2017198631A1 (fr) 2016-05-16 2017-05-15 Marqueurs à dérégulation sélective présents dans des lymphocytes t régulateurs infiltrant la tumeur

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/295,965 Continuation US20230375555A1 (en) 2016-05-16 2023-04-05 Markers selectively deregulated in tumor-infiltrating regulatory t cells

Publications (1)

Publication Number Publication Date
US20190391152A1 true US20190391152A1 (en) 2019-12-26

Family

ID=58772864

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/301,805 Abandoned US20190391152A1 (en) 2016-05-16 2017-05-15 Markers selectively deregulated in tumor-infiltrating regulatory t cells
US18/295,965 Pending US20230375555A1 (en) 2016-05-16 2023-04-05 Markers selectively deregulated in tumor-infiltrating regulatory t cells

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/295,965 Pending US20230375555A1 (en) 2016-05-16 2023-04-05 Markers selectively deregulated in tumor-infiltrating regulatory t cells

Country Status (11)

Country Link
US (2) US20190391152A1 (fr)
EP (2) EP4092045A1 (fr)
JP (2) JP7358047B2 (fr)
KR (1) KR102623927B1 (fr)
CN (1) CN109715653A (fr)
AU (1) AU2017266686B2 (fr)
BR (1) BR112018073414A2 (fr)
CA (1) CA3023980A1 (fr)
ES (1) ES2922525T3 (fr)
MX (2) MX2018013952A (fr)
WO (1) WO2017198631A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021202678A1 (fr) * 2020-04-01 2021-10-07 Xilio Development, Inc. Cytokines il-12 masquées et leurs produits de clivage
CN117079710A (zh) * 2023-08-18 2023-11-17 上海爱谱蒂康生物科技有限公司 生物标志物及其在预测和/或诊断utuc肌肉浸润中的应用
US11866476B2 (en) 2018-09-27 2024-01-09 Xilio Development, Inc. Masked IL-2-Fc fusion polypeptides

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2863412T3 (es) 2017-03-29 2021-10-11 Shionogi & Co Composición farmacéutica para el tratamiento del cáncer
MX2021002002A (es) 2018-08-23 2021-05-31 Seagen Inc Anticuerpos anti inmunorreceptor de celulas t con dominios ig y motivo inhibidor inmunorreceptor basado en tirosina (tigit).
IL263394A (en) * 2018-11-29 2020-05-31 Amit Ido Methods for activating inactive immune cells and cancer treatment
SG11202106214YA (en) 2018-12-27 2021-07-29 Shionogi & Co Novel anti-ccr8 antibody
EP3921335A4 (fr) * 2019-02-08 2023-02-08 The Regents Of The University Of California Compositions et méthodes impliquant la layiline
CN110187107A (zh) * 2019-04-26 2019-08-30 温州医科大学 一种基于肿瘤组织浸润免疫细胞特征建立的结直肠癌预后评估装置及方法
CN115280148A (zh) * 2020-02-26 2022-11-01 Pam治疗诊断有限公司 测定肽酰甘氨酸α-酰胺化单加氧酶(PAM)的方法及其用于诊断目的的用途
TW202216771A (zh) 2020-06-26 2022-05-01 德商拜耳廠股份有限公司 用於治療應用之ccr8抗體
BR112023024837A2 (pt) 2021-07-27 2024-02-20 Abbvie Inc Anticorpos anti-ccr8
CN114381528A (zh) * 2022-02-14 2022-04-22 天津市泌尿外科研究所 Mettl7a基因及其表达产物在诊断和治疗肿瘤中的用途
KR102492241B1 (ko) * 2022-02-25 2023-01-30 전남대학교산학협력단 Kitenin의 이량체 형성을 저해하는 펩타이드 및 이의 용도
CN116496417B (zh) * 2023-06-27 2023-10-10 北京市肿瘤防治研究所 含有膜型il7的融合蛋白及t细胞
CN117625792B (zh) * 2024-01-23 2024-04-16 杭州华得森生物技术有限公司 用于胃癌诊断和预后判断的生物标志物及其应用

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5075A (en) 1847-04-17 Improvement in cultivators
US109A (en) 1836-12-15 Erawijtg
US3854480A (en) 1969-04-01 1974-12-17 Alza Corp Drug-delivery system
US3832253A (en) 1973-03-21 1974-08-27 Baxter Laboratories Inc Method of making an inflatable balloon catheter
US4667014A (en) 1983-03-07 1987-05-19 Syntex (U.S.A.) Inc. Nonapeptide and decapeptide analogs of LHRH, useful as LHRH antagonists
US4452775A (en) 1982-12-03 1984-06-05 Syntex (U.S.A.) Inc. Cholesterol matrix delivery system for sustained release of macromolecules
CA1200416A (fr) 1983-05-13 1986-02-11 Societe Des Produits Nestle S.A. Procede de production de produit alimentaire
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4954617A (en) 1986-07-07 1990-09-04 Trustees Of Dartmouth College Monoclonal antibodies to FC receptors for immunoglobulin G on human mononuclear phagocytes
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
JPH04167172A (ja) 1990-10-31 1992-06-15 Nec Corp ベクトルプロセッサ
WO1999006561A2 (fr) * 1997-07-29 1999-02-11 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services, National Institutes Of Health Adn de recepteur de chemokine ccr 8, et ses utilisations
AR016681A1 (es) * 1997-11-13 2001-07-25 Schering Corp Deplecion de celulas th2; composiciones; metodos
AU2003280471B2 (en) * 2002-07-01 2009-04-23 Tufts University Methods and compositions for inhibition of multi-drug resistance by hyaluronan oligomers
WO2007044756A2 (fr) * 2005-10-11 2007-04-19 Icos Corporation Anticorps monoclonaux reconnaissant le ccr8 humain
JP2008072939A (ja) * 2006-09-21 2008-04-03 Fujifilm Corp 多発性骨髄腫の検出方法および抑制方法
US20100292303A1 (en) * 2007-07-20 2010-11-18 Birrer Michael J Gene expression profile for predicting ovarian cancer patient survival
WO2009100035A2 (fr) * 2008-02-01 2009-08-13 Wyeth Modulation de lymphocytes t régulateurs et forkhead box p3 (foxp3) par des modulateurs de l’interleukine-21 (il-21) et du récepteur de l’il-21 (il-21r)
WO2013131010A2 (fr) * 2012-03-02 2013-09-06 Icahn School Of Medicine At Mount Sinai Fonction du récepteur de chimiokine ccr8 dans des métastases de mélanome
WO2015017537A2 (fr) * 2013-07-30 2015-02-05 H. Lee Moffitt Cancer Center And Research Institute, Inc. Signature d'expression génique de la récidive du cancer colorectal

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11866476B2 (en) 2018-09-27 2024-01-09 Xilio Development, Inc. Masked IL-2-Fc fusion polypeptides
WO2021202678A1 (fr) * 2020-04-01 2021-10-07 Xilio Development, Inc. Cytokines il-12 masquées et leurs produits de clivage
CN117079710A (zh) * 2023-08-18 2023-11-17 上海爱谱蒂康生物科技有限公司 生物标志物及其在预测和/或诊断utuc肌肉浸润中的应用

Also Published As

Publication number Publication date
CA3023980A1 (fr) 2017-11-23
JP7358047B2 (ja) 2023-10-10
EP3458473B8 (fr) 2022-08-17
CN109715653A (zh) 2019-05-03
JP2023113918A (ja) 2023-08-16
KR102623927B1 (ko) 2024-01-12
MX2018013952A (es) 2019-06-24
MX2022010703A (es) 2022-09-27
AU2017266686A1 (en) 2019-01-03
US20230375555A1 (en) 2023-11-23
EP3458473A1 (fr) 2019-03-27
BR112018073414A2 (pt) 2019-08-27
KR20190006009A (ko) 2019-01-16
WO2017198631A1 (fr) 2017-11-23
JP2019518452A (ja) 2019-07-04
EP3458473B1 (fr) 2022-04-13
ES2922525T3 (es) 2022-09-16
EP4092045A1 (fr) 2022-11-23
AU2017266686B2 (en) 2023-03-09

Similar Documents

Publication Publication Date Title
US20230375555A1 (en) Markers selectively deregulated in tumor-infiltrating regulatory t cells
Gide et al. Distinct immune cell populations define response to anti-PD-1 monotherapy and anti-PD-1/anti-CTLA-4 combined therapy
US11913075B2 (en) Methods and compositions for detecting and modulating an immunotherapy resistance gene signature in cancer
EP3207063B1 (fr) Méthodes d'identification d'épuisement des lymphocytes t au moyen de modulateurs et de biomarqueurs cd39
WO2019070755A1 (fr) Procédés et compositions pour détecter et moduler une signature génétique de résistance à l'immunothérapie dans un cancer
US11740242B2 (en) Modulating biomarkers to increase tumor immunity and improve the efficacy of cancer immunotherapy
CN113260633A (zh) 用于癌症免疫疗法的诊断方法和组合物
WO2021247540A1 (fr) Méthodes permettant de moduler l'expression du cmh-i et leurs utilisations en immunothérapie
CN110945030A (zh) 使用april-taci相互作用的调节剂调节调控性t细胞、调控性b细胞和免疫响应的方法
KR20220022050A (ko) 지속적인 임상 이익을 위한 암 바이오마커
JP2019529437A (ja) 抗pd−l1抗体単独療法に対する固形腫瘍の奏効性を特徴付けるための組成物および方法
EP4093513A1 (fr) Utilisations de biomarqueurs pour améliorer une immunothérapie
US20210347847A1 (en) Therapeutic targeting of malignant cells using tumor markers
EP3759125A2 (fr) Méthodes de traitement du cancer à l'aide de combinaisons d'agents de blocage anti-btnl2 et points de contrôle immunitaires
KR20220031069A (ko) 항-종양 치료의 효과를 진단하기 위한 방법
JP2020536111A (ja) リンパ腫を処置する方法
CN115697357A (zh) T细胞细胞毒性的调节和相关治疗
US20220211848A1 (en) Modulating gabarap to modulate immunogenic cell death
Jain et al. Role of biomarkers in assessing response to immune checkpoint inhibitors in cancer treatment
McEwen et al. ISG15-GRAIL1-CD3 axis influences survival of esophageal adenocarcinoma patients
Girard et al. MAdCAM-1 co-stimulation combined with retinoic acid and TGF-β induces blood CD8+ T cells to adopt a gutCD101+ TRM phenotype

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHECKMAB S.R.L., ITALY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ABRIGNANI, SERGIO;PAGANI, MASSIMILIANO;REEL/FRAME:048729/0994

Effective date: 20190305

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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